Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
HETEROBIFUNCTIONAL COMPOUNDS AND METHODS OF TREATING DISEASE
Document Type and Number:
WIPO Patent Application WO/2024/054955
Kind Code:
A1
Abstract:
The invention provides heterobifunctional compounds, pharmaceutical compositions, and their use in treating disease, such as cancer.

Inventors:
EASTMAN KYLE J (US)
KAYSER-BRICKER KATHERINE J (US)
MOUSSEAU JAMES JOHN (US)
NEKLESA TAAVI (US)
PULEO DAVID E (US)
RAINA KANAK SHAIL (US)
ZAWARE NILESH K (US)
GERRITZ SAMUEL W (US)
BOSANAC TODD (US)
Application Number:
PCT/US2023/073697
Publication Date:
March 14, 2024
Filing Date:
September 08, 2023
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
HALDA THERAPEUTICS OPCO INC (US)
International Classes:
C07D401/04; A61K31/4545; A61K31/497; A61K31/5025; A61K31/506; A61P35/00; C07D487/04
Domestic Patent References:
WO2018144649A12018-08-09
WO2019199816A12019-10-17
WO2020214952A12020-10-22
WO2021127443A12021-06-24
WO2023059581A12023-04-13
Foreign References:
CN113582974A2021-11-02
Other References:
MU, X. ET AL.: "Protein targeting chimeric molecules specific for dual bromodomain 4 (BRD4) and Polo-like kinase 1 (PLK1) proteins in acute myeloid leukemia cells", BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS, vol. 521, 2020, pages 833 - 839, XP085990260, DOI: 10.1016/j.bbrc.2019.11.007
Attorney, Agent or Firm:
DAVIS, Chad E. et al. (US)
Download PDF:
Claims:
Claims: 1. A compound represented by Formula I: or a pharmaceutically acceptable salt thereof; wherein: R1 is phenyl substituted by cyano, halogen, and m occurrences of R4; R2 and R3 are independently hydrogen or C1-4 alkyl; R4 is C1-4 alkyl; R5 represents independently for each occurrence C1-4 alkyl or halogen; A1 is a pyridazinylene, pyrimidinylene, pyrazinylene, pyridinylene, or phenylene, each of which is substituted with n occurrences of R5; L is a linker; and A2 is one of the following: A3 is phenylene, a 3-10 membered saturated monocyclic, bicyclic or spirocyclic carbocycylene, or C1-6 alkylene; R1B represents independently for each occurrence halo, cyano, C1-4 alkoxy, C1-4 alkyl, C1-4 haloalkyl, or cyclopropyl; R2B is hydrogen, halo, C1-4 alkyl, or C1-4 haloalkyl; R3B is -N(R8B)SO2R9B, -SO2N(R8B)2, -SO2R9B, -(C1-6 alkylene)-SO2R9B, C1-6 hydroxyalkyl, -O-(phenyl), or one of the following ring groups: (a) a 4-7 membered saturated carbocyclic ring in which one CH2 is replaced with SO2, (b) a 4-7 membered saturated heterocyclic ring containing 1 or 2 heteroatoms independently selected from nitrogen, in which one CH2 is replaced with SO2, or (c) a 3-5 membered heterocyclic ring containing 1 or 2 heteroatoms independently selected from nitrogen and oxygen; wherein each phenyl and heterocyclic ring is substituted with q occurrences of R10B; R4B is hydrogen, halo, or C1-4 alkyl; R5B is C1-4 alkyl or C3-4 cycloalkyl; R6B is hydrogen, C1-4 alkyl, or C3-4 cycloalkyl; R7B represents independently for each occurrence C1-4 alkyl or C3-4 cycloalkyl; R8B represents independently for each occurrence hydrogen or C1-4 alkyl; or two occurrences of R8B are taken together with the nitrogen atom to which they are attached to form a 3-7 membered heterocyclic ring containing 1 or 2 heteroatoms independently selected from nitrogen and oxygen; or R8B and R9B are taken together with their intervening atoms to a form a 5-7 membered ring containing 1, 2, or 3 heteroatoms independently selected from nitrogen, oxygen, and sulfur; R9B is C1-6 alkyl, C1-6 haloalkyl, -(C1-6 alkylene)-(C3-6 cycloalkyl), or C3-6 cycloalkyl; R10B represents independently for each occurrence hydroxyl, halo, or C1-4 alkyl; and m, n, p, q, and t are independently 0, 1, or 2. 2. The compound of claim 1, wherein m is 0. 3. The compound of claim 1, wherein R1 is . 4. The compound of any one of claims 1-3, wherein R2 is hydrogen. 5. The compound of any one of claims 1-4, wherein R3 is hydrogen. 6. The compound of any one of claims 1-5, wherein the compound is a compound of Formula I.

7. The compound of any one of claims 1-5, wherein the compound is a compound of Formula Ia or Ib, or a pharmaceutically acceptable salt thereof: 8. The compound of any one of claims 1-5, wherein the compound is a compound of Formula Ic or a pharmaceutically acceptable salt thereof: 9. The compound of any one of claims 1-5, wherein the compound is a compound of Formula Id or a pharmaceutically acceptable salt thereof: 10. The compound of any one of claims 1-9, wherein A1 is pyridazinylene substituted with n occurrences of R5. 11. The compound of any one of claims 1-9, wherein A1 is . 12. The compound of any one of claims 1-9, wherein A1 is pyrimidinylene substituted with n occurrences of R5. 13. The compound of any one of claims 1-9, wherein A1 is , where ** is the point of attachment to L.

14. The compound of any one of claims 1-9, wherein A1 is , where ** is the point of attachment to L. 15. The compound of any one of claims 1-9, wherein A1 is pyrazinylene substituted with n occurrences of R5. 16. The compound of any one of claims 1-9, wherein A1 is . 17. The compound of any one of claims 1-9, wherein A1 is pyridinylene substituted with n occurrences of R5. 18. The compound of any one of claims 1-9, wherein A1 is , where ** is the point of attachment to L. 19. The compound of any one of claims 1-9, wherein A1 is phenylene substituted with n occurrences of R5. 20. The compound of any one of claims 1-9, wherein A1 is . 21. The compound of any one of claims 1-10, 12, 15, 17, or 19, wherein n is 0. 22. The compound of any one of claims 1-21, wherein 23. The compound of any one of claims 1-22, wherein A3 is a 3-10 membered saturated monocyclic, bicyclic or spirocyclic carbocycylene.

24. The compound of any one of claims 1-21, wherein A2 is 25. The The compound of any one of claims 1-21, wherein A2 is or . 26. The compound of any one of claims 1-25, wherein R1B is halo. 27. The compound of any one of claims 1-25, wherein R1B is fluoro. 28. The compound of any one of claims 1-27, wherein p is 1. 29. The compound of any one of claims 1-25, wherein p is 0. 30. The compound of any one of claims 1-21, wherein A2 is .

31. The compound of any one of claims 1-21, wherein A2 is . 32. The compound of any one of claims 1-31, wherein R2B is hydrogen. 33. The compound of any one of claims 1-32, wherein R3B is -N(R8B)SO2R9B. 34. The compound of any one of claims 1-32, wherein R3B is SO2R9B. 35. The compound of any one of claims 1-32, wherein R3B is -(C1-6 alkylene)-SO2R9B. 36. The compound of any one of claims 1-32, wherein R3B is -SO2N(R8B)2. 37. The compound of any one of claims 1-32, wherein R3B is C1-6 hydroxyalkyl. 38. The compound of any one of claims 1-35, wherein R9B is C1-6 alkyl or C1-6 haloalkyl. 39. The compound of any one of claims 1-38, wherein R4B is hydrogen. 40. The compound of any one of claims 1-39, wherein R5B is C1-4 alkyl. 41. The compound of any one of claims 1-39, wherein R5B is methyl. 42. The compound of any one of claims 1-41, wherein R6B is hydrogen. 43. The compound of any one of claims 1-42, wherein R7B is C1-4 alkyl. 44. The compound of any one of claims 1-42, wherein R7B is methyl. 45. The compound of any one of claims 1-44, wherein t is 2.

47. The compound of any one of claims 1-21, wherein 48. A compound represented by Formula I*: or a pharmaceutically acceptable salt thereof; wherein: R1 is phenyl substituted by cyano, halogen, and m occurrences of R4; R2 and R3 are independently hydrogen or C1-4 alkyl; R4 is C1-4 alkyl; R5 represents independently for each occurrence C1-4 alkyl or halogen; A1 is a pyridazinylene, pyrimidinylene, pyrazinylene, pyridinylene, or phenylene, each of which is substituted with n occurrences of R5; L is a linker; A3 is phenylene, a 3-10 membered saturated monocyclic, bicyclic or spirocyclic carbocycylene, or C1-6 alkylene; R1B represents independently for each occurrence halo, cyano, C1-4 alkoxy, C1-4 alkyl, C1-4 haloalkyl, or cyclopropyl; R2B is hydrogen, halo, C1-4 alkyl, or C1-4 haloalkyl; R4B is hydrogen, halo, or C1-4 alkyl; R5B is C1-4 alkyl or C3-4 cycloalkyl; R6B is hydrogen, C1-4 alkyl, or C3-4 cycloalkyl; R7B represents independently for each occurrence C1-4 alkyl or C3-4 cycloalkyl; and m, n, p, and t are independently 0, 1, or 2. 49. The compound of claim 48, wherein the compound is a compound of Formula Ie or a pharmaceutically acceptable salt thereof: 50. The compound of claim 48 or 49, wherein A1 is pyridazinylene substituted with n occurrences of R5. 51. The compound of claim 48 or 49, wherein A1 is . 52. The compound of any one of claims 48-51, wherein 53. The compound of any one of claims 1-52, wherein L is a bivalent, saturated or unsaturated, straight or branched C1-60 hydrocarbon chain, wherein 0-20 methylene units of the hydrocarbon are independently replaced with -O-, -S-, -N(H)-, -N(C1-6 alkyl)-, -OC(O)-, -C(O)O-, -S(O)-, - S(O)2-, -N(H)S(O)2-, -N(C1-6 alkyl)S(O)2-, -S(O)2N(H)-, -S(O)2N(C1-6 alkyl)-, -N(H)C(O)-, - N(C1-6 alkyl)C(O)-, -C(O)N(H)-, -C(O)N(C1-6 alkyl)-, -OC(O)N(H)-, -OC(O)N(C1-6 alkyl)- , -N(H)C(O)O-, -N(C1-6 alkyl)C(O)O-, -N(C3-6 cycloalkyl)-, -C(halo)2-, -C(H)(halo)-, phenyl, pyridinyl, pyrimidinyl, pyrazinyl, pyridazinyl, optionally substituted 3-10 membered carbocyclyl, or optionally substituted 3-10 membered heterocyclyl containing 1, 2, 3, or 4 heteroatoms independently selected from nitrogen, oxygen, and sulfur.

54. The compound of any one of claims 1-52, wherein L is a bivalent, saturated, straight or branched C3-30 hydrocarbon chain, wherein 0-15 methylene units of the hydrocarbon are independently replaced with -O-, -N(H)-, -N(C1-6 alkyl)-, -OC(O)-, -C(O)O-, -N(H)C(O)-, - N(C1-6 alkyl)C(O)-, -C(O)N(H)-, -C(O)N(C1-6 alkyl)-, 3-10 membered carbocyclyl, or 3-10 membered heterocyclyl containing 1, 2, or 3 heteroatoms independently selected from nitrogen, oxygen, and sulfur. 55. The compound of any one of claims 1-52, wherein L is a bivalent, saturated, straight or branched C3-30 hydrocarbon chain, wherein 0-15 methylene units of the hydrocarbon are independently replaced with -O-, -N(H)-, -N(C1-6 alkyl)-, -OC(O)-, -C(O)O-, -N(H)C(O)-, - N(C1-6 alkyl)C(O)-, -C(O)N(H)-, or -C(O)N(C1-6 alkyl)-. 56. The compound of any one of claims 1-52, wherein L is a bivalent, saturated or unsaturated, straight or branched C5-40 hydrocarbon chain, wherein 1-20 methylene units of the hydrocarbon are independently replaced with -O-, -N(H)-, -N(C1-6 alkyl)-, -N(H)C(O)-, -N(C1-6 alkyl)C(O)-, -C(O)N(H)-, -C(O)N(C1-6 alkyl)-, optionally substituted 3-10 membered carbocyclyl, or optionally substituted 3-10 membered heterocyclyl containing 1, 2, 3, or 4 heteroatoms independently selected from nitrogen, oxygen, and sulfur. 57. The compound of any one of claims 1-52, wherein L is -(3-7 membered, monocyclic, saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen)-(OCH2CH2)1-15-O-***, wherein *** is the point of attachment to A2. 58. The compound of any one of claims 1-52, wherein L is -(3-7 membered, monocyclic, saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen)-(OCH2CH2)1-5-O-***, wherein *** is the point of attachment to A2. 59. The compound of any one of claims 1-52, wherein L is -(3-7 membered, monocyclic, saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen)-(OCH2CH2)6-10-O-***, wherein *** is the point of attachment to A2. 60. The compound of any one of claims 1-52, wherein L is -piperidinylene-(OCH2CH2)1-15-O-***, wherein *** is the point of attachment to A2. 61. The compound of any one of claims 1-52, wherein L is , wherein *** is the point of attachment to A2.

62. The compound of any one of claims 1-52, wherein L is , wherein *** is the point of attachment to A2. 63. The compound of any one of claims 1-52, wherein L is , wherein *** is the point of attachment to A2. 64. The compound of any one of claims 1-52, wherein L is -(3-7 membered, monocyclic, saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen)-(OCH2CH2)1-15- N(H)C(O)-C1-10 alkylene-***, -(3-7 membered, monocyclic, saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen)-(OCH2CH2)1-15-N(C1-4 alkyl)C(O)-C1-10 alkylene-***, -(3-7 membered, monocyclic, saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen)-(OCH2CH2)1-15-C(O)N(H)-C1-10 alkylene-***, or -(3-7 membered, monocyclic, saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen)-(OCH2CH2)1-15-C(O)N(C1-4 alkyl)-C1-10 alkylene-***, wherein *** is the point of attachment to A2. 65. The compound of any one of claims 1-52, wherein L is -piperidinylene-(OCH2CH2)1-5- N(H)C(O)-C1-5 alkylene-***, -piperidinylene-(OCH2CH2)1-5-N(C1-4 alkyl)C(O)-C1-5 alkylene- ***, -piperidinylene-(OCH2CH2)1-5-C(O)N(H)-C1-5 alkylene-***, or -piperidinylene- (OCH2CH2)1-5-C(O)N(C1-4 alkyl)-C1-5 alkylene-***, wherein *** is the point of attachment to A2. 66. The compound of any one of claims 1-52, wherein L is -(3-7 membered, monocyclic, saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen)-(OCH2CH2)1-10-***, - (3-7 membered, monocyclic, saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen)-(C0-10 alkylene)-O-***, or -(3-7 membered, monocyclic, saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen)-C1-10 alkylene, wherein *** is the point of attachment to A2. 67. The compound of any one of claims 1-52, wherein L is -piperidinylene-(OCH2CH2)1-5-***, - piperidinylene-(C0-5 alkylene)-O-***, or -piperidinylene-(C1-5 alkylene)-***, wherein *** is the point of attachment to A2.

68. The compound of any one of claims 1-52, wherein L is -(3-7 membered monocyclic saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen)-X1-***, wherein *** is the point of attachment to A2, and X1 is (i) C1-10 alkylene where 1 or 2 methylene groups are optionally replaced by -O-, -N(H)-, or -N(C1-4 alkyl)-, (ii) a 3-7 membered monocyclic saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen, or (iii) -(3-7 membered monocyclic saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen)-(C1-10 alkylene)-. 69. The compound of any one of claims 1-52, wherein L is -(piperidinylene)-X1-***, wherein *** is the point of attachment to A2, and X1 is (i) C1-5 alkylene where 1 or 2 methylene groups are optionally replaced by -O-, -N(H)-, or -N(C1-4 alkyl)-, (ii) a 3-4 membered monocyclic saturated heterocyclic ring containing 1 heteroatom selected from nitrogen, or (iii) -(3-4 membered monocyclic saturated heterocyclic ring containing 1 heteroatom selected from nitrogen)-(C1-5 alkylene)-. 70. The compound of any one of claims 1-52, wherein L is , wherein *** is the point of attachment to A2, and X1 is (i) C1-10 alkylene where 1 or 2 methylene groups are optionally replaced by -O-, -N(H)-, or -N(C1-4 alkyl)-, (ii) a 3-7 membered monocyclic saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen, or (iii) -(3-7 membered monocyclic saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen)-(C1-10 alkylene)-. 71. The compound of any one of claims 1-52, wherein L is -(piperazinylene)-X1-***, wherein *** is the point of attachment to A2, and X1 is (i) C1-5 alkylene where 1 or 2 methylene groups are optionally replaced by -O-, (ii) a 3-4 membered monocyclic saturated heterocyclic ring containing 1 heteroatom selected from nitrogen, or (iii) -(3-4 membered monocyclic saturated heterocyclic ring containing 1 heteroatom selected from nitrogen)-(C1-5 alkylene)-. 72. The compound of any one of claims 1-52, wherein L is , wherein *** is the point of attachment to A2, and X1 is (i) C1-10 alkylene where 1 or 2 methylene groups are optionally replaced by -O-, (ii) a 3-7 membered monocyclic saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen, or (iii) -(3-7 membered monocyclic saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen)-(C1-10 alkylene)-. 73. The compound of any one of claims 1-52, wherein L is -(3-7 membered monocyclic saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen)-X2-(C1-10 alkylene)- ***, wherein *** is the point of attachment to A2, and X2 is -O-, -N(H)-, or -N(C1-6 alkyl)-. 74. The compound of any one of claims 1-52, wherein L is -(piperidinylene)-X2-(C1-10 alkylene)- ***, wherein *** is the point of attachment to A2, and X2 is -O-, -N(H)-, or -N(C1-6 alkyl)-. 75. The compound of any one of claims 1-52, wherein L is -(piperidinylene)-X2-(a 3-7 membered monocyclic saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen)- ***, wherein *** is the point of attachment to A2, and X2 is -O-, -N(H)-, or -N(C1-6 alkyl)-. 76. The compound of any one of claims 1-52, wherein L is , wherein *** is the point of attachment to A2, and X2 is -O-, -N(H)-, or -N(C1-6 alkyl)-. 77. The compound of any one of claims 1-52, wherein L is , wherein *** is the point of attachment to A2, and X2 is -O-, -N(H)-, or -N(C1-6 alkyl)-. 78. The compound of any one of claims 73-77, wherein X2 is -O-. 79. The compound of any one of claims 1-52, wherein L is -(3-7 membered monocyclic saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen)-X1-***, wherein *** is the point of attachment to A2, and X1 is -(OCH2CH2)1-10 where 1 CH2 group is optionally replaced with -C(H)(C3-6 cycloalkyl)-. 80. The compound of any one of claims 1-52, wherein L is a 7-11 membered spirocyclic or fused bicyclic saturated heterocyclic ring containing 1, 2, or 3 heteroatoms selected from nitrogen and oxygen. 81. The compound of any one of claims 1-52, wherein L is a 7-8 membered spirocyclic or fused bicyclic saturated heterocyclic ring containing 2 heteroatoms selected from nitrogen.

82. The compound of any one of claims 1-52, wherein L is -(3-7 membered monocyclic saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen)-X3-(3-7 membered monocyclic saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen)-, wherein X3 is C1-10 alkylene, -O-, -N(H)-, -N(C1-4 alkyl)-, or a bond. 83. The compound of any one of claims 1-52, wherein L is -(piperidinylene)-(C1-5 alkylene)- (piperazinylene)-***, wherein *** is the point of attachment to A2. 84. The compound of any one of claims 1-52, wherein L is -(piperazinylene)-(azetidinylene)-*** or (azetidinylene)-(piperazinylene)-***, wherein *** is the point of attachment to A2. 85. The compound of any one of claims 1-52, wherein L is -(3-7 membered monocyclic saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen)-X3-(C3-6 cycloalkylene)-O-***, -(3-7 membered monocyclic saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen)-X3-(C3-6 cycloalkylene)-N(H)-***, or -(3-7 membered monocyclic saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen)- X3-(C3-6 cycloalkylene)-N(C1-4 alkyl)-***, wherein *** is the point of attachment to A2, and X3 is C1-10 alkylene, -O-, -N(H)-, -N(C1-4 alkyl)-, or a bond. 86. The compound of any one of claims 1-52, wherein L is -(piperidinylene)-X3-(C3-6 cycloalkylene)-O-***, -(piperidinylene)-X3-(C3-6 cycloalkylene)-N(H)-***, or - (piperidinylene)-X3-(C3-6 cycloalkylene)-***, wherein *** is the point of attachment to A2, and X3 is C1-10 alkylene, -O-, -N(H)-, -N(C1-4 alkyl)-, or a bond. 87. The compound of any one of claims 1-52, wherein L has the formula –(C0-12 alkylene)- (optionally substituted 3-40 membered heteroalkylene)-(C0-12 alkylene)-. 88. The compound of any one of claims 1-52, wherein L is -(7-10 membered spirocyclic heterocyclylene containing 1, 2, 3, or 4 heteroatoms independently selected from nitrogen)-(C0- 12 alkylene)-***, wherein *** is the point of attachment to A2. 89. The compound of any one of claims 1-52, wherein L is -(piperidinylene)-(C0-12 alkylene)-O- ***, wherein *** is the point of attachment to A2. 90. The compound of any one of claims 1-52, wherein L is –(piperidinylene)-(C0-12 alkylene)- (pyrimidinylene)-O-***, wherein *** is the point of attachment to A2.

91. The compound of any one of claims 1-52, wherein L is -(5-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-O-(C1-6 alkylene)-***, -(5- 6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)- (C1-6 alkylene)-***, -(5-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-(C1-6 alkylene)-O-***, -(7-11 membered spirocyclic heterocyclyl containing 1-2 heteroatoms selected from nitrogen)-O-(C0-6 alkylene)-***, -(7-11 membered spirocyclic heterocyclyl containing 1-2 heteroatoms selected from nitrogen)-(C0-6 alkylene)- ***, -(4-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-(C0-6 alkylene)-***, -(4-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-O-***, -(6-11 membered bicyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-(C1-6 alkylene)-***, -(5-6 membered monocyclic heterocyclyl containing1 heteroatom selected from nitrogen, wherein the heterocyclyl is substituted with 0 or 1 occurrence of C1-4 alkenyl)-***, -(5-6 membered monocyclic heterocyclyl containing1 heteroatom selected from nitrogen, wherein the heterocyclyl is substituted with 0 or 1 occurrence of C1-4 alkyl)-***, -(4-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-(C0-6 alkylene)-(4-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-***, or -(4-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-(C0- 6 alkylene)-(4-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-O-***, wherein *** is the point of attachment to A2. 92. The compound of any one of claims 1-52, wherein L is -(4-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-(C0-6 alkylene)-(3-7 membered monocyclic saturated carbocyclic ring)-***, -(4-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-(C0-6 alkylene)-(3-7 membered monocyclic saturated carbocyclic ring)-O-***, -(4-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-O-(4-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-***, -(5-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-(C1- 6 alkylene)-S(O)2- ***, -N(C1-4 alkyl)-(C1-6 alkylene)-O-***, -N(C1-4 alkyl)-(C1-6 alkylene)- ***, -(5-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-(C0-6 alkylene)-(3-7 membered monocyclic saturated carbocyclic ring)-***, -(4-6 membered monocyclic heterocyclyl containing 1 heteroatom selected from nitrogen)-N(C1-4 alkyl)-***, -(4-6 membered monocyclic heterocyclyl containing 1 heteroatom selected from nitrogen)-N(C1-4 alkyl)-***, -(4-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-N(C1-4 alkyl)-(4-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-***, -(4-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-N(C3-6 cycloalkyl)-(4-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-***, or -(5-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-O-(C1-6 alkylene)-O-***, wherein *** is the point of attachment to A2. 93. The compound of any one of claims 1-52, wherein L is -(5-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-(C1-6 alkylene)-O-***, -(5- 6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)- (C1-6 alkylene)-N(C1-4 alkyl)-(3-7 membered monocyclic saturated carbocyclic ring, wherein the carbocyclic ring is substituted with 0-4 occurrences of C1-4 alkyl)-O-***, -(4-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-(C0-6 alkylene)- (7-11 membered spirocyclic heterocyclyl containing 1-2 heteroatoms selected from nitrogen)- O-***, -(4-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-(C0-6 alkylene)-O-(5-6 membered heteroaryl containing 1 or 2 heteroatoms selected from nitrogen)-O-***, -(4-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-(C0-6 alkylene)-(5-6 membered heteroaryl containing 1 or 2 heteroatoms selected from nitrogen)-O-***, -(4-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-O-(C0-6 alkylene)-(5-6 membered heteroaryl containing 1 or 2 heteroatoms selected from nitrogen)-O-***, -(4-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-O-(C1-6 alkylene)-O-(5-6 membered heteroaryl containing 1 or 2 heteroatoms selected from nitrogen)- O-***, -(5-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-O-(3-7 membered monocyclic saturated carbocyclic ring)-O-***, -(5-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-(C1-6 alkylene)- N(C1-4 alkyl)-(3-7 membered monocyclic saturated carbocyclic ring)-O-***, -(5-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-(C1-6 alkylene)- N(C3-6 cycloalkyl)-(3-7 membered monocyclic saturated carbocyclic ring)-O-***, -(5-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-(C1- 6 alkylene)-O-(3-7 membered monocyclic saturated carbocyclic ring)-O-***, or -(5-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-(C1- 6 alkylene)-(7-11 membered spirocyclic heterocyclyl containing 1-2 heteroatoms selected from nitrogen)-O-***, wherein *** is the point of attachment to A2. 94. The compound of any one of claims 1-52, wherein L is -(5-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-(C1-6 alkylene substituted with 0, 1, or 2 occurrences of C1-4 alkyl)-O-***, -(5-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-(C1-6 alkylene substituted with 1 occurrence of a spirocyclic C3-6 cycloalkyl)-O-***, -(5-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-(C1-6 alkylene substituted with 1 occurrence of C3-6 cycloalkyl)-O-***, -(4-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-(C0-6 alkylene)-O-(7-11 membered spirocyclic saturated carbocyclic ring)-O-***, -(4-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-(C0-6 alkylene)-N(C3-6 cycloalkyl)-(3-7 membered monocyclic saturated carbocyclic ring)-O-***, -(4-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-(C0-6 alkylene)-(4-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-(C1-6 alkylene substituted with 1 occurrence of C3-6 cycloalkyl)-O-***, -(4-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-(C0-6 alkylene)-(3-7 membered monocyclic saturated carbocyclic ring)-(C1-6 alkylene substituted with 1 occurrence of C3-6 cycloalkyl)-O-***, -(7-11 membered spirocyclic heterocyclyl containing 1-2 heteroatoms selected from nitrogen)-(C0-6 alkylene)-(3-7 membered monocyclic saturated carbocyclic ring)-O-***, -(4-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-(4-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-(C1-6 alkylene substituted with 1 occurrence of a spirocyclic C3-6 cycloalkyl)-O-***, -(4-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-(C0-6 alkylene)-(4-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-(C1-6 alkylene)-***, -(4-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-O-(3-7 membered monocyclic saturated carbocyclic ring)-(C1-6 alkylene substituted with 1 occurrence of C3-6 cycloalkyl)-O-***, or -(4-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-(C0-6 alkylene)-(4-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-(C1-6 haloalkyl)-***, wherein *** is the point of attachment to A2. 95. The compound of any one of claims 1-52, wherein L is -(4-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-(C0-6 alkylene)-(5-6 membered heteroaryl containing 1 or 2 heteroatoms selected from nitrogen)-(C1-6 haloalkyl)- ***, -(4-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-(C0-6 alkylene)-(5-6 membered heteroaryl containing 1 or 2 heteroatoms selected from nitrogen)-O-***, -(4-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-O-(C1-6 alkylene)-C(O)-NH-***, -(4-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-(C0-6 alkylene)-C(O)-NH- ***, -(4-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-(C0-6 alkylene)-(5-6 membered aryl substituted with 0 or 1 occurrence of fluoro)-O- ***, -(4-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-(C0-6 alkylene)-O-(5-6 membered aryl)-O-***, -(7-11 membered spirocyclic heterocyclyl containing 1-2 heteroatoms selected from nitrogen)-O-(3-7 membered monocyclic saturated carbocyclic ring)-O-***, -(7-11 membered spirocyclic heterocyclyl containing 1-2 heteroatoms selected from nitrogen)-(C0-6 alkylene)-(3-7 membered monocyclic saturated carbocyclic ring)-O-***, or -(4-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-(C0-6 alkylene)-(pyrazinonyl)-***, wherein *** is the point of attachment to A2. 96. The compound of any one of claims 1-52, wherein L is one of the following: wherein *** is the point of attachment to A2. 97. The compound of any one of claims 1-52, wherein L is one of the following: wherein *** is the point of attachment to A2. 98. The compound of any one of claims 1-52, wherein L is one of the following: wherein *** is the point of attachment to A2.

99. A compound represented by Formula II: or a pharmaceutically acceptable salt thereof; wherein: TPL is a group defined by Formula II-1 that is substituted by one occurrence of RII-1A, wherein Formula II-1 is represented by: (II-1) or a pharmaceutically acceptable salt thereof; wherein: RII-1A is a bond to L; R1 is phenyl substituted by cyano, halogen, and m occurrences of R4; R2 and R3 are independently hydrogen or C1-4 alkyl; R4 is C1-4 alkyl; R5 represents independently for each occurrence C1-4 alkyl or halogen; A1 is a pyridazinyl, pyrimidinyl, pyrazinyl, pyridinyl, or phenyl, each of which is substituted with n occurrences of R5; L is a linker; EPL is a moiety that binds to BRD4; and m and n are independently 0, 1, or 2. 100. The compound of claim 99, wherein the that is substituted by one occurrence of RII-1A.

101. The compound of claim 99, wherein the 102. The compound of any one of claims 99-101, wherein the EPL is defined by Formula II-2 that is substituted by one occurrence of RII-2A, wherein Formula II-2 is represented by: wherein RII-2A is a bond to L; A3 is phenylene, a 3-10 membered saturated monocyclic, bicyclic or spirocyclic carbocycylene, or C1-6 alkylene; R1B represents independently for each occurrence halo, C1-4 alkyl, or C1-4 haloalkyl; R2B is hydrogen, halo, C1-4 alkyl, or C1-4 haloalkyl; R3B is -N(R8B)SO2R9B, -SO2N(R8B)2, -SO2R9B, -(C1-6 alkylene)-SO2R9B, C1-6 hydroxyalkyl, -O-(phenyl), or one of the following ring groups: (a) a 4-7 membered saturated carbocyclic ring in which one CH2 is replaced with SO2, (b) a 4-7 membered saturated heterocyclic ring containing 1 or 2 heteroatoms independently selected from nitrogen, in which one CH2 is replaced with SO2, or (c) a 3-5 membered heterocyclic ring containing 1 or 2 heteroatoms independently selected from nitrogen and oxygen; wherein each phenyl and heterocyclic ring is substituted with q occurrences of R10B;R4B is hydrogen, halo, or C1-4 alkyl; R5B is C1-4 alkyl or C3-4 cycloalkyl; R6B is hydrogen, C1-4 alkyl, or C3-4 cycloalkyl; R7B represents independently for each occurrence C1-4 alkyl or C3-4 cycloalkyl; R8B represents independently for each occurrence hydrogen or C1-4 alkyl; or two occurrences of R8B are taken together with the nitrogen atom to which they are attached to form a 3-7 membered heterocyclic ring containing 1 or 2 heteroatoms independently selected from nitrogen and oxygen; or R8B and R9B are taken together with their intervening atoms to a form a 5-7 membered ring containing 1, 2, or 3 heteroatoms independently selected from nitrogen, oxygen, and sulfur; R9B is C1-6 alkyl, C1-6 haloalkyl, -(C1-6 alkylene)-(C3-6 cycloalkyl), or C3-6 cycloalkyl; R10B represents independently for each occurrence hydroxyl, halo, or C1-4 alkyl; and p, q, and t are independently 0, 1, or 2. , , ,

each of which is substituted by one occurrence of RII-2A, wherein RII-2A is a bond to L. 104. The compound of any one of claims 99-101, wherein the EPL is , , ,

. 105. The compound of any one of claims 99-104, wherein L is a bivalent, saturated or unsaturated, straight or branched C1-60 hydrocarbon chain, wherein 0-20 methylene units of the hydrocarbon are independently replaced with -O-, -S-, -N(H)-, -N(C1-6 alkyl)-, -OC(O)-, - C(O)O-, -S(O)-, -S(O)2-, -N(H)S(O)2-, -N(C1-6 alkyl)S(O)2-, -S(O)2N(H)-, -S(O)2N(C1-6 alkyl)-, -N(H)C(O)-, -N(C1-6 alkyl)C(O)-, -C(O)N(H)-, -C(O)N(C1-6 alkyl)-, -OC(O)N(H)-, - OC(O)N(C1-6 alkyl)-, -N(H)C(O)O-, -N(C1-6 alkyl)C(O)O-, optionally substituted 3-10 membered carbocyclyl, or optionally substituted 3-10 membered heterocyclyl containing 1, 2, 3, or 4 heteroatoms independently selected from nitrogen, oxygen, and sulfur. 106. The compound of any one of claims 99-104, wherein L is a bivalent, saturated, straight or branched C3-30 hydrocarbon chain, wherein 0-15 methylene units of the hydrocarbon are independently replaced with -O-, -N(H)-, -N(C1-6 alkyl)-, -OC(O)-, -C(O)O-, -N(H)C(O)-, - N(C1-6 alkyl)C(O)-, -C(O)N(H)-, -C(O)N(C1-6 alkyl)-, 3-10 membered carbocyclyl, or 3-10 membered heterocyclyl containing 1, 2, or 3 heteroatoms independently selected from nitrogen, oxygen, and sulfur. 107. The compound of any one of claims 99-104, wherein L is a bivalent, saturated, straight or branched C3-30 hydrocarbon chain, wherein 0-15 methylene units of the hydrocarbon are independently replaced with -O-, -N(H)-, -N(C1-6 alkyl)-, -OC(O)-, -C(O)O-, -N(H)C(O)-, - N(C1-6 alkyl)C(O)-, -C(O)N(H)-, or -C(O)N(C1-6 alkyl)-.

108. A compound represented by Formula If: L is one of the following: ^ -(3-7 membered monocyclic saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen)-X1-***, wherein *** is the point of attachment to A2, and X1 is (i) C1-10 alkylene where 1 or 2 methylene groups are optionally replaced by -O-, -N(H)-, or -N(C1-4 alkyl)-, (ii) a 3-7 membered monocyclic saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen, or (iii) -(3-7 membered monocyclic saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen)-(C1-10 alkylene)-***; or ^ -(4-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-(C0-6 alkylene)-(4-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-*** or -(4-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-O-(4-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-***, wherein *** is the point of attachment to A2. 109. The compound of claim 108, wherein . 110. The compound of claim 108 or 109, wherein L is -(3-7 membered monocyclic saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen)-X1-***, wherein *** is the point of attachment to A2, and X1 is (i) C1-10 alkylene where 1 or 2 methylene groups are optionally replaced by -O-, -N(H)-, or -N(C1-4 alkyl)-, (ii) a 3-7 membered monocyclic saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen, or (iii) -(3-7 membered monocyclic saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen)-(C1-10 alkylene)-***. 111. The compound of claim 108 or 109, wherein L is -(4-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-(C0-6 alkylene)-(4-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-***, wherein *** is the point of attachment to A2.

112. The compound of claim 108 or 109, wherein L is -(4-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-O-(4-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-***, wherein *** is the point of attachment to A2. 113. A compound in Table 1, or a pharmaceutically acceptable salt thereof. 114. The compound of claim 113, wherein the compound is any one of compound I-1 to I-253 in Table 1, or a pharmaceutically acceptable salt thereof. 115. A pharmaceutical composition comprising a compound of any one of claims 1-114 and a pharmaceutically acceptable carrier. 116. A method of treating cancer, comprising administering to a patient in need thereof a therapeutically effective amount of a compound of any one of claims 1-114 to treat the cancer. 117. The method of claim 116, wherein the cancer is ovarian cancer, uterine cancer, endometrial cancer, cervical cancer, prostate cancer, testicular cancer, breast cancer, brain cancer, lung cancer, oral cancer, esophageal cancer, head and neck cancer, stomach cancer, colon cancer, rectal cancer, skin cancer, sebaceous gland carcinoma, bile duct cancer, gallbladder cancer, liver cancer, pancreatic cancer, bladder cancer, urinary tract cancer, kidney cancer, eye cancer, thyroid cancer, lymphoma, or leukemia. 118. The method of claim 116, wherein the cancer is prostate cancer. 119. A method of causing death of a cancer cell, comprising contacting a cancer cell with an effective amount of a compound of any one of claims 1-114 to cause death of the cancer cell. 120. The method of claim 119, wherein the cancer cell is selected from an ovarian cancer, uterine cancer, endometrial cancer, cervical cancer, prostate cancer, testicular cancer, breast cancer, brain cancer, lung cancer, oral cancer, esophageal cancer, head and neck cancer, stomach cancer, colon cancer, rectal cancer, skin cancer, sebaceous gland carcinoma, bile duct cancer, gallbladder cancer, liver cancer, pancreatic cancer, bladder cancer, urinary tract cancer, kidney cancer, eye cancer, thyroid cancer, lymphoma, or leukemia cell. 121. The method of claim 119, wherein the cancer cell is a prostate cancer cell.

Description:
Attorney Docket No.401507-019WO (203479) HETEROBIFUNCTIONAL COMPOUNDS AND METHODS OF TREATING DISEASE CROSS-REFERENCE TO RELATED APPLICATIONS [0001] This application claims the benefit of and priority to United States Provisional Patent Application serial number 63/404,572, filed September 8, 2022, and United States Provisional Patent Application serial number 63/494,343, filed April 5, 2023; the contents of each of which are hereby incorporated by reference in their entirety. FIELD OF THE INVENTION [0002] The invention provides heterobifunctional compounds, pharmaceutical compositions, and their use in treating disease, such as cancer. BACKGROUND [0003] Cancer continues to be a significant health problem despite the substantial research efforts and scientific advances reported in the literature for treating this disease. Solid tumors, including prostate cancer, breast cancer, and lung cancer remain highly prevalent among the world population. The incidence of prostate cancer increases with age, and with increasing longevity of human subjects, there continues to be a corresponding rise in the number of patients suffering from prostate cancer. Breast cancer is one of the most common cancers among women and is a leading cause of death for women between ages 50-55. Lung cancer is a leading cause of death among cancer patients, where over 85% of lung cancers are non-small cell lung cancer (NSCLC). Many lung cancers are attributed to tobacco smoking. Current treatment options for these cancers are not effective for all patients and/or can have substantial adverse side effects. [0004] New therapies are needed to address this unmet need in cancer therapy. In particular, new therapies are needed that achieve an anti-cancer effect through a different mechanism than commonly available therapies. Exemplary mechanisms for common anti-cancer therapies include (a) alkylation of DNA which limits ability of the cell to reproduce, (b) topoisomerase inhibition, in which the therapeutic agent inhibits the activity of a topoisomerases thereby limiting separation of strands of DNA, and (c) mitotic inhibition, where the therapeutic agent reduces ability of the cell to divide. New therapies that achieve an anti-cancer effect through a different mechanism present an opportunity to treat cancers more effectively and/or to treat cancers that have become resistant to currently available medicines. [0005] The present invention addresses the foregoing needs and provides other related advantages. SUMMARY [0006] The invention provides heterobifunctional compounds, pharmaceutical compositions, and their use in treating disease, such as cancer. In particular, one aspect of the invention provides a collection of heterobifunctional compounds, such as a compound represented by Formula I: or a pharmaceutically acceptable salt thereof, where the variables are as defined in the detailed description. Further description of additional collections of heterobifunctional compounds are described in the detailed description. The compounds may be part of a pharmaceutical composition comprising a pharmaceutically acceptable carrier. [0007] Another aspect of the invention provides a collection of heterobifunctional compounds, such as a compound represented by Formula II: or a pharmaceutically acceptable salt thereof, where the variables are as defined in the detailed description. Further description of additional collections of heterobifunctional compounds are described in the detailed description. The compounds may be part of a pharmaceutical composition comprising a pharmaceutically acceptable carrier. [0008] Another aspect of the invention provides a method of treating cancer. The method comprises administering to a patient in need thereof a therapeutically effective amount of a compound described herein, such as a compound of Formula I or II, to treat the cancer. [0009] Another aspect of the invention provides a method of causing death of a cancer cell. The method comprises contacting a cancer cell with an effective amount of a compound described herein, such as a compound of Formula I or II, to cause death of the cancer cell. DETAILED DESCRIPTION [0010] The invention provides heterobifunctional compounds, pharmaceutical compositions, and their use in treating disease, such as cancer. The practice of the present invention employs, unless otherwise indicated, conventional techniques of organic chemistry, pharmacology, molecular biology (including recombinant techniques), cell biology, biochemistry, and immunology. Such techniques are explained in the literature, such as in “Comprehensive Organic Synthesis” (B.M. Trost & I. Fleming, eds., 1991-1992); “Handbook of experimental immunology” (D.M. Weir & C.C. Blackwell, eds.); “Current protocols in molecular biology” (F.M. Ausubel et al., eds., 1987, and periodic updates); and “Current protocols in immunology” (J.E. Coligan et al., eds., 1991), each of which is herein incorporated by reference in its entirety. [0011] Various aspects of the invention are set forth below in sections; however, aspects of the invention described in one particular section are not to be limited to any particular section. Further, when a variable is not accompanied by a definition, the previous definition of the variable controls. Definitions [0012] Compounds of the present invention include those described generally herein, and are further illustrated by the classes, subclasses, and species disclosed herein. As used herein, the following definitions shall apply unless otherwise indicated. These definitions apply regardless of whether a term is used by itself or in combination with other terms, unless otherwise indicated. Hence, the definition of “alkyl” applies to “alkyl” as well as the “alkyl” portions of “-O-alkyl” etc. For purposes of this invention, the chemical elements are identified in accordance with the Periodic Table of the Elements, CAS version, Handbook of Chemistry and Physics, 75 th Ed. Additionally, general principles of organic chemistry are described in “Organic Chemistry”, Thomas Sorrell, University Science Books, Sausalito: 1999, and “March’s Advanced Organic Chemistry”, 5 th Ed., Ed.: Smith, M.B. and March, J., John Wiley & Sons, New York: 2001, the entire contents of which are hereby incorporated by reference. [0013] The term “aliphatic” or “aliphatic group”, as used herein, means a straight-chain (i.e., unbranched) or branched, substituted or unsubstituted hydrocarbon chain that is completely saturated or that contains one or more units of unsaturation, or a monocyclic hydrocarbon or bicyclic hydrocarbon that is completely saturated or that contains one or more units of unsaturation, but which is not aromatic (also referred to herein as “cycloaliphatic”), that has a single point of attachment to the rest of the molecule. Unless otherwise specified, aliphatic groups contain 1-6 aliphatic carbon atoms. In some embodiments, aliphatic groups contain 1-5 aliphatic carbon atoms. In other embodiments, aliphatic groups contain 1-4 aliphatic carbon atoms. In still other embodiments, alipha ic groups contain 1-3 aliphatic carbon atoms, and in yet other embodiments, aliphatic groups contain 1-2 aliphatic carbon atoms. In some embodiments, “cycloaliphatic” refers to a monocyclic C3-C6 hydrocarbon that is completely saturated or that contains one or more units of unsaturation, but which is not aromatic, that has a single point of attachment to the rest of the molecule. Suitable aliphatic groups include, but are not limited to, linear or branched, substituted or unsubstituted alkyl, alkenyl, alkynyl groups and hybrids thereof such as (cycloalkyl)alkyl, (cycloalkenyl)alkyl or (cycloalkyl)alkenyl. [0014] As used herein, the term “bicyclic ring” or “bicyclic ring system” refers to any bicyclic ring system, i.e., carbocyclic or heterocyclic, saturated or having one or more units of unsaturation, having one or more atoms in common between the two rings of the ring system. Thus, the term includes any permissible ring fusion, such as ortho-fused or spirocyclic. As used herein, the term “heterobicyclic” is a subset of “bicyclic” that requires that one or more heteroatoms are present in one or both rings of the bicycle. Such heteroatoms may be present at ring junctions and are optionally substituted, and may be selected from nitrogen (including N-oxides), oxygen, sulfur (including oxidized forms such as sulfones and sulfonates), phosphorus (including oxidized forms such as phosphates), boron, etc. In some embodiments, a bicyclic group has 7-12 ring members and 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur. As used herein, the term “bridged bicyclic” refers to any bicyclic ring system, i.e., carbocyclic or heterocyclic, saturated or partially unsaturated, having at least one bridge. As defined by IUPAC, a “bridge” is an unbranched chain of atoms or an atom or a valence bond connecting two bridgeheads, where a “bridgehead” is any skeletal atom of the ring system which is bonded to three or more skeletal atoms (excluding hydrogen). In some embodiments, a bridged bicyclic group has 7-12 ring members and 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur. Such bridged bicyclic groups are well known in the art and include those groups set forth below where each group is attached to the rest of the molecule at any substitutable carbon or nitrogen atom. Unless otherwise specified, a bridged bicyclic group is optionally substituted with one or more substituents as set forth for aliphatic groups. Additionally or alternatively, any substitutable nitrogen of a bridged bicyclic group is optionally substituted. Exemplary bicyclic rings include: [0015] Exemplary bridged bicyclics include: . [0016] The term “lower alkyl” refers to a C 1-4 straight or branched alkyl group. Exemplary lower alkyl groups are methyl, ethyl, propyl, isopropyl, butyl, isobutyl, and tert-butyl. [0017] The term “lower haloalkyl” refers to a C 1-4 straight or branched alkyl group that is substituted with one or more halogen atoms. [0018] The term “heteroatom” means one or more of oxygen, sulfur, nitrogen, phosphorus, or silicon (including, any oxidized form of nitrogen, sulfur, phosphorus, or silicon; the quaternized form of any basic nitrogen or; a substitutable nitrogen of a heterocyclic ring, for example N (as in 3,4-dihydro-2H-pyrrolyl), NH (as in pyrrolidinyl) or NR + (as in N-substituted pyrrolidinyl)). [0019] The term “unsaturated,” as used herein, means that a moiety has one or more units of unsaturation. [0020] As used herein, the term “bivalent C 1-8 (or C 1-6 ) saturated or unsaturated, straight or branched, hydrocarbon chain”, refers to bivalent alkylene, alkenylene, and alkynylene chains that are straight or branched as defined herein. [0021] The term “alkylene” refers to a bivalent alkyl group. An “alkylene chain” is a polymethylene group, i.e., –(CH 2 ) n –, wherein n is a positive integer, preferably from 1 to 6, from 1 to 4, from 1 to 3, from 1 to 2, or from 2 to 3. A substituted alkylene chain is a polymethylene group in which one or more methylene hydrogen atoms are replaced with a substituent. Suitable substituents include those described below for a substituted aliphatic group. [0022] The term “-(C 0 alkylene)-“ refers to a bond. Accordingly, the term “-(C 0-3 alkylene)-” encompasses a bond (i.e., C0) and a -(C1-3 alkylene)- group. [0023] The term “alkenylene” refers to a bivalent alkenyl group. A substituted alkenylene chain is a polymethylene group containing at least one double bond in which one or more hydrogen atoms are replaced with a substituent. Suitable substituents include those described below for a substituted aliphatic group. [0024] The term “halogen” means F, Cl, Br, or I. [0025] The term “aryl” used alone or as part of a larger moiety as in “aralkyl,” “aralkoxy,” or “aryloxyalkyl,” refers to monocyclic or bicyclic ring systems having a total of five to fourteen ring members, wherein at least one ring in the system is aromatic and wherein each ring in the system contains 3 to 7 ring members. The term “aryl” may be used interchangeably with the term “aryl ring.” In certain embodiments of the present invention, “aryl” refers to an aromatic ring system which includes, but not limited to, phenyl, biphenyl, naphthyl, anthracyl and the like, which may bear one or more substituents. Also included within the scope of the term “aryl,” as it is used herein, is a group in which an aromatic ring is fused to one or more non–aromatic rings, such as indanyl, phthalimidyl, naphthimidyl, phenanthridinyl, or tetrahydronaphthyl, and the like. The term “haloaryl” refers to an aryl group that is substituted with at least one halogen. Exemplary haloaryl groups include chlorophenyl (e.g., 3-chlorophenyl, 4-chlorophenyl), fluorophenyl, and the like. The term “phenylene” refers to a bivalent phenyl group. [0026] The terms “heteroaryl” and “heteroar–,” used alone or as part of a larger moiety, e.g., “heteroaralkyl,” or “heteroaralkoxy,” refer to groups having 5 to 10 ring atoms, preferably 5, 6, or 9 ring atoms; having 6, 10, or 14 ^ electrons shared in a cyclic array; and having, in addition to carbon atoms, from one to five heteroatoms. The term “heteroatom” refers to nitrogen, oxygen, or sulfur, and includes any oxidized form of nitrogen or sulfur, and any quaternized form of a basic nitrogen. Heteroaryl groups include, without limitation, thienyl, furanyl, pyrrolyl, imidazolyl, pyrazolyl, triazolyl, tetrazolyl, oxazolyl, isoxazolyl, oxadiazolyl, thiazolyl, isothiazolyl, thiadiazolyl, pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, indolizinyl, purinyl, naphthyridinyl, and pteridinyl. The terms “heteroaryl” and “heteroar–”, as used herein, also include groups in which a heteroaromatic ring is fused to one or more aryl, cycloaliphatic, or heterocyclyl rings, where unless otherwise specified, the radical or point of attachment is on the heteroaromatic ring or on one of the rings to which the heteroaromatic ring is fused. Nonlimiting examples include indolyl, isoindolyl, benzothienyl, benzofuranyl, dibenzofuranyl, indazolyl, benzimidazolyl, benzthiazolyl, quinolyl, isoquinolyl, cinnolinyl, phthalazinyl, quinazolinyl, quinoxalinyl, 4H–quinolizinyl, carbazolyl, acridinyl, phenazinyl, phenothiazinyl, phenoxazinyl, tetrahydroquinolinyl, and tetrahydroisoquinolinyl. A heteroaryl group may be mono– or bicyclic. The term “heteroaryl” may be used interchangeably with the terms “heteroaryl ring,” “heteroaryl group,” or “heteroaromatic,” any of which terms include rings that are optionally substituted. The term “heteroaralkyl” refers to an alkyl group substituted by a heteroaryl, wherein the alkyl and heteroaryl portions independently are optionally substituted. The term “haloheteroaryl” refers to a heteroaryl group that is substituted with at least one halogen. Exemplary haloheteroaryl groups include chloropyridine, fluoropyridine, chloropyrazole, fluoropyrazole, and the like. The term “heteroarylene” refers to a bivalent heteroaryl group. Similarly, the terms “pyrazolylene”, “imidazolylene”, and “pyrrolylene”, respectively refer to bivalent pyrazolyl, imidazolyl, and pyrrolyl groups. Similarly, the terms “pyridazinylene,” “pyrimidinylene,” “pyrazinylene,” and “pyridinylene,” respectively refer to bivalent pyridazinyl, pyrimidinyl, pyrazinyl, and pyridinyl groups. [0027] As used herein, the terms “heterocycle,” “heterocyclyl,” “heterocyclic radical,” and “heterocyclic ring” are used interchangeably and refer to a stable 5– to 7–membered monocyclic or 7–10–membered bicyclic heterocyclic moiety that is either saturated or partially unsaturated, and having, in addition to carbon atoms, one or more, preferably one to four, heteroatoms, as defined above. When used in reference to a ring atom of a heterocycle, the term "nitrogen" includes a substituted nitrogen. As an example, in a saturated or partially unsaturated ring having 0–3 heteroatoms selected from oxygen, sulfur or nitrogen, the nitrogen may be N (as in 3,4–dihydro– 2H–pyrrolyl), NH (as in pyrrolidinyl), or + NR (as in N–substituted pyrrolidinyl). [0028] A heterocyclic ring can be attached to its pendant group at any heteroatom or carbon atom that results in a stable structure and any of the ring atoms can be optionally substituted. Examples of such saturated or partially unsaturated heterocyclic radicals include, without limitation, tetrahydrofuranyl, tetrahydrothiophenyl pyrrolidinyl, piperidinyl, pyrrolinyl, tetrahydroquinolinyl, tetrahydroisoquinolinyl, decahydroquinolinyl, oxazolidinyl, piperazinyl, dioxanyl, dioxolanyl, diazepinyl, oxazepinyl, thiazepinyl, morpholinyl, 2-oxa-6- azaspiro[3.3]heptane, and quinuclidinyl. The terms “heterocycle,” “heterocyclyl,” “heterocyclyl ring,” “heterocyclic group,” “heterocyclic moiety,” and “heterocyclic radical,” are used interchangeably herein, and also include groups in which a heterocyclyl ring is fused to one or more aryl, heteroaryl, or cycloaliphatic rings, such as indolinyl, 3H–indolyl, chromanyl, phenanthridinyl, or tetrahydroquinolinyl. A heterocyclyl group may be mono– or bicyclic. The term “heterocyclylalkyl” refers to an alkyl group substituted by a heterocyclyl, wherein the alkyl and heterocyclyl portions independently are optionally substituted. The term “heterocyclylene” refers to a bivalent heterocyclyl group. The terms “piperidinylene,” “piperazinylene,” and “azetidinylene”, respectively refer to bivalent piperidinyl, piperazinyl, and azetidinyl groups. [0029] As used herein, the term “heterocycloalkyl” refers to a saturated heterocyclyl. The term “heterocycloalkylene” refers to a bivalent heterocycloalkyl group. [0030] As used herein, the term “partially unsaturated” refers to a ring moiety that includes at least one double or triple bond. The term “partially unsaturated” is intended to encompass rings having multiple sites of unsaturation, but is not intended to include aryl or heteroaryl moieties, as herein defined. [0031] As described herein, compounds of the invention may contain “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. 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. Combinations of substituents envisioned by this invention 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. [0032] Each optional substituent on a substitutable carbon is a monovalent substituent independently selected from halogen; –(CH 2 ) 0–4 R ^; –(CH 2 ) 0–4 OR ^; -O(CH 2 ) 0-4 R o , –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°; –(CH2)0–4O(CH2)0–1-pyridyl which may be substituted with R°; –NO2; –CN; – N3; -(CH2)0–4N(R ^)2; –(CH2)0–4N(R ^)C(O)R ^; –N(R ^)C(S)R ^; –(CH2)0–4N(R ^)C(O)NR ^2; -N(R ^)C(S)NR ^2; –(CH2)0–4N(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 ^; –(CH2)0–4C(O)R ^; –C(S)R ^; –(CH2)0–4C(O)OR ^; –(CH2)0–4C(O)SR ^; -(CH2)0–4C(O)OSiR ^3; –(CH2)0–4OC(O)R ^; –OC(O)(CH2)0–4SR–, SC(S)SR°; –(CH2)0–4SC(O)R ^; – (CH2)0–4C(O)NR ^2; –C(S)NR ^2; –C(S)SR°; –SC(S)SR°, -(CH2)0–4OC(O)NR ^2; -C(O)N(OR ^)R ^; –C(O)C(O)R ^; –C(O)CH2C(O)R ^; –C(NOR ^)R ^; -(CH2)0–4SSR ^; –(CH2)0– 4S(O)2R ^; –(CH2)0–4S(O)2OR ^; –(CH2)0–4OS(O)2R ^; –S(O)2NR ^2; –S(O)(NR ^)R ^; – S(O)2N=C(NR ^2)2; -(CH2)0–4S(O)R ^; -N(R ^)S(O)2NR ^2; –N(R ^)S(O)2R ^; –N(OR ^)R ^; – C(NH)NR ^2; –P(O)2R ^; -P(O)R ^2; -OP(O)R ^2; –OP(O)(OR ^)2; SiR ^3; –(C1–4 straight or branched alkylene)O–N(R ^) 2 ; or –(C 1–4 straight or branched alkylene)C(O)O–N(R ^) 2 . [0033] Each R ^ 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 5–6–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 by a divalent substituent on a saturated carbon atom of R ^ selected from =O and =S; or each R ^ is optionally substituted with a monovalent substituent independently selected from halogen, – (CH2)0–2R ^ , –(haloR ^ ), –(CH2)0–2OH, –(CH2)0–2OR ^ , –(CH2)0–2CH(OR ^ )2; -O(haloR ^ ), –CN, –N3, –(CH2)0–2C(O)R ^ , –(CH2)0–2C(O)OH, –(CH2)0–2C(O)OR ^ , –(CH2)0–2SR ^ , –(CH2)0–2SH, –(CH2)0– 2 NH 2 , –(CH 2 ) 0–2 NHR ^ , –(CH 2 ) 0–2 NR ^ 2 , –NO 2 , –SiR ^ 3 , –OSiR ^ 3 , -C(O)SR ^ , –(C 1–4 straight or branched alkylene)C(O)OR ^ , or –SSR ^ . [0034] Each R ^ 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, and wherein each R ^ is unsubstituted or where preceded by halo is substituted only with one or more halogens; or wherein an optional substituent on a saturated carbon is a divalent substituent independently selected from =O, =S, =NNR * 2, =NNHC(O)R * , =NNHC(O)OR * , =NNHS(O)2R * , =NR * , =NOR * , –O(C(R * 2))2–3O–, or –S(C(R * 2))2– 3 S–, or a divalent substituent bound to vicinal substitutable carbons of an “optionally substituted” group is –O(CR * 2)2–3O–, wherein each independent occurrence of R * is selected from hydrogen, C1–6 aliphatic or an unsubstituted 5–6–membered saturated, partially unsaturated, or aryl ring having 0–4 heteroatoms independently selected from nitrogen, oxygen, or sulfur. [0035] When R * is C 1–6 aliphatic, R * is optionally substituted with halogen, – R ^ , -(haloR ^ ), -OH, –OR ^ , –O(haloR ^ ), –CN, –C(O)OH, –C(O)OR ^ , –NH2, –NHR ^ , –NR ^ 2, or – NO 2 , wherein each R ^ 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, and wherein each R ^ is unsubstituted or where preceded by halo is substituted only with one or more halogens. [0036] An optional substituent on a substitutable nitrogen is independently –R , –NR 2 , – C(O)R , –C(O)OR , –C(O)C(O)R , –C(O)CH2C(O)R , -S(O)2R , -S(O)2NR 2, –C(S)NR 2, – C(NH)NR 2, or –N(R )S(O)2R ; wherein each R is independently hydrogen, C1–6 aliphatic, unsubstituted –OPh, or an unsubstituted 5–6–membered saturated, partially unsaturated, or aryl ring having 0–4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or, 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; wherein when R is C 1–6 aliphatic, R is optionally substituted with halogen, –R ^ , -(haloR ^ ), -OH, –OR ^ , –O(haloR ^ ), –CN, –C(O)OH, –C(O)OR ^ , –NH 2 , –NHR ^ , –NR ^ 2 , or –NO 2 , wherein each R ^ 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, and wherein each R ^ is unsubstituted or where preceded by halo is substituted only with one or more halogens. [0037] As used herein, the term "pharmaceutically acceptable salt" refers to those salts which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower 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, S. M. Berge et al., describe pharmaceutically acceptable salts in detail in J. Pharmaceutical Sciences, 1977, 66, 1–19, incorporated herein by reference. Pharmaceutically acceptable salts of the compounds of this invention include those derived from suitable inorganic and organic acids and bases. Examples of pharmaceutically acceptable, nontoxic acid addition salts are salts of an amino group formed with inorganic acids such as hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid and perchloric acid or with organic acids such as acetic acid, oxalic acid, maleic acid, tartaric acid, citric acid, succinic acid or malonic acid or by using other methods used in the art such as ion exchange. Other pharmaceutically acceptable salts include adipate, alginate, ascorbate, aspartate, benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphorate, camphorsulfonate, citrate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, formate, fumarate, glucoheptonate, glycerophosphate, gluconate, hemisulfate, heptanoate, hexanoate, hydroiodide, 2–hydroxy–ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, malate, maleate, malonate, methanesulfonate, 2–naphthalenesulfonate, nicotinate, nitrate, oleate, oxalate, palmitate, pamoate, pectinate, persulfate, 3–phenylpropionate, phosphate, pivalate, propionate, stearate, succinate, sulfate, tartrate, thiocyanate, p– toluenesulfonate, undecanoate, valerate salts, and the like. [0038] Further, acids which are generally considered suitable for the formation of pharmaceutically useful salts from basic pharmaceutical compounds are discussed, for example, by P. Stahl et al., Camille G. (eds.) Handbook of Pharmaceutical Salts. Properties, Selection and Use. (2002) Zurich: Wiley-VCH; S. Berge et al., Journal of Pharmaceutical Sciences (1977) 66(1) 1- 19; P. Gould, International J. of Pharmaceutics (1986) 33201-217; Anderson et al., The Practice of Medicinal Chemistry (1996), Academic Press, New York; and in The Orange Book (Food & Drug Administration, Washington, D.C. on their website). These disclosures are incorporated herein by reference. [0039] Salts derived from appropriate bases include alkali metal, alkaline earth metal, ammonium and N + (C 1–4 alkyl) 4 salts. Representative alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, and the like. Further pharmaceutically acceptable salts include, when appropriate, nontoxic ammonium, quaternary ammonium, and amine cations formed using counterions such as halide, hydroxide, carboxylate, sulfate, phosphate, nitrate, loweralkyl sulfonate and aryl sulfonate. [0040] Unless otherwise stated, structures depicted herein are also meant to include all isomeric (e.g., enantiomeric, diastereomeric, and geometric (or conformational)) forms of the structure; for example, the R and S configurations for each asymmetric center, Z and E double bond isomers, and Z and E conformational isomers. Therefore, single stereochemical isomers as well as enantiomeric, diastereomeric, and geometric (or conformational) mixtures of the present compounds are within the scope of the invention. Unless otherwise stated, all tautomeric forms of the compounds of the invention are within the scope of the invention. The invention includes compounds that differ only in the presence of one or more isotopically enriched atoms. For example, compounds having the present structures including the replacement of hydrogen by deuterium or tritium, or the replacement of a carbon by a 13 C- or 14 C-enriched carbon are within the scope of this invention. Such compounds are useful, for example, as analytical tools, as probes in biological assays, or as therapeutic agents in accordance with the present invention. [0041] Diastereomeric mixtures can be separated into their individual diastereomers on the basis of their physical chemical differences by methods known to those skilled in the art, such as, for example, by chromatography and/or fractional crystallization. Enantiomers can be separated by converting the enantiomeric mixture into a diastereomeric mixture by reaction with an appropriate optically active compound (e.g., chiral auxiliary such as a chiral alcohol or Mosher’s acid chloride), separating the diastereomers and converting (e.g., hydrolyzing) the individual diastereomers to the corresponding pure enantiomers. Alternatively, a particular enantiomer of a compound of the present invention may be prepared by asymmetric synthesis. Still further, where the molecule contains a basic functional group (such as amino) or an acidic functional group (such as carboxylic acid) diastereomeric salts are formed with an appropriate optically-active acid or base, followed by resolution of the diastereomers thus formed by fractional crystallization or chromatographic means known in the art, and subsequent recovery of the pure enantiomers. [0042] Individual stereoisomers of the compounds of the invention may, for example, be substantially free of other isomers, or may be admixed, for example, as racemates or with all other, or other selected, stereoisomers. Chiral center(s) in a compound of the present invention can have the S or R configuration as defined by the IUPAC 1974 Recommendations. Further, to the extent a compound described herein may exist as a atropisomer (e.g., substituted biaryls), all forms of such atropisomer are considered part of this invention. [0043] Chemical names, common names, and chemical structures may be used interchangeably to describe the same structure. If a chemical compound is referred to using both a chemical structure and a chemical name, and an ambiguity exists between the structure and the name, the structure predominates. It should also be noted that any carbon as well as heteroatom with unsatisfied valences in the text, schemes, examples and tables herein is assumed to have the sufficient number of hydrogen atom(s) to satisfy the valences. [0044] The terms “a” and “an” as used herein mean “one or more” and include the plural unless the context is inappropriate. [0045] The term “alkyl” refers to a saturated straight or branched hydrocarbon, such as a straight or branched group of 1-12, 1-10, or 1-6 carbon atoms, referred to herein as C 1 -C 12 alkyl, C 1 -C 10 alkyl, and C 1 -C 6 alkyl, respectively. Exemplary alkyl groups include, but are not limited to, methyl, ethyl, propyl, isopropyl, 2-methyl-1-propyl, 2-methyl-2-propyl, 2-methyl-1-butyl, 3- methyl-1-butyl, 2-methyl-3-butyl, 2,2-dimethyl-1-propyl, 2-methyl-1-pentyl, 3-methyl-1-pentyl, 4- methyl-1-pentyl, 2-methyl-2-pentyl, 3-methyl-2-pentyl, 4-methyl-2-pentyl, 2,2-dimethyl-1-butyl, 3,3-dimethyl-1-butyl, 2-ethyl-1-butyl, butyl, isobutyl, t-butyl, pentyl, isopentyl, neopentyl, hexyl, heptyl, octyl, etc. [0046] The term “cycloalkyl” refers to a monovalent saturated cyclic, bicyclic, or bridged cyclic (e.g., adamantyl) hydrocarbon group of 3-12, 3-8, 4-8, or 4-6 carbons, referred to herein, e.g., as “C3-C6 cycloalkyl,” derived from a cycloalkane. Exemplary cycloalkyl groups include cyclohexyl, cyclopentyl, cyclobutyl, and cyclopropyl. The term “cycloalkylene” refers to a bivalent cycloalkyl group. [0047] The term “haloalkyl” refers to an alkyl group that is substituted with at least one halogen. Exemplary haloalkyl groups include -CH 2 F, -CHF 2 , -CF 3 , -CH 2 CF 3 , -CF 2 CF 3 , and the like. The term “chloroalkyl” refers to an alkyl group that is substituted with at least one chloro. The term “bromoalkyl” refers to an alkyl group that is substituted with at least one bromo. The term “haloalkylene” refers to a bivalent haloalkyl group. [0048] The term “hydroxyalkyl” refers to an alkyl group that is substituted with at least one hydroxyl. Exemplary hydroxyalkyl groups include -CH2CH2OH, -C(H)(OH)CH3, -CH2C(H)(OH)CH2CH2OH, and the like. [0049] The term “heteroalkyl” refers to an alkyl group in which one or more carbon atoms has been replaced by a heteroatom (e.g., N, O, or S). Exemplary heteroalkyl groups include -OCH 3 , -CH 2 OCH 3 , -CH 2 CH 2 N(CH 3 ) 2 , and -CH 2 CH 2 OH. The heteroalkyl group may contain, for example, from 2-4, 2-6, or 2-8 atoms selected from the group consisting of carbon and a heteroatom (e.g., N, O, or S). The phrase 3-8 membered heteroalkyl refers to a heteroalkyl group having from 3 to 8 atoms selected from the group consisting of carbon and a heteroatom. The term “heteroalkylene” refers to a bivalent heteroalkyl group. [0050] The terms “alkenyl” and “alkynyl” are art-recognized and refer to unsaturated aliphatic groups analogous in length and possible substitution to the alkyls described above, but that contain at least one double or triple bond respectively. The term “haloalkenyl” refers to an alkenyl group that is substituted with at least one halogen. The term “fluoroalkenyl” refers to an alkenyl group that is substituted with at least one fluoro. The term “nitroalkenyl” refers to an alkenyl group that is substituted with at least one nitro. [0051] The term “carbocyclylene” refers to a bivalent cycloaliphatic group. [0052] The terms “alkoxyl” or “alkoxy” are art-recognized and refer to an alkyl group, as defined above, having an oxygen radical attached thereto. Representative alkoxyl groups include methoxy, ethoxy, propyloxy, tert-butoxy and the like. The term “haloalkoxyl” refers to an alkoxyl group that is substituted with at least one halogen. Exemplary haloalkoxyl groups include -OCH2F, -OCHF2, -OCF3, -OCH2CF3, -OCF2CF3, and the like. [0053] The term “oxo” is art-recognized and refers to a “=O” substituent. For example, a cyclopentane susbsituted with an oxo group is cyclopentanone. [0054] The term “amino” is art-recognized and refers to both unsubstituted and substituted amines, e.g., a moiety that may be represented by the general formulas: wherein R 50 , R 51 , R 52 and R 53 each independently represent a hydrogen, an alkyl, an alkenyl, -(CH 2 ) m -R 61 , or R 50 and R 51 , taken together with the N atom to which they are attached complete a heterocycle having from 4 to 8 atoms in the ring structure; R 61 represents an aryl, a 3-7 membered cycloalkyl, a 4-7 membered cycloalkenyl, 5-10 membered heteroaryl, or 3-10 membered heterocyclyl; and m is zero or an integer in the range of 1 to 8. [0055] The term “amido” is art-recognized and refers to both unsubstituted and substituted amides, e.g., a moiety that may be represented by the general formulas: wherein R 50 and R 51 each independently represent a hydrogen, an alkyl, an alkenyl, -(CH2)m-R 61 , or R 50 and R 51 , taken together with the N atom to which they are attached complete a heterocycle having from 4 to 8 atoms in the ring structure; R 61 represents an aryl, a 3-7 membered cycloalkyl, a 4-7 membered cycloalkenyl, 5-10 membered heteroaryl, or 3-10 membered heterocyclyl; and m is zero or an integer in the range of 1 to 8; and R 52 is an alkyl, an alkenyl, or -(CH 2 ) m -R 61 . [0056] The symbol “ ” indicates a point of attachment. [0057] When any substituent or variable occurs more than one time in any constituent or the compound of the invention, its definition on each occurrence is independent of its definition at every other occurrence, unless otherwise indicated. [0058] One or more compounds of the invention may exist in unsolvated as well as solvated forms with pharmaceutically acceptable solvents such as water, ethanol, and the like, and it is intended that the invention embrace both solvated and unsolvated forms. “Solvate” means a physical association of a compound of this invention with one or more solvent molecules. This physical association involves varying degrees of ionic and covalent bonding, including hydrogen bonding. In certain instances the solvate will be capable of isolation, for example when one or more solvent molecules are incorporated in the crystal lattice of the crystalline solid. “Solvate” encompasses both solution-phase and isolatable solvates. Non-limiting examples of suitable solvates include ethanolates, methanolates, and the like. “Hydrate” is a solvate wherein the solvent molecule is H 2 O. [0059] As used herein, the terms “subject” and “patient” are used interchangeable and refer to organisms to be treated by the methods of the present invention. Such organisms preferably include, but are not limited to, mammals (e.g., murines, simians, equines, bovines, porcines, canines, felines, and the like), and most preferably includes humans. [0060] The term “IC 50 ” is art-recognized and refers to the concentration of a compound that is required to achieve 50% inhibition of the target. [0061] As used herein, the term “effective amount” refers to the amount of a compound sufficient to effect beneficial or desired results (e.g., a therapeutic, ameliorative, inhibitory or preventative result). An effective amount can be administered in one or more administrations, applications or dosages and is not intended to be limited to a particular formulation or administration route. As used herein, the term “treating” includes any effect, e.g., lessening, reducing, modulating, ameliorating or eliminating, that results in the improvement of the condition, disease, disorder, and the like, or ameliorating a symptom thereof. [0062] As used herein, the term “pharmaceutical composition” refers to the combination of an active agent with a carrier, inert or active, making the composition especially suitable for diagnostic or therapeutic use in vivo or ex vivo. [0063] As used herein, the term “pharmaceutically acceptable carrier” refers to any of the standard pharmaceutical carriers, such as a phosphate buffered saline solution, water, emulsions (e.g., such as an oil/water or water/oil emulsions), and various types of wetting agents. The compositions also can include stabilizers and preservatives. For examples of carriers, stabilizers and adjuvants, see e.g., Martin, Remington’s Pharmaceutical Sciences, 15th Ed., Mack Publ. Co., Easton, PA [1975]. [0064] For therapeutic use, salts of the compounds of the present invention are contemplated as being pharmaceutically acceptable. However, salts of acids and bases that are non- pharmaceutically acceptable may also find use, for example, in the preparation or purification of a pharmaceutically acceptable compound. [0065] In addition, when a compound of the invention contains both a basic moiety (such as, but not limited to, a pyridine or imidazole) and an acidic moiety (such as, but not limited to, a carboxylic acid) zwitterions (“inner salts”) may be formed. Such acidic and basic salts used within the scope of the invention are pharmaceutically acceptable (i.e., non-toxic, physiologically acceptable) salts. Such salts of the compounds of the invention may be formed, for example, by reacting a compound of the invention with an amount of acid or base, such as an equivalent amount, in a medium such as one in which the salt precipitates or in an aqueous medium followed by lyophilization. [0066] Throughout the description, where compositions are described as having, including, or comprising specific components, or where processes and methods are described as having, including, or comprising specific steps, it is contemplated that, additionally, there are compositions of the present invention that consist essentially of, or consist of, the recited components, and that there are processes and methods according to the present invention that consist essentially of, or consist of, the recited processing steps. [0067] As a general matter, compositions specifying a percentage are by weight unless otherwise specified. I. Heterobifunctional Compounds [0068] One aspect of the invention provides heterobifunctional compounds. The compounds may be used in the pharmaceutical compositions and therapeutic methods described herein. Exemplary compounds are described in the following sections, along with exemplary procedures for making the compounds. Without being bound by theory, the compounds can facilitate therapeutic effects by binding to both an androgen receptor and BRD4 (bromodomain-containing protein 4). Part A: Compound of Formula I [0069] One aspect of the invention provides a compound represented by Formula I: or a pharmaceutically acceptable salt thereof; wherein: R 1 is phenyl substituted by cyano, halogen, and m occurrences of R 4 ; R 2 and R 3 are independently hydrogen or C1-4 alkyl; R 4 is C 1-4 alkyl; R 5 represents independently for each occurrence C1-4 alkyl or halogen; A 1 is a pyridazinylene, pyrimidinylene, pyrazinylene, pyridinylene, or phenylene, each of which is substituted with n occurrences of R 5 ; L is a linker; and A 2 is one of the following: A 3 is phenylene, a 3-10 membered saturated monocyclic, bicyclic or spirocyclic carbocycylene, or C1-6 alkylene; R 1B represents independently for each occurrence halo, cyano, C 1-4 alkoxy, C 1-4 alkyl, C 1-4 haloalkyl, or cyclopropyl; R 2B is hydrogen, halo, C1-4 alkyl, or C1-4 haloalkyl; R 3B is -N(R 8B )SO 2 R 9B , -SO 2 N(R 8B ) 2 , -SO 2 R 9B , -(C 1-6 alkylene)-SO 2 R 9B , C 1-6 hydroxyalkyl, -O-(phenyl), or one of the following ring groups: (a) a 4-7 membered saturated carbocyclic ring in which one CH2 is replaced with SO2, (b) a 4-7 membered saturated heterocyclic ring containing 1 or 2 heteroatoms independently selected from nitrogen, in which one CH 2 is replaced with SO 2 , or (c) a 3-5 membered heterocyclic ring containing 1 or 2 heteroatoms independently selected from nitrogen and oxygen; wherein each phenyl and heterocyclic ring is substituted with q occurrences of R 10B ; R 4B is hydrogen, halo, or C1-4 alkyl; R 5B is C1-4 alkyl or C3-4 cycloalkyl; R 6B is hydrogen, C 1-4 alkyl, or C 3-4 cycloalkyl; R 7B represents independently for each occurrence C 1-4 alkyl or C 3-4 cycloalkyl; R 8B represents independently for each occurrence hydrogen or C1-4 alkyl; or two occurrences of R 8B are taken together with the nitrogen atom to which they are attached to form a 3-7 membered heterocyclic ring containing 1 or 2 heteroatoms independently selected from nitrogen and oxygen; or R 8B and R 9B are taken together with their intervening atoms to a form a 5-7 membered ring containing 1, 2, or 3 heteroatoms independently selected from nitrogen, oxygen, and sulfur; R 9B is C1-6 alkyl, C1-6 haloalkyl, -(C1-6 alkylene)-(C3-6 cycloalkyl), or C3-6 cycloalkyl; R 10B represents independently for each occurrence hydroxyl, halo, or C1-4 alkyl; and [0070] m, n, p, q, and t are independently 0, 1, or 2.The definitions of variables in Formula I above encompass multiple chemical groups. The application contemplates embodiments where, for example, i) the definition of a variable is a single chemical group selected from those chemical groups set forth above, ii) the definition of a variable is a collection of two or more of the chemical groups selected from those set forth above, and iii) the compound is defined by a combination of variables in which the variables are defined by (i) or (ii). [0071] In certain embodiments, the compound is a compound of Formula I. [0072] As define generally above, R 1 is phenyl substituted by cyano, halogen, and m occurrences of R 4 . In certain embodiments, R 1 is phenyl substituted by cyano, halogen, and 0 occurrences of R 4 . In certain embodiments, R 1 is phenyl substituted by cyano, halogen, and 1 occurrence of R 4 . In certain embodiments, R 1 is phenyl substituted by cyano, halogen, and 2 occurrences of R 4 . In certain embodiments, R 1 is phenyl substituted by cyano, Cl, and 0 occurrences of R 4 . In certain embodiments, R 1 is . In certain embodiments, R 1 is selected from the groups depicted in the compounds in Table 1 below. [0073] As defined generally above, R 2 is hydrogen or C 1-4 alkyl. In certain embodiments, R 2 is hydrogen. In certain embodiments, R 2 is C1-4 alkyl. In certain embodiments, R 2 is selected from the groups depicted in the compounds in Table 1 below. [0074] As defined generally above, R 3 is hydrogen or C 1-4 alkyl. In certain embodiments, R 3 is hydrogen. In certain embodiments, R 3 is C1-4 alkyl. In certain embodiments, R 3 is selected from the groups depicted in the compounds in Table 1 below. [0075] As defined generally above, A 1 is a pyridazinylene, pyrimidinylene, pyrazinylene, pyridinylene, or phenylene, each of which is substituted with n occurrences of R 5 . In certain embodiments, A 1 is pyridazinylene substituted with n occurrences of R 5 . In certain embodiments, A 1 is . In certain embodiments, A 1 is pyrimidinylene substituted with n occurrences of R 5 . In certain embodiments, A 1 is , where ** is the point of attachment to L. In certain embodiments, A 1 is , where ** is the point of attachment to L. In certain embodiments, A 1 is pyrazinylene substituted with n occurrences of R 5 . In certain embodiments, A 1 . In certain embodiments, A 1 is pyridinylene substituted with n occurrences of R 5 . In certain embodiments, A 1 is , where ** is the point of attachment to L. In certain embodiments, A 1 is phenylene substituted with n occurrences of R 5 . In certain embodiments, A 1 is . In certain embodiments, A 1 is a pyridazinylene substituted with 0 occurrences of R 5 . In certain embodiments, A 1 is a pyrimidinylene substituted with 0 occurrences of R 5 . In certain embodiments, A 1 is a pyrazinylene substituted with 0 occurrences of R 5 . In certain embodiments, A 1 is a pyridinylene, substituted with 0 occurrences of R 5 . In certain embodiments, A 1 is a phenylene substituted with 0 occurrences of R 5 . In certain embodiments, A 1 is selected from the groups depicted in the compounds in Table 1 below. [ [

i

[ . [0081] In certain embodiments, A 2 in Formula I is further selected from [0083] In certain embodiments, A 2 is selected from the groups depicted in the compounds in Table 1 below. [0084] As defined generally above, A 3 is phenylene, a 3-10 membered saturated monocyclic, bicyclic or spirocyclic carbocycylene, or C 1-6 alkylene. In certain embodiments, A 3 is phenylene. In certain embodiments, A 3 is a 3-10 membered saturated monocyclic, bicyclic or spirocyclic carbocycylene. In certain embodiments, A 3 is C1-6 alkylene. In certain embodiments, A 3 is methylene or ethylene. In certain embodiments, A 3 is a 3-6 membered saturated monocyclic carbocycylene. In certain embodiments, A 3 is a 6-10 membered saturated bicyclic or spirocyclic carbocycylene. In certain embodiments, A 3 is selected from the groups depicted in the compounds in Table 1 below. [0085] As defined generally above, R 1B represents independently for each occurrence halo, C1-4 alkyl, or C1-4 haloalkyl. In certain embodiments, R 1B is halo. In certain embodiments, R 1B is fluoro. In certain embodiments, R 1B is C 1-4 alkyl. In certain embodiments, R 1B is C 1-4 haloalkyl. In certain embodiments, R 1B is selected from the groups depicted in the compounds in Table 1 below. [0086] As defined generally above, R 2B is hydrogen, halo, C1-4 alkyl, or C1-4 haloalkyl. In certain embodiments, R 2B is hydrogen. In certain embodiments, R 2B is halo. In certain embodiments, R 2B is C 1-4 alkyl. In certain embodiments, R 2B is C 1-4 haloalkyl. In certain embodiments, R 2B is selected from the groups depicted in the compounds in Table 1 below.As defined generally above, R 3B is --N(R 8B )SO 2 R 9B , -SO 2 N(R 8B ) 2 , -SO 2 R 9B , -(C 1-6 alkylene)-SO 2 R 9B , C 1-6 hydroxyalkyl, -O-(phenyl), or one of the following ring groups: (a) a 4-7 membered saturated carbocyclic ring in which one CH2 is replaced with SO2, (b) a 4-7 membered saturated heterocyclic ring containing 1 or 2 heteroatoms independently selected from nitrogen, in which one CH 2 is replaced with SO 2 , or (c) a 3-5 membered heterocyclic ring containing 1 or 2 heteroatoms independently selected from nitrogen and oxygen; wherein each phenyl and heterocyclic ring is substituted with q occurrences of R 10B . In certain embodiments, R 3B is -N(R 8B )SO 2 R 9B , -SO 2 N(R 8B ) 2 , -SO 2 R 9B , or -(C 1-6 alkylene)-SO 2 R 9B . In certain embodiments, R 3B is -N(R 8B )SO2R 9B . In certain embodiments, R 3B is SO2R 9B . In certain embodiments, R 3B is -(C 1-6 alkylene)-SO 2 R 9B . In certain embodiments, R 3B is -SO 2 N(R 8B ) 2 . In certain embodiments, R 3B is C 1-6 hydroxyalkyl. In certain embodiments, R 3B is -C(OH)(CH 3 ) 2 . In certain embodiments, R 3B is or a 4-7 membered saturated carbocyclic ring in which one CH2 is replaced with SO 2 . In certain embodiments, R 3B is -O-(phenyl), wherein the phenyl substituted with q occurrences of R 10B . In certain embodiments, R 3B is a 4-7 membered saturated heterocyclic ring containing 1 or 2 heteroatoms independently selected from nitrogen, in which one CH2 is replaced with SO2, wherein the heterocyclic ring is substituted with q occurrences of R 10B . In certain embodiments, R 3B is a 3-5 membered heterocyclic ring containing 1 or 2 heteroatoms independently selected from nitrogen and oxygen, wherein the heterocyclic ring is substituted with q occurrences of R 10B . In certain embodiments, R 3B is selected from the groups depicted in the compounds in Table 1 below. [0087] As defined generally above, R 4B is hydrogen, halo, or C 1-4 alkyl. In certain embodiments, R 4B is hydrogen. In certain embodiments, R 4B is halo. In certain embodiments, R 4B is C1-4 alkyl. In certain embodiments, R 4B is selected from the groups depicted in the compounds in Table 1 below. [0088] As defined generally above, R 5B is C 1-4 alkyl or C 3-4 cycloalkyl. In certain embodiments, R 5B is C1-4 alkyl. In certain embodiments, R 5B is methyl. In certain embodiments, R 5B is C3-4 cycloalkyl. In certain embodiments, R 5B is selected from the groups depicted in the compounds in Table 1 below. [0089] As defined generally above, R 6B is hydrogen, C 1-4 alkyl, or C 3-4 cycloalkyl. In certain embodiments, R 6B is hydrogen. In certain embodiments, R 6B is C1-4 alkyl. In certain embodiments, R 6B is C 3-4 cycloalkyl. In certain embodiments, R 6B is selected from the groups depicted in the compounds in Table 1 below. [0090] As defined generally above, R 7B represents independently for each occurrence C1-4 alkyl or C 3-4 cycloalkyl. In certain embodiments, R 7B is C 1-4 alkyl. In certain embodiments, R 7B is methyl. In certain embodiments, R 7B is C 3-4 cycloalkyl. In certain embodiments, R 7B is selected from the groups depicted in the compounds in Table 1 below. [0091] As defined generally above, R 8B represents independently for each occurrence hydrogen or C 1-4 alkyl; or two occurrences of R 8B are taken together with the nitrogen atom to which they are attached to form a 3-7 membered heterocyclic ring containing 1 or 2 heteroatoms independently selected from nitrogen and oxygen; or R 8B and R 9B are taken together with their intervening atoms to a form a 5-7 membered ring containing 1, 2, or 3 heteroatoms independently selected from nitrogen, oxygen, and sulfur. In certain embodiments, R 8B represents independently for each occurrence hydrogen or C1-4 alkyl. In certain embodiments, R 8B is hydrogen. In certain embodiments, R 8B is C 1-4 alkyl. In certain embodiments, two occurrences of R 8B are taken together with the nitrogen atom to which they are attached to form a 3-7 membered heterocyclic ring containing 1 or 2 heteroatoms independently selected from nitrogen and oxygen. In certain embodiments, R 8B and R 9B are taken together with their intervening atoms to a form a 5-7 membered ring containing 1, 2, or 3 heteroatoms independently selected from nitrogen, oxygen, and sulfur. In certain embodiments, R 8B is selected from the groups depicted in the compounds in Table 1 below. [0092] As defined generally above, R 9B is C 1-6 alkyl, C 1-6 haloalkyl, -(C 1-6 alkylene)-(C 3-6 cycloalkyl), or C 3-6 cycloalkyl. In certain embodiments, R 9B is C 1-6 alkyl or C 1-6 haloalkyl. In certain embodiments, R 9B is C1-6 alkyl. In certain embodiments, R 9B is C1-6 haloalkyl. In certain embodiments, R 9B is -(C1-6 alkylene)-(C3-6 cycloalkyl). In certain embodiments, R 9B is C3-6 cycloalkyl. In certain embodiments, R 9B is selected from the groups depicted in the compounds in Table 1 below. [0093] As defined generally above, R 10B represents independently for each occurrence hydroxyl, halo, or C 1-4 alkyl. In certain embodiments, R 10B represents independently for each occurrence halo. In certain embodiments, R 10B represents independently for each occurrence C 1-4 alkyl. In certain embodiments, R 10B is hydrogen. In certain embodiments, R 10B is halo. In certain embodiments, R 10B is C 1-4 alkyl. In certain embodiments, R 10B is methyl. In certain embodiments, R 10B is selected from the groups depicted in the compounds in Table 1 below. [0094] As defined generally above, m, n, p and t are independently 0, 1, or 2. In certain embodiments, p is 2. In certain embodiments, p is 1. In certain embodiments, p is 0. In certain embodiments, t is 2. In certain embodiments, m is 0. In certain embodiments, m is 1. In certain embodiments, m is 2. In certain embodiments, n is 0. In certain embodiments, n is 1. In certain embodiments, q is 2. In certain embodiments, q is 1. In certain embodiments, q is 0. In certain embodiments, n is 2. In certain embodiments, t is 0. In certain embodiments, t is 1. In certain embodiments, p is selected from the corresponding value in the groups depicted in the compounds in Table 1 below. In certain embodiments, q is selected from the corresponding value in the groups depicted in the compounds in Table 1 below. In certain embodiments, t is selected from the corresponding value in the groups depicted in the compounds in Table 1 below. In certain embodiments, m is selected from the corresponding value in the groups depicted in the compounds in Table 1 below. In certain embodiments, n is selected from the corresponding value in the groups depicted in the compounds in Table 1 below. [0095] In certain embodiments, the compound of Formula I is further defined by Formula Ia or Ib, or a pharmaceutically acceptable salt thereof: In certain embodiments, the definition of variables R 1 , R 2 , R 3 , A 1 , and A 2 is one of the embodiments described above in connection with Formula I. In certain embodiments, the compound is a compound of Formula Ia or a pharmaceutically acceptable salt thereof. In certain embodiments, the compound is a compound of Formula Ib or a pharmaceutically acceptable salt thereof. [0096] In certain embodiments, the compound of Formula I is further defined by Formula 1c or a pharmaceutically acceptable salt thereof: (1c). In certain embodiments, the definition of variables A 1 and A 2 is one of the embodiments described above in connection with Formula I. [0097] In certain embodiments, the compound of Formula I is further defined by Formula 1d or a pharmaceutically acceptable salt thereof: (1d). In certain embodiments, the definition of variables A 1 and A 2 is one of the embodiments described above in connection with Formula I. [0098] The compounds may be further characterized according to, for example, the identity of L. Exemplary further embodiments for L are provided in Part C below. Part A-1: Compound of Formula I-1 [0099] One aspect of the invention provides a compound represented by Formula I-1: (I-1) or a pharmaceutically acceptable salt thereof; wherein: R 1 is phenyl substituted by cyano, halogen, and m occurrences of R 4 ; R 2 and R 3 are independently hydrogen or C 1-4 alkyl; R 4 is C1-4 alkyl; R 5 represents independently for each occurrence C1-4 alkyl or halogen; A 1 is a pyridazinylene, pyrimidinylene, pyrazinylene, pyridinylene, or phenylene, each of which is substituted with n occurrences of R 5 ; L is a linker; and A 2 is one of the following: A 3 is phenylene, a 3-10 membered saturated monocyclic, bicyclic or spirocyclic carbocycylene, or C 1-6 alkylene; R 1B represents independently for each occurrence halo, C1-4 alkyl, or C1-4 haloalkyl; R 2B is hydrogen, halo, C1-4 alkyl, or C1-4 haloalkyl; R 3B is -N(R 8B )SO 2 R 9B , -SO 2 N(R 8B ) 2 , -SO 2 R 9B , -(C 1-6 alkylene)-SO 2 R 9B , C 1-6 hydroxyalkyl, or a 4-7 membered saturated carbocyclic ring in which one CH 2 is replaced with SO 2 ; R 4B is hydrogen, halo, or C1-4 alkyl; R 5B is C 1-4 alkyl or C 3-4 cycloalkyl; R 6B is hydrogen, C 1-4 alkyl, or C 3-4 cycloalkyl; R 7B represents independently for each occurrence C1-4 alkyl or C3-4 cycloalkyl; R 8B represents independently for each occurrence hydrogen or C 1-4 alkyl; or two occurrences of R 8B are taken together with the nitrogen atom to which they are attached to form a 3-7 membered heterocyclic ring containing 1 or 2 heteroatoms independently selected from nitrogen and oxygen; or R 8B and R 9B are taken together with their intervening atoms to a form a 5-7 membered ring containing 1, 2, or 3 heteroatoms independently selected from nitrogen, oxygen, and sulfur; R 9B is C 1-6 alkyl, C 1-6 haloalkyl, -(C 1-6 alkylene)-(C 3-6 cycloalkyl), or C 3-6 cycloalkyl; and m, n, p and t are independently 0, 1, or 2. [0100] The definitions of variables in Formula I-1 above encompass multiple chemical groups. The application contemplates embodiments where, for example, i) the definition of a variable is a single chemical group selected from those chemical groups set forth above, ii) the definition of a variable is a collection of two or more of the chemical groups selected from those set forth above, and iii) the compound is defined by a combination of variables in which the variables are defined by (i) or (ii). [0101] In certain embodiments, the compound is a compound of Formula I-1. [0102] As define generally above, R 1 is phenyl substituted by cyano, halogen, and m occurrences of R 4 . In certain embodiments, R 1 is phenyl substituted by cyano, halogen, and 0 occurrences of R 4 . In certain embodiments, R 1 is phenyl substituted by cyano, halogen, and 1 occurrence of R 4 . In certain embodiments, R 1 is phenyl substituted by cyano, halogen, and 2 occurrences of R 4 . In certain embodiments, R 1 is phenyl substituted by cyano, Cl, and 0 occurrences of R 4 . In certain embodiments, R 1 is . In certain embodiments, R 1 is selected from the groups depicted in the compounds in Table 1 below. [0103] As defined generally above, R 2 is hydrogen or C 1-4 alkyl. In certain embodiments, R 2 is hydrogen. In certain embodiments, R 2 is C 1-4 alkyl. In certain embodiments, R 2 is selected from the groups depicted in the compounds in Table 1 below. [0104] As defined generally above, R 3 is hydrogen or C1-4 alkyl. In certain embodiments, R 3 is hydrogen. In certain embodiments, R 3 is C 1-4 alkyl. In certain embodiments, R 3 is selected from the groups depicted in the compounds in Table 1 below. [0105] As defined generally above, A 1 is a pyridazinylene, pyrimidinylene, pyrazinylene, pyridinylene, or phenylene, each of which is substituted with n occurrences of R 5 . In certain embodiments, A 1 is pyridazinylene substituted with n occurrences of R 5 . In certain embodiments, A 1 is . In certain embodiments, A 1 is pyrimidinylene substituted with n occurrences of R 5 . In certain embodiments, A 1 is , where ** is the point of attachment to L. In certain embodiments, A 1 is , where ** is the point of attachment to L. In certain embodiments, A 1 is pyrazinylene substituted with n occurrences of R 5 . In certain embodiments, A 1 . In certain embodiments, A 1 is pyridinylene substituted with n occurrences of R 5 . In certain embodiments, , where ** is the point of attachment to L. In certain embodiments, A 1 is phenylene substituted with n occurrences of R 5 . In certain embodiments, A 1 is . In certain embodiments, A 1 is a pyridazinylene substituted with 0 occurrences of R 5 . In certain embodiments, A 1 is a pyrimidinylene substituted with 0 occurrences of R 5 . In certain embodiments, A 1 is a pyrazinylene substituted with 0 occurrences of R 5 . In certain embodiments, A 1 is a pyridinylene, substituted with 0 occurrences of R 5 . In certain embodiments, A 1 is a phenylene substituted with 0 occurrences of R 5 . In certain embodiments, A 1 is selected from the groups depicted in the compounds in Table 1 below. [0106] As defined generally above,

