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/2024/054603
Kind Code:
A1
Abstract:
The invention provides heterobifunctional compounds which may bind to both an androgen receptor and BRD4 (bromodomain-containing protein 4). Also provided are pharmaceutical compositions comprising the same and their use in treating disease, such as cancer.

Inventors:
EASTMAN KYLE J (US)
KAYSER-BRICKER KATHERINE J (US)
MARTIN MICHAEL BARRINGTON (US)
MOUSSEAU JAMES JOHN (US)
PERRY MATTHEW ALEXANDER (US)
PULEO DAVID E (US)
RAINA KANAK SHAIL (US)
ZAWARE NILESH K (US)
NEKLESA TAAVI (US)
GERRITZ SAMUEL W (US)
BOSANAC TODD (US)
Application Number:
PCT/US2023/032252
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:
C07D495/14; A61K31/55; A61K31/551; A61P35/00; C07D498/04; C07D498/14; C07D519/00
Domestic Patent References:
WO2019215488A12019-11-14
WO2016149668A12016-09-22
WO2023059581A12023-04-13
Other References:
TANG, P. ET AL.: "Targeting Bromodomain and Extraterminal Proteins for Drug Discovery: From Current Progress to Technological Development", J. MED. CHEM., vol. 64, 2021, pages 2419 - 2435, XP093118352, DOI: 10.1021/acs.jmedchem.0c01487
DUAN,Y. ET AL.: "Targeting Brd4 for cancer therapy: inhibitors and degraders", MED. CHEM. COMMUN., vol. 9, 2018, pages 1779 - 1802, XP055842447, DOI: 10.1039/C8MD00198G
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 represents independently for each occurrence C1-4 alkyl; R3 is hydrogen or C1-4 alkyl; R4 is C1-4 alkyl; R5 represents independently for each occurrence C1-4 alkyl or halogen; or one occurrence of R5 is taken together with R3 to form a C1-3 alkylene; A1 is a pyridazinylene, pyrimidinylene, pyrazinylene, or pyridinylene, each of which is substituted with n occurrences of R5; A2 is one of the following: , , , (R10A)s R1A N N O N , or (R9A)q ; R1A is C1-4 alkyl or C3-4 cycloalkyl; R2A represents independently for each occurrence C1-4 alkyl or C3-4 cycloalkyl; R3A is phenyl substituted with 1, 2, or 3 substituents independently selected from halo, C1-C4 alkyl, or C1-C4 haloalkyl R4A is -(C1-6 alkylene)-C(O)N(R5A)(R6A), -(C1-6 alkylene)-N(R5A)C(O)R7A, -(C1-6 alkylene)-CO2R8A, -(C1-6 alkylene)-OC(O)R7A, -(C1-6 alkylene)-CN, -(C1-6 alkylene)-O-(C1-6 alkyl), C1-6 alkyl, C3-6 cycloalkyl, or -(C0-6 alkylene)-(5-6 membered heteroaryl containing 1, 2, 3, or 4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, wherein the heteroaryl is substituted with 0, 1, or 2 occurrences of C1-6 alkyl); R5A and R6A are independently hydrogen, C1-6 alkyl, or C3-6 cycloalkyl; or R5A and R6A are taken together with the nitrogen atom to which they are attached to form a 3-7 membered ring containing 1 nitrogen atom; R7A is C1-6 alkyl, -(C1-6 alkylene)-(C3-6 cycloalkyl), or C3-6 cycloalkyl; R8A is hydrogen, C1-6 alkyl, -(C1-6 alkylene)-(C3-6 cycloalkyl), or C3-6 cycloalkyl; R9A is halo; R10A represents independently for each occurrence C1-4 alkyl or -N(R11A)2; R11A represents independently for each occurrence hydrogen or C1-4 alkyl; R12A is -(C0-6 alkylene)-(5-6 membered heteroaryl containing 1, 2, 3, or 4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, wherein the heteroaryl is substituted with 0, 1, or 2 occurrences of C1-6 alkyl); L is a linker; k is 1, 2, 3, or 4; m, n, p, and q are independently 0, 1, or 2; and s is 1 or 2. 2. The compound of claim 1, wherein R2 is methyl. 3. The compound of claim 1 or 2, wherein R3 is hydrogen. 4. The compound of any one of claims 1-3, wherein m is 0. 5. The compound of any one of claims 1-3, wherein R1 is . 6. The compound of any one of claims 1-5, wherein k is 4. 7. The compound of any one of claims 1-6, wherein the compound is a compound of Formula I. 8. The compound of claim 1 or 2, wherein the compound is a compound of Formula Ia or a pharmaceutically acceptable salt thereof:

9. The compound of claim 1 or 2, wherein the compound is a compound of Formula Ib or a pharmaceutically acceptable salt thereof: 10. The compound of claim 1, wherein the compound is a compound of Formula Ic or a pharmaceutically acceptable salt thereof: 11. The compound of claim 1, wherein the compound is a compound of Formula Id or a pharmaceutically acceptable salt thereof: 12. The compound of any one of claims 1-11, wherein A1 is pyridazinylene substituted with n occurrences of R5. 13. The compound of any one of claims 1-11, wherein A1 is .