[0110] In certain embodiments, A 2 is selected from the groups depicted in the compounds in Table 1 below. [0111] As defined generally above, A 3 is phenylene, a 3-10 membered saturated monocyclic, bicyclic or spirocyclic carbocycylene, or C 1-6 alkylene. In certain embodiments, A 3 is phenylene. In certain embodiments, A 3 is a 3-10 membered saturated monocyclic, bicyclic or spirocyclic carbocycylene. In certain embodiments, A 3 is C1-6 alkylene. In certain embodiments, A 3 is methylene or ethylene. In certain embodiments, A 3 is a 3-6 membered saturated monocyclic carbocycylene. In certain embodiments, A 3 is a 6-10 membered saturated bicyclic or spirocyclic carbocycylene. In certain embodiments, A 3 is selected from the groups depicted in the compounds in Table 1 below. [0112] As defined generally above, R 1B represents independently for each occurrence halo, C1-4 alkyl, or C1-4 haloalkyl. In certain embodiments, R 1B is halo. In certain embodiments, R 1B is fluoro. In certain embodiments, R 1B is C 1-4 alkyl. In certain embodiments, R 1B is C 1-4 haloalkyl. In certain embodiments, R 1B is selected from the groups depicted in the compounds in Table 1 below. [0113] As defined generally above, R 2B is hydrogen, halo, C1-4 alkyl, or C1-4 haloalkyl. In certain embodiments, R 2B is hydrogen. In certain embodiments, R 2B is halo. In certain embodiments, R 2B is C 1-4 alkyl. In certain embodiments, R 2B is C 1-4 haloalkyl. In certain embodiments, R 2B is selected from the groups depicted in the compounds in Table 1 below. [0114] As defined generally above, R 3B is -N(R 8B )SO 2 R 9B , -SO 2 N(R 8B ) 2 , -SO 2 R 9B , -(C 1-6 alkylene)-SO2R 9B , C1-6 hydroxyalkyl, or a 4-7 membered saturated carbocyclic ring in which one CH 2 is replaced with SO 2 . In certain embodiments, R 3B is -N(R 8B )SO 2 R 9B , -SO 2 N(R 8B ) 2 , -SO 2 R 9B , or -(C 1-6 alkylene)-SO 2 R 9B . In certain embodiments, R 3B is -N(R 8B )SO 2 R 9B . In certain embodiments, R 3B is SO2R 9B . In certain embodiments, R 3B is -(C1-6 alkylene)-SO2R 9B . In certain embodiments, R 3B is -SO2N(R 8B )2. In certain embodiments, R 3B is C1-6 hydroxyalkyl. In certain embodiments, R 3B is -C(OH)(CH 3 ) 2 . In certain embodiments, R 3B is or a 4-7 membered saturated carbocyclic ring in which one CH2 is replaced with SO2. In certain embodiments, R 3B is selected from the groups depicted in the compounds in Table 1 below. [0115] As defined generally above, R 4B is hydrogen, halo, or C 1-4 alkyl. In certain embodiments, R 4B is hydrogen. In certain embodiments, R 4B is halo. In certain embodiments, R 4B is C1-4 alkyl. In certain embodiments, R 4B is selected from the groups depicted in the compounds in Table 1 below. [0116] As defined generally above, R 5B is C 1-4 alkyl or C 3-4 cycloalkyl. In certain embodiments, R 5B is C1-4 alkyl. In certain embodiments, R 5B is methyl. In certain embodiments, R 5B is C3-4 cycloalkyl. In certain embodiments, R 5B is selected from the groups depicted in the compounds in Table 1 below. [0117] As defined generally above, R 6B is hydrogen, C1-4 alkyl, or C3-4 cycloalkyl. In certain embodiments, R 6B is hydrogen. In certain embodiments, R 6B is C1-4 alkyl. In certain embodiments, R 6B is C 3-4 cycloalkyl. In certain embodiments, R 6B is selected from the groups depicted in the compounds in Table 1 below. [0118] As defined generally above, R 7B represents independently for each occurrence C1-4 alkyl or C 3-4 cycloalkyl. In certain embodiments, R 7B is C 1-4 alkyl. In certain embodiments, R 7B is methyl. In certain embodiments, R 7B is C 3-4 cycloalkyl. In certain embodiments, R 7B is selected from the groups depicted in the compounds in Table 1 below. [0119] As defined generally above, R 8B represents independently for each occurrence hydrogen or C 1-4 alkyl; or two occurrences of R 8B are taken together with the nitrogen atom to which they are attached to form a 3-7 membered heterocyclic ring containing 1 or 2 heteroatoms independently selected from nitrogen and oxygen; or R 8B and R 9B are taken together with their intervening atoms to a form a 5-7 membered ring containing 1, 2, or 3 heteroatoms independently selected from nitrogen, oxygen, and sulfur. In certain embodiments, R 8B represents independently for each occurrence hydrogen or C1-4 alkyl. In certain embodiments, R 8B is hydrogen. In certain embodiments, R 8B is C 1-4 alkyl. In certain embodiments, two occurrences of R 8B are taken together with the nitrogen atom to which they are attached to form a 3-7 membered heterocyclic ring containing 1 or 2 heteroatoms independently selected from nitrogen and oxygen. In certain embodiments, R 8B and R 9B are taken together with their intervening atoms to a form a 5-7 membered ring containing 1, 2, or 3 heteroatoms independently selected from nitrogen, oxygen, and sulfur. In certain embodiments, R 8B is selected from the groups depicted in the compounds in Table 1 below. [0120] As defined generally above, R 9B is C 1-6 alkyl, C 1-6 haloalkyl, -(C 1-6 alkylene)-(C 3-6 cycloalkyl), or C 3-6 cycloalkyl. In certain embodiments, R 9B is C 1-6 alkyl or C 1-6 haloalkyl. In certain embodiments, R 9B is C1-6 alkyl. In certain embodiments, R 9B is C1-6 haloalkyl. In certain embodiments, R 9B is -(C 1-6 alkylene)-(C 3-6 cycloalkyl). In certain embodiments, R 9B is C 3-6 cycloalkyl. In certain embodiments, R 9B is selected from the groups depicted in the compounds in Table 1 below. [0121] As defined generally above, m, n, p and t are independently 0, 1, or 2. In certain embodiments, p is 2. In certain embodiments, p is 1. In certain embodiments, p is 0. In certain embodiments, t is 2. In certain embodiments, m is 0. In certain embodiments, m is 1. In certain embodiments, m is 2. In certain embodiments, n is 0. In certain embodiments, n is 1. In certain embodiments, n is 2. In certain embodiments, t is 0. In certain embodiments, t is 1. In certain embodiments, p is selected from the corresponding value in the groups depicted in the compounds in Table 1 below. In certain embodiments, t is selected from the corresponding value in the groups depicted in the compounds in Table 1 below. In certain embodiments, m is selected from the corresponding value in the groups depicted in the compounds in Table 1 below. In certain embodiments, n is selected from the corresponding value in the groups depicted in the compounds in Table 1 below. [0122] In certain embodiments, the compound of Formula I-1 is further defined by Formula Ia- 1 or Ib-1, or a pharmaceutically acceptable salt thereof: [0123] In certain embodiments, the definition of variables R 1 , R 2 , R 3 , A 1 , and A 2 is one of the embodiments described above in connection with Formula I-1. In certain embodiments, the compound is a compound of Formula Ia-1 or a pharmaceutically acceptable salt thereof. In certain embodiments, the compound is a compound of Formula Ib-1 or a pharmaceutically acceptable salt thereof. [0124] In certain embodiments, the compound of Formula I-1 is further defined by Formula 1c or a pharmaceutically acceptable salt thereof: (1c-1). [0125] In certain embodiments, the definition of variables A 1 and A 2 is one of the embodiments described above in connection with Formula I-1. [0126] In certain embodiments, the compound of Formula I-1 is further defined by Formula 1d- 1 or a pharmaceutically acceptable salt thereof: (1d-1). [0127] In certain embodiments, the definition of variables A 1 and A 2 is one of the embodiments described above in connection with Formula I-1. [0128] The compounds may be further characterized according to, for example, the identity of L. Exemplary further embodiments for L are provided in Part C below. Part A-2: Compounds of Formula I* [0129] Another aspect of the invention provides a compound represented by Formula I*: or a pharmaceutically acceptable salt thereof; wherein: R 1 is phenyl substituted by cyano, halogen, and m occurrences of R 4 ; R 2 and R 3 are independently hydrogen or C1-4 alkyl; R 4 is C 1-4 alkyl; R 5 represents independently for each occurrence C 1-4 alkyl or halogen; A 1 is a pyridazinylene, pyrimidinylene, pyrazinylene, pyridinylene, or phenylene, each of which is substituted with n occurrences of R 5 ; L is a linker; A 3 is phenylene, a 3-10 membered saturated monocyclic, bicyclic or spirocyclic carbocycylene, or C 1-6 alkylene; R 1B represents independently for each occurrence halo, cyano, C1-4 alkoxy, C1-4 alkyl, C1-4 haloalkyl, or cyclopropyl; R 2B is hydrogen, halo, C 1-4 alkyl, or C 1-4 haloalkyl; R 4B is hydrogen, halo, or C1-4 alkyl; R 5B is C 1-4 alkyl or C 3-4 cycloalkyl; R 6B is hydrogen, C 1-4 alkyl, or C 3-4 cycloalkyl; R 7B represents independently for each occurrence C1-4 alkyl or C3-4 cycloalkyl; and m, n, p, and t are independently 0, 1, or 2. [0130] In certain embodiments, the compound is a compound of Formula Ie or a pharmaceutically acceptable salt thereof: [0131] In certain embodiments, A 1 is pyridazinylene substituted with n occurrences of R 5 . In certain embodiments, [0132] In certain embodiments, [0133] The compounds may be further characterized according to, for example, the identity of L. Exemplary further embodiments for L are provided in Part C below. Part A-3: Compounds of Formula If [0134] Another aspect of the invention provides a compound represented by Formula If: or a pharmaceutically acceptable salt thereof, wherein:

L is one of the following: ^ -(3-7 membered monocyclic saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen)-X 1 -***, wherein *** is the point of attachment to A 2 , and X 1 is (i) C 1-10 alkylene where 1 or 2 methylene groups are optionally replaced by -O-, -N(H)-, or -N(C1-4 alkyl)-, (ii) a 3-7 membered monocyclic saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen, or (iii) -(3-7 membered monocyclic saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen)-(C1-10 alkylene)-***; or ^ -(4-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-(C 0-6 alkylene)-(4-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-*** or -(4-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-O-(4-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-***, wherein *** is the point of attachment to A 2 . . [0136] In certain embodiments, L is -(3-7 membered monocyclic saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen)-X 1 -***, wherein *** is the point of attachment to A 2 , and X 1 is (i) C 1-10 alkylene where 1 or 2 methylene groups are optionally replaced by -O-, -N(H)-, or -N(C 1-4 alkyl)-, (ii) a 3-7 membered monocyclic saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen, or (iii) -(3-7 membered monocyclic saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen)-(C1-10 alkylene)- ***. In certain embodiments, L is -(4-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-(C0-6 alkylene)-(4-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-***, wherein *** is the point of attachment to A 2 . In certain embodiments, L is -(4-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-O-(4-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-***, wherein *** is the point of attachment to A 2 . Part B: Compound of Formula II [0137] Another aspect of the invention provides a compound represented by Formula II: or a pharmaceutically acceptable salt thereof; wherein: TPL is a group defined by Formula II-1 that is substituted by one occurrence of R II-1A , wherein Formula II-1 is represented by: or a pharmaceutically acceptable salt thereof; wherein: R II-1A is a bond to L; R 1 is phenyl substituted by cyano, halogen, and m occurrences of R 4 ; R 2 and R 3 are independently hydrogen or C1-4 alkyl; R 4 is C 1-4 alkyl; R 5 represents independently for each occurrence C 1-4 alkyl or halogen; A 1 is a pyridazinyl, pyrimidinyl, pyrazinyl, pyridinyl, or phenyl, each of which is substituted with n occurrences of R 5 ; L is a linker; EPL is a moiety that binds to BRD4; and m and n are independently 0, 1, or 2. [0138] The definitions of variables in Formula II above encompass multiple chemical groups. The application contemplates embodiments where, for example, i) the definition of a variable is a single chemical group selected from those chemical groups set forth above, ii) the definition of a variable is a collection of two or more of the chemical groups selected from those set forth above, and iii) the compound is defined by a combination of variables in which the variables are defined by (i) or (ii). [0139] In certain embodiments, the compound is a compound of Formula II. [0140] As defined generally above, R 1 is phenyl substituted by cyano, halogen, and m occurrences of R 4 . In certain embodiments, R 1 is phenyl substituted by cyano, halogen, and 0 occurrences of R 4 . In certain embodiments, R 1 is . In certain embodiments, R 1 is phenyl substituted by cyano, halogen, and 1 occurrences of R 4 . In certain embodiments, R 1 is phenyl substituted by cyano, halogen, and 2 occurrences of R 4 . In certain embodiments, R 1 is phenyl substituted by cyano, Cl, and 0 occurrences of R 4 . In certain embodiments, R 1 is selected from the groups depicted in the compounds in Table 1 below. [0141] As defined generally above, R 2 and R 3 are independently hydrogen or C 1-4 alkyl. In certain embodiments, R 2 is hydrogen. In certain embodiments, R 3 is hydrogen. In certain embodiments, R 2 is C1-4 alkyl. In certain embodiments, R 3 is C1-4 alkyl. In certain embodiments, R 2 and R 3 are each independently selected from the groups depicted in the compounds in Table 1 below. In certain embodiments, R 2 and R 3 are each independently selected from the groups depicted in the compounds in Table 1 below. [0142] As defined generally above, A 1 is a pyridazinyl, pyrimidinyl, pyrazinyl, pyridinyl, or phenyl, each of which is substituted with n occurrences of R 5 . In certain embodiments, A 1 is pyridazinyl substituted with n occurrences of R 5 . In certain embodiments, A 1 is pyrimidinyl substituted with n occurrences of R 5 . In certain embodiments, A 1 is pyrazinyl substituted with n occurrences of R 5 . In certain embodiments, A 1 is pyridinyl substituted with n occurrences of R 5 . In In certain embodiments, A 1 is phenyl substituted with n occurrences of R 5 . In certain embodiments, A 1 is selected from the groups depicted in the compounds in Table 1 below. [0143] In certain embodiments, m is 0. In certain embodiments, m is 1. In certain embodiments, m is 2. In certain embodiments, n is 0. In certain embodiments, n is 1. In certain embodiments, n is 2. In certain embodiments, m is selected from the corresponding value in the groups depicted in the compounds in Table 1 below. In certain embodiments, n is selected from the corresponding value in the groups depicted in the compounds in Table 1 below. [0144] In certain embodiments, the that is substituted by one occurrence of R II-1A . [0145] In certain embodiments, the one occurrence certain embodiments, the [0146] In certain embodiments, the EPL is defined by Formula II-2 that is substituted by one occurrence of R II-2A , wherein Formula II-2 is represented by: R II-2A is a bond to L; A 3 is phenylene, a 3-10 membered saturated monocyclic, bicyclic or spirocyclic carbocycylene, or C 1-6 alkylene; R 1B represents independently for each occurrence halo, C1-4 alkyl, or C1-4 haloalkyl; R 2B is hydrogen, halo, C1-4 alkyl, or C1-4 haloalkyl; R 3B is -N(R 8B )SO 2 R 9B , -SO 2 N(R 8B ) 2 , -SO 2 R 9B , -(C 1-6 alkylene)-SO 2 R 9B , C 1-6 hydroxyalkyl, or a 4-7 membered saturated carbocyclic ring in which one CH 2 is replaced with SO 2 ; R 4B is hydrogen, halo, or C1-4 alkyl; R 5B is C1-4 alkyl or C3-4 cycloalkyl; R 6B is hydrogen, C 1-4 alkyl, or C 3-4 cycloalkyl; R 7B represents independently for each occurrence C1-4 alkyl or C3-4 cycloalkyl; R 8B represents independently for each occurrence hydrogen or C1-4 alkyl; or two occurrences of R 8B are taken together with the nitrogen atom to which they are attached to form a 3-7 membered heterocyclic ring containing 1 or 2 heteroatoms independently selected from nitrogen and oxygen; or R 8B and R 9B are taken together with their intervening atoms to a form a 5-7 membered ring containing 1, 2, or 3 heteroatoms independently selected from nitrogen, oxygen, and sulfur; R 9B is C1-6 alkyl, C1-6 haloalkyl, -(C1-6 alkylene)-(C3-6 cycloalkyl), or C3-6 cycloalkyl; and p and t are independently 0, 1, or 2. [0147] In certain embodiments, the EPL is defined by Formula II-2 that is substituted by one occurrence of R II-2A , wherein Formula II-2 is represented by: wherein R II-2A is a bond to L; A 3 is phenylene, a 3-10 membered saturated monocyclic, bicyclic or spirocyclic carbocycylene, or C1-6 alkylene; R 1B represents independently for each occurrence halo, C1-4 alkyl, or C1-4 haloalkyl; R 2B is hydrogen, halo, C 1-4 alkyl, or C 1-4 haloalkyl; R 3B is -N(R 8B )SO 2 R 9B , -SO 2 N(R 8B ) 2 , -SO 2 R 9B , -(C 1-6 alkylene)-SO 2 R 9B , C 1-6 hydroxyalkyl, -O-(phenyl), or one of the following ring groups: (a) a 4-7 membered saturated carbocyclic ring in which one CH2 is replaced with SO2, (b) a 4-7 membered saturated heterocyclic ring containing 1 or 2 heteroatoms independently selected from nitrogen, in which one CH2 is replaced with SO2, or (c) a 3-5 membered heterocyclic ring containing 1 or 2 heteroatoms independently selected from nitrogen and oxygen; wherein each phenyl and heterocyclic ring is substituted with q occurrences of R 10B ; R 4B is hydrogen, halo, or C1-4 alkyl; R 5B is C 1-4 alkyl or C 3-4 cycloalkyl; R 6B is hydrogen, C 1-4 alkyl, or C 3-4 cycloalkyl; R 7B represents independently for each occurrence C1-4 alkyl or C3-4 cycloalkyl; R 8B represents independently for each occurrence hydrogen or C1-4 alkyl; or two occurrences of R 8B are taken together with the nitrogen atom to which they are attached to form a 3-7 membered heterocyclic ring containing 1 or 2 heteroatoms independently selected from nitrogen and oxygen; or R 8B and R 9B are taken together with their intervening atoms to a form a 5-7 membered ring containing 1, 2, or 3 heteroatoms independently selected from nitrogen, oxygen, and sulfur; R 9B is C 1-6 alkyl, C 1-6 haloalkyl, -(C 1-6 alkylene)-(C 3-6 cycloalkyl), or C 3-6 cycloalkyl; R 10B represents independently for each occurrence hydroxyl, halo, or C1-4 alkyl; and p, q, and t are independently 0, 1, or 2. [

of which is substituted by one occurrence of R II-2A , wherein R II-2A is a bond to L.

[ R II-2A , wherein R II-2A is a bond to L. [0150] In certain embodiments, the EPL is one of the following: A 3 is phenylene, a 3-10 membered saturated monocyclic, bicyclic or spirocyclic carbocycylene, or C 1-6 alkylene; R 1B represents independently for each occurrence halo, C1-4 alkyl, or C1-4 haloalkyl; R 2B is hydrogen, halo, C1-4 alkyl, or C1-4 haloalkyl; R 3B is -N(R 8B )SO 2 R 9B , -SO 2 N(R 8B ) 2 , -SO 2 R 9B , -(C 1-6 alkylene)-SO 2 R 9B , C 1-6 hydroxyalkyl, or a 4-7 membered saturated carbocyclic ring in which one CH2 is replaced with SO2; R 4B is hydrogen, halo, or C 1-4 alkyl; R 5B is C 1-4 alkyl or C 3-4 cycloalkyl; R 6B is hydrogen, C1-4 alkyl, or C3-4 cycloalkyl; R 7B represents independently for each occurrence C1-4 alkyl or C3-4 cycloalkyl; R 8B represents independently for each occurrence hydrogen or C 1-4 alkyl; or two occurrences of R 8B are taken together with the nitrogen atom to which they are attached to form a 3-7 membered heterocyclic ring containing 1 or 2 heteroatoms independently selected from nitrogen and oxygen; or R 8B and R 9B are taken together with their intervening atoms to a form a 5-7 membered ring containing 1, 2, or 3 heteroatoms independently selected from nitrogen, oxygen, and sulfur; R 9B is C 1-6 alkyl, C 1-6 haloalkyl, -(C 1-6 alkylene)-(C 3-6 cycloalkyl), or C 3-6 cycloalkyl; and p and t are independently 0, 1, or 2. [0151] embodiments, the EPL is one of the following: In certain embodiments, the [0152] [0153] In certain embodiments, the certain embodiments, the [0154] In certain embodiments, the certain embodiments, the [0157] As defined generally above, A 3 is phenylene, a 3-10 membered saturated monocyclic, bicyclic or spirocyclic carbocycylene, or C1-6 alkylene. In certain embodiments, A 3 is phenylene. In certain embodiments, A 3 is a 3-10 membered saturated monocyclic, bicyclic or spirocyclic carbocycylene. In certain embodiments, A 3 is C 1-6 alkylene. In certain embodiments, A 3 is methylene or ethylene. In certain embodiments, A 3 is a 3-6 membered saturated monocyclic carbocycylene. In certain embodiments, A 3 is a 6-10 membered saturated bicyclic or spirocyclic carbocycylene. In certain embodiments, A 3 is selected from the groups depicted in the compounds in Table 1 below. [0158] As defined generally above, R 1B represents independently for each occurrence halo, C1-4 alkyl, or C 1-4 haloalkyl. In certain embodiments, R 1B is halo. In certain embodiments, R 1B is fluoro. In certain embodiments, R 1B is C 1-4 alkyl. In certain embodiments, R 1B is C 1-4 haloalkyl. In certain embodiments, R 1B is selected from the groups depicted in the compounds in Table 1 below. [0159] As defined generally above, R 2B is hydrogen, halo, C 1-4 alkyl, or C 1-4 haloalkyl. In certain embodiments, R 2B is hydrogen. In certain embodiments, R 2B is halo. In certain embodiments, R 2B is C1-4 alkyl. In certain embodiments, R 2B is C1-4 haloalkyl. In certain embodiments, R 2B is selected from the groups depicted in the compounds in Table 1 below. [0160] As defined generally above, R 3B is --N(R 8B )SO 2 R 9B , -SO 2 N(R 8B ) 2 , -SO 2 R 9B , -(C 1-6 alkylene)-SO2R 9B , C1-6 hydroxyalkyl, -O-(phenyl), or one of the following ring groups: (a) a 4-7 membered saturated carbocyclic ring in which one CH2 is replaced with SO2, (b) a 4-7 membered saturated heterocyclic ring containing 1 or 2 heteroatoms independently selected from nitrogen, in which one CH 2 is replaced with SO 2 , or (c) a 3-5 membered heterocyclic ring containing 1 or 2 heteroatoms independently selected from nitrogen and oxygen; [0161] wherein each phenyl and heterocyclic ring is substituted with q occurrences of R 10B . In certain embodiments, R 3B is -N(R 8B )SO2R 9B , -SO2N(R 8B )2, -SO2R 9B , or -(C1-6 alkylene)-SO2R 9B . In certain embodiments, R 3B is -N(R 8B )SO 2 R 9B . In certain embodiments, R 3B is SO 2 R 9B . In certain embodiments, R 3B is -(C 1-6 alkylene)-SO 2 R 9B . In certain embodiments, R 3B is -SO 2 N(R 8B ) 2 . In certain embodiments, R 3B is C1-6 hydroxyalkyl. In certain embodiments, R 3B is -C(OH)(CH3)2. In certain embodiments, R 3B is or a 4-7 membered saturated carbocyclic ring in which one CH2 is replaced with SO 2 . In certain embodiments, R 3B is -O-(phenyl), wherein the phenyl substituted with q occurrences of R 10B . In certain embodiments, R 3B is a 4-7 membered saturated heterocyclic ring containing 1 or 2 heteroatoms independently selected from nitrogen, in which one CH 2 is replaced with SO2, wherein the heterocyclic ring is substituted with q occurrences of R 10B . In certain embodiments, R 3B is a 3-5 membered heterocyclic ring containing 1 or 2 heteroatoms independently selected from nitrogen and oxygen, wherein the heterocyclic ring is substituted with q occurrences of R 10B . In certain embodiments, R 3B is selected from the groups depicted in the compounds in Table 1 below. [0162] As defined generally above, R 4B is hydrogen, halo, or C 1-4 alkyl. In certain embodiments, R 4B is hydrogen. In certain embodiments, R 4B is halo. In certain embodiments, R 4B is C1-4 alkyl. In certain embodiments, R 4B is selected from the groups depicted in the compounds in Table 1 below. [0163] As defined generally above, R 5B is C 1-4 alkyl or C 3-4 cycloalkyl. In certain embodiments, R 5B is C1-4 alkyl. In certain embodiments, R 5B is methyl. In certain embodiments, R 5B is C3-4 cycloalkyl. In certain embodiments, R 5B is selected from the groups depicted in the compounds in Table 1 below. [0164] As defined generally above, R 6B is hydrogen, C1-4 alkyl, or C3-4 cycloalkyl. In certain embodiments, R 6B is hydrogen. In certain embodiments, R 6B is C 1-4 alkyl. In certain embodiments, R 6B is C 3-4 cycloalkyl. In certain embodiments, R 6B is selected from the groups depicted in the compounds in Table 1 below. [0165] As defined generally above, R 7B represents independently for each occurrence C1-4 alkyl or C 3-4 cycloalkyl. In certain embodiments, R 7B is C 1-4 alkyl. In certain embodiments, R 7B is methyl. In certain embodiments, R 7B is C 3-4 cycloalkyl. In certain embodiments, R 7B is selected from the groups depicted in the compounds in Table 1 below. [0166] As defined generally above, R 8B represents independently for each occurrence hydrogen or C 1-4 alkyl; or two occurrences of R 8B are taken together with the nitrogen atom to which they are attached to form a 3-7 membered heterocyclic ring containing 1 or 2 heteroatoms independently selected from nitrogen and oxygen; or R 8B and R 9B are taken together with their intervening atoms to a form a 5-7 membered ring containing 1, 2, or 3 heteroatoms independently selected from nitrogen, oxygen, and sulfur. In certain embodiments, R 8B represents independently for each occurrence hydrogen or C1-4 alkyl. In certain embodiments, R 8B is hydrogen. In certain embodiments, R 8B is C 1-4 alkyl. In certain embodiments, two occurrences of R 8B are taken together with the nitrogen atom to which they are attached to form a 3-7 membered heterocyclic ring containing 1 or 2 heteroatoms independently selected from nitrogen and oxygen. In certain embodiments, R 8B and R 9B are taken together with their intervening atoms to a form a 5-7 membered ring containing 1, 2, or 3 heteroatoms independently selected from nitrogen, oxygen, and sulfur. In certain embodiments, R 8B is selected from the groups depicted in the compounds in Table 1 below. [0167] As defined generally above, R 9B is C 1-6 alkyl, C 1-6 haloalkyl, -(C 1-6 alkylene)-(C 3-6 cycloalkyl), or C3-6 cycloalkyl. In certain embodiments, R 9B is C1-6 alkyl or C1-6 haloalkyl. In certain embodiments, R 9B is C1-6 alkyl. In certain embodiments, R 9B is C1-6 haloalkyl. In certain embodiments, R 9B is -(C 1-6 alkylene)-(C 3-6 cycloalkyl). In certain embodiments, R 9B is C 3-6 cycloalkyl. In certain embodiments, R 9B is selected from the groups depicted in the compounds in Table 1 below. [0168] As defined generally above, R 10B represents independently for each occurrence hydroxyl, halo, or C 1-4 alkyl. In certain embodiments, R 10B represents independently for each occurrence halo. In certain embodiments, R 10B represents independently for each occurrence C1-4 alkyl. In certain embodiments, R 10B is hydrogen. In certain embodiments, R 10B is halo. In certain embodiments, R 10B is C 1-4 alkyl. In certain embodiments, R 10B is methyl. In certain embodiments, R 10B is selected from the groups depicted in the compounds in Table 1 below. [0169] In certain embodiments, p is 2. In certain embodiments, p is 1. In certain embodiments, p is 0. In certain embodiments, q is 2. In certain embodiments, q is 1. In certain embodiments, q is 0. In certain embodiments, t is 2. In certain embodiments, t is 0. In certain embodiments, t is 1. In certain embodiments, p is selected from the corresponding value in the groups depicted in the compounds in Table 1 below. In certain embodiments, q is selected from the corresponding value in the groups depicted in the compounds in Table 1 below. In certain embodiments, t is selected from the corresponding value in the groups depicted in the compounds in Table 1 below. [0170] In certain embodiments, the EPL is selected from the groups depicted in the compounds in Table 1 below. [0171] In certain embodiments, the EPL is defined by variable A 2 set forth above in connection with Formula I. In certain embodiments, the EPL is defined by one or more of the embodiments for variable A 2 set forth in connection with Formula I. [0172] The compounds may be further characterized according to, for example, the identity of L. Exemplary further embodiments for L are provided in Part C below. Part C: Exemplary Further Description of Linker (L) Component of Compounds Herein Such as Formula I and II [0173] Compounds herein above, such as of Formula I and II, may be further characterized according to, for example, the identity of the linker (L) component. A variety of linkers are known to one of skill in the art and may be used in the heterobifunctional compounds described herein. For example, in certain embodiments, L comprises one or more optionally substituted groups selected from amino acids, polyether chains, aliphatic groups, and any combinations thereof. In certain embodiments, L consists of one or more optionally substituted groups selected from amino acids, polyether chains, aliphatic groups, and any combinations thereof. In certain embodiments, L consists of one or more groups selected from amino acids, polyether chains, aliphatic groups, and any combinations thereof. [0174] In some embodiments, L is symmetrical. In some embodiments, L is asymmetric. In certain embodiments, L is a bond. [0175] In certain embodiments, L is a covalent bond or a bivalent C 1-30 saturated or unsaturated, straight or branched, hydrocarbon chain, wherein 1-15 methylene units of L are optionally and independently replaced by cyclopropylene, -N(H)-, -N(C1-4 alkyl)-, -N(C3-5 cycloalkyl)-, -O-, - C(O)-, -OC(O)-, -C(O)O-, -S-, -S(O)-, -S(O)2-, -S(O)2N(H)-, -S(O)2N(C1-4 alkyl)-, -S(O)2N(C3-5 cycloalkyl)-, -N(H)C(O)-, -N(C 1-4 alkyl)C(O)-, -N(C 3-5 cycloalkyl)C(O)-, -C(O)N(H)-, -C(O)N(C 1- 4 alkyl)-, -C(O)N(C 3-5 cycloalkyl)-, phenylene, an 8-10 membered bicyclic arylene, a 4-7 membered saturated or partially unsaturated carbocyclylene, an 8-10 membered bicyclic saturated or partially unsaturated carbocyclylene, a 3-7 membered saturated or partially unsaturated heterocyclylene having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur, an 8-10 membered bicyclic saturated or partially unsaturated heterocyclylene having 1-2 heteroatoms independently selected from nitrogen, oxygen, and sulfur, a 5-6 membered heteroarylene having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, or an 8-10 membered bicyclic heteroarylene having 1-5 heteroatoms independently selected from nitrogen, oxygen, or sulfur. [0176] In certain embodiments, L is a bivalent, saturated or unsaturated, straight or branched C1-60 hydrocarbon chain, wherein 0-20 methylene units of the hydrocarbon are independently replaced with -O-, -S-, -N(R**)-, -OC(O)-, -C(O)O-, -S(O)-, -S(O) 2 -, -N(R**)S(O) 2 -, - S(O) 2 N(R**)-, -N(R**)C(O)-, -C(O)N(R**)-, -OC(O)N(R**)-, -N(R**)C(O)O-, optionally substituted 3-10 membered carbocyclyl, or optionally substituted 3-10 membered heterocyclyl containing 1, 2, 3, or 4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, wherein R** represents independently for each occurrence hydrogen, C 1-6 alkyl, or C 3-6 cycloalkyl. [0177] In certain embodiments, L is a bivalent, saturated or unsaturated, straight or branched C1-60 hydrocarbon chain, wherein 0-20 methylene units of the hydrocarbon are independently replaced with -O-, -S-, -N(H)-, -N(C 1-6 alkyl)-, -OC(O)-, -C(O)O-, -S(O)-, -S(O) 2 -, -N(H)S(O) 2 -, - N(C 1-6 alkyl)S(O) 2 -, -S(O) 2 N(H)-, -S(O) 2 N(C 1-6 alkyl)-, -N(H)C(O)-, -N(C 1-6 alkyl)C(O)-, - C(O)N(H)-, -C(O)N(C1-6 alkyl)-, -OC(O)N(H)-, -OC(O)N(C1-6 alkyl)-, -N(H)C(O)O-, -N(C1-6 alkyl)C(O)O-, -N(C 3-6 cycloalkyl)-, -C(halo) 2 -, -C(H)(halo)-, phenyl, pyridinyl, pyrimidinyl, pyrazinyl, pyridazinyl, optionally substituted 3-10 membered carbocyclyl, or optionally substituted 3-10 membered heterocyclyl containing 1, 2, 3, or 4 heteroatoms independently selected from nitrogen, oxygen, and sulfur. [0178] In certain embodiments, L is a bivalent, saturated or unsaturated, straight or branched C1-60 hydrocarbon chain, wherein 0-20 methylene units of the hydrocarbon are independently replaced with -O-, -S-, -N(H)-, -N(C1-6 alkyl)-, -OC(O)-, -C(O)O-, -S(O)-, -S(O)2-, -N(H)S(O)2-, - N(C 1-6 alkyl)S(O) 2 -, -S(O) 2 N(H)-, -S(O) 2 N(C 1-6 alkyl)-, -N(H)C(O)-, -N(C 1-6 alkyl)C(O)-, - C(O)N(H)-, -C(O)N(C 1-6 alkyl)-, -OC(O)N(H)-, -OC(O)N(C 1-6 alkyl)-, -N(H)C(O)O-, -N(C 1-6 alkyl)C(O)O-, optionally substituted 3-10 membered carbocyclyl, or optionally substituted 3-10 membered heterocyclyl containing 1, 2, 3, or 4 heteroatoms independently selected from nitrogen, oxygen, and sulfur. [0179] In certain embodiments, L is a bivalent, saturated, straight or branched C3-30 hydrocarbon chain, wherein 0-15 methylene units of the hydrocarbon are independently replaced with -O-, -N(H)-, -N(C 1-6 alkyl)-, -OC(O)-, -C(O)O-, -N(H)C(O)-, -N(C 1-6 alkyl)C(O)-, - C(O)N(H)-, -C(O)N(C1-6 alkyl)-, 3-10 membered carbocyclyl, or 3-10 membered heterocyclyl containing 1, 2, or 3 heteroatoms independently selected from nitrogen, oxygen, and sulfur. [0180] In certain embodiments, L is a bivalent, saturated, straight or branched C 3-30 hydrocarbon chain, wherein 0-15 methylene units of the hydrocarbon are independently replaced with -O-, -N(H)-, -N(C 1-6 alkyl)-, -OC(O)-, -C(O)O-, -N(H)C(O)-, -N(C 1-6 alkyl)C(O)-, - C(O)N(H)-, or -C(O)N(C1-6 alkyl)-. [0181] In yet other embodiments, L comprises a polyethylene glycol chain ranging in size from about 1 to about 12 ethylene glycol units, from about 1 to about 10 ethylene glycol units, from about 2 to about 6 ethylene glycol units, from about 2 to about 5 ethylene glycol units, or from about 2 to about 4 ethylene glycol units. In yet other embodiments, L is a diradical of a polyethylene glycol chain ranging in size from about 1 to about 12 ethylene glycol units, from about 1 to about 10 ethylene glycol units, from about 2 to about 6 ethylene glycol units, from about 2 to about 5 ethylene glycol units, or from about 2 to about 4 ethylene glycol units. [0182] In certain embodiments, L is a heteroalkylene having from 4 to 30 atoms selected from carbon, oxygen, nitrogen, and sulfur. In certain embodiments, L is a heteroalkylene having from 4 to 20 atoms selected from carbon, oxygen, nitrogen, and sulfur. In certain embodiments, L is a heteroalkylene having from 4 to 10 atoms selected from carbon, oxygen, nitrogen, and sulfur. In certain embodiments, L is a heteroalkylene having from 4 to 30 atoms selected from carbon, oxygen, and nitrogen. In certain embodiments, L is a heteroalkylene having from 4 to 20 atoms selected from carbon, oxygen, and nitrogen. In certain embodiments, L is a heteroalkylene having from 4 to 10 atoms selected from carbon, oxygen, and nitrogen. In certain embodiments, L is a heteroalkylene having from 4 to 30 atoms selected from carbon and oxygen. In certain embodiments, L is a heteroalkylene having from 4 to 20 atoms selected from carbon and oxygen. In certain embodiments, L is a heteroalkylene having from 4 to 10 atoms selected from carbon and oxygen. [0183] In additional embodiments, the L is an optionally substituted (poly)ethyleneglycol having between 1 and about 100 ethylene glycol units, between about 1 and about 50 ethylene glycol units, between 1 and about 25 ethylene glycol units, between about 1 and about 10 ethylene glycol units, between 1 and about 8 ethylene glycol units, between 1 and about 6 ethylene glycol units, between 2 and about 4 ethylene glycol units, or optionally substituted alkyl groups interdispersed with optionally substituted, O, N, S, P or Si atoms. In certain embodiments, L is substituted with an aryl, phenyl, benzyl, alkyl, alkylene, or heterocycle group. [0184] In certain embodiments, L is a bivalent, saturated or unsaturated, straight or branched C 1-45 hydrocarbon chain, wherein 0-10 methylene units of the hydrocarbon are independently replaced with -O-, -S-, -N(R**)-, -OC(O)-, -C(O)O-, -S(O)-, -S(O) 2 -, -N(R**)S(O) 2 -, - S(O) 2 N(R**)-, -N(R**)C(O)-, -C(O)N(R**)-, -OC(O)N(R**)-, -N(R**)C(O)O-, optionally substituted carbocyclyl, or optionally substituted heterocyclyl, wherein R** represents independently for each occurrence hydrogen, C 1-6 alkyl, or C 3-6 cycloalkyl. [0185] In certain embodiments, L is a bivalent, saturated or unsaturated, straight or branched C1-45 hydrocarbon chain, wherein 0-10 methylene units of the hydrocarbon are independently replaced with -O-, -S-, -N(R**)-, -OC(O)-, -C(O)O-, -S(O)-, -S(O)2-, -N(R**)S(O)2-, - S(O) 2 N(R**)-, -N(R**)C(O)-, -C(O)N(R**)-, -OC(O)N(R**)-, -N(R**)C(O)O-, optionally substituted 3-10 membered carbocyclyl, or optionally substituted 3-10 membered heterocyclyl containing 1, 2, 3, or 4 heteroatoms selected from nitrogen, oxygen, and sulfur, wherein R** represents independently for each occurrence hydrogen, C 1-6 alkyl, or C 3-6 cycloalkyl. [0186] In certain embodiments, L has the formula -N(R)-(optionally substituted 3-20 membered heteroalkylene)p-CH2-C(O)-, wherein R is hydrogen or optionally substituted C1-C6 alkyl, and p is 0 or 1. [0187] In certain embodiments, L has the formula -N(R)-(3-20 membered heteroalkylene) p - CH2-C(O)-; wherein the 3-20 membered heteroalkylene is optionally substituted with 1, 2, 3, or 4 substituents independently selected from halogen, C 1 -C 6 haloalkyl, C 3 -C 6 cycloalkyl, hydroxyl, and cyano; R is hydrogen or optionally substituted C 1 -C 6 alkyl; and p is 0 or 1. [0188] In certain embodiments, L has the formula -N(R)-(3-20 membered heteroalkylene)p- CH2-C(O)-; wherein the 3-20 membered heteroalkylene is optionally substituted with 1, 2, or 3 substituents independently selected from halogen and C 1 -C 6 haloalkyl; R is hydrogen or C 1 -C 6 alkyl; and p is 0 or 1. [0189] In certain embodiments, L is a bivalent, saturated or unsaturated, straight or branched C 1-60 hydrocarbon chain, wherein 0-20 methylene units of the hydrocarbon are independently replaced with -O-, -S-, -N(H)-, -N(C 1-6 alkyl)-, -OC(O)-, -C(O)O-, -S(O)-, -S(O) 2 -, -N(H)S(O) 2 -, - N(C1-6 alkyl)S(O)2-, -S(O)2N(H)-, -S(O)2N(C1-6 alkyl)-, -N(H)C(O)-, -N(C1-6 alkyl)C(O)-, - C(O)N(H)-, -C(O)N(C 1-6 alkyl)-, -OC(O)N(H)-, -OC(O)N(C 1-6 alkyl)-, -N(H)C(O)O-, -N(C 1-6 alkyl)C(O)O-, optionally substituted 3-10 membered carbocyclyl, or optionally substituted 3-10 membered heterocyclyl containing 1, 2, 3, or 4 heteroatoms independently selected from nitrogen, oxygen, and sulfur. [0190] In certain embodiments, L is a bivalent, saturated, straight or branched C 3-30 hydrocarbon chain, wherein 0-15 methylene units of the hydrocarbon are independently replaced with -O-, -N(H)-, -N(C1-6 alkyl)-, -OC(O)-, -C(O)O-, -N(H)C(O)-, -N(C1-6 alkyl)C(O)-, - C(O)N(H)-, -C(O)N(C 1-6 alkyl)-, 3-10 membered carbocyclyl, or 3-10 membered heterocyclyl containing 1, 2, or 3 heteroatoms independently selected from nitrogen, oxygen, and sulfur. [0191] In certain embodiments, L is a bivalent, saturated, straight or branched C3-30 hydrocarbon chain, wherein 0-15 methylene units of the hydrocarbon are independently replaced with -O-, -N(H)-, -N(C 1-6 alkyl)-, -OC(O)-, -C(O)O-, -N(H)C(O)-, -N(C 1-6 alkyl)C(O)-, - C(O)N(H)-, or -C(O)N(C1-6 alkyl)-. [0192] In certain embodiments, L is a bivalent, saturated or unsaturated, straight or branched C 5-40 hydrocarbon chain, wherein 1-20 methylene units of the hydrocarbon are independently replaced with -O-, -N(H)-, -N(C 1-6 alkyl)-, -N(H)C(O)-, -N(C 1-6 alkyl)C(O)-, -C(O)N(H)-, - C(O)N(C1-6 alkyl)-, optionally substituted 3-10 membered carbocyclyl, or optionally substituted 3- 10 membered heterocyclyl containing 1, 2, 3, or 4 heteroatoms independently selected from nitrogen, oxygen, and sulfur. [0193] In certain embodiments, L is -(3-7 membered, monocyclic, saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen)-(OCH 2 CH 2 ) 1-15 -O-***, wherein *** is the point of attachment to A 2 . [0194] In certain embodiments, L is -(3-7 membered, monocyclic, saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen)-(OCH2CH2)1-5-O-***, wherein *** is the point of attachment to A 2 . [0195] In certain embodiments, L is -(3-7 membered, monocyclic, saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen)-(OCH2CH2)6-10-O-***, wherein *** is the point of attachment to A 2 . [0196] In certain embodiments, L is -piperidinylene-(OCH 2 CH 2 ) 1-15 -O-***, wherein *** is the point of attachment to A 2 . [0197] In certain embodiments, L is , wherein *** is the point of attachment to A 2 . In certain embodiments, L is , wherein *** is the point of attachment to A 2 . In certain embodiments, L is , wherein *** is the point of attachment to A 2 . [0198] In certain embodiments, L is -(3-7 membered, monocyclic, saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen)-(OCH 2 CH 2 ) 1-15 -N(H)C(O)-C 1-10 alkylene- ***, -(3-7 membered, monocyclic, saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen)-(OCH2CH2)1-15-N(C1-4 alkyl)C(O)-C1-10 alkylene-***, -(3-7 membered, monocyclic, saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen)- (OCH2CH2)1-15-C(O)N(H)-C1-10 alkylene-***, or -(3-7 membered, monocyclic, saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen)-(OCH2CH2)1-15-C(O)N(C1- 4 alkyl)-C 1-10 alkylene-***, wherein *** is the point of attachment to A 2 . [0199] In certain embodiments, L is -(3-7 membered, monocyclic, saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen)-(OCH2CH2)1-10-N(H)C(O)-C1-5 alkylene- ***, -(3-7 membered, monocyclic, saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen)-(OCH 2 CH 2 ) 1-10 -N(C 1-4 alkyl)C(O)-C 1-5 alkylene-***, -(3-7 membered, monocyclic, saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen)- (OCH2CH2)1-10-C(O)N(H)-C1-5 alkylene-***, or -(3-7 membered, monocyclic, saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen)-OCH 2 CH 2 ) 1-10 -C(O)N(C 1-4 alkyl)-C1-5 alkylene-***, wherein *** is the point of attachment to A 2 . [0200] In certain embodiments, L is -piperidinylene-(OCH2CH2)1-5-N(H)C(O)-C1-5 alkylene- ***, -piperidinylene-(OCH 2 CH 2 ) 1-5 -N(C 1-4 alkyl)C(O)-C 1-5 alkylene-***, -piperidinylene- (OCH 2 CH 2 ) 1-5 -C(O)N(H)-C 1-5 alkylene-***, or -piperidinylene-(OCH 2 CH 2 ) 1-5 -C(O)N(C 1-4 alkyl)- C1-5 alkylene-***, wherein *** is the point of attachment to A 2 . [0201] In certain embodiments, L is -(3-7 membered, monocyclic, saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen)-(OCH 2 CH 2 ) 1-10 -***, -(3-7 membered, monocyclic, saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen)-(C0- 10 alkylene)-O-***, or -(3-7 membered, monocyclic, saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen)-C 1-10 alkylene, wherein *** is the point of attachment to A 2 . [0202] In certain embodiments, L is -piperidinylene-(OCH2CH2)1-5-***, -piperidinylene-(C0-5 alkylene)-O-***, or -piperidinylene-(C1-5 alkylene)-***, wherein *** is the point of attachment to A 2 . [0203] In certain embodiments, L is -(3-7 membered monocyclic saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen)-X 1 -***, wherein *** is the point of attachment to A 2 , and X 1 is (i) C 1-10 alkylene where 1 or 2 methylene groups are optionally replaced by -O-, -N(H)-, or -N(C 1-4 alkyl)-, (ii) a 3-7 membered monocyclic saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen, or (iii) -(3-7 membered monocyclic saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen)-(C1-10 alkylene)-. [0204] In certain embodiments, L is -(piperidinylene)-X 1 -***, wherein *** is the point of attachment to A 2 , and X 1 is (i) C1-5 alkylene where 1 or 2 methylene groups are optionally replaced by -O-, -N(H)-, or -N(C1-4 alkyl)-, (ii) a 3-4 membered monocyclic saturated heterocyclic ring containing 1 heteroatom selected from nitrogen, or (iii) -(3-4 membered monocyclic saturated heterocyclic ring containing 1 heteroatom selected from nitrogen)-(C 1-5 alkylene)-. [0205] In certain embodiments, L is , wherein *** is the point of attachment to A 2 , and X 1 is (i) C 1-10 alkylene where 1 or 2 methylene groups are optionally replaced by -O-, - N(H)-, or -N(C 1-4 alkyl)-, (ii) a 3-7 membered monocyclic saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen, or (iii) -(3-7 membered monocyclic saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen)-(C1-10 alkylene)-. [0206] In certain embodiments, L is -(piperazinylene)-X 1 -***, wherein *** is the point of attachment to A 2 , and X 1 is (i) C1-5 alkylene where 1 or 2 methylene groups are optionally replaced by -O-, (ii) a 3-4 membered monocyclic saturated heterocyclic ring containing 1 heteroatom selected from nitrogen, or (iii) -(3-4 membered monocyclic saturated heterocyclic ring containing 1 heteroatom selected from nitrogen)-(C 1-5 alkylene)-. [0207] In certain embodiments, L is , wherein *** is the point of attachment to A 2 , and X 1 is (i) C1-10 alkylene where 1 or 2 methylene groups are optionally replaced by -O-, (ii) a 3-7 membered monocyclic saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen, or (iii) -(3-7 membered monocyclic saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen)-(C1-10 alkylene)-. [0208] In certain embodiments, L is -(3-7 membered monocyclic saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen)-X 2 -(C1-10 alkylene)-***, wherein *** is the point of attachment to A 2 , and X 2 is -O-, -N(H)-, or -N(C1-6 alkyl)-. In certain embodiments, L is - (3-7 membered monocyclic saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen)-X 2 -(C1-10 alkylene)-***, wherein *** is the point of attachment to A 2 , and X 2 is -O-. [0209] In certain embodiments, L is -(piperidinylene)-X 2 -(C 1-10 alkylene)-***, wherein *** is the point of attachment to A 2 , and X 2 is -O-, -N(H)-, or -N(C 1-6 alkyl)-. In certain embodiments, L is -(piperidinylene)-X 2 -(C1-10 alkylene)-***, wherein *** is the point of attachment to A 2 , and X 2 is is -O-. [0210] In certain embodiments, L is -(piperidinylene)-X 2 -(a 3-7 membered monocyclic saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen)-***, wherein *** is the point of attachment to A 2 , and X 2 is -O-, -N(H)-, or -N(C1-6 alkyl)-. In certain embodiments, L is -(piperidinylene)-X 2 -(a 3-7 membered monocyclic saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen)-***, wherein *** is the point of attachment to A 2 , and X 2 is -O-. [ point of attachment to A 2 , and X 2 is -O-, -N(H)-, or -N(C 1-6 alkyl)-. In certain embodiments, L is [0212] In certain embodiments, L is , wherein *** is the point of attachment to A 2 , and X 2 is -O-, -N(H)-, or -N(C 1-6 alkyl)-. In certain embodiments, L is , wherein *** is the point of attachment to A 2 , and X 2 is -O-. [0213] In certain embodiments, L is -(3-7 membered monocyclic saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen)-X 1 -***, wherein *** is the point of attachment to A 2 , and X 1 is -(OCH2CH2)1-10 where 1 CH2 group is optionally replaced with - C(H)(C3-6 cycloalkyl)-. [0214] In certain embodiments, L is a 7-11 membered spirocyclic or fused bicyclic saturated heterocyclic ring containing 1, 2, or 3 heteroatoms selected from nitrogen and oxygen. In certain embodiments, L is a 7-8 membered spirocyclic or fused bicyclic saturated heterocyclic ring containing 2 heteroatoms selected from nitrogen. [0215] In certain embodiments, L is -(3-7 membered monocyclic saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen)-X 3 -(3-7 membered monocyclic saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen)-, wherein X 3 is C1-10 alkylene, -O-, -N(H)-, -N(C1-4 alkyl)-, or a bond. [0216] In certain embodiments, L is -(piperidinylene)-(C 1-5 alkylene)-(piperazinylene)-***, wherein *** is the point of attachment to A 2 . [0217] In certain embodiments, L is -(piperazinylene)-(azetidinylene)-*** or (azetidinylene)- (piperazinylene)-***, wherein *** is the point of attachment to A 2 . [0218] In certain embodiments, L is -(3-7 membered monocyclic saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen)-X 3 -(C3-6 cycloalkylene)-O-***, -(3-7 membered monocyclic saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen)-X 3 -(C 3-6 cycloalkylene)-N(H)-***, or -(3-7 membered monocyclic saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen)-X 3 -(C3-6 cycloalkylene)-N(C1-4 alkyl)- ***, wherein *** is the point of attachment to A 2 , and X 3 is C 1-10 alkylene, -O-, -N(H)-, -N(C 1-4 alkyl)-, or a bond. [0219] In certain embodiments, L is -(piperidinylene)-X 3 -(C3-6 cycloalkylene)-O-***, - (piperidinylene)-X 3 -(C3-6 cycloalkylene)-N(H)-***, or -(piperidinylene)-X 3 -(C3-6 cycloalkylene)- ***, wherein *** is the point of attachment to A 2 , and X 3 is C 1-10 alkylene, -O-, -N(H)-, -N(C 1-4 alkyl)-, or a bond. [0220] In certain embodiments, L has the formula –(C0-12 alkylene)-(optionally substituted 3-40 membered heteroalkylene)-(C 0-12 alkylene)-. [0221] In certain embodiments, L is -(7-10 membered spirocyclic heterocyclylene containing 1, 2, 3, or 4 heteroatoms independently selected from nitrogen)-(C0-12 alkylene)-***, wherein *** is the point of attachment to A 2 . [0222] In certain embodiments, L is -(piperidinylene)-(C0-12 alkylene)-O-***, wherein *** is the point of attachment to A 2 . [0223] In certain embodiments, L is –(piperidinylene)-(C 0-12 alkylene)-(pyrimidinylene)-O-***, wherein *** is the point of attachment to A 2 . [ the point of attachment to A 2 . wherein *** is the point of attachment to A 2 . [0226] In certain embodiments, L is one of the following: wherein *** is the point of attachment to A 2 . Additional Exemplary Embodiments for L [0227] In certain embodiments, L is -N(H)-(C2-9 alkylene)-O-(C1-6 alkylene)-C(O)-***, -N(H)- (C 10-20 alkylene)-O-(C 1-6 alkylene)-C(O)-***, -N(H)-[(C 2-4 alkylene)-O-] 2-6 -(C 1-6 alkylene)-C(O)- ***, -N(H)-[(C 2-4 alkylene)-O-] 7-15 -(C 1-6 alkylene)-C(O)-***, -N(H)-(C 1-6 alkylene)-C(O)-***, - N(H)-(C7-15 alkylene)-C(O)-***, -N(H)-[(C2-4 alkylene)-O-]2-6-(C1-6 alkylene)-***, -N(H)-[(C2-4 alkylene)-O-]7-15-(C1-6 alkylene)-***, -N(H)-(C2-9 alkylene)-O-(C1-6 alkylene)-C(O)N(C1-6 alkyl)- (C 1-6 alkylene)-***, -N(H)-(C 2-9 alkylene)-O-(C 1-6 alkylene)-C(O)N(H)-(C 1-6 alkylene)-***, -N(H)- [(C 2-4 alkylene)-O-] 2-6 -(C 1-6 alkylene)-N(H)-(C 1-6 alkylene)-***, -N(H)-[(C 2-4 alkylene)-O-] 7-15 -(C 1-6 alkylene)-N(H)-(C1-6 alkylene)-***, -N(H)-[(C2-4 alkylene)-O-]2-6-(C1-6 alkylene)-N(C1-6 alkyl)-(C1- 6 alkylene)-***, or -N(H)-[(C 2-4 alkylene)-O-] 7-15 -(C 1-6 alkylene)-N(C 1-6 alkyl)-(C 1-6 alkylene)-***, where *** is a point of attachment to A 2 . [0228] In certain embodiments, L is -N(H)-(C2-9 alkylene)-O-(C1-6 alkylene)-C(O)-***, -N(H)- (C 10-20 alkylene)-O-(C 1-6 alkylene)-C(O)-***, -N(H)-[CH 2 CH 2 -O-] 2-6 -(C 1-6 alkylene)-C(O)-***, - N(H)-[CH 2 CH 2 -O-] 7-15 -(C 1-6 alkylene)-C(O)-***, -N(H)-(C 1-6 alkylene)-C(O)-***, -N(H)-(C 7-15 alkylene)-C(O)-***, -N(H)-[CH2CH2-O-]2-6-(C1-6 alkylene)-***, -N(H)-[CH2CH2-O-]7-15-(C1-6 alkylene)-***, -N(H)-(C2-9 alkylene)-O-(C1-6 alkylene)-C(O)N(C1-6 alkyl)-(C1-6 alkylene)-***, - N(H)-(C 2-9 alkylene)-O-(C 1-6 alkylene)-C(O)N(H)-(C 1-6 alkylene)-***, -N(H)-[CH 2 CH 2 -O-] 2-6 -(C 1-6 alkylene)-N(H)-(C 1-6 alkylene)-***, -N(H)-[CH 2 CH 2 -O-] 7-15 -(C 1-6 alkylene)-N(H)-(C 1-6 alkylene)- ***, -N(H)-[CH 2 CH 2 -O-] 2-6 -(C 1-6 alkylene)-N(C 1-6 alkyl)-(C 1-6 alkylene)-***, or -N(H)-[CH 2 CH 2 - O-]7-15-(C1-6 alkylene)-N(C1-6 alkyl)-(C1-6 alkylene)-***, where *** is a point of attachment to A 2 . [0229] In certain embodiments, L is -N(H)-[(C 2-4 alkylene)-O-] 2-6 -(C 1-6 alkylene)-C(O)-***, - N(H)-[(C 2-4 alkylene)-O-] 7-15 -(C 1-6 alkylene)-C(O)-***, -N(H)-(C 1-6 alkylene)-N(C 1-6 alkyl)C(O)- (C1-6 alkylene)***, -N(H)-(C1-6 alkylene)-N(H)C(O)-(C1-6 alkylene)***, -N(H)-(C2-6 alkylene)-***, -N(H)-(C7-15 alkylene)-***, -N(C1-6 alkyl)-(C2-6 alkylene)-***, -N(C1-6 alkyl)-(C7-15 alkylene)-***, - N(H)-[(C 2-4 alkylene)-O-] 2-6 -(C 1-6 alkylene)-***, -N(H)-[(C 2-4 alkylene)-O-] 7-15 -(C 1-6 alkylene)-***, -N(H)-(C1-6 alkylene)-(3-6 membered heterocycloalkylene)-(C1-6 alkylene)-N(C1-6 alkyl)-(C1-6 alkylene)-***, -N(H)-(C1-6 alkylene)-(3-6 membered heterocycloalkylene)-(C1-6 alkylene)-N(H)- (C 1-6 alkylene)-***, -N(H)-(C 2-6 alkylene)-N(H)-(C 1-6 alkylene)-***, or -N(H)-(C 2-6 alkylene)- N(C 1-6 alkyl)-(C 1-6 alkylene)-***, where *** is a point of attachment to A 2 . [0230] In certain embodiments, L is -N(H)-[CH2CH2-O-]2-6-(C1-6 alkylene)-C(O)-***, -N(H)- [CH 2 CH 2 -O-] 7-15 -(C 1-6 alkylene)-C(O)-***, -N(H)-(C 1-6 alkylene)-N(C 1-6 alkyl)C(O)-(C 1-6 alkylene)***, -N(H)-(C 1-6 alkylene)-N(H)C(O)-(C 1-6 alkylene)***, -N(H)-(C 2-6 alkylene)-***, - N(H)-(C7-15 alkylene)-***, -N(C1-6 alkyl)-(C2-6 alkylene)-***, -N(C1-6 alkyl)-(C7-15 alkylene)-***, - N(H)-[CH 2 CH 2 -O-] 2-6 -(C 1-6 alkylene)-***, -N(H)-[CH 2 CH 2 -O-] 7-15 -(C 1-6 alkylene)-***, -N(H)-(C 1- 6 alkylene)-(3-6 membered heterocycloalkylene)-(C 1-6 alkylene)-N(C 1-6 alkyl)-(C 1-6 alkylene)-***, - N(H)-(C1-6 alkylene)-(3-6 membered heterocycloalkylene)-(C1-6 alkylene)-N(H)-(C1-6 alkylene)- ***, -N(H)-(C2-6 alkylene)-N(H)-(C1-6 alkylene)-***, or -N(H)-(C2-6 alkylene)-N(C1-6 alkyl)-(C1-6 alkylene)-***, where *** is a point of attachment to A 2 . [0231] In certain embodiments, L is -[(C 2-4 alkylene)-O-] 2-6 -(C 1-6 alkylene)-***, -[(C 2-4 alkylene)-O-]7-15-(C1-6 alkylene)-***, -[(C2-4 alkylene)-O-]2-6-(C1-6 alkylene)-N(C1-6 alkyl)(C1-6 alkylene)-***, -[(C 2-4 alkylene)-O-] 7-15 -(C 1-6 alkylene)-N(C 1-6 alkyl)(C 1-6 alkylene)-***, -[(C 2-4 alkylene)-O-] 2-6 -(C 1-6 alkylene)-N(H)(C 1-6 alkylene)-***, -[(C 2-4 alkylene)-O-] 7-15 -(C 1-6 alkylene)- N(H)(C1-6 alkylene)-***, -(C1-9 alkylene)-C(O)N(H)-(C1-6 alkylene)-***, -(C1-9 alkylene)- N(H)C(O)-(C 1-6 alkylene)-***, -(C 1-9 alkylene)-C(O)N(H)-[(C 2-4 alkylene)-O-] 2-6 -(C 1-6 alkylene)- ***, -(C 1-9 alkylene)-N(H)C(O)-[(C 2-4 alkylene)-O-] 2-6 -(C 1-6 alkylene)-***, -(C 1-9 alkylene)- C(O)N(H)-[(C2-4 alkylene)-O-]7-15-(C1-6 alkylene)-***, -(C1-9 alkylene)-N(H)C(O)-[(C2-4 alkylene)- O-]7-15-(C1-6 alkylene)-***, -(C1-9 alkylene)-C(O)N(H)-[(C2-4 alkylene)-O-]2-6-(C1-6 alkylene)-N(C1-6 alkyl)-(C 1-6 alkylene)-***, -(C 1-9 alkylene)-N(H)C(O)-[(C 2-4 alkylene)-O-] 2-6 -(C 1-6 alkylene)-N(C 1-6 alkyl)-(C 1-6 alkylene)-***, -(C 1-9 alkylene)-C(O)N(H)-[(C 2-4 alkylene)-O-] 7-15 -(C 1-6 alkylene)-N(C 1- 6 alkyl)-(C 1-6 alkylene)-***, or -(C 1-9 alkylene)-N(H)C(O)-[(C 2-4 alkylene)-O-] 7-15 -(C 1-6 alkylene)- N(C1-6 alkyl)-(C1-6 alkylene)-***, where *** is a point of attachment to A 2 . [0232] In certain embodiments, L is -[CH 2 CH 2 -O-] 2-6 -(C 1-6 alkylene)-***, -[CH 2 CH 2 -O-] 7-15 - (C 1-6 alkylene)-***, -[CH 2 CH 2 -O-] 2-6 -(C 1-6 alkylene)-N(C 1-6 alkyl)(C 1-6 alkylene)-***, -[CH 2 CH 2 - O-]7-15-(C1-6 alkylene)-N(C1-6 alkyl)(C1-6 alkylene)-***, -[CH2CH2-O-]2-6-(C1-6 alkylene)-N(H)(C1-6 alkylene)-***, -[CH2CH2-O-]7-15-(C1-6 alkylene)-N(H)(C1-6 alkylene)-***, -(C1-9 alkylene)- C(O)N(H)-(C 1-6 alkylene)-***, -(C 1-9 alkylene)-N(H)C(O)-(C 1-6 alkylene)-***, -(C 1-9 alkylene)- C(O)N(H)-[CH2CH2-O-]2-6-(C1-6 alkylene)-***, -(C1-9 alkylene)-N(H)C(O)-[CH2CH2-O-]2-6-(C1-6 alkylene)-***, -(C1-9 alkylene)-C(O)N(H)-[CH2CH2-O-]7-15-(C1-6 alkylene)-***, -(C1-9 alkylene)- N(H)C(O)-[CH 2 CH 2 -O-] 7-15 -(C 1-6 alkylene)-***, -(C 1-9 alkylene)-C(O)N(H)-[CH 2 CH 2 -O-] 2-6 -(C 1-6 alkylene)-N(C 1-6 alkyl)-(C 1-6 alkylene)-***, -(C 1-9 alkylene)-N(H)C(O)-[CH 2 CH 2 -O-] 2-6 -(C 1-6 alkylene)-N(C1-6 alkyl)-(C1-6 alkylene)-***, -(C1-9 alkylene)-C(O)N(H)-[CH2CH2-O-]7-15-(C1-6 alkylene)-N(C 1-6 alkyl)-(C 1-6 alkylene)-***, or -(C 1-9 alkylene)-N(H)C(O)-[(CH 2 CH 2 -O-] 7-15 -(C 1-6 alkylene)-N(C 1-6 alkyl)-(C 1-6 alkylene)-***, where *** is a point of attachment to A 2 . [0233] In certain embodiments, L is -N(H)-[(C2-4 alkylene)-O-]2-6-(C1-6 alkylene)-N(H)-***, - N(H)-[(C 2-4 alkylene)-O-] 7-15 -(C 1-6 alkylene)-N(H)-***, -N(C 1-6 alkyl)-[(C 2-4 alkylene)-O-] 2-6 -(C 1-6 alkylene)-N(H)-***, -N(C 1-6 alkyl)-[(C 2-4 alkylene)-O-] 7-15 -(C 1-6 alkylene)-N(H)-***, -N(C 1-6 alkyl)-[(C2-4 alkylene)-O-]2-6-(C1-6 alkylene)-N(C1-6 alkyl)-***, or -N(C1-6 alkyl)-[(C2-4 alkylene)-O- ]7-15-(C1-6 alkylene)-N(C1-6 alkyl)-***, where *** is a point of attachment to A 2 . [0234] In certain embodiments, L is -N(H)-[CH 2 CH 2 -O-] 2-6 -(C 1-6 alkylene)-N(H)-***, -N(H)- [CH 2 CH 2 -O-] 7-15 -(C 1-6 alkylene)-N(H)-***, -N(C 1-6 alkyl)-[CH 2 CH 2 -O-] 2-6 -(C 1-6 alkylene)-N(H)- ***, -N(C1-6 alkyl)-[CH2CH2-O-]7-15-(C1-6 alkylene)-N(H)-***, -N(C1-6 alkyl)-[CH2CH2-O-]2-6-(C1-6 alkylene)-N(C 1-6 alkyl)-***, or -N(C 1-6 alkyl)-[CH 2 CH 2 -O-] 7-15 -(C 1-6 alkylene)-N(C 1-6 alkyl)-***, where *** is a point of attachment to A 2 .

[0235] In some embodiments, L is one of the following:

; wherein a dashed bond indicates a point of attachment. [0236] In certain embodiments, L is -C(H)(R 100 )-, -C(R 100 )2-, O, -N(R 101 )-, -S(O)2-, an optionally substituted C 3-7 cycloalkylene, an optionally substituted C 4-7 cycloalkenylene, or an optionally substituted 3-7 membered heterocyclylene containing 1, 2 or 3 heteroatoms selected from oxygen, nitrogen, and sulfur; wherein R 100 represents independently for each occurrence hydrogen, halogen, C 1-6 alkyl, or C 3-6 cycloalkyl, and R 101 is hydrogen, C 1-6 alkyl, or C 3-6 cycloalkyl. In certain embodiments, R 100 represents independently for each occurrence hydrogen, methyl, ethyl, isopropyl, cyclopropyl, cyclobutyl, or cyclopentyl. In certain embodiments, R 101 is hydrogen, methyl, ethyl, isopropyl, cyclopropyl, cyclobutyl, or cyclopentyl. [0237] In certain embodiments, L is -CH 2 -Y 20 -, -C(H)(R 100 )-Y 20 -, -C(R 100 ) 2 -Y 20 -, -O-Y 20 -, - N(R 101 )-Y 20 -, -S(O)2-Y 20 -, -C(O)-Y 20 -, -(optionally substituted C3-7 cycloalkylene)-Y 20 -, - (optionally substituted C4-7 cycloalkenylene)-Y 20 -, -(optionally substituted 3-7 membered heterocyclylene containing 1, 2 or 3 heteroatoms selected from oxygen, nitrogen, and sulfur)-Y 20 -, -Y 20 -CH 2 -, -Y 20 -C(H)(R 100 )-, -Y 20 -C(R 100 ) 2 -, -Y 20 -O-, -Y 20 -N(R 101 )-, -Y 20 -S(O) 2 -, -Y 20 -C(O)-, - Y 20 -(optionally substituted C 3-7 cycloalkylene)-, -Y 20 -(optionally substituted C 4-7 cycloalkenylene)- , or -Y 20 -(optionally substituted 3-7 membered heterocyclylene containing 1, 2 or 3 heteroatoms selected from oxygen, nitrogen, and sulfur)-; wherein R 100 represents independently for each occurrence hydrogen, halogen, C 1-6 alkyl, or C 3-6 cycloalkyl, and R 101 is hydrogen, C 1-6 alkyl, or C 3- 6 cycloalkyl. In certain embodiments, R 100 represents independently for each occurrence hydrogen, methyl, ethyl, isopropyl, cyclopropyl, cyclobutyl, or cyclopentyl. In certain embodiments, R 101 is hydrogen, methyl, ethyl, isopropyl, cyclopropyl, cyclobutyl, or cyclopentyl. [0238] In certain embodiments, L is one of the following: [0239] wherein X 20 , Y 20 , and Z 20 are independently -CH 2 -, -C(H)(R 100 )-, -C(R 100 ) 2 -, O, - N(R 101 )-, -S(O) 2 -, -C(O)-, an optionally substituted C 3-7 cycloalkylene, or an optionally substituted C4-7 cycloalkenylene; wherein R 100 represents independently for each occurrence hydrogen, halogen, C 1-6 alkyl, or C 3-6 cycloalkyl, and R 101 is hydrogen, C 1-6 alkyl, or C 3-6 cycloalkyl. In certain embodiments, R 100 represents independently for each occurrence hydrogen, methyl, ethyl, isopropyl, cyclopropyl, cyclobutyl, or cyclopentyl. In certain embodiments, R 101 is hydrogen, methyl, ethyl, isopropyl, cyclopropyl, cyclobutyl, or cyclopentyl. [0240] In certain embodiments, L is one of the following: [0241] wherein X 20 , Y 20 , and Z 20 are independently -C(R 100 )- or -N-; wherein R 100 represents independently for each occurrence hydrogen, halogen, C1-6 alkyl, or C3-6 cycloalkyl. In certain embodiments, R 100 represents independently for each occurrence hydrogen, methyl, ethyl, isopropyl, cyclopropyl, cyclobutyl, or cyclopentyl. [0242] In certain embodiments, L is -X 20 -Y 20 -Z 20 -, wherein X 20 , Y 20 , and Z 20 are independently -CH 2 -, -C(H)(R 100 )-, -C(R 100 ) 2 -, O, -N(R 101 )-, -S(O) 2 -, -C(O)-, an optionally substituted C 3-7 cycloalkylene, an optionally substituted C 4-7 cycloalkenylene, or optionally substituted 3-7 membered heterocyclylene containing 1, 2 or 3 heteroatoms selected from oxygen, nitrogen, and sulfur; wherein R 100 represents independently for each occurrence hydrogen, halogen, C 1-6 alkyl, or C 3-6 cycloalkyl, and R 101 is hydrogen, C 1-6 alkyl, or C 3-6 cycloalkyl. In certain embodiments, R 100 represents independently for each occurrence hydrogen, methyl, ethyl, isopropyl, cyclopropyl, cyclobutyl, or cyclopentyl. In certain embodiments, R 101 is hydrogen, methyl, ethyl, isopropyl, cyclopropyl, cyclobutyl, or cyclopentyl. [0243] In certain embodiments, L is -X 20 =Y 20 -Z 21 -, wherein X 20 and Y 20 are independently - C(R 100 )- or -N-, and Z 21 is -CH2-, -C(H)(R 100 )-, -C(R 100 )2-, O, -N(R 101 )-, -S(O)2-, -C(O)-, an optionally substituted C 3-7 cycloalkylene, or an optionally substituted C 4-7 cycloalkenylene; wherein R 100 represents independently for each occurrence hydrogen, halogen, C 1-6 alkyl, or C 3-6 cycloalkyl, and R 101 is hydrogen, C1-6 alkyl, or C3-6 cycloalkyl. In certain embodiments, R 100 represents independently for each occurrence hydrogen, methyl, ethyl, isopropyl, cyclopropyl, cyclobutyl, or cyclopentyl. In certain embodiments, R 101 is hydrogen, methyl, ethyl, isopropyl, cyclopropyl, cyclobutyl, or cyclopentyl. [0244] In certain embodiments, L is -C≡C-Z 20 -, wherein Z 20 is -CH 2 -, -C(H)(R 100 )-, -C(R 100 ) 2 -, O, -N(R 101 )-, -S(O) 2 -, -C(O)-, an optionally substituted C 3-7 cycloalkylene, or an optionally substituted C4-7 cycloalkenylene; wherein R 100 represents independently for each occurrence hydrogen, halogen, C1-6 alkyl, or C3-6 cycloalkyl, and R 101 is hydrogen, C1-6 alkyl, or C3-6 cycloalkyl. In certain embodiments, R 100 represents independently for each occurrence hydrogen, methyl, ethyl, isopropyl, cyclopropyl, cyclobutyl, or cyclopentyl. In certain embodiments, R 101 is hydrogen, methyl, ethyl, isopropyl, cyclopropyl, cyclobutyl, or cyclopentyl. [0245] In certain embodiments, L is one of the following: [0246] wherein X 20 , Y 20 , and Z 20 are independently -CH2-, -C(H)(R 100 )-, -C(R 100 )2-, O, - N(R 101 )-, -S(O)2-, -C(O)-, an optionally substituted C3-7 cycloalkylene, an optionally substituted C4- 7 cycloalkenylene, or optionally substituted 3-7 membered heterocyclylene containing 1, 2 or 3 heteroatoms selected from oxygen, nitrogen, and sulfur; wherein R 100 represents independently for each occurrence hydrogen, halogen, C1-6 alkyl, or C3-6 cycloalkyl, and R 101 is hydrogen, C1-6 alkyl, or C 3-6 cycloalkyl. In certain embodiments, R 100 represents independently for each occurrence hydrogen, methyl, ethyl, isopropyl, cyclopropyl, cyclobutyl, or cyclopentyl. In certain embodiments, R 101 is hydrogen, methyl, ethyl, isopropyl, cyclopropyl, cyclobutyl, or cyclopentyl. [0247] In certain embodiments, L is one of the following:

[0248] wherein V 20 , W 20 , X 20 , Y 20 , and Z 20 are independently -CH 2 -, -C(H)(R 100 )-, -C(R 100 ) 2 -, O, -N(R 101 )-, -S(O)2-, -C(O)-, an optionally substituted C3-7 cycloalkylene, an optionally substituted C4-7 cycloalkenylene, or optionally substituted 3-7 membered heterocyclylene containing 1, 2 or 3 heteroatoms selected from oxygen, nitrogen, and sulfur; wherein R 100 represents independently for each occurrence hydrogen, halogen, C1-6 alkyl, or C3-6 cycloalkyl, and R 101 is hydrogen, C1-6 alkyl, or C3-6 cycloalkyl. In certain embodiments, R 100 represents independently for each occurrence hydrogen, methyl, ethyl, isopropyl, cyclopropyl, cyclobutyl, or cyclopentyl. In certain embodiments, R 101 is hydrogen, methyl, ethyl, isopropyl, cyclopropyl, cyclobutyl, or cyclopentyl. [0249] In certain embodiments, L is one of the following: [0250] wherein W 20 , X 20 , Y 20 , and Z 20 are independently -C(R 100 )- or -N-; wherein R 100 represents independently for each occurrence hydrogen, halogen, C1-6 alkyl, or C3-6 cycloalkyl. In certain embodiments, R 100 represents independently for each occurrence hydrogen, methyl, ethyl, isopropyl, cyclopropyl, cyclobutyl, or cyclopentyl. [0251] In certain embodiments, L is one of the following: [0252] wherein W 20 , X 20 , Y 20 , and Z 20 are independently -C(R 100 )- or -N-; wherein R 100 represents independently for each occurrence hydrogen, halogen, C 1-6 alkyl, or C 3-6 cycloalkyl. In certain embodiments, R 100 represents independently for each occurrence hydrogen, methyl, ethyl, isopropyl, cyclopropyl, cyclobutyl, or cyclopentyl. [0253] In certain embodiments, L is one of the following:

[0254] wherein U, V, W, X, Y, and Z are independently -CH2-, -C(H)(R 100 )-, -C(R 100 )2-, O, - N(R 101 )-, -S(O) 2 -, -C(O)-, an optionally substituted C 3-7 cycloalkylene, an optionally substituted C 4- 7 cycloalkenylene, or optionally substituted 3-7 membered heterocyclylene containing 1, 2 or 3 heteroatoms selected from oxygen, nitrogen, and sulfur; wherein R 100 represents independently for each occurrence hydrogen, halogen, C 1-6 alkyl, or C 3-6 cycloalkyl; and R 101 is hydrogen, C 1-6 alkyl, or C 3-6 cycloalkyl; and a dashed bond indicates a point of attachment. In certain embodiments, R 100 represents independently for each occurrence hydrogen, methyl, ethyl, isopropyl, cyclopropyl, cyclobutyl, or cyclopentyl. In certain embodiments, R 101 is hydrogen, methyl, ethyl, isopropyl, cyclopropyl, cyclobutyl, or cyclopentyl. [0255] In certain embodiments, L is one of the following: [0256] wherein X, Y, and Z are independently -C(R 100 )- or -N-; V and W are independently - CH2-, -C(H)(R 100 )-, -C(R 100 )2-, O, -N(R 101 )-, -S(O)2-, -C(O)-, an optionally substituted C3-7 cycloalkylene, an optionally substituted C 4-7 cycloalkenylene, or optionally substituted 3-7 membered heterocyclylene containing 1, 2 or 3 heteroatoms selected from oxygen, nitrogen, and sulfur; wherein R 100 represents independently for each occurrence hydrogen, halogen, C1-6 alkyl, or C3-6 cycloalkyl; R 101 is hydrogen, C1-6 alkyl, or C3-6 cycloalkyl; and a dashed bond indicates a point of attachment. In certain embodiments, R 100 represents independently for each occurrence hydrogen, methyl, ethyl, isopropyl, cyclopropyl, cyclobutyl, or cyclopentyl. In certain embodiments, R 101 is hydrogen, methyl, ethyl, isopropyl, cyclopropyl, cyclobutyl, or cyclopentyl. [0257] In certain embodiments, L is one of the following: [0258] wherein W, X, Y, and Z are independently -C(R 100 )- or -N-; V is -CH 2 -, -C(H)(R 100 )-, - C(R 100 ) 2 -, O, -N(R 101 )-, -S(O) 2 -, -C(O)-, an optionally substituted C 3-7 cycloalkylene, an optionally substituted C4-7 cycloalkenylene, or optionally substituted 3-7 membered heterocyclylene containing 1, 2 or 3 heteroatoms selected from oxygen, nitrogen, and sulfur; wherein R 100 represents independently for each occurrence hydrogen, halogen, C 1-6 alkyl, or C 3-6 cycloalkyl; R 101 is hydrogen, C1-6 alkyl, or C3-6 cycloalkyl; and a dashed bond indicates a point of attachment. In certain embodiments, R 100 represents independently for each occurrence hydrogen, methyl, ethyl, isopropyl, cyclopropyl, cyclobutyl, or cyclopentyl. In certain embodiments, R 101 is hydrogen, methyl, ethyl, isopropyl, cyclopropyl, cyclobutyl, or cyclopentyl. [0259] In certain embodiments, L is one of the following:

[0260] wherein T, U, V, W, X, Y, and Z are independently -CH2-, -C(H)(R 100 )-, -C(R 100 )2-, O, - N(R 101 )-, -S(O)2-, -C(O)-, an optionally substituted C3-7 cycloalkylene, an optionally substituted C4- 7 cycloalkenylene, or optionally substituted 3-7 membered heterocyclylene containing 1, 2 or 3 heteroatoms selected from oxygen, nitrogen, and sulfur; wherein R 100 represents independently for each occurrence hydrogen, halogen, C 1-6 alkyl, or C 3-6 cycloalkyl; R 101 is hydrogen, C 1-6 alkyl, or C 3-6 cycloalkyl; and a dashed bond indicates a point of attachment. In certain embodiments, R 100 represents independently for each occurrence hydrogen, methyl, ethyl, isopropyl, cyclopropyl, cyclobutyl, or cyclopentyl. In certain embodiments, R 101 is hydrogen, methyl, ethyl, isopropyl, cyclopropyl, cyclobutyl, or cyclopentyl. [0261] In certain embodiments, L is one of the following: [0262] wherein W, X, Y, and Z are independently -C(R 100 )- or -N-; U and V are independently -CH2-, -C(H)(R 100 )-, -C(R 100 )2-, O, -N(R 101 )-, -S(O)2-, -C(O)-, an optionally substituted C3-7 cycloalkylene, an optionally substituted C 4-7 cycloalkenylene, or optionally substituted 3-7 membered heterocyclylene containing 1, 2 or 3 heteroatoms selected from oxygen, nitrogen, and sulfur; R 100 represents independently for each occurrence hydrogen, halogen, C1-6 alkyl, or C3-6 cycloalkyl; R 101 is hydrogen, C1-6 alkyl, or C3-6 cycloalkyl; and a dashed bond indicates a point of attachment. In certain embodiments, R 100 represents independently for each occurrence hydrogen, methyl, ethyl, isopropyl, cyclopropyl, cyclobutyl, or cyclopentyl. In certain embodiments, R 101 is hydrogen, methyl, ethyl, isopropyl, cyclopropyl, cyclobutyl, or cyclopentyl. [0263] In certain embodiments, L is one of the following: [0264] wherein X, Y, and Z are independently -C(R 100 )- or -N-; U, V, and W are independently -CH2-, -C(H)(R 100 )-, -C(R 100 )2-, O, -N(R 101 )-, -S(O)2-, -C(O)-, an optionally substituted C3-7 cycloalkylene, an optionally substituted C 4-7 cycloalkenylene, or optionally substituted 3-7 membered heterocyclylene containing 1, 2 or 3 heteroatoms selected from oxygen, nitrogen, and sulfur; wherein R 100 represents independently for each occurrence hydrogen, halogen, C 1-6 alkyl, or C 3-6 cycloalkyl; R 101 is hydrogen, C 1-6 alkyl, or C 3-6 cycloalkyl; and a dashed bond indicates a point of attachment. In certain embodiments, R 100 represents independently for each occurrence hydrogen, methyl, ethyl, isopropyl, cyclopropyl, cyclobutyl, or cyclopentyl. In certain embodiments, R 101 is hydrogen, methyl, ethyl, isopropyl, cyclopropyl, cyclobutyl, or cyclopentyl. [0265] In certain embodiments, L is one of the following:

[0266] variables m, n, o, p, and q are independently 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20. [0267] In certain embodiments, L is one of the following:

wherein any m or n are independently 0, 1, 2, 3, 4, 5, or 6; and any X is H or F. [0268] In certain embodiments, L is one of the following:

wherein any m or n are independently 0, 1, 2, 3, 4, 5, or 6. [0269] In certain embodiments, L is one of the following:

[0270] wherein any m or n are independently 0, 1, 2, 3, 4, 5, or 6. [0271] In certain embodiments, L is one of the following:

[0272] In certain embodiments, L is one of the following:

[0273] In certain embodiments, L has the formula –(C0-12 alkylene)-(optionally substituted 3-40 membered heteroalkylene)-(C0-12 alkylene)-. In certain embodiments, L is C4-14 alkylene. In certain embodiments, L is -(CH 2 ) 6-10 -. [0274] In certain embodiments, L is -CH2CH2(OCH2CH2)-***, -CH2CH2(OCH2CH2)2-***, - CH2CH2(OCH2CH2)3-***, -CH2CH2(OCH2CH2)4-***, -CH2CH2(OCH2CH2)5-***, - CH 2 CH 2 (OCH 2 CH 2 ) 6 -***, -CH 2 CH 2 (OCH 2 CH 2 ) 7 -***, -CH 2 CH 2 (OCH 2 CH 2 ) 8 -***, - CH 2 CH 2 (OCH 2 CH 2 ) 9 -***, -CH 2 CH 2 (OCH 2 CH 2 ) 10 -***, -CH 2 CH 2 (OCH 2 CH 2 ) 11 -***, - CH2CH2(OCH2CH2)12-***, -CH2CH2(OCH2CH2)13-***, -CH2CH2(OCH2CH2)14-***, - point of attachment to [0275] In certain embodiments, L is -(C2-20 alkylene)-(OCH2CH2)2-4-(C0-4 alkylene)-***, -(C2-20 alkylene)-(OCH2CH2)5-7-(C0-4 alkylene)-***, -(C2-20 alkylene)-(OCH2CH2)8-10-(C0-4 alkylene)-***, -(C 2-20 alkylene)-(OCH 2 CH 2 ) 11-13 -(C 0-4 alkylene)-***, -(C 2-20 alkylene)-(OCH 2 CH 2 ) 14-16 -(C 0-4 alkylene)-***, -(C2-20 alkylene)-(OCH2CH2)17-20-(C0-4 alkylene)-***, -(C1-20 alkylene)- (OCH2CH2)1-10-(C0-4 alkylene)-C(O)-***, or -(C1-20 alkylene)-(OCH2CH2)11-20-(C0-4 alkylene)- C(O)-***, where *** is a point of attachment to A 2 . [0276] In certain embodiments, L is -O(CH 2 CH 2 O) 2-4 -(C 0-4 alkylene)-***, -O(CH 2 CH 2 O) 5-7 - (C0-4 alkylene)-***, -O(CH2CH2O)8-10-(C0-4 alkylene)-***, -O(CH2CH2O)11-13-(C0-4 alkylene)-***, -O(CH 2 CH 2 O) 14-16 -(C 0-4 alkylene)-***, -O(CH 2 CH 2 O) 16-20 -(C 0-4 alkylene)-***, -O(CH 2 CH 2 O) 2-10 - (C 0-4 alkylene)C(O)-***, or -O(CH 2 CH 2 O) 11-20 -(C 0-4 alkylene)C(O)-***, where *** is a point of attachment to A 2 . [0277] In certain embodiments, L is -(C0-20 alkylene)-(OCH2CH2)1-10-(N(C1-4 alkyl))-***, -(C0-20 alkylene)-(OCH 2 CH 2 ) 11-20 -(N(C 1-4 alkyl))-***, -(C 0-20 alkylene)-(CH 2 CH 2 O) 1-10 -(C 2-10 alkylene)- (N(C1-4 alkyl))-(C0-10 alkylene)-***, or -(C0-20 alkylene)-(CH2CH2O)11-20-(C2-10 alkylene)-(N(C1-4 alkyl))-(C0-10 alkylene)-***, where *** is a point of attachment to A 2 . [0278] In certain embodiments, L is -(5-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-O-(C 1-6 alkylene)-***, -(5-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-(C1-6 alkylene)-***, -(5-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-(C 1-6 alkylene)-O-***, -(7-11 membered spirocyclic heterocyclyl containing 1-2 heteroatoms selected from nitrogen)-O-(C0-6 alkylene)-***, -(7-11 membered spirocyclic heterocyclyl containing 1-2 heteroatoms selected from nitrogen)-(C 0-6 alkylene)-***, -(4-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-(C 0-6 alkylene)-***, -(4-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-O-***, -(6-11 membered bicyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-(C1-6 alkylene)-***, -(5-6 membered monocyclic heterocyclyl containing1 heteroatom selected from nitrogen, wherein the heterocyclyl is substituted with 0 or 1 occurrence of C 1-4 alkenyl)-***, -(5-6 membered monocyclic heterocyclyl containing1 heteroatom selected from nitrogen, wherein the heterocyclyl is substituted with 0 or 1 occurrence of C 1-4 alkyl)-***, -(4-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-(C 0-6 alkylene)-(4-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-***, or -(4-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-(C 0-6 alkylene)-(4-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-O-***, wherein *** is the point of attachment to A 2 . [0279] In certain embodiments, L is -(4-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-(C 0-6 alkylene)-(3-7 membered monocyclic saturated carbocyclic ring)-***, -(4-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-(C 0-6 alkylene)-(3-7 membered monocyclic saturated carbocyclic ring)-O- ***, -(4-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-O-(4-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-***, -(5-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-(C1-6 alkylene)-S(O)2- ***, -N(C1-4 alkyl)-(C1-6 alkylene)-O-***, -N(C1-4 alkyl)-(C1- 6 alkylene)-***, -(5-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-(C 0-6 alkylene)-(3-7 membered monocyclic saturated carbocyclic ring)-***, -(4-6 membered monocyclic heterocyclyl containing 1 heteroatom selected from nitrogen)-N(C1-4 alkyl)- ***, -(4-6 membered monocyclic heterocyclyl containing 1 heteroatom selected from nitrogen)- N(C 1-4 alkyl)-***, -(4-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-N(C 1-4 alkyl)-(4-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-***, -(4-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-N(C 3-6 cycloalkyl)-(4-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-***, or -(5-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-O-(C1-6 alkylene)-O-***, wherein *** is the point of attachment to A 2 . [0280] In certain embodiments, L is -(5-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-(C1-6 alkylene)-O-***, -(5-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-(C1-6 alkylene)-N(C1-4 alkyl)- (3-7 membered monocyclic saturated carbocyclic ring, wherein the carbocyclic ring is substituted with 0-4 occurrences of C 1-4 alkyl)-O-***, -(4-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-(C0-6 alkylene)-(7-11 membered spirocyclic heterocyclyl containing 1-2 heteroatoms selected from nitrogen)-O-***, -(4-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-(C 0-6 alkylene)-O-(5-6 membered heteroaryl containing 1 or 2 heteroatoms selected from nitrogen)-O-***, -(4-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-(C 0-6 alkylene)-(5-6 membered heteroaryl containing 1 or 2 heteroatoms selected from nitrogen)-O-***, -(4-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-O- (C0-6 alkylene)-(5-6 membered heteroaryl containing 1 or 2 heteroatoms selected from nitrogen)- O-***, -(4-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-O-(C 1-6 alkylene)-O-(5-6 membered heteroaryl containing 1 or 2 heteroatoms selected from nitrogen)-O-***, -(5-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-O-(3-7 membered monocyclic saturated carbocyclic ring)-O-***, -(5-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-(C1-6 alkylene)-N(C 1-4 alkyl)-(3-7 membered monocyclic saturated carbocyclic ring)-O-***, -(5-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-(C 1-6 alkylene)-N(C3-6 cycloalkyl)-(3-7 membered monocyclic saturated carbocyclic ring)-O-***, -(5-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-(C1-6 alkylene)-O-(3-7 membered monocyclic saturated carbocyclic ring)-O-***, or -(5-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-(C1-6 alkylene)-(7- 11 membered spirocyclic heterocyclyl containing 1-2 heteroatoms selected from nitrogen)-O-***, wherein *** is the point of attachment to A 2 . [0281] In certain embodiments, L is -(5-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-(C1-6 alkylene substituted with 0, 1, or 2 occurrences of C1-4 alkyl)-O-***, -(5-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-(C 1-6 alkylene substituted with 1 occurrence of a spirocyclic C 3-6 cycloalkyl)-O- ***, -(5-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-(C 1-6 alkylene substituted with 1 occurrence of C 3-6 cycloalkyl)-O-***, -(4-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-(C 0-6 alkylene)-O- (7-11 membered spirocyclic saturated carbocyclic ring)-O-***, -(4-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-(C0-6 alkylene)-N(C3-6 cycloalkyl)-(3-7 membered monocyclic saturated carbocyclic ring)-O-***, -(4-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-(C 0-6 alkylene)-(4- 6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-(C1-6 alkylene substituted with 1 occurrence of C 3-6 cycloalkyl)-O-***, -(4-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-(C 0-6 alkylene)-(3-7 membered monocyclic saturated carbocyclic ring)-(C1-6 alkylene substituted with 1 occurrence of C3-6 cycloalkyl)-O-***, -(7-11 membered spirocyclic heterocyclyl containing 1-2 heteroatoms selected from nitrogen)-(C 0-6 alkylene)-(3-7 membered monocyclic saturated carbocyclic ring)-O-***, -(4-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-(4-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-(C1-6 alkylene substituted with 1 occurrence of a spirocyclic C 3-6 cycloalkyl)-O-***, -(4-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-(C 0-6 alkylene)-(4- 6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-(C1-6 alkylene)-***, -(4-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-O-(3-7 membered monocyclic saturated carbocyclic ring)-(C1-6 alkylene substituted with 1 occurrence of C 3-6 cycloalkyl)-O-***, or -(4-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-(C 0-6 alkylene)-(4-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-(C1-6 haloalkyl)-***, wherein *** is the point of attachment to A 2 . [0282] In certain embodiments, L is -(4-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-(C0-6 alkylene)-(5-6 membered heteroaryl containing 1 or 2 heteroatoms selected from nitrogen)-(C1-6 haloalkyl)-***, -(4-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-(C 0-6 alkylene)-(5-6 membered heteroaryl containing 1 or 2 heteroatoms selected from nitrogen)-O-***, -(4-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-O-(C1-6 alkylene)- C(O)-NH-***, -(4-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-(C 0-6 alkylene)-C(O)-NH-***, -(4-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-(C0-6 alkylene)-(5-6 membered aryl substituted with 0 or 1 occurrence of fluoro)-O-***, -(4-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-(C 0-6 alkylene)-O-(5-6 membered aryl)-O-***, -(7-11 membered spirocyclic heterocyclyl containing 1-2 heteroatoms selected from nitrogen)-O-(3-7 membered monocyclic saturated carbocyclic ring)-O-***, -(7-11 membered spirocyclic heterocyclyl containing 1-2 heteroatoms selected from nitrogen)-(C 0-6 alkylene)-(3-7 membered monocyclic saturated carbocyclic ring)-O-***, or -(4-6 membered monocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-(C0-6 alkylene)-(pyrazinonyl)-***, wherein *** is the point of attachment to A 2 . [0283] In certain embodiments, L is selected from those depicted in the compounds in Table 1, below. Exemplary Specific Compounds [0284] In certain embodiments, the compound is a compound in Table 1, or a pharmaceutically acceptable salt thereof. In certain embodiments, the compound is a compound in Table 1. In certain embodiments, the compound is a compound selected from compounds I-1 through I-36 in Table 1. In certain embodiments, the compound is a compound selected from compounds I-1 through I-36 in Table 1, or a pharmaceutically acceptable salt thereof. In certain embodiments, the compound is a compound selected from compounds I-37 through I-253 in Table 1. In certain embodiments, the compound is a compound selected from compounds I-37 through I-253 in Table 1, or a pharmaceutically acceptable salt thereof. In certain embodiments, the compound is a compound selected from compounds I-1 through I-253 in Table 1, or a pharmaceutically acceptable salt thereof. In certain embodiments, the compound is a compound selected from compounds I-1 through I-253 in Table 1.

e r u t c . 1 u r t E S L B A T d n u o p . o 1 - I 2 - I 3 - m N I o C

4 2 1

5 2 1 8- I 9- 0 I 1 - I

6 2 1 1 1 2 3 - I 1 - I 1 - I

7 2 1 4 1 5 6 - I 1 - I 1 - I

8 2 1 7 1 8 9 - I 1 - I 1 - I

0 2 1 2 2 2 3- I - I - I 2 - I

0 3 1 4 2 5 6 - I 2 - I 2 - I

7 2 8 2 9 2 0- I - I - I 3 - I

N l C 1 3 2 3 3 4 - I - I 3 - I 3 - I

O 1 N l C 5 3 6 - 7 8 I 3 - I 3 - I 3 - I

9 3 0 4 1 4 2 - I - I - I 4 - I

O S H N O N O F O N N N O N H O l C N 3 4 4 4 5 4 6 - I - I - I 4 - I

1 7 4 8 4 9 4 0 - I - I - I 5 - I

N l C 1 5 2 5 3 4 - I - I 5 - I 5 - I

N l C 5 5 6 5 7 8 - I - I 5 - I 5 - I

9 5 0 6 1 6 2 - I - I - I 6 - I

3 6 4 6 5 6 6 - I - I - I 6 - I

7 6 8 6 9 6 0 - I - I - I 7 - I

1 7 2 7 3 7 4 - I - I - I 7 - I

3 4 1 5 7 6 7 - I 7 - I 7 - I

O H N O N O F N N N N O N H O l C N 8 7 9 7 0 8 1 - I - I - I 8 - I

2 8 3 8 4 8 5 - I - I - I 8 - I

6 8 7 8 8 8 9 - I - I - I 8 - I

7 4 1 0 9 1 2 - I 9 - I 9 - I

8 4 1 3 9 4 5 - I 9 - I 9 - I

6 9 7 9 8 9 9 - I - I - I 9 - I

H O N O N N N N O H N O l C l N C N 0 0 1 2 3 1 0 1 0 0 - I - I 1 - I 1 - I

l C N 4 0 5 1 0 6 0 7 0 - I 1 - I 1 - I 1 - I

8 0 9 0 1 1 0 1 1 - I 1 - I 1 - I 1 - I

2 1 3 4 5 1 1 1 1 - I 1 - I 1 - I 1 - I

6 1 7 8 9 1 1 1 1 - I 1 - I 1 - I 1 - I

0 2 1 2 3 1 2 2 2 - I 1 - I 1 - I 1 - I

4 2 5 6 7 1 2 2 2 - I 1 - I 1 - I 1 - I

8 2 9 0 1 1 2 3 3 - I 1 - I 1 - I 1 - I

2 3 3 4 5 1 3 3 3 - I 1 - I 1 - I 1 - I

l C N 6 3 7 1 3 8 3 9 3 - I 1 - I 1 - I 1 - I

0 6 1 0 4 1 4 2 1 I 1 4 - - I 1 - I

1 6 1 3 4 4 4 5 1 I 1 4 - - I 1 - I

6 4 7 8 9 1 4 4 4 - I 1 - I 1 - I 1 - I

6 1 0 5 1 1 5 2 5 3 5 - I 1 - I 1 - I 1 - I

6 1 4 5 5 1 5 6 5 7 5 - I 1 - I 1 - I 1 - I

1 l C N 8 5 9 5 0 6 1 1 I 1 - I 1 6 - - I 1 - I

2 6 3 4 5 1 6 6 6 - I 1 - I 1 - I 1 - I

l C N N l C 6 6 7 6 8 9 1 6 6 - I 1 - I 1 - I 1 - I

0 7 1 2 3 1 7 7 7 - I 1 - I 1 - I 1 - I

9 6 1 4 7 5 7 6 1 I 1 7 - - I 1 - I

0 7 1 N l C 7 7 8 7 9 0 1 7 8 - I 1 - I 1 - I 1 - I

1 8 2 3 4 1 8 8 8 - I 1 - I 1 - I 1 - I

H N O N O N N N N H N O O N l C 5 8 6 7 8 1 8 1 8 8 - I - I 1 - I 1 - I

N l C l C N 9 8 0 9 1 2 1 9 9 - I 1 - I 1 - I 1 - I

N l C 3 9 4 1 9 5 9 6 9 - I 1 - I 1 - I 1 - I

l C N 7 9 8 1 9 9 9 0 0 - I 1 - I 1 - I 2 - I

H N O N O N N N N O N H O l C l C N N 1 0 2 3 4 2 0 2 0 0 - I - I 2 - I 2 - I

N l C 5 0 6 2 0 7 0 8 0 - I 2 - I 2 - I 2 - I

N l C 9 0 0 2 1 1 1 2 1 - I 2 - I 2 - I 2 - I

9 7 1 3 1 4 1 5 2 I 2 1 - - I 2 - I

O 0 8 1 l C N 6 1 7 8 2 1 - I 2 1 - I 2 - I

9 1 0 1 2 2 2 2 2 - I 2 - I 2 - I 2 - I

l C N 3 2 4 2 2 5 2 6 2 - I 2 - I 2 - I 2 - I

O 3 8 1 l C N 7 2 8 2 9 0 2 2 3 - I 2 - I 2 - I 2 - I

O 4 8 1 N l C 1 3 2 3 3 4 2 3 3 - I 2 - I 2 - I 2 - I

5 8 1 5 3 6 3 7 2 I 2 3 - - I 2 - I

8 3 9 0 1 2 3 4 4 - I 2 - I 2 - I 2 - I

2 4 3 4 5 2 4 4 4 - I 2 - I 2 - I 2 - I

8 8 1 6 4 7 4 8 2 I 2 4 - - I 2 - I

9 8 1 9 4 0 5 1 2 I 2 5 - - I 2 - I

0 9 1 2 5 3 5 4 2 I 2 5 - - I 2 - I

1 9 1 5 5 2 - I Synthetic Methods [0285] Methods for preparing compounds described herein are illustrated in the following synthetic Schemes. The Schemes are given for the purpose of illustrating the invention, and are not intended to limit the scope or spirit of the invention. Starting materials shown in the Schemes can be obtained from commercial sources or can be prepared based on procedures described in the literature. [0286] In the Schemes, it is understood by one skilled in the art of organic synthesis that the functionality present on various portions of the molecule should be compatible with the reagents and reactions proposed. Substituents not compatible with the reaction conditions will be apparent to one skilled in the art, and alternate methods are therefore indicated (for example, use of protecting groups or alternative reactions). Protecting group chemistry and strategy is well known, such as described in, for example, “Protecting Groups in Organic Synthesis”, T. W. Greene and P. G. M. Wuts, 3 rd edition, John Wiley & Sons, 1999, the entire contents of which are hereby incorporated by reference. [0287] The synthetic route illustrated in Scheme 1 is a general method for preparing compounds of Formula H. Coupling fluoro-nitrobenzene compound A with substituted phenol B provides phenyl ether compound C. Reduction of the nitro group in compound C provides substituted aniline D. Reaction of substituted aniline D with ClSO2R i provides sulfonamide E. Removal of the protection group (Pg) from compound E provides compound F. The Pg may be, for example, a Boc protecting group that can be removed by treating the compound with trifluoroacetic acid. Coupling compound F with compound G (such as a nucleophilic aromatic substitution reaction when X is an amino group and the leaving group in compound E is chloro) provides the final compound of Formula H.

SCHEME 1. [0288] The modular synthetic route illustrated in Scheme 1 can be readily modified to provide additional compounds by conducting functional group transformations on the intermediate and/or final compounds. Such functional group transformations are well known in the art, as described in, for example, Comprehensive Organic Synthesis (B.M. Trost & I. Fleming, eds., 1991-1992); Organic Synthesis, 3 rd Ed. (Michael B. Smith, Wavefunction, Inc., Irvine: 2010); Modern Methods of Organic Synthesis, 4 th Ed. (William Carruthers and Iain Coldham, Cambridge University Press, Cambridge: 2004); March’s Advanced Organic Chemistry: Reactions, Mechanisms, and Structure, 8 th Ed., (Michael B. Smith, John Wiley & Sons, New York: 2020); and Comprehensive Organic Transformations: A Guide to Functional Group Preparations, 3rd Ed. (Richard C. Larock, ed., John Wiley & Sons, New York: 2018). Protecting group strategies may be deployed as appropriate to accommodate differing functional groups in the molecules used in the synthetic route. Protecting group chemistry and strategy is described in, for example, Protecting Groups in Organic Synthesis, 3 rd Edition, T. W. Greene and P. G. M. Wuts, John Wiley & Sons, 1999 and Greene's Protective Groups in Organic Synthesis, 5th Ed., (Peter G. M. Wuts, John Wiley & Sons: 2014). II. Therapeutic Applications [0289] The heterobifunctional compounds described herein, such as a compound of Formula I, II, or other compounds in Section I, provide therapeutic benefits to patients suffering from cancer. Accordingly, one aspect of the invention provides a method of treating cancer. The method comprises administering to a patient in need thereof a therapeutically effective amount of a compound described herein, such as a compound of Formula I, II, or other compound in Section I, to treat the cancer. In certain embodiments, the compound is a compound of Formula I. In certain embodiments, the particular compound of Formula I is a compound defined by one of the embodiments described above. Cancer [0290] In certain embodiments, the cancer is ovarian cancer, uterine cancer, endometrial cancer, cervical cancer, prostate cancer, testicular cancer, breast cancer, brain cancer, lung cancer, oral cancer, esophageal cancer, head and neck cancer, stomach cancer, colon cancer, rectal cancer, skin cancer, sebaceous gland carcinoma, bile duct and gallbladder cancers, liver cancer, pancreatic cancer, bladder cancer, urinary tract cancer, kidney cancer, eye cancer, thyroid cancer, lymphoma, or leukemia. In certain embodiments, the cancer is prostate cancer. [0291] In certain embodiments, the cancer is squamous cell cancer, lung cancer including small cell lung cancer, non-small cell lung cancer, vulval cancer, thyroid cancer, adenocarcinoma of the lung and squamous carcinoma of the lung, cancer of the peritoneum, hepatocellular cancer, gastric or stomach cancer including gastrointestinal cancer, pancreatic cancer, glioblastoma, cervical cancer, ovarian cancer, liver cancer, bladder cancer, hepatoma, breast cancer, colon cancer, rectal cancer, colorectal cancer, endometrial or uterine carcinoma, salivary gland carcinoma, kidney or renal cancer, prostate cancer, hepatic carcinoma, anal carcinoma, penile carcinoma, and head and neck cancer. In certain embodiments, the cancer is at least one selected from the group consisting of ALL, T-lineage Acute lymphoblastic Leukemia (T-ALL), T-lineage lymphoblastic Lymphoma (T-LL), Peripheral T-cell lymphoma, Adult T-cell Leukemia, Pre-B ALL, Pre-B Lymphomas, Large B-cell Lymphoma, Burkitts Lymphoma, B-cell ALL, Philadelphia chromosome positive ALL, Philadelphia chromosome positive CML, lymphoma, leukemia, multiple myeloma myeloproliferative diseases, large B cell lymphoma, or B cell Lymphoma. [0292] In certain embodiments, the cancer is a solid tumor or leukemia. In certain other embodiments, the cancer is colon cancer, pancreatic cancer, breast cancer, ovarian cancer, prostate cancer, squamous cell carcinoma, basal cell carcinoma, adenocarcinoma, sweat gland carcinoma, sebaceous gland carcinoma, lung cancer, leukemia, bladder cancer, stomach cancer, cervical cancer, testicular cancer, skin cancer, rectal cancer, thyroid cancer, kidney cancer, uterus cancer, espophagus cancer, liver cancer, an acoustic neuroma, oligodendroglioma, meningioma, neuroblastoma, or retinoblastoma. In certain other embodiments, the cancer is small cell lung cancer, non-small cell lung cancer, melanoma, cancer of the central nervous system tissue, brain cancer, Hodgkin’s lymphoma, non-Hodgkin’s lymphoma, cutaneous T-Cell lymphoma, cutaneous B-Cell lymphoma, or diffuse large B-Cell lymphoma. In certain other embodiments, the cancer is breast cancer, colon cancer, small-cell lung cancer, non-small cell lung cancer, prostate cancer, renal cancer, ovarian cancer, leukemia, melanoma, or cancer of the central nervous system tissue. In certain other embodiments, the cancer is colon cancer, small-cell lung cancer, non-small cell lung cancer, renal cancer, ovarian cancer, renal cancer, or melanoma. [0293] In certain embodiments, the cancer is a fibrosarcoma, myxosarcoma, liposarcoma, chondrosarcoma, osteogenic sarcoma, chordoma, angiosarcoma, endotheliosarcoma, lymphangiosarcoma, lymphangioendotheliosarcoma, Ewing’s tumor, leiomyosarcoma, rhabdomyosarcoma, squamous cell carcinoma, basal cell carcinoma, adenocarcinoma, sweat gland carcinoma, sebaceous gland carcinoma, papillary carcinoma, papillary adenocarcinomas, cystadenocarcinoma, medullary carcinoma, bronchogenic carcinoma, renal cell carcinoma, hepatoma, bile duct carcinoma, choriocarcinoma, seminoma, embryonal carcinoma, Wilms’ tumor, epithelial carcinoma, glioma, astrocytoma, medulloblastoma, or hemangioblastoma. [0294] In certain embodiments, the cancer is a neuroblastoma, meningioma, hemangiopericytoma, multiple brain metastase, glioblastoma multiforms, glioblastoma, brain stem glioma, poor prognosis malignant brain tumor, malignant glioma, anaplastic astrocytoma, anaplastic oligodendroglioma, neuroendocrine tumor, rectal adeno carcinoma, Dukes C & D colorectal cancer, unresectable colorectal carcinoma, metastatic hepatocellular carcinoma, Kaposi’s sarcoma, karotype acute myeloblastic leukemia, Hodgkin’s lymphoma, non-Hodgkin’s lymphoma, cutaneous T-Cell lymphoma, cutaneous B-Cell lymphoma, diffuse large B-Cell lymphoma, low grade follicular lymphoma, metastatic melanoma, localized melanoma, malignant mesothelioma, malignant pleural effusion mesothelioma syndrome, peritoneal carcinoma, papillary serous carcinoma, gynecologic sarcoma, soft tissue sarcoma, scelroderma, cutaneous vasculitis, Langerhans cell histiocytosis, leiomyosarcoma, fibrodysplasia ossificans progressive, hormone refractory prostate cancer, resected high-risk soft tissue sarcoma, unrescectable hepatocellular carcinoma, Waidenstrom’s macroglobulinemia, smoldering myeloma, indolent myeloma, fallopian tube cancer, androgen independent prostate cancer, androgen dependent stage IV non-metastatic prostate cancer, hormone-insensitive prostate cancer, chemotherapy-insensitive prostate cancer, castrate resistant prostate cancer, castrate resistant metastatic prostate cancer, papillary thyroid carcinoma, follicular thyroid carcinoma, medullary thyroid carcinoma, or leiomyoma. [0295] In certain embodiments, the cancer is bone cancer, pancreatic cancer, skin cancer, cancer of the head or neck, cutaneous or intraocular melanoma, ovarian cancer, colon cancer, rectal cancer, cancer of the anal region, stomach cancer, gastrointestinal (gastric, colorectal, and duodenal), uterine cancer, carcinoma of the fallopian tubes, carcinoma of the endometrium, carcinoma of the cervix, carcinoma of the vagina, carcinoma of the vulva, Hodgkin's Disease, cancer of the esophagus, cancer of the small intestine, cancer of the endocrine system, cancer of the thyroid gland, cancer of the parathyroid gland, cancer of the adrenal gland, sarcoma of soft tissue, cancer of the urethra, cancer of the penis, prostate cancer, testicular cancer, chronic or acute leukemia, chronic myeloid leukemia, lymphocytic lymphomas, cancer of the bladder, cancer of the kidney or ureter, renal cell carcinoma, carcinoma of the renal pelvis, non-Hodgkins's lymphoma, spinal axis tumors, brain stem glioma, pituitary adenoma, adrenocortical cancer, gall bladder cancer, multiple myeloma, cholangiocarcinoma, fibrosarcoma, neuroblastoma, retinoblastoma, or a combination of one or more of the foregoing cancers. [0296] In certain embodiments, the cancer is hepatocellular carcinoma, ovarian cancer, ovarian epithelial cancer, or fallopian tube cancer; papillary serous cystadenocarcinoma or uterine papillary serous carcinoma (UPSC); prostate cancer; testicular cancer; gallbladder cancer; hepatocholangiocarcinoma; soft tissue and bone synovial sarcoma; rhabdomyosarcoma; osteosarcoma; chondrosarcoma; Ewing sarcoma; anaplastic thyroid cancer; adrenocortical adenoma; pancreatic cancer; pancreatic ductal carcinoma or pancreatic adenocarcinoma; gastrointestinal/stomach (GIST) cancer; lymphoma; squamous cell carcinoma of the head and neck (SCCHN); salivary gland cancer; glioma, or brain cancer; neurofibromatosis-1 associated malignant peripheral nerve sheath tumors (MPNST); Waldenstrom's macroglobulinemia; or medulloblastoma. [0297] In certain embodiments, the cancer is hepatocellular carcinoma (HCC), hepatoblastoma, colon cancer, rectal cancer, ovarian cancer, ovarian epithelial cancer, fallopian tube cancer, papillary serous cystadenocarcinoma, uterine papillary serous carcinoma (UPSC), hepatocholangiocarcinoma, soft tissue and bone synovial sarcoma, rhabdomyosarcoma, osteosarcoma, anaplastic thyroid cancer, adrenocortical adenoma, pancreatic cancer, pancreatic ductal carcinoma, pancreatic adenocarcinoma, glioma, neurofibromatosis-1 associated malignant peripheral nerve sheath tumors (MPNST), Waldenstrom's macroglobulinemia, or medulloblastoma. [0298] In certain embodiments, the cancer is a solid tumor, such as a sarcoma, carcinoma, or lymphoma. In certain embodiments, the cancer is kidney cancer; hepatocellular carcinoma (HCC) or hepatoblastoma, or liver cancer; melanoma; breast cancer; colorectal carcinoma, or colorectal cancer; colon cancer; rectal cancer; anal cancer; lung cancer, such as non-small cell lung cancer (NSCLC) or small cell lung cancer (SCLC); ovarian cancer, ovarian epithelial cancer, ovarian carcinoma, or fallopian tube cancer; papillary serous cystadenocarcinoma or uterine papillary serous carcinoma (UPSC); prostate cancer; testicular cancer; gallbladder cancer; hepatocholangiocarcinoma; soft tissue and bone synovial sarcoma; rhabdomyosarcoma; osteosarcoma; chondrosarcoma; Ewing sarcoma; anaplastic thyroid cancer; adrenocortical carcinoma; pancreatic cancer; pancreatic ductal carcinoma or pancreatic adenocarcinoma; gastrointestinal/stomach (GIST) cancer; lymphoma; squamous cell carcinoma of the head and neck (SCCHN); salivary gland cancer; glioma, or brain cancer; neurofibromatosis-1 associated malignant peripheral nerve sheath tumors (MPNST); Waldenstrom's macroglobulinemia; or medulloblastoma. [0299] In certain embodiments, the cancer is renal cell carcinoma, hepatocellular carcinoma (HCC), hepatoblastoma, colorectal carcinoma, colorectal cancer, colon cancer, rectal cancer, anal cancer, ovarian cancer, ovarian epithelial cancer, ovarian carcinoma, fallopian tube cancer, papillary serous cystadenocarcinoma, uterine papillary serous carcinoma (UPSC), hepatocholangiocarcinoma, soft tissue and bone synovial sarcoma, rhabdomyosarcoma, osteosarcoma, chondrosarcoma, anaplastic thyroid cancer, adrenocortical carcinoma, pancreatic cancer, pancreatic ductal carcinoma, pancreatic adenocarcinoma, glioma, brain cancer, neurofibromatosis-1 associated malignant peripheral nerve sheath tumors (MPNST), Waldenstrom's macroglobulinemia, or medulloblastoma. [0300] In certain embodiments, the cancer is hepatocellular carcinoma (HCC), hepatoblastoma, colon cancer, rectal cancer, ovarian cancer, ovarian epithelial cancer, ovarian carcinoma, fallopian tube cancer, papillary serous cystadenocarcinoma, uterine papillary serous carcinoma (UPSC), hepatocholangiocarcinoma, soft tissue and bone synovial sarcoma, rhabdomyosarcoma, osteosarcoma, anaplastic thyroid cancer, adrenocortical carcinoma, pancreatic cancer, pancreatic ductal carcinoma, pancreatic adenocarcinoma, glioma, neurofibromatosis-1 associated malignant peripheral nerve sheath tumors (MPNST), Waldenstrom's macroglobulinemia, or medulloblastoma. [0301] In certain embodiments, the cancer is hepatocellular carcinoma (HCC). In some embodiments, the cancer is hepatoblastoma. In some embodiments, the cancer is colon cancer. In some embodiments, the cancer is rectal cancer. In some embodiments, the cancer is ovarian cancer, or ovarian carcinoma. In some embodiments, the cancer is ovarian epithelial cancer. In some embodiments, the cancer is fallopian tube cancer. In some embodiments, the cancer is papillary serous cystadenocarcinoma. In some embodiments, the cancer is uterine papillary serous carcinoma (UPSC). In some embodiments, the cancer is hepatocholangiocarcinoma. In some embodiments, the cancer is soft tissue and bone synovial sarcoma. In some embodiments, the cancer is rhabdomyosarcoma. In some embodiments, the cancer is osteosarcoma. In some embodiments, the cancer is anaplastic thyroid cancer. In some embodiments, the cancer is adrenocortical carcinoma. In some embodiments, the cancer is pancreatic cancer, or pancreatic ductal carcinoma. In some embodiments, the cancer is pancreatic adenocarcinoma. In some embodiments, the cancer is glioma. In some embodiments, the cancer is malignant peripheral nerve sheath tumors (MPNST). In some embodiments, the cancer is neurofibromatosis-1 associated MPNST. In some embodiments, the cancer is Waldenstrom's macroglobulinemia. In some embodiments, the cancer is medulloblastoma. Causing Death of Cancer Cell [0302] Another aspect of the invention provides a method of causing death of a cancer cell. The method comprises contacting a cancer cell with an effective amount of a compound described herein, such as a compound of Formula I or II, or other compound in Section I, to cause death of the cancer cell. In certain embodiments, the particular compound of Formula I or II is a compound defined by one of the embodiments described above. [0303] In certain embodiments, the cancer cell is selected from ovarian cancer, uterine cancer, endometrial cancer, cervical cancer, prostate cancer, testicular cancer, breast cancer, brain cancer, lung cancer, oral cancer, esophageal cancer, head and neck cancer, stomach cancer, colon cancer, rectal cancer, skin cancer, sebaceous gland carcinoma, bile duct and gallbladder cancers, liver cancer, pancreatic cancer, bladder cancer, urinary tract cancer, kidney cancer, eye cancer, thyroid cancer, lymphoma, or leukemia. In certain embodiments, the cancer cell is one or more of the cancers recited in the section above entitled “Cancer.” In certain embodiments, the cancer cell is a prostate cancer cell. Combination Therapies [0304] The compounds useful within the methods of the invention may be used in combination with one or more additional therapeutic agents useful for treating any disease contemplated herein. These additional therapeutic agents may comprise compounds that are commercially available or synthetically accessible to those skilled in the art. These additional therapeutic agents are known to treat, prevent, or reduce the symptoms, of a disease or disorder contemplated herein. [0305] Accordingly, in certain embodiments, the method further comprises administering to the subject an additional therapeutic agent that treats the disease contemplated herein. [0306] In certain embodiments, administering the compound of the invention to the subject allows for administering a lower dose of the additional therapeutic agent as compared to the dose of the additional therapeutic agent alone that is required to achieve similar results in treating the disease contemplated herein. For example, in certain embodiments, the compound of the invention enhances the therapeutic activity of the additional therapeutic compound, thereby allowing for a lower dose of the additional therapeutic compound to provide the same effect. [0307] A synergistic effect may be calculated, for example, using suitable methods such as, for example, the Sigmoid-E max equation (Holford & Scheiner, 1981, Clin. Pharmacokinet.6:429-453), the equation of Loewe additivity (Loewe & Muischnek, 1926, Arch. Exp. Pathol Pharmacol. 114:313-326) and the median-effect equation (Chou & Talalay, 1984, Adv. Enzyme Regul.22:27- 55). Each equation referred to above may be applied to experimental data to generate a corresponding graph to aid in assessing the effects of the drug combination. The corresponding graphs associated with the equations referred to above are the concentration-effect curve, isobologram curve and combination index curve, respectively. [0308] In certain embodiments, the compound of the invention and the therapeutic agent are co- administered to the subject. In other embodiments, the compound of the invention and the therapeutic agent are coformulated and co-administered to the subject. [0309] In certain embodiments, the compound is administered in combination with a second therapeutic agent having activity against cancer. In certain embodiments, the second therapeutic agent is mitomycin, tretinoin, ribomustin, gemcitabine, vincristine, etoposide, cladribine, mitobronitol, methotrexate, doxorubicin, carboquone, pentostatin, nitracrine, zinostatin, cetrorelix, letrozole, raltitrexed, daunorubicin, fadrozole, fotemustine, thymalfasin, sobuzoxane, nedaplatin, cytarabine, bicalutamide, vinorelbine, vesnarinone, aminoglutethimide, amsacrine, proglumide, elliptinium acetate, ketanserin, doxifluridine, etretinate, isotretinoin, streptozocin, nimustine, vindesine, flutamide, drogenil, butocin, carmofur, razoxane, sizofilan, carboplatin, mitolactol, tegafur, ifosfamide, prednimustine, picibanil, levamisole, teniposide, improsulfan, enocitabine, lisuride, oxymetholone, tamoxifen, progesterone, mepitiostane, epitiostanol, formestane, interferon-alpha, interferon-2 alpha, interferon-beta, interferon-gamma, colony stimulating factor- 1, colony stimulating factor-2, denileukin diftitox, interleukin-2, and leutinizing hormone releasing factor. [0310] In certain embodiments, the second therapeutic agent is an mTOR inhibitor, which inhibits cell proliferation, angiogenesis and glucose uptake. Approved mTOR inhibitors useful in the present invention include everolimus (Afinitor®, Novartis); temsirolimus (Torisel®, Pfizer); and sirolimus (Rapamune®, Pfizer). [0311] In certain embodiments, the second therapeutic agent is a Poly ADP ribose polymerase (PARP) inhibitor. Approved PARP inhibitors useful in the present invention include olaparib (Lynparza®, AstraZeneca); rucaparib (Rubraca®, Clovis Oncology); and niraparib (Zejula®, Tesaro). Other PARP inhibitors being studied which may be used in the present invention include talazoparib (MDV3800/BMN 673/LT00673, Medivation/Pfizer/Biomarin); veliparib (ABT-888, AbbVie); and BGB-290 (BeiGene, Inc.). [0312] In certain embodiments, the second therapeutic agent is a phosphatidylinositol 3 kinase (PI3K) inhibitor. Approved PI3K inhibitors useful in the present invention include idelalisib (Zydelig®, Gilead). Other PI3K inhibitors being studied which may be used in the present invention include alpelisib (BYL719, Novartis); taselisib (GDC-0032, Genentech/Roche); pictilisib (GDC-0941, Genentech/Roche); copanlisib (BAY806946, Bayer); duvelisib (formerly IPI-145, Infinity Pharmaceuticals); PQR309 (Piqur Therapeutics, Switzerland); and TGR1202 (formerly RP5230, TG Therapeutics). [0313] In certain embodiments, the second therapeutic agent is a proteasome inhibitor. Approved proteasome inhibitors useful in the present invention include bortezomib (Velcade®, Takeda); carfilzomib (Kyprolis®, Amgen); and ixazomib (Ninlaro®, Takeda). [0314] In certain embodiments, the second therapeutic agent is a histone deacetylase (HDAC) inhibitor. Approved HDAC inhibitors useful in the present invention include vorinostat (Zolinza®, Merck); romidepsin (Istodax®, Celgene); panobinostat (Farydak®, Novartis); and belinostat (Beleodaq®, Spectrum Pharmaceuticals). Other HDAC inhibitors being studied which may be used in the present invention include entinostat (SNDX-275, Syndax Pharmaceuticals) (NCT00866333); and chidamide (Epidaza®, HBI-8000, Chipscreen Biosciences, China). [0315] In certain embodiments, the second therapeutic agent is a CDK inhibitor, such as a CDK 4/6 inhibitor. Approved CDK 4/6 inhibitors useful in the present invention include palbociclib (Ibrance®, Pfizer); and ribociclib (Kisqali®, Novartis). Other CDK 4/6 inhibitors being studied which may be used in the present invention include abemaciclib (Ly2835219, Eli Lilly); and trilaciclib (G1T28, G1 Therapeutics). [0316] In certain embodiments, the second therapeutic agent is an indoleamine (2,3)- dioxygenase (IDO) inhibitor. IDO inhibitors being studied which may be used in the present invention include epacadostat (INCB024360, Incyte); indoximod (NLG-8189, NewLink Genetics Corporation); capmanitib (INC280, Novartis); GDC-0919 (Genentech/Roche); PF-06840003 (Pfizer); BMS:F001287 (Bristol-Myers Squibb); Phy906/KD108 (Phytoceutica); and an enzyme that breaks down kynurenine (Kynase, Kyn Therapeutics). [0317] In certain embodiments, the second therapeutic agent is a growth factor antagonist, such as an antagonist of platelet-derived growth factor (PDGF), or epidermal growth factor (EGF) or its receptor (EGFR). Approved PDGF antagonists which may be used in the present invention include olaratumab (Lartruvo®; Eli Lilly). Approved EGFR antagonists which may be used in the present invention include cetuximab (Erbitux®, Eli Lilly); necitumumab (Portrazza®, Eli Lilly), panitumumab (Vectibix®, Amgen); and osimertinib (targeting activated EGFR, Tagrisso®, AstraZeneca). [0318] In certain embodiments, the second therapeutic agent is an aromatase inhibitor. Approved aromatase inhibitors which may be used in the present invention include exemestane (Aromasin®, Pfizer); anastazole (Arimidex®, AstraZeneca) and letrozole (Femara®, Novartis). [0319] In certain embodiments, the second therapeutic agent is an antagonist of the hedgehog pathway. Approved hedgehog pathway inhibitors which may be used in the present invention include sonidegib (Odomzo®, Sun Pharmaceuticals); and vismodegib (Erivedge®, Genentech), both for treatment of basal cell carcinoma. [0320] In certain embodiments, the second therapeutic agent is a folic acid inhibitor. Approved folic acid inhibitors useful in the present invention include pemetrexed (Alimta®, Eli Lilly). [0321] In certain embodiments, the second therapeutic agent is a CC chemokine receptor 4 (CCR4) inhibitor. CCR4 inhibitors being studied that may be useful in the present invention include mogamulizumab (Poteligeo®, Kyowa Hakko Kirin, Japan). [0322] In certain embodiments, the second therapeutic agent is an isocitrate dehydrogenase (IDH) inhibitor. IDH inhibitors being studied which may be used in the present invention include AG120 (Celgene; NCT02677922); AG221 (Celgene, NCT02677922; NCT02577406); BAY1436032 (Bayer, NCT02746081); IDH305 (Novartis, NCT02987010). [0323] In certain embodiments, the second therapeutic agent is an arginase inhibitor. Arginase inhibitors being studied which may be used in the present invention include AEB1102 (pegylated recombinant arginase, Aeglea Biotherapeutics), which is being studied in Phase 1 clinical trials for acute myeloid leukemia and myelodysplastic syndrome (NCT02732184) and solid tumors (NCT02561234); and CB-1158 (Calithera Biosciences). [0324] In certain embodiments, the second therapeutic agent is a glutaminase inhibitor. Glutaminase inhibitors being studied which may be used in the present invention include CB-839 (Calithera Biosciences). [0325] In certain embodiments, the second therapeutic agent is an antibody that binds to tumor antigens, that is, proteins expressed on the cell surface of tumor cells. Approved antibodies that bind to tumor antigens which may be used in the present invention include rituximab (Rituxan®, Genentech/BiogenIdec); ofatumumab (anti-CD20, Arzerra®, GlaxoSmithKline); obinutuzumab (anti-CD20, Gazyva®, Genentech), ibritumomab (anti-CD20 and Yttrium-90, Zevalin®, Spectrum Pharmaceuticals); daratumumab (anti-CD38, Darzalex®, Janssen Biotech), dinutuximab (anti- glycolipid GD2, Unituxin®, United Therapeutics); trastuzumab (anti-HER2, Herceptin®, Genentech); ado-trastuzumab emtansine (anti-HER2, fused to emtansine, Kadcyla®, Genentech); and pertuzumab (anti-HER2, Perjeta®, Genentech); and brentuximab vedotin (anti-CD30-drug conjugate, Adcetris®, Seattle Genetics). [0326] In certain embodiments, the second therapeutic agent is a topoisomerase inhibitor. Approved topoisomerase inhibitors useful in the present invention include irinotecan (Onivyde®, Merrimack Pharmaceuticals); topotecan (Hycamtin®, GlaxoSmithKline). Topoisomerase inhibitors being studied which may be used in the present invention include pixantrone (Pixuvri®, CTI Biopharma). [0327] In certain embodiments, the second therapeutic agent is a nucleoside inhibitor, or other therapeutic that interfere with normal DNA synthesis, protein synthesis, cell replication, or will otherwise inhibit rapidly proliferating cells. Such nucleoside inhibitors or other therapeutics include trabectedin (guanidine alkylating agent, Yondelis®, Janssen Oncology), mechlorethamine (alkylating agent, Valchlor®, Aktelion Pharmaceuticals); vincristine (Oncovin®, Eli Lilly; Vincasar®, Teva Pharmaceuticals; Marqibo®, Talon Therapeutics); temozolomide (prodrug to alkylating agent 5-(3-methyltriazen-1-yl)-imidazole-4-carboxamide (MTIC) Temodar®, Merck); cytarabine injection (ara-C, antimetabolic cytidine analog, Pfizer); lomustine (alkylating agent, CeeNU®, Bristol-Myers Squibb; Gleostine®, NextSource Biotechnology); azacitidine (pyrimidine nucleoside analog of cytidine, Vidaza®, Celgene); omacetaxine mepesuccinate (cephalotaxine ester) (protein synthesis inhibitor, Synribo®; Teva Pharmaceuticals); asparaginase Erwinia chrysanthemi (enzyme for depletion of asparagine, Elspar®, Lundbeck; Erwinaze®, EUSA Pharma); eribulin mesylate (microtubule inhibitor, tubulin-based antimitotic, Halaven®, Eisai); cabazitaxel (microtubule inhibitor, tubulin-based antimitotic, Jevtana®, Sanofi-Aventis); capacetrine (thymidylate synthase inhibitor, Xeloda®, Genentech); bendamustine (bifunctional mechlorethamine derivative, believed to form interstrand DNA cross-links, Treanda®, Cephalon/Teva); ixabepilone (semi-synthetic analog of epothilone B, microtubule inhibitor, tubulin-based antimitotic, Ixempra®, Bristol-Myers Squibb); nelarabine (prodrug of deoxyguanosine analog, nucleoside metabolic inhibitor, Arranon®, Novartis); clorafabine (prodrug of ribonucleotide reductase inhibitor, competitive inhibitor of deoxycytidine, Clolar®, Sanofi- Aventis); and trifluridine and tipiracil (thymidine-based nucleoside analog and thymidine phosphorylase inhibitor, Lonsurf®, Taiho Oncology). [0328] In certain embodiments, the second therapeutic agent is a platinum-based therapeutic, also referred to as platins. Platins cause cross-linking of DNA, such that they inhibit DNA repair and/or DNA synthesis, mostly in rapidly reproducing cells, such as cancer cells. Approved platinum-based therapeutics which may be used in the present invention include cisplatin (Platinol®, Bristol-Myers Squibb); carboplatin (Paraplatin®, Bristol-Myers Squibb; also, Teva; Pfizer); oxaliplatin (Eloxitin® Sanofi-Aventis); and nedaplatin (Aqupla®, Shionogi). Other platinum-based therapeutics which have undergone clinical testing and may be used in the present invention include picoplatin (Poniard Pharmaceuticals); and satraplatin (JM-216, Agennix). [0329] In certain embodiments, the second therapeutic agent is a taxane compound, which causes disruption of microtubules, which are essential for cell division. Approved taxane compounds which may be used in the present invention include paclitaxel (Taxol®, Bristol-Myers Squibb), docetaxel (Taxotere®, Sanofi-Aventis; Docefrez®, Sun Pharmaceutical), albumin-bound paclitaxel (Abraxane®; Abraxis/Celgene), and cabazitaxel (Jevtana®, Sanofi-Aventis). Other taxane compounds which have undergone clinical testing and may be used in the present invention include SID530 (SK Chemicals, Co.) (NCT00931008). [0330] In certain embodiments, the second therapeutic agent is an inhibitor of anti-apoptotic proteins, such as BCL-2. Approved anti-apoptotics which may be used in the present invention include venetoclax (Venclexta®, AbbVie/Genentech); and blinatumomab (Blincyto®, Amgen). Other therapeutic agents targeting apoptotic proteins which have undergone clinical testing and may be used in the present invention include navitoclax (ABT-263, Abbott), a BCL-2 inhibitor (NCT02079740). [0331] In certain embodiments, the second therapeutic agent is a selective estrogen receptor modulator (SERM), which interferes with the synthesis or activity of estrogens. Approved SERMs useful in the present invention include raloxifene (Evista®, Eli Lilly). [0332] In certain embodiments, the second therapeutic agent is an inhibitor of interaction between the two primary p53 suppressor proteins, MDMX and MDM2. Inhibitors of p53 suppression proteins being studied which may be used in the present invention include ALRN- 6924 (Aileron), a stapled peptide that equipotently binds to and disrupts the interaction of MDMX and MDM2 with p53. ALRN-6924 is currently being evaluated in clinical trials for the treatment of AML, advanced myelodysplastic syndrome (MDS) and peripheral T-cell lymphoma (PTCL) (NCT02909972; NCT02264613). [0333] In certain embodiments, the second therapeutic agent is an inhibitor of transforming growth factor-beta (TGF-beta or TGFβ). Inhibitors of TGF-beta proteins being studied which may be used in the present invention include NIS793 (Novartis), an anti-TGF-beta antibody being tested in the clinic for treatment of various cancers, including breast, lung, hepatocellular, colorectal, pancreatic, prostate and renal cancer (NCT 02947165). In some embodiments, the inhibitor of TGF-beta proteins is fresolimumab (GC1008; Sanofi-Genzyme), which is being studied for melanoma (NCT00923169); renal cell carcinoma (NCT00356460); and non-small cell lung cancer (NCT02581787). Additionally, in some embodiments, the additional therapeutic agent is a TGF-beta trap, such as described in Connolly et al. (2012) Int'l J. Biological Sciences 8:964- 978. One therapeutic compound currently in clinical trials for treatment of solid tumors is M7824 (Merck KgaA—formerly MSB0011459X), which is a bispecific, anti-PD-L1/TGFβ trap compound (NCT02699515); and (NCT02517398). M7824 is comprised of a fully human IgG1 antibody against PD-L1 fused to the extracellular domain of human TGF-beta receptor II, which functions as a TGFβ “trap.” [0334] In certain embodiments, the second therapeutic agent is a cancer vaccine. In some embodiments, the cancer vaccine is selected from sipuleucel-T (Provenge®, Dendreon/Valeant Pharmaceuticals), which has been approved for treatment of asymptomatic, or minimally symptomatic metastatic castrate-resistant (hormone-refractory) prostate cancer; and talimogene laherparepvec (Imlygic®, BioVex/Amgen, previously known as T-VEC), a genetically modified oncolytic viral therapy approved for treatment of unresectable cutaneous, subcutaneous and nodal lesions in melanoma. In some embodiments, the additional therapeutic agent is selected from an oncolytic viral therapy such as pexastimogene devacirepvec (PexaVec/JX-594, SillaJen/formerly Jennerex Biotherapeutics), a thymidine kinase- (TK-) deficient vaccinia virus engineered to express GM-CSF, for hepatocellular carcinoma (NCT02562755) and melanoma (NCT00429312); pelareorep (Reolysin®, Oncolytics Biotech), a variant of respiratory enteric orphan virus (reovirus) which does not replicate in cells that are not RAS-activated, in numerous cancers, including colorectal cancer (NCT01622543); prostate cancer (NCT01619813); head and neck squamous cell cancer (NCT01166542); pancreatic adenocarcinoma (NCT00998322); and non-small cell lung cancer (NSCLC) (NCT 00861627); enadenotucirev (NG-348, PsiOxus, formerly known as ColoAd1), an adenovirus engineered to express a full length CD80 and an antibody fragment specific for the T-cell receptor CD3 protein, in ovarian cancer (NCT02028117); metastatic or advanced epithelial tumors such as in colorectal cancer, bladder cancer, head and neck squamous cell carcinoma and salivary gland cancer (NCT02636036); ONCOS-102 (Targovax/formerly Oncos), an adenovirus engineered to express GM-CSF, in melanoma (NCT03003676); and peritoneal disease, colorectal cancer or ovarian cancer (NCT02963831); GL-ONC1 (GLV- 1h68/GLV-1h153, Genelux GmbH), vaccinia viruses engineered to express beta-galactosidase (beta-gal)/beta-glucoronidase or beta-gal/human sodium iodide symporter (hNIS), respectively, were studied in peritoneal carcinomatosis (NCT01443260); fallopian tube cancer, ovarian cancer (NCT 02759588); or CG0070 (Cold Genesys), an adenovirus engineered to express GM-CSF, in bladder cancer (NCT02365818). [0335] In certain embodiments, the second therapeutic agent is an immune checkpoint inhibitor selected from a PD-1 antagonist, a PD-L1 antagonist, or a CTLA-4 antagonist. In some embodiments, a compound disclosed herein or a pharmaceutically acceptable salt thereof is administered in combination with nivolumab (anti-PD-1 antibody, Opdivo®, Bristol-Myers Squibb); pembrolizumab (anti-PD-1 antibody, Keytruda®, Merck); ipilimumab (anti-CTLA-4 antibody, Yervoy®, Bristol-Myers Squibb); durvalumab (anti-PD-L1 antibody, Imfinzi®, AstraZeneca); or atezolizumab (anti-PD-L1 antibody, Tecentriq®, Genentech). Other immune checkpoint inhibitors suitable for use in the present invention include REGN2810 (Regeneron), an anti-PD-1 antibody tested in patients with basal cell carcinoma (NCT03132636); NSCLC (NCT03088540); cutaneous squamous cell carcinoma (NCT02760498); lymphoma (NCT02651662); and melanoma (NCT03002376); pidilizumab (CureTech), also known as CT- 011, an antibody that binds to PD-1, in clinical trials for diffuse large B-cell lymphoma and multiple myeloma; avelumab (Bavencio®, Pfizer/Merck KGaA), also known as MSB0010718C), a fully human IgG1 anti-PD-L1 antibody, in clinical trials for non-small cell lung cancer, Merkel cell carcinoma, mesothelioma, solid tumors, renal cancer, ovarian cancer, bladder cancer, head and neck cancer, and gastric cancer; and PDR001 (Novartis), an inhibitory antibody that binds to PD-1, in clinical trials for non-small cell lung cancer, melanoma, triple negative breast cancer and advanced or metastatic solid tumors. Tremelimumab (CP-675,206; Astrazeneca) is a fully human monoclonal antibody against CTLA-4 that has been in studied in clinical trials for a number of indications, including: mesothelioma, colorectal cancer, kidney cancer, breast cancer, lung cancer and non-small cell lung cancer, pancreatic ductal adenocarcinoma, pancreatic cancer, germ cell cancer, squamous cell cancer of the head and neck, hepatocellular carcinoma, prostate cancer, endometrial cancer, metastatic cancer in the liver, liver cancer, large B-cell lymphoma, ovarian cancer, cervical cancer, metastatic anaplastic thyroid cancer, urothelial cancer, fallopian tube cancer, multiple myeloma, bladder cancer, soft tissue sarcoma, and melanoma. AGEN-1884 (Agenus) is an anti-CTLA4 antibody that is being studied in Phase 1 clinical trials for advanced solid tumors (NCT02694822). [0336] Another aspect of the invention provides for the use of a compound described herein (such as a compound of Formula I, Formula II, or other compounds in Section I) in the manufacture of a medicament. In certain embodiments, the medicament is for treating a disease described herein, such as cancer. [0337] Another aspect of the invention provides for the use of a compound described herein (such as a compound of Formula I, Formula II, or other compounds in Section I) for treating a medical disease, such a disease described herein (e.g., cancer). Evaluation of Cellular Growth Inhibition of HEK293 cells and HeLa cells [0338] Compounds can be evaluated for ability to inhibit the proliferation of HEK293 cells or HeLa cells according to the following procedure. HEK293 and HeLa cells are cultured in DMEM medium supplemented with 10% fetal bovine serum and 1% Penn/Strep. Cells are seeded in white 384-well plates at 500 cells/well in 25 ^L complete medium. Following seeding, plates are spun at 300 × g^for three minutes and cultured at 37°C with 5% CO 2 ^in a humidified tissue culture incubator. After 24 hours, compounds are titrated in 100% DMSO and diluted in complete cell culture medium. A 25 ^L aliquot of compound/media mixture is added to cells to bring total volume in well to 50 ^L. DMSO alone is used as a negative control. Plates are then spun at 300×g^for three minutes and stored at 37°C with 5% CO2 for three days. On Day 0 and Day 3 of compound treatment, cell viability is quantified with CellTiter-Glo 2.0 reagent (Promega). After equilibrating microplates at room temperature for 30 minutes, 25 µL CellTiter-Glo 2.0 reagent is dispensed into each well to bring total volume to 75 ^L. Plates are mixed on shaker for 2 minutes at 500rpm, followed by a 10-minute incubation at room temperature. Following a quick spin, luminescence readings are measured with an EnVision Plate Reader. Data is normalized to DMSO treated Day 0 and Day 3 readings. A four-parameter non-linear regression curve fit is applied to dose-response data in GraphPad Prism data analysis software to determine the half maximal growth inhibitory concentration (GI 50 ) for each compound. III. Pharmaceutical Compositions and Dosing Considerations [0339] As indicated above, the invention provides pharmaceutical compositions, which comprise a therapeutically-effective amount of one or more of the compounds described above, formulated together with one or more pharmaceutically acceptable carriers (additives) and/or diluents. The pharmaceutical compositions may be specially formulated for administration in solid or liquid form, including those adapted for the following: (1) 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; (2) parenteral administration, for example, by subcutaneous, intramuscular, intravenous or epidural injection as, for example, a sterile solution or suspension, or sustained-release formulation; (3) topical application, for example, as a cream, ointment, or a controlled-release patch or spray applied to the skin; (4) intravaginally or intrarectally, for example, as a pessary, cream or foam; (5) sublingually; (6) ocularly; (7) transdermally; or (8) nasally. In certain embodiments, the invention provides a pharmaceutical composition comprising a compound described herein (e.g., a compound of Formula I) and a pharmaceutically acceptable carrier. [0340] The phrase “therapeutically effective amount” as used herein means that amount of a compound, material, or composition comprising a compound of the present invention which is effective for producing some desired therapeutic effect in at least a sub-population of cells in an animal at a reasonable benefit/risk ratio applicable to any medical treatment. [0341] The phrase “pharmaceutically acceptable” is employed herein to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio. [0342] Wetting agents, emulsifiers and lubricants, such as sodium lauryl sulfate and magnesium stearate, as well as coloring agents, release agents, coating agents, sweetening, flavoring and perfuming agents, preservatives and antioxidants can also be present in the compositions. [0343] Examples of pharmaceutically-acceptable antioxidants include: (1) water soluble antioxidants, such as ascorbic acid, cysteine hydrochloride, sodium bisulfate, sodium metabisulfite, sodium sulfite and the like; (2) oil-soluble antioxidants, such as ascorbyl palmitate, butylated hydroxyanisole (BHA), butylated hydroxytoluene (BHT), lecithin, propyl gallate, alpha- tocopherol, and the like; and (3) metal chelating agents, such as citric acid, ethylenediamine tetraacetic acid (EDTA), sorbitol, tartaric acid, phosphoric acid, and the like. [0344] Formulations of the present invention include those suitable for oral, nasal, topical (including buccal and sublingual), rectal, vaginal and/or parenteral administration. The formulations may conveniently be presented in unit dosage form and may be prepared by any methods well known in the art of pharmacy. The amount of active ingredient which can be combined with a carrier material to produce a single dosage form will vary depending upon the host being treated, the particular mode of administration. The amount of active ingredient which can be combined with a carrier material to produce a single dosage form will generally be that amount of the compound which produces a therapeutic effect. Generally, out of one hundred percent, this amount will range from about 0.1 percent to about ninety-nine percent of active ingredient, preferably from about 5 percent to about 70 percent, most preferably from about 10 percent to about 30 percent. [0345] In certain embodiments, a formulation of the present invention comprises an excipient selected from the group consisting of cyclodextrins, celluloses, liposomes, micelle forming agents, e.g., bile acids, and polymeric carriers, e.g., polyesters and polyanhydrides; and a compound of the present invention. In certain embodiments, an aforementioned formulation renders orally bioavailable a compound of the present invention. [0346] Methods of preparing these formulations or compositions include the step of bringing into association a compound of the present invention with the carrier and, optionally, one or more accessory ingredients. In general, the formulations are prepared by uniformly and intimately bringing into association a compound of the present invention with liquid carriers, or finely divided solid carriers, or both, and then, if necessary, shaping the product. [0347] Formulations of the invention suitable for oral administration may be in the form of capsules, cachets, pills, tablets, lozenges (using a flavored basis, usually sucrose and acacia or tragacanth), powders, granules, or as a solution or a suspension in an aqueous or non-aqueous liquid, or as an oil-in-water or water-in-oil liquid emulsion, or as an elixir or syrup, or as pastilles (using an inert base, such as gelatin and glycerin, or sucrose and acacia) and/or as mouth washes and the like, each containing a predetermined amount of a compound of the present invention as an active ingredient. A compound of the present invention may also be administered as a bolus, electuary or paste. [0348] In solid dosage forms of the invention for oral administration (capsules, tablets, pills, dragees, powders, granules, trouches and the like), the active ingredient is mixed with one or more pharmaceutically-acceptable carriers, such as sodium citrate or dicalcium phosphate, and/or any of the following: (1) fillers or extenders, such as starches, lactose, sucrose, glucose, mannitol, and/or silicic acid; (2) binders, such as, for example, carboxymethylcellulose, alginates, gelatin, polyvinyl pyrrolidone, sucrose and/or acacia; (3) humectants, such as glycerol; (4) disintegrating agents, such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate; (5) solution retarding agents, such as paraffin; (6) absorption accelerators, such as quaternary ammonium compounds and surfactants, such as poloxamer and sodium lauryl sulfate; (7) wetting agents, such as, for example, cetyl alcohol, glycerol monostearate, and non-ionic surfactants; (8) absorbents, such as kaolin and bentonite clay; (9) lubricants, such as talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate, zinc stearate, sodium stearate, stearic acid, and mixtures thereof; (10) coloring agents; and (11) controlled release agents such as crospovidone or ethyl cellulose. In the case of capsules, tablets and pills, the pharmaceutical compositions may also comprise buffering agents. Solid compositions of a similar type may also be employed as fillers in soft and hard-shelled gelatin capsules using such excipients as lactose or milk sugars, as well as high molecular weight polyethylene glycols and the like. [0349] A tablet may be made by compression or molding, optionally with one or more accessory ingredients. Compressed tablets may be prepared using binder (for example, gelatin or hydroxypropylmethyl cellulose), lubricant, inert diluent, preservative, disintegrant (for example, sodium starch glycolate or cross-linked sodium carboxymethyl cellulose), surface-active or dispersing agent. Molded tablets may be made by molding in a suitable machine a mixture of the powdered compound moistened with an inert liquid diluent. [0350] The tablets, and other solid dosage forms of the pharmaceutical compositions of the present invention, such as dragees, capsules, pills and granules, may optionally be scored or prepared with coatings and shells, such as enteric coatings and other coatings well known in the pharmaceutical-formulating art. They may also be formulated so as to provide slow or controlled release of the active ingredient therein using, for example, hydroxypropylmethyl cellulose in varying proportions to provide the desired release profile, other polymer matrices, liposomes and/or microspheres. They may be formulated for rapid release, e.g., freeze-dried. They may be sterilized by, for example, filtration through a bacteria-retaining filter, or by incorporating sterilizing agents in the form of sterile solid compositions which can be dissolved in sterile water, or some other sterile injectable medium immediately before use. These compositions may also optionally contain opacifying agents and may be of a composition that they release the active ingredient(s) only, or preferentially, in a certain portion of the gastrointestinal tract, optionally, in a delayed manner. Examples of embedding compositions which can be used include polymeric substances and waxes. The active ingredient can also be in micro-encapsulated form, if appropriate, with one or more of the above-described excipients. [0351] Liquid dosage forms for oral administration of the compounds of the invention include pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs. In addition to the active ingredient, the liquid dosage forms may contain inert diluents commonly used in the art, such as, for example, water or other solvents, solubilizing agents and emulsifiers, such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor and sesame oils), glycerol, tetrahydrofuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof. [0352] Besides inert diluents, the oral compositions can also include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, coloring, perfuming and preservative agents. [0353] Suspensions, in addition to the active compounds, may contain suspending agents as, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar and tragacanth, and mixtures thereof. [0354] Formulations of the pharmaceutical compositions of the invention for rectal or vaginal administration may be presented as a suppository, which may be prepared by mixing one or more compounds of the invention with one or more suitable nonirritating excipients or carriers comprising, for example, cocoa butter, polyethylene glycol, a suppository wax or a salicylate, and which is solid at room temperature, but liquid at body temperature and, therefore, will melt in the rectum or vaginal cavity and release the active compound. [0355] Formulations of the present invention which are suitable for vaginal administration also include pessaries, tampons, creams, gels, pastes, foams or spray formulations containing such carriers as are known in the art to be appropriate. [0356] Dosage forms for the topical or transdermal administration of a compound of this invention include powders, sprays, ointments, pastes, creams, lotions, gels, solutions, patches and inhalants. The active compound may be mixed under sterile conditions with a pharmaceutically- acceptable carrier, and with any preservatives, buffers, or propellants which may be required. [0357] The ointments, pastes, creams and gels may contain, in addition to an active compound of this invention, excipients, such as animal and vegetable fats, oils, waxes, paraffins, starch, tragacanth, cellulose derivatives, polyethylene glycols, silicones, bentonites, silicic acid, talc and zinc oxide, or mixtures thereof. [0358] Powders and sprays can contain, in addition to a compound of this invention, excipients such as lactose, talc, silicic acid, aluminum hydroxide, calcium silicates and polyamide powder, or mixtures of these substances. Sprays can additionally contain customary propellants, such as chlorofluorohydrocarbons and volatile unsubstituted hydrocarbons, such as butane and propane. [0359] Transdermal patches have the added advantage of providing controlled delivery of a compound of the present invention to the body. Such dosage forms can be made by dissolving or dispersing the compound in the proper medium. Absorption enhancers can also be used to increase the flux of the compound across the skin. The rate of such flux can be controlled by either providing a rate controlling membrane or dispersing the compound in a polymer matrix or gel. [0360] Ophthalmic formulations, eye ointments, powders, solutions and the like, are also contemplated as being within the scope of this invention. [0361] Pharmaceutical compositions of this invention suitable for parenteral administration comprise one or more compounds of the invention in combination with one or more pharmaceutically-acceptable sterile isotonic aqueous or nonaqueous solutions, dispersions, suspensions or emulsions, or sterile powders which may be reconstituted into sterile injectable solutions or dispersions just prior to use, which may contain sugars, alcohols, antioxidants, buffers, bacteriostats, solutes which render the formulation isotonic with the blood of the intended recipient or suspending or thickening agents. [0362] Examples of suitable aqueous and nonaqueous carriers which may be employed in the pharmaceutical compositions of the invention include water, ethanol, polyols (such as glycerol, propylene glycol, polyethylene glycol, and the like), and suitable mixtures thereof, vegetable oils, such as olive oil, and injectable organic esters, such as ethyl oleate. Proper fluidity can be maintained, for example, by the use of coating materials, such as lecithin, by the maintenance of the required particle size in the case of dispersions, and by the use of surfactants. [0363] These compositions may also contain adjuvants such as preservatives, wetting agents, emulsifying agents and dispersing agents. Prevention of the action of microorganisms upon the subject compounds may be ensured by the inclusion of various antibacterial and antifungal agents, for example, paraben, chlorobutanol, phenol sorbic acid, and the like. It may also be desirable to include isotonic agents, such as sugars, sodium chloride, and the like into the compositions. In addition, prolonged absorption of the injectable pharmaceutical form may be brought about by the inclusion of agents which delay absorption such as aluminum monostearate and gelatin. [0364] In some cases, in order to prolong the effect of a drug, it is desirable to slow the absorption of the drug from subcutaneous or intramuscular injection. This may be accomplished by the use of a liquid suspension of crystalline or amorphous material having poor water solubility. The rate of absorption of the drug then depends upon its rate of dissolution which, in turn, may depend upon crystal size and crystalline form. Alternatively, delayed absorption of a parenterally- administered drug form is accomplished by dissolving or suspending the drug in an oil vehicle. [0365] Injectable depot forms are made by forming microencapsule matrices of the subject compounds in biodegradable polymers such as polylactide-polyglycolide. Depending on the ratio of drug to polymer, and the nature of the particular polymer employed, the rate of drug release can be controlled. Examples of other biodegradable polymers include poly(orthoesters) and poly(anhydrides). Depot injectable formulations are also prepared by entrapping the drug in liposomes or microemulsions which are compatible with body tissue. [0366] When the compounds of the present invention are administered as pharmaceuticals, to humans and animals, they can be given per se or as a pharmaceutical composition containing, for example, 0.1 to 99% (more preferably, 10 to 30%) of active ingredient in combination with a pharmaceutically acceptable carrier. [0367] The preparations of the present invention may be given orally, parenterally, topically, or rectally. They are of course given in forms suitable for each administration route. For example, they are administered in tablets or capsule form, by injection, inhalation, eye lotion, ointment, suppository, etc. administration by injection, infusion or inhalation; topical by lotion or ointment; and rectal by suppositories. Oral administrations are preferred. [0368] The phrases “parenteral administration” and “administered parenterally” as used herein means modes of administration other than enteral and topical administration, usually by injection, and includes, without limitation, intravenous, intramuscular, intraarterial, intrathecal, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intraarticular, subcapsular, subarachnoid, intraspinal and intrasternal injection and infusion. [0369] The phrases “systemic administration,” “administered systemically,” “peripheral administration” and “administered peripherally” as used herein mean the administration of a compound, drug or other material other than directly into the central nervous system, such that it enters the patient’s system and, thus, is subject to metabolism and other like processes, for example, subcutaneous administration. [0370] These compounds may be administered to humans and other animals for therapy by any suitable route of administration, including orally, nasally, as by, for example, a spray, rectally, intravaginally, parenterally, intracisternally and topically, as by powders, ointments or drops, including buccally and sublingually. [0371] Regardless of the route of administration selected, the compounds of the present invention, which may be used in a suitable hydrated form, and/or the pharmaceutical compositions of the present invention, are formulated into pharmaceutically-acceptable dosage forms by conventional methods known to those of skill in the art. [0372] Actual dosage levels of the active ingredients in the pharmaceutical compositions of this invention may be varied so as to obtain an amount of the active ingredient which is effective to achieve the desired therapeutic response for a particular patient, composition, and mode of administration, without being toxic to the patient. [0373] The selected dosage level will depend upon a variety of factors including the activity of the particular compound of the present invention employed, or the ester, salt or amide thereof, the route of administration, the time of administration, the rate of excretion or metabolism of the particular compound being employed, the rate and extent of absorption, the duration of the treatment, other drugs, compounds and/or materials used in combination with the particular compound employed, the age, sex, weight, condition, general health and prior medical history of the patient being treated, and like factors well known in the medical arts. [0374] A physician or veterinarian having ordinary skill in the art can readily determine and prescribe the effective amount of the pharmaceutical composition required. For example, the physician or veterinarian could start doses of the compounds of the invention employed in the pharmaceutical composition at levels lower than that required in order to achieve the desired therapeutic effect and gradually increase the dosage until the desired effect is achieved. [0375] In general, a suitable daily dose of a compound of the invention will be that amount of the compound which is the lowest dose effective to produce a therapeutic effect. Such an effective dose will generally depend upon the factors described above. Preferably, the compounds are administered at about 0.01 mg/kg to about 200 mg/kg, more preferably at about 0.1 mg/kg to about 100 mg/kg, even more preferably at about 0.5 mg/kg to about 50 mg/kg. When the compounds described herein are co-administered with another agent (e.g., as sensitizing agents), the effective amount may be less than when the agent is used alone. [0376] If desired, the effective daily dose of the active compound may be administered as two, three, four, five, six or more sub-doses administered separately at appropriate intervals throughout the day, optionally, in unit dosage forms. Preferred dosing is one administration per day. [0377] The invention further provides a unit dosage form (such as a tablet or capsule) comprising a heterobifunctional substituted phenylpyrimidinone or related compound described herein in a therapeutically effective amount for the treatment of a medical disorder described herein. IV. MEDICAL KITS [0378] Another aspect of this invention is a kit comprising (i) a compound described herein, such as a compound of Formula I, and (ii) instructions for use, such as treating cancer. V. ENUMERATED EMBODIMENTS [0379] Another aspect of the invention provides the following enumerated embodiments: Embodiment No.1. A compound represented by Formula I: or a pharmaceutically acceptable salt thereof; wherein: R 1 is phenyl substituted by cyano, halogen, and m occurrences of R 4 ; R 2 and R 3 are independently hydrogen or C1-4 alkyl; R 4 is C1-4 alkyl; R 5 represents independently for each occurrence C 1-4 alkyl or halogen; A 1 is a pyridazinylene, pyrimidinylene, pyrazinylene, pyridinylene, or phenylene, each of which is substituted with n occurrences of R 5 ; L is a linker; and A 2 is one of the following: A 3 is phenylene, a 3-10 membered saturated monocyclic, bicyclic or spirocyclic carbocycylene, or C 1-6 alkylene; R 1B represents independently for each occurrence halo, C1-4 alkyl, or C1-4 haloalkyl; R 2B is hydrogen, halo, C1-4 alkyl, or C1-4 haloalkyl; R 3B is -N(R 8B )SO 2 R 9B , -SO 2 N(R 8B ) 2 , -SO 2 R 9B , -(C 1-6 alkylene)-SO 2 R 9B , C 1-6 hydroxyalkyl, or a 4-7 membered saturated carbocyclic ring in which one CH2 is replaced with SO2; R 4B is hydrogen, halo, or C 1-4 alkyl; R 5B is C 1-4 alkyl or C 3-4 cycloalkyl; R 6B is hydrogen, C1-4 alkyl, or C3-4 cycloalkyl; R 7B represents independently for each occurrence C1-4 alkyl or C3-4 cycloalkyl; R 8B represents independently for each occurrence hydrogen or C 1-4 alkyl; or two occurrences of R 8B are taken together with the nitrogen atom to which they are attached to form a 3-7 membered heterocyclic ring containing 1 or 2 heteroatoms independently selected from nitrogen and oxygen; or R 8B and R 9B are taken together with their intervening atoms to a form a 5-7 membered ring containing 1, 2, or 3 heteroatoms independently selected from nitrogen, oxygen, and sulfur; R 9B is C 1-6 alkyl, C 1-6 haloalkyl, -(C 1-6 alkylene)-(C 3-6 cycloalkyl), or C 3-6 cycloalkyl; and m, n, p and t are independently 0, 1, or 2. Embodiment No.2. The compound of embodiment 1, wherein m is 0. Embodiment No.3. The compound of embodiment 1, wherein R 1 is . Embodiment No.4. The compound of any one of embodiments 1-3, wherein R 2 is hydrogen. Embodiment No.5. The compound of any one of embodiments 1-4, wherein R 3 is hydrogen. Embodiment No.6. The compound of any one of embodiments 1-5, wherein the compound is a compound of Formula I. Embodiment No.7. The compound of any one of embodiments 1-5, wherein the compound is a compound of Formula Ia or Ib, or a pharmaceutically acceptable salt thereof: (1a) (1b). Embodiment No.8. The compound of any one of embodiments 1-5, wherein the compound is a compound of Formula Ic or a pharmaceutically acceptable salt thereof: Embodiment No.9. The compound of any one of embodiments 1-5, wherein the compound is a compound of Formula Id or a pharmaceutically acceptable salt thereof: Embodiment No.10. The compound of any one of embodiments 1-9, wherein A 1 is pyridazinylene substituted with n occurrences of R 5 . Embodiment No.11. The compound of any one of embodiments 1-9, wherein A 1 is . Embodiment No.12. The compound of any one of embodiments 1-9, wherein A 1 is pyrimidinylene substituted with n occurrences of R 5 . Embodiment No.13. The compound of any one of embodiments 1-9, wherein A 1 is the point of attachment to L. Embodiment No.14. The compound of any one of embodiments 1-9, wherein A 1 is the point of attachment to L. Embodiment No.15. The compound of any one of embodiments 1-9, wherein A 1 is pyrazinylene substituted with n occurrences of R 5 . Embodiment No.16. The compound of any one of embodiments 1-9, wherein A 1 is . Embodiment No.17. The compound of any one of embodiments 1-9, wherein A 1 is pyridinylene substituted with n occurrences of R 5 . Embodiment No.18. The compound of any one of embodiments 1-9, wherein A 1 is the point of attachment to L. Embodiment No.19. The compound of any one of embodiments 1-9, wherein A 1 is phenylene substituted with n occurrences of R 5 . Embodiment No.20. The compound of any one of embodiments 1-9, wherein A 1 is . Embodiment No.21. The compound of any one of embodiments 1-10, 12, 15, 17, or 19, wherein n is 0. Embodiment No.22. The compound of any one of embodiments 1-21, wherein A 2 is Embodiment No.23. The compound of any one of embodiments 1-22, wherein A 3 is a 3-10 membered saturated monocyclic, bicyclic or spirocyclic carbocycylene. Embodiment No.24. The compound of any one of embodiments 1-21, wherein A 2 is .