14. The compound of any one of claims 1-11, wherein A1 is pyrimidinylene substituted with n occurrences of R5. 15. The compound of any one of claims 1-11, wherein A1 is , where ** is the point of attachment to L. 16. The compound of any one of claims 1-11, wherein A1 is , where ** is the point of attachment to L. 17. The compound of any one of claims 1-11, wherein A1 is pyrazinylene substituted with n occurrences of R5. 18. The compound of any one of claims 1-11, wherein A1 is . 19. The compound of any one of claims 1-11, wherein A1 is pyridinylene substituted with n occurrences of R5. 20. The compound of any one of claims 1-11, wherein A1 where ** is the point of attachment to L. 21. The compound of any one of claims 1-12, 14, 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 R3A is phenyl substituted with halo.

24. The compound of any one of claims 1-21, wherein . 25. The compound of any one of claims 1-21 or 24, wherein R4A is -(C1-6 alkylene)- C(O)N(R5A)(R6A). 26. The compound of any one of claims 1-21 or 24, wherein R4A is -(C1-6 alkylene)-CO2R8A. 27. The compound of any one of claims 1-21 or 24, wherein R4A is -(C0-6 alkylene)-(5-6 membered heteroaryl containing 1, 2, or 3 heteroatoms independently selected from nitrogen, oxygen, and sulfur).. 28. The compound of any one of claims 1-21 or 24, wherein R4A is -(C1-3 alkylene)-(5-membered heteroaryl containing 1 or 2 heteroatoms independently selected from nitrogen, oxygen, and sulfur). 29. The compound of any one of claims 1-21 or 24, wherein R4A is -(C1-3 alkylene)-(oxazolyl). 30. The compound of any one of claims 1-29, wherein R1A is C1-4 alkyl. 31. The compound of any one of claims 1-29, wherein R1A is methyl. 32. The compound of any one of claims 1-31, wherein R2A is C1-4 alkyl. 33. The compound of any one of claims 1-31, wherein R2A is methyl. 34. The compound of any one of claims 1-33, wherein p is 2.

35. The compound of any one of claims 1-21, wherein A2 is . 6. The compound of any one of claims 1-21, wherein A2 3 is , , or . 37. The compound of any one of claims 1-21, wherein A2 is , , , or .

38. The compound of any one of claims 1-21, wherein A2 is . 39. The compound of any one of claims 1-21, wherein A2 is . 40. The compound of any one of claims 1-21, wherein A2 is . 41. The compound of any one of claims 1-21, wherein A2 is , wherein q is 1.

42. The compound of any one of claims 1-21, wherein A2 is or . 43. The compound of any one of claims 1-21, wherein A2 is . 44. The compound of any one of claims 1-21, wherein A2 is . 45. The compound of any one of claims 1-21, wherein A2 is one of the following:

. 46. 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 represents independently for each occurrence C1-4 alkyl; R3 is hydrogen or C1-4 alkyl; R4 is C1-4 alkyl; R5 represents independently for each occurrence C1-4 alkyl or halogen; or one occurrence of R5 is taken together with R3 to form a C1-3 alkylene; A1 is a pyridazinylene, pyrimidinylene, pyrazinylene, or pyridinylene, each of which is substituted with n occurrences of R5; A2 is ; R1A is C1-4 alkyl or C3-4 cycloalkyl; R2A represents independently for each occurrence C1-4 alkyl or C3-4 cycloalkyl; R3A is phenyl substituted with 1, 2, or 3 substituents independently selected from halo, C1-C4 alkyl, or C1-C4 haloalkyl R4A is C1-6 hydroxyalkyl, C1-4 haloalkyl, -(C1-6 alkylene)-C(O)N(R5A)(R6A), -(C1-6 alkylene)-N(R5A)C(O)R7A, -(C1-6 alkylene)-CO2R8A, -(C1-6 alkylene)-OC(O)R7A, -(C1-6 alkylene)-CN, -(C1-6 alkylene)-O-(C1-6 alkyl), C1-6 alkyl, C3-6 cycloalkyl, or -(C0-6 alkylene)- (5-6 membered heteroaryl containing 1, 2, 3, or 4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, wherein the heteroaryl is substituted with 0, 1, or 2 occurrences of C1-6 alkyl); R5A and R6A are independently hydrogen, C1-6 alkyl, or C3-6 cycloalkyl; or R5A and R6A are taken together with the nitrogen atom to which they are attached to form a 3-7 membered ring containing 1 nitrogen atom; R7A is C1-6 alkyl, -(C1-6 alkylene)-(C3-6 cycloalkyl), or C3-6 cycloalkyl; R8A is hydrogen, C1-6 alkyl, -(C1-6 alkylene)-(C3-6 cycloalkyl), or C3-6 cycloalkyl; R9A is halo; R10A is hydrogen or C1-4 alkyl; or R4A and R10A are taken together with the carbon atom to which they are attached to form a 3-5 membered saturated carbocylic ring; L is a linker; k is 1, 2, 3, or 4; and m, n, p, and q are independently 0, 1, or 2. 47. The compound of claim 46, wherein A2 is one of the following: 48. The compound of any one of claims 1-47, 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(C3-7 cycloalkyl)-, 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. 49. The compound of any one of claims 1-47, 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. 50. The compound of any one of claims 1-47, 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)-. 51. The compound of any one of claims 1-47, 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. 52. The compound of any one of claims 1-47, 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. 53. The compound of any one of claims 1-47, 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.