Embodiment No.25. The compound of any one of embodiments 1-21, wherein A 2 is Embodiment No.26. The compound of any one of embodiments 1-25, wherein R 1B is halo. Embodiment No.27. The compound of any one of embodiments 1-25, wherein R 1B is fluoro. Embodiment No.28. The compound of any one of embodiments 1-27, wherein p is 1. Embodiment No.29. The compound of any one of embodiments 1-25, wherein p is 0. Embodiment No.30. The compound of any one of embodiments 1-21, wherein A 2 is . Embodiment No.31. The compound of any one of embodiments 1-21, wherein A 2 is . Embodiment No.32. The compound of any one of embodiments 1-31, wherein R 2B is hydrogen. Embodiment No.33. The compound of any one of embodiments 1-32, wherein R 3B is - N(R 8B )SO 2 R 9B . Embodiment No.34. The compound of any one of embodiments 1-32, wherein R 3B is SO 2 R 9B . Embodiment No.35. The compound of any one of embodiments 1-32, wherein R 3B is -(C1-6 alkylene)-SO2R 9B . Embodiment No.36. The compound of any one of embodiments 1-32, wherein R 3B is - SO2N(R 8B )2. Embodiment No.37. The compound of any one of embodiments 1-32, wherein R 3B is C 1-6 hydroxyalkyl. Embodiment No.38. The compound of any one of embodiments 1-35, wherein R 9B is C1-6 alkyl or C1-6 haloalkyl. Embodiment No.39. The compound of any one of embodiments 1-38, wherein R 4B is hydrogen. Embodiment No.40. The compound of any one of embodiments 1-39, wherein R 5B is C1-4 alkyl. Embodiment No.41. The compound of any one of embodiments 1-39, wherein R 5B is methyl. Embodiment No.42. The compound of any one of embodiments 1-41, wherein R 6B is hydrogen. Embodiment No.43. The compound of any one of embodiments 1-42, wherein R 7B is C 1-4 alkyl. Embodiment No.44. The compound of any one of embodiments 1-42, wherein R 7B is methyl. Embodiment No.45. The compound of any one of embodiments 1-44, wherein t is 2. Embodiment No.46. The compound of any one of embodiments 1-21, wherein A 2 is

Embodiment No.47. The compound of any one of embodiments 1-46, wherein L is a bivalent, saturated or unsaturated, straight or branched C 1-60 hydrocarbon chain, wherein 0-20 methylene units of the hydrocarbon are independently replaced with -O-, -S-, -N(H)-, - N(C 1-6 alkyl)-, -OC(O)-, -C(O)O-, -S(O)-, -S(O) 2 -, -N(H)S(O) 2 -, -N(C 1-6 alkyl)S(O) 2 -, - S(O) 2 N(H)-, -S(O) 2 N(C 1-6 alkyl)-, -N(H)C(O)-, -N(C 1-6 alkyl)C(O)-, -C(O)N(H)-, - C(O)N(C1-6 alkyl)-, -OC(O)N(H)-, -OC(O)N(C1-6 alkyl)-, -N(H)C(O)O-, -N(C1-6 alkyl)C(O)O-, optionally substituted 3-10 membered carbocyclyl, or optionally substituted 3-10 membered heterocyclyl containing 1, 2, 3, or 4 heteroatoms independently selected from nitrogen, oxygen, and sulfur. Embodiment No.48. The compound of any one of embodiments 1-46, wherein L is a bivalent, saturated, straight or branched C 3-30 hydrocarbon chain, wherein 0-15 methylene units of the hydrocarbon are independently replaced with -O-, -N(H)-, -N(C 1-6 alkyl)-, -OC(O)-, - C(O)O-, -N(H)C(O)-, -N(C1-6 alkyl)C(O)-, -C(O)N(H)-, -C(O)N(C1-6 alkyl)-, 3-10 membered carbocyclyl, or 3-10 membered heterocyclyl containing 1, 2, or 3 heteroatoms independently selected from nitrogen, oxygen, and sulfur. Embodiment No.49. The compound of any one of embodiments 1-46, wherein L is a bivalent, saturated, straight or branched C 3-30 hydrocarbon chain, wherein 0-15 methylene units of the hydrocarbon are independently replaced with -O-, -N(H)-, -N(C 1-6 alkyl)-, -OC(O)-, - C(O)O-, -N(H)C(O)-, -N(C1-6 alkyl)C(O)-, -C(O)N(H)-, or -C(O)N(C1-6 alkyl)-. Embodiment No.50. The compound of any one of embodiments 1-46, wherein L is a bivalent, saturated or unsaturated, straight or branched C5-40 hydrocarbon chain, wherein 1-20 methylene units of the hydrocarbon are independently replaced with -O-, -N(H)-, -N(C 1-6 alkyl)-, -N(H)C(O)-, -N(C 1-6 alkyl)C(O)-, -C(O)N(H)-, -C(O)N(C 1-6 alkyl)-, optionally substituted 3-10 membered carbocyclyl, or optionally substituted 3-10 membered heterocyclyl containing 1, 2, 3, or 4 heteroatoms independently selected from nitrogen, oxygen, and sulfur. Embodiment No.51. The compound of any one of embodiments 1-46, wherein L is -(3-7 membered, monocyclic, saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen)-(OCH 2 CH 2 ) 1-15 -O-***, wherein *** is the point of attachment to A 2 . Embodiment No.52. The compound of any one of embodiments 1-46, wherein L is -(3-7 membered, monocyclic, saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen)-(OCH 2 CH 2 ) 1-5 -O-***, wherein *** is the point of attachment to A 2 . Embodiment No.53. The compound of any one of embodiments 1-46, wherein L is -(3-7 membered, monocyclic, saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen)-(OCH 2 CH 2 ) 6-10 -O-***, wherein *** is the point of attachment to A 2 . Embodiment No.54. The compound of any one of embodiments 1-46, wherein L is - piperidinylene-(OCH2CH2)1-15-O-***, wherein *** is the point of attachment to A 2 . Embodiment No.55. The compound of any one of embodiments 1-46, wherein L is , wherein *** is the point of attachment to A 2 . Embodiment No.56. The compound of any one of embodiments 1-46, wherein L is , wherein *** is the point of attachment to A 2 . Embodiment No.57. The compound of any one of embodiments 1-46, wherein L is , wherein *** is the point of attachment to A 2 . Embodiment No.58. The compound of any one of embodiments 1-46, wherein L is -(3-7 membered, monocyclic, saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen)-(OCH 2 CH 2 ) 1-15 -N(H)C(O)-C 1-10 alkylene-***, -(3-7 membered, monocyclic, saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen)- (OCH2CH2)1-15-N(C1-4 alkyl)C(O)-C1-10 alkylene-***, -(3-7 membered, monocyclic, saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen)- (OCH2CH2)1-15-C(O)N(H)-C1-10 alkylene-***, or -(3-7 membered, monocyclic, saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen)-(OCH 2 CH 2 ) 1-15 - C(O)N(C 1-4 alkyl)-C 1-10 alkylene-***, wherein *** is the point of attachment to A 2 . Embodiment No.59. The compound of any one of embodiments 1-46, wherein L is - piperidinylene-(OCH2CH2)1-5-N(H)C(O)-C1-5 alkylene-***, -piperidinylene-(OCH2CH2)1-5- N(C 1-4 alkyl)C(O)-C 1-5 alkylene-***, -piperidinylene-(OCH 2 CH 2 ) 1-5 -C(O)N(H)-C 1-5 alkylene-***, or -piperidinylene-(OCH2CH2)1-5-C(O)N(C1-4 alkyl)-C1-5 alkylene-***, wherein *** is the point of attachment to A 2 . Embodiment No.60. The compound of any one of embodiments 1-46, wherein L is -(3-7 membered, monocyclic, saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen)-(OCH2CH2)1-10-***, -(3-7 membered, monocyclic, saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen)-(C 0-10 alkylene)-O-***, or -(3-7 membered, monocyclic, saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen)-C1-10 alkylene, wherein *** is the point of attachment to A 2 . Embodiment No.61. The compound of any one of embodiments 1-46, wherein L is - piperidinylene-(OCH 2 CH 2 ) 1-5 -***, -piperidinylene-(C 0-5 alkylene)-O-***, or - piperidinylene-(C1-5 alkylene)-***, wherein *** is the point of attachment to A 2 . Embodiment No.62. The compound of any one of embodiments 1-46, wherein L is -(3-7 membered monocyclic saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen)-X 1 -***, wherein *** is the point of attachment to A 2 , and X 1 is (i) C 1-10 alkylene where 1 or 2 methylene groups are optionally replaced by -O-, -N(H)-, or -N(C1-4 alkyl)-, (ii) a 3-7 membered monocyclic saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen, or (iii) -(3-7 membered monocyclic saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen)-(C1-10 alkylene)-. Embodiment No.63. The compound of any one of embodiments 1-46, wherein L is - (piperidinylene)-X 1 -***, wherein *** is the point of attachment to A 2 , and X 1 is (i) C 1-5 alkylene where 1 or 2 methylene groups are optionally replaced by -O-, -N(H)-, or -N(C1-4 alkyl)-, (ii) a 3-4 membered monocyclic saturated heterocyclic ring containing 1 heteroatom selected from nitrogen, or (iii) -(3-4 membered monocyclic saturated heterocyclic ring containing 1 heteroatom selected from nitrogen)-(C 1-5 alkylene)-. Embodiment No.64. The compound of any one of embodiments 1-46, wherein L is , wherein *** is the point of attachment to A 2 , and X 1 is (i) C1-10 alkylene where 1 or 2 methylene groups are optionally replaced by -O-, -N(H)-, or -N(C1-4 alkyl)-, (ii) a 3-7 membered monocyclic saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen, or (iii) -(3-7 membered monocyclic saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen)-(C1-10 alkylene)-. Embodiment No.65. The compound of any one of embodiments 1-46, wherein L is - (piperazinylene)-X 1 -***, wherein *** is the point of attachment to A 2 , and X 1 is (i) C1-5 alkylene where 1 or 2 methylene groups are optionally replaced by -O-, (ii) a 3-4 membered monocyclic saturated heterocyclic ring containing 1 heteroatom selected from nitrogen, or (iii) -(3-4 membered monocyclic saturated heterocyclic ring containing 1 heteroatom selected from nitrogen)-(C1-5 alkylene)-. Embodiment No.66. The compound of any one of embodiments 1-46, wherein L is , wherein *** is the point of attachment to A 2 , and X 1 is (i) C 1-10 alkylene where 1 or 2 methylene groups are optionally replaced by -O-, (ii) a 3-7 membered monocyclic saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen, or (iii) -(3-7 membered monocyclic saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen)-(C1-10 alkylene)-. Embodiment No.67. The compound of any one of embodiments 1-46, wherein L is -(3-7 membered monocyclic saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen)-X 2 -(C 1-10 alkylene)-***, wherein *** is the point of attachment to A 2 , and X 2 is -O-, -N(H)-, or -N(C1-6 alkyl)-. Embodiment No.68. The compound of any one of embodiments 1-46, wherein L is - (piperidinylene)-X 2 -(C 1-10 alkylene)-***, wherein *** is the point of attachment to A 2 , and X 2 is -O-, -N(H)-, or -N(C1-6 alkyl)-. Embodiment No.69. The compound of any one of embodiments 1-46, wherein L is - (piperidinylene)-X 2 -(a 3-7 membered monocyclic saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen)-***, wherein *** is the point of attachment to A 2 , and X 2 is -O-, -N(H)-, or -N(C1-6 alkyl)-. Embodiment No.70. The compound of any one of embodiments 1-46, wherein L is , wherein *** is the point of attachment to A 2 , and X 2 is - O-, -N(H)-, or -N(C1-6 alkyl)-. Embodiment No.71. The compound of any one of embodiments 1-46, wherein L is , wherein *** is the point of attachment to A 2 , and X 2 is -O-, -N(H)-, or -N(C1-6 alkyl)-. Embodiment No.72. The compound of any one of embodiments 67-71, wherein X 2 is -O-. Embodiment No.73. The compound of any one of embodiments 1-46, wherein L is -(3-7 membered monocyclic saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen)-X 1 -***, wherein *** is the point of attachment to A 2 , and X 1 is - (OCH 2 CH 2 ) 1-10 where 1 CH 2 group is optionally replaced with -C(H)(C 3-6 cycloalkyl)-. Embodiment No.74. The compound of any one of embodiments 1-46, wherein L is a 7-11 membered spirocyclic or fused bicyclic saturated heterocyclic ring containing 1, 2, or 3 heteroatoms selected from nitrogen and oxygen. Embodiment No.75. The compound of any one of embodiments 1-46, wherein L is a 7-8 membered spirocyclic or fused bicyclic saturated heterocyclic ring containing 2 heteroatoms selected from nitrogen. Embodiment No.76. The compound of any one of embodiments 1-46, wherein L is -(3-7 membered monocyclic saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen)-X 3 -(3-7 membered monocyclic saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen)-, wherein X 3 is C 1-10 alkylene, -O-, -N(H)-, -N(C 1-4 alkyl)-, or a bond. Embodiment No.77. The compound of any one of embodiments 1-46, wherein L is - (piperidinylene)-(C 1-5 alkylene)-(piperazinylene)-***, wherein *** is the point of attachment to A 2 . Embodiment No.78. The compound of any one of embodiments 1-46, wherein L is - (piperazinylene)-(azetidinylene)-*** or (azetidinylene)-(piperazinylene)-***, wherein *** is the point of attachment to A 2 . Embodiment No.79. The compound of any one of embodiments 1-46, wherein L is -(3-7 membered monocyclic saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen)-X 3 -(C 3-6 cycloalkylene)-O-***, -(3-7 membered monocyclic saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen)-X 3 -(C 3-6 cycloalkylene)-N(H)-***, or -(3-7 membered monocyclic saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen)-X 3 -(C3-6 cycloalkylene)-N(C1-4 alkyl)-***, wherein *** is the point of attachment to A 2 , and X 3 is C 1-10 alkylene, -O-, - N(H)-, -N(C1-4 alkyl)-, or a bond. Embodiment No.80. The compound of any one of embodiments 1-46, wherein L is - (piperidinylene)-X 3 -(C 3-6 cycloalkylene)-O-***, -(piperidinylene)-X 3 -(C 3-6 cycloalkylene)- N(H)-***, or -(piperidinylene)-X 3 -(C 3-6 cycloalkylene)-***, wherein *** is the point of attachment to A 2 , and X 3 is C1-10 alkylene, -O-, -N(H)-, -N(C1-4 alkyl)-, or a bond. Embodiment No.81. The compound of any one of embodiments 1-46, wherein L has the formula –(C 0-12 alkylene)-(optionally substituted 3-40 membered heteroalkylene)-(C 0-12 alkylene)-. Embodiment No.82. The compound of any one of embodiments 1-46, wherein L is one of the following: wherein *** is the point of attachment to A 2 . Embodiment No.83. The compound of any one of embodiments 1-46, wherein L is one of the following:

wherein *** is the point of attachment to A 2 . EXAMPLES [0380] The invention now being generally described, will be more readily understood by reference to the following examples, which are included merely for purposes of illustration of certain aspects and embodiments of the present invention, and is not intended to limit the invention. General Methods [0381] All reactions were carried out under an atmosphere of dry nitrogen or argon. Glassware was oven-dried prior to use. Unless otherwise indicated, common reagents or materials were obtained from commercial sources and used without further purification. N,N- Diisopropylethylamine (DIPEA) was obtained anhydrous by distillation over potassium hydroxide. Tetrahydrofuran (THF), dichloromethane (CH2Cl2), and dimethylformamide (DMF) was dried by a PureSolv TM solvent drying system. PTLC refers to preparatory thin layer chromatographic separation. Abbreviations: HFIP (hexafluoroisopropanol), HEPES (4-(2-hydroxyethyl)-1- piperazineethanesulfonic acid. Flash column chromatography was performed using silica gel 60 (230-400 mesh). Analytical thin layer chromatography (TLC) was carried out on Merck silica gel plates with QF-254 indicator and visualized by UV or KMnO4. 1 H and 13 C NMR spectra were recorded on an Agilent DD 2 500 (500 MHz 1 H; 125 MHz 13 C) or Agilent DD 2 600 (600 MHz 1 H; 150 MHz 13 C) or Agilent DD 2 400 (400 MHz 1 H; 100 MHz 13 C) spectrometer at room temperature. Chemical shifts were reported in ppm relative to the residual CDCl3 (δ 7.26 ppm 1 H; δ 77.0 ppm 13 C), CD3OD (δ 3.31 ppm 1 H; δ 49.00 ppm 13 C), or d6-DMSO (δ 2.50 ppm 1 H; δ 39.52 ppm 13 C). NMR chemical shifts were expressed in ppm relative to internal solvent peaks, and coupling constants were measured in Hz. (bs = broad signal). In most cases, only peaks of the major rotamer are reported. [0382] Mass spectra were obtained using Agilent 1100 series LC/MSD spectrometers. Analytical HPLC analyses were carried out on 250 x 4.6 mm C-18 column using gradient conditions (10-100% B, flow rate = 1.0 mL/min, 20 min), or as as described in the LC-MS Method tables. [0383] Unless indicated otherwise, preparative HPLC was carried out on 250 x 21.2 mm C-18 column using gradient conditions (10-100% B, flow rate = 10.0 mL/min, 20 min). The eluents used were: solvent A (H 2 O with 0.1% TFA) and solvent B (CH 3 CN with 0.1% TFA). Final products were typically purified via reversed-phase HPLC, PTLC, or flash column chromatography.

[0384] The following abbreviations are used herein: ACN: acetonitrile; Bn: benzyl; Boc: tert- butoxycarbonyl; DCM: dichloromethane; DIEA: diisopropylethylamine; DMF: dimethylformamide; DMSO: dimethylsulfoxide; EtOH: ethanol; EA or EtOAc: ethyl acetate; equiv. or eq.: molar equivalents; FA: formic acid; h: hour or hours; HATU: 1- [bis(dimethylamino)methylene]-1H-1,2,3-triazolo[4,5-b]pyridi nium 3-oxide hexafluoro-phosphate; HPLC: high-pressure liquid chromatography; LCMS or LC-MS: liquid chromatography-mass spectrometry; MeCN: acetonitrile; MeOH: methanol; MS: mass spectrometry; NMP: N- methylpyrrolidone; NMR: nuclear magnetic resonance; PE: petroleum ether; rt: room temperature; TEA: triethylamine; TFA: trifluoroacetic acid; THF: tetrahydrofuran; TLC: thin-layer chromatography; psi: pounds-per-square inch; and Tos or Ts: p-toluenesulfonyl.

EXAMPLE 1 – Synthesis of N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]-6-[4-[2-[3-[4- (ethylsulfonylamino)-2-(6-methyl-7-oxo-1H-pyrrolo[2,3-c]pyri din-4-yl)phenoxy] phenoxy]ethoxy]-1-piperidyl]pyridazine-3-carboxamide (I-1)

[0385] Step 1: Preparation of tert-butyl 4-[2-(p-tolylsulfonyloxy)ethoxy]piperidine-1- carboxylate. To a solution of tert-butyl 4-(2-hydroxyethoxy)piperidine-1-carboxylate (5.0 g, 20 mmol, 1.0 equiv) in DCM (50 mL) was added TEA (7.2 g, 71 mmol, 3.5 equiv) and TosCl (9.7 g, 51 mmol, 2.5 equiv) at 0 ℃. The mixture was stirred at 25 ℃ for 2 hours and concentrated. The residue was purified by silica column chromatography on silica gel (PE:EA from 10/1 to 3/1) to afford tert-butyl 4-[2-(p-tolylsulfonyloxy)ethoxy]piperidine-1-carboxylate (7.0 g,86% yield) as a colorless oil. [0386] Step 2: Preparation of tert-butyl 4-[2-(3-benzyloxyphenoxy)ethoxy]piperidine-1- carboxylate. To a solution of tert-butyl 4-[2-(p-tolylsulfonyloxy)ethoxy]piperidine-1-carboxylate (3.0 g, 7.5 mmol, 1.0 equiv) and 3-benzyloxyphenol (1.5 g, 7.5 mmol, 1.0 equiv) in MeCN (30 mL) was added Cs 2 CO 3 (7.3 g, 23 mmol, 3.0 equiv). The mixture was stirred at 90 ℃ for 12 hours and concentrated. The residue was purified by silica column chromatography on silica gel (PE:EA from 10/1 to 4/1) to afford tert-butyl 4-[2-(3-benzyloxyphenoxy)ethoxy] piperidine-1-carboxylate (2.8 g,87% yield) as a yellow oil. [0387] Step 3: Preparation of tert-butyl 4-[2-(3-hydroxyphenoxy)ethoxy]piperidine-1- carboxylate. To a solution of tert-butyl 4-[2-(3-benzyloxyphenoxy)ethoxy]piperidine-1- carboxylate (3.1 g, 7.3 mmol, 1.0 equiv) in MeOH (60 mL) was added Pd/C (0.3 g, 10% purity) under N 2 . The suspension was degassed under vacuum and purged with H 2 for several times. The mixture was stirred under H 2 (50 psi) at 30 ℃ for 12 hours. The mixture was filtered and concentrated to afford tert-butyl 4-[2-(3-hydroxyphenoxy)ethoxy]piperidine-1-carboxylate (2 g, crude) as a colorless oil. [0388] Step 4: Preparation of tert-butyl 4-[2-[3-[2-(6-methyl-7-oxo-1H-pyrrolo[2,3- c]pyridin-4-yl)-4-nitro-phenoxy]phenoxy]ethoxy]piperidine-1- carboxylate. To a solution of tert-butyl 4-[2-(3-hydroxyphenoxy)ethoxy]piperidine-1-carboxylate (0.5 g, 1.5 mmol, 1.0 equiv) and 4-(2-fluoro-5-nitro-phenyl)-6-methyl-1-(p-tolylsulfonyl)pyrr olo[2,3-c]pyridin-7-one (0.6 g, 1.5 mmol, 1.0 equiv) in DMSO (10 mL) was added Cs2CO3 (1.5 g, 4.5 mmol, 3.0 equiv). The mixture was stirred at 70 ℃ for 12 hours. The mixture was diluted with water (100 mL) and extracted with EtOAc (3 x 10 mL). The combined organic layers were washed with brine (10 mL), dried over sodium sulfate, filtered, and concentrated. The residue was purified by prep-TLC (DCM:MeOH = 10:1) to afford tert-butyl 4-[2-[3-[2-(6-methyl-7-oxo-1H-pyrrolo[2,3-c]pyridin-4-yl)-4- nitro- phenoxy]phenoxy]ethoxy]piperidine-1-carboxylate (0.15 g, 17% yield) as a yellow oil. [0389] Step 5: Preparation of tert-butyl 4-[2-[3-[4-amino-2-(6-methyl-7-oxo-1H- pyrrolo[2,3-c]pyridin-4-yl)phenoxy]phenoxy]ethoxy]piperidine -1-carboxylate. To a solution of tert-butyl 4-[2-[3-[2-(6-methyl-7-oxo-1H-pyrrolo[2,3-c]pyridin-4-yl)-4- nitro- phenoxy]phenoxy]ethoxy]piperidine-1-carboxylate (150 mg, 248 ^mol, 1.0 equiv) in THF (5 mL) was added Pd/C (30 mg, 10% purity) under N2. The suspension was degassed under vacuum and purged with H 2 for several times. The mixture was stirred under H 2 (15 psi) at 25 ℃ for 12 hours. The mixture was filtered and concentrated to afford tert-butyl 4-[2-[3-[4-amino-2-(6-methyl-7- oxo-1H-pyrrolo[2,3-c]pyridin-4-yl)phenoxy]phenoxy]ethoxy]pip eridine-1-carboxylate (60 mg, 42% yield) as a colorless oil. [0390] Step 6: Preparation of tert-butyl 4-[2-[3-[4-(ethylsulfonylamino)-2-(6-methyl-7-oxo- 1H-pyrrolo[2,3-c]pyridin-4-yl)phenoxy]phenoxy]ethoxy]piperid ine-1-carboxylate. To a solution of tert-butyl 4-[2-[3-[4-amino-2-(6-methyl-7-oxo-1H-pyrrolo[2,3-c]pyridin- 4- yl)phenoxy]phenoxy]ethoxy]piperidine-1-carboxylate (60 mg, 104 ^mol, 1.0 equiv) in DCM (5 mL) was added TEA (106 mg, 1.0 mmol, 10.0 equiv) and ethanesulfonyl chloride (27 mg, 208 ^mol, 2.0 equiv) at 0 ℃. The mixture was stirred at 25 ℃ for 2 hours and concentrated. The residue was purified by prep-TLC (DCM:MeOH = 10:1) to afford tert-butyl 4-[2-[3-[4- (ethylsulfonylamino)-2-(6-methyl-7-oxo-1H-pyrrolo[2,3-c]pyri din-4-yl)phenoxy]phenoxy] ethoxy]piperidine-1-carboxylate (38 mg, 55% yield) as a colorless oil. [0391] Step 7: Preparation of N-[3-(6-methyl-7-oxo-1H-pyrrolo[2,3-c]pyridin-4-yl)-4-[3-[2- (4-piperidyloxy)ethoxy]phenoxy]phenyl]ethanesulfonamide. To a solution of tert-butyl 4-[2-[3- [4-(ethylsulfonylamino)-2-(6-methyl-7-oxo-1H-pyrrolo[2,3-c]p yridin-4-yl)phenoxy] phenoxy]ethoxy]piperidine-1-carboxylate (45 mg, 0.1 mmol, 1.0 equiv) in DCM (1 mL) was added TFA (0.5 mL). The mixture was stirred at 25 ℃ for 1 hour. The mixture was concentrated for the next step without further purification. [0392] Step 8: Preparation of N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]-6-[4-[2-[3-[4- (ethylsulfonylamino)-2-(6-methyl-7-oxo-1H-pyrrolo[2,3-c]pyri din-4-yl)phenoxy]phenoxy] ethoxy]-1-piperidyl]pyridazine-3-carboxamide (I-1). To a solution of N-[3-(6-methyl-7-oxo-1H- pyrrolo[2,3-c]pyridin-4-yl)-4-[3-[2-(4-piperidyloxy)ethoxy]p henoxy]phenyl] ethanesulfonamide (38 mg, 67 ^mol, 1.0 equiv) and 6-chloro-N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]pyridazin e- 3-carboxamide (26 mg, 67 ^mol, 1.0 equiv) in NMP (2 mL) was added K2CO3 (28 mg, 201 ^mol, 3.0 equiv). The mixture was stirred at 50 ℃ for 2 hours. The residue was purified by prep-HPLC (column: Phenomenex luna C18150 x 25 mm, 10 ^m; mobile phase: [water(FA)-ACN]; B%: 52%-82%,10 min) to give N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]-6-[4-[2-[3-[4- (ethylsulfonylamino)-2-(6-methyl-7-oxo-1H-pyrrolo[2,3-c]pyri din-4- yl)phenoxy]phenoxy]ethoxy]-1-piperidyl]pyridazine-3-carboxam ide (I-3) (18 mg, 28% yield) as a white solid. 1 H NMR (400 MHz, CD3OD): δ 12.04 (s, 1H), 9.90-9.69 (m, 1H), 8.59 (d, J = 8.2 Hz, 1H), 7.83 (d, J = 17.2 Hz, 1H), 7.39 (d, J = 5.6 Hz, 2H), 7.35 (d, J = 9.6 Hz, 1H), 7.30-7.26 (m, 2H), 7.22 (d, J = 8.28 Hz, 1H), 7.13 (d, J = 8.6 Hz, 2H), 7.06 (d, J = 8.8 Hz, 1H), 6.61-6.55 (m, 1H), 6.44-6.37 (m, 2H), 6.28-6.25 (m, 1H), 4.58-4.50 (m, 1H), 4.10-4.04 (m, 2H), 4.01-3.96 (m, 2H), 3.90-3.82 (m, 1H), 3.76-3.71 (m, 2H), 3.65 (t, J = 7.8 Hz, 1H), 3.48 (s, 3H), 3.12 (d, J = 7.4 Hz, 2H), 2.71-2.65 (m, 1H), 2.14-2.07 (m, 2H) , 1.94-1.86 (m, 4H), 1.70-1.59 (m, 2H), 1.57-1.43 (m, 4H), 1.23 (t, J = 7.4 Hz, 3H). LC-MS: MS (ES + ): RT = 3.037 min, m/z = 461.1, 921.3; LC-MS Method 10. EXAMPLE 2 – Synthesis of N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]-6-[4-[2-[2-[2-[3- [4- (ethylsulfonylamino)-2-(6-methyl-7-oxo-1H-pyrrolo[2,3-c]pyri din-4-yl)phenoxy] phenoxy]ethoxy]ethoxy]ethoxy]-1-piperidyl]pyridazine-3-carbo xamide (I-2)

[0393] Step 1: Preparation of 2-[2-(2-benzyloxyethoxy)ethoxy]ethyl 4-methyl benzenesulfonate. To a solution of 2-[2-(2-benzyloxyethoxy)ethoxy]ethanol (30 g, 125 mmol, 1.0 equiv) and TEA (36 g, 359 mmol, 2.9 equiv) in DCM (250 mL) was added TsCl (40 g, 210 mmol, 1.7 equiv). The mixture was stirred at 25 ℃ for 12 hours. The mixture was concentrated and purified by column chromatography (SiO 2 , PE/EA = 5/1 to 1/1) to give 2-[2-(2- benzyloxyethoxy)ethoxy]ethyl 4-methylbenzenesulfonate (45.5 g, 92% yield) as a white oil. [0394] Step 2: Preparation of tert-butyl 4-[2-[2-(2-benzyloxyethoxy)ethoxy]ethoxy] piperidine-1-carboxylate. To a solution of tert-butyl 4-hydroxypiperidine-1-carboxylate (12.2 g, 61 mmol, 1.2 equiv) in THF (200 mL) was added NaH (3.1 g, 78 mmol, 60% purity, 1.5 equiv) at 0 ℃. The mixture was stirred at 25 ℃ for 0.5 hour. Then 2-[2-(2-benzyloxyethoxy) ethoxy]ethyl 4- methylbenzenesulfonate (20 g, 50.7 mmol, 1.0 equiv) was added. The mixture was stirred at 25 ℃ for 12 hours and quenched by addition of saturated salt solution (10 mL) and H2O (50 mL). The mixture was extracted with EtOAc (4 x 25 mL). The combined organic layers were washed with H 2 O (60 mL), dried over anhydrous sodium sulfate, filtered, and concentrated. The residue was purified by column chromatography (SiO2, PE/EA = 30/1 to 5/1) to give tert-butyl 4-[2-[2-(2- benzyloxyethoxy)ethoxy]ethoxy] piperidine-1-carboxylate (21.4 g, 100% yield) as a yellow oil. [0395] Step 3: Preparation of tert-butyl 4-[2-[2-(2-hydroxyethoxy)ethoxy]ethoxy] piperidine-1-carboxylate. To a solution of tert-butyl 4-[2-[2-(2-benzyloxyethoxy)ethoxy] ethoxy]piperidine-1-carboxylate (10 g, 24 mmol, 1.0 equiv) in MeOH (100 mL) was added Pd/C (2 g, 10% purity) under N2. The mixture was stirred under H2 (50 psi) at 20 ℃ for 12 hours. The mixture was filtered and concentrated to give tert-butyl 4-[2-[2-(2-hydroxyethoxy)ethoxy] ethoxy]piperidine-1-carboxylate (8 g, crude) as a brown oil. 1 H NMR (400 MHz, CDCl 3 ): δ 3.95- 3.60 (m, 12H), 3.54-3.46 (m, 1H), 3.16-3.00 (m, 2H), 1.93-1.81 (m, 2H), 1.61-1.41 (m, 13H). [0396] Step 4: Preparation of tert-butyl 4-[2-[2-[2-(p-tolylsulfonyloxy)ethoxy]ethoxy] ethoxy]piperidine-1-carboxylate. A solution of tert-butyl 4-[2-[2-(2-hydroxyethoxy)ethoxy] ethoxy]piperidine-1-carboxylate (4 g, 12 mmol, 1.0 equiv), TEA (6 g, 60 mmol, 5.0 equiv), and TsCl (6.86 g, 36 mmol, 3.0 equiv) in DCM (40 mL) was stirred at 25 °C for 12 hours. The mixture was concentrated and purified by column chromatography (SiO 2 , PE/EA = 20/1 to 1/1) to give tert- butyl 4-[2-[2-[2-(p-tolylsulfonyloxy)ethoxy] ethoxy]ethoxy]piperidine-1-carboxylate (4.3 g, 74% yield) as a light-yellow oil. 1 H NMR (400 MHz, CDCl 3 ): δ 7.73 (d, J = 8.4 Hz, 2H), 7.27 (d, J = 8.0 Hz, 2H), 4.15-4.07 (m, 2H), 3.83-3.59 (m, 4H), 3.59-3.48 (m, 7H), 3.46-3.35 (m, 1H), 3.05- 2.92 (m, 2H), 2.38 (s, 3H), 1.83-1.71 (m, 2H), 1.49-1.33 (m, 12H). [0397] Step 5: Preparation of tert-butyl 4-[2-[2-[2-(3-benzyloxyphenoxy)ethoxy] ethoxy]ethoxy]piperidine-1-carboxylate. A mixture of tert-butyl 4-[2-[2-[2-(p- tolylsulfonyloxy)ethoxy]ethoxy]ethoxy]piperidine-1-carboxyla te (2.3 g, 4.7 mmol, 1.0 equiv), 3- benzyloxyphenol (1.0 g, 5.2 mmol, 1.1 equiv), and K 2 CO 3 (1.96 g, 14.2 mmol, 3.0 equiv) in ACN (20 mL) was stirred at 80 ℃ for 12 hours. The mixture was concentrated and purified by column chromatography (SiO2, PE/EA = 20/1 to 3/1) to give tert-butyl 4-[2-[2-[2-(3- benzyloxyphenoxy)ethoxy]ethoxy]ethoxy]piperidine-1-carboxyla te (1.9 g, 78% yield) as a brown oil. 1 H NMR (400 MHz, CDCl3): δ 7.54-7.30 (m, 5H), 7.26-7.14 (m, 1H), 6.69-6.43 (m, 3H), 5.13-5.00 (m, 2H), 4.24-4.05 (m, 3H), 3.88-3.61 (m, 11H), 3.55-3.42 (m, 1H), 3.11-3.00 (m, 2H), 1.94-1.78 (m, 2H), 1.56-1.49 (m, 2H), 1.48-1.43 (m, 9H). [0398] Step 6: Preparation of tert-butyl 4-[2-[2-[2-(3-hydroxyphenoxy)ethoxy] ethoxy]ethoxy]piperidine-1-carboxylate. To a solution of tert-butyl 4-[2-[2-[2-(3-benzyloxy- phenoxy)ethoxy]ethoxy]ethoxy]piperidine-1-carboxylate (1.9 g, 3.7 mmol, 1.0 equiv) in MeOH (20 mL) was added Pd/C (0.3 g, 10% purity) under N2. The mixture was stirred under H2 (15 psi) at 25 ℃ for 12 hours. The mixture was filtered, and the filtrate was concentrated under reduced pressure to give tert-butyl 4-[2-[2-[2-(3-hydroxyphenoxy)ethoxy] ethoxy]ethoxy]piperidine-1- carboxylate (1.4 g, 89% yield) as a brown oil. 1 H NMR (400 MHz, CDCl3): δ 7.13 (t, J = 8.0 Hz, 1H), 6.58- 6.41 (m, 2H), 4.20- 4.04 (m, 2H), 3.92-3.63 (m, 12H), 3.60-3.52 (m, 1H), 3.24-3.02 (m, 2H), 1.90-1.82 (m, 3H), 1.62-1.57 (m, 2H), 1.48 (s, 9H). [0399] Step 7: Preparation of tert-butyl 4-[2-[2-[2-[3-[2-(6-methyl-7-oxo-1H-pyrrolo[2,3- c]pyridin-4-yl)-4-nitro-phenoxy]phenoxy]ethoxy]ethoxy]ethoxy ]piperidine-1-carboxylate. A mixture of tert-butyl 4-[2-[2-[2-(3-hydroxyphenoxy)ethoxy]ethoxy]ethoxy]piperidine -1- carboxylate (420 mg, 987 ^mol, 1.0 equiv), 4-(2-fluoro-5-nitro-phenyl)-6-methyl-1-(p- tolylsulfonyl)pyrrolo[2,3-c]pyridin-7-one (436 mg, 987 ^mol, 1.0 equiv), and Cs 2 CO 3 (805 mg, 2.47 mmol, 2.5 equiv) in DMSO (8 mL) was stirred at 70 ℃ for 12 hours. The mixture was concentrated and purified by prep-HPLC (column: Waters Xbridge C18150 x 50 mm, 10 ^m; mobile phase: [water(NH 4 HCO 3 )-ACN]; B%: 43%-73%, 10 min) to give tert-butyl 4-[2-[2-[2-[3- [2-(6-methyl-7-oxo-1H-pyrrolo[2,3-c]pyridin-4-yl)-4-nitro-ph enoxy]phenoxy]ethoxy] ethoxy]ethoxy]piperidine-1-carboxylate (200 mg, 29% yield) as a brown oil. 1 H NMR (400 MHz, CDCl 3 ): δ 10.01-9.88 (m, 1H), 8.44 (d, J = 2.8 Hz, 1H), 8.21-8.15 (m, 1H), 7.31-7.29 (m, 1H), 7.28-7.24 (m, 1H), 7.03 (d, J = 9.2 Hz, 1H), 6.80-6.73 (m, 1H), 6.66-6.53 (m, 2H), 6.40 (t, J = 2.8 Hz, 1H), 4.18-4.02 (m, 2H), 3.87-3.61 (m, 15H), 3.54-3.44 (m, 1H), 3.12-3.02 (m, 2H), 1.90-1.78 (m, 2H), 1.58-1.43 (m, 11H). [0400] Step 8: Preparation of tert-butyl 4-[2-[2-[2-[3-[4-amino-2-(6-methyl-7-oxo-1H- pyrrolo[2,3-c]pyridin-4-yl)phenoxy]phenoxy]ethoxy]ethoxy]eth oxy]piperidine-1-carboxylate. To a solution of tert-butyl 4-[2-[2-[2-[3-[2-(6-methyl-7-oxo-1H-pyrrolo[2,3-c]pyridin-4- yl)-4- nitro-phenox ]phenoxy]ethoxy]ethoxy]ethoxy]piperidine-1-carboxylate (200 mg, 289 ^mol, 1.0 equiv) in THF (2 mL) was added Pd/C (100 mg, 10% purity) under N 2 . The mixture was stirred under H2 (15 psi) at 25 ℃ for 2 hours. The mixture was filtered, and the filtrate was concentrated under reduced pressure to give tert-butyl 4-[2-[2-[2-[3-[4-amino-2-(6-methyl-7-oxo-1H- pyrrolo[2,3-c]pyridin-4-yl)phenoxy]phenoxy]ethoxy]ethoxy]eth oxy] piperidine-1-carboxylate (191 mg, crude) as a brown oil. 1 H NMR (400 MHz, CDCl3): δ 7.63-7.26 (m, 1H), 7.11-6.80 (m, 4H), 6.57-6.07 (m, 3H), 5.23 (s, 2H), 4.01-3.85 (m, 2H), 3.83-3.28 (m, 16H), 3.04-2.86 (m, 2H), 1.76- 1.65 (m, 2H), 1.46-1.30 (m, 14H). [0401] Step 9: Preparation of tert-butyl 4-[2-[2-[2-[3-[4-(ethylsulfonylamino)-2-(6-methyl- 7-oxo-1H-pyrrolo[2,3-c]pyridin-4-yl)phenoxy]phenoxy] ethoxy]ethoxy]ethoxy ]piperidine-1- carboxylate. To a solution of tert-butyl 4-[2-[2-[2-[3-[4-amino-2-(6-methyl-7-oxo-1H- pyrrolo[2,3-c]pyridin-4-yl)phenoxy]phenoxy]ethoxy]ethoxy]eth oxy]piperidine-1-carboxylate (100 mg, 151 ^mol, 1.0 equiv) and TEA (160 mg, 1.58 mmol, 10.5 equiv) in DCM (1 mL) was added ethanesulfonyl chloride (27 mg, 212 ^mol, 1.4 equiv) at 0 ℃. The mixture was stirred at 25 ℃ for 1 hour. The solution was washed with H 2 O (2 mL), and the organic layer was concentrated. The residue was purified by prep-TLC (SiO 2 , PE:EA = 0:1) to give tert-butyl 4-[2-[2-[2-[3-[4- (ethylsulfonylamino)-2-(6-methyl-7-oxo-1H-pyrrolo[2,3-c]pyri din-4-yl)phenoxy]phenoxy] ethoxy]ethoxy]ethoxy]piperidine-1-carboxylate (60 mg, 53% yield) as a brown oil. [0402] Step 10: Preparation of N-[3-(6-methyl-7-oxo-1H-pyrrolo[2,3-c]pyridin-4-yl)-4-[3- [2-[2-[2-(4-piperidyloxy)ethoxy]ethoxy]ethoxy]phenoxy]phenyl ]ethanesulfonamide. A solution of tert-butyl 4-[2-[2-[2-[3-[4-(ethylsulfonylamino)-2-(6-methyl-7-oxo-1H-p yrrolo[2,3- c]pyridin-4-yl)phenoxy]phenoxy]ethoxy]ethoxy]ethoxy]piperidi ne-1-carboxylate (60 mg, 79 ^mol, 1.0 equiv) in DCM (1.5 mL) and TFA (0.5 mL) was stirred at 25 ℃ for 1 hour. The solution was concentrated to give crude N-[3-(6-methyl-7-oxo-1H-pyrrolo[2,3-c]pyridin-4-yl)-4-[3-[2- [2- [2-(4-piperidyloxy)ethoxy]ethoxy]ethoxy]phenoxy]phenyl]ethan esulfonamide (61 mg, TFA salt) as a brown oil. [0403] Step 11: Preparation of N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]-6-[4-[2-[2-[2- [3-[4-(ethylsulfonylamino)-2-(6-methyl-7-oxo-1H-pyrrolo[2,3- c]pyridin-4-yl)phenoxy] phenoxy]ethoxy]ethoxy]ethoxy]-1-piperidyl]pyridazine-3-carbo xamide (I-2). A mixture of N- [3-(6-methyl-7-oxo-1H-pyrrolo[2,3-c]pyridin-4-yl)-4-[3-[2-[2 -[2-(4-piperidyloxy) ethoxy]ethoxy]ethoxy]phenoxy]phenyl]ethanesulfonamide (61 mg, 79 ^mol, 1.0 equiv, TFA salt), 6-chloro-N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]pyridazin e-3-carboxamide (34 mg, 87 ^mol, 1.1 equiv), and K2CO3 (55 mg, 396 ^mol, 5.0 equiv) in NMP (1 mL) was stirred at 60 ℃ for 3 hours. The mixture was purified by prep-HPLC (column: Waters Xbridge 150 x 25 mm, 5 ^m; mobile phase: [water(NH4HCO3)-ACN]; B%: 46%-76%,8min) to give N-[4-(3-chloro-4-cyano- phenoxy)cyclohexyl]-6-[4-[2-[2-[2-[3-[4-(ethylsulfonylamino) -2-(6-methyl-7-oxo-1H-pyrrolo[2,3- c]pyridin-4-yl)phenoxy]phenoxy]ethoxy]ethoxy]ethoxy]-1-piper idyl] pyridazine-3-carboxamide (8.8 mg, 11% yield) as a white solid. 1 H NMR (400 MHz, MeOD): δ 7.80 (d, J = 9.6 Hz, 1H), 7.69 (d, J = 8.8 Hz, 1H), 7.47 (d, J = 2.4 Hz, 1H), 7.35-7.25 (m, 2H), 7.23-7.18 (m, 2H), 7.16-7.00 (m, 4H), 6.56-6.48 (m, 1H), 6.42-6.31 (m, 3H), 4.58 (s, 2H), 4.05-3.91 (m, 4H), 3.80-3.73 (m, 2H), 3.70-3.61 (m, 7H), 3.58 (s, 3H), 3.50-3.38 (m, 2H), 3.22-3.09 (m, 3H), 2.24-2.14 (m, 2H), 2.11- 2.01 (m, 2H), 1.99-1.86 (m, 2H), 1.70-1.54 (m, 6H), 1.36 (t, J = 7.2 Hz, 3H). LC-MS: MS (ES + ): RT = 2.966 min, m/z = 505.0, 1009.2; LC-MS Method 10. EXAMPLE 3 – Synthesis of N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]-6-[4-[2-[2-[2-[2- [2- [3-[4-(ethylsulfonylamino)-2-(6-methyl-7-oxo-1H-pyrrolo[2,3- c]pyridin-4-yl)phenoxy] phenoxy]ethoxy]ethoxy]ethoxy]ethoxy]ethoxy]-1-piperidyl]pyri dazine-3-carboxamide (I-3) [0404] To a solution of N-[3-(6-methyl-7-oxo-1H-pyrrolo[2,3-c]pyridin-4-yl)-4-[3-[2- [2-[2-[2- [2-(4-piperidyloxy)ethoxy]ethoxy]ethoxy]ethoxy]ethoxy] phenoxy]phenyl]ethanesulfonamide (60 mg, 70 ^mol, 1.0 eq, TFA salt) in NMP (3 mL) was added K 2 CO 3 (97 mg, 700 ^mol, 10.0 eq) and 6-chloro-N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]pyridazin e-3-carboxamide (27 mg, 70 ^mol, 1.0 equiv). The mixture was stirred at 70 ℃ for 3 hours and purified by prep-HPLC (column: Unisil 3-100 C18 Ultra 150 x 50 mm, 3 ^m; mobile phase: [water(FA)-ACN]; B%: 42%- 62%, 5 min) to give N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]-6-[4-[2-[2-[2-[2- [2-[3-[4- (ethylsulfonylamino)-2-(6-methyl-7-oxo-1H-pyrrolo[2,3-c]pyri din-4-yl)phenoxy] phenoxy]ethoxy]ethoxy]ethoxy]ethoxy]ethoxy]-1-piperidyl]pyri dazine-3-carboxamide (25 mg, 32% yield) as an off-white solid. 1 H NMR (400 MHz, MeOD): δ 8.60-8.42 (m, 1H), 7.90 (s, 1H), 7.74-7.66 (m, 1H), 7.51-7.46 (m, 1H), 7.37-7.34 (m, 1H), 7.32-7.28 (m, 1H), 7.22 (t, J = 3.2 Hz, 3H), 7.12-7.03 (m, 3H), 6.56-6.51 (m, 1H), 6.43-6.34 (m, 3H), 4.55-4.45 (m, 1H), 4.09-4.02 (m, 2H), 4.00-3.94 (m, 3H), 3.77-3.74 (m, 2 H), 3.71-3.60 (m, 20H), 3.51-3.45 (m, 2H), 3.21-3.15 (m, 2H), 2.23-2.16 (m, 2H), 2.13-2.06 (m, 2H), 1.98-1.91 (m, 2H), 1.67-1.59 (m, 6H), 1.38 (t, J = 7.2 Hz, 3H). LC-MS: MS (ES + ): RT = 2.941 min, m/z = 549.2, 1097.3; LC-MS Method 10. EXAMPLE 4 – Synthesis of N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]-6-[4-[2-[2-[2-[2- [2- [2-[2-[3-[4-(ethylsulfonylamino)-2-(6-methyl-7-oxo-1H-pyrrol o[2,3-c]pyridin-4- yl)phenoxy]phenoxy]ethoxy]ethoxy]ethoxy]ethoxy]ethoxy]ethoxy ]ethoxy]-1- piperidyl]pyridazine-3-carboxamide (I-4)

[0405] Step 1: Preparation of tert-butyl 4-[2-[2-[2-[2-[2-[2-[2-(3-benzyloxyphenoxy) ethoxy]ethoxy]ethoxy]ethoxy]ethoxy]ethoxy]ethoxy]piperidine- 1-carboxylate. To a solution of tert-butyl 4-hydroxypiperidine-1-carboxylate (2 g, 9.96 mmol, 1 equiv) in THF (60 mL) was added NaH (478 mg, 12 mmol, 60% purity, 1.2 equiv), and the mixture was stirred at 0 ℃ for 0.5 hour. Then, 2-[2-[2-[2-[2-[2-[2-(3-benzyloxyphenoxy)ethoxy]ethoxy]ethoxy ]ethoxy] ethoxy]ethoxy]ethyl 4-methylbenzenesulfonate (6.6 g, 9.96 mmol, 1 equiv) was added at 0 ℃, and the mixture was stirred at 20 ℃ for 12 hours. The mixture was quenched by addition of NH 4 Cl (100 mL) and extracted with ethyl acetate (3 x 100mL). The combined organic layers were washed with brine, dried over anhydrous sodium sulfate, filtered, and concentrated. The residue was purified by prep-HPLC (column: Kromasil Eternity XT 250 x 80 mm, 10 ^m; mobile phase: [water(NH 4 HCO 3 )- ACN]; B%: 55%-85%, 20 min) to give tert-butyl 4-[2-[2-[2-[2-[2-[2-[2-(3- benzyloxyphenoxy)ethoxy]ethoxy]ethoxy]ethoxy]ethoxy]ethoxy] ethoxy]piperidine-1-carboxylate (4 g, 58% yield). [0406] Step 2: Preparation of tert-butyl 4-[2-[2-[2-[2-[2-[2-[2-(3-hydroxyphenoxy) ethoxy]ethoxy]ethoxy] ethoxy]ethoxy]ethoxy]ethoxy]piperidine-1-carboxylate. To a solution of tert-butyl 4-[2-[2-[2-[2-[2-[2-[2-(3-benzyloxyphenoxy)ethoxy]ethoxy]eth oxy]ethoxy] ethoxy]ethoxy]ethoxy]piperidine-1-carboxylate (4 g, 6 mmol, 1 equiv) in THF (80 mL) was added Pd(OH)2 (0.5 g, 10% purity) under N2 atmosphere. The mixture was stirred under H2 (15 Psi) at 20 ℃ for 3 hours. The mixture was filtered and concentrated. The residue was purified by prep-HPLC (FA condition; column: Phenomenex luna C18150 x 40 mm, 15 ^m; mobile phase: [water(FA)- ACN]; B%: 40%-60%, 10 min) to give tert-butyl 4-[2-[2-[2-[2-[2-[2-[2-(3- hydroxyphenoxy)ethoxy]ethoxy]ethoxy]ethoxy]ethoxy]ethoxy]eth oxy]piperidine-1-carboxylate (1.85 g, 53% yield). 1 H NMR (400 MHz, CDCl3) δ 7.07 (m, 1H), 6.53 (t, J = 2.2 Hz, 1H), 6.49 - 6.43 (m, 2H), 4.16 - 4.10 (m, 2H), 3.86 - 3.63 (m, 28H), 3.51 - 3.44 (m, 1H), 3.05 (m, 2H), 1.87 - 1.78 (m, 2H), 1.56 - 1.48 (m, 2H), 1.46 (s, 9H). [0407] Step 3: Preparation of tert-butyl 4-[2-[2-[2-[2-[2-[2-[2-[3-[2-[6-methyl-7-oxo-1-(p- tolylsulfonyl)pyrrolo[2,3-c] pyridin-4-yl]-4-nitro-phenoxy]phenoxy]ethoxy]ethoxy] ethoxy]ethoxy]ethoxy]ethoxy]ethoxy]piperidine-1-carboxylate. To a solution of tert-butyl 4-[2- [2-[2-[2-[2-[2-[2-(3-hydroxyphenoxy)ethoxy]ethoxy]ethoxy]eth oxy]ethoxy]ethoxy] ethoxy]piperidine-1-carboxylate (400 mg, 665 ^mol, 1 equiv) and 4-(2-fluoro-5-nitro-phenyl)-6- methyl-1-(p-tolylsulfonyl)pyrrolo[2,3-c]pyridin-7-one (294 mg, 665 ^mol, 1 equiv) in NMP (6 mL) was added Cs 2 CO 3 (542 mg, 1.66 mmol, 2.5 equiv). The mixture was stirred at 70 ℃ for 3 hours and concentrated. The mixture was partitioned between water (50 mL), extracted with ethyl acetate (3 x 40 mL), dried, filtered, and concentrated. The residue was purified by prep-TLC (SiO2, EA:MeOH = 10:1) to give tert-butyl 4-[2-[2-[2-[2-[2-[2-[2-[3-[2-[6-methyl-7-oxo-1-(p- tolylsulfonyl)pyrrolo[2,3-c] pyridin-4-yl]-4-nitro-phenoxy]phenoxy]ethoxy]ethoxy]ethoxy] ethoxy]ethoxy]ethoxy]ethoxy]piperidine-1-carboxylate (647 mg, 54% yield). [0408] Step 4: Preparation of tert-butyl 4-[2-[2-[2-[2-[2-[2-[2-[3-[4-amino-2-[6-methyl-7- oxo-1-(p-tolylsulfonyl)pyrrolo[2,3-c]pyridin-4-yl]phenoxy]ph enoxy]ethoxy]ethoxy]ethoxy] ethoxy]ethoxy]ethoxy]ethoxy]piperidine-1-carboxylate. To a solution of tert-butyl 4-[2-[2-[2- [2-[2-[2-[2-[3-[2-[6-methyl-7-oxo-1-(p-tolylsulfonyl)pyrrolo [2,3-c] pyridin-4-yl]-4-nitro- phenoxy]phenoxy]ethoxy]ethoxy]ethoxy] ethoxy]ethoxy]ethoxy]ethoxy]piperidine-1-carboxylate (647 mg, 361 ^mol, 1 equiv) in EtOH (10 mL) and H 2 O (5 mL) was added Fe (201 mg, 3.6 mmol, 10 equiv) and NH 4 Cl (193 mg, 3.6 mmol, 10 equiv). The mixture was stirred at 85 ℃ for 3 hours and concentrated. The residue was purified by prep-TLC (SiO 2 , EA:MeOH = 10:1) to give tert- butyl 4-[2-[2-[2-[2-[2-[2-[2-[3-[4-amino-2-[6-methyl-7-oxo-1-(p-to lylsulfonyl)pyrrolo[2,3- c]pyridin-4-yl]phenoxy]phenoxy]ethoxy]ethoxy]ethoxy]ethoxy] ethoxy]ethoxy]ethoxy]piperidine- 1-carboxylate (611 mg, 85% yield). [0409] Step 5: Preparation of tert-butyl 4-[2-[2-[2-[2-[2-[2-[2-[3-[4-(ethylsulfonylamino)-2- [6-methyl-7-oxo-1-(p-tolylsulfonyl)pyrrolo[2,3-c]pyridin-4-y l]phenoxy]phenoxy]ethoxy] ethoxy]ethoxy]ethoxy]ethoxy]ethoxy]ethoxy]piperidine-1-carbo xylate. To a solution of tert- butyl 4-[2-[2-[2-[2-[2-[2-[2-[3-[4-amino-2-[6-methyl-7-oxo-1-(p-to lylsulfonyl)pyrrolo [2,3- c]pyridin-4-yl]phenoxy]phenoxy]ethoxy]ethoxy]ethoxy]ethoxy]e thoxy]ethoxy]ethoxy] piperidine- 1-carboxylate (511 mg, 515 ^mol, 1 equiv) in DCM (7 mL) was added Et 3 N (1.04 g, 10 mmol, 20 equiv) and ethanesulfonyl chloride (133 mg, 1.03 mmol, 2 equiv). The mixture was stirred at 25 ℃ for 12 hours. The mixture was partitioned between water (50 mL), extracted with ethyl acetate (3 x 40 mL), dried over anhydrous sodium sulfate, filtered, and concentrated. The residue was purified by prep-TLC (SiO2, EA:MeOH = 10:1) to give tert-butyl 4-[2-[2-[2-[2-[2-[2-[2-[3-[4- (ethylsulfonylamino)-2-[6-methyl-7-oxo-1-(p-tolylsulfonyl)py rrolo[2,3-c]pyridin-4- yl]phenoxy]phenoxy]ethoxy]ethoxy]ethoxy] ethoxy]ethoxy]ethoxy]ethoxy]piperidine-1- carboxylate (170 mg, 31% yield). [0410] Step 6: Preparation of N-[3-(6-methyl-7-oxo-1H-pyrrolo[2,3-c]pyridin-4-yl)-4-[3-[2- [2-[2-[2-[2-[2-[2-(4-piperidyloxy)ethoxy]ethoxy]ethoxy]ethox y]ethoxy]ethoxy]ethoxy] phenoxy]phenyl]ethanesulfonamide. To a solution of tert-butyl 4-[2-[2-[2-[2-[2-[2-[2-[3-[4- (ethylsulfonylamino)-2-(6-methyl-7-oxo-1H-pyrrolo [2,3-c]pyridin-4-yl)phenoxy]phenoxy] ethoxy]ethoxy]ethoxy]ethoxy]ethoxy]ethoxy]ethoxy]piperidine- 1-carboxylate (107 mg, 115 ^mol, 1 equiv) in DCM (5 mL) was added TFA (2 mL). The mixture was stirred at 25 ℃ for 0.5 hour and concentrated to give N-[3-(6-methyl-7-oxo-1H-pyrrolo[2,3-c]pyridin-4-yl)-4-[3-[2- [2-[2-[2-[2-[2- [2-(4-piperidyloxy)ethoxy]ethoxy]ethoxy]ethoxy]ethoxy]ethoxy ]ethoxy]phenoxy] phenyl]ethanesulfonamide (108 mg, 99% yield). [0411] Step 7: Preparation of N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]-6-[4-[2-[2-[2-[2- [2-[2-[2-[3-[4-(ethylsulfonylamino)-2-(6-methyl-7-oxo-1H-pyr rolo[2,3-c]pyridin-4- yl)phenoxy]phenoxy]ethoxy]ethoxy]ethoxy]ethoxy]ethoxy]ethoxy ]ethoxy]-1- piperidyl]pyridazine-3-carboxamide (I-4). To a solution of N-[3-(6-methyl-7-oxo-1H- pyrrolo[2,3-c]pyridin-4-yl)-4-[3-[2-[2-[2-[2-[2-[2-[2-(4-pip eridyloxy)ethoxy]ethoxy] ethoxy]ethoxy]ethoxy]ethoxy]ethoxy]phenoxy]phenyl]ethanesulf onamide (108 mg, 114 ^mol, 1 equiv, TFA) and 6-chloro-N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl] pyridazine-3-carboxamide (49 mg, 126 ^mol, 1.1 equiv) in NMP (1.5 mL) was added K2CO3 (158 mg, 1.14 mmol, 10 equiv). The mixture was stirred at 50 ℃ for 3 hours and purified by prep-HPLC (FA condition; column: Waters Xbridge 150 x 25mm, 5 ^m; mobile phase: [water(NH4HCO3)-ACN]; B%: 41%-71%, 9 min) to give N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]-6-[4-[2-[2-[2-[2- [2-[2-[2-[3-[4- (ethylsulfonylamino)-2-(6-methyl-7-oxo-1H-pyrrolo[2,3-c]pyri din-4- yl)phenoxy]phenoxy]ethoxy]ethoxy]ethoxy]ethoxy]ethoxy] ethoxy]ethoxy]-1-piperidyl] pyridazine-3-carboxamide (I-4) (13 mg, 10% yield). 1 H NMR (400 MHz, CDCl3): δ 9.99 - 9.89 (m, 1H), 8.01 - 7.94 (m, 2H), 7.58 (d, J = 8.8 Hz, 1H), 7.53 - 7.46 (m, 2H), 7.30 (d, J = 2.8 Hz, 1H), 7.25 (t, J = 2.8 Hz, 1H), 7.13 (t, J = 8.4 Hz, 1H), 7.09 - 7.05 (m, 2H), 7.03 (d, J = 2.4 Hz, 1H), 6.98 (d, J = 9.6 Hz, 1H), 6.88 (m, 1H), 6.56 (m, 1H), 6.48 (m, 1H), 6.41 (m, 2H), 4.37 - 4.31 (m, 1H), 4.09 - 3.99 (m, 5H), 3.81 - 3.77 (m, 2H), 3.68 - 3.63 (m, 25H), 3.60 (s, 3H), 3.47 (m, 2H), 3.22 (m, 2H), 2.23 - 2.16 (m, 4H), 1.96 - 1.88 (m, 2H), 1.76 - 1.67 (m, 4H), 1.56 - 1.49 (m, 2H), 1.45 (t, J = 7.6 Hz, 3H). LC-MS: MS (ES + ): RT = 2.93 min, m/z = 593.2, 1185.3; LC-MS Method 10. EXAMPLE 5 – Synthesis of N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]-2-[4-[2-[3-[4- (ethylsulfonylamino)-2-(6-methyl-7-oxo-1H-pyrrolo[2,3-c]pyri din-4-yl)phenoxy]phenoxy] ethoxy]-1-piperidyl]pyrimidine-5-carboxamide (I-5) [0412] To a solution of N-[3-(6-methyl-7-oxo-1H-pyrrolo[2,3-c]pyridin-4-yl)-4-[3-[2- (4- piperidyloxy)ethoxy]phenoxy]phenyl]ethanesulfonamide (30 mg, 51 ^mol, 1.0 equiv) and 2- chloro-N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]pyrimidine- 5-carboxamide (20 mg, 51 ^mol, 1.0 equiv) in NMP (2 mL) was added K2CO3 (21 mg, 154 ^mol, 3.0 equiv). The mixture was stirred at 50 ℃ for 2 hours and purified by prep-HPLC (column: Unisil 3-100 C18 Ultra 150 x 50 mm, 3 ^m; mobile phase: [water(FA)-ACN]; B%: 48%-78%, 7 min) to give N-[4-(3-chloro-4- cyano-phenoxy)cyclohexyl]-2-[4-[2-[3-[4-(ethylsulfonylamino) -2-(6-methyl-7-oxo-1H- pyrrolo[2,3-c]pyridin-4-yl)phenoxy]phenoxy]ethoxy]-1-piperid yl]pyrimidine-5-carboxamide (I-5) (18 mg, 33% yield) as a white solid. 1 H NMR (400 MHz, MeOD): δ 8.72 (s, 2H), 7.69 (d, J = 8.8 Hz, 1H), 7.47 (d, J = 2.6 Hz, 1H), 7.34-7.31 (m, 1H), 7.28 (d, J = 8.6 Hz, 1H), 7.22-7.17 (m, 2H), 7.12-7.01 (m, 3H), 6.53 (d, J = 8.0 Hz, 1H), 6.41-6.32 (m, 3H), 4.51-4.42 (m, 1H), 4.23 (d, J = 13.2 Hz, 2H), 4.01-3.96 (m, 2H), 3.95-3.87 (m, 1H), 3.82-3.75 (m, 2H), 3.68 (t, J = 7.6 Hz, 1H), 3.61-3.54 (m, 4H), 3.15 (d, J = 7.2 Hz, 2H), 2.20 (d, J = 8.4 Hz, 2H), 2.10-2.03 (m, 2H), 1.99 (d, J = 9.8 Hz, 2H), 1.63-1.52 (m, 6H), 1.35 (t, J = 7.4 Hz, 3H). LC-MS: MS (ES + ): RT = 3.164 min, m/z = 921.2, 461.1; LC-MS Method 10. EXAMPLE 6 – Synthesis of N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]-4-[4-[2-[3-[4- (ethylsulfonylamino)-2-(6-methyl-7-oxo-1H-pyrrolo[2,3-c]pyri din-4-yl)phenoxy] phenoxy]ethoxy]-1-piperidyl]benzamide (I-6) [0413] To a solution of 4-[4-[2-[3-[4-(ethylsulfonylamino)-2-(6-methyl-7-oxo-1H-pyrr olo[2,3- c]pyridin-4-yl)phenoxy]phenoxy]ethoxy]-1-piperidyl]benzoic acid (53 mg, 66 ^mol, 1.0 equiv) in DMF (1 mL) was added DIEA (43 mg, 331 ^mol, 5.0 equiv), 4-(4-aminocyclo-hexoxy)-2-chloro- benzonitrile (24 mg, 66 ^mol, 1.0 equiv, TFA salt), and HATU (33 mg, 86 ^mol, 1.3 equiv). The mixture was stirred at 25 ℃ for 0.5 hour and concentrated. The residue was purified by prep- HPLC (column: Phenomenex C1875 x 30 mm, 3 ^m; mobile phase: [water(FA)-ACN]; B%: 55%- 85%, 7 min) to give N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]-4-[4-[2-[3-[4- (ethylsulfonylamino)-2-(6-methyl-7-oxo-1H-pyrrolo[2,3-c]pyri din-4-yl)phenoxy] phenoxy]ethoxy]-1-piperidyl]benzamide (I-8) (27 mg, 44% yield) as a white solid. 1 H NMR (400 MHz, CDCl3): δ 10.36-10.51 (m, 1 H), 7.83-8.05 (m, 1 H), 7.66 (d, J = 8.6 Hz, 2 H), 7.55 (d, J = 8.6 Hz, 1 H), 7.50 (s, 1 H), 7.26 (d, J = 2.4 Hz, 1 H), 7.23 (t, J = 2.8 Hz, 1 H), 7.12 (t, J = 8.2 Hz, 1 H), 7.01-7.06 (m, 2 H), 6.99 (d, J = 2.2 Hz, 1 H), 6.86-6.96 (m, 2 H), 6.83-6.86 (m, 1 H), 6.54-6.57 (m, 1 H), 6.46-6.48 (m, 1 H), 6.39-6.42 (m, 2 H), 6.06-6.20 (m, 1 H), 4.25-4.30 (m, 1 H), 4.01-4.08 (m, 3 H), 3.75-3.80 (m, 2 H), 3.56 (s, 3 H), 3.50-3.54 (m, 1 H), 3.12-3.22 (m, 2 H), 3.04-3.08 (m, 2 H), 2.12-2.22 (m, 4 H), 1.96-2.04 (m, 2 H), 1.60-1.75 (m, 6 H), 1.43 (t, J = 7.4 Hz, 5 H). LC-MS: MS (ES + ): RT = 2.764 min, m/z = 460.4, 919.3; LC-MS Method 25. EXAMPLE 7 – Synthesis of N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]-6-[4-[2-[3-[4- (ethylsulfonylamino)-2-(6-methyl-7-oxo-1H-pyrrolo[2,3-c]pyri din-4-yl)phenoxy] phenoxy]ethoxy]-1-piperidyl]pyridine-3-carboxamide (I-7) [0414] To a solution of 6-[4-[2-[3-[4-(ethylsulfonylamino)-2-(6-methyl-7-oxo-1H-pyrr olo[2,3- c]pyridin-4-yl)phenoxy]phenoxy]ethoxy]-1-piperidyl]pyridine- 3-carboxylic acid (23 mg, 33 ^mol, 1.0 equiv) in DMF (1 mL) was added HATU (16.5 mg, 44 ^mol, 1.3 equiv), DIEA (22 mg, 167 ^mol, 5.0 equiv), and 4-(4-aminocyclohexoxy)-2-chloro-benzonitrile (9 mg, 33 ^mol, 1.0 equiv). The mixture was stirred at 25 ℃ for 0.5 hour and concentrated. The residue was purified by prep- HPLC (column: Phenomenex C1875 x 30 mm, 3 ^m; mobile phase: [water (FA)-ACN]; B%: 35%-65%, 7 min) and (column: Phenomenex C1875 x 30 mm, 3 ^m; mobile phase: [water(FA)- ACN]; B%: 40%-70%, 7 min) to give N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]-6-[4-[2-[3-[4- (ethylsulfonylamino)-2-(6-methyl-7-oxo-1H-pyrrolo[2,3-c]pyri din-4- yl)phenoxy]phenoxy]ethoxy]-1-piperidyl]pyridine-3-carboxamid e (I-6) (20 mg, 64% yield). 1 H NMR (400 MHz, DMSO-d6) δ 12.05 (s, 1H), 9.82 (s, 1H), 8.58 (s, 1H), 8.16- 8.02 (m, 1H), 7.97 (d, J = 9.2 Hz, 1H), 7.87 (d, J = 8.8 Hz, 1H), 7.40 (m, 2H), 7.33- 7.19 (m, 3H), 7.17- 7.11 (m, 2H), 7.06 (d, J = 8.8 Hz, 1H), 6.90 (m, 1H), 6.59 (m, 1H), 6.44- 6.37 (m, 2H), 6.27 (s, 1H), 4.61- 4.47 (m, 1H), 4.08- 3.89 (m, 4H), 3.84 - 3.76 (m, 1H), 3.73 (m, 2H), 3.62 (m, 1H), 3.49 (s, 3H), 3.31- 3.21 (m, 2H), 3.13 (m, 2H), 2.10 (m, 2H), 1.95- 1.84 (m, 5H), 1.56- 1.48 (m, 3H), 1.45- 1.37 (m, 2H), 1.24 (m, 3H). LC-MS: MS (ES + ): RT = 2.413 min, m/z = 460.7, 920.3; LC-MS Method 25. EXAMPLE 8 – Synthesis of N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]-5-[4-[2-[3-[4- (ethylsulfonylamino)-2-(6-methyl-7-oxo-1H-pyrrolo[2,3-c]pyri din-4-yl)phenoxy] phenoxy]ethoxy]-1-piperidyl]pyrazine-2-carboxamide (I-8) [0415] To a solution of 5-[4-[2-[3-[4-(ethylsulfonylamino)-2-(6-methyl-7-oxo-1H-pyrr olo[2,3- c]pyridin-4-yl)phenoxy]phenoxy]ethoxy]-1-piperidyl]pyrazine- 2-carboxylic acid (60 mg, 74 ^mol, 1.0 equiv, TFA salt) and 4-(4-aminocyclohexoxy)-2-chloro-benzonitrile (18 mg, 74 ^mol, 1.0 equiv) in DMF (1 mL) was added DIEA (48 mg, 373 ^mol, 65 μL, 5.0 equiv) and HATU (36 mg, 97 ^mol, 1.3 equiv). The mixture was stirred at 25 ℃ for 1 hour and concentrated. The residue was purified by prep-HPLC (column: Phenomenex C1875 x 30mm, 3 ^m; mobile phase: [water(FA)- ACN]; B%: 55%-85%, 7 min) to give N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]-5-[4-[2-[3-[4- (ethylsulfonylamino)-2-(6-methyl-7-oxo-1H-pyrrolo[2,3-c]pyri din-4- yl)phenoxy]phenoxy]ethoxy]-1-piperidyl]pyrazine-2-carboxamid e (I-9) (49 mg, 70% yield). 1 H NMR (400 MHz, CDCl 3 ) δ 10.38-10.10 (m, 1H), 8.81 (s, 1H), 7.97 (s, 1H), 7.56 (d, J = 8.4 Hz, 1H), 7.51-7.45 (m, 1H), 7.44-7.35 (m, 2H), 7.31-7.28 (m, 1H), 7.27-7.22 (m, 1H), 7.16-7.03 (m, 3H), 7.00 (d, J = 2.0 Hz, 1H), 6.90-6.81 (m, 1H), 6.63-6.52 (m, 1H), 6.50-6.36 (m, 3H), 4.38-4.25 (m, 1H), 4.11-3.91 (m, 5H), 3.85-3.78 (m, 2H), 3.73-3.66 (m, 1H), 3.62 (s, 3H), 3.56-3.44 (m, 2H), 3.20 (q, J = 7.2 Hz, 2H), 2.30-2.05(m, 5H), 2.05-1.84 (m, 5H), 1.54-1.41 (m, 5H). LC-MS: MS (ES + ): RT = 2.982 min, m/z = 921.3; LC-MS Method 25. EXAMPLE 9 – Synthesis of N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]-5-[4-[2-[3-[4- (ethylsulfonylamino)-2-(6-methyl-7-oxo-1H-pyrrolo[2,3-c]pyri din-4-yl)phenoxy] phenoxy]ethoxy]-1-piperidyl]pyridine-2-carboxamide (I-9) [0416] To a solution of 5-[4-[2-[3-[4-(ethylsulfonylamino)-2-(6-methyl-7-oxo-1H-pyrr olo[2,3- c]pyridin-4-yl)phenoxy]phenoxy]ethoxy]-1-piperidyl]pyridine- 2-carboxylic acid (50 mg, 62 ^mol, 1.0 equiv,) in DMF (1 mL) was added DIEA (24 mg, 187 ^mol, 3.0 equiv), 4-(4- aminocyclohexoxy)-2-chloro-benzonitrile (23 mg, 63 ^mol, 1.0 equiv, TFA salt), and HATU (31 mg, 81 ^mol, 1.3 equiv). The mixture was stirred at 20 ℃ for 0.5 hour and concentrated. The residue was purified by prep-HPLC (column: Phenomenex C1875 x 30mm, 3 ^m; mobile phase: [water(FA)-ACN]; B%: 52%-82%, 7 min) to give N-[4-(3-chloro-4-cyano-phenoxy) cyclohexyl]- 5-[4-[2-[3-[4-(ethylsulfonylamino)-2-(6-methyl-7-oxo-1H-pyrr olo[2,3-c]pyridin-4- yl)phenoxy]phenoxy]ethoxy]-1-piperidyl]pyridine-2-carboxamid e (I-7) (35 mg, 61% yield). 1 H NMR (400 MHz, DMSO-d6): δ 12.06 (s, 1H), 9.83 (s, 1H), 8.32-8.23 (m, 1H), 8.18 (d, J = 8.2 Hz, 1H), 7.91-7.79 (m, 2H), 7.44-7.35 (m, 3H), 7.32-7.25 (m, 2H), 7.25-7.19 (m, 1H), 7.17-7.09 (m, 2H), 7.08-7.02 (m, 1H), 6.63-6.53 (m, 1H), 6.45-6.35 (m, 2H), 6.26 (t, J = 2.0 Hz, 1H), 4.60-4.45 (m, 1H), 4.03-3.92 (m, 2H), 3.87-3.77 (m, 1H), 3.73-3.61 (m, 4H), 3.60-3.54 (m, 1H), 3.49 (s, 3H), 3.17-3.02 (m, 4H), 2.13-2.04 (m, 2H), 1.96-1.84 (m, 4H), 1.65-1.45 (m, 6H), 1.27-1.18 (m, 3H). LC-MS: MS (ES + ): RT = 2.86 min, m/z = 460.8, 920.3; LC-MS Method 25. EXAMPLE 10 – Synthesis of N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]-5-[4-[2-[3-[4- (ethylsulfonylamino)-2-(6-methyl-7-oxo-1H-pyrrolo[2,3-c]pyri din-4-yl)phenoxy] phenoxy]ethoxy]-1-piperidyl]pyrimidine-2-carboxamide (I-10) [0417] To a solution of 5-[4-[2-[3-[4-(ethylsulfonylamino)-2-(6-methyl-7-oxo-1H-pyrr olo[2,3- c]pyridin-4-yl)phenoxy]phenoxy]ethoxy]-1-piperidyl]pyrimidin e-2-carboxylic acid (40 mg, 50 ^mol, 1.0 equiv) in DMF (1 mL) was added DIEA (32 mg, 249 ^mol, 5.0 equiv), 4-(4- aminocyclohexoxy)-2-chloro-benzonitrile (18 mg, 50 ^mol, 1.0 equiv, TFA salt), and HATU (25 mg, 65 ^mol, 1.3 equiv). The mixture was stirred at 25 ℃ for 0.5 hour and concentrated. The residue was purified by prep-HPLC (column: Phenomenex C1875 x 30 mm, 3 ^m; mobile phase: [water(FA)-ACN]; B%: 50%-80%, 7 min) to give N-[4-(3-chloro-4-cyano-phenoxy) cyclohexyl]- 5-[4-[2-[3-[4-(ethylsulfonylamino)-2-(6-methyl-7-oxo-1H-pyrr olo[2,3-c]pyridin-4- yl)phenoxy]phenoxy]ethoxy]-1-piperidyl]pyrimidine-2-carboxam ide (I-10) (17 mg, 37% yield) as a yellow solid. 1 H NMR (400 MHz, CDCl 3 ): δ 10.73 (s, 1 H), 8.33 (s, 3 H), 7.75 (d, J = 8.4 Hz, 1 H), 7.54-7.57 (m, 2 H), 7.30-7.33 (m, 1 H), 7.25-7.27 (m, 1 H), 7.03-7.13 (m, 3 H), 6.99 (d, J = 2.4 Hz, 1 H), 6.84-6.86 (m, 1 H), 6.52-6.54 (m, 1 H), 6.45-6.47 (m, 1 H), 6.40 (t, J = 2.4 Hz, 1 H), 6.36 (t, J = 2.2 Hz, 1 H), 4.27-4.34 (m, 1 H), 4.05-4.13 (m, 1 H), 3.99-4.03 (m, 2 H), 3.74-3.78 (m, 2 H), 3.61-3.67 (m, 1 H), 3.51-3.56 (m, 2 H), 3.19-3.25 (m, 4 H), 2.13-2.23 (m, 5 H), 1.91-1.97 (m, 2 H), 1.61-1.83 (m, 5 H), 1.40-1.54 (m, 6 H). LC-MS: MS (ES + ): RT = 2.862 min, m/z = 461.2, 921.3; LC-MS Method 25. EXAMPLE 11 – Synthesis of Additional Compounds [0418] The following additional compounds were prepared based on procedures described herein: I-11, I-12, I-13, I-14, I-15, I-16, I-17, I-18, I-19, I-20, I-21, I-22, I-23, I-24, I-25, I-26, I-27, I-28, I-29, I-30, I-31, I-32, I-33, I-34, I-35, and I-36. EXAMPLE 12 – Synthesis of N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]-6-[4-[3-[3-[4- (ethylsulfonylamino)-2-(6-methyl-7-oxo-1H-pyrrolo[2,3-c]pyri din-4-yl)phenoxy]-2-fluoro- phenyl]propoxy]-1-piperidyl]pyridazine-3-carboxamide (I-37)

[0419] Step 1: Preparation of tert-butyl 4-[3-(2-fluoro-3-tetrahydropyran-2-yloxy- phenyl)prop-2-ynoxy]piperidine-1-carboxylate. To a solution of tert-butyl 4-prop-2- ynoxypiperidine-1-carboxylate (4.00 g, 16.7 mmol, 1.5 equiv),2-(3-bromo-2-fluoro- phenoxy)tetrahydropyran (3.10 g, 11.3 mmol, 1.0 equiv) in DMF (45.0 mL) and MeCN (15.0 mL) was added [2-(2-aminophenyl)phenyl]palladium(1+);bis(1-adamantyl)-buty l- phosphane;methanesulfonate (800 mg, 1.10 mmol, 0.1 equiv) and Cs 2 CO 3 (9.10 g, 27.9 mmol, 2.5 equiv) .The mixture was stirred at 90 °C for 2 h. LC-MS showed desired compound was detected. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by column chomatography (SiO 2 , Petroleum ether/Ethyl acetate=6/1 to 1/1). Compound tert-butyl 4-[3-(2-fluoro-3-tetrahydropyran-2-yloxy-phenyl)prop-2-ynoxy ]piperidine-1-carboxylate (3.30 g, 7.61 mmol, 68% yield) was obtained as a yellow oil. [0420] Step 2: Preparation of tert-butyl 4-[3-(2-fluoro-3-tetrahydropyran-2-yloxy- phenyl)propoxy]piperidine-1-carboxylate. To Pd/C (3.10 g, 10% purity) was added into a 100 mL single-necked round bottom flask under N 2 , and then CF3CH 2 OH (30.0 mL) was added at 25°C under N 2 . After addition, tert-butyl 4-[3-(2-fluoro-3-tetrahydropyran-2-yloxy-phenyl)prop-2- ynoxy]piperidine-1-carboxylate (3.10 g, 7.15 mmol, 1.0 equiv) was added under N2. The suspension was degassed under vacuum and purged with H2 several times. The mixture was stirred under H 2 (15 psi) at 25°C for 2 h. LC-M showed compound was detected. The reaction mixture was filtered and the filter was concentrated. The residue was purified by column chomatography (SiO 2 , Petroleum ether/Ethyl acetate=5:1 to 1:1). Compound tert-butyl 4-[3-(2-fluoro-3- tetrahydropyran-2-yloxy-phenyl)propoxy]piperidine-1-carboxyl ate (2.00 g, 4.11 mmol, 58% yield, 90% purity) was obtained as a yellow oil. [0421] Step 3: Preparation of tert-butyl 4-[3-(2-fluoro-3-hydroxy-phenyl)propoxy] piperidine-1-carboxylate. To a solution of tert-butyl 4-[3-(2-fluoro-3-tetrahydropyran-2-yloxy- phenyl)propoxy]piperidine-1-carboxylate (2.00 g, 4.57 mmol, 1.0 equiv) in MeOH (30.0 mL) was added TsOH (1.05 g, 6.11 mmol, 1.3 equiv). The mixture was stirred at 25°C for 12 h. LC-MS showed desired compound was detected. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-TLC (SiO 2 , Petroleum ether/Ethyl acetate=3/1). Compound tert-butyl 4-[3-(2-fluoro-3-hydroxy-phenyl)propoxy]piperidine-1- carboxylate (1.10 g, 2.80 mmol, 61% yield, 90% purity) was obtained as a yellow oil. [0422] Step 4: Preparation of tert-butyl 4-[3-[2-fluoro-3-[2-(6-methyl-7-oxo-1H- pyrrolo[2,3-c]pyridin-4-yl)-4-nitro-phenoxy]phenyl]propoxy]p iperidine-1-carboxylate. To a solution of tert-butyl 4-[3-(2-fluoro-3-hydroxy-phenyl)propoxy]piperidine-1-carboxy late (200 mg, 566 μmol, 1.0 equiv) and 4-(2-fluoro-5-nitro-phenyl)-6-methyl-1-(p-tolylsulfonyl) pyrrolo[2,3- c]pyridin-7-one (252 mg, 571 μmol, 1.0 equiv) in DMSO (4.00 mL) was added Cs 2 CO 3 (461 mg, 1.41 mmol, 2.5 equiv). The mixture was stirred at 100 °C for 12 h. LC-MS showed desired compound was detected. The reaction mixture was diluted with water 20.0 mL and extracted with EA 90.0 mL (30.0 mL x 3). The combined organic layers were washed with brine 90.0 mL (30.0 mL x 3), dried over [Na 2 SO 4 ], filtered and concentrated under reduced pressure to give a residue. The residue was purified or by prep-TLC (SiO 2 , DCM: MeOH = 10:1). Compound tert-butyl 4-[3- [2-fluoro-3-[2-(6-methyl-7-oxo-1H-pyrrolo[2,3-c]pyridin-4-yl )-4-nitro- phenoxy]phenyl]propoxy]piperidine-1-carboxylate (100 mg, 161 μmol, 28% yield) was obtained as a yellow solid. [0423] Step 5: Preparation of tert-butyl 4-[3-[3-[4-amino-2-(6-methyl-7-oxo-1H- pyrrolo[2,3-c]pyridin-4-yl)phenoxy]-2-fluoro-phenyl]propoxy] piperidine-1-carboxylate. To a solution of tert-butyl 4-[3-[2-fluoro-3-[2-(6-methyl-7-oxo-1H-pyrrolo[2,3-c]pyridin -4-yl)-4-nitro- phenoxy]phenyl]propoxy]piperidine-1-carboxylate (120 mg, 193 μmol, 1.0 equiv) in EtOH (20.0 mL) and H 2 O (10.0 mL) was added Fe (54.0 mg, 967 μmol, 5.0 equiv) and NH 4 Cl (51.7 mg, 967 μmol, 5.0 equiv). The mixture was stirred at 80°C for 12 h. LC-MS showed desired compound was detected. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-TLC (SiO 2 , DCM: MeOH =10:1). Compound tert-butyl 4-[3-[3-[4-amino-2-(6-methyl-7-oxo-1H-pyrrolo[2,3-c]pyridin- 4-yl)phenoxy]-2-fluoro- phenyl]propoxy]piperidine-1-carboxylate (50.0 mg, 84.7 μmol, 44% yield) was obtained as a yellow solid. [0424] Step 6: Preparation of tert-butyl 4-[3-[3-[4-[bis(ethylsulfonyl)amino]-2-(6-methyl- 7-oxo-1H-pyrrolo[2,3-c]pyridin-4-yl)phenoxy]-2-fluoro-phenyl ]propoxy]piperidine-1- carboxylate. To a solution of tert-butyl 4-[3-[3-[4-amino-2-(6-methyl-7-oxo-1H-pyrrolo[2,3- c]pyridin-4-yl)phenoxy]-2-fluoro-phenyl]propoxy]piperidine-1 -carboxylate (50.0 mg, 84.7 μmol, 1.0 equiv) and ethanesulfonyl chloride (21.8 mg, 169 μmol, 16.0 μL, 2.0 equiv) in DCM (2.00 mL) was added TEA (85.7 mg, 847 μmol, 118 μL, 10 equiv). The mixture was stirred at 25 °C for 12 h. LC-MS showed desired compound was detected. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-TLC (SiO 2 , DCM: MeOH =10:1). Compound tert-butyl 4-[3-[3-[4-[bis(ethylsulfonyl)amino]-2-(6-methyl-7-oxo-1H- pyrrolo[2,3-c]pyridin-4-yl)phenoxy]-2-fluoro-phenyl]propoxy] piperidine-1-carboxylate (60.0 mg, 77.4 μmol, 91% yield) was obtained as a yellow oil. [0425] Step 7: Preparation of tert-butyl 4-[3-[3-[4-(ethylsulfonylamino)-2-(6-methyl-7-oxo- 1H-pyrrolo[2,3-c]pyridin-4-yl)phenoxy]-2-fluoro-phenyl]propo xy]piperidine-1-carboxylate. To a solution of tert-butyl 4-[3-[3-[4-[bis(ethylsulfonyl)amino]-2-(6-methyl-7-oxo-1H-py rrolo[2,3- c]pyridin-4-yl)phenoxy]-2-fluoro-phenyl]propoxy]piperidine-1 -carboxylate (60.0 mg, 77.4 μmol, 1.0 equiv) in dioxane (2.00 mL) was added NaOH (30.9 mg, 774 μmol, 10 equiv), H 2 O (2.00 mL). The mixture was stirred at 80°C for 2 h. LC-MS showed desired compound was detected. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-TLC (SiO 2 , DCM: MeOH =10:1). Compound tert-butyl 4-[3-[3-[4- (ethylsulfonylamino)-2-(6-methyl-7-oxo-1H-pyrrolo[2,3-c]pyri din-4-yl)phenoxy]-2-fluoro- phenyl]propoxy]piperidine-1-carboxylate (35.0 mg, 51.3 μmol, 66 % yield) was obtained as a yellow oil. [0426] Step 8: Preparation of N-[4-[2-fluoro-3-[3-(4-piperidyloxy)propyl]phenoxy]-3-(6- methyl-7-oxo-1H-pyrrolo[2,3-c]pyridin-4-yl)phenyl]ethanesulf onamide. To a solution of tert- butyl 4-[3-[3-[4-(ethylsulfonylamino)-2-(6-methyl-7-oxo-1H-pyrrolo [2,3-c]pyridin-4-yl)phenoxy]- 2-fluoro-phenyl]propoxy]piperidine-1-carboxylate (35.0 mg, 51.3 μmol, 1.0 equiv) in DCM (2.00 mL) was added TFA (1.75 g, 15.3 mmol, 1.13 mL, 299 equiv). The mixture was stirred at 25°C for 2 h. LC-MS showed desired compound was detected. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was used into the next step without further purification. Compound N-[4-[2-fluoro-3-[3-(4-piperidyloxy)propyl] phenoxy]-3-(6-methyl-7-oxo- 1H-pyrrolo[2,3-c]pyridin-4-yl)phenyl]ethanesulfonamide (30.0 mg, 43.1 μmol, 84% yield, TFA) was obtained as a yellow solid. [0427] Step 9: Preparation of N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]-6-[4-[3-[3-[4- (ethylsulfonylamino)-2-(6-methyl-7-oxo-1H-pyrrolo[2,3-c]pyri din-4-yl)phenoxy]-2-fluoro- phenyl]propoxy]-1-piperidyl]pyridazine-3-carboxamide. To a solution of N-[4-[2-fluoro-3-[3- (4-piperidyloxy)propyl]phenoxy]-3-(6-methyl-7-oxo-1H-pyrrolo [2,3-c]pyridin-4-yl)phenyl] ethanesulfonamide (75.0 mg, 108 μmol, 1.0 equiv, TFA) and 6-chloro-N-[4-(3-chloro-4-cyano- phenoxy)cyclohexyl]pyridazine-3-carboxamide (42.1 mg, 108 μmol, 1.0 equiv) in NMP (4.00 mL) was added K 2 CO 3 (29.8 mg, 215 μmol, 2.0 equiv). The mixture was stirred at 50 °C for 12 h. LC- MS showed desired compound was detected. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (column: Unisil 3-100 C18 Ultra 150x50mmx3 μm; mobile phase: [water(FA)-ACN];B%: 57%-87%,7min). Compound N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]-6-[4-[3-[3-[4-(et hylsulfonylamino)-2-(6- methyl-7-oxo-1H-pyrrolo[2,3-c]pyridin-4-yl)phenoxy]-2-fluoro -phenyl]propoxy]-1- piperidyl]pyridazine-3-carboxamide (32.4 mg, 34.2 μmol, 32% yield, 99% purity) was obtained as a off-white solid. LC-MS: MS (ES + ): RT =2.808 min, m/z = 937.2 [M + H + ] LCMS method: . 1 H NMR (400 MHz, METHANOL-d4) δ ppm 1.27 - 1.43 (m, 3 H) 1.53 - 1.75 (m, 5 H) 1.77 - 1.99 (m, 4 H) 2.02 - 2.26 (m, 4 H) 2.63 - 2.81 (m, 3 H) 3.05 - 3.21 (m, 3 H) 3.43 - 3.56 (m, 6 H) 3.58 - 3.71 (m, 5 H) 3.88 - 4.11 (m, 5 H) 4.44 - 4.54 (m, 2 H) 6.33 - 6.45 (m, 1 H) 6.60 - 6.76 (m, 1 H) 6.84 - 6.93 (m, 1 H) 6.95 - 7.10 (m, 1 H) 7.17 - 7.39 (m, 4 H) 7.42 - 7.55 (m, 1 H) 7.65 - 7.74 (m, 1 H) 7.86 (s, 1 H).

EXAMPLE 13 – Synthesis of N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]-6-[4-[2-[3-[4- ethylsulfonyl-2-(6-methyl-7-oxo-1H-pyrrolo[2,3-c]pyridin-4-y l)phenoxy]-2-fluoro- phenoxy]ethyl]piperazin-1-yl]pyridazine-3-carboxamide (I-40) [0428] Step 1: Preparation of tert-butyl 4-[2-(2-fluoro-3-hydroxy-phenoxy)ethyl] piperazine-1-carboxylate. To a mixture of tert-butyl 4-(2-chloroethyl)piperazine-1-carboxylate (4.00 g, 16.0 mmol, 1.0 equiv) and 2-fluorobenzene-1,3-diol (4.12 g, 32.1 mmol, 2.0 equiv) in MeCN (50 mL) was added K 2 CO 3 (4.44 g, 32.1 mmol, 2.0 equiv), the mixture was stirred at 50 °C for 2 h. The mixture was filtered and concentrated. The residue was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate=10/1 to 1/1) to give tert-butyl 4-[2-(2-fluoro- 3-hydroxy-phenoxy)ethyl]piperazine-1-carboxylate (2.3 g, 42 % yield). [0429] Step 2: Preparation of tert-butyl 4-[2-[3-(2-bromo-4-ethylsulfonyl-phenoxy)-2- fluoro-phenoxy]ethyl]piperazine-1-carboxylate. To a mixture of tert-butyl 4-[2-(2-fluoro-3- hydroxy-phenoxy)ethyl]piperazine-1-carboxylate (600 mg, 1.76 mmol, 1.0 equiv) and 2-bromo-4- ethylsulfonyl-1-fluoro-benzene (470 mg, 1.76 mmol, 1.0 equiv) in DMSO (10 mL) was added Cs 2 CO 3 (1.15 g, 3.53 mmol, 2.0 equiv), the mixture was stirred at 50 °C for 2 h. The mixture was diluted with water (20 mL) and extracted with EtOAc (30 mL * 3). The combined organic layers were washed with brine (50 mL), dried over Na2SO4, filtered and concentrated. The residue was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate=10/1 to 0/1) to give tert- butyl 4-[2-[3-(2-bromo-4-ethylsulfonyl-phenoxy)-2-fluoro-phenoxy]e thyl]piperazine-1- carboxylate (900 mg, 1.53 mmol, 86 % yield) as a white solid. [0430] Step 3: Preparation of a tert-butyl 4-[2-[3-[4-ethylsulfonyl-2-[6-methyl-7-oxo-1-(p- tolylsulfonyl)pyrrolo[2,3-c]pyridin-4-yl]phenoxy]-2-fluoro-p henoxy]ethyl]piperazine-1- carboxylate. To a mixture of tert-butyl 4-[2-[3-(2-bromo-4-ethylsulfonyl-phenoxy)-2-fluoro- phenoxy]ethyl]piperazine-1-carboxylate (900 mg, 1.53 mmol, 1.0 equiv) and 6-methyl-1-(p- tolylsulfonyl)-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-y l)pyrrolo[2,3-c]pyridin-7-one (787 mg, 1.84 mmol, 1.2 equiv) in THF (15 mL) and H2O (3 mL) was added K3PO4 (975 mg, 4.60 mmol, 3.0 equiv) and SPhos Pd G3 (119 mg, 153 μmol, 0.1 equiv), the mixture was stirred at 60 °C for 12 h under N 2 . The mixture was diluted with water (20 mL) and extracted with EtOAc (30 mL * 3). The combined organic layers were washed with brine (50 mL), dried over Na2SO4, filtered and concentrated to give a tert-butyl 4-[2-[3-[4-ethylsulfonyl-2-[6-methyl-7-oxo-1-(p- tolylsulfonyl)pyrrolo[2,3-c]pyridin-4-yl]phenoxy]-2-fluoro-p henoxy]ethyl]piperazine-1- carboxylate (1.2 g, crude). [0431] Step 4: Preparation of tert-butyl 4-[2-[3-[4-ethylsulfonyl-2-(6-methyl-7-oxo-1H- pyrrolo[2,3-c]pyridin-4-yl)phenoxy]-2-fluoro-phenoxy]ethyl]p iperazine-1-carboxylate. To a mixture of tert-butyl 4-[2-[3-[4-ethylsulfonyl-2-[6-methyl-7-oxo-1-(p-tolylsulfony l)pyrrolo[2,3- c]pyridin-4-yl]phenoxy]-2-fluoro-phenoxy]ethyl]piperazine-1- carboxylate (1.20 g, 1.48 mmol, 1.0 equiv) in MeOH (15 mL) was added KOH (249 mg, 4.45 mmol, 3.0 equiv), the mixture was stirred at 25 °C for 0.5 h. The reaction mixture was diluted with water (30 mL) and extracted with EtOAc (40 mL * 3). The combined organic layers were washed with brine (100 mL), dried over Na 2 SO 4 , filtered and concentrated. The residue was purified by prep-HPLC (column: Phenomenex luna C18 150*40mm* 15um;mobile phase: [water(FA)-ACN];B%: 17%-47%,10min) to give tert-butyl 4-[2- [3-[4-ethylsulfonyl-2-(6-methyl-7-oxo-1H-pyrrolo[2,3-c]pyrid in-4-yl)phenoxy]-2-fluoro- phenoxy]ethyl]piperazine-1-carboxylate (700 mg, 72 % yield). [0432] Step 5: Preparation of 4-[5-ethylsulfonyl-2-[2-fluoro-3-(2-piperazin-1- ylethoxy)phenoxy]phenyl]-6-methyl-1H-pyrrolo[2,3-c]pyridin-7 -one. To a mixture of tert-butyl 4-[2-[3-[4-ethylsulfonyl-2-(6-methyl-7-oxo-1H-pyrrolo[2,3-c] pyridin-4-yl)phenoxy]-2-fluoro- phenoxy]ethyl]piperazine-1-carboxylate (100 mg, 152 μmol, 1.0 equiv) in DCM (2 mL) was added TFA (1.54 g, 13.5 mmol, 1 mL, 88 equiv), the mixture was stirred at 25 °C for 0.5 h. The mixture was concentrated to give 4-[5-ethylsulfonyl-2-[2-fluoro-3-(2-piperazin-1- ylethoxy)phenoxy]phenyl]-6-methyl-1H-pyrrolo[2,3-c]pyridin-7 -one (100 mg, TFA salt) as a colorless gum. [0433] Step 6: Preparation of N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]-6-[4-[2-[3-[4- ethylsulfonyl-2-(6-methyl-7-oxo-1H-pyrrolo[2,3-c]pyridin-4-y l)phenoxy]-2-fluoro- phenoxy]ethyl]piperazin-1-yl]pyridazine-3-carboxamide. To a solution of 4-[5-ethylsulfonyl-2- [2-fluoro-3-(2-piperazin-1-ylethoxy)phenoxy]phenyl]-6-methyl -1H-pyrrolo[2,3-c]pyridin-7-one (84 mg, 151 μmol, 1 equiv) and 6-chloro-N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl] pyridazine- 3-carboxamide (65 mg, 166 μmol, 1.1 equiv) in NMP (2 mL) was added K2CO3 (105 mg, 757 μmol, 5 equiv). The mixture was stirred at 50 °C for 1 h. The mixture was filtered and concentrated. The residue was purified by prep-HPLC (column: Phenomenex C 18 75*30mm*3um; mobile phase: [water (FA)-ACN]; B%: 22%-52%, 7 min) to give N-[4-(3-chloro- 4-cyano-phenoxy)cyclohexyl]-6-[4-[2-[3-[4-ethylsulfonyl-2-(6 -methyl-7-oxo-1H-pyrrolo[2,3- c]pyridin-4-yl)phenoxy]-2-fluoro-phenoxy]ethyl]piperazin-1-y l]pyridazine-3-carboxamide (75 mg, 54% yield). 1 H NMR (400 MHz, DMSO-d6) δ 12.20 - 12.05 (m, 1H), 8.64 - 8.58 (m, 1H), 7.93 (d, J = 2.2 Hz, 1H), 7.88 - 7.84 (m, 2H), 7.82 ( d, J = 2.2 Hz, 1H), 7.43 (s, 1H), 7.39 (d, J = 2.3 Hz, 2H), 7.33 - 7.29 (m, 1H), 7.24 - 7.09 (m, 3H), 7.03 - 6.98 (m, 1H), 6.92 - 6.83 (m, 1H), 6.28 - 6.24 (m, 1H), 4.57 - 4.49 (m, 1H), 4.29 - 4.20 (m, 2H), 3.94 - 3.76 (m, 2H), 3.75 - 3.67 (m, 4H), 3.58 (s, 3H), 3.37 - 3.35 (m, 1H), 2.85 - 2.75 (m, 2H), 2.65 - 2.58 (m, 4H), 2.15 - 2.02 (m, 2H), 1.94 - 1.86 (m, 2H), 1.72 - 1.44 (m, 4H), 1.14 (t, J = 7.3 Hz, 3H). LC-MS: MS (ES + ): RT =2.219 min, m/z =909.2 [M + 1]; LCMS Method 25. EXAMPLE 14 – Synthesis of N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]-6-[4-[2-[4-[4- ethylsulfonyl-2-(6-methyl-7-oxo-1H-pyrrolo[2,3-c]pyridin-4-y l)phenoxy]-3-fluoro- phenyl]ethyl]-1-piperidyl]pyridazine-3-carboxamide (I-45)

[0434] Step 1: Preparation of tert-butyl 4-[2-(4-benzyloxy-3-fluoro- phenyl)ethynyl]piperidine-1-carboxylate. To a solution of 1-benzyloxy-4-bromo-2-fluoro- benzene (2.00 g, 7.11 mmol, 1.0 equiv) and tert-butyl 4-ethynylpiperidine-1-carboxylate (1.49 g, 7.11 mmol, 1.0 equiv) in DMF (20 mL) was added TEA (2.16 g, 21.3 mmol, 3.0 equiv), dichloropalladium;triphenylphosphane (249 mg, 355 μmol, 0.05 equiv) and CuI (135 mg, 711 μmol, 0.1 equiv). The mixture was stirred at 80 °C for 12 h under N 2 . The residue was diluted with H 2 O (50 mL) and extracted with EtOAc (50 mL * 3). The combined organic layers were washed with brine (50 mL), dried over Na2SO4, filtered and concentrated. The residue was purified by column chromatography (SiO 2 , Petroleum ether/Ethyl acetate=50/1 to 5/1) to give tert-butyl 4-[2- (4-benzyloxy-3-fluoro-phenyl)ethynyl]piperidine-1-carboxylat e (2.1 g, 72 % yield). [0435] Step 2: Preparation of tert-butyl 4-[2-(3-fluoro-4-hydroxy-phenyl)ethyl] piperidine- 1-carboxylate. To a solution of tert-butyl 4-[2-(4-benzyloxy-3-fluoro-phenyl)ethynyl]piperidine- 1-carboxylate (2.10 g, 5.13 mmol, 1.0 equiv) in MeOH (20 mL) was added Pd/C (200 mg, 10 % purity) under N 2 . The mixture was stirred under H 2 (15 psi) at 30 °C for 1 h. The mixture was filtered and concentrated to give tert-butyl 4-[2-(3-fluoro-4-hydroxy-phenyl)ethyl]piperidine-1- carboxylate (1.66 g, 5.13 mmol). [0436] Step 3: Preparation of tert-butyl 4-[2-[4-(2-bromo-4-ethylsulfonyl-phenoxy)-3- fluoro-phenyl]ethyl]piperidine-1-carboxylate. To a mixture of tert-butyl 4-[2-(3-fluoro-4- hydroxy-phenyl)ethyl]piperidine-1-carboxylate (300 mg, 927 μmol, 1.0 equiv) and 2-bromo-4- ethylsulfonyl-1-fluoro-benzene (247 mg, 927 μmol, 1.0 equiv) in DMSO (5 mL) was added Cs 2 CO 3 (604 mg, 1.86 mmol, 2.0 equiv), the mixture was stirred at 50 °C for 2 h. The mixture was diluted with water (80 mL) and extracted with EtOAc (50 mL * 3). The combined organic layers were washed with brine (50 mL), dried over Na 2 SO 4 , filtered and concentrated. The residue was purified by column chromatography (SiO 2 , Petroleum ether/Ethyl acetate=50/1 to 5/1) to give tert- butyl 4-[2-[4-(2-bromo-4-ethylsulfonyl-phenoxy)-3-fluoro-phenyl]et hyl]piperidine-1-carboxylate (380 mg, 71 % yield). [0437] Step 4: Preparation of tert-butyl 4-[2-[4-[4-ethylsulfonyl-2-[6-methyl-7-oxo-1-(p- tolylsulfonyl)pyrrolo[2,3-c]pyridin-4-yl]phenoxy]-3-fluoro-p henyl]ethyl]piperidine-1- carboxylate. To a mixture of tert-butyl 4-[2-[4-(2-bromo-4-ethylsulfonyl-phenoxy)-3-fluoro- phenyl]ethyl]piperidine-1-carboxylate (380 mg, 666 μmol, 1.0 equiv) and 6-methyl-1-(p- tolylsulfonyl)-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-y l)pyrrolo[2,3-c]pyridin-7-one (313 mg, 732 μmol, 1.1 equiv) in THF (10 mL) and H 2 O (2 mL) was added SPhos Pd G3 (51 mg, 66 μmol, 0.1 equiv) and K3PO4 (424 mg, 2 mmol, 3.0 equiv), the mixture was stirred at 60 °C for 12 h under N2. The mixture was diluted with water (20 mL) and extracted with EtOAc (30 mL * 3). The combined organic layers were washed with brine (50 mL), dried over Na 2 SO 4 , filtered and concentrated to give tert-butyl 4-[2-[4-[4-ethylsulfonyl-2-[6-methyl-7-oxo-1-(p- tolylsulfonyl)pyrrolo[2,3-c]pyridin-4-yl]phenoxy]-3-fluoro-p henyl]ethyl]piperidine-1-carboxylate (527 mg, crude). [0438] Step 5: Preparation of tert-butyl 4-[2-[4-[4-ethylsulfonyl-2-(6-methyl-7-oxo-1H- pyrrolo[2,3-c]pyridin-4-yl)phenoxy]-3-fluoro-phenyl]ethyl]pi peridine-1-carboxylate. To a mixture of tert-butyl 4-[2-[4-[4-ethylsulfonyl-2-[6-methyl-7-oxo-1-(p-tolylsulfony l)pyrrolo[2,3- c]pyridin-4-yl]phenoxy]-3-fluoro-phenyl]ethyl]piperidine-1-c arboxylate (527 mg, 665 μmol, 1.0 equiv) in MeOH (10 mL) was added KOH (186 mg, 3.33 mmol, 5.0 equiv), the mixture was stirred at 25 °C for 1 h. The reaction mixture was diluted with water (50 mL) and extracted with EtOAc (50 mL * 3). The combined organic layers were washed with brine (50 mL), dried over Na2SO4, filtered and concentrated. The residue was purified by column chromatography (SiO 2 , Petroleum ether/Ethyl acetate=20/1 to 0/1) to give tert-butyl 4-[2-[4-[4-ethylsulfonyl-2-(6-methyl-7-oxo-1H- pyrrolo[2,3-c]pyridin-4-yl)phenoxy]-3-fluoro-phenyl]ethyl]pi peridine-1-carboxylate (240 mg, 56 % yield). [0439] Step 6: Preparation of 4-[5-ethylsulfonyl-2-[2-fluoro-4-[2-(4-piperidyl)ethyl] phenoxy]phenyl]-6-methyl-1H-pyrrolo[2,3-c]pyridin-7-one. To a mixture of tert-butyl 4-[2-[4- [4-ethylsulfonyl-2-(6-methyl-7-oxo-1H-pyrrolo[2,3-c]pyridin- 4-yl)phenoxy]-3-fluoro- phenyl]ethyl]piperidine-1-carboxylate (240 mg, 376 μmol, 1.0 equiv) in DCM (4 mL) was added TFA (2 mL), the mixture was stirred at 30 °C for 0.5 h. The mixture was concentrated to give 4-[5- ethylsulfonyl-2-[2-fluoro-4-[2-(4-piperidyl)ethyl]phenoxy]ph enyl]-6-methyl-1H-pyrrolo[2,3- c]pyridin-7-one (245 mg, TFA salt). [0440] Step 7: Preparation of N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]-6-[4-[2-[4-[4- ethylsulfonyl-2-(6-methyl-7-oxo-1H-pyrrolo[2,3-c]pyridin-4-y l)phenoxy]-3-fluoro- phenyl]ethyl]-1-piperidyl]pyridazine-3-carboxamide. To a mixture of 4-[5-ethylsulfonyl-2-[2- fluoro-4-[2-(4-piperidyl)ethyl]phenoxy]phenyl]-6-methyl-1H-p yrrolo[2,3-c]pyridin-7-one (245 mg, 375 μmol, 1.0 equiv, TFA salt) and 6-chloro-N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl] pyridazine-3-carboxamide (161 mg, 413 μmol, 1.1 equiv) in NMP (3 mL) was added K2CO3 (259 mg, 1.88 mmol, 5.0 equiv), the mixture was stirred at 50 °C for 12 h. The mixture was filtered and concentrated. The residue was purified by prep-HPLC (column: UniSil 3-100 C18 UItra (150*25mm*3um);mobile phase: [water(FA)-ACN];B%: 59%-89%,7min) to give N-[4-(3-chloro- 4-cyano-phenoxy)cyclohexyl]-6-[4-[2-[4-[4-ethylsulfonyl-2-(6 -methyl-7-oxo-1H-pyrrolo[2,3- c]pyridin-4-yl)phenoxy]-3-fluoro-phenyl]ethyl]-1-piperidyl]p yridazine-3-carboxamide (56 mg, 16 % yield). 1 H NMR (400 MHz, DMSO-d6): δ 12.18 - 12.05 (m, 1H), 8.63 - 8.55 (m, 1H), 7.94 - 7.90 (m, 1H), 7.88 - 7.77 (m, 3H), 7.44 (s, 1H), 7.41 - 7.37 (m, 1H), 7.36 - 7.22 (m, 4H), 7.16 - 7.08 (m, 2H), 6.95 - 6.88 (m, 1H), 6.32 - 6.20 (m, 1H), 4.59 - 4.43 (m, 3H), 3.90 - 3.80 (m, 1H), 3.58 (s, 3H), 3.30 - 3.26 (m, 2H), 3.04 - 2.91 (m, 2H), 2.68 - 2.62 (m, 2H), 2.15 - 2.05 (m, 2H), 1.93 - 1.87 (m, 2H), 1.85 - 1.77 (m, 2H), 1.70 - 1.47 (m, 7H), 1.20 - 1.10 (m, 5H). LC-MS: MS (ES + ): RT = 2.533 min, m/z = 892.2 [M + 1]; LCMS Method 25. EXAMPLE 15 – Synthesis of N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]-6-[2-[4-[4- ethylsulfonyl-2-(6-methyl-7-oxo-1H-pyrrolo[2,3-c]pyridin-4-y l)phenoxy]phenyl]-2,7- diazaspiro[3.5]nonan-7-yl]pyridazine-3-carboxamide (I-90)

[0441] Step 1: Preparation of tert-butyl 2-(4-benzyloxyphenyl)-2,7-diazaspiro[3.5] nonane- 7-carboxylate. A mixture of tert-butyl 2,7-diazaspiro[3.5]nonane-7-carboxylate (1.5 g, 6.63 mmol, 1.0 equiv), 1-benzyloxy-4-bromo-benzene (1.7 g, 6.63 mmol, 1.0 equiv), Pd 2 (dba) 3 (303 mg, 331 μmol, 0.05 equiv), BINAP (412 mg, 663 μmol, 0.1 equiv) and NaOtBu (2.1 g, 22 mmol, 3.3 equiv), TEA (738 mg, 7.29 mmol, 1.1 equiv) in Tol. (8 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 110 °C for 12 h under N2 atmosphere. The residue was purified by column chromatography (SiO 2 , Petroleum ether/Ethyl acetate=10:1 to 5:1) to give tert-butyl 2- (4-benzyloxyphenyl)-2,7-diazaspiro[3.5]nonane-7-carboxylate (2.5 g, 92% yield) as a yellow solid. [0442] Step 2: Preparation of tert-butyl 2-(4-hydroxyphenyl)-2,7-diazaspiro[3.5]nonane-7- carboxylate. To a solution of tert-butyl 2-(4-benzyloxyphenyl)-2,7-diazaspiro[3.5]nonane-7- carboxylate (2.5 g, 6.12 mmol, 1.0 equiv) in THF (30 mL) was added Pd/C (300 mg, 10% purity). The mixture was stirred under H 2 (15 psi) at 20°C for 3 h. The mixture was filtered and the filter was concentrated to give tert-butyl 2-(4-hydroxyphenyl)-2,7-diazaspiro[3.5]nonane-7-carboxylate (1.9 g, 98% yield) as a yellow solid. [0443] Step 3: Preparation of tert-butyl 2-[4-(2-bromo-4-ethylsulfonyl-phenoxy)phenyl]- 2,7-diazaspiro[3.5]nonane-7-carboxylate. To a solution of tert-butyl 2-(4-hydroxyphenyl)-2,7- diazaspiro[3.5]nonane-7-carboxylate (0.5 g, 1.57 mmol, 1.0 equiv) in DMSO (5 mL) was added Cs2CO3 (1.53 g, 4.71 mmol, 3.0 equiv) and 2-bromo-4-ethylsulfonyl-1-fluoro-benzene (420 mg, 1.57 mmol, 1.0 equiv). The mixture was stirred at 50 °C for 1 h. The residue was purified by column chromatography (SiO 2 , Petroleum ether/Ethyl acetate = 10/1 to 1/1) to give tert-butyl 2-[4- (2-bromo-4-ethylsulfonyl-phenoxy)phenyl]-2,7-diazaspiro[3.5] nonane-7-carboxylate (680 mg, 77% yield) as a white solid. [0444] Step 4: Preparation tert-butyl 2-[4-[4-ethylsulfonyl-2-[6-methyl-7-oxo-1-(p- tolylsulfonyl)pyrrolo[2,3-c]pyridin-4-yl]phenoxy]phenyl]-2,7 -diazaspiro[3.5]nonane-7- carboxylate. To a solution of 6-methyl-1-(p-tolylsulfonyl)-4-(4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2-yl)pyrrolo[2,3-c]pyridin-7-one (113 mg, 265 μmol, 1.0 equiv) and tert-butyl 2-[4- (2-bromo-4-ethylsulfonyl-phenoxy)phenyl]-2,7-diazaspiro[3.5] nonane-7-carboxylate (150 mg, 265 μmol, 1.0 equiv) in THF (10 mL) was added H 2 O (1 mL), dicyclohexyl-[2-(2,6- dimethoxyphenyl)phenyl]phosphane;methanesulfonate;(2-phenyla nilino)palladium(1+) (20.7 mg, 26 μmol, 0.1 eq) and K3PO4 (169 mg, 796 μmol, 3 eq). The mixture was stirred at 60 °C for 12 h. The residue was purified by prep-TLC (PE/EA = 1/2) to give tert-butyl 2-[4-[4-ethylsulfonyl-2-[6- methyl-7-oxo-1-(p-tolylsulfonyl)pyrrolo[2,3-c]pyridin-4-yl]p henoxy]phenyl]-2,7- diazaspiro[3.5]nonane-7-carboxylate (200 mg, 96% yield) as a yellow solid. [0445] Step 5: Preparation of tert-butyl 2-[4-[4-ethylsulfonyl-2-(6-methyl-7-oxo-1H- pyrrolo[2,3-c]pyridin-4-yl)phenoxy]phenyl]-2,7-diazaspiro[3. 5]nonane-7-carboxylate. To a solution of tert-butyl 2-[4-[4-ethylsulfonyl-2-[6-methyl-7-oxo-1-(p-tolylsulfonyl)p yrrolo[2,3- c]pyridin-4-yl]phenoxy]phenyl]-2,7-diazaspiro[3.5]nonane-7-c arboxylate (200 mg, 254 μmol, 1.0 equiv) in MeOH (3 mL) was added KOH (285 mg, 5.08 mmol, 20.0 equiv). The mixture was stirred at 20 °C for 1 h. The residue was purified by prep-TLC (DCM/MeOH = 10/1) to give tert- butyl 2-[4-[4-ethylsulfonyl-2-(6-methyl-7-oxo-1H-pyrrolo[2,3-c]pyr idin-4-yl)phenoxy]phenyl]- 2,7-diazaspiro[3.5]nonane-7-carboxylate (160 mg, 100% yield) as a yellow solid. [0446] Step 6: Preparation of 4-[2-[4-(2,7-diazaspiro[3.5]nonan-2-yl)phenoxy]-5- ethylsulfonyl-phenyl]-6-methyl-1H-pyrrolo[2,3-c]pyridin-7-on e. To a solution of tert-butyl 2- [4-[4-ethylsulfonyl-2-(6-methyl-7-oxo-1H-pyrrolo[2,3-c]pyrid in-4-yl)phenoxy]phenyl]-2,7- diazaspiro[3.5]nonane-7-carboxylate (160 mg, 252 μmol, 1.0 equiv) in DCM (3 mL) was added TFA (1 mL). The mixture was stirred at 10 °C for 0.5 h. The mixture was concentrated to give 4- [2-[4-(2,7-diazaspiro[3.5]nonan-2-yl)phenoxy]-5-ethylsulfony l-phenyl]-6-methyl-1H-pyrrolo[2,3- c]pyridin-7-one (160 mg, TFA salt) as a yellow solid. [0447] Step 7: Preparation of N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]-6-[2-[4-[4- ethylsulfonyl-2-(6-methyl-7-oxo-1H-pyrrolo[2,3-c]pyridin-4-y l)phenoxy]phenyl]-2,7- diazaspiro[3.5]nonan-7-yl]pyridazine-3-carboxamide.4-[2-[4-( 2,7-diazaspiro[3.5]nonan-2- yl)phenoxy]-5-ethylsulfonyl4-[2-[4-(2,7-diazaspiro[3.5]nonan -2-yl)phenoxy]-5-ethylsulfonyl- phenyl]-6-methyl-1H-pyrrolo[2,3-c]pyridin-7-one (160 mg, 247 μmol, 1.0 equiv, TFA salt) in NMP (5 mL) was added K2CO3 (170 mg, 1.24 mmol, 5.0 equiv) and 6-chloro-N-[4-(3-chloro-4- cyano-phenoxy)cyclohexyl]pyridazine-3-carboxamide (96 mg, 247 μmol, 1.0 equiv). The mixture was stirred at 50 °C for 12 h. The residue was purified by prep-HPLC (column: YMC Triart C18 150*25mm*5um;mobile phase: [water(TFA)-ACN];B%:63%-83%,10min) and triturated with MeOH (3 mL) at 20 o C for 30 min to give N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]-6-[2-[4- [4-ethylsulfonyl-2-(6-methyl-7-oxo-1H-pyrrolo[2,3-c]pyridin- 4-yl)phenoxy]phenyl]-2,7- diazaspiro[3.5]nonan-7-yl]pyridazine-3-carboxamide (100 mg, 46% yield). 1 H NMR (400 MHz, CDCl3): δ 9.80-9.72 (m, 1H), 8.03 (dd, J = 6.0, 3.6 Hz, 2H), 7.92-7.85 (m, 1H), 7.81-7.73 (m, 1H), 7.58 (d, J = 8.8 Hz, 1H), 7.19 (s, 1H), 7.07-7.02 (m, 2H), 6.97-6.93 (m, 3H), 6.90-6.86 (m, 1H), 6.54-6.45 (m, 2H), 6.44-6.39 (m, 1H), 4.41-4.29 (m, 1H), 4.14-4.02 (m, 1H), 3.85-3.79 (m, 4H), 3.75-3.73 (m, 7H), 3.20-3.14 (m, 2H), 2.25-2.16 (m, 4 H), 2.03-1.97 (m, 4H), 1.71 (d, J=11.6 Hz, 2H), 1.52-1.44 (m, 2H), 1.35 (t, J = 7.6 Hz, 3H) LC-MS: MS (ES + ): RT = 3.132 min, m/z = 887.2 [M + H + ]; LCMS Method 10. EXAMPLE 16 – Synthesis of compound N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]-6-[4-[4- [4-(2,4-difluorophenoxy)-3-(6-methyl-7-oxo-1H-pyrrolo[2,3-c] pyridin-4- yl)phenyl]sulfonylbutoxy]-1-piperidyl]pyridazine-3-carboxami de (I-92)

[0448] Step 1: Preparation of tert-butyl 4-(4-bromobutoxy)piperidine-1-carboxylate. To a solution of tert-butyl 4-hydroxypiperidine-1-carboxylate (10.0 g, 49.7 mmol, 1.0 equiv) in THF (100 mL) was added NaH (2.98 g, 74.5 mmol, 60% purity, 1.5 equiv) at 0 °C under N2 atmosphere. The mixture was stirred at 0 °C for 0.5 h, and then 1,4-dibromobutane (10.7 g, 49.7 mmol, 1.0 equiv) was added at 0 °C. The mixture was stirred at 25 °C for 12 h. The reaction mixture was poured into NH4Cl aq.200 mL at 0 °C, and then diluted with water 300 mL extracted with EA (300 mL x 3). The combined organic layers were washed with brine (200 mL x 3), dried over Na 2 SO 4 , filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate=10/1 to 2/1). The desired compound tert-butyl 4-(4-bromobutoxy)piperidine-1-carboxylate (2.0 g, 6.0 mmol, 12% yield) was obtained as yellow oil. [0449] Step 2: Preparation of tert-butyl 4-[4-(3-chloro-4-fluoro-phenyl)sulfanyl- butoxy]piperidine-1-carboxylate. To a solution of tert-butyl 4-(4-bromobutoxy)piperidine-1- carboxylate (1.04 g, 3.09 mmol, 1.0 equiv) and 3-chloro-4-fluoro-benzenethiol (604 mg, 3.71 mmol, 1.2 equiv) in DMF (10 mL) was added K2CO3 (1.28 g, 9.28 mmol, 3.0 equiv). The mixture was stirred at 50 °C for 12 h. The reaction mixture was poured into water 50 mL, and then diluted with water 40 mL and extracted with EA (50 mL x 3). The combined organic layers were washed with brine (50 mL x 2), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO 2 , Petroleum ether/Ethyl acetate=10/1 to 9/1). The desired compound tert-butyl 4-[4-(3-chloro-4-fluoro- phenyl)sulfanylbutoxy]piperidine-1-carboxylate (1.0 g, 2.4 mmol, 77% yield) was obtained as yellow oil. [0450] Step 3: Preparation of tert-butyl 4-[4-(3-chloro-4-fluoro-phenyl)sulfonylbutoxy] piperidine-1-carboxylate. A mixture of tert-butyl 4-[4-(3-chloro-4-fluoro-phenyl)sulfanyl- butoxy]piperidine-1-carboxylate (500 mg, 1.20 mmol, 1.0 equiv) in DCM (5 mL) was added m- CPBA (607 mg, 2.99 mmol, 85% purity, 2.5 equiv) at 0 °C. The mixture was stirred at 25 °C for 12 h. The reaction mixture was poured into Na2SO3 aq.10 mL at 0 °C, and then diluted with water 20 mL and extracted with EA (30 mL x 3). The combined organic layers were washed with Na2SO3 aq. (30 mL x 2), dried over Na 2 SO 4 , filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate=5/1 to 3/1). The desired compound tert-butyl 4-[4-(3-chloro-4-fluoro- phenyl)sulfonylbutoxy]piperidine-1-carboxylate (510 mg, 1.13 mmol, 95% yield) was obtained as yellow oil. [0451] Step 4: Preparation of tert-butyl 4-[4-[3-chloro-4-(2,4-difluorophenoxy) phenyl]sulfonylbutoxy]piperidine-1-carboxylate. To a solution oftert-butyl 4-[4-(3-chloro-4- fluoro-phenyl)sulfonylbutoxy]piperidine-1-carboxylate (920 mg, 2.04 mmol, 1.0 equiv) in ACN (10 mL) was added 2,4-difluorophenol (319 mg, 2.45 mmol, 1.2 equiv) and K 2 CO 3 (848 mg, 6.13 mmol, 3.0 equiv). The mixture was stirred at 60 °C for 12 h. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO 2 , Petroleum ether/Ethyl acetate=10/1 to 3/1). The desired compound tert- butyl 4-[4-[3-chloro-4-(2,4-difluorophenoxy)phenyl]sulfonylbutoxy] piperidine-1-carboxylate (1.10 g, 1.96 mmol, 96% yield) was obtained as colourless oil. [0452] Step 5: Preparation of tert-butyl 4-[4-[4-(2,4-difluorophenoxy)-3-[6-methyl-7-oxo- 1-(p-tolylsulfonyl)pyrrolo[2,3-c]pyridin-4-yl]phenyl]sulfony lbutoxy]piperidine-1-carboxylate. To a solution of 6-methyl-1-(p-tolylsulfonyl)-4-(4,4,5,5-tetramethyl-1,3,2-di oxaborolan-2- yl)pyrrolo[2,3-c]pyridin-7-one (150 mg, 350 μmol, 1.0 equiv) and tert-butyl 4-[4-[3-chloro-4-(2,4- difluorophenoxy)phenyl]sulfonylbutoxy]piperidine-1-carboxyla te (196 mg, 350 μmol, 1.0 equiv) in THF (4 mL) and H 2 O (0.8 mL) was added K 3 PO 4 (223 mg, 1.05 mmol, 3.0 equiv), and then SPhos Pd G3 (55.0 mg, 70.0 μmol, 0.2 equiv) was added and purged with N 2 for 3 times. The mixture was stirred at 60 °C for 12 h. The reaction mixture was quenched by addition water 20 mL, and then extracted with EA (20 mL x 3). The combined organic layers were washed with brine (20 mL x 3), dried over Na 2 SO 4 , filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (FA condition;column: UniSil 3-100 C18 UItra (150*25mm*3um);mobile phase: [water(FA)-ACN];B%: 62%-92%,7min). The desired compound tert-butyl 4-[4-[4-(2,4-difluorophenoxy)-3-[6-methyl-7-oxo-1-(p-tolylsu lfonyl)pyrrolo[2,3- c]pyridin-4-yl]phenyl]sulfonylbutoxy]piperidine-1-carboxylat e (150 mg, 182 μmol, 52% yield) was obtained as off-white solid. [0453] Step 6: Preparation of tert-butyl 4-[4-[4-(2,4-difluorophenoxy)-3-(6-methyl-7-oxo- 1H-pyrrolo[2,3-c]pyridin-4-yl)phenyl]sulfonylbutoxy]piperidi ne-1-carboxylate. To a solution of tert-butyl 4-[4-[4-(2,4-difluorophenoxy)-3-[6-methyl-7-oxo-1-(p-tolylsu lfonyl) pyrrolo[2,3- c]pyridin-4-yl]phenyl]sulfonylbutoxy]piperidine-1-carboxylat e (150 mg, 182 μmol, 1.0 equiv) in MeOH (1.5 mL) was added KOH (102 mg, 1.82 mmol, 10.0 equiv) and THF (1 mL). The mixture was stirred at 25 °C for 1 h. The reaction mixture was quenched by addition water 30 mL, and then extracted with EA (50 mL x 3). The combined organic layers were washed with water (50 mL x 3), dried over Na 2 SO 4 , filtered and concentrated under reduced pressure to give a residue. The reaction mixture was used into the next step without further purification. The desired compound tert-butyl 4-[4-[4-(2,4-difluorophenoxy)-3-(6-methyl-7-oxo-1H-pyrrolo[2 ,3-c]pyridin-4- yl)phenyl]sulfonylbutoxy]piperidine-1-carboxylate (110 mg, 164 μmol, 90% yield) was obtained as colourless oil. [0454] Step 7: Preparation of 4-[2-(2,4-difluorophenoxy)-5-[4-(4-piperidyloxy) butylsulfonyl]phenyl]-6-methyl-1H-pyrrolo[2,3-c]pyridin-7-on e. To a solution of tert-butyl 4- [4-[4-(2,4-difluorophenoxy)-3-(6-methyl-7-oxo-1H-pyrrolo[2,3 -c]pyridin-4-yl)phenyl] sulfonylbutoxy]piperidine-1-carboxylate (110 mg, 164 μmol, 1.0 equiv) in DCM (1 mL) was added TFA (1.54 g, 13.5 mmol, 82.5 equiv). The mixture was stirred at 25 °C for 1 h. The reaction mixture was concentrated under reduced pressure to give a residue. The reaction mixture was used into the next step without further purification. The desired compound 4-[2-(2,4-difluorophenoxy)- 5-[4-(4-piperidyloxy)butylsulfonyl]phenyl]-6-methyl-1H-pyrro lo[2,3-c]pyridin-7-one (93 mg, crude) was obtained as brown oil. [0455] Step 8: Preparation of compound N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]-6- [4-[4-[4-(2,4-difluorophenoxy)-3-(6-methyl-7-oxo-1H-pyrrolo[ 2,3-c]pyridin-4-yl)phenyl] sulfonylbutoxy]-1-piperidyl]pyridazine-3-carboxamide. To a solution of 4-[2-(2,4- difluorophenoxy)-5-[4-(4-piperidyloxy)butylsulfonyl]phenyl]- 6-methyl-1H-pyrrolo[2,3-c]pyridin- 7-one (93.0 mg, 0.160 mmol, 1.0 equiv) and 6-chloro-N-[4-(3-chloro-4-cyano- phenoxy)cyclohexyl]pyridazine-3-carboxamide (64.0 mg, 0.160 mmol, 1.0 equiv) in NMP (1.5 mL) was added K 2 CO 3 (225 mg, 1.63 mmol, 10.0 equiv) The reaction mixture was stirred at 50 °C for 12 h. The reaction mixture was filtered. The residue was purified by prep-HPLC (FA condition; column: Phenomenex Luna C18150*25mm*10um;mobile phase: [water(FA)-ACN];B%: 53%- 83%,10min). The desired compound N-[4-(3-chloro-4-cyano-phenoxy) cyclohexyl]-6-[4-[4-[4- (2,4-difluorophenoxy)-3-(6-methyl-7-oxo-1H-pyrrolo[2,3-c]pyr idin-4-yl)phenyl]sulfonylbutoxy]- 1-piperidyl]pyridazine-3-carboxamide (67 mg, 72 μmol, 44% yield) was obtained as off-white solid. 1 H NMR (400 MHz, METHANOL-d 4 ) δ = 8.03 (d, J = 2.4 Hz, 1H), 7.91 - 7.85 (m, 2H), 7.71 - 7.67 (m, 1H), 7.38 (s, 1H), 7.34 (d, J = 2.8 Hz, 1H), 7.24 - 7.18 (m, 3H), 7.16 - 7.10 (m, 1H), 7.06 - 7.00 (m, 2H), 6.99 - 6.94 (m, 1H), 6.35 (d, J = 2.8 Hz, 1H), 4.54 - 4.46 (m, 1H), 3.99 - 3.89 (m, 3H), 3.69 (s, 3H), 3.58 (d, J = 3.6 , 1H), 3.54 - 3.49 (m, 3H), 3.47 (d, J = 4.8 Hz, 1H), 3.34 (s, 1H), 2.22 - 2.16 (m, 2H), 2.13 - 2.06 (m, 2H), 1.92 - 1.78 (m, 5H), 1.70 - 1.61 (m, 6H), 1.57 - 1.50 (m, 2H) LC-MS: MS (ES + ): RT = 2.782 min, m/z = 926.5 [M + H + ]; LCMS Method 25. EXAMPLE 17 – Synthesis of N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]-6-[3-[4-[4-(1- hydroxy-1-methyl-ethyl)-2-(6-methyl-7-oxo-1H-pyrrolo[2,3-c]p yridin-4-yl)phenoxy] phenoxy]azetidin-1-yl]pyridazine-3-carboxamide (I-95)

[0456] Step 1: Preparation of tert-butyl 3-(4-benzyloxyphenoxy)azetidine-1-carboxylate. To a solution of tert-butyl 3-bromoazetidine-1-carboxylate (5 g, 21.18 mmol, 1.0 equiv) in ACN (50 mL) was added K 2 CO 3 (8.78 g, 63.53 mmol, 3.0 equiv) and 4-benzyloxyphenol (4.24 g, 21.18 mmol, 1.0 equiv). The mixture was stirred at 80 °C for 12 h. The mixture was diluted with water (50 mL) and extracted with Ethyl acetate (3 x 50 mL). The combined organic layers were dried over sodium sulfate, filtered and concentrated. The residue was purified by prep-HPLC (column: Kromasil Eternity XT 250*80mm*10um;mobile phase: [water( NH4HCO3)-ACN];B%: 50%- 80%,20min) to give tert-butyl 3-(4-benzyloxyphenoxy)azetidine-1-carboxylate (2.8 g, 37% yield) as a yellow solid. [0457] Step 2: Preparation of tert-butyl 3-(4-hydroxyphenoxy)azetidine-1-carboxylate. To a solution of tert-butyl 3-(4-benzyloxyphenoxy)azetidine-1-carboxylate (1.4 g, 3.94 mmol, 1.0 equiv) in THF (14 mL) was added Pd/C (140 mg, 10% purity) under N2 atmosphere. The mixture was stirred under H2 (15 psi) at 25 °C for 2 h. The mixture was filtered and concentrated to give tert-butyl 3-(4-hydroxyphenoxy)azetidine-1-carboxylate (1 g, 95 % yield) as a white solid. [0458] Step 3: Preparation of tert-butyl 3-[4-(2-bromo-4-methoxycarbonyl-phenoxy) phenoxy]azetidine-1-carboxylate. To a solution of tert-butyl 3-(4-hydroxyphenoxy) azetidine-1- carboxylate (1 g, 3.77 mmol, 1.0 equiv) in DMSO (10 mL) was added Cs2CO3 (2.46 g, 7.54 mmol, 2.0 equiv) and methyl 3-bromo-4-fluoro-benzoate (878 mg, 3.77 mmol, 1.0 equiv). The mixture was stirred at 50 °C for 1 h. The mixture was diluted with water (10 mL) and extracted with Ethyl acetate (3 x 30 mL). The combined organic layers were dried over sodium sulfate, filtered and concentrated. The residue was purified by silica column chromatography on silica gel (Petroleum ether: Ethyl acetate from 10/1 to 4/1) to give tert-butyl 3-[4-(2-bromo-4-methoxycarbonyl- phenoxy)phenoxy]azetidine-1-carboxylate (1.6 g, 88% yield) as a yellow solid. [0459] Step 4: Preparation of methyl 4-[4-(azetidin-3-yloxy)phenoxy]-3-bromo-benzoate. To a solution of tert-butyl 3-[4-(2-bromo-4-methoxycarbonyl-phenoxy)phenoxy]azetidine-1- carboxylate (500 mg, 1.05 mmol, 1.0 equiv) in DCM (5 mL) was added TFA (1 mL). The mixture was stirred at 25 °C for 0.5 h. The solution was concentrated under reduced pressure to give methyl 4-[4-(azetidin-3-yloxy)phenoxy]-3-bromo-benzoate (514 mg, crude, TFA salt) as a yellow oil. [0460] Step 5: Preparation of 2-[4-[4-(azetidin-3-yloxy)phenoxy]-3-bromo-phenyl] propan-2-ol. To a solution of methyl 4-[4-(azetidin-3-yloxy)phenoxy]-3-bromo-benzoate (514 mg, 1.04 mmol, 1.0 equiv, TFA) in THF (10 mL) was added MeMgBr (3 M, 5.2 mL, 15.0 equiv) at 0 °C. The mixture was quenched by addition NH 4 Cl 10 mL and water (10 mL). The mixture was extracted with Ethyl acetate (3 x 40 mL). The combined organic layers were dried over sodium sulfate, filtered and concentrated to give 2-[4-[4-(azetidin-3-yloxy)phenoxy]-3-bromo- phenyl]propan-2-ol (390 mg crude) as a yellow solid. [0461] Step 6: Preparation of 4-[2-[4-(azetidin-3-yloxy)phenoxy]-5-(1-hydroxy-1-methyl- ethyl)phenyl]-6-methyl-1-(p-tolylsulfonyl)pyrrolo[2,3-c]pyri din-7-one. To a solution of 2-[4- [4-(azetidin-3-yloxy)phenoxy]-3-bromo-phenyl]propan-2-ol (200 mg, 528 μmol, 1.0 equiv) in THF (20 mL) and H2O (4 mL) was added 6-methyl-1-(p-tolylsulfonyl)-4-(4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2-yl)pyrrolo[2,3-c]pyridin-7-one (249 mg, 581 μmol, 1.1 equiv), K 3 PO 4 (337 mg, 1.59 mmol, 3.0 equiv) and SPhos Pd G3 (42 mg, 53 μmol, 0.1 equiv). The mixture was stirred at 60 °C for 12 h. The mixture was diluted with water (10 mL) and extracted with Ethyl acetate (3 x 30 mL). The combined organic layers were dried over sodium sulfate, filtered and concentrated. The residue was purified by prep-HPLC (column: Waters Xbridge C18150*50mm* 10um;mobile phase: [water( NH 4 HCO 3 )-ACN];B%: 35%-65%,10min) to give 4-[2-[4-(azetidin-3- yloxy)phenoxy]-5-(1-hydroxy-1-methyl-ethyl)phenyl]-6-methyl- 1-(p-tolylsulfonyl)pyrrolo[2,3- c]pyridin-7-one (125 mg, 39% yield) as a white solid. [0462] Step 7: Preparation of 4-[2-[4-(azetidin-3-yloxy)phenoxy]-5-(1-hydroxy-1-methyl- ethyl)phenyl]-6-methyl-1H-pyrrolo[2,3-c]pyridin-7-one. To a solution of 4-[2-[4-(azetidin-3- yloxy)phenoxy]-5-(1-hydroxy-1-methyl-ethyl)phenyl]-6-methyl- 1-(p-tolylsulfonyl)pyrrolo[2,3- c]pyridin-7-one (125 mg, 208 μmol, 1.0 equiv) in MeOH (1 mL) was added KOH (175 mg, 3.13 mmol, 15.0 equiv). The mixture was stirred at 25 °C for 1 h. The mixture was diluted with water (5 mL) and extracted with DCM (3 x 10 mL). The combined organic layers were dried over sodium sulfate, filtered and concentrated. The residue was purified by prep-HPLC (c column: Waters Xbridge C18150*50mm* 10um;mobile phase: [water( NH4HCO3)-ACN];B%: 16%-46%,10min) to give 4-[2-[4-(azetidin-3-yloxy)phenoxy]-5-(1-hydroxy-1-methyl-eth yl)phenyl]-6-methyl-1H- pyrrolo[2,3-c]pyridin-7-one (40 mg, 43% yield) as a white solid. Step 8: Preparation of N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]-6-[3-[4-[4-(1-hyd roxy-1- methyl-ethyl)-2-(6-methyl-7-oxo-1H-pyrrolo[2,3-c]pyridin-4-y l)phenoxy] phenoxy] azetidin- 1-yl]pyridazine-3-carboxamide. To a solution of 4-[2-[4-(azetidin-3-yloxy)phenoxy]-5-(1- hydroxy-1-methyl-ethyl)phenyl]-6-methyl-1H-pyrrolo[2,3-c]pyr idin-7-one (40 mg, 90 μmol, 1.0 equiv) in NMP (1 mL) was added K 2 CO 3 (37 mg, 269 μmol, 3.0 equiv) and 6-chloro-N-[4-(3- chloro-4-cyano-phenoxy)cyclohexyl]pyridazine-3-carboxamide (35 mg, 89 μmol, 1.0 equiv). The mixture was stirred at 50 °C for 12 h and concentrated. The residue was purified by prep-HPLC (column: Waters Xbridge 150*25mm* 5um;mobile phase: [water (ammonia hydroxide v/v)- ACN];B%: 44%-74%,9min) to give N-[4-(3-chloro-4-cyano-phenoxy) cyclohexyl]-6-[3-[4-[4-(1- hydroxy-1-methyl-ethyl)-2-(6-methyl-7-oxo-1H-pyrrolo[2,3-c]p yridin-4- yl)phenoxy]phenoxy]azetidin-1-yl]pyridazine-3-carboxamide (20 mg, 26% yield) as an off-white solid. 1 H NMR (400 MHz, CDCl 3 ) δ 10.24-9.97 (m, 1H), 8.03-7.97 (m, 1H), 7.92-7.85 (m, 1H), 7.67-7.63 (m, 1H), 7.60-7.54 (m, 1H), 7.48-7.41 (m, 1H), 7.26-7.22 (m, 1H), 7.07 (s, 1H), 7.03- 6.94 (m, 2H), 6.90-6.82 (m, 3H), 6.71-6.60 (m, 3H), 6.42-6.37 (m, 1H), 5.14-5.06 (m, 1H), 4.62- 4.53 (m, 2H), 4.38-4.29 (m, 1H), 4.28-4.19 (m, 2H), 4.12-4.00 (m, 1H), 3.65 (s, 3H), 2.24-2.13 (m, 4H), 2.01-1.95 (m, 1H), 1.69-1.66 (m, 2H), 1.65 (s, 6H), 1.53-1.45 (m, 2H). LC-MS: MS (ES + ): RT = 2.642 min, m/z = 800.2 [M + H + ]; LCMS Method 25.

EXAMPLE 18 – Synthesis of N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]-6-[2-[4-[4-(1- hydroxy-1-methyl-ethyl)-2-(6-methyl-7-oxo-1H-pyrrolo[2,3-c]p yridin-4-yl)phenoxy] phenyl]ethyl-methyl-amino]pyridazine-3-carboxamide (I-100) [0463] Step 1: Preparation of methyl 3-bromo-4-[4-[2-[tert-butoxycarbonyl(methyl) amino]ethyl]phenoxy]benzoate. To a solution of methyl 3-bromo-4-fluoro-benzoate (862 mg, 3.7 mmol, 1 equiv) and tert-butyl N-[2-(4-hydroxyphenyl) ethyl]-N-methyl-carbamate (930 mg, 3.7 mmol, 1 equiv) in DMSO (8 mL) was added Cs2CO3 (2.4 g, 7.4 mmol, 2 equiv). The mixture was stirred at 50 °C for 1 h. The residue was purified by prep-HPLC (column: Waters Xbridge C18 150*50mm* 10um;mobile phase: [water( NH 4 HCO 3 )-ACN];B%: 67%-97%,10min) to give methyl 3-bromo-4-[4-[2-[tert-butoxycarbonyl(methyl)amino]ethyl]phen oxy]benzoate (1.5 g, 90% yield). [0464] Step 2: Preparation of methyl 3-bromo-4-[4-[2-(methylamino) ethyl] phenoxy] benzoate. To a solution of methyl 3-bromo-4-[4-[2-[tert-butoxycarbonyl (methyl) amino] ethyl] phenoxy] benzoate (1.4 g, 3.0 mmol, 1 equiv) in DCM (10 mL) was added TFA (4 mL). The mixture was stirred at 25 °C for 0.5 h. The mixture was concentrated under reduced pressure to give methyl 3-bromo-4-[4-[2-(methylamino) ethyl] phenoxy] benzoate (1.4 g, crude, TFA) as a yellow oil. [0465] Step 3: Preparation of 2-[3-bromo-4-[4-[2-(methylamino)ethyl]phenoxy]phenyl] propan-2-ol. To a solution of methyl 3-bromo-4-[4-[2-(methylamino) ethyl] phenoxy] benzoate (1.4 g, 3.0 mmol, 1 equiv, TFA) in THF (15 mL) was added MeMgBr (3 M, 15.2 mL, 15 equiv) at 0°C. The mixture was stirred at 25 °C for 12 h. The mixture was purified by prep-HPLC (column: Waters Xbridge C18150*50mm* 10um;mobile phase: [water( NH4HCO3)-ACN];B%: 32%- 62%,10min) to give 2-[3-bromo-4-[4-[2-(methylamino)ethyl]phenoxy]phenyl]propan- 2-ol (400 mg, 35% yield) as a yellow oil. [0466] Step 4: Preparation of 4-[5-(1-hydroxy-1-methyl-ethyl)-2-[4-[2-(methylamino) ethyl]phenoxy]phenyl]-6-methyl-1-(p-tolylsulfonyl)pyrrolo[2, 3-c]pyridin-7-one. To a solution of 6-methyl-1-(p-tolylsulfonyl)-4-(4,4,5,5-tetramethyl-1,3,2-di oxaborolan-2-yl)pyrrolo[2,3- c]pyridin-7-one (444 mg, 1.0 mmol, 1.1 equiv) and 2-[3-bromo-4-[4-[2- (methylamino)ethyl]phenoxy]phenyl]propan-2-ol (343 mg, 942 μmol, 1 equiv) in THF (5 mL) H2O (1 mL) was added K3PO4 (599 mg, 2.8 mmol, 3 equiv) and SPhos Pd G3 (73.4 mg, 94 μmol, 0.1 equiv). The mixture was stirred at 60 °C for 12 h. The mixture was purified by prep-HPLC (column: Waters Xbridge C18150*50mm* 10um;mobile phase: [water( NH 4 HCO 3 )-ACN];B%: 38%-58%,10min) to give 4-[5-(1-hydroxy-1-methyl-ethyl)-2-[4-[2- (methylamino)ethyl]phenoxy]phenyl]-6-methyl-1-(p-tolylsulfon yl)pyrrolo[2,3-c]pyridin-7-one (245 mg, 44% yield) as a yellow oil. [0467] Step 5: Preparation of 4-[5-(1-hydroxy-1-methyl-ethyl)-2-[4-[2-(methylamino) ethyl]phenoxy]phenyl]-6-methyl-1H-pyrrolo[2,3-c]pyridin-7-on e. To a solution of 4-[5-(1- hydroxy-1-methyl-ethyl)-2-[4-[2-(methylamino) ethyl] phenoxy] phenyl]-6-methyl-1-(p- tolylsulfonyl) pyrrolo [2, 3-c] pyridin-7-one (240 mg, 410 μmol, 1 equiv) in MeOH (2 mL) was added KOH (115 mg, 2.0 mmol, 5 equiv). The mixture was stirred at 25 °C for 0.5 h. The mixture was diluted with water (10 mL) and extracted with DCM (20 mL * 3). The combined organic layers were washed with brine (20 mL), dried over Na 2 SO 4 , filtered and concentrated to give 4-[5- (1-hydroxy-1-methyl-ethyl)-2-[4-[2-(methylamino)ethyl]phenox y]phenyl]-6-methyl-1H- pyrrolo[2,3-c]pyridin-7-one (150 mg, crude). Step 6: Preparation of N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]-6-[2-[4-[4-(1-hyd roxy-1- methyl-ethyl)-2-(6-methyl-7-oxo-1H-pyrrolo[2,3-c]pyridin-4-y l)phenoxy]phenyl]ethyl- methyl-amino]pyridazine-3-carboxamide. To a solution of 6-chloro-N-[4-(3-chloro-4-cyano- phenoxy)cyclohexyl]pyridazine-3-carboxamide (75mg, 191 μmol, 1.1 equiv) and 4-[5-(1-hydroxy- 1-methyl-ethyl)-2-[4-[2-(methylamino)ethyl]phenoxy]phenyl]-6 -methyl-1H-pyrrolo[2,3-c]pyridin- 7-one (75 mg, 174 μmol, 1 equiv) in NMP (6 mL) was added K 2 CO 3 (120 mg, 869 μmol, 5 equiv). The mixture was stirred at 50 °C for 1 h. The residue was purified by prep-HPLC (column: Unisil 3-100 C18 Ultra 150*50mm*3 μm;mobile phase: [water(FA)-ACN];B%: 54%-84%,7min) to give N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]-6-[2-[4-[4-(1-hyd roxy-1-methyl-ethyl)-2-(6-methyl- 7-oxo-1H-pyrrolo[2,3-c]pyridin-4-yl)phenoxy]phenyl] ethyl-methyl-amino]pyridazine-3- carboxamide (21.5 mg, 14% yield) as an off-white solid. 1 H NMR (400 MHz, MeOD) δ 7.89 - 7.83 (m, 1H), 7.74 - 7.67 (m, 2H), 7.54 - 7.47 (m, 1H), 7.36 - 7.32 (m, 1H), 7.24 - 7.18 (m, 2H), 7.09 - 7.02 (m, 3H), 6.99 - 6.94 (m, 2H), 6.70 - 6.66 (m, 2H), 6.39 - 6.35 (m, 1H), 4.72 - 4.47 (m, 2H), 4.05 - 3.95 (m, 1H), 3.85 (br t, J = 6.7 Hz, 2H), 3.62 (s, 3H), 2.98 - 2.92 (m, 3H), 2.87-2.85 (m, 2H), 2.25 - 2.09 (m, 4H), 1.66-1.64 (m, 4H), 1.61 (s, 6H). LC-MS: MS (ES + ): RT = 2.659 min, m/z = 786.2 [M + H + ]; LCMS Method 25. EXAMPLE 19 – Synthesis of N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]-6-[4-[4-[4-(1- hydroxy-1-methyl-ethyl)-2-(6-methyl-7-oxo-1H-pyrrolo[2,3-c]p yridin-4-yl)phenoxy]-N- methyl-anilino]-1-piperidyl]pyridazine-3-carboxamide (I-137)

[0468] Step 1: Preparation of tert-butyl 4-(4-hydroxyanilino)piperidine-1-carboxylate. To a solution of tert-butyl 4-oxopiperidine-1-carboxylate (5 g, 25.09 mmol, 1 equiv) in DCE (50 mL) was added NaBH(OAc)3 (15.96 g, 75.28 mmol, 3 equiv) and 4-aminophenol (2.74 g, 25.09 mmol, 1 equiv). The mixture was stirred at 20 °C for 1 h and concentrated. The residue was purified by prep-HPLC (column: Kromasil Eternity XT 250*80mm*10um; mobile phase: [water( NH 4 HCO 3 )- ACN]; B %: 25 %-55 %, 20 min) to give tert-butyl 4-(4-hydroxyanilino) piperidine-1-carboxylate (3 g, 41 % yield). [0469] Step 2: Preparation of tert-butyl 4-(4-hydroxy-N-methyl-anilino)piperidine-1- carboxylate. To a solution of tert-butyl 4-(4-hydroxyanilino)piperidine-1-carboxylate (2.2 g, 7.52 mmol, 1 equiv) in MeOH (30 mL) was added NaBH 3 CN (946 mg, 15.05 mmol, 2 equiv) and formaldehyde (2.26 g, 75.25 mmol, 10 equiv). The mixture was stirred at 20 °C for 1 h. The mixture was filtered and concentrated to give crude product tert-butyl 4-(4-hydroxy-N-methyl- anilino)piperidine-1-carboxylate (2.3 g). [0470] Step 3: Preparation of tert-butyl 4-[4-(2-bromo-4-methoxycarbonyl-phenoxy)-N- methyl-anilino]piperidine-1-carboxylate. To a solution of tert-butyl 4-(4-hydroxy-N-methyl- anilino)piperidine-1-carboxylate (3 g, 9.79 mmol, 1 equiv) in DMSO (2 mL) was added Cs 2 CO 3 (6.38 g, 19.58 mmol, 2 equiv) and methyl 3-bromo-4-fluoro-benzoate (2.28 g, 9.79 mmol, 1 equiv). The mixture was stirred at 50 °C for 1 h and concentrated. The residue was purified by prep-HPLC (column: Waters Xbridge C18150*50mm* 10um; mobile phase: [water( NH 4 HCO 3 )- ACN]; B %: 67 %-97 %, 10 min) to give tert-butyl 4-[4-(2-bromo-4-methoxycarbonyl-phenoxy)- N-methyl-anilino]piperidine-1-carboxylate (2.2 g, 43 % yield). [0471] Step 4: Preparation of methyl 3-bromo-4-[4-[methyl(4-piperidyl)amino]phenoxy] benzoate. To a solution of tert-butyl 4-[4-(2-bromo-4-methoxycarbonyl-phenoxy)-N-methyl- anilino]piperidine-1-carboxylate (2.2 g, 4.24 mmol, 1 equiv) in DCM (20 mL) was added TFA (10 mL).The mixture was stirred at 20 °C for 0.5 h. The mixture was filtered and concentrated to give crude product methyl 3-bromo-4-[4-[methyl(4-piperidyl)amino]phenoxy]benzoate (2.25 g, TFA salt). [0472] Step 5: Preparation of 2-[3-bromo-4-[4-[methyl(4-piperidyl)amino]phenoxy] phenyl]propan-2-ol. To a solution of methyl 3-bromo-4-[4-[methyl(4-piperidyl)amino] phenoxy]benzoate (2.25 g, 4.22 mmol, 1 equiv, TFA salt) in THF (20 mL) was added MeMgBr (3 M, 35.2 mL, 25 equiv) at 0 °C. The mixture was stirred at 20 °C for 1 h. The residue was purified by prep-HPLC (column: Waters Xbridge C18150*50mm* 10um; mobile phase: [water( NH 4 HCO 3 )-ACN]; B %: 51 %-81 %, 10 min) to give 2-[3-bromo-4-[4-[methyl(4- piperidyl)amino]phenoxy]phenyl]propan-2-ol (730 mg, 41 % yield). [0473] Step 6: Preparation of 4-[5-(1-hydroxy-1-methyl-ethyl)-2-[4-[methyl(4-piperidyl) amino]phenoxy]phenyl]-6-methyl-1-(p-tolylsulfonyl)pyrrolo[2, 3-c]pyridin-7-one. To a solution of 2-[3-bromo-4-[4-[methyl(4-piperidyl)amino]phenoxy]phenyl]pro pan-2-ol (680 mg, 1.62 mmol, 1 equiv) in THF (10 mL) and H 2 O (2 mL) was added SPhos Pd G3 (127 mg, 162 μmol, 0.1 equiv), 6-methyl-1-(p-tolylsulfonyl)-4-(4,4,5,5-tetramethyl-1,3,2-di oxaborolan-2- yl)pyrrolo[2,3-c]pyridin-7-one (833 mg, 1.95 mmol, 1.2 equiv) and K3PO4 (1.03 g, 4.86 mmol, 3 equiv). The mixture was stirred at 60 °C for 1 h. The residue was purified by prep-HPLC (column: Waters Xbridge C18150*50mm* 10um; mobile phase: [water( NH 4 HCO 3 )-ACN]; B %: 57 %-87 %, 10 min) to give 4-[5-(1-hydroxy-1-methyl-ethyl)-2-[4-[methyl(4-piperidyl) amino]phenoxy]phenyl]-6-methyl-1-(p-tolylsulfonyl)pyrrolo[2, 3-c]pyridin-7-one (360 mg, 35 % yield). [0474] Step 7: Preparation of 4-[5-(1-hydroxy-1-methyl-ethyl)-2-[4-[methyl(4-piperidyl) amino]phenoxy]phenyl]-6-methyl-1H-pyrrolo[2,3-c]pyridin-7-on e. To a solution of 4-[5-(1- hydroxy-1-methyl-ethyl)-2-[4-[methyl(4-piperidyl)amino]pheno xy]phenyl]-6-methyl-1-(p- tolylsulfonyl)pyrrolo[2,3-c]pyridin-7-one (180 mg, 281 μmol, 1 equiv) in MeOH (3 mL) was added KOH (79 mg, 1.40 mmol, 5 equiv). The mixture was stirred at 20 °C for 0.5 h and concentrated to give 4-[5-(1-hydroxy-1-methyl-ethyl)-2-[4-[methyl(4-piperidyl)ami no] phenoxy] phenyl]-6-methyl-1H-pyrrolo[2,3-c]pyridin-7-one (116 mg, 85 % yield). Step 8: Preparation of N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]-6-[4-[4-[4-(1-hyd roxy-1- methyl-ethyl)-2-(6-methyl-7-oxo-1H-pyrrolo[2,3-c]pyridin-4-y l)phenoxy]-N-methyl-anilino]- 1-piperidyl]pyridazine-3-carboxamide. To a solution of 4-[5-(1-hydroxy-1-methyl-ethyl)-2-[4- [methyl(4-piperidyl)amino]phenoxy]phenyl]-6-methyl-1H-pyrrol o[2,3-c]pyridin-7-one (116 mg, 239 μmol, 1 equiv) in NMP (2 mL) was added K2CO3 (99 mg, 715 μmol, 3 equiv) and 6-chloro-N- [4-(3-chloro-4-cyano-phenoxy)cyclohexyl]pyridazine-3-carboxa mide (93 mg, 238 μmol, 1 equiv). The mixture was stirred at 50 °C for 12 h and concentrated. The residue was purified by prep- HPLC (column: Phenomenex C1875*30mm*3um; mobile phase: [water(FA)-ACN]; B %: 25 %- 55 %, 7 min) to give N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]-6-[4-[4-[4-(1-hyd roxy-1- methyl-ethyl)-2-(6-methyl-7-oxo-1H-pyrrolo[2,3-c]pyridin-4-y l)phenoxy]-N-methyl-anilino]-1- piperidyl]pyridazine-3-carboxamide (50 mg, 24 % yield). 1 H NMR (400 MHz, MeOD) δ 7.90 (d, J=9.64 Hz, 1 H), 7.67 - 7.70 (m, 1 H), 7.63 (d, J=2.52 Hz, 1 H), 7.55 - 7.58 (m, 1 H), 7.43-7.46 (m, 1 H), 7.29 - 7.36 (m, 2 H), 7.17 - 7.23 (m, 2 H), 7.02-7.05 (m, 1 H), 6.96 (d, J=8.52 Hz, 1 H), 6.80 - 6.88 (m, 2 H), 6.70 - 6.78 (m, 2 H), 6.36 (d, J=2.88 Hz, 1 H), 4.59 (d, J=13.36 Hz, 3 H), 3.89 - 4.04 (m, 1 H), 3.68 - 3.79 (m, 1 H), 3.63 (s, 3 H), 3.01 - 3.16 (m, 2 H), 2.66 (s, 3 H), 2.16 - 2.27 (m, 2 H), 2.05 - 2.14 (m, 2 H), 1.75 - 1.84 (m, 2 H), 1.61 - 1.73 (m, 6 H), 1.58 (s, 6 H). LC-MS: MS (ES + ): RT = 2.178 min, m/z = 841.2 [M + H + ]; LCMS Method 25. EXAMPLE 20 – Synthesis of N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]-2-[4-[2-[3-[4- (ethylsulfonylmethyl)-2-(6-methyl-7-oxo-1H-pyrrolo[2,3-c]pyr idin-4-yl)phenoxy] phenoxy]ethoxy]-1-piperidyl]pyrimidine-5-carboxamide (I-151)

[0475] Step 1: Preparation of tert-butyl 4-(2-benzyloxyethoxy)piperidine-1-carboxylate. To a mixture of NaH (1.9 g, 48.9 mmol, 60 % purity, 3.0 equiv) in THF (50 mL) was added tert- butyl 4-hydroxypiperidine-1-carboxylate (9.8 g, 48.9 mmol, 3.0 equiv) at 0 °C. The mixture was stirred at 0 °C for 0.5 h. Then 2-benzyloxyethyl 4-methylbenzenesulfonate (5 g, 16.3 mmol, 1.0 equiv) was added at 0 °C. The mixture was stirred at 25 °C for 12 h under N2. The mixture was poured into ice saturated NH4Cl solution (50 mL). The solution was extracted with EA (3 x 50 mL). The combined organic layers were washed with saturated brine (50 mL), dried, filtered and concentrated to give a residue. The residue was purified by column chromatography (SiO 2 , Petroleum ether/Ethyl acetate = 5/1 to 3/1) to give the tert-butyl 4-(2-benzyloxyethoxy) piperidine- 1-carboxylate (4.2 g, 12.5 mmol, 76 % yield) as a yellow oil. [0476] Step 2: Preparation of tert-butyl 4-(2-hydroxyethoxy)piperidine-1-carboxylate. To a solution of tert-butyl 4-(2-benzyloxyethoxy)piperidine-1-carboxylate (4.2 g, 12.5 mmol, 1.0 equiv) in MeOH (40 mL) was added Pd/C (1 g, 10 % purity). The mixture was stirred at 25 °C for 12 h under H2. The mixture was cooled to 25 °C and filtered. The filtrate was concentrated to get the tert-butyl 4-(2-hydroxyethoxy)piperidine-1-carboxylate (2.8 g, 11.6 mmol, 93 % yield) as a yellow oil. [0477] Step 3: Preparation of tert-butyl 4-[2-(p-tolylsulfonyloxy)ethoxy]piperidine-1- carboxylate. To a solution of tert-butyl 4-(2-hydroxyethoxy) piperidine-1-carboxylate (1.5 g, 6.1 mmol, 1.0 equiv) in DCM (15 mL) was added TEA (1.8 g, 18.3 mmol, 2.5 mL, 3.0 equiv) and TosCl (1.7 g, 9.1 mmol, 1.5 equiv). The mixture was stirred at 25 °C for 12 h. The mixture was added 30 mL water, the aqueous phase was extracted with DCM (30 mL × 3).The combined organic phase was washed with brine (30 mL × 3), dried with anhydrous sodium sulfate, filtered and concentrated in vacuum. The residue was purified by column chromatography (SiO 2 , Petroleum ether/Ethyl acetate = 5/1 to 3/1) to give the tert-butyl 4-[2-(p-tolylsulfonyloxy) ethoxy]piperidine-1-carboxylate (2 g, 5.0 mmol, 81 % yield) as a yellow oil. [0478] Step 4: Preparation of tert-butyl 4-[2-(3-hydroxyphenoxy)ethoxy]piperidine-1- carboxylate. A mixture of tert-butyl 4-[2-(p-tolylsulfonyloxy)ethoxy]piperidine-1-carboxylate (3.3 g, 8.3 mmol, 1.0 equiv), benzene-1,3-diol (2.7 g, 24 mmol, 4.0 mL, 3.0 equiv), Cs 2 CO 3 (8.0 g, 25 mmol, 3.0 equiv) in DMSO (35 mL) was degassed and purged with N 2 for 3 times, and then the mixture was stirred at 60 °C for 12 h under N2 atmosphere. The mixture was concentrated to give a residue and was purified by prep-HPLC (column: YMC Triart C18250*50mm*7um;mobile phase: [water( NH 4 HCO 3 )-ACN];B%: 40%-70%,22min) to give the tert-butyl 4-[2-(3- hydroxyphenoxy)ethoxy]piperidine-1-carboxylate (1.8 g, 5.0 mmol, 66% yield) as a white solid. [0479] Step 5: Preparation of tert-butyl 4-[2-[3-(2-bromo-4-formyl-phenoxy)phenoxy] ethoxy]piperidine-1-carboxylate. A mixture of tert-butyl 4-[2-(3-hydroxyphenoxy)ethoxy] piperidine-1-carboxylate (1.8 g, 5.3 mmol, 1.0 equiv), 3-bromo-4-fluoro-benzaldehyde (1.3 g, 6.0 mmol, 1.2 equiv), Cs 2 CO 3 (5.0 g, 15 mmol, 3.0 equiv) in DMSO (20 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 60 °C for 2 h under N2 atmosphere. The mixture was poured into H2O (100 mL) and extracted with EA(50 mL × 2). The combined organic layer was washed with brine (100 mL × 2) and dried over Na 2 SO 4 . The mixture was concentrated to get the residue and was purified by column chromatography (SiO 2 , Petroleum ether/Ethyl acetate = 3/1 to 1/1) to give the tert-butyl 4-[2-[3-(2-bromo-4-formyl- phenoxy)phenoxy]ethoxy]piperidine-1-carboxylate (2.3 g, 4.3 mmol, 83% yield) as a yellow oil. [0480] Step 6: Preparation of tert-butyl 4-[2-[3-[2-bromo-4-(hydroxymethyl)phenoxy] phenoxy]ethoxy]piperidine-1-carboxylate. A mixture of tert-butyl 4-[2-[3-(2-bromo-4-formyl- phenoxy)phenoxy]ethoxy]piperidine-1-carboxylate (1.8 g, 3.4 mmol, 1.0 equiv), NaBH 4 (510 mg, 13.0 mmol, 4.0 equiv) in EtOH (20 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 25 °C for 1 h under N 2 atmosphere. The mixture was poured into NH 4 Cl (30 mL) and extracted with EA (20 mL x 2), organic layer was washed with brine (40 mL) and dried over Na2SO4. Then the organic phase was concentrated to give the residue and was purified by column chromatography (SiO 2 , Petroleum ether/Ethyl acetate = 3/1 to 1/1) to give the tert-butyl 4- [2-[3-[2-bromo-4-(hydroxymethyl)phenoxy]phenoxy]ethoxy]piper idine-1-carboxylate (1.6 g, 3.0 mmol, 88% yield) as a colorless oil. [0481] Step 7: Preparation of tert-butyl 4-[2-[3-[2-bromo-4-(bromomethyl)phenoxy] phenoxy]ethoxy]piperidine-1-carboxylate. A mixture of tert-butyl 4-[2-[3-[2-bromo-4- (hydroxymethyl)phenoxy]phenoxy]ethoxy]piperidine-1-carboxyla te (600 mg, 1.1 mmol, 1.0 equiv), PBr 3 (466 mg, 1.7 mmol, 1.5 equiv) in NMP (11 mL) was degassed and purged with N 2 for 3 times, and then the mixture was stirred at 0 °C for 2 h under N2 atmosphere. The mixture was poured into saturated NaHCO 3 (100 mL) and extracted with EA (80 mL x 2). The combined organic layers were washed with brine (60 mL x 2), dried over Na 2 SO 4 , filtered and concentrated under reduced pressure to give a residue. The residue was used for next step without further purification to give the tert-butyl 4-[2-[3-[2-bromo-4-(bromomethyl) phenoxy]phenoxy] ethoxy]piperidine-1-carboxylate (630 mg, 1.0 mmol, 94% yield) as a yellow oil. [0482] Step 8: Preparation of tert-butyl 4-[2-[3-[2-bromo-4-(ethylsulfonylmethyl) phenoxy]phenoxy]ethoxy]piperidine-1-carboxylate. A mixture of tert-butyl 4-[2-[3-[2-bromo-4- (bromomethyl)phenoxy]phenoxy]ethoxy]piperidine-1-carboxylate (630 mg, 1.0 mmol, 1.0 equiv), sodium;ethanesulfinate (250 mg, 2.1 mmol, 2.0 equiv) in DMF (9.0 mL) was degassed and purged with N 2 for 3 times, and then the mixture was stirred at 60 °C for 1 h under N 2 atmosphere. The mixture was poured into H2O (50 mL x 2) and extracted with EA (15 mL x 2). The combined organic layer was washed with brine(30 mL x 2) and dried over Na2SO4 The mixture was concentrated to get the residue and was purified by column chromatography (SiO 2 , Petroleum ether/Ethyl acetate = 3/1 to 1/1) to give the tert-butyl 4-[2-[3-[2-bromo-4- (ethylsulfonylmethyl)phenoxy]phenoxy]ethoxy]piperidine-1-car boxylate (500 mg, 835 μmol, 78% yield) as a yellow oil. [0483] Step 9: Preparation of tert-butyl 4-[2-[3-[4-(ethylsulfonylmethyl)-2-[6-methyl-7- oxo-1-(p-tolylsulfonyl)pyrrolo[2,3-c]pyridin-4-yl]phenoxy]ph enoxy]ethoxy]piperidine-1- carboxylate. A mixture of tert-butyl 4-[2-[3-[2-bromo-4-(ethylsulfonylmethyl)phenoxy] phenoxy]ethoxy]piperidine-1-carboxylate (150 mg, 250 μmol, 1.0 equiv), 6-methyl-1-(p- tolylsulfonyl)-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-y l)pyrrolo[2,3-c]pyridin-7-one (129 mg, 301 μmol, 1.2 equiv)6-methyl-1-(p-tolylsulfonyl)-4-(4,4,5,5-tetramethyl-1, 3,2-dioxaborolan- 2-yl)pyrrolo[2,3-c]pyridin-7-one (129 mg, 301 μmol, 1.2 equiv), K3PO4 (160 mg, 752 μmol, 3.0 equiv), SPhos Pd G3 (20 mg, 25 μmol, 0.1 equiv) and in THF (5.0 mL) ,H 2 O (1.0 mL) was degassed and purged with N 2 for 2mins, and then the mixture was stirred at 60 °C for 3 h under N 2 atmosphere. The residue was used for next step without further purification to give the tert-butyl 4- [2-[3-[4-(ethylsulfonylmethyl)-2-[6-methyl-7-oxo-1-(p-tolyls ulfonyl)pyrrolo[2,3-c]pyridin-4- yl]phenoxy]phenoxy]ethoxy]piperidine-1-carboxylate ( crude) as a brown liquid. [0484] Step 10: Preparation of tert-butyl 4-[2-[3-[4-(ethylsulfonylmethyl)-2-(6-methyl-7- oxo-1H-pyrrolo[2,3-c]pyridin-4-yl)phenoxy]phenoxy]ethoxy]pip eridine-1-carboxylate. A mixture of tert-butyl 4-[2-[3-[4-(ethylsulfonylmethyl)-2-[6-methyl-7-oxo-1-(p-toly lsulfonyl) pyrrolo[2,3-c]pyridin-4-yl]phenoxy]phenoxy]ethoxy]piperidine -1-carboxylate (120 mg, 146 μmol, 1.0 equiv), NaOH (29 mg, 732 μmol, 5.0 equiv) H 2 O (1.0 mL) in MeOH (1.0 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 60 °C for 12 h under N2 atmosphere. The mixture was poured into H2O (20 mL x 2) and extracted with DCM (20 mL x 2). The combined organic layer was washed with brine (30 mL) and dried over Na 2 SO 4 . The residue was purified by prep-TLC (SiO2, Dichloromethane : Methanol = 10:1) to give the tert-butyl 4-[2- [3-[4-(ethylsulfonylmethyl)-2-(6-methyl-7-oxo-1H-pyrrolo[2,3 -c]pyridin-4- yl)phenoxy]phenoxy]ethoxy]piperidine-1-carboxylate (180 mg, 270 μmol, 92% yield) as a yellow gum. [0485] Step 11: Preparation of 4-[5-(ethylsulfonylmethyl)-2-[3-[2-(4-piperidyloxy) ethoxy]phenoxy]phenyl]-6-methyl-1H-pyrrolo[2,3-c]pyridin-7-o ne. A mixture of tert-butyl 4- [2-[3-[4-(ethylsulfonylmethyl)-2-(6-methyl-7-oxo-1H-pyrrolo[ 2,3-c]pyridin-4-yl)phenoxy] phenoxy]ethoxy]piperidine-1-carboxylate (120 mg, 180 μmol, 1.0 equiv) in TFA (1.0 mL), DCM (3.0 mL) was degassed and purged with N 2 for 3 times, and then the mixture was stirred at 25 °C for 1 h under N2 atmosphere. The mixture was concentrated to give a residue. The mixture is used for the next step of the reaction to give the 4-[5-(ethylsulfonylmethyl)-2-[3-[2-(4- piperidyloxy)ethoxy]phenoxy]phenyl]-6-methyl-1H-pyrrolo[2,3- c]pyridin-7-one (102 mg, 180 μmol) as a yellow gum. Step 12: Preparation of N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]-2-[4-[2-[3-[4- (ethylsulfonylmethyl)-2-(6-methyl-7-oxo-1H-pyrrolo[2,3-c]pyr idin-4-yl)phenoxy] phenoxy]ethoxy]-1-piperidyl]pyrimidine-5-carboxamide. A mixture of 4-[5- (ethylsulfonylmethyl)-2-[3-[2-(4-piperidyloxy)ethoxy]phenoxy ]phenyl]-6-methyl-1H-pyrrolo[2,3- c]pyridin-7-one (100 mg, 147 μmol, 1.0 equiv, TFA), 2-chloro-N-[4-(3-chloro-4-cyano- phenoxy)cyclohexyl]pyrimidine-5-carboxamide (69 mg, 176 μmol, 1.2 equiv), K 2 CO 3 (102 mg, 736 μmol, 5 equiv) in DMF (2.0 mL) was degassed and purged with N 2 for 3 times, and then the mixture was stirred at 60 °C for 2 h under N2 atmosphere. Filter mixture ,and the residue was purified by prep-HPLC (column: Phenomenex luna C18150*25mm* 10um;mobile phase: [water(FA)-ACN];B%: 48%-78%,8min) to give the N-[4-(3-chloro-4-cyano-phenoxy) cyclohexyl]-2-[4-[2-[3-[4-(ethylsulfonylmethyl)-2-(6-methyl- 7-oxo-1H-pyrrolo[2,3-c]pyridin-4- yl)phenoxy]phenoxy]ethoxy]-1-piperidyl]pyrimidine-5-carboxam ide (59 mg, 63 μmol, 43% yield, 98% purity) as a white solid. 1 H NMR: (400 MHz, MeOD-d4) δ = 8.72 (d, J = 1.6 Hz, 2H), 7.73 - 7.62 (m, 2H), 7.45 (d, J = 7.6 Hz, 1H), 7.36 - 7.29 (m, 1H), 7.24 (s, 1H), 7.19 (s, 1H), 7.14 - 7.07 (m, 2H), 7.04 (d, J = 8.0 Hz, 1H), 6.59 (d, J = 7.6 Hz, 1H), 6.43 (d, J = 9.2 Hz, 3H), 4.59 (s, 1H), 4.50 - 4.43 (m, 3H), 4.28 - 4.20 (m, 2H), 4.00 (d, J = 3.6 Hz, 2H), 3.95 - 3.88 (m, 1H), 3.79 (s, 2H), 3.73 - 3.66 (m, 1H), 3.63 - 3.57 (m, 4H), 3.14 - 3.08 (m, 2H), 2.24 - 2.16 (m, 2H), 2.09 (s, 2H), 1.96 - 1.85 (m, 2H), 1.63 - 1.51 (m, 6H), 1.42 - 1.35 (m, 3H) LC-MS: MS (ES + ): RT = 2.909 min, m/z = 920.5 [M +H + ] . LC-MS Method: 25. EXAMPLE 21 – Synthesis of N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]-6-[4-[2-[4-[4- (ethylsulfonylmethyl)-2-(6-methyl-7-oxo-1H-pyrrolo[2,3-c]pyr idin-4-yl)phenoxy] phenyl]ethyl]-1-piperidyl]pyridazine-3-carboxamide (I-154)