54. The compound of any one of claims 1-47, 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. 55. The compound of any one of claims 1-47, wherein L is -piperidinylene-(OCH2CH2)1-15-O- ***, wherein *** is the point of attachment to A2. 56. The compound of any one of claims 1-47, wherein L is , wherein *** is the point of attachment to A2. 57. The compound of any one of claims 1-47, wherein L is , wherein *** is the point of attachment to A2. 58. The compound of any one of claims 1-47, wherein L is , wherein *** is the point of attachment to A2. 59. The compound of any one of claims 1-47, 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. 60. The compound of any one of claims 1-47, 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.

61. The compound of any one of claims 1-47, 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. 62. The compound of any one of claims 1-47, 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. 63. The compound of any one of claims 1-47, 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)-. 64. The compound of any one of claims 1-47, 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)-. 65. The compound of any one of claims 1-47, wherein L is , wherein *** is the point of attachment to A2, and X1 is (i) C1-10 alkylen e 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)-.

66. The compound of any one of claims 1-47, 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)-. 67. The compound of any one of claims 1-47, 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)-. 68. The compound of any one of claims 1-47, 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)-. 69. The compound of any one of claims 1-47, 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)-. 70. The compound of any one of claims 1-47, 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)-. N X2 C1-10 alkylene *** 71. The compound of any one of claims 1-47, wherein L is , wherein *** is the point of attachment to A2, and X2 is - O-, -N(H)-, or -N(C1-6 alkyl)-. 72. The compound of any one of claims 1-47, wherein L is , wherein *** is the point of attachment to A2, and X2 is -O-, -N(H)-, or -N(C1-6 alkyl)-.

73. The compound of any one of claims 68-72, wherein X2 is -O-. 74. The compound of any one of claims 1-47, 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)-. 75. The compound of any one of claims 1-47 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. 76. The compound of any one of claims 1-47, wherein L is a 7-8 membered spirocyclic or fused bicyclic saturated heterocyclic ring containing 2 heteroatoms selected from nitrogen. 77. The compound of any one of claims 1-47, wherein L is a -(8-10 membered spirocyclic saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen)-O-***, where *** is the point of attachment to A2. 78. The compound of any one of claims 1-47, wherein L is a -(9-membered spirocyclic saturated heterocyclic ring containing 1 heteroatom selected from nitrogen)-O-***, where *** is the point of attachment to A2. 79. The compound of any one of claims 1-47, 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. 80. The compound of any one of claims 1-47, wherein L is -(piperidinylene)-(C1-5 alkylene)- (piperazinylene)-***, wherein *** is the point of attachment to A2. 81. The compound of any one of claims 1-47, wherein L is -(piperazinylene)-(azetidinylene)-*** or (azetidinylene)-(piperazinylene)-***, wherein *** is the point of attachment to A2. 82. The compound of any one of claims 1-47, 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)-(C2-6 alkylene)-N(H)C(O)-(C1-6 alkylene)-***, -(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)-(C2-6 alkylene)-N(C1-6 alkyl)C(O)-(C1-6 alkylene)-***, 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)-(C2-6 alkylene)-C(O)N(H)-(C1-6 alkylene)-***, -(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)-(C2-6 alkylene)-C(O)N(C1-6 alkyl)-(C1-6 alkylene)-***, wherein X3 is C1-10 alkylene, -O-, -N(H)-, -N(C1-4 alkyl)-, or a bond. 83. The compound of any one of claims 1-47, wherein L is -(piperidinylene)-(C1-5 alkylene)- (piperazinylene)-(C2-5 alkylene)-N(H)C(O)-(C1-6 alkylene)-***, wherein *** is the point of attachment to A2. 84. The compound of any one of claims 1-47, 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. 85. The compound of any one of claims 1-47, 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. 86. The compound of any one of claims 1-47, wherein L is -N(C1-3 alkyl)-(C2-7 alkylene)- N(H)C(O)-(C1-6 alkylene)-***, -N(C1-3 alkyl)-(C2-7 alkylene)-N(C1-3 alkyl)C(O)-(C1-6 alkylene)-***, -N(H)-(C2-7 alkylene)-N(H)C(O)-(C1-6 alkylene)-***, -N(H)-(C2-7 alkylene)- N(C1-3 alkyl)C(O)-(C1-6 alkylene)-***, -N(C1-3 alkyl)-(C2-7 alkylene)-C(O)N(H)-(C1-6 alkylene)-***, -N(C1-3 alkyl)-(C2-7 alkylene)-C(O)N(C1-3 alkyl)-(C1-6 alkylene)-***, -N(H)- (C2-7 alkylene)-C(O)N(H)-(C1-6 alkylene)-***, or -N(H)-(C2-7 alkylene)-C(O)N(C1-3 alkyl)- (C1-6 alkylene)-***, where *** is a point of attachment to A2.

87. The compound of any one of claims 1-47, wherein L is -N(C1-3 alkyl)-[(C2-4 alkylene)-O-]2-8- (C2-6 alkylene)-N(H)C(O)-(C1-6 alkylene)-, -N(C1-3 alkyl)-[(C2-4 alkylene)-O-]2-8-(C2-6 alkylene)-N(C1-3 alkyl)C(O)-(C1-6 alkylene)-, -N(H)-[(C2-4 alkylene)-O-]2-8-(C2-6 alkylene)- N(H)C(O)-(C1-6 alkylene)-, -N(H)-[(C2-4 alkylene)-O-]2-8-(C2-6 alkylene)-N(C1-3 alkyl)C(O)- (C1-6 alkylene)-, -N(C1-3 alkyl)-[(C2-4 alkylene)-O-]2-8-(C2-6 alkylene)-C(O)N(H)-(C1-6 alkylene)-, -N(C1-3 alkyl)-[(C2-4 alkylene)-O-]2-8-(C2-6 alkylene)-C(O)N(C1-3 alkyl)-(C1-6 alkylene)-, or -N(H)-[(C2-4 alkylene)-O-]2-8-(C2-6 alkylene)-C(O)N(H)-(C1-6 alkylene)-, - N(H)-[(C2-4 alkylene)-O-]2-8-(C2-6 alkylene)-C(O)N(C1-3 alkyl)-(C1-6 alkylene)-, where *** is a point of attachment to A2. 88. The compound of any one of claims 1-47, wherein L is -N(CH3)-[(CH2CH2)-O-]2-8-(C2-6 alkylene)-N(H)C(O)-(C1-6 alkylene)-***, -N(CH3)-[(CH2CH2)-O-]2-8-(C2-6 alkylene)- N(CH3)C(O)-(C1-6 alkylene)-***, -N(H)-[(CH2CH2)-O-]2-8-(C2-6 alkylene)-N(H)C(O)-(C1-6 alkylene)-***, or -N(H)-[(CH2CH2)-O-]2-8-(C2-6 alkylene)-N(H)C(O)-(C1-6 alkylene)-***, where *** is a point of attachment to A2. 89. The compound of any one of claims 1-47, wherein L has the formula –(C0-12 alkylene)- (optionally substituted 3-40 membered heteroalkylene)-(C0-12 alkylene)-. 90. The compound of any one of claims 1-47, wherein L is one of the following: wherein *** is the point of attachment to A2.