[0486] Step 1: Preparation of tert-butyl 4-[2-[4-(2-bromo-4-formyl-phenoxy) phenyl] ethyl]piperidine-1-carboxylate. To a solution of tert-butyl 4-[2-(4-hydroxyphenyl)ethyl] piperidine-1-carboxylate (1 g, 3.27 mmol, 1 equiv) in DMSO (5 mL) was added Cs 2 CO 3 (2.13 g, 6.55 mmol, 2 equiv) and 3-bromo-4-fluoro-benzaldehyde (665 mg, 3.27 mmol, 1 equiv). The mixture was stirred at 80 °C for 1 h and concentrated. The residue was purified by column chromatography (SiO 2 , Petroleum ether/Ethyl acetate=5/1 to 3/1) to give tert-butyl 4-[2-[4-(2- bromo-4-formyl-phenoxy)phenyl]ethyl]piperidine-1-carboxylate (1.17 g, 73 % yield). [0487] Step 2: Preparation of tert-butyl 4-[2-[4-[2-bromo-4-(hydroxymethyl) phenoxy] phenyl]ethyl]piperidine-1-carboxylate. To a solution of tert-butyl 4-[2-[4-(2-bromo-4-formyl- phenoxy)phenyl]ethyl]piperidine-1-carboxylate (1.17 g, 2.40 mmol, 1 equiv) in DCM (10 mL) was added NaBH 4 (100 mg, 2.64 mmol, 1.1 equiv). The mixture was stirred at 20 °C for 0.5 h. The mixture was filtered and concentrated to give crude tert-butyl 4-[2-[4-[2-bromo-4- (hydroxymethyl)phenoxy]phenyl]ethyl]piperidine-1-carboxylate (1.15 g) was a yellow oil [0488] Step 3: Preparation of 4-[2-[4-[2-bromo-4-(bromomethyl)phenoxy]phenyl] ethyl]piperidine. To a solution of tert-butyl 4-[2-[4-[2-bromo-4-(hydroxymethyl)phenoxy] phenyl]ethyl]piperidine-1-carboxylate (800 mg, 1.63 mmol, 1 equiv) in DCM (10 mL) was added PBr3 (662 mg, 2.45 mmol, 1.5 equiv) at 0 °C. The mixture was stirred at 20 °C for 0.5 h and concentrated to give crude product 4-[2-[4-[2-bromo-4-(bromomethyl)phenoxy]phenyl] ethyl]piperidine (400 mg) was a yellow oil. [0489] Step 4: Preparation of tert-butyl 4-[2-[4-[2-bromo-4-(bromomethyl)phenoxy] phenyl]ethyl]piperidine-1-carboxylate. To a solution of 4-[2-[4-[2-bromo-4-(bromomethyl) phenoxy]phenyl]ethyl]piperidine (400 mg, 883 μmol, 1 equiv) in DCM (5 mL) was added Boc2O (385 mg, 1.77 mmol, 2 equiv) and DIEA (342 mg, 2.65 mmol, 3 equiv) .The mixture was stirred at 20 °C for 0.5 h. The mixture was filtered and concentrated. The residue was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate=10/1 to 5/1) to give tert-butyl 4-[2-[4-[2- bromo-4-(bromomethyl)phenoxy]phenyl]ethyl]piperidine-1-carbo xylate (180 mg, 37 % yield) was a yellow oil. [0490] Step 5: Preparation of tert-butyl 4-[2-[4-[2-bromo-4-(ethylsulfonylmethyl) phenoxy]phenyl]ethyl]piperidine-1-carboxylate. To a solution of tert-butyl 4-[2-[4-[2-bromo-4- (bromomethyl)phenoxy]phenyl]ethyl]piperidine-1-carboxylate (180 mg, 325 μmol, 1 eq) in DMF (5 mL) was added sodium;ethanesulfinate (189 mg, 1.63 mmol, 5 equiv). The mixture was stirred at 20 °C for 0.5 h. The mixture was filtered and concentrated to give crude tert-butyl 4-[2-[4-[2- bromo-4-(ethylsulfonylmethyl)phenoxy]phenyl]ethyl]piperidine -1-carboxylate (184 mg). [0491] Step 6: Preparation of tert-butyl 4-[2-[4-[4-(ethylsulfonylmethyl)-2-[6-methyl-7- oxo-1-(p-tolylsulfonyl)pyrrolo[2,3-c]pyridin-4-yl]phenoxy]ph enyl]ethyl]piperidine-1- carboxylate. To a solution of tert-butyl 4-[2-[4-[2-bromo-4-(ethylsulfonylmethyl)phenoxy] phenyl]ethyl]piperidine-1-carboxylate (184 mg, 325 μmol, 1 equiv) in THF (5 mL), H 2 O (1 mL) was added K3PO4 (207 mg, 974 μmol, 3 equiv), 6-methyl-1-(p-tolylsulfonyl)-4-(4,4,5,5- tetramethyl-1,3,2-dioxaborolan-2-yl)pyrrolo[2,3-c]pyridin-7- one (209 mg, 487 μmol, 1.5 equiv) and SPhos Pd G3 (25 mg, 32 μmol, 0.1 equiv). The mixture was stirred at 60 °C for 3 h. The residue was purified by prep-TLC (SiO2, PE: EA = 1:2) to give tert-butyl 4-[2-[4-[4- (ethylsulfonylmethyl)-2-[6-methyl-7-oxo-1-(p-tolylsulfonyl)p yrrolo[2,3-c]pyridin-4- yl]phenoxy]phenyl]ethyl]piperidine-1-carboxylate (200 mg, 78 % yield). [0492] Step 7: Preparation of tert-butyl 4-[2-[4-[4-(ethylsulfonylmethyl)-2-(6-methyl-7- oxo-1H-pyrrolo[2,3-c]pyridin-4-yl)phenoxy]phenyl]ethyl]piper idine-1-carboxylate. To a solution of tert-butyl 4-[2-[4-[4-(ethylsulfonylmethyl)-2-[6-methyl-7-oxo-1-(p-toly lsulfonyl) pyrrolo[2,3-c]pyridin-4-yl]phenoxy]phenyl]ethyl]piperidine-1 -carboxylate (200 mg, 254 μmol, 1 equiv) in MeOH (5 mL) was added KOH (171 mg, 3.05 mmol, 12 equiv). The mixture was stirred at 20 °C for 0.5 h and concentrated to give crude product tert-butyl 4-[2-[4-[4- (ethylsulfonylmethyl)-2-(6-methyl-7-oxo-1H-pyrrolo[2,3-c]pyr idin-4-yl)phenoxy]phenyl] ethyl]piperidine-1-carboxylate (160 mg, 99 % yield). [0493] Step 8: Preparation of 4-[5-(ethylsulfonylmethyl)-2-[4-[2-(4-piperidyl)ethyl] phenoxy]phenyl]-6-methyl-1H-pyrrolo[2,3-c]pyridin-7-one. To a solution of tert-butyl 4-[2-[4- [4-(ethylsulfonylmethyl)-2-(6-methyl-7-oxo-1H-pyrrolo[2,3-c] pyridin-4-yl)phenoxy]phenyl] ethyl]piperidine-1-carboxylate (100 mg, 158 μmol, 1 eq) in DCM (2 mL) was added TFA (1 mL). The mixture was stirred at 20 °C for 0.5 h and concentrated to give crude 4-[5- (ethylsulfonylmethyl)-2-[4-[2-(4-piperidyl)ethyl]phenoxy]phe nyl]-6-methyl-1H-pyrrolo[2,3- c]pyridin-7-one (100 mg, TFA salt). [0494] Step 9: Preparation of N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]-6-[4-[2-[4-[4- (ethylsulfonylmethyl)-2-(6-methyl-7-oxo-1H-pyrrolo[2,3-c]pyr idin-4-yl)phenoxy] phenyl] ethyl]-1-piperidyl]pyridazine-3-carboxamide. To a solution of 4-[5-(ethylsulfonylmethyl)-2-[4- [2-(4-piperidyl)ethyl]phenoxy]phenyl]-6-methyl-1H-pyrrolo[2, 3-c]pyridin-7-one (100 mg, 154 μmol, 1 equiv, TFA salt) in NMP (1 mL) was added K2CO3 (64 mg, 463 μmol, 3 equiv) and 6- chloro-N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]pyridazine- 3-carboxamide (66 mg, 170 μmol, 1.1 equiv). The mixture was stirred at 50 °C for 3 h and purified by prep-HPLC (column: Phenomenex luna C18150*25mm* 10um; mobile phase: [water(FA)-ACN]; B %: 65 %-95 %, 10 min) to give N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]-6-[4-[2-[4-[4-(et hylsulfonylmethyl)-2- (6-methyl-7-oxo-1H-pyrrolo[2,3-c]pyridin-4-yl)phenoxy] phenyl]ethyl]-1-piperidyl]pyridazine-3- carboxamide (40 mg, 28 % yield). 1 H NMR (400 MHz, MeOD) δ 7.86 - 7.97 (m, 1 H), 7.59 - 7.72 (m, 2 H), 7.39 - 7.47 (m, 1 H), 7.30 - 7.37 (m, 2 H), 7.21 (d, J=18.00 Hz, 2 H), 7.03-7.07 (m, 4 H), 6.76 (d, J=8.24 Hz, 2 H), 6.43 (d, J=2.24 Hz, 1 H), 4.38 - 4.51 (m, 5 H), 3.90 - 4.00 (m, 1 H), 3.61 (s, 3 H), 3.00 - 3.14 (m, 4 H), 2.57-2.61 (m, 2 H), 2.03 - 2.21 (m, 5 H), 1.81 - 1.91 (m, 2 H), 1.61- 1.65 (m, 4 H), 1.52 - 1.58 (m, 2 H), 1.35-1.39 (m, 3 H), 1.22 - 1.30 (m, 2 H). LC-MS: MS (ES + ): RT = 2.422 min, m/z = 888.2 [M + H + ]; LCMS Method 25. EXAMPLE 22 – Synthesis of N-((1r,4r)-4-(3-chloro-4-cyanophenoxy)cyclohexyl)-6-(4- ((((1r,3r)-3-(4-(2-hydroxypropan-2-yl)-2-(6-methyl-7-oxo-6,7 -dihydro-1H-pyrrolo[2,3- c]pyridin-4-yl)phenoxy)cyclobutyl)(methyl)amino)methyl)piper idin-1-yl)pyridazine-3- carboxamide (I-158)

[0495] Step 1: Preparation of methyl 3-bromo-4-((1r,3r)3-((tert-butoxycarbonyl) amino)cyclobutoxy) benzoate. To a solution of methyl 3-bromo-4-hydroxybenzoate (3.70 g, 16.0 mmol, 1.5 equiv), tert-butyl ((1s,3s)-3-hydroxycyclobutyl) carbamate (2.00 g, 10.7 mmol, 1.0 equiv) and PPh 3 (5.60 g, 21.4 mmol, 2.0 equiv) in THF (100 mL) was added dropwise DIAD (4.32 g, 21.4 mmol, 4.15 mL, 2.0 equiv) at 0°C. After addition, the mixture was stirred at 20 °C for 12 h. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (column: Welch Ultimate XB-SiOH 250*70*10um; mobile phase: [Heptane-EtOH(0.1%NH3H2O)];B%: 1%-35%,15min). methyl 3-bromo-4-((1r,3r)3-((tert- butoxycarbonyl)amino)cyclobutoxy) benzoate (4 g, 9.99 mmol, 93% yield) was obtained as a white solid. [0496] Step 2: Preparation of methyl 4-((1r,3r)-3-aminocyclobutoxy)-3-bromobenzoate. To a solution of methyl methyl 3-bromo-4-((1r,3r)3-((tert-butoxycarbonyl)amino)cyclobutoxy) benzoate (1.8 g, 4.5 mmol, 1.0 equiv) in DCM (20 mL) was added TFA (10 mL). The mixture was stirred at 20 °C for 1 h. The reaction mixture was concentrated under reduced pressure to give a crude methyl 4-((1r,3r)-3-aminocyclobutoxy)-3-bromobenzoate (1.8 g, 4.5 mmol, TFA salt) as a yellow gum. [0497] Step 3: Preparation of tert-butyl-4-((((1r,3r)-3-(2-bromo-4-(methoxycarbonyl) phenoxy)cyclobutyl)(methyl)amino)methyl)piperidine-1-carboxy late. To a solution of methyl 4-((1r,3r)-3-aminocyclobutoxy)-3-bromobenzoate (1.8 g, 4.4 mmol, 1.0 equiv, TFA salt) in DCM (40 mL) was added NaBH(OAc) 3 (2.76 g, 13.05 mmol, 3.0 equiv), TEA (2.20 g, 21.8 mmol, 3.0 mL, 5.0 equiv) and tert-butyl 4-formylpiperidine-1-carboxylate (926 mg, 4.35 mmol, 1.0 equiv). The mixture was stirred at 20 °C for 0.5 h. Then formaldehyde (388 mg, 4.79 mmol, 37% purity, 1.1 equiv) was added to the mixture. The mixture was stirred at 20 °C for 1 h. The reaction mixture was quenched by addition NaHCO 3 (20 mL) and then extracted with DCM (2 x 20 mL). The combined organic layers were washed with brine (20 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO 2 , EA/ MeOH = 10/1). tert-butyl-4-((((1r,3r)-3-(2-bromo-4- (methoxycarbonyl)phenoxy)cyclobutyl)(methyl)amino)methyl)pip eridine-1-carboxylate (2.2 g, 4.35 mmol, crude) was obtained as a yellow gum. [0498] Step 4: Preparation of methyl 3-bromo-4-((1r,3r)-3-(methyl(piperidin-4- ylmethyl)amino)cyclobutoxy)benzoate. To a solution of tert-butyl-4-((((1r,3r)-3-(2-bromo-4- (methoxycarbonyl)phenoxy)cyclobutyl)(methyl)amino)methyl)pip eridine-1-carboxylate ((2.4 g, 4.7 mmol, 1.0 equiv) in DCM (10 mL) was added TFA (5 mL).The mixture was stirred at 20 °C for 1 h. The reaction mixture was quenched by addition NaHCO3 (50 mL) at 0 °C, and then extracted with DCM (2 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 to give crude methyl 3- bromo-4-((1r,3r)-3-(methyl(piperidin-4-ylmethyl)amino)cyclob utoxy)benzoate (2 g, crude) as a yellow solid. [0499] Step 5: Preparation of 2-(3-bromo-4-((1r,3r)-3-(methyl(piperidin-4-ylmethyl) amino)cyclobutoxy)phenyl)propan-2-ol. To a solution of methyl 3-bromo-4-((1r,3r)-3- (methyl(piperidin-4-ylmethyl)amino)cyclobutoxy)benzoate (800 mg, 1.94 mmol, 1.0 equiv) in DCM (20 mL) was added MeMgBr (3 M, 6.48 mL, 10.0 equiv) at -78 °C. The mixture was stirred at 20 °C for 12 h. The reaction mixture was quenched by addition NH 4 Cl (20 mL) at 0 °C, and then diluted with extracted with EtOAc (20 mL). The combined organic layers were washed with brine (20mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (column: Waters Xbridge C18150*50mm*10um; mobile phase: [water (NH 4 HCO 3 )-ACN]; B%: 30%-60%, 10min) to give 2-(3-bromo-4-((1r,3r)-3- (methyl(piperidin-4-ylmethyl)amino)cyclobutoxy)phenyl)propan -2-ol (70 mg, 170 μmol, 8% yield) as a yellow gum. [0500] Step 6: Preparation of 4-(5-(2-hydroxypropan-2-yl)-2-((1r,3r)-3-(methyl (piperidin- 4-ylmethyl)amino)cyclobutoxy)phenyl)-6-methyl-1-tosyl-1,6-di hydro-7 H-pyrrolo[2,3- c]pyridin-7-one. A mixture of 2-(3-bromo-4-((1r,3r)-3-(methyl(piperidin-4- ylmethyl)amino)cyclobutoxy)phenyl)propan-2-ol (70 mg, 0.17 mmol, 1.0 equiv), 6-methyl-4- (4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1-tosyl-1,6-di hydro-7 H-pyrrolo[2,3-c]pyridin-7-one (109 mg, 255 μmol, 1.5 equiv), K 3 PO 4 (108 mg, 510 μmol, 3.0 equiv) and SPhos Pd G3 (26 mg, 34 μmol, 0.2 equiv) in THF (2 mL) and H2O (0.5 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 60 °C for 12 h under N2 atmosphere. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (column: Phenomenex C1875*30mm*3um; mobile phase: [water (FA)-ACN]; B%: 5%-35%, 7 min) to give desired 4-(5-(2-hydroxypropan-2-yl)-2-((1r,3r)-3-(methyl(piperidin-4 - ylmethyl)amino)cyclobutoxy)phenyl)-6-methyl-1-tosyl-1,6-dihy dro-7 H-pyrrolo[2,3-c]pyridin-7- one (55 mg, 86 μmol, 51% yield) as a white solid. [0501] Step 7: Preparation of 4-(5-(2-hydroxypropan-2-yl)-2-((1r,3r)-3-(methyl (piperidin- 4-ylmethyl)amino)cyclobutoxy)phenyl)-6-methyl-1,6-dihydro-7 H-pyrrolo[2,3-c]pyridin-7- one. To a solution of 4-(5-(2-hydroxypropan-2-yl)-2-((1r,3r)-3-(methyl(piperidin-4 - ylmethyl)amino)cyclobutoxy)phenyl)-6-methyl-1-tosyl-1,6-dihy dro-7 H-pyrrolo[2,3-c]pyridin-7- one (55 mg, 86 μmol, 1.0 equiv) in MeOH (1.3 mL) was added KOH (1 M, 1.30 mL, 15 equiv) t 20 °C. The mixture was stirred at 20 °C for 2 h. The reaction mixture was quenched by addition of AcOH (0.1 mL) at 0 °C, and then freeze-dried to give a residue. The residue was dissolved in NMP (0.5 mL ) 4-(5-(2-hydroxypropan-2-yl)-2-((1r,3r)-3-(methyl(piperidin-4 -ylmethyl)amino) cyclobutoxy)phenyl)-6-methyl-1,6-dihydro-7 H-pyrrolo[2,3-c]pyridin-7-one (40 mg, crude) was obtained as a yellow solution in NMP. [0502] Step 8: Preparation of N-((1r,4r)-4-(3-chloro-4-cyanophenoxy)cyclohexyl)-6-(4- ((((1r,3r)-3-(4-(2-hydroxypropan-2-yl)-2-(6-methyl-7-oxo-6,7 -dihydro-1H-pyrrolo[2,3- c]pyridin-4-yl)phenoxy)cyclobutyl)(methyl)amino)methyl)piper idin-1-yl)pyridazine-3- carboxamide. A mixture of 4-(5-(2-hydroxypropan-2-yl)-2-((1r,3r)-3-(methyl(piperidin-4 - ylmethyl)amino)cyclobutoxy)phenyl)-6-methyl-1,6-dihydro-7 H-pyrrolo[2,3-c]pyridin-7-one (40 mg, 83 μmol, 1.0 equiv), 6-chloro-N-((1r,4r)-4-(3-chloro-4-cyanophenoxy)cyclohexyl) pyridazine- 3-carboxamide (32 mg, 83 μmol, 1.0 equiv) and K2CO3 (34 mg, 250 μmol, 3.0 equiv) in NMP (1 mL) was stirred at 50 °C for 12 h. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (column: Phenomenex luna C18150*25mm* 10um; mobile phase: [water (FA)-ACN];B%: 20%-50%,10min) to give desired compound N-((1r,4r)-4-(3-chloro-4-cyanophenoxy)cyclohexyl)-6-(4-((((1 r,3r)-3-(4-(2- hydroxypropan-2-yl)-2-(6-methyl-7-oxo-6,7-dihydro-1H-pyrrolo [2,3-c]pyridin-4- yl)phenoxy)cyclobutyl)(methyl)amino)methyl)piperidin-1-yl)py ridazine-3-carboxamide (29 mg, 35 μmol, 41% yield, 98% purity) as an off-white solid. 1 H NMR (400 MHz, CD 3 OD): δ 7.88 (d, J = 9.6 Hz, 1H), 7.68 (d, J = 8.9 Hz, 1H), 7.50 (d, J = 2.4 Hz, 1H), 7.43 (dd, J = 2.4, 8.6 Hz, 1H), 7.32 (d, J = 2.9 Hz, 1H), 7.28 (d, J = 9.6 Hz, 1H), 7.22 - 7.17 (m, 2H), 7.04 (dd, J = 2.4, 8.8 Hz, 1H), 6.81 (d, J = 8.6 Hz, 1H), 6.24 (d, J = 2.8 Hz, 1H), 4.79 - 4.70 (m, 1H), 4.60 - 4.56 (m, 1H), 4.50 (d, J = 12.5 Hz, 3H), 4.06 - 3.87 (m, 1H), 3.69 (s, 3H), 3.19 - 2.97 (m, 3H), 2.44 - 2.30 (m, 2H), 2.25 (s, 2H), 2.24 - 2.14 (m, 5H), 2.13 - 2.04 (m, 2H), 1.89 (m, 3H), 1.71 - 1.59 (m, 4H), 1.54 (s, 6H), 1.32 - 1.14 (m, 2H) LC-MS: MS (ES+): RT = 2.029 min, m/z = 833.6 [M + H+]; LCMS method: 10 EXAMPLE 23 – Synthesis of N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]-6-[4-[2-[4-[[4-(1 - hydroxy-1-methyl-ethyl)-2-(6-methyl-7-oxo-1H-pyrrolo[2,3-c]p yridin-4-yl)phenyl]methyl] phenyl]ethyl]-1-piperidyl]pyridazine-3-carboxamide (I-205) [0503] Step 1: Preparation of tert-butyl 4-[2-[4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2- yl)phenyl]ethyl]piperidine-1-carboxylate. To a solution of tert-butyl 4-[2-(4-bromophenyl) ethyl]piperidine-1-carboxylate (2.00 g, 5.43 mmol, 1.0 equiv), 4,4,5,5-tetramethyl-2-(4,4,5,5- tetramethyl-1,3,2-dioxaborolan-2-yl)-1,3,2-dioxaborolane (2.80 g, 11.0 mmol, 2.0 equiv), cyclopentyl(diphenyl)phosphane;dichloropalladium;iron (400 mg, 547 μmol, 0.1 equiv) in dioxane (20 mL) was added potassium;acetate (1.10 g, 11.2 mmol, 2.1 equiv) at 25°C. The mixture was stirred at 80 °C for 12 h under N2. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate=10/1 to 1/1). Compound tert-butyl 4-[2-[4-(4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2-yl)phenyl]ethyl]piperidine-1-carboxylate (2.20 g, 5.30 mmol, 98% yield) was obtained as a white solid. [0504] Step 2: Preparation of tert-butyl 4-[2-[4-[(2-bromo-4-ethoxycarbonyl-phenyl) methyl]phenyl]ethyl]piperidine-1-carboxylate. To a solution of tert-butyl 4-[2-[4-(4,4,5,5- tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl]ethyl]piperidine- 1-carboxylate (2.20 g, 5.30 mmol, 1.0 equiv), ethyl 3-bromo-4-(bromomethyl)benzoate (2.60 g, 8.07 mmol, 1.5 equiv) in EtOH (12 mL), Water (2.4 mL), toluene (12 mL) was de-gassed and then added Na2CO3 (1 M, 9.62 mL, 1.8 equiv) and Pd(PPh3)4 (890 mg, 770 μmol, 1.5e-1 equiv) under N 2 . then heated to 80 °C for 12 h. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate=10/1 to 1/1). Compound tert-butyl 4-[2-[4-[(2-bromo-4-ethoxycarbonyl-phenyl)methyl]phenyl] ethyl]piperidine-1- carboxylate (1.20 g, 2.26 mmol, 43% yield) was obtained as a white solid. [0505] Step 3: Preparation of 2-[3-bromo-4-[[4-[2-(4-piperidyl)ethyl]phenyl]methyl] phenyl]propan-2-ol. To a solution of tert-butyl 4-[2-[4-[(2-bromo-4-ethoxycarbonyl- phenyl)methyl]phenyl]ethyl]piperidine-1-carboxylate (1.10 g, 2.07 mmol, 1.0 equiv) in DCM (22 mL) was de-gassed and then added MeMgBr (3 M, 11.00 mL, 15.9 equiv) at -78 °C under N2. Then heated to 25 °C for 12 h. The reaction mixture was quenched by addition water 50 mL at 0 °C, and then diluted with DCM 25 mL and extracted with DCM (50 mL x 3). The combined organic layers were washed with brine 50 mL, dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (FA condition; column: Phenomenex luna C18150*40mm* 15um;mobile phase: [water(FA)-ACN];B%: 16%- 46%,10min). Compound 2-[3-bromo-4-[[4-[2-(4-piperidyl)ethyl]phenyl]methyl]phenyl] propan-2- ol (290 mg, 696 μmol, 34% yield) was obtained as a white solid. [0506] Step 4: Preparation of 4-[5-(1-hydroxy-1-methyl-ethyl)-2-[[4-[2-(4-piperidyl) ethyl]phenyl]methyl]phenyl]-6-methyl-1-(p-tolylsulfonyl)pyrr olo[2,3-c]pyridin-7-one.2-[3- bromo-4-[[4-[2-(4-piperidyl)ethyl]phenyl]methyl]phenyl]propa n-2-ol (190 mg, 456 μmol, 1.0 equiv), 6-methyl-1-(p-tolylsulfonyl)-4-(4,4,5,5-tetramethyl-1,3,2-di oxaborolan-2-yl)pyrrolo[2,3- c]pyridin-7-one (285 mg, 665 μmol, 1.5 equiv), K3PO4 (285 mg, 1.34 mmol, 2.9 equiv) and SPhos Pd G3 (76.0 mg, 97.4 μmol, 0.2 equiv) in THF (10 mL) and Water (2 mL) was de-gassed and then heated to 60 °C for 12 h under N2. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (FA condition; column: YMC Triart C18150*25mm*5um;mobile phase: [water(FA)-ACN];B%: 26%-56%,8.5min). Compound 4-[5- (1-hydroxy-1-methyl-ethyl)-2-[[4-[2-(4-piperidyl)ethyl] phenyl]methyl]phenyl]-6-methyl-1-(p- tolylsulfonyl)pyrrolo[2,3-c]pyridin-7-one (125 mg, 196 μmol, 43% yield) was obtained as a white solid. [0507] Step 5: Preparation of 4-[5-(1-hydroxy-1-methyl-ethyl)-2-[[4-[2-(4-piperidyl) ethyl]phenyl]methyl]phenyl]-6-methyl-1H-pyrrolo[2,3-c]pyridi n-7-one. To a solution of 4-[5- (1-hydroxy-1-methyl-ethyl)-2-[[4-[2-(4-piperidyl)ethyl]pheny l]methyl]phenyl]-6-methyl-1-(p- tolylsulfonyl)pyrrolo[2,3-c]pyridin-7-one (60 mg, 94 μmol, 1.0 equiv) in MeOH (1.5 mL) was added NaOH (1 M, 1.41 mL, 15.0 equiv) and The mixture was stirred at 25 °C for 1 h. The reaction mixture was adjust PH=7.0 with 2M HOAc,then concentrated under reduced pressure to give a residue. The crude product was used into the next step without further purification. Compound 4-[5-(1-hydroxy-1-methyl-ethyl)-2-[[4-[2-(4-piperidyl)ethyl] phenyl]methyl]phenyl]-6- methyl-1H-pyrrolo[2,3-c]pyridin-7-one (45 mg, 93 μmol, 99% yield) was obtained as a white solid. Step 6: Preparation of N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]-6-[4-[2-[4-[[4-(1 -hydroxy- 1-methyl-ethyl)-2-(6-methyl-7-oxo-1H-pyrrolo[2,3-c]pyridin-4 -yl)phenyl] methyl]phenyl]ethyl]-1-piperidyl]pyridazine-3-carboxamide. To a solution of 4-[5-(1-hydroxy- 1-methyl-ethyl)-2-[[4-[2-(4-piperidyl)ethyl]phenyl]methyl]ph enyl]-6-methyl-1H-pyrrolo[2,3- c]pyridin-7-one (45 mg, 93 μmol, 1.0 equiv) and 6-chloro-N-[4-(3-chloro-4-cyano- phenoxy)cyclohexyl]pyridazine-3-carboxamide (37 mg, 94 μmol, 1.0 equiv) in NMP (0.5 mL) was de-gassed and then added K 2 CO 3 (39 mg, 279 μmol, 3.0 equiv) and heated to 50 °C for 12 h. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (FA condition; column: Unisil 3-100 C18 Ultra 150*50mm*3 μm;mobile phase: [water(FA)-ACN];B%: 62%-92%,7min). Compound N-[4-(3-chloro-4-cyano-phenoxy) cyclohexyl]-6-[4-[2-[4-[[4-(1-hydroxy-1-methyl-ethyl)-2-(6-m ethyl-7-oxo-1H-pyrrolo[2,3- c]pyridin-4-yl)phenyl]methyl]phenyl]ethyl]-1-piperidyl]pyrid azine-3-carboxamide (5.92 mg, 6.99 μmol, 8% yield, 99% purity) was obtained as a white solid. 1 H NMR: (400 MHz, MeOH-d4) δ = 7.88 (d, J = 9.6 Hz, 1H), 7.69 (d, J = 8.8 Hz, 1H), 7.49 - 7.45 (m, 1H), 7.40 (d, J = 2.0 Hz, 1H), 7.31 (d, J = 2.8 Hz, 1H), 7.28 (d, J = 1.6 Hz, 1H), 7.26 (d, J = 3.6 Hz, 1H), 7.20 (d, J = 2.4 Hz, 1H), 7.07 - 7.02 (m, 1H), 6.96 (d, J = 8.0 Hz, 2H), 6.73 (d, J = 8.0 Hz, 2H), 6.68 (s, 1H), 6.05 (d, J = 2.8 Hz, 1H), 4.52 - 4.45 (m, 3H), 4.03 - 3.92 (m, 1H), 3.84 (s, 2H), 3.53 (s, 3H), 3.50 - 3.47 (m, 1H), 3.06 - 2.97 (m, 2H), 2.66 (s, 2H), 2.59 (br t, J = 7.2 Hz, 2H), 2.26 - 2.16 (m, 2H), 2.14 - 2.07 (m, 2H), 1.92 - 1.84 (m, 2H), 1.64 (br t, J = 9.6 Hz, 4H), 1.55 (s, 6H), 1.31 - 1.23 (m, 2H) LC-MS: MS (ES + ): RT = 2.591 min, m/z = 838.4 [M + H + ], LC-MS Method 25. EXAMPLE 24 – Synthesis of N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]-6-[4-[3-[4-(2,4- difluorophenoxy)-3-(6-methyl-7-oxo-1H-pyrrolo[2,3-c]pyridin- 4-yl)anilino]-3-oxo-propyl]-1- piperidyl]pyridazine-3-carboxamide (I-212)

[0508] Step 1: Preparation of 4-[2-(2,4-difluorophenoxy)-5-nitro-phenyl]-6-methyl-1H- pyrrolo[2,3-c]pyridin-7-one. To a solution of 4-(2-fluoro-5-nitro-phenyl)-6-methyl-1-(p- tolylsulfonyl)pyrrolo[2,3-c]pyridin-7-one (2.50 g, 5.66 mmol, 1.0 equiv) in DMSO (30 mL) was added Cs2CO3 (5.54 g, 17.0 mmol, 3.0 equiv) and then 2,4-difluorophenol (737 mg, 5.66 mmol, 1.0 equiv). The mixture was stirred at 80 °C for 12 h. The reaction mixture was poured into ice water 150 mL, and then extracted with EA (100 mL x 3). The combined organic layers were washed with brine (50 mL x 3), dried over Na 2 SO 4 , filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (ISCO®; 40 g SepaFlash® Silica Flash Column, Eluent of 0~50% Ethyl acetate/Petroleum ethergradient @ 50 mL/min). The desired compound 4-[2-(2,4-difluorophenoxy)-5-nitro-phenyl]-6-methyl-1H- pyrrolo[2,3-c]pyridin-7-one (1.50 g, 3.68 mmol, 65% yield) was obtained as yellow solid. [0509] Step 2: Preparation of 4-[5-amino-2-(2,4-difluorophenoxy)phenyl]-6-methyl-1H- pyrrolo[2,3-c]pyridin-7-one. To a solution of 4-[2-(2,4-difluorophenoxy)-5-nitro-phenyl]-6- methyl-1H-pyrrolo[2,3-c]pyridin-7-one (1.40 g, 3.52 mmol, 1.0 equiv) and NH 4 Cl (565 mg, 10.6 mmol, 3.0 equiv) in H 2 O (7 mL) and EtOH (14 mL) was added Zn (1.33 g, 20.3 mmol, 5.8 equiv). The mixture was stirred at 50 °C for 3 h. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate=1/1 to 1/5). The desired compound 4-[5-amino-2-(2,4- difluorophenoxy)phenyl]-6-methyl-1H-pyrrolo[2,3-c]pyridin-7- one (1.10 g, 2.79 mmol, 79% yield) was obtained as yellow solid. [0510] Step 3: Preparation of tert-butyl 4-[3-[4-(2,4-difluorophenoxy)-3-(6-methyl-7-oxo- 1H-pyrrolo[2,3-c]pyridin-4-yl)anilino]-3-oxo-propyl]piperidi ne-1-carboxylate. To a solution of 3-(1-tert-butoxycarbonyl-4-piperidyl)propanoic acid (126 mg, 490 μmol, 1.2 equiv) in DMF (2 mL) was added DIEA (158 mg, 1.22 mmol, 3.0 equiv) and HATU (186 mg, 490 μmol, 1.2 equiv) the mixture was stirred for 30 min, and then 4-[5-amino-2-(2,4-difluorophenoxy)phenyl]-6-methyl- 1H-pyrrolo[2,3-c]pyridin-7-one (150 mg, 408 μmol, 1.0 equiv) was added. The mixture was stirred at 25 °C for 12 hr. The reaction mixture was pour into NaCl aq. water 10 mL, and then extracted with EA (10 mL x 3). The combined organic layers were washed with brine (10 mL x 3), dried over Na 2 SO 4 , filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-TLC (SiO 2 , PE: EA = 0:1). The desired compound tert-butyl 4-[3-[4-(2,4- difluorophenoxy)-3-(6-methyl-7-oxo-1H-pyrrolo[2,3-c]pyridin- 4-yl)anilino]-3-oxo- propyl]piperidine-1-carboxylate (240 mg, 394 μmol, 97% yield) was obtained as white solid. [0511] Step 4: Preparation of N-[4-(2,4-difluorophenoxy)-3-(6-methyl-7-oxo-1H- pyrrolo[2,3-c]pyridin-4-yl)phenyl]-3-(4-piperidyl)propanamid e. To a solution of tert-butyl 4- [3-[4-(2,4-difluorophenoxy)-3-(6-methyl-7-oxo-1H-pyrrolo[2,3 -c]pyridin-4-yl)anilino]-3-oxo- propyl]piperidine-1-carboxylate (160 mg, 264 μmol, 1.0 equiv) in DCM (1 mL) was TFA (211 mg, 1.85 mmol, 7.0 equiv). The mixture was stirred at 25 °C for 1 h. The reaction mixture was concentrated under reduced pressure to give a residue. The reaction mixture was used into the next step without further purification. The desired compound N-[4-(2,4-difluorophenoxy)-3-(6-methyl- 7-oxo-1H-pyrrolo[2,3-c]pyridin-4-yl)phenyl]-3-(4-piperidyl)p ropanamide (130 mg, crude) was obtained as brown oil. [0512] Step 5: Preparation of N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]-6-[4-[3-[4-(2,4- difluorophenoxy)-3-(6-methyl-7-oxo-1H-pyrrolo[2,3-c]pyridin- 4-yl)anilino]-3-oxo-propyl]-1- piperidyl]pyridazine-3-carboxamide. To a solution of N-[4-(2,4-difluorophenoxy)-3-(6-methyl- 7-oxo-1H-pyrrolo[2,3-c]pyridin-4-yl)phenyl]-3-(4-piperidyl)p ropanamide (129 mg, 256 μmol, 1.0 equiv) and 6-chloro-N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]pyridazin e-3-carboxamide (100 mg, 256 μmol, 1.0 equiv) in NMP (2 mL) was added K2CO3 (353 mg, 2.56 mmol, 10.0 equiv) The reaction mixture was stirred at 50 °C for 12 h. The reaction mixture was filtered. The residue was purified by prep-HPLC (FA condition; column: Phenomenex Luna C18150*25mm*10um;mobile phase: [water(FA)-ACN];B%: 54%-84%,10min). The desired compound N-[4-(3-chloro-4-cyano- phenoxy)cyclohexyl]-6-[4-[3-[4-(2,4-difluorophenoxy)-3-(6-me thyl-7-oxo-1H-pyrrolo[2,3- c]pyridin-4-yl)anilino]-3-oxo-propyl]-1-piperidyl]pyridazine -3-carboxamide (115 mg, 129 μmol, 50% yield) was obtained as yellow solid. 1 H NMR (400 MHz, MeOH-d4) δ = 7.94 (d, J = 9.6 Hz, 1H), 7.81 - 7.78 (m, 1H), 7.72 - 7.66 (m, 1H), 7.56 - 7.50 (m, 1H), 7.44 - 7.38 (m, 1H), 7.33 - 7.30 (m, 1H), 7.29 - 7.27 (m, 1H), 7.21 - 7.19 (m, 1H), 7.06 - 7.02 (m, 1H), 7.01 - 6.94 (m, 2H), 6.91 - 6.84 (m, 1H), 6.79 - 6.73 (m, 1H), 6.41 (d, J = 2.8 Hz, 1H), 4.55 - 4.46 (m, 3H), 4.03 - 3.92 (m, 1H), 3.65 (s, 3H), 3.16 - 3.05 (m, 2H), 2.50 - 2.43 (m, 2H), 2.25 - 2.17 (m, 2H), 2.14 - 2.06 (m, 2H), 1.98 - 1.91 (m, 2H), 1.75 - 1.69 (m, 3H), 1.64 (t, J = 9.6 Hz, 4H), 1.35 - 1.28 (m, 2H) LC-MS: MS (ES + ): RT = 2.745 min, m/z = 861.2 [M + H + ], m/z = 431.2 [1/2M + H + ]; LCMS Method 25. EXAMPLE 25 – Synthesis of N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]-6-[4-[2-[5-[4- ethylsulfonyl-2-(6-methyl-7-oxo-1H-pyrrolo[2,3-c]pyridin-4-y l)phenoxy]-2-pyridyl]ethyl]-1- piperidyl]pyridazine-3-carboxamide (I-218)

[0513] Step 1: Preparation of tert-butyl 4-[2-(5-hydroxy-2-pyridyl)ethynyl]piperidine-1- carboxylate. To a solution of tert-butyl 2-[(9S)-7-(4-chlorophenyl)-4,5,13-trimethyl-3-thia- 1,8,11,12-tetrazatricyclo[8.3.0.02,6]trideca-2(6),4,7,10,12- pentaen-9-yl]acetate (250 mg, 547 μmol, 1.0 equiv) in dioxane (3 mL) was added Cs2CO3 (535 mg, 2 mmol, 3.0 equiv) and SPhos Pd G3 (43 mg, 55 μmol, 0.1 equiv).The mixture was stirred at 90 °C for 12 h. The reaction mixture was concentrated to afford crude product. The residue was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate=10/1 to 1/1). Compound tert-butyl 4-[2-(5-hydroxy-2- pyridyl)ethynyl]piperidine-1-carboxylate (4.0 g, 12 mmol, 64% yield) was obtained as a yellow solid. [0514] Step 2: Preparation of tert-butyl 4-[2-(5-hydroxy-2-pyridyl)ethyl]piperidine-1- carboxylate. To a solution of tert-butyl 4-[2-(5-hydroxy-2-pyridyl)ethynyl]piperidine-1- carboxylate (4 g, 124 mmol, 1.0 equiv) in TFE (60 mL) was added Pd/C (1.0 g, 10% purity). The suspension was degassed and purged with H2 for 3 times. The mixture was stirred under H2 (15 psi) at 25 °C for 12 h. The reaction mixture was filtered and the filtrate was concentrated to afford crude product. Compound tert-butyl 4-[2-(5-hydroxy-2-pyridyl)ethyl]piperidine-1-carboxylate (4 g, 12 mmol, 96 % yield) was obtained as a yellow oil . [0515] Step 3: Preparation of tert-butyl 4-[2-[5-(2-bromo-4-ethylsulfonyl-phenoxy)-2- pyridyl]ethyl]piperidine-1-carboxylate. To a solution of tert-butyl 4-[2-(5-hydroxy-2- pyridyl)ethyl]piperidine-1-carboxylate (1 g, 3 mmol, 1.0 equiv), 2-bromo-4-ethylsulfonyl-1-fluoro- benzene (871 mg, 3 mmol, 1.0 equiv) in DMSO (10 mL) was added Cs 2 CO 3 (3 g, 10 mmol, 3.0 equiv). The mixture was stirred at 70 °C for 2 h. The residue was diluted with 20 mL water and extracted with EA (20 mL x 2). The combined organic layers were dried over Na 2 SO 4 , filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate=10/1 to 2/1). Compound tert-butyl 4-[2-[5- (2-bromo-4-ethylsulfonyl-phenoxy)-2-pyridyl]ethyl]piperidine -1-carboxylate (1.0 g, 2.0 mmol, 61%yield) was obtained as a colorless oil. [0516] Step 4: Preparation of tert-butyl 4-[2-[5-[4-ethylsulfonyl-2-[6-methyl-7-oxo-1-(p- tolylsulfonyl)pyrrolo[2,3-c]pyridin-4-yl]phenoxy]-2-pyridyl] ethyl]piperidine-1-carboxylate. To a solution of tert-butyl 4-[2-[5-(2-bromo-4-ethylsulfonyl-phenoxy)-2-pyridyl]ethyl] piperidine- 1-carboxylate (200 mg, 361 μmol, 1.0 equiv),6-methyl-1-(p-tolylsulfonyl)-4-(4,4,5,5-tetramethyl- 1,3,2-dioxaborolan-2-yl)pyrrolo[2,3-c]pyridin-7-one (201 mg, 470 μmol, 1.0 equiv) in THF (2 mL) and H2O (0.4 mL) was added SPhos Pd G3 (28 mg, 36 μmol, 0.1 equiv) and K3PO4 (230 g, 1 mmol, 3.0 equiv). The mixture was stirred at 60 °C for 3 h. The residue was diluted with 20 mL water and extracted with EA (20 mL x 2). The combined organic layers were dried over Na 2 SO 4 , filtered and concentrated under reduced pressure to give a residue. Compound tert-butyl 4-[2-[5- [4-ethylsulfonyl-2-[6-methyl-7-oxo-1-(p-tolylsulfonyl) pyrrolo[2,3-c]pyridin-4-yl]phenoxy]-2- pyridyl]ethyl]piperidine-1-carboxylate (280 mg, crude) was obtained as a yellow oil. [0517] Step 5: Preparation of tert-butyl 4-[2-[5-[4-ethylsulfonyl-2-(6-methyl-7-oxo-1H- pyrrolo[2,3-c]pyridin-4-yl)phenoxy]-2-pyridyl]ethyl]piperidi ne-1-carboxylate. To a solution of tert-butyl 4-[2-[5-[4-ethylsulfonyl-2-[6-methyl-7-oxo-1-(p-tolylsulfony l)pyrrolo[2,3-c]pyridin -4- yl]phenoxy]-2-pyridyl]ethyl]piperidine-1-carboxylate (280 mg, 361 μmol, 1.0 equiv) in MeOH (2 mL) was added KOH (1 M, 2 mL, 5.5 equiv). The mixture was stirred at 20 °C for 3 h. The reaction mixture was acidified by HOAc to pH = 5~6. The residue was purified by prep- HPLC(column: Phenomenex luna C18150*25mm* 10um;mobile phase: [water(FA)-ACN];B%: 38%-68%,10min).. Compound tert-butyl 4-[2-[5-[4-ethylsulfonyl-2-(6-methyl-7-oxo-1H- pyrrolo[2,3-c]pyridin-4-yl)phenoxy]-2-pyridyl]ethyl]piperidi ne-1-carboxylate (160 mg, 258 μmol, 71% ield) was obtained as a yellow oil. [0518] Step 6: Preparation of 4-[5-ethylsulfonyl-2-[[6-[2-(4-piperidyl)ethyl]-3-pyridyl] oxy]phenyl]-6-methyl-1H-pyrrolo[2,3-c]pyridin-7-one. To a solution of tert-butyl 4-[2-[5-[4- ethylsulfonyl-2-(6-methyl-7-oxo-1H-pyrrolo[2,3-c]pyridin-4-y l)phenoxy]-2-pyridyl]ethyl] piperidine-1-carboxylate (100 mg, 161 μmol, 1.0 equiv) in DCM (1 mL) was added TFA (0.5 mL). The mixture was stirred at 20 °C for 2 h. The reaction was concentrated to afford crude product. Compound 4-[5-ethylsulfonyl-2-[[6-[2-(4-piperidyl)ethyl]-3-pyridyl]ox y]phenyl]-6-methyl-1H- pyrrolo[2,3-c]pyridin-7-one (100 mg, 158 μmol, 99% yield, TFA salt) was obtained as a yellow oil. [0519] Step 7: Preparation of N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]-6-[4-[2-[5-[4- ethylsulfonyl-2-(6-methyl-7-oxo-1H-pyrrolo[2,3-c]pyridin-4-y l)phenoxy]-2-pyridyl]ethyl]-1- piperidyl]pyridazine-3-carboxamide. To a solution of 4-[5-ethylsulfonyl-2-[[6-[2-(4- piperidyl)ethyl]-3-pyridyl]oxy]phenyl]-6-methyl-1H-pyrrolo[2 ,3-c]pyridin-7-one (100 mg, 192 μmol, 1.0 equiv), 6-chloro-N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]pyridazin e-3-carboxamide (75 mg, 192 μmol, 1.0 equiv) in NMP (1 mL) was added K 2 CO 3 (80 mg, 576 μmol, 3.0 equiv). The mixture was stirred at 50 °C for 12 h. The reaction mixture was concentrated to afford crude product. The residue was purified by prep-HPLC(column: Phenomenex luna C18150*25mm* 10um;mobile phase: [water(FA)-ACN];B%: 50%-80%,10min). Compound N-[4-(3-chloro-4- cyano-phenoxy)cyclohexyl]-6-[4-[2-[5-[4-ethylsulfonyl-2-(6-m ethyl-7-oxo-1H-pyrrolo[2,3- c]pyridin-4-yl)phenoxy]-2-pyridyl]ethyl]-1-piperidyl]pyridaz ine-3-carboxamide (44 mg, 50 μmol, 26%yield, 98%purity) was obtained as a yellow solid. 1 H NMR: (CD3OD, 400 MHz) 8.08 (m, 2 H) 7.91 - 8.00 (m, 2 H) 7.66 - 7.72 (m, 1 H) 7.27 - 7.45 (m, 6 H) 7.20 (m,1 H) 7.05 (m, 1 H) 6.33 (m, H) 4.42 - 4.55 (m, 3 H) 3.91 - 4.04 (m, 1 H) 3.61 - 3.68 (m, 3 H) 3.48 (s, 1 H) 3.02 - 3.17 (m, 3 H) 2.81 (m, 2 H) 2.07 - 2.25 (m, 4 H) 1.86 - 1.95 (m, 2 H) 1.60 - 1.68 (m, 6 H) 1.26 - 1.33 (m, 5 H) 1.24 - 1.35 (m, 1 H) LC-MS: MS (ES+): RT =2.266, m/z =875.2[M+H] + . LCMS Method 25. EXAMPLE 26 – Synthesis of N-[4-(3-chloro-4-cyano-phenoxy) cyclohexyl]-6-[4-[2-[4-[2-(6- methyl-7-oxo-1H- pyrrolo [2, 3-c] pyridin-4-yl)-4-methylsulfonyl-phenoxy] phenyl] ethyl]-1- piperidyl] pyridazine-3-carboxamide (I-231) [0520] Step 1: Preparation of tert-butyl 4-[2-(4-benzyloxyphenyl)ethynyl]piperidine-1- carboxylate. A mixture of tert-butyl 4-ethynylpiperidine-1-carboxylate (337 mg, 1.61 mmol, 1.0 eqiuv), 1-benzyloxy-4-iodo-benzene (500 mg, 1.61 mmol, 1.0 equiv), Pd(PPh3)2Cl2 (56 mg, 80 μmol, 0.05 equiv), CuI (30 mg, 161 μmol, 0.1 equiv) and TEA (489 mg, 4.84 mmol, 3.0 equiv) in DMF (5 mL) was degassed and purged with N2 for 3 times and the mixture was stirred at 80 °C for 12 h under N 2 atmosphere. The mixture was partitioned between H 2 O 20 mL and EA 100 mL. The organic phase was separated, washed with H2O 30 mL (30 mL * 3), dried over [Na2SO4], filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO 2 , Petroleum ether/Ethyl acetate=20/1 to 15/1) to give compound tert-butyl 4- [2-(4-benzyloxyphenyl)ethynyl]piperidine-1-carboxylate (360 mg, 57% yield). [0521] Step 2: Preparation of tert-butyl 4-[2-(4-hydroxyphenyl)ethyl]piperidine-1- carboxylate. To a solution of tert-butyl 4-[2-(4-benzyloxyphenyl)ethynyl]piperidine-1-carboxylate (360 mg, 919umol, 1.0 equiv) in MeOH (5 mL) was addedPd/C (150 mg, 10% purity) under N2 atmosphere. The mixture was stirred under H 2 (15 Psi) at 25 °C for 2 h. The mixture filtered and concentrated. The residue was used for next step directly to give tert-butyl 4-[2-(4- hydroxyphenyl)ethyl]piperidine-1-carboxylate (270 mg, 96% yield). [0522] Step 3: Preparation of tert-butyl 4-[2-[4-(2-bromo-4-methylsulfonyl-phenoxy) phenyl]ethyl]piperidine-1-carboxylate. To a solution of tert-butyl 4-[2-(4-hydroxyphenyl) ethyl]piperidine-1-carboxylate (270 mg, 884 μmol, 1.0 equiv) and 2-bromo-1-fluoro-4- methylsulfonyl-benzene (223 mg, 884 μmol, 1.0 equiv) in NMP (5 mL) was added Cs2CO3 (720 mg, 2.21 mmol, 2.5 equiv). The mixture was stirred at 70 °C for 2 h. The mixture was partitioned between H 2 O 20 mL and EA 100 mL. The organic phase was separated, washed with H 2 O 30 mL (30 mL * 3), dried over [Na 2 SO 4 ], filtered and concentrated. The residue was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate=15/1 to 10/1) to give tert-butyl 4-[2-[4-(2- bromo-4-methylsulfonyl-phenoxy)phenyl]ethyl]piperidine-1-car boxylate (400 mg, 84% yield). [0523] Step 4: Preparation of tert-butyl 4-[2-[4-[2-[6-methyl-7-oxo-1-(p-tolylsulfonyl) pyrrolo[2,3-c]pyridin-4-yl]-4-methylsulfonyl-phenoxy]phenyl] ethyl]piperidine-1-carboxylate. A mixture of tert-butyl 4-[2-[4-(2-bromo-4-methylsulfonyl-phenoxy) phenyl]ethyl]piperidine-1- carboxylate (400 mg, 742 μmol, 1.0 equiv), 6-methyl-1-(p-tolylsulfonyl)-4-(4,4,5,5-tetramethyl- 1,3,2-dioxaborolan-2-yl)pyrrolo[2,3-c]pyridin-7-one (318 mg, 742 μmol, 1.0 equiv), dicyclohexyl- [2-(2,6-dimethoxyphenyl)phenyl] phosphane ;methanesulfonate;(2-phenylanilino)palladium(1+) (57 mg, 74 μmol, 0.1 equiv), K 3 PO 4 (473 mg, 2.23 mmol, 3.0 equiv) in THF (10 mL) and H 2 O (2 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 60 °C for 12 h under N2 atmosphere. The mixture was partitioned between H2O 20 mL and EA 100 mL. The organic phase was separated, washed with H 2 O (30 mL * 3), dried over [Na 2 SO 4 ], filtered and concentrated. The residue was purified by column chromatography (SiO 2 , Petroleum ether/Ethyl acetate=1/1 to 0/1) to give compound tert-butyl 4-[2-[4-[2-[6-methyl-7-oxo-1-(p- tolylsulfonyl)pyrrolo[2,3-c]pyridin-4-yl]-4-methylsulfonyl-p henoxy]phenyl]ethyl]piperidine-1- carboxylate (400 mg, 70% yield). [0524] Step 5: Preparation of tert-butyl 4-[2-[4-[2-(6-methyl-7-oxo-1H-pyrrolo[2,3- c]pyridin-4-yl)-4-methylsulfonyl-phenoxy]phenyl]ethyl]piperi dine-1-carboxylate. To a solution of tert-butyl 4-[2-[4-[2-[6-methyl-7-oxo-1-(p-tolylsulfonyl)pyrrolo[2,3-c] pyridin-4-yl]-4- methylsulfonyl-phenoxy]phenyl]ethyl]piperidine-1-carboxylate (200 mg, 263 μmol, 1.0 equiv) in MeOH (2 mL) was added KOH (73 mg, 1.32 mmol, 5.0 equiv). The mixture was stirred at 25 °C for 1 h. The mixture was partitioned between H2O 20 mL and EA 100 mL. The organic phase was separated, washed with H 2 O (30 mL * 3), dried over [Na 2 SO 4 ], filtered and concentrated. The residue was purified by column chromatography (SiO 2 , Petroleum ether/Ethyl acetate=1/1 to 0/1) to give tert-butyl 4-[2-[4-[2-(6-methyl-7-oxo-1H-pyrrolo[2,3-c]pyridin-4-yl)-4- methylsulfonyl- phenoxy]phenyl]ethyl]piperidine-1-carboxylate (120 mg, 75% yield). [0525] Step 6: Preparation of 6-methyl-4-[5-methylsulfonyl-2-[4-[2-(4-piperidyl)ethyl] phenoxy]phenyl]-1H-pyrrolo[2,3-c]pyridin-7-one. To a solution of tert-butyl 4-[2-[4-[2-(6- methyl-7-oxo-1H-pyrrolo[2,3-c]pyridin-4-yl)-4-methylsulfonyl -phenoxy]phenyl]ethyl] piperidine- 1-carboxylate (120 mg, 198 μmol, 1.0 equiv) in DCM (4 mL) was added TFA (1.5 g, 13.5 mmol, 1 mL, 68 equiv). The mixture was stirred at 25 °C for 0.5 h. The mixture filtered and concentrated. The residue was used for next step directly to give compound 6-methyl-4-[5-methylsulfonyl-2-[4- [2-(4-piperidyl)ethyl]phenoxy]phenyl]-1H-pyrrolo[2,3-c]pyrid in-7-one (100 mg, 99% yield). [0526] Step 7: Preparation of N-[4-(3-chloro-4-cyano-phenoxy) cyclohexyl]-6-[4-[2-[4-[2- (6-methyl-7-oxo-1H- pyrrolo [2, 3-c] pyridin-4-yl)-4-methylsulfonyl-phenoxy] phenyl] ethyl]- 1-piperidyl] pyridazine-3-carboxamide. To a solution of 6-methyl-4-[5-methylsulfonyl-2-[4-[2- (4-piperidyl)ethyl]phenoxy]phenyl]-1H-pyrrolo[2,3-c]pyridin- 7-one (100 mg, 197 μmol, 1.0 equiv) and 6-chloro-N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]pyridazin e-3-carboxamide (77 mg, 197 μmol, 1.0 equiv) in NMP (2 mL) was added K2CO3 (82 mg, 593 μmol, 3.0 equiv). The mixture was stirred at 50 °C for 12 h. The mixture was filtered and concentrated. The residue was purified by prep-HPLC (column: Waters Xbridge C18150*50mm* 10um; mobile phase: [water (NH4HCO3)-ACN]; B%: 52%-82%, 10min). Compound N-[4-(3-chloro-4-cyano-phenoxy) cyclohexyl]-6-[4-[2-[4-[2-(6-methyl-7-oxo-1H- pyrrolo [2, 3-c] pyridin-4-yl)-4-methylsulfonyl- phenoxy] phenyl] ethyl]-1-piperidyl] pyridazine-3-carboxamide (70 mg, 40% yield). 1 H NMR (400 MHz, DMSO): δ 12.17 - 12.00 (m, 1H), 8.61 (d, J = 7.8 Hz, 1H), 7.97 (d, J = 2.3 Hz, 1H), 7.90 - 7.83 (m, 2H), 7.81 (d, J = 9.5 Hz, 1H), 7.43 (s, 1H), 7.40 (d, J = 2.1 Hz, 1H), 7.35 (d, J = 9.8 Hz, 1H), 7.31 - 7.23 (m, 3H), 7.15 - 7.12 (m, 1H), 7.05 (d, J = 8.3 Hz, 2H), 6.99 (d, J = 8.8 Hz, 1H), 6.29 (d, J = 2.1 Hz, 1H), 4.58 - 4.45 (m, 3H), 3.91 - 3.81 (m, 1H), 3.58 (s, 3H), 3.25 (s, 3H), 2.97 (t, J = 12.3 Hz, 2H), 2.14 - 2.07 (m, 2H), 1.97 - 1.72 (m, 5H), 1.69 - 1.46 (m, 8H), 1.26 - 1.13 (m, 2H). LC-MS: MS (ES+): RT = 2.694 min, m/z = 860.5 [M + H+]; LCMS method: 10 EXAMPLE 27 – Synthesis of N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]-6-[4-[[4-[4- ethylsulfonyl-2-(1-methyl-6-oxo-3-pyridyl)phenoxy]phenoxy]me thyl]-1-piperidyl] pyridazine- 3-carboxamide (I-234)

[0527] Step 1: Preparation of tert-butyl 4-[[4-(2-bromo-4-ethylsulfonyl-phenoxy) phenoxy]methyl]piperidine-1-carboxylate. To a solution of tert-butyl 4-[(4-hydroxyphenoxy) methyl]piperidine-1-carboxylate (1.0 g, 3.3 mmol, 1.0 equiv) and 2-bromo-4-ethylsulfonyl-1- fluoro-benzene (1.04 g, 3.90 mmol, 1.2 equiv) in DMSO (10 mL) was added Cs 2 CO 3 (2.65 g, 8.13 mmol, 2.5 equiv). The mixture was stirred at 50 °C for 12 h. The residue was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate=50/1 to 3/1) to give tert-butyl 4-[[4-(2- bromo-4-ethylsulfonyl-phenoxy)phenoxy]methyl]piperidine-1-ca rboxylate (1.7 g, 94% yield) as a colorless oil. [0528] Step 2: Preparation of tert-butyl 4-[[4-[4-ethylsulfonyl-2-(1-methyl-6-oxo-3- pyridyl)phenoxy]phenoxy]methyl]piperidine-1-carboxylate. To a solution of tert-butyl 4-[[4-(2- bromo-4-ethylsulfonyl-phenoxy)phenoxy]methyl]piperidine-1-ca rboxylate (300 mg, 541 μmol, 1.0 equiv) and 1-methyl-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyri din-2-one (152 mg, 649 μmol, 1.2 equiv) in THF (5 mL) and H2O (1 mL) was added K3PO4 (344 mg, 1.62 mmol, 3.0 equiv) and SPhos Pd G3 (126 mg, 162 μmol, 0.3 equiv). The mixture was stirred at 60 °C for 12 h. The residue was purified by column chromatography (SiO 2 , Petroleum ether/Ethyl acetate=10/1 to 1/1) to give tert-butyl 4-[[4-[4-ethylsulfonyl-2-(1-methyl-6-oxo-3-pyridyl) phenoxy]phenoxy]methyl]piperidine-1-carboxylate (300 mg, 95% yield) as a colorless oil. [0529] Step 3: Preparation of of 5-[5-ethylsulfonyl-2-[4-(4-piperidylmethoxy) phenoxy] phenyl]-1-methyl-pyridin-2-one. To a solution of tert-butyl 4-[[4-[4-ethylsulfonyl-2-(1-methyl-6- oxo-3-pyridyl)phenoxy]phenoxy]methyl]piperidine-1-carboxylat e (100 mg, 171 μmol, 1.0 eq) in DCM (3 mL) was added TFA (1.5 mL). The mixture was stirred at 25 °C for 0.5 h. The reaction mixture was concentrated to afford crude product. [0530] Step 4: Preparation of N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]-6-[4-[[4-[4- ethylsulfonyl-2-(1-methyl-6-oxo-3-pyridyl)phenoxy]phenoxy]me thyl]-1-piperidyl] pyridazine- 3-carboxamide. To a solution of 5-[5-ethylsulfonyl-2-[4-(4-piperidylmethoxy) phenoxy]phenyl]- 1-methyl-pyridin-2-one (100 mg, 167 μmol, 1.0 equiv, TFA salt) and 6-chloro-N-[4-(3-chloro-4- cyano-phenoxy)cyclohexyl]pyridazine-3-carboxamide (65 mg, 0.17 mmol, 1.0 equiv) in NMP (2 mL) was added K 2 CO 3 (69 mg, 0.50 mmol, 3.0 equiv). The mixture was stirred at 50 °C for 1 h. The residue was purified by prep-HPLC (column: Unisil 3-100 C18 Ultra 150*50mm*3um; mobile phase: [water(FA)-ACN]; B%: 56%-86%,7min) to give N-[4-(3-chloro-4-cyano- phenoxy)cyclohexyl]-6-[4-[[4-[4-ethylsulfonyl-2-(1-methyl-6- oxo-3-pyridyl) phenoxy]phenoxy]methyl]-1-piperidyl]pyridazine-3-carboxamide (72 mg, 49% yield, 96.55% purity) as a white solid. 1 H NMR (400 MHz, DMSO-d6) δ 8.60 (d, J=8.24 Hz, 1 H), 8.06 (d, J=2.56 Hz, 1 H), 7.73 - 7.88 (m, 5 H), 7.35 - 7.40 (m, 2 H), 7.11 - 7.15 (m, 3 H), 7.02 (d, J=9.12 Hz, 2 H), 6.87 (d, J=8.64 Hz, 1 H), 6.46 (d, J=9.36 Hz, 1 H), 4.55 (d, J=11.26 Hz, 3 H), 3.88 (d, J=6.00 Hz, 3 H), 3.51 (s, 3 H), 3.06 (t, J=11.88 Hz, 2 H), 2.04 - 2.21 (m, 4 H), 1.90 (d, J=12.26 Hz, 4 H), 1.46 - 1.69 (m, 5 H), 1.28 - 1.38 (m, 2 H), 1.13 (t, J=7.38 Hz, 3 H) LC-MS: MS (ES + ): RT =2.864 min, m/z = 837.3 [M+ H + ]; LCMS Method 25. EXAMPLE 28 – Synthesis of N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]-6-[4-[[4-[4-(1- hydroxy-1-methyl-ethyl)-2-(1-methyl-6-oxo-3-pyridyl)phenoxy] phenoxy]methyl]-1- piperidyl]pyridazine-3-carboxamide (I-235)

[0531] Step 1: Preparation of tert-butyl 4-[[4-(2-bromo-4-methoxycarbonyl- phenoxy)phenoxy]methyl]piperidine-1-carboxylate. To a solution of tert-butyl 4-[(4- hydroxyphenoxy)methyl]piperidine-1-carboxylate (2.0 g, 6.0 mmol, 1.0 equiv) in DMSO (20 mL) was added Cs 2 CO 3 (6.4 g, 19 mmol, 3.0 equiv) and methyl 3-bromo-4-fluoro-benzoate (1.5 g, 6.5 mmol, 1.0 equiv) . The mixture was stirred at 70 °C for 2 h. To the reaction mixture was added water (50 mL) and the mixture was extracted with EtOAc (50 mL). The combined organic phase was washed with brine (50 mL x 3), dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuum. Compound tert-butyl 4-[[4-(2-bromo-4-methoxycarbonyl- phenoxy)phenoxy]methyl]piperidine-1-carboxylate (3.4 g, crude) was obtained as a yellow oil. [0532] Step 2: Preparation of methyl 3-bromo-4-[4-(4-piperidylmethoxy)phenoxy] benzoate. To a solution of tert-butyl 4-[[4-(2-bromo-4-methoxycarbonyl-phenoxy)phenoxy] methyl]piperidine-1-carboxylate (3.4 g, 6.5 mmol, 1.0 equiv) in DCM (10 mL) was added TFA (7.4 g, 65 mmol, 4.8 mL, 10 equiv) .The mixture was stirred at 25 °C for 1 h . To the reaction mixture was added water (50 mL) and the mixture was extracted with EtOAc (50 mL). The combined organic phase was washed with brine (50 mL x 3), dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuum. The residue was purified by prep-HPLC (column: Phenomenex luna C18150*40mm* 15um;mobile phase: [water(FA)-ACN];B%: 23%- 53%,10min). Compound methyl 3-bromo-4-[4-(4-piperidylmethoxy)phenoxy]benzoate (2.6 g, 5.5 mmol, 85% yield, FA salt) was obtained as a yellow oil. [0533] Step 3: Preparation of 2-[3-bromo-4-[4-(4-piperidylmethoxy)phenoxy]phe nyl]propan-2-ol. To a solution of methyl 3-bromo-4-[4-(4-piperidylmethoxy) phenoxy]benzoate (1.0 g, 2.4 mmol, 1.0 equiv) in DCM (50 mL) was added bromo(methyl) magnesium (3.0 M, 24 mL, 30 equiv) at -78-25 °C .The mixture was stirred at -78-25 °C for 12 h . The reaction mixture was added to 1 ml NH 4 Cl at 0 °C. To the reaction mixture was added water (20 mL) and the mixture was extracted with DCM (20 mL). The combined organic phase was washed with brine (20 mL x 3), dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuum The residue was purified by prep-HPLC (column: 3_Phenomenex Luna C1875*30mm*3um; mobile phase: [water(FA)-ACN];B%: 20%-40%,8min). Compound 2-[3-bromo-4-[4-(4- piperidylmethoxy)phenoxy]phe nyl]propan-2-ol (0.23 g, 0.55 mmol, 23% yield) was obtained as a white solid. [0534] Step 4: Preparation of 5-[5-(1-hydroxy-1-methyl-ethyl)-2-[4-(4-piperidylmethoxy) phenoxy]phenyl]-1-methyl-pyridin-2-one. To a solution of 2-[3-bromo-4-[4-(4- piperidylmethoxy)phenoxy]phenyl]propan-2-ol (0.2 g, 0.5 mmol, 1.0 equiv) in THF (3 mL) and H 2 O (0.6 mL) was added K 3 PO 4 (0.3 g, 1.4 mmol, 3.0 equiv) , 1-methyl-5-(4,4,5,5-tetramethyl- 1,3,2-dioxaborolan-2-yl)pyridin-2-one (0.17 mg, 0.70 mmol, 1.5 equiv) and SPhos Pd G3 (37 mg, 48 μmol, 0.1 equiv) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 60 °C for 3 h under N 2 . The reaction mixture was filtered and the filtrate was concentrated to afford crude product The residue was purified by prep-TLC (DCM:MeOH = 7:1) to afford 5-[5-(1- hydroxy-1-methyl-ethyl)-2-[4-(4-piperidylmethoxy)phenoxy]phe nyl]-1-methyl-pyridin-2-one (0.1 g, 0.2 mmol, 46% yield) as a white solid. [0535] Step 5: Preparation of N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]-6-[4-[[4-[4-(1- hydroxy-1-methyl-ethyl)-2-(1-methyl-6-oxo-3-pyridyl)phenoxy] phenoxy]methyl]-1- piperidyl]pyridazine-3-carboxamide. To a solution of 5-[5-(1-hydroxy-1-methyl-ethyl)-2-[4-(4- piperidylmethoxy)phenoxy]phenyl]-1-methyl-pyridin-2-one (0.1 g, 0.2 mmol, 1.0 equiv) in NMP (1 mL) was added K 2 CO 3 (92 mg, 0.67 mmol, 3.0 equiv) and 6-chloro-N-[4-(3-chloro-4-cyano- phenoxy)cyclohexyl]pyridazine-3-carboxamide (87 mg, 0.22 mmol, 1.0 equiv).The mixture was stirred at 50 °C for 12 h. The reaction mixture was filtered and the filtrate was concentrated to afford crude product The residue was purified by prep-HPLC (column: Phenomenex C18 75*30mm*3um;mobile phase: [water(FA)-ACN];B%: 60%-90%,7min) and by prep-HPLC (column: Waters Xbridge 150*25mm* 5um;mobile phase: [water( NH4HCO3)-ACN];B%: 50%- 80%,8min). Compound N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]-6-[4-[[4-[4-(1-hy droxy-1- methyl-ethyl)-2-(1-methyl-6-oxo-3-pyridyl)phenoxy]phenoxy]me thyl]-1-piperidyl]pyridazine-3- carboxamide (14 mg, 17 μmol, 8% yield) was obtained as a white solid. 1 H NMR: (CD3OD, 400 MHz) 7.90 (d, J = 9.7 Hz, 1H), 7.86 - 7.82 (m, 1H), 7.81 - 7.76 (m, 1H), 7.72 - 7.66 (m, 1H), 7.56 - 7.50 (m, 1H), 7.45 - 7.38 (m, 1H), 7.35 - 7.28 (m, 1H), 7.23 - 7.18 (m, 1H), 7.08 - 7.01 (m, 1H), 6.90 - 6.78 (m, 5H), 6.59 - 6.45 (m, 1H), 4.58 (s, 3H), 4.06 - 3.94 (m, 1H), 3.93 - 3.81 (m, 2H), 3.62 - 3.54 (m, 3H), 3.22 - 3.07 (m, 2H), 2.26 - 1.97 (m, 7H), 1.71 - 1.60 (m, 4H), 1.58 - 1.52 (m, 6H), 1.50 - 1.41 (m, 2H) LC-MS: MS (ES + ): RT = 2.395 min, m/z = 803.2 [M + H + ]; LCMS Method 25. EXAMPLE 29– Synthesis of N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]-6-[4-[[4-[4-(3- hydroxyoxetan-3-yl)-2-(6-methyl-7-oxo-1H-pyrrolo[2,3-c]pyrid in-4-yl)phenoxy] phenoxy]methyl]-1-piperidyl]pyridazine-3-carboxamide (I-236) [0536] Step 1: Preparation of 2-[4-(2-bromo-4-iodo-phenoxy)phenoxy]tetrahydropyran. A mixture of 4-tetrahydropyran-2-yloxyphenol (5 g, 26 mmol, 1.0 equiv), 2-bromo-1-fluoro-4-iodo- benzene (8 g, 26 mmol, 1.0 equiv) and Cs2CO3 (17 g, 52 mmol, 2.0 equiv) in DMSO (50 mL) was stirred at 110 °C for 12 h. The reaction mixture was poured into H 2 O (300 mL) at 0°C, and then extracted with EtOAc (200 mL x 2). The organic layers were washed with brine (100 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to afford crude product. The residue was purified by column chromatography (SiO 2 , Petroleum ether/Ethyl acetate=10/1 to 5/1). Compound 2-[4-(2-bromo-4-iodo-phenoxy)phenoxy]tetrahydropyran (11 g, 23 mmol, 88% yield) was obtained as a yellow oil. [0537] Step 2: Preparation of 3-[3-bromo-4-(4-tetrahydropyran-2-yloxyphenoxy) phenyl]oxetan-3-ol. To a solution of 2-[4-(2-bromo-4-iodo-phenoxy)phenoxy]tetrahydropyran (10 g, 21 mmol, 1.0 equiv) in THF (180 mL) was addeddropwise n-BuLi (2.5 M, 10 mL, 1.3 equiv) at -78°C, and then it was stirred for 1 h. oxetan-3-one (3 g, 41 mmol, 2.0 equiv) in THF (20 mL) was added dropwise at -78°C, and then it was stirred at 20 °C for 11 h. The reaction mixture was quenched by sat.NH 4 Cl (50 mL) at 0°C, and then extracted with EtOAc (50 mL x 2). The organic layers were washed with brine (50 mL), dried over Na 2 SO 4 , filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate=5/1 to 3/1). Compound 3-[3-bromo-4-(4-tetrahydropyran-2- yloxyphenoxy)phenyl]oxetan-3-ol (4 g, 8 mmol, 37% yield, 80%purity) was obtained as a yellow oil. [0538] Step 3: Preparation of 3-[3-bromo-4-(4-hydroxyphenoxy)phenyl]oxetan-3-ol. To a solution of 3-[3-bromo-4-(4-tetrahydropyran-2-yloxyphenoxy)phenyl]oxetan -3-ol (3 g, 8 mmol, 1.0 equiv) in MeOH (40 mL) was added 4-methylbenzenesulfonic acid (3 g, 16 mmol, 2.0 equiv).The mixture was stirred at 20 °C for 2 h. The residue was quenched with sat.NaHCO3 (50 mL) and extracted with EA (40 mL x 2). The combined organic layers were dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO 2 , Petroleum ether/Ethyl acetate=10/1 to 1/1) to afford 3-[3-bromo-4- (4-hydroxyphenoxy)phenyl]oxetan-3-ol (2.0 g, 7.0 mmol, 70% yield) as a colorless oil. [0539] Step 4: Preparation of tert-butyl 4-[[4-[2-bromo-4-(3-hydroxyoxetan-3- yl)phenoxy]phenoxy]methyl]piperidine-1-carboxylate. To a solution of 3-[3-bromo-4-(4- hydroxyphenoxy)phenyl]oxetan-3-ol (2.0 g, 6.0 mmol, 1.0 equiv), tert-butyl 4-(bromomethyl) piperidine-1-carboxylate (2.0 g, 7.0 mmol, 1.2 equiv) in MeCN (20 mL) was added K 2 CO 3 (2.0 g, 17 mmol, 3.0 equiv), and then it was stirred at 80 °C for 12 h. The residue was concentrated under reduced pressure to afford crude product. The residue was purified by column chromatography (SiO 2 , Petroleum ether/Ethyl acetate=10/1 to 1/1) and further purified by prep-HPLC (column: Waters Xbridge C18150*50mm* 10um;mobile phase: [water( NH4HCO3)-ACN];B%: 49%- 79%,10min). Compound tert-butyl 4-[[4-[2-bromo-4-(3-hydroxyoxetan-3- yl)phenoxy]phenoxy]methyl]piperidine-1-carboxylate (900 mg, 2 mmol, 30% yield) was obtained as a white solid. [0540] Step 5: Preparation of tert-butyl 4-[[4-[4-(3-hydroxyoxetan-3-yl)-2-[6-methyl-7- oxo-1-(p-tolylsulfonyl)pyrrolo[2,3-c]pyridin-4-yl]phenoxy]ph enoxy]methyl]piperidine-1- carboxylate. To a solution of tert-butyl 4-[[4-[2-bromo-4-(3-hydroxyoxetan-3-yl)phenoxy] phenoxy]methyl]piperidine-1-carboxylate (300 mg, 561 μmol, 1.0 equiv), 6-methyl-1-(p- tolylsulfonyl)-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-y l)pyrrolo[2,3-c]pyridin-7-one (240 mg, 561 μmol, 1.0 equiv) in THF (3 mL) and H2O (0.6 mL) was added SPhos Pd G3 (44 mg, 56 μmol, 0.1 equiv) and K 3 PO 4 (357 mg, 2.0 mmol, 3.0 equiv). The mixture was stirred at 60 °C for 12 h. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by prep-TLC (SiO2, Petroleum ether/Ethyl acetate= 1:30). Compound tert-butyl 4-[[4- [4-(3-hydroxyoxetan-3-yl)-2-[6-methyl-7-oxo-1-(p-tolylsulfon yl)pyrrolo[2,3-c]pyridin-4- yl]phenoxy]phenoxy]methyl]piperidine-1-carboxylate (200 mg, 265 μmol, 47 % yield) was obtained as a yellow oil. [0541] Step 6: Preparation of tert-butyl 4-[[4-[4-(3-hydroxyoxetan-3-yl)-2-(6-methyl-7- oxo-1H-pyrrolo[2,3-c]pyridin-4-yl)phenoxy]phenoxy]methyl]pip eridine-1-carboxylate. To a solution of tert-butyl 4-[[4-[4-(3-hydroxyoxetan-3-yl)-2-[6-methyl-7-oxo-1-(p-tolyl sulfonyl) pyrrolo[2,3-c]pyridin-4-yl]phenoxy]phenoxy]methyl]piperidine -1-carboxylate (200 mg, 265 μmol, 1.0 equiv) in MeOH (2 mL) was added KOH (1 M, 2 mL, 7.8 equiv). The mixture was stirred at 20 °C for 3 h. The reaction mixture was quenched by water (20 mL) and extracted with EA (10 mL x 2), dried over anhydrous Na 2 SO 4 , filtered to afford tert-butyl 4-[[4-[4-(3-hydroxyoxetan-3-yl)-2- (6-methyl-7-oxo-1H-pyrrolo[2,3-c]pyridin-4-yl)phenoxy] phenoxy]methyl]piperidine-1- carboxylate (159 mg, 264 μmol, 99% yield) as a white solid. [0542] Step 7: Preparation of 4-[5-(3-hydroxyoxetan-3-yl)-2-[4-(4-piperidylmethoxy) phenoxy]phenyl]-6-methyl-1H-pyrrolo[2,3-c]pyridin-7-one. To a solution of tert-butyl 4-[[4-[4- (3-hydroxyoxetan-3-yl)-2-(6-methyl-7-oxo-1H-pyrrolo[2,3-c]py ridin-4-yl)phenoxy]phenoxy] methyl]piperidine-1-carboxylate (130 mg, 216 μmol, 1.0 equiv) in DCM (1 mL) was TFA (0.4 mL), and then it was stirred at 20 °C for 2 h. pH was adjusted to 7~8 by sat.NaHCO 3 , and then it was freeze-dried to afford crude product.4-[5-(3-hydroxyoxetan-3-yl)-2-[4-(4- piperidylmethoxy)phenoxy]phenyl]-6-methyl-1H-pyrrolo[2,3-c]p yridin-7-one (110 mg, 211 μmol, 98% yield) was obtained as a yellow oil. [0543] Step 8: Preparation of N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]-6-[4-[[4-[4-(3- hydroxyoxetan-3-yl)-2-(6-methyl-7-oxo-1H-pyrrolo[2,3-c]pyrid in-4-yl)phenoxy]phenoxy] methyl]-1-piperidyl]pyridazine-3-carboxamide. To a solution of 4-[5-(3-hydroxyoxetan-3-yl)-2- [4-(4-piperidylmethoxy)phenoxy]phenyl]-6-methyl-1H-pyrrolo[2 ,3-c]pyridin-7-one (110 mg, 259 μmol, 1.0 equiv), 6-chloro-N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]pyridazin e-3-carboxamide (101 mg, 259 μmol, 1.0 equiv) in NMP (1 mL) was added K 2 CO 3 (107 mg, 778 μmol, 3.0 equiv). The mixture was stirred at 50 °C for 12 h. The reaction mixture was filtered. The filtrate was purified by prep-HPLC (column: Phenomenex luna C18150*25mm* 10um;mobile phase: [water(FA)-ACN];B%: 63%-76%,6min) to afford Compound N-[4-(3-chloro-4-cyano- phenoxy)cyclohexyl]-6-[4-[[4-[4-(3-hydroxyoxetan-3-yl)-2-(6- methyl-7-oxo-1H-pyrrolo[2,3- c]pyridin-4-yl)phenoxy]phenoxy]methyl]-1-piperidyl]pyridazin e-3-carboxamide (6 mg, 7 μmol, 3% yield, 95% purity) as an off-white solid. 1 H NMR: (CD 3 OD, 400 MHz) 12.14 (m, 1 H) 12.07 - 12.21 (m, 1 H) 8.54 - 8.68 (m, 1 H) 7.76 - 7.92 (m, 2 H) 7.27 - 7.54 (m, 7 H) 7.12 - 7.13 (m, 1 H) 6.94 - 7.17 (m, 6 H) 6.70 - 6.79 (m, 1 H) 6.38 - 6.50 (m, 1 H) 6.13 - 6.28 (m, 1 H) 5.04 - 5.27 (m, 2 H) 4.48 - 4.60 (m, 3 H) 3.79 - 3.93 (m, 3 H) 3.52 - 3.61 (m, 3 H) 2.05 - 2.16 (m, 1 H) 2.05 - 2.15 (m, 2 H) 1.47 - 1.68 (m, 4 H) 1.26 - 1.38 (m, 2 H) LC-MS: MS (ES + ): RT =2.224min, m/z =856.4 [M+ H + ]; LCMS Method 25. EXAMPLE 30 – Synthesis of N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]-6-[4-[2-[4-[2-(6- methyl-7-oxo-1H-pyrrolo[2,3-c]pyridin-4-yl)-4-(methylsulfony lmethyl)phenoxy] phenyl] ethyl]-1-piperidyl]pyridazine-3-carboxamide (I-237) [0544] Step 1: Preparation of tert-butyl 4-[2-[4-(2-bromo-4-formyl-phenoxy)phenyl] ethyl]piperidine-1-carboxylate. To a solution of tert-butyl 4-[2-(4-hydroxyphenyl)ethyl] piperidine-1-carboxylate (1 g, 3.27 mmol, 1 equiv) in DMSO (5 mL) was added Cs 2 CO 3 (2.13 g, 6.55 mmol, 2 equiv) and 3-bromo-4-fluoro-benzaldehyde (665 mg, 3.27 mmol, 1 equiv). The mixture was stirred at 80 °C for 1 h and concentrated. The residue was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate=5/1 to 3/1) to give tert-butyl 4-[2-[4-(2- bromo-4-formyl-phenoxy)phenyl]ethyl]piperidine-1-carboxylate (1.17 g, 73 % yield). [0545] Step 2: Preparation of tert-butyl 4-[2-[4-[2-bromo-4-(hydroxymethyl)phenoxy] phenyl]ethyl]piperidine-1-carboxylate. To a solution of tert-butyl 4-[2-[4-(2-bromo-4-formyl- phenoxy)phenyl]ethyl]piperidine-1-carboxylate (1.17 g, 2.40 mmol, 1 equiv) in DCM (10 mL) was added NaBH4 (100 mg, 2.64 mmol, 1.1 equiv). The mixture was stirred at 20 °C for 0.5 h. The mixture was filtered and concentrated to give crude tert-butyl 4-[2-[4-[2-bromo-4- (hydroxymethyl)phenoxy]phenyl]ethyl]piperidine-1-carboxylate (1.15 g) was a yellow oil [0546] Step 3: Preparation of 4-[2-[4-[2-bromo-4-(bromomethyl)phenoxy]phenyl] ethyl]piperidine. To a solution of tert-butyl 4-[2-[4-[2-bromo-4-(hydroxymethyl)phenoxy] phenyl]ethyl]piperidine-1-carboxylate (800 mg, 1.63 mmol, 1 equiv) in DCM (10 mL) was added PBr3 (662 mg, 2.45 mmol, 1.5 equiv) at 0 °C. The mixture was stirred at 20 °C for 0.5 h and concentrated to give crude product 4-[2-[4-[2-bromo-4-(bromomethyl)phenoxy]phenyl] ethyl]piperidine (400 mg) was a yellow oil. [0547] Step 4: Preparation of tert-butyl 4-[2-[4-[2-bromo-4-(bromomethyl)phenoxy] phenyl]ethyl]piperidine-1-carboxylate. To a solution of 4-[2-[4-[2-bromo-4-(bromomethyl) phenoxy]phenyl]ethyl]piperidine (400 mg, 883 μmol, 1 equiv) in DCM (5 mL) was added Boc2O (385 mg, 1.77 mmol, 2 equiv) and DIEA (342 mg, 2.65 mmol, 3 equiv) .The mixture was stirred at 20 °C for 0.5 h. The mixture was filtered and concentrated. The residue was purified by column chromatography (SiO 2 , Petroleum ether/Ethyl acetate=10/1 to 5/1) to give tert-butyl 4-[2-[4-[2- bromo-4-(bromomethyl)phenoxy]phenyl]ethyl]piperidine-1-carbo xylate (180 mg, 37 % yield) was a yellow oil. [0548] Step 5: Preparation of tert-butyl 4-[2-[4-[2-bromo-4-(methylsulfonylmethyl) phenoxy]phenyl]ethyl]piperidine-1-carboxylate. To a solution of tert-butyl 4-[2-[4-[2-bromo-4- (bromomethyl)phenoxy]phenyl]ethyl]piperidine-1-carboxylate (360 mg, 650 μmol, 1 eq) in DMF (2 mL) was addedsodium; methanesulfinate (199 mg, 1.95 mmol, 3 equiv). The mixture was stirred at 20 °C for 0.5 h and concentrated to give crude tert-butyl 4-[2-[4-[2-bromo-4- (methylsulfonylmethyl)phenoxy]phenyl]ethyl]piperidine-1-carb oxylate (350 mg). [0549] Step 6: Preparation of tert-butyl 4-[2-[4-[2-[6-methyl-7-oxo-1-(p-tolylsulfonyl) pyrrolo[2,3-c]pyridin-4-yl]-4-(methylsulfonylmethyl)phenoxy] phenyl]ethyl]piperidine-1- carboxylate. To a solution of tert-butyl 4-[2-[4-[2-bromo-4-(methylsulfonylmethyl)phenoxy] phenyl]ethyl]piperidine-1-carboxylate (250 mg, 452 μmol, 1 equiv) in THF (5 mL) and H 2 O (1 mL) was added K3PO4 (288 mg, 1.36 mmol, 3 equiv) , 6-methyl-1-(p-tolylsulfonyl)-4-(4,4,5,5- tetramethyl-1,3,2-dioxaborolan-2-yl)pyrrolo[2,3-c]pyridin-7- one (194 mg, 452umol, 1 equiv) and SPhos Pd G3 (35 mg, 45 μmol, 0.1 equiv). The mixture was stirred at 60 °C for 3 h. The residue was purified by prep-TLC (SiO2, PE:EA= 1:3) to give tert-butyl 4-[2-[4-[2-[6-methyl-7-oxo-1-(p- tolylsulfonyl)pyrrolo[2,3-c]pyridin-4-yl]-4-(methylsulfonylm ethyl)phenoxy] phenyl]ethyl]piperidine-1-carboxylate (200 mg, 57% yield). [0550] Step 7: Preparation of tert-butyl 4-[2-[4-[2-(6-methyl-7-oxo-1H-pyrrolo[2,3- c]pyridin-4-yl)-4-(methylsulfonylmethyl)phenoxy]phenyl]ethyl ]piperidine-1-carboxylate. To a solution of tert-butyl 4-[2-[4-[2-[6-methyl-7-oxo-1-(p-tolylsulfonyl)pyrrolo[2,3-c] pyridin-4-yl]- 4-(methylsulfonylmethyl)phenoxy]phenyl]ethyl]piperidine-1-ca rboxylate (200 mg, 258 μmol, 1 equiv) in MeOH (2 mL) was added KOH (72 mg, 1.29 mmol, 5 equiv). The mixture was stirred at 20 °C for 0.5 h and concentrated to give crude tert-butyl 4-[2-[4-[2-(6-methyl-7-oxo-1H- pyrrolo[2,3-c]pyridin-4-yl)-4-(methylsulfonylmethyl)phenoxy] phenyl]ethyl]piperidine-1- carboxylate (150 mg). [0551] Step 8: Preparation of 6-methyl-4-[5-(methylsulfonylmethyl)-2-[4-[2-(4-piperidyl) ethyl]phenoxy]phenyl]-1H-pyrrolo[2,3-c]pyridin-7-one. To a solution of tert-butyl 4-[2-[4-[2- (6-methyl-7-oxo-1H-pyrrolo[2,3-c]pyridin-4-yl)-4-(methylsulf onylmethyl)phenoxy] phenyl] ethyl]piperidine-1-carboxylate (150 mg, 242 μmol, 1 equiv) in DCM (2 mL) was added TFA (1 mL).The mixture was stirred at 20 °C for 0.5 h. The mixture was filtered and concentrated to give crude 6-methyl-4-[5-(methylsulfonylmethyl)-2-[4-[2-(4-piperidyl)et hyl] phenoxy]phenyl]-1H- pyrrolo[2,3-c]pyridin-7-one (120 mg, crude) was used into the next step without further purification, which was a yellow oil. [0552] Step 9: Preparation of N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]-6-[4-[2-[4-[2-(6- methyl-7-oxo-1H-pyrrolo[2,3-c]pyridin-4-yl)-4-(methylsulfony lmethyl)phenoxy]phenyl] ethyl]-1-piperidyl]pyridazine-3-carboxamide. To a solution of 6-methyl-4-[5- (methylsulfonylmethyl)-2-[4-[2-(4-piperidyl)ethyl]phenoxy]ph enyl]-1H-pyrrolo[2,3-c]pyridin-7- one (120 mg, 231 μmol, 1 equiv) in NMP (2 mL) was added K2CO3 (96 mg, 693 μmol, 3 equiv) and 6-chloro-N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]pyridazin e-3-carboxamide (90 mg, 231 μmol, 1 equiv). The mixture was stirred at 50 °C for 3 hr. The residue was purified by prep-HPLC (column: Phenomenex luna C18150*25mm* 10um; mobile phase: [water(FA)-ACN]; B %: 59 %- 79 %, 9 min). to give N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]-6-[4-[2-[4-[2-(6- methyl-7- oxo-1H-pyrrolo[2,3-c]pyridin-4-yl)-4-(methylsulfonylmethyl)p henoxy]phenyl]ethyl]-1- piperidyl]pyridazine-3-carboxamide (60 mg, 29 % yield). 1 H NMR (400 MHz, DMSO) δ 12.04 (s, 1 H), 8.55 - 8.63 (m, 1 H), 7.85 (d, J=8.80 Hz, 1 H), 7.79 (d, J=9.56 Hz, 1 H), 7.57 (d, J=2.08 Hz, 1 H), 7.27 - 7.40 (m, 5 H), 7.10 - 7.18 (m, 3 H), 6.95 (d, J=8.32 Hz, 1 H), 6.87 (d, J=8.44 Hz, 2 H), 6.30-6.31 (m, 1 H), 4.41 - 4.57 (m, 5 H), 3.80 - 3.92 (m, 1 H), 3.52 (s, 3 H), 2.89 - 3.01 (m, 5 H), 2.54-2.58 ( m, 2 H), 2.05 - 2.15 (m, 2 H), 1.89 (d, J=10.04 Hz, 2 H), 1.79 (d, J=10.76 Hz, 2 H), 1.59 - 1.70 (m, 2 H), 1.42 - 1.57 (m, 5 H), 1.07 - 1.21 (m, 2 H). LC-MS: MS (ES + ): RT = 2.325 min, m/z = 874.2 [M +H + ]; LCMS Method 25. EXAMPLE 31 – Synthesis of N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]-6-[4-[2-[4-[5- ethylsulfonyl-2-(6-methyl-7-oxo-1H-pyrrolo[2,3-c]pyridin-4-y l)phenyl]-5-oxo-pyrazin-2- yl]ethyl]-1-piperidyl]pyridazine-3-carboxamide (I-238)