91. The compound of any one of claims 1-47, wherein L is one of the following: wherein is the point of attachment to A .

92. The compound of any one of claims 1-47, wherein L is wherein *** is the point of attachment to A2. 93. The compound of any one of claims 1-47, wherein L is wherein *** is the point of attachment to A2. 94. The compound of any one of claims 1-47, wherein L is -(8-12 membered spirocyclic heterocyclyl containing 1, 2 or 3 heteroatoms independently selected from nitrogen and oxygen)-(C3-4 cycloalkylene)-***, -(8-12 membered spirocyclic heterocyclyl containing 1, 2 or 3 heteroatoms independently selected from nitrogen and oxygen)-(C0–4 alkylene)-***, -(8- 12 membered spirocyclic heterocyclyl containing 1, 2 or 3 heteroatoms independently selected from nitrogen and oxygen)-C(O)-(C1-4 alkylene)-***, -(5-6 membered saturated monocyclic heterocyclylene containing 1 or 2 heteroatoms independently selected from nitrogen and oxygen)-(3-5 membered saturated monocyclic heterocyclylene containing 1 or 2 heteroatoms independently selected from nitrogen and oxygen)-***, -(5-6 membered saturated monocyclic heterocyclylene containing 1 or 2 heteroatoms independently selected from nitrogen and oxygen)-O-***, -(5-6 membered saturated monocyclic heterocyclylene containing 1 or 2 heteroatoms independently selected from nitrogen and oxygen)-(C0-4 alkylene)-***, -(5-6 membered saturated monocyclic heterocyclylene containing 1 or 2 heteroatoms )-(C0–4 alkylene)-O-***, -(8-12 membered spirocyclic heterocyclyl containing 1, 2 or 3 heteroatoms independently selected from nitrogen and oxygen)-C(O))-***, -(8-12 membered spirocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-O- (C0-6 alkylene)-***, -(8-12 membered spirocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-(C1-6 alkylene)-O-(C0-6 alkylene)-***, -(5-6 membered saturated monocyclic heterocyclylene containing 1 or 2 heteroatoms )-C(O)N(H)-(C0-6 alkylene)-***, - (N(C1-6 alkyl)-(C0-6 alkylene)-C(O)N(H)-(C0-6 alkylene)-***, -(8-12 membered spirocyclic heterocyclyl containing 1, 2 or 3 heteroatoms independently selected from nitrogen and oxygen)-(C2-4 alkynylene)-***, -(5-6 membered saturated monocyclic heterocyclylene containing 1 or 2 heteroatoms independently selected from nitrogen and oxygen)-(C1-6 alkyl)- (3-5 membered saturated monocyclic heterocyclylene containing 1 or 2 heteroatoms independently selected from nitrogen and oxygen)-***, -(5-6 membered saturated monocyclic heterocyclylene containing 1 or 2 heteroatoms selected from nitrogen)- C(O)N(H)-(C0-6 alkylene)-N(H))-***, -(C(O)N(H)-(C0-6 alkylene)-(5-6 membered saturated monocyclic heterocyclylene containing 1 or 2 heteroatoms )-***, -(5-6 membered saturated monocyclic heterocyclylene containing 1 or 2 heteroatoms independently selected from nitrogen and oxygen)-N(C1-6 alkyl)-(5-6 membered saturated monocyclic heterocyclylene containing 1 or 2 heteroatoms independently selected from nitrogen and oxygen)-O-***, - C(O)-(5-6 membered saturated monocyclic heterocyclylene containing 1 or 2 heteroatoms selected from nitrogen)-C(O)-***, -(C0-6 alkylene)-(5-6 membered saturated monocyclic heterocyclylene containing 1 or 2 heteroatoms selected from nitrogen)-C(O)-***, -(5-6 membered saturated monocyclic heterocyclylene containing 1 or 2 heteroatoms selected from nitrogen)-C(O)-(C0-6 alkylene)***, -C(O)-(5-6 membered saturated monocyclic heterocyclylene containing 1 or 2 heteroatoms selected from nitrogen)-(C1-6 alkylene)-***, - (C(O)N(H)-(C1-6 alkylene)-C(O)N(H)-(C0-6 alkylene)-***, or -(8-11 membered fused bicyclic heterocyclyl containing 1, 2 or 3 heteroatoms independently selected from nitrogen and oxygen)-(C1-4 alkylene)-***, wherein *** is the point of attachment to A2. 95. The compound of any one of claims 1-47, wherein L is -N(C1-6 alkyl)-(5-6 membered saturated monocyclic heterocyclylene containing 1 or 2 heteroatoms selected from nitrogen)- C(O)-(C0-6 alkylene)-***, -(5-6 membered saturated monocyclic heterocyclylene containing 1 or 2 heteroatoms selected from nitrogen)-(5-6 membered saturated monocyclic heterocyclylene containing 1 or 2 heteroatoms selected from nitrogen)-(C0-4 alkylene)-O-***, -(C0-6 alkylene)-N(H)C(O)N(H)-(C0-6 alkylene)-***, -N(H)-(C0-6 alkylene)-(5-6 membered saturated monocyclic heterocyclylene containing 1 or 2 heteroatoms selected from nitrogen)- (C0-6 alkylene)-***, -(C0-6 alkylene)-C(O)-(5-6 membered saturated monocyclic heterocyclylene containing 1 or 2 heteroatoms selected from nitrogen)-(C0-6 alkylene)-***, - (C0-6 alkylene)-(5-6 membered saturated monocyclic heterocyclylene containing 1 or 2 heteroatoms selected from nitrogen)-C(O)N(C1-6 alkyl)-(C0-6 alkylene)-***, -(C0-6 alkylene)- (5-6 membered saturated monocyclic heterocyclylene containing 1 or 2 heteroatoms selected from nitrogen)-(C0-6 alkylene)-N(C1-6 alkyl)-(3-6 membered saturated monocyclic heterocyclylene containing 1 or 2 heteroatoms selected from nitrogen)-***, -(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)-(C0-6 alkylene)-(5-6 membered heteroaryl containing 1 or 2 heteroatoms selected from nitrogen)-***, -(8-12 membered spirocyclic heterocyclyl substituted with 1 or 2 fluoro containing 1, 2 or 3 heteroatoms independently selected from nitrogen and oxygen)-(C1-4 alkylene)-***, -(5-6 membered saturated monocyclic heterocyclylene containing 1 or 2 heteroatoms selected from nitrogen)- (C0-4 alkylene)-(C3-6 cycloalkylene)-(C0-4 alkylene)-O-***, -(5-6 membered saturated monocyclic heterocyclylene containing 1 or 2 heteroatoms selected from nitrogen)-(C0-4 alkylene)-(C3-6 cycloalkylene)-(C0–4 alkylene)-***, -(C0–4 alkylene)-(8-12 membered spirocyclic heterocyclyl containing 1, 2 or 3 heteroatoms independently selected from nitrogen and oxygen)-***, -(C0-4 alkylene)-(C3-6 cycloalkylene)-(C2-4 alkynylene)-***, -(C0-4 alkylene)-(8-10 membered fused bicyclic heterocyclyl substituted with 1 or two fluoro containing 1 or 2 heteroatoms selected from nitrogen)-(C0–4 alkylene)-***, -(8-12 membered spirocyclic heterocyclyl containing 1, 2 or 3 heteroatoms independently selected from nitrogen and oxygen)-O-***, -(8-12 membered spirocyclic heterocyclyl containing 1, 2 or 3 heteroatoms independently selected from nitrogen and oxygen)-(C0–4 alkylene)-(5-6 membered saturated monocyclic heterocyclylene containing 1 or 2 heteroatoms selected from nitrogen)-***, -(C0-4 alkylene)-(4-6 membered saturated heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-(phenylene substituted with trifluoromethyl)-(C0–4 alkylene)-N(H)-***, or is -(5-6 membered saturated monocyclic heterocyclylene containing 1 or 2 heteroatoms selected from nitrogen)-C(O)-(5-6 membered saturated monocyclic heterocyclylene containing 1 or 2 heteroatoms selected from nitrogen)-(C0-6 alkylene)-***, - (C3-6 cycloalkylene)-C(O)N(C1-6 alkyl)(C0-6 alkylene)-***, -(5-6 membered saturated monocyclic heterocyclylene containing 1 or 2 heteroatoms selected from nitrogen)- (phenylene substituted with 0 or 1 occurrence of methyl or halo)-(C0-6 alkylene)-***, -(5-6 membered saturated monocyclic heterocyclylene containing 1 or 2 heteroatoms selected from nitrogen)-(C0-6 alkylene)-(5-6 membered saturated monocyclic oxo-substituted heterocyclylene containing 1 or 2 heteroatoms selected from nitrogen)-(C0-6 alkylene)-***, - (8-12 membered spirocyclic C1-4 alkyl substituted heterocyclyl containing 1, 2 or 3 heteroatoms independently selected from nitrogen and oxygen)-(C0-6 alkylene)-(O)0-1***, - (C2-4 alkynylene)-(8-12 membered spirocyclic heterocyclyl containing 1, 2 or 3 heteroatoms independently selected from nitrogen and oxygen)-(C0-4 alkylene)-***, -(C0-4 alkylene)-(C3-7 cycloalkylene)-(C2-4 alkynylene)-***, -(C1-4 alkylene)-(8-12 membered spirocyclic heterocyclyl containing 1, 2 or 3 heteroatoms independently selected from nitrogen and oxygen)-(C0-4 alkylene)-***, -(C1-4 alkylene)-(5-7 membered saturated heterocyclyl containing 1, 2 or 3 heteroatoms independently selected from nitrogen and oxygen)-(C0–4 alkylene)-***, -(C0–4 alkylene)-(5-7 membered saturated heterocyclyl containing 1 or 2 heteroatoms independently selected from nitrogen and oxygen)-(C2-4 alkenylene)-***, or - (C0-4 alkylene)-(6-8 membered saturated heterocyclyl substituted with 1 or 2 fluoro containing 1 or 2 heteroatoms independently selected from nitrogen and oxygen)-(C0–4 alkylene)-***, wherein *** is the point of attachment to A2. 96. The compound of claim 1, wherein the compound is a compound of Formula Ie or a pharmaceutically acceptable salt thereof: wherein L is (i) Ψ-(8-10 membered spirocyclic saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen)-O-, where Ψ is the point of attachment to the pyrimidinylene in Formula Ie or (ii) a 9-11 membered spirocyclic saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen, wherein the heterocyclic ring is substituted with 0 or 1 occurrences of C1-4 alkyl.

97. The compound of claim 96, wherein L is Ψ-(8-10 membered spirocyclic saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen)-O-, where Ψ is the point of attachment to the pyrimidinylene in Formula Ie. 98. The compound of claim 96, wherein L is Ψ-(9-membered spirocyclic saturated heterocyclic ring containing 1 heteroatom selected from nitrogen)-O-, where Ψ is the point of attachment to the pyrimidinylene in Formula Ie. 99. The compound of claim 96, wherein L is a 9-11 membered spirocyclic saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen. 100. The compound of claim 96, wherein L is a 10-membered spirocyclic saturated heterocyclic ring containing 1 heteroatom selected from nitrogen. 101. The compound of claim 1, wherein the compound is a compound of Formula If or a pharmaceutically acceptable salt thereof: 102. The compound of claim 1, wherein the compound is a compound of Formula Ig or a pharmaceutically acceptable salt thereof: 1g. 103. 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) wherein: RII-1A is a bond to L; R1 is phenyl substituted by cyano, halogen, and m occurrences of R4; R2 represents independently for each occurrence C1-4 alkyl; R3 is hydrogen or C1-4 alkyl; R4 is C1-4 alkyl; R5 represents independently for each occurrence C1-4 alkyl or halogen; or one occurrence of R5 is taken together with R3 to form a C1-3 alkylene; A1 is a pyridazinylene, pyrimidinylene, pyrazinylene, or pyridinylene, each of which is substituted with n occurrences of R5; L is a linker; EPL is a moiety that binds to BRD4; k is 1, 2, 3, or 4; and m and n are independently 0, 1, or 2.