[0553] Step 1: Preparation of tert-butyl 4-[2-[5-(2-bromo-4-ethylsulfonyl-phenoxy) pyrazin-2-yl] ethyl] piperidine-1-carboxylate. To a solution of tert-butyl 4-[2-(5- hydroxypyrazin-2-yl) ethyl] piperidine-1-carboxylate (0.77 g, 2.50 mmol, 1 equiv), 2-bromo-4- ethylsulfonyl-1-fluoro-ben zene (736.3 mg, 2.7 mmol, 1.1 equiv) in DMSO (3 mL) was added Cs2CO3 (2.5 g, 7.5 mmol, 3.0 equiv). The mixture was stirred at 70 °C for 1 h. After filtration, the filtrate was purified by prep-HPLC (column: Waters Xbridge C18150*50mm*10um; mobile phase: [water (NH 4 HCO 3 )-ACN]; B%: 44%-74%, 10min) to give a compound tert-butyl 4-[2-[5- (2-bromo-4-ethylsulfonyl-phenoxy) pyrazin-2-yl] ethyl] piperidine-1-carboxylate (0.35 g, 25 % yield) as a white solid. [0554] Step 2: Preparation of tert-butyl 4-[2-[4-[5-ethylsulfonyl-2-[6-methyl-7-oxo-1-(p- tolylsulfonyl)pyrrolo[2,3-c]pyridin-4-yl]phenyl]-5-oxo-pyraz in-2-yl]ethyl]piperidine-1- carboxylate. A mixture of tert-butyl 4-[2-[4-(3-bromo-5-ethylsulfonyl-phenyl)-5-oxo-pyrazin-2- yl]ethyl]piperidine-1-carboxylate (28 mg, 50.5 μmol, 1.0 equiv), 6-methyl-1-(p-tolylsulfonyl)-4- (4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyrrolo[2,3-c]p yridin-7-one (28.2 mg, 65.6 μmol, 1.3 equiv), K 3 PO 4 (32.1 mg, 151.4 μmol, 3.0 equiv) and SPhos PdG3 (3.9 mg, 5.1 μmol, 0.1 equiv) in H2O (0.3 mL), THF (1.5 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 60 °C for 3 h under N2 atmosphere. After concentrated, the residue was purified by prep-HPLC (column: YMC Triart C18150*25mm*5um; mobile phase: [water (FA)-ACN]; B%: 57%-87%, 8.5min) to give a compound tert-butyl 4-[2-[4-[5-ethylsulfonyl-2-[6-methyl-7-oxo-1-(p- tolylsulfonyl)pyrrolo[2,3-c]pyridin-4-yl]phenyl]-5-oxo-pyraz in-2-yl]ethyl]piperidine-1- carboxylate (0.18 g, 64 % yield) as a white solid. [0555] Step 3: Preparation of tert-butyl 4-[2-[4-[5-ethylsulfonyl-2-(6-methyl-7-oxo-1H- pyrrolo [2, 3-c] pyridin-4-yl) phenyl]-5-oxo-pyrazin-2-yl] ethyl] piperidine-1-carboxylate. To a solution of tert-butyl 4-[2-[4-[5-ethylsulfonyl-2-[6-methyl-7-oxo-1-(p-tolylsulfony l) pyrrolo[2,3- c]pyridin-4-yl]phenyl]-5-oxo-pyrazin-2-yl]ethyl]piperidine-1 -carboxylate (140.0 mg, 180.4 μmol, 1.0 equiv) in IPA (2 mL) was added KOH (1 M, 4.87 mL, 27.0 equiv). The mixture was stirred at 25 °C for 1 h. The residue was adjusted to pH = 5~6 by AcOH, diluted with H2O (20 mL) and freeze-dried to give a crude compound. tert-butyl 4-[2-[4-[5-ethylsulfonyl-2-(6-methyl-7-oxo-1H- pyrrolo [2, 3-c] pyridin-4-yl) phenyl]-5-oxo-pyrazin-2-yl] ethyl] piperidine-1-carboxylate (112 mg, 99 % yield) was obtained as a colorless oil. [0556] Step 4: Preparation of compound 4-[4-ethylsulfonyl-2-[2-oxo-5-[2-(4-piperidyl) ethyl] pyrazin-1-yl] phenyl]-6-methyl-1H-pyrrolo [2, 3-c] pyridin-7-one. To a solution of tert- butyl 4-[2-[4-[5-ethylsulfonyl-2-(6-methyl-7-oxo-1H-pyrrolo[2,3-c] pyridin-4-yl)phenyl]-5-oxo- pyrazin-2-yl]ethyl]piperidine-1-carboxylate (0.11 g, 176.9 μmol, 1.0 equiv) in DCM (1 mL) was added TFA (0.5 mL). The mixture was stirred at 25 °C for 0.5 h. The reaction mixture was concentrated under reduced pressure to remove solvent to give compound 4-[4-ethylsulfonyl-2-[2- oxo-5-[2-(4-piperidyl) ethyl] pyrazin-1-yl] phenyl]-6-methyl-1H-pyrrolo [2, 3-c] pyridin-7-one (112 mg, 99 % yield, TFA salt) as a colorless oil. [0557] Step 5: Preparation of N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]-6-[4-[2-[4-[5- ethylsulfonyl-2-(6-methyl-7-oxo-1H-pyrrolo[2,3-c]pyridin-4-y l)phenyl]-5-oxo-pyrazin-2- yl]ethyl]-1-piperidyl]pyridazine-3-carboxamide. To a solution of 4-[4-ethylsulfonyl-2-[2-oxo-5- [2-(4-piperidyl)ethyl]pyrazin-1-yl]phenyl]-6-methyl-1H-pyrro lo[2,3-c]pyridin-7-one (0.11 g, 173.5 μmol, 1.0 equiv, TFA salt), 6-chloro-N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl] pyridazine-3- carboxamide (101.5 mg, 259.8 μmol, 1.5 equiv) in NMP (1 mL) was added K2CO3 (71.7 mg, 519.5 μmol, 3.0 equiv). The mixture was stirred at 50 °C for 12 h. After filtered, the filtrate was purified by prep-HPLC (column: Waters Xbridge 150*25mm* 5um; mobile phase: [water (NH 4 HCO 3 )- ACN]; B%: 38%-68%, 8min) to give a compound N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]- 6-[4-[2-[4-[5-ethylsulfonyl-2-(6-methyl-7-oxo-1H-pyrrolo[2,3 -c]pyridin-4-yl)phenyl]-5-oxo- pyrazin-2-yl]ethyl]-1-piperidyl]pyridazine-3-carboxamide (48 mg, 29.0 % yield) as a yellow solid. 1 H NMR (400 MHz, DMSO-d6) δ 12.10 (s, 1 H), 8.52 - 8.71 (m, 1 H), 8.02 - 8.09 (m, 2 H), 7.96 (d, J = 1.00 Hz, 1 H), 7.76 - 7.91 (m, 3 H), 7.39 (d, J = 2.38 Hz, 1 H) ,7.32 (d, J = 9.63 Hz, 1 H), 7.01 - 7.28 (m, 4 H), 5.91 - 5.99 (m, 1 H), 4.30 - 4.66 (m, 3 H), 3.81 - 3.97 (m, 1 H), 3.44 - 3.54 (m, 5 H), 2.90 (d, J = 2.88 Hz, 2 H), 2.29 - 2.33 (m, 2 H), 2.02 - 2.15 (m, 2 H), 1.90 (d, J = 10.26 Hz, 2 H), 1.58 - 1.74 (m, 4 H), 1.46 - 1.56 (m, 2 H), 1.25 - 1.34 (m, 1 H), 1.12 - 1.24 (m, 5 H), 0.96 - 1.08 (m, 2 H). LC-MS: MS (ES + ): RT = 2.21 min, m/z = 876.2 [M + H + ]; LCMS method: EXAMPLE 32 – Synthesis of N-[4-(3-chloro-4- cyano-phenoxy)cyclohexyl]-6-[4-[2-[4-[4-(1,1- dioxo-1,2-thiazolidin-2-yl)-2-(6-methyl-7-oxo-1H-pyrrolo[2,3 -c]pyridin-4- yl)phenoxy]phenyl]ethyl]-1-piperidyl]pyridazine-3-carboxamid e (I-239)

[0558] Step 1: Preparation of tert-butyl 4-[2-[4-[2-[6-methyl-7-oxo-1-(p-tolylsulfonyl) pyrrolo[2,3-c] pyridin-4-yl]- 4- nitro-phenoxy]phenyl]ethyl]piperidine-1-carboxylate. To a solution of tert-butyl 4-[2-(4-hydroxyphenyl)ethyl]piperidine-1-carboxylate (500 mg, 1.64 mmol, 1 equiv) in NMP (5 mL) was added Cs2CO3 (1.33 g, 4.09 mmol, 2.5 equiv) and 4-(2-fluoro-5-nitro- phenyl)-6-methyl-1-(p-tolylsulfonyl)pyrrolo[2,3-c]pyridin-7- one (795 mg, 1.80 mmol, 1.1 equiv). The mixture was stirred at 70 °C for 1 h. The crude product was used into the next step without further purification. [0559] Step 2: Preparation of tert-butyl 4-[2-[4-[2-(6-methyl-7-oxo-1H-pyrrolo[2,3-c] pyridin-4-yl)- 4-nitro- phenoxy]phenyl]ethyl]piperidine-1-carboxylate. To a solution of tert- butyl 4-[2-[4-[2-[6-methyl-7-oxo-1-(p-tolylsulfonyl)pyrrolo[2,3-c] pyridin-4-yl]- 4- nitro- phenoxy]phenyl]ethyl]piperidine-1-carboxylate (1.2 g, 1.65 mmol, 1 equiv) in MeOH (20 mL) was added KOH (1.32 g, 23.5 mmol, 14 equiv). The mixture was stirred at 20 °C for 0.5 h. The reaction mixture was partitioned between water 30 mL. The organic phase was separated, washed with Ethyl acetate 60 mL (20mL* 3), dried over [anhydrous sodium sulfate], filtered and concentrated. The residue was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate=1/1 to 1/2) to give tert-butyl 4-[2-[4-[2-(6-methyl-7-oxo-1H-pyrrolo[2,3-c] pyridin-4-yl)- 4-nitro- phenoxy]phenyl]ethyl]piperidine-1-carboxylate (900 mg, 95% yield). [0560] Step 3: Preparation of tert-butyl 4-[2-[4-[4-amino-2-(6-methyl-7-oxo-1H - pyrrolo[2,3-c]pyridin-4-yl)phenoxy]phenyl]ethyl]piperidine-1 -carboxylate. A mixture of tert- butyl 4-[2-[4-[2-(6-methyl-7-oxo-1H-pyrrolo[2,3-c]pyridin-4-yl)-4- nitro-phenoxy] phenyl] ethyl]piperidine-1-carboxylate (900 mg, 1.57 mmol, 1 equiv), H2, Pd/C (10% purity) in THF (20 mL) was degassed and purged with N 2 for 3 times, and then the mixture was stirred at 20 °C for 1 h under N2 atmosphere. The mixture was filtered and purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate=1/3 to 0/1) to give tert-butyl 4-[2-[4-[4-amino-2-(6-methyl-7-oxo- 1H - pyrrolo[2,3-c]pyridin-4-yl)phenoxy]phenyl]ethyl]piperidine-1 -carboxylate (800 mg, 94% yield). [0561] Step 4: Preparation of tert-butyl 4-[2-[4-[4-(3-chloropropylsulfonylamino)-2- (6- methyl-7 -oxo-1H –pyrrolo [2,3-c]pyridin-4-yl)phenoxy]phenyl]ethyl]piperidine-1- carboxylate. To a solution of tert-butyl 4-[2-[4-[4-amino-2-(6-methyl-7-oxo-1H-pyrrolo[2,3-c] pyridin-4-yl) phenoxy] phenyl]ethyl]piperidine-1-carboxylate (700 mg, 1.29 mmol, 1 equiv) in DCM (10 mL) was added Et3N (1.31 g, 12.9 mmol, 10 equiv) and 3-chloropropane-1-sulfonyl chloride (685 mg, 3.87 mmol, 470 μL, 3 equiv) at 0 °C. The mixture was stirred at 20 °C for 12 h. The crude product was used into the next step without further purification. [0562] Step 5: Preparation of tert-butyl 4-[2-[4-[4-(1,1-dioxo-1,2- thiazolidin-2-yl)-2-(6- methyl-7-oxo-1H-pyrrolo[2,3-c]pyridin-4-yl)phenoxy]phenyl]et hyl]piperidine-1-carboxylate. To a solution of tert-butyl 4-[2-[4-[4-(3-chloropropylsulfonylamino)-2- (6-methyl-7 -oxo-1H – pyrrolo [2,3-c]pyridin-4-yl)phenoxy]phenyl]ethyl]piperidine-1-carbox ylate (300 mg, 439 μmol, 1 equiv) in MeOH (10 mL) was added NaOH (88 mg, 2.20 mmol, 5 equiv) and Et3N (222 mg, 2.2 mmol, 5 equiv). The mixture was stirred at 80 °C for 12 h. The reaction mixture filtered. The residue was purified by prep-TLC (SiO 2 , Petroleum ether : Ethyl acetate = 0:1) to give tert-butyl 4- [2-[4-[4-(1,1-dioxo-1,2- thiazolidin-2-yl)-2-(6-methyl-7-oxo-1H-pyrrolo[2,3-c]pyridin -4- yl)phenoxy]phenyl]ethyl]piperidine-1-carboxylate (60 mg, 21% yield). [0563] Step 6: Preparation of 4-[5-(1,1-dioxo-1,2-thiazolidin-2-yl)-2-[4-[2-(4-piperidyl) ethyl]phenoxy]phenyl] -6- methyl-1H-pyrrolo[2,3-c]pyridin-7-one. To a solution of tert-butyl 4-[2-[4-[4-(1,1-dioxo-1,2-thiazolidin-2-yl)-2- (6-methyl-7- oxo-1H- pyrrolo [2,3-c]pyridin-4- yl)phenoxy]phenyl]ethyl]piperidine-1-carboxylate (60 mg, 93 μmol, 1 equiv) in DCM (5 mL) was added TFA (1 mL). The mixture was stirred at 20 °C for 0.5 h. The crude product was used into the next step without further purification. [0564] Step 7: Preparation of N-[4-(3-chloro-4- cyano-phenoxy)cyclohexyl]-6-[4-[2-[4-[4- (1,1-dioxo-1,2-thiazolidin-2-yl)-2-(6-methyl-7-oxo-1H-pyrrol o[2,3-c]pyridin-4-yl)phenoxy] phenyl]ethyl]-1-piperidyl]pyridazine-3-carboxamide. To a solution of 4-[5-(1,1-dioxo-1,2- thiazolidin-2-yl)-2-[4-[2-(4-piperidyl) ethyl]phenoxy]phenyl] -6- methyl-1H-pyrrolo[2,3-c]pyridin- 7-one (50 mg, 91 μmol, 1 equiv) in NMP (1 mL) was added K2CO3 (126 mg, 914 μmol, 10 equiv) and 6-chloro-N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl] pyridazine -3-carboxamide (36 mg, 92 μmol, 1 equiv). The mixture was stirred at 50 °C for 12 h. The reaction mixture was filtered. The residue was purified by prep-HPLC (FA condition; column: Phenomenex luna C18150*25mm* 10um;mobile phase: [water(FA)-ACN];B%: 65%-95%,10min) to give N-[4-(3-chloro-4- cyano- phenoxy)cyclohexyl]-6-[4-[2-[4-[4-(1,1-dioxo-1,2-thiazolidin -2-yl)-2-(6-methyl-7-oxo-1H- pyrrolo[2,3-c]pyridin-4-yl)phenoxy]phenyl]ethyl]-1-piperidyl ]pyridazine-3-carboxamide (25 mg, 29% yield). 1 H NMR (400 MHz, CDCl3): δ 10.33 - 10.05 (m, 1H), 7.97 (m, 1H), 7.89 (m, 1H), 7.57 (d, J = 8.8 Hz, 1H), 7.46 (d, J = 2.4 Hz, 1H), 7.25 (m, 1H), 7.12 (s, 1H), 7.07 - 6.98 (m, 5H), 6.86 (m, 1H), 6.78 (d, J = 8.4 Hz, 2H), 6.48 (s, 1H), 4.56 - 4.42 (m, 2H), 4.36 - 4.28 (m, 1H), 4.06 (m, 1H), 3.80 (t, J = 6.4 Hz, 2H), 3.64 (s, 3H), 3.42 (t, J = 7.6 Hz, 2H), 3.02 (m, 2H), 2.62 - 2.53 (m, 4H), 2.21 - 2.14 (m, 4H), 1.71 - 1.43 (m, 8H), 1.32 - 1.23 (m, 3H) LC-MS: MS (ES+): RT = 2.417 min, m/z = 901.4 [M + H+]; LCMS Method 25. EXAMPLE 33 – Synthesis of N-((1r,4r)-4-(3-chloro-4-cyanophenoxy)cyclohexyl)-6-(4- (((1r,3r)-3-(4-((ethylsulfonyl)methyl)-2-(6-methyl-7-oxo-6,7 -dihydro-1H-pyrrolo[2,3- c]pyridin-4-yl)phenoxy)cyclobutoxy)methyl)piperidin-1-yl)pyr idazine-3-carboxamide (I-240) [0565] Step 1: Preparation of tert-butyl 4-(((1r,3r)-3-(2-bromo-4-((ethylsulfonyl)methyl) phenoxy)cyclobutoxy)methyl)piperidine-1-carboxylate. To a solution of 2-bromo-4- (ethylsulfonylmethyl)phenol (293 mg, 1.05 mmol, 1.0 equiv) and tert-butyl-4-(((1s,3s)-3- hydroxycyclobutoxy)methyl)piperidine-1-carboxylate (300 mg, 1.05 mmol, 1.0 equiv) in THF (4 mL) was added PPh3 (551 mg, 2.10 mmol, 2.0 equiv), and then DIAD (425 mg, 2.10 mmol, 0.4 mL, 2.0 equiv) was added at 0°C.The mixture was stirred at 0~25 °C for 8 h.The reaction was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (column: Phenomenex luna C18150*40mm* 15um;mobile phase: [water(FA)- ACN];B%: 55%-85%,15min) to give tert-butyl 4-(((1r,3r)-3-(2-bromo-4-((ethylsulfonyl)methyl) phenoxy)cyclobutoxy)methyl)piperidine-1-carboxylate (300 mg, 548 μmol, 52% yield) was obtained as a off-white solid. [0566] Step 2: Preparation of tert-butyl 4-(((1r,3r)-3-(4-((ethylsulfonyl)methyl)-2-(6- methyl-7-oxo-1-tosyl-6,7-dihydro-1H-pyrrolo[2,3-c]pyridin-4- yl)phenoxy)cyclobutoxy) methyl)piperidine-1-carboxylate. To a solution of 6-methyl-1-(p-tolylsulfonyl)-4-(4,4,5,5- tetramethyl-1,3,2-dioxaborolan-2-yl)pyrrolo[2,3-c]pyridin-7- one (305 mg, 713 μmol, 1.3 equiv) and tert-butyl 4-(((1r,3r)-3-(2-bromo-4-((ethylsulfonyl)methyl)phenoxy)cycl obutoxy)methyl) piperidine-1-carboxylate (300 mg, 548 μmol, 1.0 equiv) in THF (5 mL) and H 2 O (1 mL) was added K3PO4 (349 mg, 1.65 mmol, 3.0 equiv) and SPhos Pd G3 (42 mg, 54 μmol, 0.1 equiv) .The mixture was stirred at 60 °C for 3 h.The reaction mixture was filtered and concentrated under reduced pressure to give a reside. The residue was purified by prep-HPLC (column: Phenomenex luna C18150*25mm* 10um;mobile phase: [water(FA)-ACN];B%: 60%-90%,10min) to give tert- butyl 4-(((1r,3r)-3-(4-((ethylsulfonyl)methyl)-2-(6-methyl-7-oxo-1 -tosyl-6,7-dihydro-1H- pyrrolo[2,3-c]pyridin-4-yl)phenoxy)cyclobutoxy)methyl)piperi dine-1-carboxylate (260 mg, 338 μmol, 61% yield) was obtained as a off-white solid. [0567] Step 3: Preparation of tert-butyl 4-(((1r,3r)-3-(4-((ethylsulfonyl)methyl)-2-(6- methyl-7-oxo-6,7-dihydro-1H-pyrrolo[2,3-c]pyridin-4-yl)pheno xy)cyclobutoxy)methyl) piperidine-1-carboxylate. To a solution of tert-butyl 4-(((1r,3r)-3-(4-((ethylsulfonyl)methyl)-2-(6- methyl-7-oxo-1-tosyl-6,7-dihydro-1H-pyrrolo[2,3-c]pyridin-4- yl)phenoxy)cyclobutoxy) methyl)piperidine-1-carboxylate (260 mg, 338 μmol, 1.0 equiv) in i-PrOH (5 mL) was added KOH (1 M, 5 mL, 15.0 equiv). The mixture was stirred at 25 °C for 2 h. The reaction mixture was extracted with EA 20 mL. The combined organic layers were washed with H2O (2 x 10 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give tert-butyl 4-(((1r,3r) -3-(4- ((ethylsulfonyl)methyl)-2-(6-methyl-7-oxo-6,7-dihydro-1H-pyr rolo[2,3-c]pyridin-4- yl)phenoxy)cyclobutoxy)methyl)piperidine-1-carboxylate (200 mg, 325 μmol, 96% yield) was obtained as a yellow oil. [0568] Step 4: Preparation of 4-(5-((ethylsulfonyl)methyl)-2-((1r,3r)-3-(piperidin-4- ylmethoxy)cyclobutoxy)phenyl)-6-methyl-1,6-dihydro-7H-pyrrol o[2,3-c]pyridin-7-one. To a solution of tert-butyl 4-(((1r,3r)-3-(4-((ethylsulfonyl)methyl)-2-(6-methyl-7-oxo-6 ,7-dihydro-1H- pyrrolo[2,3-c]pyridin-4-yl)phenoxy)cyclobutoxy)methyl)piperi dine-1-carboxylate (200 mg, 325 μmol, 1.0 equiv) in DCM (1 mL) was added TFA (0.5 mL). The mixture was stirred at 25 °C for 1 h. The reaction mixture was filtered and concentrated under reduced pressure to give 4-(5- ((ethylsulfonyl)methyl)-2-((1r,3r)-3-(piperidin-4-ylmethoxy) cyclobutoxy)phenyl)-6-methyl-1,6- dihydro-7H-pyrrolo[2,3-c]pyridin-7-one (200 mg, 318 μmol, 97% yield, TFA salt) was obtained as a red oil. [0569] Step 5: Preparation of N-((1r,4r)-4-(3-chloro-4-cyanophenoxy)cyclohexyl)-6-(4- (((1r,3r)-3-(4-((ethylsulfonyl)methyl)-2-(6-methyl-7-oxo-6,7 -dihydro-1H-pyrrolo[2,3- c]pyridin-4-yl)phenoxy)cyclobutoxy)methyl)piperidin-1-yl)pyr idazine-3-carboxamide. To a solution of 6-chloro-N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]pyridazin e-3-carboxamide (149 mg, 382 μmol, 1.2 equiv) and 4-(5-((ethylsulfonyl)methyl)-2-((1r,3r)-3-(piperidin-4- ylmethoxy)cyclobutoxy)phenyl)-6-methyl-1,6-dihydro-7H-pyrrol o[2,3-c]pyridin-7-one (200 mg, 318 μmol, 1.0 equiv, TFA salt) in NMP (3 mL) was added K2CO3 (440 mg, 3.19 mmol, 10.0 equiv). The mixture was stirred at 50 °C for 8 h. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (column: Phenomenex luna C18150*25mm* 10um;mobile phase: [water(FA)-ACN];B%: 60%- 90%,10min) to give N-((1r,4r)-4-(3-chloro-4-cyanophenoxy)cyclohexyl)-6-(4-(((1r ,3r)-3-(4- ((ethylsulfonyl)methyl)-2-(6-methyl-7-oxo-6,7-dihydro-1H-pyr rolo[2,3-c]pyridin-4-yl)phenoxy) cyclobutoxy)methyl)piperidin-1-yl)pyridazine-3-carboxamide (110 mg, 122 μmol, 38% yield, 96.11% purity) was obtained as a yellow solid . 1 H NMR (400 MHz, DMSO-d 6 ) δ 12.00 (br s, 1H), 8.58 (d, J = 8.3 Hz, 1H), 7.83 (dd, J = 9.2, 18.4 Hz, 2H), 7.43 (d, J = 2.1 Hz, 1H), 7.39 - 7.23 (m, 5H), 7.13 (dd, J = 2.3, 8.8 Hz, 1H), 6.88 (d, J = 8.5 Hz, 1H), 6.24 - 6.19 (m, 1H), 4.89 - 4.78 (m, 1H), 4.48 (br d, J = 13.3 Hz, 4H), 4.13 - 4.03 (m, 1H), 3.90 - 3.80 (m, 2H), 3.61 (m, 4H), 3.15 (br d, J = 6.1 Hz, 2H), 3.08 - 2.95 (m, 4H), 2.35 (br d, J = 4.6 Hz, 2H), 2.26 - 2.17 (m, 2H), 2.15 - 2.04 (m, 2H), 1.96 - 1.83 (m, 3H), 1.76 (td, J = 3.1, 6.7 Hz, 3H), 1.64 (br d, J = 13.1 Hz, 2H), 1.52 (br s, 2H), 1.22 (br t, J = 7.4 Hz, 3H) LC-MS: MS (ES + ): RT = 2.54 min, m/z = 868.4 [M +H + ]; LCMS method: 10. EXAMPLE 34 – Synthesis of 6-[4-[2-[4-[4-bromo-2-(6-methyl-7-oxo-1H-pyrrolo[2,3- c]pyridin-4-yl)phenoxy]phenyl]ethyl]-1-piperidyl]-N-[4-(3-ch loro-4-cyano- phenoxy)cyclohexyl]pyridazine-3-carboxamide (I-241)

[0570] Step 1: Preparation of tert-butyl 4-[2-[4-(2-bromo-4-nitro-phenoxy)phenyl] ethyl]piperidine-1-carboxylate. The reaction was conducted by three batches in parallel. A mixture of tert-butyl 4-[2-(4-hydroxyphenyl)ethyl]piperidine-1-carboxylate (48.0 g, 157 mmol, 1 equiv), 2-bromo-1-fluoro-4-nitro-benzene (34.6 g, 157 mmol, 1 equiv) and Cs 2 CO 3 (102 g, 314 mmol, 2 equiv) in CH3CN (500 mL) was stirred at 60 °C for 12 h. The reaction was filtered and the filter cake was washed with ethyl acetate (500 mL). The combined filtrate was concentrated in vacuo to afford tert-butyl 4-[2-[4-(2-bromo-4-nitro-phenoxy)phenyl] ethyl]piperidine-1- carboxylate (234 g, 463 mmol, 98% yield) as a black brown solid. [0571] Step 2: Preparation of tert-butyl 4-[2-[4-(4-amino-2-bromo-phenoxy)phenyl] ethyl]piperidine-1-carboxylate. This reaction was conducted by three batches in parallel. To a solution of tert-butyl 4-[2-[4-(2-bromo-4-nitro-phenoxy)phenyl]ethyl]piperidine-1-c arboxylate (39.0 g, 77.2 mmol, 1 equiv) and NH 4 Cl (24.8 g, 463 mmol, 6 equiv) in EtOH (450 mL) and H 2 O (50 mL) was added Fe (25.9 g, 463 mmol, 6 equiv) at 25 °C. The reaction was stirred for 10 h at 80 °C. The reaction was filtered and the filter cake was washed with EtOH (600 mL). The combined organic phase was concentrated in vacuo. The residue was triturated with mixture solution (800 mL, Petroleum ether/Ethyl acetate = 6/1). The solid was collected by filtration and dried in vacuo to afford tert-butyl 4-[2-[4-(4-amino-2-bromo-phenoxy)phenyl]ethyl]piperidine-1- carboxylate (102 g, 214 mmol, 93% yield) as a brown solid. [0572] Step 3: Preparation of tert-butyl 4-[2-[4-[4-amino-2-[6-methyl-7-oxo-1-(p- tolylsulfonyl)pyrrolo[2,3-c]pyridin-4-yl]phenoxy]phenyl]ethy l]piperidine-1-carboxylate. A mixture of tert-butyl 4-[2-[4-(4-amino-2-bromo-phenoxy)phenyl]ethyl]piperidine-1-c arboxylate (29.1 g, 61.3 mmol, 1.05 equiv), 6-methyl-1-(p-tolylsulfonyl)-4-(4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2-yl)pyrrolo[2,3-c]pyridin-7-one (25.0 g, 58.4 mmol, 1 equiv), SPhos Pd G3 (4.55 g, 5.84 mmol, 0.1 equiv) and K 3 PO 4 (37.2 g, 175 mmol, 3 eq) in dioxane (610 mL) and H 2 O (61 mL) was stirred at 70 °C for 10 h under N 2 . The reaction mixture was evaporated to remove the solvent. The residue was purified by column chromatography on silica gel (Petroleum ether/Ethyl acetate = 100/1 to 1/1) to afford tert-butyl 4-[2-[4-[4-amino-2-[6-methyl-7-oxo-1-(p- tolylsulfonyl)pyrrolo[2,3-c]pyridin-4-yl]phenoxy]phenyl]ethy l]piperidine-1-carboxylate (31.0 g, 44.5 mmol, 76% yield) as a yellow solid. [0573] Step 4: Preparation of tert-butyl 4-[2-[4-[4-bromo-2-[6-methyl-7-oxo-1-(p- tolylsulfonyl)pyrrolo[2,3-c]pyridin-4-yl]phenoxy]phenyl]ethy l]piperidine-1-carboxylate. This reaction was conducted by three batches in parallel. A mixture of CuBr (4.94 g, 34.4 mmol, 2.0 equiv) and tert-butyl nitrite (3.55 g, 34.4 mmol 2.0 equiv) in ACN (150 mL) was stirred at 25 °C for 0.5 h under N2. Then to the reaction mixture was added tert-butyl 4-[2-[4-[4-amino-2-[6- methyl-7-oxo-1-(p-tolylsulfonyl)pyrrolo[2,3-c]pyridin-4-yl]p henoxy]phenyl]ethyl]piperidine-1- carboxylate (12.0 g, 17.2 mmol, 1 equiv) at 25 °C. The reaction mixture was stirred at 50 °C for 1.5 h under N 2 . The reaction was evaporated to remove the solvents. The residue was diluted with ethyl acetate (1000 mL) and water (500 mL). The pH of the mixture was adjusted to 8 by addition of saturated aqueous NaHCO3 solution. The mixture was filtered and the filtrate was extracted with ethyl acetate (500 mL*4). The combined organic phase was dried with anhydrous Na 2 SO 4 , filtered and concentrated in vacuo. The residue was purified by column chromatography on silica gel (Petroleum ether/Ethyl acetate = 40/1 to 1/1) to afforded tert-butyl 4-[2-[4-[4-bromo-2-[6- methyl-7-oxo-1-(p-tolylsulfonyl)pyrrolo[2,3-c]pyridin-4-yl]p henoxy]phenyl] ethyl]piperidine-1- carboxylate (18.0 g, 23.7 mmol, 46% yield) as a brown solid. [0574] Step 5: Preparation of tert-butyl 4-[2-[4-[4-bromo-2-(6-methyl-7-oxo-1H- pyrrolo[2,3-c]pyridin-4-yl)phenoxy]phenyl]ethyl]piperidine-1 -carboxylate. To a mixture of tert-butyl 4-[2-[4-[4-bromo-2-[6-methyl-7-oxo-1-(p-tolylsulfonyl)pyrrol o[2,3-c]pyridin-4- yl]phenoxy]phenyl]ethyl]piperidine-1-carboxylate (20.0 g, 26.3 mmol, 1 equiv) in MeOH (500 mL) was added KOH (7.38 g, 131 mmol, 5 equiv) at 25 °C. The reaction mixture was stirred at 25 °C for 1.5 h. The reaction was evaporated to remove the solvents. The residue was diluted with water (500 mL) and Ethyl acetate (500 mL). The mixture was extracted with ethyl acetate (300 mL*4). The combined organic phase was dried with anhydrous Na 2 SO 4 , filtered and concentrated in vacuo to afford tert-butyl 4-[2-[4-[4-bromo-2-(6-methyl-7-oxo-1H-pyrrolo[2,3-c]pyridin- 4- yl)phenoxy]phenyl]ethyl]piperidine-1-carboxylate (15.4 g, 25.5 mmol, 97% yield) as a yellow solid. [0575] Step 6: Preparation of 4-[5-bromo-2-[4-[2-(4-piperidyl)ethyl]phenoxy]phenyl]-6- methyl-1H-pyrrolo[2,3-c]pyridin-7-one. A mixture of tert-butyl 4-[2-[4-[4-bromo-2-(6-methyl-7- oxo-1H-pyrrolo[2,3-c]pyridin-4-yl)phenoxy]phenyl]ethyl]piper idine-1-carboxylate (15.4 g, 25.46 mmol, 1 equiv) and HCl/dioxane (4 N, 200 mL, 31 equiv) in CH2Cl2 (50 mL) was stirred at 25 °C for 1 h. The reaction was dried in vacuo to afford 4-[5-bromo-2-[4-[2-(4-piperidyl) ethyl]phenoxy]phenyl]-6-methyl-1H-pyrrolo[2,3-c]pyridin-7-on e (17.0 g, crude, HCl) as yellow solid. [0576] Step 7: Preparation of 6-[4-[2-[4-[4-bromo-2-(6-methyl-7-oxo-1H-pyrrolo[2,3- c]pyridin-4-yl)phenoxy]phenyl]ethyl]-1-piperidyl]-N-[4-(3-ch loro-4-cyano-phenoxy) cyclohexyl]pyridazine-3-carboxamide. A mixture of 4-[5-bromo-2-[4-[2-(4- piperidyl)ethyl]phenoxy]phenyl]-6-methyl-1H-pyrrolo[2,3-c]py ridin-7-one (12.0 g, 22.1 mmol, 1 equiv, HCl), 6-chloro-N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]pyridazin e-3-carboxamide (9.08 g, 23.2 mmol, 1.05 equiv) and DIPEA (17.1 g, 133 mmol, 6 equiv) in NMP (130 mL) was stirred at 80 °C for 10 h. The combined mixture was poured into water (1000 mL) and stirred at 25 °C for 0.5 h. The precipitate solid was collected by filtration. The filtrate was extracted with ethyl acetate (200 mL*6). The combined organic phase was washed with brine (100 mL*5), dried with anhydrous Na 2 SO 4 , filtered and concentrated in vacuo. The residue and the filter cake was purified by column chromatography on silica gel (Petroleum ether/Ethyl acetate = 10/1 to 1/5 and Ethyl acetate/Dichloromethan = 5/1 to 1/1). The crude product was triturated with Ethyl acetate/MeOH (20/1, 400 mL). The solid was collected by filtration and dried in vacuo to afford 6-[4-[2-[4-[4- bromo-2-(6-methyl-7-oxo-1H-pyrrolo[2,3-c]pyridin-4-yl)phenox y]phenyl]ethyl]-1-piperidyl]-N- [4-(3-chloro-4-cyano-phenoxy)cyclohexyl]pyridazine-3-carboxa mide (11.9 g, 13.5 mmol, 61% yield, 98% purity) was obtained as an off-white solid 1 H NMR (400 MHz, DMSO-d 6 ) δ 12.03 (s, 1H), 8.58 (d, 1H, J = 8.4 Hz), 7.85 (d, 1H, J = 8.8 Hz), 7.79 (d, 1H, J = 9.6 Hz), 7.62 (d, 1H, J = 2.4 Hz), 7.51 (dd, 1H, J = 2.4, 8.8 Hz), 7.38 (d, 1H, J = 2.4 Hz), 7.3-7.3 (m, 2H), 7.26 (t, 1H, J = 2.8 Hz), 7.30-7.25 (m, 3H), 6.95-6.80 (m, 3H), 6.23 (t, 1H, J = 2.0 Hz), 4.78-4.62 (m, 3H), 3.98- 3.82 (m, 1H), 3.52 (s, 3H), 3.10-2.90 (m, 2H), 2.60-2.50 (m, 2H), 2.25-2.08 (m, 2H), 1.95-1.85 (m, 2H), 1.85-1.75 (m, 2H), 1.85-1.65 (m, 7H), 1.30-1.15 (m, 2H). LC-MS: MS (ES + ): RT = 2.688 min, m/z = 860.2 [M + H + ]; LCMS Method 25. EXAMPLE 35 – Synthesis of N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]-6-[4-[2-[4-[4- (ethylsulfonylamino)-2-(2-methyl-1-oxo-4-isoquinolyl)phenoxy ]-2-fluoro-phenyl]ethyl]-1- piperidyl]pyridazine-3-carboxamide (I-242) [0577] Step 1: Preparation of 4-(2-fluoro-5-nitro-phenyl)-2-methyl-isoquinolin-1-one. A mixture of 4-bromo-2-methyl-isoquinolin-1-one (1 g, 4 mmol, 1 equiv) , (2-fluoro-5-nitro- phenyl)boronic acid (1.55 g, 8 mmol, 2 equiv) , [2-(2-aminophenyl)phenyl]palladium(1+);bis(1- adamantyl)-butyl-phosphane;methanesulfonate (152 mg,210 μmol, 0.05 equiv) and K3PO4 (2.67 g, 12 mmol, 3 equiv) in THF (20 mL) and H2O (2 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 60 °C for 12 h under N 2 atmosphere. The reaction mixture was diluted with H2O 30 mL and extracted with EA 50 ml. The combined organic layers were washed with brina 100 mL, filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-TLC (SiO 2 , EA: PE = 1: 1) to afford 4-(2-fluoro-5-nitro-phenyl)-2-methyl- isoquinolin-1-one (1 g, 79 % yield) as a yellow solid. [0578] Step 2: Preparation of tert-butyl 4-[2-[2-fluoro-4-[2-(2-methyl-1-oxo-4-isoquinolyl)- 4-nitro-phenoxy]phenyl]ethyl]piperidine-1-carboxylate. To a solution of tert-butyl 4-[2-(2- fluoro-4-hydroxy-phenyl)ethyl]piperidine-1-carboxylate (250 mg, 773 μmol, 1 equiv) and 4-(2- fluoro-5-nitro-phenyl)-2-methyl-isoquinolin-1-one (230 mg, 773 μmol, 1 equiv) in NMP (4 mL) was added Cs2CO3 (503 mg, 1 mmol, 2 equiv).The mixture was stirred at 70 °C for 1 h. After filtration, The residue was purified by prep-HPLC (column: Phenomenex luna C18150*25mm* 10um;mobile phase: [water(FA)-ACN];B%: 77 %-100 %,10 min) to afford compound tert-butyl 4- [2-[2-fluoro-4-[2-(2-methyl-1-oxo-4-isoquinolyl)-4-nitro-phe noxy] phenyl]ethyl]piperidine-1- carboxylate (268 mg, 57 % yield) as a yellow oil. [0579] Step 3: Preparation of tert-butyl 4-[2-[4-[4-amino-2-(2-methyl-1-oxo-4- isoquinolyl)phenoxy]-2-fluoro-phenyl] ethyl] piperidine-1-carboxylate. To a solution of tert- butyl 4-[2-[2-fluoro-4-[2-(2-methyl-1-oxo-4-isoquinolyl)-4-nitro-p henoxy]phenyl]ethyl] piperidine-1-carboxylate (260 mg, 432 μmol, 1 equiv) in MeOH (5 mL) was added Pd /C (100 mg, 10% purity) under N2 atmosphere. The mixture was stirred under H2 (15 Psi or atm.) at 25 °C for 4 h. After filtration, the residue was purified by prep-TLC (SiO 2 , EA: PE = 1: 1) to afford tert-butyl 4-[2-[4-[4-amino-2-(2-methyl-1-oxo-4-isoquinolyl) phenoxy]-2-fluoro-phenyl] ethyl] piperidine-1- carboxylate (140 mg, 56 % yield). [0580] Step 4: Preparation of tert-butyl 4-[2-[4-[4-(ethylsulfonylamino)-2-(2-methyl-1-oxo- 4-isoquinolyl) phenoxy]-2-fluoro-phenyl] ethyl] iperidine-1-carboxylate. To a solution of tert- butyl 4-[2-[4-[4-amino-2-(2-methyl-1-oxo-4-isoquinolyl)phenoxy]-2- fluoro-phenyl]ethyl] piperidine-1-carboxylate (100 mg, 174 μmol, 1 equiv) and ethanesulfonyl chloride (67 mg, 524 μmol, 3 equiv) in DCM (5 mL) was added TEA (88 mg, 874 μmol, 5 equiv). The mixture was stirred at 25 °C for 12 h. The mixture was diluted with H 2 O 20 mL and extracted with DCM 60 mL. The combined organic layers were washed with brine 50 mL, filtered and concentrated. The residue was purified by prep-TLC (SiO2, EA: PE = 3: 1) to afford compound tert-butyl 4-[2-[4-[4- (ethylsulfonylamino)-2-(2-methyl-1-oxo-4-isoquinolyl) phenoxy]-2-fluoro-phenyl] ethyl] iperidine-1-carboxylate (crude, TFA). [0581] Step 5: Preparation of N-[4-[3-fluoro-4-[2-(4-piperidyl)ethyl]phenoxy]-3-(2-methyl- 1-oxo-4-isoquinolyl) phenyl]ethanesulfonamide. To a solution of tert-butyl 4-[2-[4-[4- (ethylsulfonylamino)-2-(2-methyl-1-oxo-4-isoquinolyl)phenoxy ]-2-fluoro-phenyl]ethyl] piperidine-1-carboxylate (100 mg, 150 μmol, 1 equiv) in DCM (3 mL) was added TFA (500 μL).The mixture was stirred at 25 °C for 30 min. After filtration, the filtrate was concentrated to afford crude product. [0582] Step 6: Preparation of N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]-6-[4-[2-[4-[4- (ethylsulfonylamino)-2-(2-methyl-1-oxo-4-isoquinolyl)phenoxy ]-2-fluoro-phenyl]ethyl]-1- piperidyl]pyridazine-3-carboxamide. To a solution of N-[4-[3-fluoro-4-[2-(4-piperidyl)ethyl] phenoxy]-3-(2-methyl-1-oxo-4-isoquinolyl) phenyl]ethanesulfonamide (80 mg, 118 μmol, 1 equiv, TFA salt) and 6-chloro-N-[4-(3-chloro-4-cyano-phenoxy) cyclohexyl] pyridazine-3-carboxamide (48 mg, 123 μmol, 1 equiv) in NMP (2 mL) was added K 2 CO 3 (81 mg, 590 μmol, 5 equiv). The mixture was stirred at 50 °C for 12 h. After filtration, The residue was purified by prep-HPLC (column: Phenomenex luna C18150*25mm* 10um;mobile phase: [water(FA)-ACN];B%: 69 %- 99 %,10 min) to afford N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]-6-[4-[2-[4-[4- (ethylsulfonylamino)-2-(2-methyl-1-oxo-4-isoquinolyl)phenoxy ]-2-fluoro-phenyl]ethyl]-1- piperidyl]pyridazine-3-carboxamide (62 mg, 56 % yield). 1 H NMR (DMSO-d6, 400 MHz) δ 9.89 (s, 1H), 8.59 (d, 1H, J=8.1 Hz), 8.24 (d, 1H, J=7.6 Hz), 7.86 (d, 1H, J=8.8 Hz), 7.80 (d, 1H, J=9.5 Hz), 7.7-7.6 (m, 1H), 7.5-7.5 (m, 2H), 7.40 (d, 1H, J=2.3 Hz), 7.4-7.3 (m, 1H), 7.3-7.3 (m, 2H), 7.24 (d, 1H, J=2.6 Hz), 7.2-7.1 (m, 1H), 7.1-7.1 (m, 2H), 6.6-6.5 (m, 2H), 4.6-4.4 (m, 3H), 3.9-3.8 (m, 1H), 3.50 (s, 3H), 3.2-3.1 (m, 2H), 3.0-2.9 (m, 2H), 2.1-2.1 (m, 2H), 1.9-1.9 (m, 2H), 1.78 (d, 2H, J=13.0 Hz), 1.7-1.6 (m, 2H), 1.7-1.4 (m, 4H), 1.4-1.4 (m, 2H), 1.25 (t, 5H, J=7.3 Hz), 1.2-1.1 (m, 2H). LC-MS: MS (ES+): RT = 2.399 min, m/z =918.4 [M+ H + ]; LCMS Method 25.

EXAMPLE 36 – Synthesis of N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]-6-[4-[2-[4-[4- ethylsulfonyl-2-(2-methyl-1-oxo-4-isoquinolyl)phenoxy]-2-flu oro-phenyl]ethyl]-1- piperidyl]pyridazine-3-carboxamide (I-243) [0583] Step 1: Preparation of tert-butyl 4-[2-[4-[4-ethylsulfonyl-2-(2-methyl-1-oxo-4- isoquinolyl)phenoxy]-2-fluoro-phenyl]ethyl]piperidine-1-carb oxylate. To a solution of tert- butyl 4-[2-[4-(2-bromo-4-ethylsulfonyl-phenoxy)-2-fluoro-phenyl]et hyl]piperidine-1-carboxylate (150 mg, 262 μmol, 1 equiv) and 2-methyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2- yl)isoquinolin-1-one (89 mg, 315 μmol, 1 equiv) in THF (4 mL) and H 2 O (0.8 mL) was added SPhos Pd G3 (20 mg, 26 μmol, 0.1 equiv) and K3PO4 (167 mg, 788 μmol, 3 equiv). The mixture was stirred at 60 °C for 12 h. After filtration, The residue was purified by prep-TLC (SiO2, EA: PE = 1: 1) to afford tert-butyl 4-[2-[4-[4-ethylsulfonyl-2-(2-methyl-1-oxo-4-isoquinolyl)phe noxy]-2- fluoro-phenyl]ethyl]piperidine-1-carboxylate (60 mg, 35 % yield). [0584] Step 2: Preparation of 4-[5-ethylsulfonyl-2-[3-fluoro-4-[2-(4-piperidyl)ethyl] phenoxy]phenyl]-2-methyl-isoquinolin-1-one. To a solution of tert-butyl 4-[2-[4-[4- ethylsulfonyl-2-(2-methyl-1-oxo-4-isoquinolyl)phenoxy]-2-flu oro-phenyl]ethyl]piperidine-1- carboxylate (60 mg, 92 μmol, 1 equiv) in DCM (3 mL) was added TFA (500 μL). The mixture was stirred at 25 °C for 30 min. After filtration, the filtrate was concentrated to afford crude product. [0585] Step 3: Preparation of N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]-6-[4-[2-[4-[4- ethylsulfonyl-2-(2-methyl-1-oxo-4-isoquinolyl)phenoxy]-2-flu oro-phenyl]ethyl]-1-piperidyl] pyridazine-3-carboxamide. To a solution of 4-[5-ethylsulfonyl-2-[3-fluoro-4-[2-(4-piperidyl) ethyl]phenoxy]phenyl]-2-methyl-isoquinolin-1-one (50 mg, 75 μmol, 1 equiv, TFA salt) and 6- chloro-N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]pyridazine- 3-carboxamide (31 mg, 79 μmol, 1 equiv) in NMP (1.5 mL) was added K2CO3 (52 mg, 377 μmol, 5 equiv).The mixture was stirred at 50 °C for 12 h. After filtration, The residue was purified by prep-HPLC (column: Phenomenex luna C18150*25mm* 10um;mobile phase: [water(FA)-ACN];B %: 71 %-100 %,10 min) to afford N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]-6-[4-[2-[4-[4-eth ylsulfonyl-2-(2-methyl-1-oxo-4- isoquinolyl)phenoxy]-2-fluoro-phenyl]ethyl]-1-piperidyl] pyridazine-3-carboxamide (21 mg, 29 % yield). 1 H NMR (DMSO-d 6 , 400 MHz) δ 8.59 (d, 1H, J=8.1 Hz), 8.29 (d, 1H, J=8.0 Hz), 7.95 (d, 1H, J=2.4, 8.6 Hz), 7.91 (d, 1H, J=2.4 Hz), 7.86 (d, 1H, J=8.8 Hz), 7.80 (d, 1H, J=9.5 Hz), 7.7-7.7 (m, 1H), 7.63 (s, 1H), 7.53 (t, 1H, J=7.5 Hz), 7.4-7.4 (m, 2H), 7.34 ( d, 1H, J=9.8 Hz), 7.3-7.3 (m, 1H), 7.14 (d, 2H, J=8.5 Hz), 6.96 (d, 1H, J=2.3, 10.8 Hz), 6.84 (d, 1H, J=1.9, 8.4 Hz), 4.6-4.5 (m, 3H), 3.9-3.8 (m, 1H), 3.56 (s, 3H), 2.98 ( t, 2H, J=11.9 Hz), 2.61 ( d, 3H, J=6.3, 8.1 Hz), 2.1-2.1 (m, 2H), 1.9-1.9 (m, 2H), 1.8-1.8 (m, 2H), 1.7-1.4 (m, 8H), 1.17 (t, 5H, J=7.3 Hz). LC-MS: MS (ES+): RT = 2.483 min, m/z =906.4 [M+ H + ]; LCMS Method 25.

EXAMPLE 37 – Synthesis of N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]-6-[4-[5- cyclopropyl-5-[2-(6-methyl-7-oxo-1H-pyrrolo[2,3-c]pyridin-4- yl)-4-methylsulfonyl- phenoxy]pentyl]piperazin-1-yl]pyridazine-3-carboxamide (I-244) [0586] Step 1: Preparation of tert-butyl 4-(5-hydroxypentyl)piperazine-1-carboxylate. A mixture of tert-butyl piperazine-1-carboxylate (10.0 g, 53.7 mmol, 1.0 equiv), 5-bromopentan-1-ol (8.97 g, 53.7 mmol, 1.0 equiv), DIEA (20.8 g, 161 mmol, 28 mL, 3.0 equiv) in MeCN (100 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 50 °C for 12 h under N2 atmosphere. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO 2 , Petroleum ether/Ethyl acetate=1/1 to 0/1). Compound tert-butyl 4-(5-hydroxypentyl)piperazine-1-carboxylate (7.00 g, 25.7 mmol, 48% yield) was obtained as a yellow solid. [0587] Step 2: Preparation of tert-butyl 4-(5-oxopentyl)piperazine-1-carboxylate. DMSO (1.50 g, 19.2 mmol, 1.5 mL, 5.2 equiv) was slowly added to a solution ofoxalyl dichloride (1.45 g, 11.4 mmol, 1 mL, 3.1 equiv) in distilled DCM (15 mL) at -78 °C and the mixture was stirred at -78 °C for 0.5 h. Then tert-butyl 4-(5-hydroxypentyl)piperazine-1-carboxylate (1.00 g, 3.67 mmol, 1.0 equiv) was added and the mixture was stirred at -78 °C for 1 h. There after DIPEA (3.34 g, 25.8 mmol, 4.5 mL, 7.0 equiv) was slowly added at -78 °C, and the mixture was stirred at 25 °C for 0.5 h. The crude product tert-butyl 4-(5-oxopentyl)piperazine-1-carboxylate (0.99 g, 3.66 mmol, 100% yield) as a colorless oil was used into the next step without further purification. [0588] Step 3: Preparation of tert-butyl 4-(5-cyclopropyl-5-hydroxy-pentyl)piperazine-1- carboxylate. To a solution of tert-butyl 4-(5-oxopentyl)piperazine-1-carboxylate (0.99 g, 3.66 mmol, 1.0 equiv) in DCM (15 mL) was added bromo(cyclopropyl)magnesium (0.5 M, 150 mL, 20.5 equiv) at -78 °C. The mixture was stirred at -78 °C-25 °C for 12 h. The reaction mixture was quenched by addition water 50 mL at 25 °C, and then diluted with DCM 50 mL and washed with brine 50 mL, dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (FA condition; column: Shim-pack C18 150*25*10um;mobile phase: [water(FA)-ACN];B%: 2%-32%,10min). Compound tert-butyl 4-(5- cyclopropyl-5-hydroxy-pentyl)piperazine-1-carboxylate (0.50 g, 1.60 mmol, 44% yield) was obtained as a white solid. [0589] Step 4: Preparation of tert-butyl 4-[5-(2-bromo-4-methylsulfonyl-phenoxy)-5- cyclopropyl-pentyl]piperazine-1-carboxylate. To a stirred solution of tert-butyl 4-(5- cyclopropyl-5-hydroxy-pentyl)piperazine-1-carboxylate (200 mg, 640 μmol, 1.0 equiv) in DMF (4 mL) was added NaH (38 mg, 0.96 mmol, 60% purity, 1.5 equiv) at 0 °C. The mixture was stirred at 0 °C for 1 h.2-bromo-1-fluoro-4-methylsulfonyl-benzene (194 mg, 768 μmol, 1.2 equiv) was then added at 0 °C. The mixture was warmed to room temperature (25 °C) and stirred at 25 °C for 11 h. To the reaction mixture was added water (30 mL) and the mixture was extracted with ethyl acetate (30 mL x 3). The combined organic phase was washed with brine (20 mL), dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo. The crude product was purified by preparative HPLC (column: Phenomenex luna C18150*25mm* 10um;mobile phase: [water(FA)- ACN];B%: 17%-47%,10min as additive) to yield. Compound tert-butyl 4-[5-(2-bromo-4- methylsulfonyl-phenoxy)-5-cyclopropyl-pentyl]piperazine-1-ca rboxylate (180 mg, 330 μmol, 52% yield) was obtained as a white solid. [0590] Step 5: Preparation of tert-butyl4-[5-cyclopropyl-5-[2-[6-methyl-7-oxo-1-(p- tolylsulfonyl)pyrrolo[2,3-c]pyridin-4-yl]-4-methylsulfonyl-p henoxy]pentyl]piperazine-1- carboxylate. A mixture of tert-butyl 4-[5-(2-bromo-4-methylsulfonyl-phenoxy)-5-cyclopropyl- pentyl]piperazine-1-carboxylate (180 mg, 330 μmol,1 equiv), 6-methyl-1-(p-tolylsulfonyl)-4- (4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyrrolo[2,3-c]p yridin-7-one (184 mg, 429 μmol, 1.3 equiv), SPhos Pd G3 (26 mg, 33 μmol, 0.1 equiv), K3PO4 (1.5 M, 660 μL, 3.0 equiv) in THF (10 mL) was degassed and purged with N 2 for 3 times, and then the mixture was stirred at 60 °C for 12 h under N2 atmosphere. The reaction was filtered and the filtrate was concentrated in vacuo to give the crude product. The crude product was purified by preparative HPLC (column: YMC Triart C18 150*25mm*5um;mobile phase: [water(FA)-ACN];B%: 30%-60%,8.5min as additive) to yield. Compound tert-butyl4-[5-cyclopropyl-5-[2-[6-methyl-7-oxo-1-(p-tolylsu lfonyl)pyrrolo [2,3- c]pyridin-4-yl]-4-methylsulfonyl-phenoxy]pentyl]piperazine-1 -carboxylate (120 mg, 156 μmol, 47% yield) was obtained as a white solid. [0591] Step 6: Preparation of tert-butyl4-[5-cyclopropyl-5-[2-(6-methyl-7-oxo-1H- pyrrolo[2,3-c]pyridin-4-yl)-4-methylsulfonyl-phenoxy]pentyl] piperazine-1-carboxylate. To a solution of tert-butyl 4-[5-cyclopropyl-5-[2-[6-methyl-7-oxo-1-(p-tolylsulfonyl)pyr rolo[2,3- c]pyridin-4-yl]-4-methylsulfonyl-phenoxy]pentyl]piperazine-1 -carboxylate (120 mg, 156 μmol, 1.0 equiv) in MeOH (3 mL) was added NaOH (1 M, 2.4 mL, 15.0 equiv). The mixture was stirred at 25 °C for 1 h. To the reaction mixture was added water (10 mL) and the mixture was extracted with ethyl acetate (20 mL x 3). The combined organic phase was washed with brine (20 mL), dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo. Compound tert-butyl4-[5-cyclopropyl- 5-[2-(6-methyl-7-oxo-1H-pyrrolo[2,3-c]pyridin-4-yl)-4-methyl sulfonyl- phenoxy]pentyl]piperazine-1-carboxylate (90 mg, crude) was obtained as a white solid. [0592] Step 7: Preparation of 4-[2-(1-cyclopropyl-5-piperazin-1-yl-pentoxy)-5- methylsulfonyl-phenyl]-6-methyl-1H-pyrrolo[2,3-c]pyridin-7-o ne. To a solution of tert-butyl 4- [5-cyclopropyl-5-[2-(6-methyl-7-oxo-1H-pyrrolo[2,3-c]pyridin -4-yl)-4-methylsulfonyl- phenoxy]pentyl]piperazine-1-carboxylate (90 mg, 0.15 mmol, 1.0 equiv) in DCM (3 mL) was added 2,6-dimethylpyridine (315 mg, 2.94 mmol, 342 μL, 20.0 equiv) and TMSOTf (326 mg, 1.47 mmol, 265 μL, 10.0 equiv). The mixture was stirred at -78-25 °C for 1 h. The reaction mixture was concentrated in vacuo to give the crude product. Compound 4-[2-(1-cyclopropyl-5-piperazin-1-yl- pentoxy)-5-methylsulfonyl-phenyl]-6-methyl-1H-pyrrolo[2,3-c] pyridin-7-one (70 mg, crude) was obtained as a white solid. [0593] Step 8: Preparation of N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]-6-[4-[5- cyclopropyl-5-[2-(6-methyl-7-oxo-1H-pyrrolo[2,3-c]pyridin-4- yl)-4-methylsulfonyl- phenoxy]pentyl]piperazin-1-yl]pyridazine-3-carboxamide. To a solution of 4-[2-(1- cyclopropyl-5-piperazin-1-yl-pentoxy)-5-methylsulfonyl-pheny l]-6-methyl-1H-pyrrolo[2,3- c]pyridin-7-one (70 mg, 0.14 mmol, 1.0 equiv) in NMP (2 mL) was added K2CO3 (94 mg, 0.68 mmol, 5.0 equiv) and 6-chloro-N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]pyridazin e-3- carboxamide (53 mg, 0.14 mmol, 1.0 equiv). The mixture was stirred at 50 °C for 1 h. The reaction was filtered and the filtrate was concentrated in vacuo to give the crude product. The crude product was purified by preparative HPLC (column: Phenomenex Luna C18150*25mm*10um;mobile phase: [water(FA)-ACN];B%: 25%-55%,10min as additive) to yield. Compound N-[4-(3-chloro-4- cyano-phenoxy)cyclohexyl]-6-[4-[5-cyclopropyl-5-[2-(6-methyl -7-oxo-1H-pyrrolo[2,3-c]pyridin- 4-yl)-4-methylsulfonyl-phenoxy]pentyl]piperazin-1-yl]pyridaz ine-3-carboxamide (61 mg, 69 μmol, 51% yield, 99% purity) was obtained as a yellow solid. 1 H NMR (400 MHz, DMSO-d 6 ) δ 12.06 (s, 1H), 8.62 (d, J = 8.4 Hz, 1H), 7.88 - 7.80 (m, 4H), 7.41 - 7.27 (m, 5H), 7.13 (s, 1H), 6.14 (t, J = 2.4 Hz, 1H), 4.59 - 4.47 (m, 1H), 4.17 - 4.08 (m, 1H), 3.85 (s, 1H), 3.73 - 3.59 (m, 3H), 3.57 (s, 3H), 3.31 - 3.29 (m, 2H), 3.21 (s, 3H), 2.48 - 2.20 (m, 5H), 2.14 - 2.06 (m, 2H), 1.89 (d, J = 10.4 Hz, 2H), 1.70 - 1.58 (m, 4H), 1.56-1.46 (m, 2H), 1.44 - 1.25 (m, 4H), 0.96 (s, 1H), 0.51-0.36 (m, 2H), 0.33-0.20 (m, 2H). LC-MS: MS (ES + ): RT = 2.094 min, m/z = 867.4 [M + H + ]; LCMS Method 25. EXAMPLE 38 – Synthesis of N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]-6-[4-[[3-[4-(1,1- dioxothiolan-2-yl)-2-(6-methyl-7-oxo-1H-pyrrolo[2,3-c]pyridi n-4-yl)phenoxy]cyclobutoxy] methyl]-1-piperidyl]pyridazine-3-carboxamide (I-245)

[0594] Step 1: Preparation of 2-chloro-4-(1,1-dioxothiolan-2-yl)phenol. To a solution of thiolane 1,1-dioxide (4.95 g, 41.2 mmol, 3.9 mL, 2.0 equiv) in THF (60 mL) was added n-BuLi (2.5 M, 16.5 mL, 2.0 equiv) at -78°C. The mixture was stirred at -78 °C for 0.5 h. ZnCl2 (2 M, 20.6 mL, 2.0 equiv) was added at -78 °C. The mixture was stirred at 25 °C for 0.5 h.2-(4-bromo-2- chloro-phenoxy)tetrahydropyran (6 g, 20.6 mmol, 1.0 equiv), Pd(OAc)2 (462 mg, 2.06 mmol, 0.1 equiv) and XPhos (1.96 g, 4.12 mmol, 0.2 equiv) in THF (20 mL) was added and stirred at 65 °C for 11 h. The mixture was quenched with Saturate the ammonium chloride solution (150 mL) and extracted with ethyl acetate (150 mL * 3). The combined organic layers were washed with brine (150 mL * 3), dried over sodium sulfate, filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (column: Phenomenex Luna C18150 * 25mm * 10 μm; mobile phase: [water (FA)-ACN]; B%: 38%-68%, 10 min) to give 2-chloro-4-(1,1- dioxothiolan-2-yl)phenol (2.6 g, 51% yield) as a yellow solid. [0595] Step 2: Preparation of tert-butyl 4-[[3-[2-chloro-4-(1,1-dioxothiolan-2- yl)phenoxy]cyclobutoxy]methyl]piperidine-1-carboxylate. To a solution of tert-butyl 4-[(3- hydroxycyclobutoxy)methyl]piperidine-1-carboxylate (400 mg, 1.40 mmol, 1.5 equiv) in THF (6 mL) was added PPh3 (0.25 M, 11.2 mL, 3.0 equiv), 2-chloro-4-(1,1-dioxothiolan-2-yl)phenol (231 mg, 934 μmol, 1.0 equiv) and DIAD (378 mg, 1.87 mmol, 0.4 mL, 2.0 equiv) at 0°C. The mixture was stirred at 25 °C for 12 h. The mixture was concentrated to give a residue. The residue was purified by prep-HPLC (column: Phenomenex luna C18150 * 25mm * 10 μm; mobile phase:[water(FA)-ACN]; B%: 56%-86%, min) to give tert-butyl 4-[[3-[2-chloro-4-(1,1- dioxothiolan-2-yl)phenoxy]cyclobutoxy]methyl]piperidine-1-ca rboxylate (400 mg, 83% yield) as a yellow solid. [0596] Step 3: Preparation of tert-butyl 4-[[3-[4-(1,1-dioxothiolan-2-yl)-2-[6-methyl-7-oxo- 1-(p-tolylsulfonyl)pyrrolo[2,3-c]pyridin-4-yl]phenoxy]cyclob utoxy]methyl]piperidine-1- carboxylate. To a solution of 6-methyl-1-(p-tolylsulfonyl)-4-(4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2-yl)pyrrolo[2,3-c]pyridin-7-one (367 mg, 856 μmol, 1.1 equiv) in THF (10 mL) and H 2 O (2 mL) was added SPhos Pd G3 (61 mg, 78 μmol, 0.1 equiv), tert-butyl 4-[[3-[2-chloro-4- (1,1-dioxothiolan-2-yl)phenoxy]cyclobutoxy]methyl]piperidine -1-carboxylate (400 mg, 778 μmol, 1.0 equiv) and K3PO4 (496 mg, 2.33 mmol, 3.0 equiv). The mixture was stirred at 60 °C for 2 h. The reaction mixture was diluted with water (10 mL) and extracted with ethyl acetate (20 mL * 3). The combined organic layers were washed with brine (20 mL), dried over sodium sulfate, filtered and concentrated under reduced pressure to give tert-butyl 4-[[3-[4-(1,1-dioxothiolan-2-yl)-2-[6- methyl-7-oxo-1-(p-tolylsulfonyl)pyrrolo[2,3-c]pyridin-4- yl]phenoxy]cyclobutoxy]methyl]piperidine-1-carboxylate (500 mg, crude) as a yellow oil. [0597] Step 4: Preparation of tert-butyl 4-[[3-[4-(1,1-dioxothiolan-2-yl)-2-(6-methyl-7-oxo- 1H-pyrrolo[2,3-c]pyridin-4-yl)phenoxy]cyclobutoxy]methyl]pip eridine-1-carboxylate. To a solution of tert-butyl 4-[[3-[4-(1,1-dioxothiolan-2-yl)-2-[6-methyl-7-oxo-1-(p- tolylsulfonyl)pyrrolo[2,3-c]pyridin-4-yl]phenoxy]cyclobutoxy ]methyl]piperidine-1-carboxylate (500 mg, 641 μmol, 1.0 equiv) in MeOH (5 mL) was added KOH (360 mg, 6.41 mmol, 38.5 mL, 10.0 equiv). The mixture was stirred at 25 °C for 1 h. The mixture was adjusted pH to 7 with AcOH and concentrated to give a residue. The residue was purified by prep-HPLC (column: Phenomenex Luna C18150 * 25mm * 10um; mobile phase: [water (FA)-ACN]; B%: 48%-78%, 9 min) to give tert-butyl 4-[[3-[4-(1,1-dioxothiolan-2-yl)-2-(6-methyl-7-oxo-1H-pyrrol o[2,3- c]pyridin-4-yl)phenoxy]cyclobutoxy]methyl]piperidine-1-carbo xylate (190 mg, 47% yield) as a yellow solid. [0598] Step 5: Preparation of 4-[5-(1,1-dioxothiolan-2-yl)-2-[3-(4-piperidylmethoxy) cyclobutoxy]phenyl]-6-methyl-1H-pyrrolo[2,3-c]pyridin-7-one. To a solution of tert-butyl 4- [[3-[4-(1,1-dioxothiolan-2-yl)-2-(6-methyl-7-oxo-1H-pyrrolo[ 2,3-c]pyridin-4- yl)phenoxy]cyclobutoxy]methyl]piperidine-1-carboxylate (190 mg, 304 μmol, 1.0 equiv) in DCM (2 mL) was added TFA (616 mg, 5.40 mmol, 0.4 mL, 17.8 equiv). The mixture was stirred at 25 °C for 1 h. The mixture was concentrated to give 4-[5-(1,1-dioxothiolan-2-yl)-2-[3-(4- piperidylmethoxy)cyclobutoxy]phenyl]-6-methyl-1H-pyrrolo[2,3 -c]pyridin-7-one (190 mg, 98% yield, TFA salt) as a yellow oil. [0599] Step 6: Preparation of N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]-6-[4-[[3-[4-(1,1- dioxothiolan-2-yl)-2-(6-methyl-7-oxo-1H-pyrrolo[2,3-c]pyridi n-4-yl)phenoxy] cyclobutoxy]methyl]-1-piperidyl]pyridazine-3-carboxamide. To a solution of 4-[5-(1,1- dioxothiolan-2-yl)-2-[3-(4-piperidylmethoxy)cyclobutoxy]phen yl]-6-methyl-1Hpyrrolo[2,3- c]pyridin-7-one (190 mg, 297 μmol, 1.0 equiv, TFA salt) in NMP (2.5 mL) was added K2CO3 (205 mg, 1.49 mmol, 5.0 equiv) and 6-chloro-N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]pyridazin e- 3-carboxamide (116 mg, 297 μmol, 1.0 equiv). The mixture was stirred at 50 °C for 12 h. The mixture was concentrated to give a residue. The residue was purified by prep-HPLC (column: Waters Xbridge 150 * 25mm * 5um; mobile phase: [water (NH4HCO3)-ACN]; B%: 48%-78%, 10 min) to give N-[4-(3-chloro-4-cyano-phenoxy) cyclohexyl]-6-[4-[[3-[4-(1,1-dioxothiolan-2-yl)-2- (6-methyl-7-oxo-1H-pyrrolo[2,3-c]pyridin-4-yl)phenoxy]cyclob utoxy]methyl]-1- piperidyl]pyridazine-3-carboxamide (34 mg, 12% yield, 95% purity) as an off-white solid. 1 H NMR (400 MHz, DMSO-d 6 ): δ 11.99 (s, 1H), 8.58 (d, J = 8.4 Hz, 1H), 7.85 (d, J = 8.8 Hz, 1H), 7.80 (d, J = 9.6 Hz, 1H), 7.38 (m, J = 2.0, 8.4 Hz, 2H), 7.33 (d, J = 9.6 Hz, 1H), 7.31-7.26 (m, 2H), 7.23 (s, 1H), 7.13 (m, J = 2.4, 8.8 Hz, 1H), 6.89 (d, J = 8.4 Hz, 1H), 6.19 (d, J = 2.0 Hz, 1H), 4.91- 4.76 (m, 1H), 4.56-4.43 (m, 3H), 4.33 (m, J = 6.8, 12.0 Hz, 1H), 4.07 (t, J = 4.8 Hz, 1H), 3.92-3.80 (m, 1H), 3.57 (s, 3H), 3.19-3.13 (m, 3H), 3.04-2.96 (m, 2H), 2.39-2.30 (m, 3H), 2.28-2.16 (m, 4H), 2.14-2.07 (m, 3H), 1.89 (m, J = 3.2, 9.2 Hz, 3H), 1.79-1.73 (m, 2H), 1.68-1.59 (m, 2H), 1.56-1.48 (m, 2H), 1.27-1.13 (m, 3H) LC-MS: MS (ES + ): RT = 2.553 min, m/z = 880.4 [M + H + ]; LCMS method: 10. EXAMPLE 39 – Synthesis of N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]-6-[4-[2-[4-[4-(1, 1- dioxothiazinan-2-yl)-2-(6-methyl-7-oxo-1H-pyrrolo[2,3-c]pyri din-4-yl)phenoxy]phenyl] ethyl]-1-piperidyl]pyridazine-3-carboxamide (I-246)

[0600] Step 1: Preparation of tert-butyl 4-[2-(4-benzyloxyphenyl)ethynyl]piperidine-1- carboxylate. To a solution of tert-butyl 4-ethynylpiperidine-1-carboxylate (5.0 g, 23.8 mmol, 1.0 equiv), 1-benzyloxy-4-bromo-benzene (6.9 g, 26.2 mmol, 1.1 equiv), CuI (910 mg, 4.7 mmol, 0.2 equiv), TEA (12.0 g, 119.4 mmol, 16.6 mL, 5.0 equiv) and Pd(PPh3)4 (2.7 g, 2.3 mmol, 0.1 equiv) in DMF (100 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 90 °C for 3 h under N2 atmosphere. The residue was diluted with H2O (10 mL) and extracted with DCM (3 x 10 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 to give a residue. The residue was purified by column chromatography (SiO2, Petroleum ether/ Ethyl acetate = 50/1 to 3/1) to give compound tert-butyl 4-[2-(4-benzyloxyphenyl)ethynyl]piperidine-1-carboxylate (5.6 g, 59% yield). [0601] Step 2: Preparation of tert-butyl 4-[2-(4-hydroxyphenyl)ethyl]piperidine-1- carboxylate. To a solution of tert-butyl 4-[2-(4-benzyloxyphenyl)ethynyl]piperidine-1-carboxylate (6.7 g, 17.1 mmol, 1.0 equiv), Pd/C (3.0 g, 10% purity) in MeOH (70 mL) was stirred at 40 °C for 12 h under H2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure. The residue was used for next step without further purification to give compound tert- butyl 4-[2-(4-hydroxyphenyl)ethyl]piperidine-1-carboxylate (4.8 g, 93% yield). [0602] Step 3: Preparation of tert-butyl 4-[2-[4-(2-bromo-4-nitro-phenoxy)phenyl] ethyl]piperidine-1-carboxylate. To a solution of tert-butyl 4-[2-(4-hydroxyphenyl)ethyl] piperidine-1-carboxylate (4.5 g, 14.7 mmol, 1.0 equiv), 2-bromo-1-fluoro-4-nitro-benzene (3.8 g, 17.6 mmol, 1.2 equiv), K 2 CO 3 (4.0 g, 29.4 mmol, 2.0 equiv) in ACN (70 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 80 °C for 12 h under N2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure. The residue was purified by column chromatography (SiO 2 , Petroleum ether/ Ethyl acetate = 50/1 to 10/1) to give compound tert-butyl 4-[2-[4-(2-bromo-4-nitro-phenoxy)phenyl]ethyl]piperidine-1- carboxylate (6.8 g, 92% yield). [0603] Step 4: Preparation of tert-butyl 4-[2-[4-(4-amino-2-bromo-phenoxy)phenyl] ethyl]piperidine-1-carboxylate. To a solution of tert-butyl 4-[2-[4-(2-bromo-4-nitro-phenoxy) phenyl]ethyl]piperidine-1-carboxylate (6.8 g, 13.6 mmol, 1.0 equiv), Fe (3.8 g, 68.1 mmol, 5.0 equiv), NH4Cl (7.2 g, 136.3 mmol, 10.0 equiv) in EtOH (35 mL), H2O (35 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 85 °C for 12 h under N2 atmosphere. The reaction mixture was filtered under reduced pressure. The residue was diluted with H2O (20 mL) and extracted with DCM (3 x 20 mL). The combined organic layers were washed with brine (3 x 10 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO 2 , Petroleum ether/ Ethyl acetate = 50/1 to 10/1) to give compound tert-butyl 4-[2-[4-(4-amino-2-bromo- phenoxy)phenyl]ethyl]piperidine-1-carboxylate (2.2 g, 33% yield). [0604] Step 5: Preparation of tert-butyl 4-[2-[4-[4-[bis(4-chlorobutylsulfonyl)amino]-2- bromo-phenoxy]phenyl]ethyl]piperidine-1-carboxylate. To a solution of tert-butyl 4-[2-[4-(4- amino-2-bromo-phenoxy)phenyl]ethyl]piperidine-1-carboxylate (600 mg, 1.2 mmol, 1.0 equiv), 4- chlorobutane-1-sulfonyl chloride (723 mg, 3.7 mmol, 3.0 equiv), TEA (638 mg, 6.3 mmol, 878 μL, 5.0 equiv) in DCM (10 mL) was degassed and purged with N2 for 3 times at 0 °C, and then the mixture was stirred at 25 °C for 12 h under N 2 atmosphere. The residue was diluted with H 2 O (10 mL) and extracted with DCM (3 x 10 mL). The combined organic layers were washed with brine (3 x 10 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was used for next step without further purification to give compound tert- butyl 4-[2-[4-[4-[bis(4-chlorobutylsulfonyl)amino]-2-bromo-phenoxy ]phenyl]ethyl]piperidine-1- carboxylate (990 mg, 99 % yield). [0605] Step 6: Preparation of tert-butyl 4-[2-[4-[2-bromo-4-(1,1-dioxothiazinan-2- yl)phenoxy]phenyl]ethyl]piperidine-1-carboxylate. A solution of tert-butyl 4-[2-[4-[4-[bis(4- chlorobutylsulfonyl)amino]-2-bromo-phenoxy]phenyl]ethyl]pipe ridine-1-carboxylate (990 mg, 1.2 mmol, 1.0 equiv), NaOH (252 mg, 6.3 mmol, 5.0 equiv), TEA (638 mg, 6.3 mmol, 878 μL, 5.0 equiv) in MeOH (10 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 80 °C for 12 h under N 2 atmosphere. The residue was diluted with H 2 O (10 mL) and extracted with DCM (3 x 10 mL). The combined organic layers were washed with brine (3 x 10 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, Petroleum ether/ Ethyl acetate = 50/1 to 5/1) to give compound tert-butyl 4-[2-[4-[2-bromo-4-(1,1-dioxothiazinan-2-yl)phenoxy] phenyl]ethyl]piperidine-1-carboxylate (612 mg, 81% yield). [0606] Step 7: Preparation of tert-butyl 4-[2-[4-[4-(1,1-dioxothiazinan-2-yl)-2-(6-methyl-7- oxo-1H-pyrrolo[2,3-c]pyridin-4-yl)phenoxy]phenyl]ethyl]piper idine-1-carboxylate. To a solution of tert-butyl 4-[2-[4-[2-bromo-4-(1,1-dioxothiazinan-2-yl)phenoxy]phenyl]e thyl] piperidine-1-carboxylate (300 mg, 505 μmol, 1.0 equiv), 6-methyl-1-(p-tolylsulfonyl)-4-(4,4,5,5- tetramethyl-1,3,2-dioxaborolan-2-yl)pyrrolo[2,3-c]pyridin-7- one (259 mg, 606 μmol, 1.2 equiv), K3PO4 (321 mg, 1.5 mmol, 3.0 equiv), SPhos Pd G3 (39 mg, 50 μmol, 0.1 equiv) and NaOH (101 mg, 2.5 mmol, 5.0 equiv) in THF (6 mL), H 2 O (1.2 mL), MeOH (2 mL) was degassed and purged with N 2 for 3 times, and then the mixture was stirred at 60 °C for 2 h under N 2 atmosphere. The residue was diluted with H2O (10 mL) and extracted with DCM (3 x 10 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 to give a residue. The residue was purified by column chromatography (SiO2, Petroleum ether/ Ethyl acetate = 50/1 to 0/1) to give compound tert-butyl 4-[2-[4-[4-(1,1- dioxothiazinan-2-yl)-2-(6-methyl-7-oxo-1H-pyrrolo[2,3-c]pyri din-4- yl)phenoxy]phenyl]ethyl]piperidine-1-carboxylate (330 mg, 98 % yield). [0607] Step 8: Preparation of 4-[5-(1,1-dioxothiazinan-2-yl)-2-[4-[2-(4-piperidyl)ethyl] phenoxy]phenyl]-6-methyl-1H-pyrrolo[2,3-c]pyridin-7-one. To a solution of tert-butyl 4-[2-[4- [4-(1,1-dioxothiazinan-2-yl)-2-(6-methyl-7-oxo-1H-pyrrolo[2, 3-c]pyridin-4-yl)phenoxy] phenyl]ethyl]piperidine-1-carboxylate (320 mg, 484 μmol, 1.0 equiv) in TFA (1 mL), DCM (3 mL) was degassed and purged with N 2 for 3 times, and then the mixture was stirred at 25 °C for 1 h under N2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure. The residue was used for next step without further purification to give compound 4-[5- (1,1-dioxothiazinan-2-yl)-2-[4-[2-(4-piperidyl)ethyl]phenoxy ]phenyl]-6-methyl-1H-pyrrolo[2,3- c]pyridin-7-one (326 mg, 99% yield, TFA). [0608] Step 9: Preparation of N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]-6-[4-[2-[4-[4- (1,1-dioxothiazinan-2-yl)-2-(6-methyl-7-oxo-1H-pyrrolo[2,3-c ]pyridin-4-yl)phenoxy] phenyl]ethyl]-1-piperidyl]pyridazine-3-carboxamide. To a solution of 4-[5-(1,1-dioxothiazinan- 2-yl)-2-[4-[2-(4-piperidyl)ethyl]phenoxy]phenyl]-6-methyl-1H -pyrrolo[2,3-c]pyridin-7-one (326 mg, 483 μmol, 1.0 equiv, TFA), 6-chloro-N-[4-(3-chloro-4-cyano-phenoxy) cyclohexyl]pyridazine-3-carboxamide (189 mg, 483 μmol, 1.0 equiv), K2CO3 (333 mg, 2.4 mmol, 5.0 equiv) in DMF (1 mL) was degassed and purged with N 2 for 3 times, and then the mixture was stirred at 60 °C for 6 h under N2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure. The residue was purified by prep-HPLC(EC6043-353-P1A1) (column: Phenomenex luna C18150*25mm* 10um;mobile phase: [water(FA)-ACN]; B%: 64%- 94%,10min) to give compound N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]-6-[4-[2-[4-[4-(1, 1- dioxothiazinan-2-yl)-2-(6-methyl-7-oxo-1H-pyrrolo[2,3-c]pyri din-4-yl)phenoxy] phenyl]ethyl]-1- piperidyl]pyridazine-3-carboxamide (70 mg, 98% yield). 1 H NMR: (400 MHz, CDCl 3 ) δ 10.30 (s, 1H), 7.98 (d, J = 9.6 Hz, 1H), 7.90 (d, J = 8.0 Hz, 1H), 7.63 - 7.51 (m, 2H), 7.28 - 7.23 (m, 2H), 7.14 (s, 1H), 7.08 (d, J = 8.4 Hz, 2H), 7.04 - 6.94 (m, 3H), 6.92 - 6.79 (m, 3H), 6.49 (t, J = 2.4 Hz, 1H), 4.51 (d, J = 13.1 Hz, 2H), 4.41 - 4.28 (m, 1H), 4.15 - 4.00 (m, 1H), 3.82 - 3.70 (m, 2H), 3.69 - 3.58 (m, 3H), 3.32 - 3.21 (m, 2H), 3.02 (t, J = 11.8 Hz, 2H), 2.69 - 2.54 (m, 2H), 2.36 (td, J = 5.9, 11.8 Hz, 2H), 2.20 (t, J = 10.5 Hz, 4H), 1.98 - 1.88 (m, 4H), 1.73 - 1.66 (m, 2H), 1.65 - 1.55 (m, 3H), 1.49 (d, J = 12.3 Hz, 2H), 1.33 - 1.24 (m, 2H). LC-MS: MS (ES + ): RT = 2.493 min, m/z = 915.4 [M + H + ]; LCMS Method 25. EXAMPLE 40 – Synthesis of N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]-6-[4-[[3-[4-(1- hydroxy-1-methyl-ethyl)-2-(2-methyl-1-oxo-4-isoquinolyl)phen oxy]cyclobutoxy]methyl]-1- piperidyl]pyridazine-3-carboxamide (I-247)