104. The compound of claim 103, wherein the TPL is that is substituted by one occurrence of RII-1A. 105. The compound of claim 103, wherein the TPL is . 106. The compound of any one of claims 103-105, 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; R1A is C1-4 alkyl or C3-4 cycloalkyl; R2A represents independently for each occurrence C1-4 alkyl or C3-4 cycloalkyl; R3A is phenyl substituted with 1, 2, or 3 substituents independently selected from halo, C1-C4 alkyl, or C1-C4 haloalkyl; R4A is -(C1-6 alkylene)-C(O)N(R5A)(R6A), -(C1-6 alkylene)-N(R5A)C(O)R7A, -(C1-6 alkylene)- CO2R8A, -(C1-6 alkylene)-OC(O)R7A, -(C1-6 alkylene)-CN, -(C1-6 alkylene)-O-(C1-6 alkyl), C1- 6 alkyl, C3-6 cycloalkyl, or -(C0-6 alkylene)-(5-6 membered heteroaryl containing 1, 2, 3, or 4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, wherein the heteroaryl is substituted with 0, 1, or 2 occurrences of C1-6 alkyl); R5A and R6A are independently hydrogen, C1-6 alkyl, or C3-6 cycloalkyl; or R5A and R6A are taken together with the nitrogen atom to which they are attached to form a 3-7 membered ring containing 1 nitrogen atom; R7A is C1-6 alkyl, -(C1-6 alkylene)-(C3-6 cycloalkyl), or C3-6 cycloalkyl; R8A is hydrogen, C1-6 alkyl, -(C1-6 alkylene)-(C3-6 cycloalkyl), or C3-6 cycloalkyl; R9A is halo; R10A represents independently for each occurrence C1-4 alkyl or -N(R11A)2; R11A represents independently for each occurrence hydrogen or C1-4 alkyl; R12A is -(C0-6 alkylene)-(5-6 membered heteroaryl containing 1, 2, 3, or 4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, wherein the heteroaryl is substituted with 0, 1, or 2 occurrences of C1-6 alkyl); p and q are independently 0, 1, or 2; and s is 1 or 2. 107. The compound of any one of claims 103-105, wherein the EPL is ,

each of which is substituted by one occurrence of RII-2A, wherein RII-2A is a bond to L. 108. The compound of any one of claims 103-105, wherein the EPL is . 109. The compound of any one of claims 103-105, wherein the EPL is , 110. The compound of any one of claims 103-109, 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. 111. The compound of any one of claims 103-109, 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. 112. The compound of any one of claims 103-109, 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)-. 113. A compound in Table 1, or a pharmaceutically acceptable salt thereof. 114. A pharmaceutical composition comprising a compound of any one of claims 1-113 and a pharmaceutically acceptable carrier. 115. 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 1-113 to treat the cancer. 116. The method of claim 115, 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. 117. The method of claim 115, wherein the cancer is prostate cancer.

118. 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 1-113 to cause death of the cancer cell. 119. The method of claim 118, 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. 120. The method of claim 118, wherein the cancer cell is a prostate cancer cell.