[0609] Step 1: Preparation of 2-methyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2- yl)isoquinolin-1-one. To a solution of 4-bromo-2-methyl-isoquinolin-1-one (1 g, 4.20 mmol, 1 equiv) in dioxane (15 mL) was added dichloropalladium;triphenylphosphane (295 mg, 420 μmol, 0.1 equiv), 4,4,5,5-tetramethyl-2-(4,4,5,5-tetramethyl-1,3,2-dioxaborola n-2-yl)-1,3,2- dioxaborolane (2.13 g, 8.40 mmol, 2 equiv) and KOAc (1.24 g, 12.6 mmol, 3 equiv) .The mixture was stirred at 110 °C for 12 h. The residue was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate=10/1 to 4/1) to give 2-methyl-4-(4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2-yl)isoquinolin-1-one (1.07 g, 89 % yield) was a white solid. [0610] Step 2: Preparation of tert-butyl 4-[[3-(2-bromo-4-methoxycarbonyl-phenoxy) cyclobutoxy]methyl]piperidine-1-carboxylate. To a solution of tert-butyl 4-[(3- hydroxycyclobutoxy)methyl]piperidine-1-carboxylate (600 mg, 2.10 mmol, 1 equiv) in THF (15 mL) was added PPh3 (1.65 g, 6.30 mmol, 3 equiv), methyl 3-bromo-4-hydroxy-benzoate (485.19 mg, 2.10 mmol, 1 equiv) and DIAD (1.27 g, 6.30 mmol, 1.22 mL, 3 equiv) at 0 °C. The mixture was stirred at 20 °C for 3 h. The residue was purified by prep-HPLC (column: Phenomenex luna C18150*40mm* 15um; mobile phase: [water(FA)-ACN]; B %: 70 %-100 %, 10 min) to give tert- butyl 4-[[3-(2-bromo-4-methoxycarbonyl-phenoxy)cyclobutoxy]methyl] piperidine-1-carboxylate (600 mg, 57 % yield) was a white solid. [0611] Step 3: Preparation of 2-[3-bromo-4-[3-(4-piperidylmethoxy) cyclobutoxy] phenyl]propan-2-ol. To a solution of tert-butyl 4-[[3-(2-bromo-4-methoxycarbonyl- phenoxy)cyclobutoxy]methyl]piperidine-1-carboxylate (500 mg, 1.00 mmol, 1 equiv) in DCM (10 mL) was added MeMgBr (3 M, 5.02 mL, 15 equiv). The mixture was stirred at -78 °C for 1 h. The residue was purified by prep-HPLC (column: Phenomenex luna C18150*40mm* 15um; mobile phase: [water(FA)-ACN]; B %: 8 %-38 %, 15 min) to give 2-[3-bromo-4-[3-(4- piperidylmethoxy)cyclobutoxy]phenyl]propan-2-ol (300 mg, 75% yield) was a white solid. [0612] Step 4: Preparation of 4-[5-(1-hydroxy-1-methyl-ethyl)-2-[3-(4-piperidylmethoxy) cyclobutoxy]phenyl]-2-methyl-isoquinolin-1-one. To a solution of 2-[3-bromo-4-[3-(4- piperidylmethoxy)cyclobutoxy]phenyl]propan-2-ol (270 mg, 678 μmol, 1 equiv) in THF (5 mL) and H 2 O (1 mL) was added SPhos Pd G3 (53 mg, 68 μmol, 0.1 equiv), 2-methyl-4-(4,4,5,5- tetramethyl-1,3,2-dioxaborolan-2-yl)isoquinolin-1-one (251 mg, 881 μmol, 1.3 equiv) and K 3 PO 4 (432 mg, 2.03 mmol, 3 equiv). The mixture was stirred at 60 °C for 3 h. The residue was purified by prep-HPLC (column: Phenomenex luna C18150*25mm* 10um; mobile phase: [water(FA)- ACN]; B %: 15 %-35 %, 9 min) to give 4-[5-(1-hydroxy-1-methyl-ethyl)-2-[3-(4- piperidylmethoxy)cyclobutoxy]phenyl]-2-methyl-isoquinolin-1- one (80 mg, 25 yield) was a white solid. [0613] Step 5: Preparation of N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]-6-[4-[[3-[4-(1- hydroxy-1-methyl-ethyl)-2-(2-methyl-1-oxo-4-isoquinolyl)phen oxy]cyclobutoxy]methyl]-1- piperidyl]pyridazine-3-carboxamide. To a solution of 4-[5-(1-hydroxy-1-methyl-ethyl)-2-[3-(4- piperidylmethoxy)cyclobutoxy]phenyl]-2-methyl-isoquinolin-1- one (80 mg, 168 μmol, 1 equiv) in NMP (1.5 mL) was added K2CO3 (70 mg, 504 μmol, 3 equiv) and 6-chloro-N-[4-(3-chloro-4- cyano-phenoxy)cyclohexyl]pyridazine-3-carboxamide (72 mg, 185 μmol, 1.1 equiv). The mixture was stirred at 50 °C for 2 h. The residue was purified by prep-HPLC (column: Phenomenex luna C18150*25mm* 10um; mobile phase: [water(FA)-ACN]; B %: 65 %-95 %, 10 min) to give N-[4- (3-chloro-4-cyano-phenoxy)cyclohexyl]-6-[4-[[3-[4-(1-hydroxy -1-methyl-ethyl)-2-(2-methyl-1- oxo-4-isoquinolyl)phenoxy]cyclobutoxy]methyl]-1-piperidyl]py ridazine-3-carboxamide (41 mg, 29 % yield) was a yellow solid. 1 H NMR (400 MHz, MeOD) δ 8.36 - 8.42 (m, 1 H), 7.88 (d, J=9.64 Hz, 1 H), 7.67 - 7.70 (m, 1 H), 7.59 - 7.65 (m, 1 H), 7.49 - 7.56 (m, 2 H), 7.41 (d, J=2.32 Hz, 1 H), 7.24 - 7.30 (m, 3 H), 7.18 - 7.21 (m, 1 H), 7.02-7.06 (m, 1 H), 6.84 (d, J=8.68 Hz, 1 H), 4.76-4.81 (m, 1 H), 4.50 (d, J=13.08 Hz, 3 H), 3.87 - 4.02 (m, 2 H), 3.67 (s, 3 H), 3.14 (d, J=5.88 Hz, 2 H), 3.00-3.06 (m, 2 H), 2.24 - 2.32 (m, 2 H), 2.04 - 2.21 (m, 6 H), 1.76 - 1.91 (m, 3 H), 1.61- 1.65 (m, 4 H), 1.55 (s, 6 H), 1.22 - 1.32 (m, 2 H). LC-MS: MS (ES + ): RT = 2.26. min, m/z = 831.4 [M + H + ]; LCMS Method 25. EXAMPLE 41 – Synthesis of N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]-6-[4-[[1-[[4-(1- hydroxy-1-methyl-ethyl)-2-(6-methyl-7-oxo-1H-pyrrolo[2,3-c]p yridin-4-yl)phenyl]methyl] azetidin-3-yl]methyl]-1-piperidyl]pyridazine-3-carboxamide (I-248) [0614] Step 1: Preparation of tert-butyl 3-[[1-(2,2,2-trifluoroacetyl)-4-piperidyl]methyl] azetidine-1-carboxylate. To a solution of tert-butyl 3-(4-piperidylmethyl)azetidine-1-carboxylate (4.5 g, 17.7 mmol, 1.0 equiv) in DCM (100 mL) was added TEA (5.37 g, 53.1 mmol, 7.4 mL, 3.0 equiv) and (2,2,2-trifluoroacetyl) 2,2,2-trifluoroacetate (7.43 g, 35.4 mmol, 4.9 mL, 2.0 equiv) at 0 °C. The mixture was stirred at 25 °C for 12 h. The reaction mixture was diluted with water (80 mL) and extracted with DCM (80 mL * 3). The combined organic layers were washed with brine (100 mL * 3), dried over sodium sulfate, filtered and concentrated under reduced pressure to give a residue. The residue was purified by silica column chromatography on silica gel (Petroleum ether: Ethyl acetate from 50/1 to 3/1) to give tert-butyl 3-[[1-(2,2,2-trifluoroacetyl)-4- piperidyl]methyl]azetidine-1-carboxylate (2.3 g, 37% yield) as a yellow oil. [0615] Step 2: Preparation of 1-[4-(azetidin-3-ylmethyl)-1-piperidyl]-2,2,2-trifluoro- ethanone. To a solution of tert-butyl 3-[[1-(2,2,2-trifluoroacetyl)-4-piperidyl]methyl]azetidine-1 - carboxylate (2.1 g, 6.0 mmol, 1.0 equiv) in DCM (20 mL) was added TFA (4 mL). The mixture was stirred at 25 °C for 0.5 h. The mixture was concentrated to give 1-[4-(azetidin-3-ylmethyl)-1- piperidyl]-2,2,2-trifluoro-ethanone (2.1 g, 96% yield, TFA salt) as a yellow oil. [0616] Step 3: Preparation of methyl 3-bromo-4-[[3-[[1-(2,2,2-trifluoroacetyl)-4- piperidyl]methyl]azetidin-1-yl]methyl]benzoate. To a solution of 1-[4-(azetidin-3-ylmethyl)-1- piperidyl]-2,2,2-trifluoro-ethanone (2.1 g, 5.76 mmol, 1.0 equiv, TFA salt) in DCM (40 mL) was added NaBH(OAc)3 (2.44 g, 11.5 mmol, 2.0 equiv), methyl 3-bromo-4-formyl-benzoate (1.47 g, 6.05 mmol, 1.1 equiv) and TEA (2.92 g, 28.8 mmol, 4 mL, 5.0 equiv). The mixture was stirred at 25 °C for 12 h. The reaction mixture was diluted with water (50 mL) and extracted with DCM (50 mL * 3). The combined organic layers were washed with brine (50 mL * 3), dried over sodium sulfate, filtered and concentrated under reduced pressure to give a residue. The residue was purified by silica column chromatography on silica gel (Petroleum ether: Ethyl acetate from 50/1 to 1/1) to give methyl 3-bromo-4-[[3-[[1-(2,2,2-trifluoroacetyl)-4-piperidyl]methyl ]azetidin-1- yl]methyl]benzoate (2.6 g, 94% yield). [0617] Step 4: Preparation of 2-[3-bromo-4-[[3-(4-piperidylmethyl)azetidin-1- yl]methyl]phenyl]propan-2-ol. To a solution of methyl 3-bromo-4-[[3-[[1-(2,2,2-trifluoroacetyl)- 4-piperidyl]methyl]azetidin-1-yl]methyl]benzoate (1.2 g, 2.51 mmol, 1.0 equiv) in DCM (20 mL) was added MeMgBr (3 M, 12.6 mL, 15.0 equiv) at -78 °C. The mixture was stirred at 25°C for 12 h. The mixture was quenched with water (50 mL) at 0 °C and stirred at 25 °C for 0.5 h. The mixture was concentrated and lyophilized to give a residue. The residue was purified by prep- HPLC (column: Phenomenex luna C18150 * 40 mm * 15um; mobile phase: [water (TFA)-ACN]; B%: 1%-30%, 10 min) to give 2-[3-bromo-4-[[3-(4-piperidylmethyl) azetidin-1- yl]methyl]phenyl]propan-2-ol (1.1 g, crude) as a colorless oil. [0618] Step 5: Preparation of 4-[5-(1-hydroxy-1-methyl-ethyl)-2-[[3-(4-piperidylmethyl) azetidin-1-yl]methyl]phenyl]-6-methyl-1-(ptolylsulfonyl)pyrr olo[2,3-c]pyridin-7-one. To a solution of 6-methyl-1-(p-tolylsulfonyl)-4-(4,4,5,5-tetramethyl-1,3,2-di oxaborolan-2-yl)pyrrolo [2,3-c]pyridin-7-one (472 mg, 1.10 mmol, 1.2 equiv) in THF (9 mL) was added K 3 PO 4 (1.5 M, 1.8 mL, 3.0 equiv), 2-[3-bromo-4-[[3-(4-piperidylmethyl)azetidin-1-yl]methyl]phe nyl]propan-2-ol (350 mg, 918 μmol, 1.0 equiv) and [2-(2-aminophenyl)phenyl]palladium(1+);bis(1-adamantyl)- butyl-phosphane;methanesulfonate (67 mg, 92 μmol, 0.1 equiv). The mixture was stirred at 60 °C for 12 h. The mixture was filtered and concentrated to give a residue. The residue was purified by prep-HPLC (column: Phenomenex luna C18150 * 25 mm * 10 μm; mobile phase: [water (FA)- ACN]; B%: 12%-42%, 10 min) to give 4-[5-(1-hydroxy-1-methyl-ethyl)-2-[[3-(4- piperidylmethyl)azetidin-1-yl]methyl]phenyl]-6-methyl-1-(pto lylsulfonyl)pyrrolo[2,3-c]pyridin-7- one (90 mg, 16% yield) as a yellow oil. [0619] Step 6: Preparation of 4-[5-(1-hydroxy-1-methyl-ethyl)-2-[[3-(4-piperidylmethyl) azetidin-1-yl]methyl]phenyl]-6-methyl-1H-pyrrolo[2,3-c]pyrid in-7-one. To a solution of 4-[5- (1-hydroxy-1-methyl-ethyl)-2-[[3-(4-piperidylmethyl)azetidin -1-yl]methyl]phenyl]-6-methyl-1-(p- tolylsulfonyl)pyrrolo[2,3-c]pyridin-7-one (80 mg, 133 μmol, 1.0 equiv) in MeOH (1 mL) was added KOH (112 mg, 1.99 mmol, 15.0 equiv). The mixture was stirred at 25 °C for 0.5 h. The mixture was adjusted pH to 7 with AcOH at 0 °C. Water (30 mL) was added to the mixture solution. The mixture was lyophilized to give 4-[5-(1-hydroxy-1-methyl-ethyl)-2-[[3-(4- piperidylmethyl)azetidin-1-yl]methyl]phenyl]-6-methyl-1H-pyr rolo[2,3-c]pyridin-7-one (60 mg, crude) as a white solid. [0620] Step 7: Preparation of N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]-6-[4-[[1-[[4-(1- hydroxy-1-methyl-ethyl)-2-(6-methyl-7-oxo-1H-pyrrolo[2,3-c]p yridin-4-yl)phenyl] methyl]azetidin-3-yl]methyl]-1-piperidyl]pyridazine-3-carbox amide. To a solution of 6-chloro- N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]pyridazine-3-carbo xamide (52 mg, 134 μmol, 1.0 equiv) in NMP (1 mL) was added K2CO3 (92 mg, 669 μmol, 5.0 equiv) and 4-[5-(1-hydroxy-1- methyl-ethyl)-2-[[3-(4-piperidylmethyl)azetidin-1-yl]methyl] phenyl]-6-methyl-1H-pyrrolo[2,3- c]pyridin-7-one (60 mg, 134 μmol, 1.0 equiv). The mixture was stirred at 50 °C for 12 h. The mixture was concentrated to give a residue. The residue was purified by prep-HPLC (column: Phenomenex luna C18150 * 25mm * 10 μm; mobile phase: [water (FA)-ACN]; B%: 18%-48%, 10 min) to give N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]-6-[4-[[1-[[4-(1-h ydroxy-1-methyl- ethyl)-2-(6-methyl-7-oxo-1H-pyrrolo[2,3-c]pyridin-4-yl)pheny l]methyl] azetidin-3-yl]methyl]-1- piperidyl]pyridazine-3-carboxamide (37 mg, 32% yield, 96% purity, formate) as a pink solid. 1 H NMR (400 MHz, DMSO-d 6 ): δ 12.07 (s, 1H), 8.56 (d, J = 8.4 Hz, 1H), 8.23-8.10 (m, 1H), 7.85 (d, J = 8.8 Hz, 1H), 7.78 (d, J = 9.6 Hz, 1H), 7.45-7.36 (m, 4H), 7.34-7.21 (m, 3H), 7.13 (m, J = 2.4, 8.8 Hz, 1H), 5.97 (t, J = 2.4 Hz, 1H), 5.15-4.81 (m, 1H), 4.58-4.49 (m, 1H), 4.43 (d, J = 12.4 Hz, 2H), 3.87-3.79 (m, 1H), 3.47 (s, 3H), 3.35 (t, J = 6.8 Hz, 4H), 2.91 (t, J = 12.4 Hz, 2H), 2.69-2.64 (m, 2H), 2.44 (s, 2H), 2.10 (d, J = 10.0 Hz, 2H), 1.89 (d, J = 10.8 Hz, 2H), 1.69-1.57 (m, 4H), 1.55-1.37 (m, 10H), 1.10-0.97 (m, 2H) LC-MS: MS (ES + ): RT = 1.972 min, m/z = 804.4 [M + H + ]; LCMS method: 10. EXAMPLE 42 – Synthesis of compound N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]-6-[4-[2- [4-[4-(1,1-dioxothian-4-yl)-2-(6-methyl-7-oxo-1H-pyrrolo[2,3 -c]pyridin-4-yl)phenoxy] phenyl]ethyl]-1-piperidyl]pyridazine-3-carboxamide (I-250)

[0621] Step 1: Preparation of tert-butyl 4-[2-[4-[4-(3,6-dihydro-2H-thiopyran-4-yl)-2-[6- methyl-7-oxo-1-(p-tolylsulfonyl)pyrrolo[2,3-c]pyridin-4-yl]p henoxy]phenyl] ethyl] piperidine-1-carboxylate. A mixture of tert-butyl 4-[2-[4-[4-bromo-2-[6-methyl-7-oxo-1-(p- tolylsulfonyl)pyrrolo[2,3-c]pyridin-4-yl]phenoxy]phenyl]ethy l]piperidine-1-carboxylate (380 mg, 499 μmol, 1.0 equiv), 2-(3,6-dihydro-2H-thiopyran-4-yl)-4,4,5,5-tetramethyl-1,3,2- dioxaborolane (225 mg, 999 μmol, 2.0 equiv), Pd(dppf)Cl2 (36 mg, 49 μmol, 0.1 equiv), Na2CO3 (105 mg, 999 μmol, 2.0 equiv) in dioxane (5 mL) and H2O (1 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 90 C for 3 h under N 2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate=4/1 to 2/1) to give compound tert- butyl 4-[2-[4-[4-(3,6-dihydro-2H-thiopyran-4-yl)-2-[6-methyl-7-oxo -1-(p- tolylsulfonyl)pyrrolo[2,3-c]pyridin-4-yl]phenoxy]phenyl]ethy l]piperidine-1-carboxylate (260 mg, 66% yield) as a white solid. [0622] Step 2: Preparation of tert-butyl 4-[2-[4-[2-[6-methyl-7-oxo-1-(p-tolylsulfonyl) pyrrolo[2,3-c]pyridin-4-yl]-4-tetrahydrothiopyran-4-yl-pheno xy]phenyl]ethyl]piperidine-1- carboxylate. To a solution of tert-butyl 4-[2-[4-[4-(3,6-dihydro-2H-thiopyran-4-yl)-2-[6-methyl-7- oxo-1-(p-tolylsulfonyl)pyrrolo[2,3-c]pyridin-4-yl]phenoxy]ph enyl]ethyl]piperidine-1-carboxylate (260 mg, 333 μmol, 1.0 equiv) in THF (5 mL) was added Pd/C (130 mg, 10% purity) under N2 atmosphere. The suspension was degassed and purged with H 2 for 3 times. The mixture was stirred under H2 (50 Psi.) at 25 C for 12 h. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was used for next step directly to give compound tert-butyl 4-[2-[4-[2-[6-methyl-7-oxo-1-(p-tolylsulfonyl)pyrrolo[2,3-c] pyridin-4-yl]-4- tetrahydrothiopyran-4-yl-phenoxy]phenyl]ethyl]piperidine-1-c arboxylate (165 mg, 63% yield). [0623] Step 3: Preparation of tert-butyl 4-[2-[4-[4-(1,1-dioxothian-4-yl)-2-[6-methyl-7-oxo- 1-(p-tolylsulfonyl)pyrrolo[2,3-c]pyridin-4-yl]phenoxy]phenyl ]ethyl]piperidine-1-carboxylate. To a solution of tert-butyl 4-[2-[4-[2-[6-methyl-7-oxo-1-(p-tolylsulfonyl) pyrrolo[2,3-c]pyridin-4- yl]-4-tetrahydrothiopyran-4-yl-phenoxy]phenyl]ethyl] piperidine-1-carboxylate (155 mg, 198 μmol, 1.0 equiv) in MeOH (2 mL) and H2O (2 mL) was added Oxone (243 mg, 396 μmol, 2.0 equiv). The mixture was stirred at 25 C for1 h. The mixture was stirred under H2 (50 Psi.) at 25 C for 12 h. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was used for next step directly to give compound tert-butyl 4-[2-[4-[4-(1,1- dioxothian-4-yl)-2-[6-methyl-7-oxo-1-(p-tolylsulfonyl)pyrrol o[2,3-c]pyridin-4- yl]phenoxy]phenyl]ethyl]piperidine-1-carboxylate (140 mg, 86% yield). [0624] Step 4: Preparation of tert-butyl 4-[2-[4-[4-(1,1-dioxothian-4-yl)-2-(6-methyl-7-oxo- 1H-pyrrolo[2,3-c]pyridin-4-yl)phenoxy]phenyl]ethyl]piperidin e-1-carboxylate. To a solution of tert-butyl 4-[2-[4-[4-(1,1-dioxothian-4-yl)-2-[6-methyl-7-oxo-1-(p-toly lsulfonyl) pyrrolo[2,3- c]pyridin-4-yl]phenoxy]phenyl]ethyl]piperidine-1-carboxylate (140 mg, 171 μmol, 1.0 equiv) in MeOH (2 mL) was added KOH (28 mg, 515 μmol, 3.0 equiv). The mixture was stirred at 0.5 or 1 h. The reaction mixture was partitioned between H 2 O 30 mL and EA 100 mL. The organic phase was separated, washed with H2O 30 mL, dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-TLC (SiO 2 , Petroleum ether/Ethyl acetate=1/1) to give compound tert-butyl 4-[2-[4-[4-(1,1-dioxothian-4-yl)-2-(6-methyl- 7-oxo-1H-pyrrolo[2,3-c]pyridin-4-yl)phenoxy]phenyl] ethyl]piperidine-1-carboxylate (10 mg, 8% yield). [0625] Step 5: Preparation of 4-[5-(1, 1-dioxothian-4-yl)-2-[4-[2-(4-piperidyl)ethyl] phenoxy]phenyl]-6-methyl-1H-pyrrolo[2,3-c]pyridin-7-one. To a solution of tert-butyl 4-[2-[4- [4-(1,1-dioxothian-4-yl)-2-(6-methyl-7-oxo-1H-pyrrolo[2,3-c] pyridin-4-yl)phenoxy]phenyl] ethyl]piperidine-1-carboxylate (20 mg, 30 μmol, 1.0 equiv) in DCM (1 mL) was added TFA (462 mg, 4.05 mmol, 0.3 mL, 133 equiv).The mixture was stirred at 25 °C for 0.5 h. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was used for next step directly to give compound 4-[5-(1, 1-dioxothian-4-yl)-2-[4-[2-(4- piperidyl)ethyl]phenoxy]phenyl]-6-methyl-1H-pyrrolo[2,3-c]py ridin-7-one (16 mg, 94% yield). [0626] Step 6: Preparation of compound N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]-6- [4-[2-[4-[4-(1,1-dioxothian-4-yl)-2-(6-methyl-7-oxo-1H-pyrro lo[2,3-c]pyridin-4-yl)phenoxy] phenyl]ethyl]-1-piperidyl]pyridazine-3-carboxamide. To a solution of 4-[5-(1,1-dioxothian-4- yl)-2-[4-[2-(4-piperidyl)ethyl]phenoxy]phenyl]-6-methyl-1H-p yrrolo[2,3-c]pyridin-7-one (16 mg, 28 μmol, 1.0 equiv) and 6-chloro-N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]pyridazin e-3- carboxamide (11 mg, 28 μmol, 1.0 equiv) in NMP (0.5 mL) was added K 2 CO 3 (19 mg, 142 μmol, 5.0 equiv). The mixture was stirred at 50 °C for 12 h. The reaction mixture filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (column: Waters Xbridge 150*25mm* 5um; mobile phase: [water (NH 4 HCO 3 )-ACN]; B%: 57%- 87%, 9min). compound N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]-6-[4-[2-[4-[4-(1, 1- dioxothian-4-yl)-2-(6-methyl-7-oxo-1H-pyrrolo[2,3-c]pyridin- 4-yl)phenoxy]phenyl]ethyl]-1- piperidyl]pyridazine-3-carboxamide (4 mg, 14% yield). 1 H NMR (400 MHz, CDCl 3 ): δ = 9.53 (d, J = 1.6 Hz, 1H), 7.89 (d, J = 9.6 Hz, 1H), 7.80 (d, J = 8.0 Hz, 1H), 7.51 - 7.36 (m, 2H), 7.30 (d, J = 2.4 Hz, 1H), 7.16 (m, 1H), 7.09- 7.06 (m, 1H), 6.98 (m, 3H), 6.94 - 6.88 (m, 3H), 6.79 - 6.76 (m, 1H), 6.74 (d, J = 8.4 Hz, 2H), 6.29 (m, 1H), 4.41 (d, J = 13.2 Hz, 2H), 4.31 - 4.19 (m, 1H), 4.09 - 3.87 (m, 1H), 3.56 (s, 3H), 3.14 - 3.04 (m, 4H), 3.00 - 2.85 (m, 2H), 2.82 - 2.67 (m, 1H), 2.61 - 2.47 (m, 2H), 2.43 - 2.29 (m, 2H), 2.21 (d, J = 13.2 Hz, 2H), 2.16 - 2.07 (m, 4H), 1.84 - 1.74 (m, 2H), 1.48 - 1.31 (m, 4H), 1.29 - 1.05 (m, 4H). LC-MS: MS (ES + ): RT = 2.433 min, m/z = 914.4 [M + H+]; LCMS Method 25. EXAMPLE 43 – Synthesis of N-((1r,4r)-4-(3-chloro-4-cyanophenoxy)cyclohexyl)-6-(4-(4-(( 4- (ethylsulfonyl)-2-(6-methyl-7-oxo-6,7-dihydro-1H-pyrrolo[2,3 -c]pyridin-4-yl)phenyl) difluoromethyl)phenethyl)piperidin-1-yl)pyridazine-3-carboxa mide (I-251)

[0627] Step 1: Preparation of tert-butyl 4-((4-formylphenyl)ethynyl)piperidine-1- carboxylate. A mixture of 4-bromobenzaldehyde (8.62 g, 46.59 mmol, 1.3 equiv), tert-butyl 4- ethynylpiperidine-1-carboxylate (7.5 g, 35.84 mmol, 1.0 equiv), CuI (341 mg, 1.79 mmol, 0.05 equiv), TEA (7.25 g, 71.67 mmol, 9.98 mL, 2.0 equiv) and Pd(PPh 3 ) 2 Cl 2 (1.26 g, 1.79 mmol, 0.05 equiv) in MeCN (40 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 70 °C for 12 h under N2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate = 10/1 to 8/1). Tert-butyl 4-((4- formylphenyl)ethynyl)piperidine-1-carboxylate (10.0 g, 31.91 mmol, 89% yield) was obtained as a yellow oil. [0628] Step 2: Preparation of tert-butyl 4-(4-(hydroxymethyl)phenethyl)piperidine-1- carboxylate. To a solution of tert-butyl 4-((4-formylphenyl)ethynyl)piperidine-1-carboxylate (10.0 g, 31.91 mmol, 1.0 equiv) in MeOH (200 mL) was added Pd/C (2.0 g, 10% purity) under N2 atmosphere. The suspension was degassed and purged with H2 for 3 times. The mixture was stirred under H 2 (15 psi) at 20 °C for 2 h. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO 2 , Petroleum ether/Ethyl acetate = 5/1 to 2/1). Tert-butyl 4-(4-(hydroxymethyl) phenethyl)piperidine- 1-carboxylate (6.0 g, 18.78 mmol, 58% yield) was obtained as a white solid. [0629] Step 3: Preparation of tert-butyl 4-(4-formylphenethyl)piperidine-1-carboxylate. To a solution of tert-butyl 4-(4-(hydroxymethyl)phenethyl)piperidine-1-carboxylate (6.0 g, 18.78 mmol, 1.0 equiv) in DCM (60 mL) was added DMP (11.95 g, 28.17 mmol, 8.72 mL, 1.5 equiv). The mixture was stirred at 20 °C for 1 h. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate = 3/1). Tert-butyl 4-(4-formylphenethyl)piperidine-1-carboxylate (5.9 g, crude) was obtained as a white solid. [0630] Step 4: Preparation of tert-butyl 4-(4-((4-bromo-2- chlorophenyl)(hydroxy)methyl)phenethyl)piperidine-1-carboxyl ate. To a solution of 4-bromo- 2-chloro-1-iodobenzene (12.0 g, 37.8 mmol, 2.0 equiv) in THF (100 mL) was added n-BuLi (2.5 M, 17.29 mL, 2.29 equiv) at -78 °C. After 0.5 h, tert-butyl 4-(4-formylphenethyl) piperidine-1- carboxylate (6.0 g, 18.90 mmol, 94.51 mL, 1.0 equiv) in THF (10 mL) stirred at -78 -30 °C for 11.5 h. The reaction mixture was quenched by addition NH4Cl (100 mL) at 0 °C, and then extracted with EtOAc (2 x 100 mL). The combined organic layers were washed with brine (100 mL), dried over Na 2 SO 4 , filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO 2 , Petroleum ether/Ethyl acetate=10/1 to 5/1). Tert-butyl 4-(4-((4-bromo-2-chlorophenyl)(hydroxy)methyl)phenethyl)pipe ridine-1-carboxylate (8.3 g, 16.31 mmol, 86% yield) was obtained as a yellow gum. [0631] Step 5: Preparation of tert-butyl 4-(4-(4-bromo-2-chlorobenzoyl) phenethyl) piperidine-1-carboxylate. To a solution of tert-butyl 4-(4-((4-bromo-2-chlorophenyl) (hydroxy)methyl)phenethyl)piperidine-1-carboxylate (8.3 g, 16.31 mmol, 1.0 equiv) in DCM (100 mL) was added DMP (10.38 g, 24.47 mmol, 7.57 mL, 1.5 equiv). The mixture was stirred at 20 °C for 1 h. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by silica chromatography (SiO 2 , Petroleum ether/Ethyl acetate = 3/1). Tert-butyl 4-(4-(4-bromo-2-chlorobenzoyl)phenethyl)piperidine-1-carboxy late (8.0 g, 15.78 mmol, 96% yield) was obtained as a white solid. [0632] Step 6: Preparation of crude 4-(4-(2-(4-bromo-2-chlorophenyl)-1,3-dithian-2- yl)phenethyl)piperidine. To a solution of propane-1,3-dithiol (27.0 g, 249.48 mmol, 25 mL, 15.8 equiv) and BF 3 .Et 2 O (22.4 g, 157.83 mmol, 19.48 mL, 10.0 equiv) in DCM (50 mL) was added tert-butyl 4-(4-(4-bromo-2-chlorobenzoyl)phenethyl)piperidine-1-carboxy late (8.0 g, 15.78 mmol, 1.0 equiv) in DCM (20 mL). The mixture was stirred at 30 °C for 72 h. The reaction mixture was partitioned between Na 2 CO 3 (50 mL) and DCM (10 mL). The organic phase was separated, washed with brine (50 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The crude 4-(4-(2-(4-bromo-2-chlorophenyl)-1,3-dithian-2- yl)phenethyl)piperidine (7.8 g, 15.70 mmol) was used into the next step without further purification as a yellow gum. [0633] Step 7: Preparation of tert-butyl 4-(4-(2-(4-bromo-2-chlorophenyl)-1,3-dithian-2- yl)phenethyl)piperidine-1-carboxylate. To a solution of crude 4-(4-(2-(4-bromo-2- chlorophenyl)-1,3-dithian-2-yl)phenethyl)piperidine (7.8 g, 15.70 mmol, 1.0 equiv) in THF (50 mL) was added (Boc) 2 O (6.85 g, 31.39 mmol, 7.21 mL, 2.0 equiv) and Et 3 N (15.88 g, 156.96 mmol, 21.85 mL, 10 equiv).The mixture was stirred at 30 °C for 1 h. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by silica chromatography (SiO 2 , Petroleum ether/Ethyl acetate = 10/1 to 4/1). tert-butyl 4-(4-(2-(4-bromo-2- chlorophenyl)-1,3-dithian-2-yl)phenethyl)piperidine-1-carbox ylate (8.7 g, 14.57 mmol, 92% yield) was obtained as a white gum. [0634] Step 8: Preparation of 4-(4-((4-bromo-2-chlorophenyl)difluoromethyl)phenethyl) piperidine. To a solution of Select F (30.97 g, 87.43 mmol, 6.0 equiv) and pyridine; hydrofluoride (27.5 g, 166.49 mmol, 25 mL, 60% purity, 11.43 equiv) in DCM (10 mL) was added tert-butyl 4- (4-(2-(4-bromo-2-chlorophenyl)-1,3-dithian-2-yl)phenethyl)pi peridine-1-carboxylate (8.7 g, 14.57 mmol, 1.0 equiv) in DCM (10 mL) 0 °C. The mixture was stirred at 30 °C for 12 h. The reaction mixture was poured into NaHCO 3 (500 mL) at 0 °C, and then extracted with EtOAc (2 x 500 mL). The combined organic layers were washed with brine (200 mL), dried over Na 2 SO 4 , filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (column: Phenomenex luna C18150*40mm* 15um; mobile phase: [water (TFA)-ACN]; B%: 35%-65%, 10 min).4-(4-((4-bromo-2-chlorophenyl)difluoromethyl) phenethyl)piperidine (5.0 g, 9.21 mmol, 63% yield, TFA salt) was obtained as a white solid. [0635] Step 9: Preparation of tert-butyl 4-(4-((4-bromo-2-chlorophenyl)difluoromethyl) phenethyl)piperidine-1-carboxylate. To a solution of 4-(4-((4-bromo-2-chlorophenyl) difluoromethyl)phenethyl)piperidine (5.0 g, 9.21 mmol, 1.0 equiv, TFA salt) in THF (5 mL) was added TEA (3.73 g, 36.85 mmol, 5.13 mL, 4.0 equiv) and (Boc) 2 O (4.02 g, 18.42 mmol, 4.23 mL, 2.0 equiv). The mixture was stirred at 20 °C for 12 h. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO 2 , Petroleum ether/Ethyl acetate = 20/1 to 10/1). tert-butyl 4-(4-((4-bromo-2-chlorophenyl) difluoromethyl)phenethyl)piperidine-1-carboxylate (4.5 g, 8.51 mmol, 92% yield) was obtained as a yellow oil. [0636] Step 10: Preparation of tert-butyl 4-(4-((2-chloro-4-(ethylthio)phenyl) difluoromethyl)phenethyl)piperidine-1-carboxylate. A mixture of tert-butyl 4-(4-((4-bromo-2- chlorophenyl)difluoromethyl)phenethyl)piperidine-1-carboxyla te (1.0 g, 1.89 mmol, 1.0 equiv), ethanethiol (4.47 g, 71.94 mmol, 5.32 mL, 38 equiv), Pd(PPh3)4 (437 mg, 378 μmol, 0.2 equiv) and Na 2 CO 3 (601 mg, 5.67 mmol, 3.0 equiv) in dioxane (5 mL) was degassed and purged with N 2 for 3 times, and then the mixture was stirred at 80 °C for 12 h under N 2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate = 20/1 to 10/1). tert-butyl 4-(4-((2-chloro-4-(ethylthio)phenyl)difluoromethyl)phenethyl )piperidine-1-carboxylate (1.0 g, crude) was obtained as a yellow gum. [0637] Step 11: Preparation of tert-butyl 4-(4-((2-chloro-4-(ethylsulfonyl)phenyl) difluoromethyl)phenethyl)piperidine-1-carboxylate. To a solution of tert-butyl 4-(4-((2-chloro- 4-(ethylthio)phenyl)difluoromethyl)phenethyl)piperidine-1-ca rboxylate (500 mg, 980 μmol, 1.0 equiv) in DCM (5 mL) was added m-CPBA (634 mg, 2.94 mmol, 80% purity, 3.0 equiv). The mixture was stirred at 30 °C for 2 h. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (column: Phenomenex luna C18150*40mm* 15um; mobile phase: [water (FA)-ACN]; B%: 65%-95%, 15 min). tert-butyl 4-(4-((2-chloro-4-(ethylsulfonyl)phenyl)difluoromethyl) phenethyl)piperidine-1-carboxylate (270 mg, 498 μmol, 50% yield) was obtained as a white solid. [0638] Step 12: Preparation of tert-butyl 4-(4-((4-(ethylsulfonyl)-2-(6-methyl-7-oxo-1- tosyl-6,7-dihydro-1H-pyrrolo[2,3-c]pyridin-4-yl)phenyl)diflu oromethyl)phenethyl) piperidine-1-carboxylate. A mixture of tert-butyl 4-(4-((2-chloro-4-(ethylsulfonyl)phenyl) difluoromethyl)phenethyl)piperidine-1-carboxylate (270 mg, 498 μmol, 1.0 equiv), 6-methyl-4- (4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1-tosyl-1,6-di hydro-7H-pyrrolo[2,3-c]pyridin-7-one (256 mg, 597 μmol, 1.2 equiv), K 3 PO 4 (1.5 M, 996 μL, 3.0 equiv), Xphos Pd G4 (128 mg, 149 μmol, 0.3 equiv) in n-BuOH (6 mL) and H 2 O (1 mL) was degassed and purged with N 2 for 3 times, and then the mixture was stirred at 60 °C for 16 h under N2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (column: Phenomenex luna C18150*40mm* 15um; mobile phase: [water (FA)- ACN]; B%: 65%-95%, 15 min). tert-butyl 4-(4-((4-(ethylsulfonyl)-2-(6-methyl-7-oxo-1-tosyl-6,7- dihydro-1H-pyrrolo[2,3-c]pyridin-4-yl)phenyl)difluoromethyl) phenethyl )piperidine-1-carboxylate (270 mg, 334 μmol, 67% yield) was obtained as a white solid. [0639] Step 13: Preparation of product tert-butyl 4-(4-((4-(ethylsulfonyl)-2-(6-methyl-7- oxo-6,7-dihydro-1H-pyrrolo[2,3-c]pyridin-4-yl)phenyl)difluor omethyl)phenethyl) piperidine- 1-carboxylate. A mixture of tert-butyl 4-(4-((4-(ethylsulfonyl)-2-(6-methyl-7-oxo-1-tosyl-6,7- dihydro-1H-pyrrolo[2,3-c]pyridin-4-yl)phenyl)difluoromethyl) phenethyl)piperidine-1-carboxylate (270 mg, 334 μmol, 1.0 equiv) and KOH (1.0 M, 5.0 mL, 15 equiv) in i-PrOH (5 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 20 °C for 1 h under N2 atmosphere. The reaction mixture was diluted with H2O (10 mL) and extracted with EtOAc (10 mL). The combined organic layers were washed with brine (10 mL), dried over Na 2 SO 4 , filtered and concentrated under reduced pressure to give crude product tert-butyl 4-(4-((4-(ethylsulfonyl)- 2-(6-methyl-7-oxo-6,7-dihydro-1H-pyrrolo[2,3-c]pyridin-4-yl) phenyl) difluoromethyl)phenethyl)piperidine-1-carboxylate (220 mg, crude) was used into the next step without further. [0640] Step 14: Preparation of 4-(2-(difluoro(4-(2-(piperidin-4-yl)ethyl)phenyl)methyl)-5- (ethylsulfonyl)phenyl)-6-methyl-1,6-dihydro-7H-pyrrolo[2,3-c ]pyridin-7-one. To a solution of product tert-butyl 4-(4-((4-(ethylsulfonyl)-2-(6-methyl-7-oxo-6,7-dihydro-1H-py rrolo[2,3- c]pyridin-4-yl)phenyl)difluoromethyl)phenethyl)piperidine-1- carboxylate (220 mg, 336 μmol, 1.0 equiv) in DCM (2.0 mL) was added TFA (1.0 mL). The mixture was stirred at 20 °C for 1 h. The reaction mixture was concentrated under reduced pressure to give a residue. The crude product product 4-(2-(difluoro(4-(2-(piperidin-4-yl)ethyl)phenyl)methyl)-5-( ethylsulfonyl) phenyl)-6- methyl-1,6-dihydro-7H-pyrrolo[2,3-c]pyridin-7-one (224 mg, TFA salt) was used into the next step without further purification as a yellow gum. [0641] Step 15: Preparation of N-((1r,4r)-4-(3-chloro-4-cyanophenoxy)cyclohexyl)-6-(4-(4- ((4-(ethylsulfonyl)-2-(6-methyl-7-oxo-6,7-dihydro-1H-pyrrolo [2,3-c]pyridin-4- yl)phenyl)difluoromethyl)phenethyl)piperidin-1-yl)pyridazine -3-carboxamide. To a solution of 4-(2-(difluoro(4-(2-(piperidin-4-yl)ethyl)phenyl)methyl)-5-( ethylsulfonyl)phenyl)-6-methyl-1,6- dihydro-7H-pyrrolo[2,3-c]pyridin-7-one (250 mg, 374 μmol, 1.0 equiv, TFA salt) in NMP (2.0 mL) was added K 2 CO 3 (258 mg, 1.87 mmol, 348 μL, 5.0 equiv) and 6-chloro-N-((1r,4r)-4-(3- chloro-4-cyanophenoxy)cyclohexyl)pyridazine-3-carboxamide (146 mg, 374 μmol, 1.0 equiv). The mixture was stirred at 50 °C for 12 h. The reaction mixture was filtered and the filtrate was purified by prep-HPLC (column: Phenomenex luna C18150*25mm* 10um; mobile phase: [water (FA)-ACN]; B%: 66%-93%, 9 min) and (column: Waters xbridge 150*25mm 10um; mobile phase: [water (NH4HCO3)-ACN]; B%: 56%-86%, 10 min). Compound N-((1r,4r)-4-(3-chloro-4- cyanophenoxy)cyclohexyl)-6-(4-(4-((4-(ethylsulfonyl)-2-(6-me thyl-7-oxo-6,7-dihydro-1H- pyrrolo[2,3-c]pyridin-4-yl)phenyl)difluoromethyl)phenethyl)p iperidin-1-yl)pyridazine-3- carboxamide (74 mg, 80 μmol, 21% yield, 98% purity) was obtained as a yellow solid. 1 H NMR (400 MHz, DMSO-d6): ^ δ 11.95 (s, 1H), 8.58 (d, J = 8.3 Hz, 1H), 8.24 - 8.17 (m, 1H), 8.15 - 8.09 (m, 1H), 7.89 - 7.77 (m, 2H), 7.75 (s, 1H), 7.38 (d, J = 1.8 Hz, 1H), 7.36 - 7.30 (m, 1H), 7.17 - 7.09 (m, 2H), 7.09 - 6.94 (m, 4H), 6.44 (s, 1H), 5.62 (s, 1H), 4.61 - 4.40 (m, 3H), 3.94 - 3.76 (m, 1H), 3.41 (q, J = 7.3 Hz, 2H), 3.32 - 3.28 (m, 3H), 2.97 (t, J = 12.5 Hz, 2H), 2.57 - 2.52 (m, 2H), 2.10 (d, J = 10.8 Hz, 2H), 1.90 (d, J = 11.1 Hz, 2H), 1.80 (d, J = 12.3 Hz, 2H), 1.71 - 1.58 (m, 2H), 1.57 - 1.45 (m, 3H), 1.44 - 1.34 (m, 2H), 1.14 (t, J = 7.3 Hz, 5H) LC-MS: MS (ES + ): RT = 2.379 min, m/z = 908.4 [M + H + ]; LCMS Method 25. EXAMPLE 44 – Synthesis of N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]-6-[4-[2-[4-[4-(1, 1- dioxothiolan-2-yl)-2-(6-methyl-7-oxo-1Hpyrrolo[2,3-c]pyridin -4-yl)phenoxy]phenyl]ethyl]-1- piperidyl]pyridazine-3-carboxamide (I-252)

[0642] Step 1: Preparation of 2-chloro-4-(1,1-dioxothiolan-2-yl)phenol. To a solution of thiolane 1,1-dioxide (4.95 g, 41.2 mmol, 3.9 mL, 2.0 equiv) in THF (60 mL) was added n-BuLi (2.5 M, 16.5 mL, 2.0 equiv) at -78°C. The mixture was stirred at -78 °C for 0.5 h. ZnCl 2 (2 M, 20.6 mL, 2.0 equiv) was added at -78 °C. The mixture was stirred at 25 °C for 0.5 h.2-(4-bromo-2- chloro-phenoxy)tetrahydropyran (6 g, 20.6 mmol, 1.0 equiv), Pd(OAc)2 (462 mg, 2.06 mmol, 0.1 equiv) and XPhos (1.96 g, 4.12 mmol, 0.2 equiv) in THF (20 mL) was added and stirred at 65 °C for 11 h. The mixture was quenched with Saturate the ammonium chloride solution (150 mL) and extracted with ethyl acetate (150 mL * 3). The combined organic layers were washed with brine (150 mL * 3), dried over sodium sulfate, filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (column: Phenomenex Luna C18150 * 25mm * 10 μm; mobile phase: [water (FA)-ACN]; B%: 38%-68%, 10 min) to give 2-chloro-4-(1,1- dioxothiolan-2-yl)phenol (2.6 g, 51% yield) as a yellow solid. [0643] Step 2: Preparation of tert-butyl 4-[2-[4-[2-chloro-4-(1,1-dioxothiolan-2- yl)phenoxy]phenyl]ethyl]piperidine-1-carboxylate. To a solution of 2-chloro-4-(1,1- dioxothiolan-2-yl)phenol (1 g, 4.07 mmol, 1.5 equiv) in DMAC (15 mL) was added iodocopper;tetrabutylammonium;diiodide (608 mg, 543 μmol, 0.2 equiv), tert-butyl 4-[2-(4- bromophenyl)ethyl]piperidine-1-carboxylate (1 g, 2.72 mmol, 1.0 equiv), Cs 2 CO 3 (2.65 g, 8.15 mmol, 3.0 equiv) and 2-(dimethylamino)acetic acid (112 mg, 1.09 mmol, 0.4 equiv). The mixture was stirred at 120 °C for 12 h under N2. The mixture was filtered and concentrated to give a residue. The residue was purified by prep-HPLC (column: Waters Xbridge 150 * 25 mm* 5um; mobile phase: [water (NH 4 HCO 3 )-ACN]; B%: 59%-89%, 9 min) tert-butyl 4-[2-[4-[2-chloro-4- (1,1-dioxothiolan-2-yl)phenoxy]phenyl]ethyl]piperidine-1-car boxylate (220 mg, 15% yield) as a yellow solid. [0644] Step 3: Preparation of tert-butyl 4-[2-[4-[4-(1,1-dioxothiolan-2-yl)-2-[6-methyl-7- oxo-1-(p-tolylsulfonyl)pyrrolo[2,3-c]pyridin-4-yl]phenoxy]ph enyl]ethyl] piperidine-1- carboxylate. To a solution of 6-methyl-1-(p-tolylsulfonyl)-4-(4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2-yl)pyrrolo[2,3-c]pyridin-7-one (194 mg, 453 μmol, 1.1 equiv) in n-BuOH (3 mL) was added K3PO4 (2 M, 0.6 mL, 3.0 equiv), tert-butyl 4-[2-[4-[2-chloro-4-(1,1-dioxothiolan-2- yl)phenoxy]phenyl]ethyl]piperidine-1-carboxylate (220 mg, 412 μmol, 1.0 equiv) and dicyclohexyl-[2-(2,4,6-triisopropylphenyl)phenyl] phosphane;methanesulfonate;[2-[2- (methylamino)phenyl]phenyl]palladium(1+) (35 mg, 41 μmol, 0.1 equiv). The mixture was stirred at 60 °C for 12 h. The reaction mixture was diluted with water (15 mL) and extracted with ethyl acetate (20 mL * 3). The combined organic layers were washed with brine (20 mL), dried over sodium sulfate, filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-TLC (Petroleum ether: Ethyl acetate=1/1) to give tert-butyl 4-[2-[4-[4-(1,1- dioxothiolan-2-yl)-2-[6-methyl-7-oxo-1-(p-tolylsulfonyl)pyrr olo[2,3-c]pyridin-4- yl]phenoxy]phenyl]ethyl]piperidine-1-carboxylate (50 mg, 15% yield) as a brown solid. [0645] Step 4: Preparation of tert-butyl 4-[2-[4-[4-(1,1-dioxothiolan-2-yl)-2-(6-methyl-7- oxo-1H-pyrrolo[2,3-c]pyridin-4-yl)phenoxy]phenyl]ethyl]piper idine-1-carboxylate. To a solution of tert-butyl 4-[2-[4-[4-(1,1-dioxothiolan-2-yl)-2-[6-methyl-7-oxo-1-(p-to lylsulfonyl) pyrrolo[2,3-c]pyridin-4-yl]phenoxy]phenyl]ethyl]piperidine-1 -carboxylate (50 mg, 63 μmol, 1.0 equiv) in MeOH (0.5 mL) was added KOH (1 M, 0.9 mL, 15.0 equiv). The mixture was stirred at 25 °C for 1 h. The reaction mixture was diluted with water (10 mL) and extracted with ethyl acetate (15 mL * 3). The combined organic layers were washed with brine (20 mL), dried over sodium sulfate, filtered and concentrated. The residue was purified by prep-TLC (Dichloromethane: Methanol=10/1) to give tert-butyl 4-[2-[4-[4-(1,1-dioxothiolan-2-yl)-2-(6- methyl-7-oxo-1H-pyrrolo[2,3-c]pyridin-4-yl)phenoxy]phenyl]et hyl]piperidine-1-carboxylate (38 mg, 94% yield). [0646] Step 5: Preparation of 4-[5-(1,1-dioxothiolan-2-yl)-2-[4-[2-(4-piperidyl)ethyl] phenoxy]phenyl]-6-methyl-1H-pyrrolo[2,3-c]pyridin-7-one. To a solution of tert-butyl 4-[2-[4- [4-(1,1-dioxothiolan-2-yl)-2-(6-methyl-7-oxo-1H-pyrrolo[2,3- c]pyridin-4- yl)phenoxy]phenyl]ethyl]piperidine-1-carboxylate (38 mg, 59 μmol, 1.0 equiv) in DCM (1 mL) was added TFA (0.2 mL). The mixture was stirred at 25 °C for 0.5 h. The mixture was concentrated to give 4-[5-(1,1-dioxothiolan-2-yl)-2-[4-[2-(4-piperidyl)ethyl]phen oxy]phenyl]-6- methyl-1H-pyrrolo[2,3-c]pyridin-7-one (38 mg, 98% yield, TFA salt) as a yellow oil. [0647] Step 6: Preparation of N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]-6-[4-[2-[4-[4- (1,1-dioxothiolan-2-yl)-2-(6-methyl-7-oxo-1Hpyrrolo[2,3-c]py ridin-4-yl)phenoxy]phenyl] ethyl]-1-piperidyl]pyridazine-3-carboxamide. To a solution of 4-[5-(1,1-dioxothiolan-2-yl)-2- [4-[2-(4-piperidyl)ethyl]phenoxy]phenyl]-6-methyl-1H-pyrrolo [2,3-c]pyridin-7-one (38 mg, 58 μmol, 1.0 equiv, TFA salt) in NMP (1 mL) was added K2CO3 (40 mg, 288 μmol, 5.0 equiv) and 6- chloro-N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]pyridazine- 3-carboxamide (23 mg, 58 μmol, 1.0 equiv). The mixture was stirred at 50 °C for 12 h. The mixture was concentrated to give a residue. The residue was purified by prep-HPLC (column: Waters Xbridge 150 * 25mm * 5um; mobile phase: [water (NH4HCO3)-ACN]; B%: 52%-82%, 10 min) to give N-[4-(3-chloro-4-cyano- phenoxy)cyclohexyl]-6-[4-[2-[4-[4-(1,1-dioxothiolan-2-yl)-2- (6-methyl-7-oxo-1Hpyrrolo[2,3- c]pyridin-4-yl)phenoxy]phenyl]ethyl]-1-piperidyl]pyridazine- 3-carboxamide (5 mg, 9% yield). 1 H NMR (400 MHz, MeOD-d4): δ 7.92-7.79 (m, 3H), 7.69 (d, J = 8.8 Hz, 1H), 7.46-7.41 (m, 2H), 7.28 (d, J = 13.2 Hz, 2H), 7.22-7.18 (m, 3H), 7.11 (d, J = 10.0 Hz, 2H), 6.83 (d, J = 8.8 Hz, 1H), 6.34-6.22 (m, 2H), 5.87-5.77 (m, 1H), 4.65-4.59 (m, 3H), 4.52-4.46 (m, 2H), 4.01-3.92 (m, 1H), 3.68 (s, 3H), 3.39-3.34 (m, 2H), 3.08-2.96 (m, 2H), 2.72-2.65 (m, 2H), 2.60-2.51 (m, 2H), 2.24- 2.17 (m, 2H), 2.13-2.06 (m, 2H), 1.91-1.80 (m, 2H), 1.68-1.59 (m, 5H), 1.59-1.54 (m, 2H), 1.31- 1.19 (m, 3H). LC-MS: MS (ES + ): RT = 2.243 min, m/z = 900.4 [M + H + ]; LCMS Method 25. EXAMPLE 45 – Synthesis of N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]-6-[4-[[4-[2-(6- methyl-7-oxo-1H-pyrrolo[2,3-c]pyridin-4-yl)phenoxy]phenoxy]m ethyl]-1- piperidyl]pyridazine-3-carboxamide (I-255) [0648] Step 1: Preparation of [4-(2-bromophenoxy)phenoxy]-tert-butyl-dimethyl-silane. The solution of 2-bromophenol (13 g, 75.14 mmol, 8.72 mL, 1 eq) and [4-[tert-butyl (dimethyl)silyl]oxyphenyl]boronic acid (22.74 g, 90.17 mmol, 1.2 eq) and Py (29.72 g, 375.71 mmol, 30.32 mL, 5 eq) and Cu(OAc) 2 (13.65 g, 75.14 mmol, 1 eq) and DIEA (48.56 g, 375.71 mmol, 65.44 mL, 5 eq) in DCM (150 mL) was stirred at 25 °C for 12 h under O2. TLC (petroleum ether: ethyl acetate=10:1) showed the reaction was completed. The mixture was filtered and the filtrate was concentrated under reduced pressure to get the residue. The crude product was purified by flash silica gel chromatography (ISCO®; 80 g SepaFlash® Silica Flash Column, Eluent of 0~5 % Ethylacetate/Petroleum ether gradient @ 70mL/min). Compound [4-(2- bromophenoxy)phenoxy]-tert-butyl-dimethyl-silane (23 g, 60.63 mmol, 80.69% yield) was obtained as a white oil. [0649] Step 2: Preparation of 4-(2-bromophenoxy)phenol. To a solution of [4-(2- bromophenoxy)phenoxy]-tert-butyl-dimethyl-silane (12 g, 31.63 mmol, 1 eq) was dissolved in HCl (1 M, 251.78 mL, 7.96 eq) in ethanol (120 mL). The mixture was stirred at 25 °C for 12 h. The reaction mixture was concentrated under reduced pressure to give a residue. The crude product was purified by flash silica gel chromatography (ISCO®; 80 g SepaFlash® Silica FlasColumn, Eluent of 0~5 % Ethylacetate/Petroleum ether gradient @ 70mL/min). Compound 4-(2- bromophenoxy)phenol (6 g, 22.63 mmol, 71.55% yield) was obtained as a white solid. [0650] Step 3: Preparation of tert-butyl 4-[[4-(2-bromophenoxy)phenoxy] methyl]piperidine-1-carboxylate. A mixture of 4-(2-bromophenoxy)phenol (1 g, 3.77 mmol, 1 eq) ,Cs2CO3 (3.69 g, 11.32 mmol, 3 eq) , KI (626.18 mg, 3.77 mmol, 1 eq) and tert-butyl 4- (bromomethyl)piperidine-1-carboxylate (3.15 g, 11.32 mmol, 3 eq) in MeCN (20 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 80 °C for 12 h under N2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The crude product was purified by flash silica gel chromatography (ISCO®; 12 g SepaFlash® Silica Flash Column, Eluent of 0~5 % Ethylacetate/Petroleum ether gradient @ 70mL/min). Compound tert-butyl 4-[[4-(2-bromophenoxy)phenoxy]methyl] piperidine-1- carboxylate (1.5 g, 3.24 mmol, 86.00% yield) was obtained as a white solid. [0651] Step 4: Preparation of tert-butyl4-[[4-[2-[6-methyl-7-oxo-1-(p-tolylsulfonyl) pyrrolo[2,3-c]pyridin-4-yl]phenoxy]phenoxy]methyl]piperidine -1-carboxylate. To a mixture of tert-butyl 4-[[4-(2-bromophenoxy)phenoxy]methyl]piperidine-1-carboxylat e (200 mg, 432.55μmol,1 eq) ,6-methyl-1-(p-tolylsulfonyl)-4-(4,4,5,5-tetramethyl-1,3,2-d ioxaborolan-2- yl)pyrrolo[2,3-c]pyridin-7-one (222.32 mg, 519.06 μmol, 1.2 eq) and SPhos Pd G3 (67.50 mg, 86.51 μmol, 0.2 eq) in THF (5 mL) was dropwise added a solution of K 3 PO 4 (275.45 mg, 1.30 mmol, 3 eq) in H 2 O (1 mL) under N 2 The reaction mixture was stirred at 60 °C for 12 h under N 2 . To the reaction mixture was added water (20 mL) and the mixture was extracted with ethyl acetate (30 mL x 3). The combined organic phase was washed with brine (20 mL), dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo. The crude product was purified by flash silica gel chromatography (ISCO®; 20 g SepaFlash® Silica Flash Column, Eluent of 0~60 % Ethylacetate/Petroleum ether gradient @70mL/min).Compound tert-butyl4-[[4-[2-[6-methyl-7- oxo-1-(p-tolylsulfonyl)pyrrolo[2,3-c]pyridin-4-yl]phenoxy]ph enoxy]methyl]piperidine-1- carboxylate (280 mg, 409.47 μmol, 94.66% yield) was obtained as a light yellow solid. [0652] Step 5: Preparation of tert-butyl 4-[[4-[2-(6-methyl-7-oxo-1H-pyrrolo[2,3- c]pyridin-4-yl)phenoxy]phenoxy]methyl]piperidine-1-carboxyla te. To a solution of tert-butyl 4-[[4-[2-[6-methyl-7-oxo-1-(p-tolylsulfonyl)pyrrolo[2,3-c]py ridin-4-yl]phenoxy]phenoxy] methyl]piperidine-1-carboxylate (280 mg, 409.47 μmol, 1 eq) in MeOH (10 mL) was added KOH (2 M, 2.66 mL, 13 eq). The mixture was stirred at 25 °C for 0.5 h. To the reaction mixture was added water (20 mL) and the mixture was extracted with ethyl acetate (30 mL x 3). The combined organic phase was washed with brine (20 mL), dried over anhydrous Na 2 SO 4 , filtered and concentrated in vacuo. The residue was purified by prep-TLC (SiO 2 , DCM: MeOH = 10:1).Compound tert-butyl 4-[[4-[2-(6-methyl-7-oxo-1H-pyrrolo[2,3-c]pyridin-4- yl)phenoxy]phenoxy]methyl]piperidine-1-carboxylate (215 mg, 405.95 μmol, 99.14% yield) was obtained as a light yellow solid. [0653] Step 6: Preparation of 6-methyl-4-[2-[4-(4-piperidylmethoxy)phenoxy]phenyl]-1H- pyrrolo[2,3-c]pyridin-7-one. To a solution of tert-butyl 4-[[4-[2-(6-methyl-7-oxo-1H-pyrrolo[2,3- c]pyridin-4-yl)phenoxy]phenoxy]methyl]piperidine-1-carboxyla te (100 mg, 188.81 μmol, 1 eq) in DCM (10 mL) was added TFA (21.53 mg, 188.81 μmol, 14.03 μL, 1 eq). The mixture was stirred at 25 °C for 0.5 h. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was taken to the next step without purification. Compound 6-methyl-4-[2-[4-(4- piperidylmethoxy)phenoxy]phenyl]-1H-pyrrolo[2,3-c]pyridin-7- one (80 mg, 186.26 μmol, 98.65% yield) was obtained as a yellow solid. [0654] Step 7: Preparation of N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]-6-[4-[[4-[2-(6- methyl-7-oxo-1H-pyrrolo[2,3-c]pyridin-4-yl)phenoxy]phenoxy]m ethyl]-1-piperidyl] pyridazine-3-carboxamide. A mixture of 6-methyl-4-[2-[4-(4-piperidylmethoxy)phenoxy] phenyl]-1H-pyrrolo[2,3-c]pyridin-7-one (80 mg, 186.26 μmol, 1 eq) , DIEA (481.45 mg, 3.73 mmol, 648.86 μL, 20 eq) and 6-chloro-N-[4-(3-chloro-4-cyano-phenoxy)cyclohexyl]pyridazin e-3- carboxamide (85 mg, 217.25 μmol, 1.17 eq) in DMF (10 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 50 °C for 12h under N 2 atmosphere. To the reaction mixture was added water (20 mL) and the mixture was extracted with ethyl acetate (30 mL x 3). The combined organic phase was washed with brine (20 mL), dried over anhydrous Na2SO4, filtered and concentrated in vacuo.The crude product was purified by prep-HPLC (column: YMC- Actus Triart C18150*30mm*7um; mobile phase: [water(FA)-ACN]; gradient: 68%-98% B over 10 min). Compound N-[4-(3-chloro-4-cyano-phenoxy) cyclohexyl]-6-[4-[[4-[2-(6-methyl-7-oxo- 1H-pyrrolo[2,3-c]pyridin-4-yl)phenoxy] phenoxy]methyl]-1-piperidyl]pyridazine-3-carboxamide (38.77 mg, 49.43 μmol, 26.54% yield) was obtained as a yellow solid. 1 H NMR (400 MHz, MeOD-d4): δ 12.07 - 11.92 (m, 1H), 8.60 (d, J = 8.1 Hz, 1H), 7.84 (dd, J = 9.1, 19.5 Hz, 2H), 7.46 (dd, J = 1.7, 7.6 Hz, 1H), 7.41 - 7.31 (m, 3H), 7.28 - 7.24 (m, 2H), 7.20 - 7.12 (m, 2H), 6.91 - 6.83 (m, 5H), 6.23 (t, J = 2.3 Hz, 1H), 4.59 - 4.49 (m, 3H), 3.82 (br d, J = 6.1 Hz, 2H), 3.54 (s, 3H), 3.11 - 3.00 (m, 2H), 2.70 - 2.66 (m, 1H), 2.36 - 2.30 (m, 1H), 2.11 (br d, J = 10.4 Hz, 3H), 1.93 - 1.84 (m, 4H), 1.68 - 1.49 (m, 4H), 1.33 - 1.28 (m, 1H). LC-MS: MS (ES + ): RT = 0.607 min, m/z = 784.3 [M + H + ]; LCMS Method: 5-95. EXAMPLE 46 – Synthesis and Characterization of Additional Compounds [0655] The following additional compounds were were prepared based on procedures described herein: I-38, I-39, I-41, I-42, I-43, I-44, I-46, I-47, I-48, I-49, I-50, I-51, I-52, I-53, I-54, I-55, I-56, I-57, I-58, I-59, I-60, I-61, I-62, I-63, I-64, I-65, I-66, I-67, I-68, I-69, I-70, I-71, I-72, I-73, I-74, I- 75, I-76, I-77, I-78, I-79, I-80, I-81, I-82, I-83, I-84, I-85, I-86, I-87, I-88, I-89, I-91, I-93, I-94, I- 96, I-97, I-98, I-99, I-101, I-102, I-103, I-104, I-105, I-106, I-107, I-108, I-109, I-110, I-111, I-112, I-113, I-114, I-115, I-116, I-117, I-118, I-119, I-120, I-121, I-122, I-123, I-124, I-125, I-126, I-127, I-128, I-129, I-130, I-131, I-132, I-133, I-134, I-135, I-136, I-138, I-139, I-140, I-141, I-142, I-143, I-144, I-145, I-146, I-147, I-148, I-149, I-150, I-152, I-153, I-155, I-156, I-157, I-159, I-160, I-161, I-162, I-163, I-164, I-165, I-166, I-167, I-168, I-169, I-170, I-171, I-172, I-173, I-174, I-175, I-176, I-177, I-178, I-179, I-180, I-181, I-182, I-183, I-184, I-185, I-186, I-187, I-188, I-189, I-190, I-191, I-192, I-193, I-194, I-195, I-196, I-197, I-198, I-199, I-200, I-201, I-202, I-203, I-204, I-206, I-207, I-208, I-209, I-210, I-211, I-213, I-214, I-215, I-216, I-217, I-219, I-220, I-221, I-222, I-223, I-224, I-225, I-226, I-227, I-228, I-229, I-230, I-232, I-233, I-249 and I-254. [0656] Exemplary compounds were characterized by LCMS. Characterization results are provided below in Table 2. TABLE 2.

EXAMPLE 47 – Assay for Binding Affinity to Androgen Receptor [0657] Exemplary compounds were tested for ability to bind to the androgen receptor. Experimental procedures and results are provided below. Part I – Experimental Procedure [0658] Fractions of cell cytosol (106 cell/point) were incubated for 24 hr at 4°C with 1 nM [ 3 H]methyltrienolone in the absence or presence of the test compound in a buffer containing 25 mM Hepes-Tris (pH 7.4), 1 mM EDTA, 10 mM Na2MoO4, 2 mM DTT, 5 μM triamcinolone acetonide, and 10% glycerol. Nonspecific binding was determined in the presence of 1 μM testosterone. Following incubation, the samples were filtered rapidly under vacuum through glass fiber filters (GF/B, Packard) presoaked with 0.3% PEI and rinsed several times with ice-cold 50 mM Tris-HCl using a 96-sample cell harvester (Unifilter, Packard). The filters were dried then counted for radioactivity in a scintillation counter (Topcount, Packard) using a scintillation cocktail (Microscint 0, Packard). The results are expressed as a percent inhibition of the control radioligand specific binding. The standard reference compound is testosterone, which is tested in each experiment at several concentrations to obtain a competition curve from which its IC50 is calculated. Part II – Results [0659] Results showing ability of exemplary compounds to bind to the androgen receptor are provided in Table 3 below. The symbol “++++” indicates a Kd less than 0.05 ^M. The symbol “+++” indicates an Kd in the range of 0.05 ^M to 0.5 ^M. The symbol “++” indicates a Kd in the range of greater than 0.5 ^M to 2.5 ^M. The symbol “+” indicates a Kd greater than 2.5 ^M. TABLE 3. EXAMPLE 48 – Assay for Binding Affinity to BRD4-BD1 [0660] Exemplary compounds were tested for ability to bind to BRD4-BD1. Experimental procedures and results are provided below. Part I – Experimental Procedure [0661] Compounds were tested using a bromoKdELECT assay. T7 phage strains displaying bromodomains were grown in parallel in 24-well blocks in an E. coli host derived from the BL21 strain. E. coli were grown to log-phase and infected with T7 phage from a frozen stock (multiplicity of infection = 0.4) and incubated with shaking at 32°C until lysis (90-150 minutes). The lysates were centrifuged (5,000 x g) and filtered (0.2μm) to remove cell debris. Streptavidin- coated magnetic beads were treated with biotinylated small molecule or acetylated peptide ligands for 30 minutes at room temperature to generate affinity resins for bromodomain assays. The liganded beads were blocked with excess biotin and washed with blocking buffer (SeaBlock (Pierce), 1% BSA, 0.05% Tween 20, 1 mM DTT) to remove unbound ligand and to reduce nonspecific phage binding. Binding reactions were assembled by combining bromodomains, liganded affinity beads, and test compounds in 1x binding buffer (17% SeaBlock, 0.33x PBS, 0.04% Tween 20, 0.02% BSA, 0.004% Sodium azide, 7.4 mM DTT). Test compounds were prepared as 1000X stocks in 100% DMSO. Kds were determined using an 11-point 3-fold compound dilution series with one DMSO control point. All compounds for Kd measurements are distributed by acoustic transfer (non-contact dispensing) in 100% DMSO. The compounds were then diluted directly into the assays such that the final concentration of DMSO was 0.09%. All reactions performed in polypropylene 384-well plates. Each was a final volume of 0.02 ml. The assay plates were incubated at room temperature with shaking for 1 hour and the affinity beads were washed with wash buffer (1x PBS, 0.05% Tween 20). The beads were then resuspended in elution buffer (1x PBS, 0.05% Tween 20, 2 μM non-biotinylated affinity ligand) and incubated at room temperature with shaking for 30 minutes. The bromodomain concentration in the eluates was measured by qPCR. Part II – Results [0662] Results showing ability of exemplary compounds to bind to BRD4-BD1 are provided in Table 4 below. The symbol “++++” indicates a Kd less than 0.05 ^M. The symbol “+++” indicates a Kd in the range of 0.05 ^M to 0.5 ^M. The symbol “++” indicates a Kd in the range of greater than 0.5 ^M to 2.5 ^M. The symbol “+” indicates a Kd greater than 2.5 ^M. TABLE 4.

EXAMPLE 49 – Assay for Binding Affinity to BRD4-BD2 [0663] Exemplary compounds were tested for ability to bind to BRD4-BD2. Experimental procedures and results are provided below. Part I – Experimental Procedure [0664] Compounds were tested using a bromoKdELECT assay. T7 phage strains displaying bromodomains were grown in parallel in 24-well blocks in an E. coli host derived from the BL21 strain. E. coli were grown to log-phase and infected with T7 phage from a frozen stock (multiplicity of infection = 0.4) and incubated with shaking at 32°C until lysis (90-150 minutes). The lysates were centrifuged (5,000 x g) and filtered (0.2μm) to remove cell debris. Streptavidin- coated magnetic beads were treated with biotinylated small molecule or acetylated peptide ligands for 30 minutes at room temperature to generate affinity resins for bromodomain assays. The liganded beads were blocked with excess biotin and washed with blocking buffer (SeaBlock (Pierce), 1% BSA, 0.05% Tween 20, 1 mM DTT) to remove unbound ligand and to reduce nonspecific phage binding. Binding reactions were assembled by combining bromodomains, liganded affinity beads, and test compounds in 1x binding buffer (17% SeaBlock, 0.33x PBS, 0.04% Tween 20, 0.02% BSA, 0.004% Sodium azide, 7.4 mM DTT). Test compounds were prepared as 1000X stocks in 100% DMSO. Kds were determined using an 11-point 3-fold compound dilution series with one DMSO control point. All compounds for Kd measurements are distributed by acoustic transfer (non-contact dispensing) in 100% DMSO. The compounds were then diluted directly into the assays such that the final concentration of DMSO was 0.09%. All reactions performed in polypropylene 384-well plates. Each was a final volume of 0.02 ml. The assay plates were incubated at room temperature with shaking for 1 hour and the affinity beads were washed with wash buffer (1x PBS, 0.05% Tween 20). The beads were then resuspended in elution buffer (1x PBS, 0.05% Tween 20, 2 μM non-biotinylated affinity ligand) and incubated at room temperature with shaking for 30 minutes. The bromodomain concentration in the eluates was measured by qPCR. Part II – Results [0665] Results showing ability of exemplary compounds to bind to BRD4-BD2 are provided in Table 5 below. The symbol “++++” indicates a Kd less than 0.05 ^M. The symbol “+++” indicates a Kd in the range of 0.05 ^M to 0.5 ^M. The symbol “++” indicates a Kd in the range of greater than 0.5 ^M to 2.5 ^M. The symbol “+” indicates a Kd greater than 2.5 ^M. TABLE 5.

EXAMPLE 50 – Cellular Growth Inhibition Assay Using T-Rex 293 Cells [0666] Exemplary compounds were tested for ability to inhibit the proliferation of the following types of cells: (i) a T-Rex 293 cell line having increased expression of androgen receptor protein due to exposure to doxycycline and (ii) a T-Rex 293 cell line lacking increased expression of androgen receptor protein. Experimental procedures and results are provided below. Part I – Experimental Procedure [0667] The following types of cells were prepared for this experiment: (i) a T-Rex 293 cell line having increased expression of androgen receptor protein due to exposure to doxycycline and (ii) a T-Rex 293 cell line lacking increased expression of androgen receptor protein. Ability of the test compounds to inhibit proliferation of the foregoing cell types was evaluated according to the procedures set forth below. [0668] The doxycycline-inducible androgen receptor protein expressing cell line was established using the following protocol: T-Rex 293 cells were purchased from Invitrogen (Cat#R71007) and transfected using Lipofectamine 2000 with the wild-type androgen receptor protein sequence cloned into the pcDNA4/TO vector. Transfected cells were selected using 400 µg/mL Zeocin (Invitrogen Cat#R25001). Following selection, single clones were raised and maintained in Dulbecco's Modified Eagle Medium (DMEM) supplemented with 10% w/w Tetracycline-free fetal bovine serum (FBS) and 250 µg/mL Zeocin. Clones were analyzed for expression of androgen receptor protein in the presence and absence of 10 ng/mL doxycycline (Sigma Cat#D9891), and a single doxycycline-inducible clone (hereinafter “SC3”) was selected for use in downstream assays. [0669] The SC3 cells were seeded on poly-D-lysine coated, black clear-bottom 384-well plates at 2500/well, in 25 ^L Phenol Red Free Dulbecco's Modified Eagle Medium (DMEM) supplemented with 10% w/w charcoal-dextran treated fetal bovine serum (FBS) and 1% w/w pen- strep, with or without 10 ng/mL doxycycline. Pen-Strep is a commercially available mixture of penicillin G and streptomycin, which is used in mammalian cell culture media to prevent bacterial contamination. Phenol Red Free Dulbecco's Modified Eagle Medium (DMEM) supplemented with 10% w/w charcoal-dextran treated fetal bovine serum (FBS) and 1% w/w pen-strep, with or without 10 ng/mL doxycycline is herein referred to as Treatment Medium. Following seeding of cells in the plates, the plates were spun at 300 × g^for 30 seconds, then equilibrated to room temperature for 30 minutes, and then deposited^in a humidified tissue culture incubator maintained at 37°C with 5% CO2. [0670] At 24 hours after seeding of the cells, dilutions of test compound were prepared in DMSO and dissolved in Treatment Medium, to achieve a final DMSO concentration of 0.5% w/w, thereby providing the Test Compound Solution. A 25 ^L aliquot of the Test Compound Solution was added to cells in the well plates. An equal volume of a solution containing DMSO and Treatment Medium was used as a negative control. Following treatment of cells with Test Compound Solution or said equal volume of a solution containing DMSO and Treatment Medium, the plates were spun at 300 × g for 30 seconds, and then left in an incubator for 72 hours. [0671] At the end of the treatment duration, cell viability was quantified with CellTiter-Glo 2.0 reagent (Promega Cat#G9243). For this purpose, plates were equilibrated to room temperature for 30 minutes, and then 25 µL of CellTiter-Glo 2.0 reagent was added to cells in the plate wells. Plates were then agitated on a shaker for two minutes at 500 rpm and subsequently incubated at room temperature for 10 minutes. Following incubation, the plates were spun at 3000 x g for 30 seconds, then sealed with an optical adhesive cover, and luminescence readings were measured with an EnVision Plate Reader. [0672] Data was normalized using zero luminescence for baseline. A four-parameter non-linear regression curve fit was applied to dose-response data in GraphPad Prism data analysis software to determine the half-maximal growth inhibitory concentration (GI50) for each test compound. Part II – Results [0673] The half-maximal growth inhibitory concentration (GI50) results are provided in Tables 6 and 7 below for exemplary compounds. Table 6 provides results from the experiment analyzing ability of test compounds to inhibit proliferation of the T-Rex 293 cell line SC3 cells having increased expression of androgen receptor protein due to exposure to doxycycline. The symbol “++++” indicates a GI50 less than 0.5 ^M. The symbol “+++” indicates an GI50 in the range of 0.5 ^M to 1.5 ^M. The symbol “++” indicates a GI50 in the range of greater than 1.5 ^M to 3 ^M. The symbol “+” indicates a GI50 greater than 3 ^M. TABLE 6.

[0674] Table 7 provides results from the experiment analyzing ability of test compounds to inhibit proliferation of the T-Rex 293 cell line SC3 cells lacking increased expression of androgen receptor protein since such cells were not exposed to doxycycline. The symbol “++++” indicates a GI50 less than 0.5 ^M. The symbol “+++” indicates an GI50 in the range of 0.5 ^M to 1.5 ^M. The symbol “++” indicates a GI50 in the range of greater than 1.5 ^M to 3 ^M. The symbol “+” indicates a GI50 greater than 3 ^M. TABLE 7.

EXAMPLE 51 – Cellular Growth Inhibition Assay for VCaP Cells [0675] Exemplary compounds were tested for ability to inhibit the proliferation of VCaP cells. VCap cells are a commercially available human prostate cancer cell line. Experimental procedures and results are provided below. Part I – Experimental Procedure [0676] VCaP cells were purchased from American Type Cell Culture (ATCC Cat#CRL2876) and then seeded on poly-D-lysine coated, black clear-bottom 384-well plates at 5000/well in 25 ^L Dulbecco's Modified Eagle Medium (DMEM) supplemented with 10% w/w Fetal bovine serum (FBS) and 1% w/w Pen-Strep. Pen-Strep is a commercially available mixture of penicillin G and streptomycin, which is used in mammalian cell culture media to prevent bacterial contamination. Dulbecco's Modified Eagle Medium (DMEM) supplemented with 10% w/w fetal bovine serum (FBS) and 1% w/w Pen-Strep is herein referred to as Treatment Medium. Following seeding of cells in the plates, the plates were spun at 300 × g^for 30 seconds, then equilibrated to room temperature for 30 minutes, and then deposited^in a humidified tissue culture incubator maintained at 37°C with 5% CO2. [0677] At 24 hours after seeding of the cells, dilutions of test compound were prepared in DMSO and dissolved in Treatment Medium, to achieve a final DMSO concentration of 0.5% w/w, thereby providing the Test Compound Solution. A 25 ^L aliquot of the Test Compound Solution was added to cells in the well plates. An equal volume of a solution containing DMSO and Treatment Medium was used as a negative control. Following treatment of cells with Test Compound Solution or said equal volume of a solution containing DMSO and Treatment Medium, the plates were spun at 300 × g for 30 seconds, and then left in an incubator for 72 hours. [0678] At the end of the treatment duration, cell viability was quantified with CellTiter-Glo 2.0 reagent (Promega Cat#G9243). For this purpose, plates were equilibrated to room temperature for 30 minutes, and then 25 µL of CellTiter-Glo 2.0 reagent was added to cells in the plate wells. Plates were then agitated on a shaker for two minutes at 500 rpm and subsequently incubated at room temperature for 10 minutes. Following incubation, the plates were spun at 3000 x g for 30 seconds, then sealed with an optical adhesive cover, and luminescence readings were measured with an EnVision Plate Reader. [0679] Data was normalized using zero luminescence for baseline. A four-parameter non-linear regression curve fit was applied to dose-response data in GraphPad Prism data analysis software to determine the half-maximal growth inhibitory concentration (GI 50 ) for each test compound. Part II – Results [0680] The half-maximal growth inhibitory concentration (GI50) results are provided in Table 8 below for exemplary compounds. The symbol “++++” indicates a GI50 less than 0.5 ^M. The symbol “+++” indicates an GI50 in the range of 0.5 ^M to 1.5 ^M. The symbol “++” indicates a GI50 in the range of greater than 1.5 ^M to 3 ^M. The symbol “+” indicates a GI50 greater than 3 ^M. TABLE 8.

EXAMPLE 52– Cellular Growth Inhibition Assay Using T-Rex 293 Cells [0681] Exemplary compounds were tested for ability to inhibit the proliferation of the following types of cells: (i) a T-Rex 293 cell line having increased expression of progesterone receptor isoform B protein due to exposure of doxycycline and (ii) a T-Rex 293 cell line lacking increased expression of progesterone receptor isoform B protein. Experimental procedures and results are provided below. Part I – Experimental Procedure [0682] The following types of cells were prepared for this experiment: (i) a T-Rex 293 cell line having increased expression of progesterone receptor isoform B protein due to exposure to doxycycline and (ii) a T-Rex 293 cell line lacking increased expression of progesterone receptor isoform B protein. Ability of the test compounds to inhibit proliferation of the foregoing cell types was evaluated according to the procedures set forth below. [0683] The doxycycline-inducible progesterone receptor isoform B protein expressing cell line was established using the following protocol: T-Rex 293 cells were purchased from Invitrogen (Cat#R71007) and transfected using Lipofectamine 2000 with the wild-type progesterone receptor B protein sequence cloned into the pcDNA4/TO vector. Transfected cells were selected using 400 µg/mL Zeocin (Invitrogen Cat#R25001). Following selection, single clones were raised and maintained in Dulbecco's Modified Eagle Medium (DMEM) supplemented with 10% w/w Tetracycline-free fetal bovine serum (FBS) and 250 µg/mL Zeocin. Clones were analyzed for expression of progesterone receptor isoform B protein in the presence and absence of 10 ng/mL doxycycline (Sigma Cat#D9891), and a single doxycycline-inducible clone (hereinafter “SC”) was selected for use in downstream assays. [0684] The SC cells were seeded on poly-D-lysine coated, black clear-bottom 384-well plates at 2500/well, in 25 ^L Phenol Red Free Dulbecco's Modified Eagle Medium (DMEM) supplemented with 10% w/w charcoal-dextran treated fetal bovine serum (FBS) and 1% w/w pen- strep, with or without 10 ng/mL doxycycline. Pen-Strep is a commercially available mixture of penicillin G and streptomycin, which is used in mammalian cell culture media to prevent bacterial contamination. Phenol Red Free Dulbecco's Modified Eagle Medium (DMEM) supplemented with 10% w/w charcoal-dextran treated fetal bovine serum (FBS) and 1% w/w pen-strep, with or without 10 ng/mL doxycycline is herein referred to as Treatment Medium. Following seeding of cells in the plates, the plates were spun at 300 × g^for 30 seconds, then equilibrated to room temperature for 30 minutes, and then deposited^in a humidified tissue culture incubator maintained at 37 °C with 5% CO 2 . [0685] At 24 hours after seeding of the cells, dilutions of test compound were prepared in DMSO and dissolved in Treatment Medium, to achieve a final DMSO concentration of 0.5% w/w, thereby providing the Test Compound Solution. A 25 ^L aliquot of the Test Compound Solution was added to cells in the well plates. An equal volume of a solution containing DMSO and Treatment Medium was used as a negative control. Following treatment of cells with Test Compound Solution or said equal volume of a solution containing DMSO and Treatment Medium, the plates were spun at 300 × g for 30 seconds, and then left in an incubator for 72 hours. [0686] At the end of the treatment duration, cell viability was quantified with CellTiter-Glo 2.0 reagent (Promega Cat#G9243). For this purpose, plates were equilibrated to room temperature for 30 minutes, and then 25 µL of CellTiter-Glo 2.0 reagent was added to cells in the plate wells. Plates were then agitated on a shaker for two minutes at 500 rpm and subsequently incubated at room temperature for 10 minutes. Following incubation, the plates were spun at 3000 x g for 30 seconds, then sealed with an optical adhesive cover, and luminescence readings were measured with an EnVision Plate Reader (Perkin Elmer). [0687] Data was normalized using zero luminescence for baseline. A four-parameter non-linear regression curve fit was applied to dose-response data in GraphPad Prism data analysis software to determine the half-maximal growth inhibitory concentration (GI50) for each test compound. Part II – Results [0688] The half-maximal growth inhibitory concentration (GI 50 ) results are provided in Tables 9 and 10 below for exemplary compounds. Table 9 provides results from the experiment analyzing ability of test compounds to inhibit proliferation of the T-Rex 293 cell line SC cells having increased expression of progesterone receptor isoform B protein due to exposure to doxycycline. The symbol “++++” indicates a GI50 less than 0.5 ^M. The symbol “+++” indicates an GI50 in the range of 0.5 ^M to 1.5 ^M. The symbol “++” indicates a GI50 in the range of greater than 1.5 ^M to 3 ^M. The symbol “+” indicates a GI50 greater than 3 ^M. TABLE 9. [0689] [0690] Table 10 provides results from the experiment analyzing ability of test compounds to inhibit proliferation of the T-Rex 293 cell line SC cells lacking increased expression of progesterone receptor isoform B protein since such cells were not exposed to doxycycline. The symbol “++++” indicates a GI50 less than 0.5 ^M. The symbol “+++” indicates an GI50 in the range of 0.5 ^M to 1.5 ^M. The symbol “++” indicates a GI50 in the range of greater than 1.5 ^M to 3 ^M. The symbol “+” indicates a GI50 greater than 3 ^M. TABLE 10.

EXAMPLE 53 – Cellular Growth Inhibition Assay for T47D Cells [0691] Exemplary compounds were tested for ability to inhibit the proliferation of T47D cells. T47D cells are a commercially available human breast cancer cell line. Experimental procedures and results are provided below. Part I – Experimental Procedure [0692] T47D cells were purchased from American Type Cell Culture (ATCC Cat# HTB-133) and then seeded on poly-D-lysine coated, black clear-bottom 384-well plates at 3000/well in 25 ^L Dulbecco's Modified Eagle Medium (DMEM) supplemented with 10% w/w Fetal bovine serum (FBS), 0.2 units/mL recombinant human insulin (Gibco Cat# 12585014), and 1% w/w Pen-Strep. Pen-Strep is a commercially available mixture of penicillin G and streptomycin, which is used in mammalian cell culture media to prevent bacterial contamination. Dulbecco's Modified Eagle Medium (DMEM) supplemented with 10% w/w fetal bovine serum (FBS), 0.2 units/mL recombinant human insulin, and 1% w/w Pen-Strep is herein referred to as Treatment Medium. Following seeding of cells in the plates, the plates were spun at 300 × g^for 30 seconds, then equilibrated to room temperature for 30 minutes, and then deposited^in a humidified tissue culture incubator maintained at 37 °C with 5% CO 2 . [0693] At 24 hours after seeding of the cells, dilutions of test compound were prepared in DMSO and dissolved in Treatment Medium, to achieve a final DMSO concentration of 0.5% w/w, thereby providing the Test Compound Solution. A 25 ^L aliquot of the Test Compound Solution was added to cells in the well plates. An equal volume of a solution containing DMSO and Treatment Medium was used as a negative control. Following treatment of cells with Test Compound Solution or said equal volume of a solution containing DMSO and Treatment Medium, the plates were spun at 300 × g for 30 seconds, and then left in an incubator for 72 hours. [0694] At the end of the treatment duration, cell viability was quantified with CellTiter-Glo 2.0 reagent (Promega Cat#G9243). For this purpose, plates were equilibrated to room temperature for 30 minutes, and then 25 µL of CellTiter-Glo 2.0 reagent was added to cells in the plate wells. Plates were then agitated on a shaker for two minutes at 500 rpm and subsequently incubated at room temperature for 10 minutes. Following incubation, the plates were spun at 3000 x g for 30 seconds, then sealed with an optical adhesive cover, and luminescence readings were measured with an EnVision Plate Reader (Perkin Elmer). [0695] Data was normalized using zero luminescence for baseline. A four-parameter non-linear regression curve fit was applied to dose-response data in GraphPad Prism data analysis software to determine the half-maximal growth inhibitory concentration (GI50) for each test compound. Part II – Results [0696] The half-maximal growth inhibitory concentration (GI50) results are provided in Table 11 below for exemplary compounds. The symbol “++++” indicates a GI50 less than 0.5 ^M. The symbol “+++” indicates an GI50 in the range of 0.5 ^M to 1.5 ^M. The symbol “++” indicates a GI50 in the range of greater than 1.5 ^M to 3 ^M. TABLE 11.

EXAMPLE 54 – Assay for Binding Affinity to Progesterone Receptor [0697] Exemplary compounds were tested for ability to bind to the progesterone receptor. Experimental procedures and results are provided below. Part I – Experimental Procedure [0698] Compounds were tested using the PR Human Progesterone NHR Binding (Agonist Radioligand) Assay, Cerep at Eurofins Discovery using a modified version of the protocol from Sarup et al. (Cancer Res 1988;48:5071-5078). Briefly, cytosolic fractions of T47D cells were incubated with 0.5 nM [ 3 H]progesterone for 1 hour at 4°C in the absence or presence of the test compound. Nonspecific binding was determined in the presence of 1 μM promegestone. Reactions were subjected to scintillation counting. Results are expressed as a percent inhibition of the control radioligand specific binding. Part II – Results [0699] Results showing ability of exemplary compounds to bind to the progesterone receptor are provided in Table 12 below. The symbol “++++” indicates a Kd less than 0.05 ^M. The symbol “+++” indicates an Kd in the range of 0.05 ^M to 0.5 ^M. The symbol “++” indicates a Kd in the range of greater than 0.5 ^M to 2.5 ^M. TABLE 12. EXAMPLE 55 – FP Assay for Binding Affinity to Progesterone Receptor [0700] Exemplary compounds were tested for ability to bind to the progesterone receptor ligand binding domain (LBD). Experimental procedures and results are provided below. Part I – Experimental Procedure [0701] Compounds were tested using the PolarScreen™ Progesterone Receptor Competitor Assay Kit (Life Technologies Cat# A15905 or a15906), as per the manufacturer’s instructions. Briefly, GST-tagged PR LBD, Fluormone tracer, and compound were mixed and incubated for 2 hours at room temperature in the dark. Following incubation, the plates were spun at 3000 x g for 30 seconds, then sealed with an optical adhesive cover, and fluorescence polarization was measured using appropriate excitation and emission filters with an EnVision Plate Reader (Perkin Elmer). Compound concentration vs. mP value were plotted in Prism (GraphPad). A four- parameter non-linear regression curve fit was applied to dose-response data to determine the half- maximal inhibitory concentration (IC 50 ) for each test compound. Part II – Results [0702] The half-maximal inhibitory concentration (IC50) results are provided in Table 13 below for exemplary compounds. The symbol “++++” indicates a IC 50 less than 0.05 ^M. The symbol “+++” indicates an IC 50 in the range of 0.05 ^M to 0.5 ^M. The symbol “++” indicates a IC 50 in the range of greater than 0.5 ^M to 2.5 ^M. TABLE 13. INCORPORATION BY REFERENCE [0703] The entire disclosure of each of the patent documents and scientific articles referred to herein is incorporated by reference for all purposes. EQUIVALENTS [0704] The invention may be embodied in other specific forms without departing from the spirit or essential characteristics thereof. The foregoing embodiments are therefore to be considered in all respects illustrative rather than limiting the invention described herein. Scope of the invention is thus indicated by the appended claims rather than by the foregoing description, and all changes that come within the meaning and range of equivalency of the claims are intended to be embraced therein.