Description:
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 63/494,364, filed April 5, 2023; United States Provisional Patent Application serial number 63/444,828, filed February 10, 2023; and United States Provisional Patent Application serial number 63/404,567, filed September 8, 2022; the contents of each of which are hereby incorporated by reference. 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 accep table 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 , iables 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. BRIEF DESCRIPTION OF FIGURES [0010] Figure 1 is a graph that shows the cell viability dose-response curve for Compound I- 49 in two cell lines, LnCap95 Parental cells and LnCap95AR-FL KO cells. [0011] Figure 2 is a graph that shows the cell viability dose-response curves for Compound I- 49 and Compound I-50 in LnCap95 Parental cells. [0012] Figure 3 is a graph that shows the relative Caspase 3/7 activity dose response curve for Compound I-49 in two cell lines, 22RV1 Parental(FL-AR hi ) and 22RV1 Parental(FL-AR low ) [0013] Figure 4 is a graph that shows the relative Caspase 3/7 activity dose response curves of Compound I-49 and Compound I-50 in 22RV1 Parental(FL-AR hi ) cells. [0014] Figure 5 is a graph that shows the relative total Gene X mRNA dose response curve for Compound I-49 in TReX293 cells in the presence and absence of doxycycline. [0015] Figure 6 is a graph that shows tumor growth curves for Compound I-49 treated mice, Enzalutimide treated mice, and vehicle treated mice in Arm 1 of the Ar amp , V7 + Castrate VcaP Model. [0016] Figure 7 is a graph thaat shows tumor growth curves for Compound I-49 treated mice, Enzalutimide treated mice, and vehicle treated mice in Arm 2 of the Ar amp , V7 + Castrate VcaP Model. [0017] Figure 8 is a graph that shows the relative total Gene X mRNA present in tumors collected at the end of the PK/PD Ar amp , V7 + Castrate VcaP Model, for varying doses of Compound I-49 treatment. [0018] Figure 9 is a graph that shows the relative total Gene X mRNA present in tumors collected at the end of the PK/PD Ar amp , V7 + Castrate VcaP Model, with Compound I-49 and Compound I-50 treatment. [0019] Figure 10 is a graph that shows the plasma PSA concentration at varying Compound I- 49 doses for samples collected from Arm 1 of the Castrate VCaP Tumor Xenograft Model described above. [0020] Figure 11 is a graph that shows the plasma PSA concentration at varying Compound I- 49 doses, for samples collected from Arm 2 of the Castrate VCaP Tumor Xenograft Model described above. [0021] Figure 12 is a graph that shows the in-vitro relative ternary complex formation dose response curve for Compound I-49 versus free AR ligand and free EP ligand. [0022] Figure 13 is a graph that shows the in-vitro relative ternary complex formation dose response curve for Compound I-49 and Compound I-50. [0023] Figure 14 is a graph that shows the relative ternary complex formation present with treatment of Compound I-49 at varying doses in samples collected at the end of the PK/PD Castrate VCaP Tumor Xenograft Model. [0024] Figure 15 is a graph that shows the relative ternary complex formation present with treatment of Compound I-49 in samples collected from Arm 2 of the Castrate VCaP Tumor Xenograft Model. [0025] Figure 16 is a graph that shows the relative ternary complex formation present with treatment of Compound I-49 and Compund I-50 in samples collected at the end of the PK/PD Castrate VCaP Tumor Xenograft Model. DETAILED DESCRIPTION [0026] 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. [0027] 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 [0028] 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. [0029] 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, aliphatic 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 C 3 -C 6 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. [0030] 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:

[0031] Exemplary bridged bicyclics include: . [0032] 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. [0033] The term “lower haloalkyl” refers to a C 1-4 straight or branched alkyl group that is substituted with one or more halogen atoms. [0034] 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)). [0035] The term “unsaturated,” as used herein, means that a moiety has one or more units of unsaturation. [0036] 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. [0037] 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. [0038] The term “-(C0 alkylene)-“ refers to a bond. Accordingly, the term “-(C0-3 alkylene)-” encompasses a bond (i.e., C 0 ) and a -(C 1-3 alkylene)- group. [0039] 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. [0040] The term “halogen” means F, Cl, Br, or I. [0041] 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. [0042] 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. [0043] 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). [0044] 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. [0045] As used herein, the term “heterocycloalkyl” refers to a saturated heterocyclyl. The term “heterocycloalkylene” refers to a bivalent heterocycloalkyl group. [0046] 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. [0047] 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. [0048] 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– 4C(O)OR°; –(CH 2 ) 0–4 CH(OR °) 2 ; –(CH 2 ) 0–4 SR °; –(CH 2 ) 0–4 Ph, which may be substituted with R°; –(CH 2 ) 0–4 O(CH 2 ) 0–1 Ph which may be substituted with R°; –CH=CHPh, which may be substituted with R°; –(CH 2 ) 0–4 O(CH 2 ) 0–1 -pyridyl which may be substituted with R°; –NO 2 ; –CN; – N 3 ; -(CH 2 ) 0–4 N(R °) 2 ; –(CH 2 ) 0–4 N(R °)C(O)R °; –N(R °)C(S)R °; –(CH 2 ) 0–4 N(R °)C(O)NR ° 2 ; -N(R °)C(S)NR ° 2 ; –(CH 2 ) 0–4 N(R °)C(O)OR °; –N(R °)N(R °)C(O)R °; -N(R °)N(R °)C(O)NR ° 2 ; -N(R °)N(R °)C(O)OR °; –(CH 2 ) 0–4 C(O)R °; –C(S)R °; –(CH 2 ) 0–4 C(O)OR °; –(CH 2 ) 0–4 C(O)SR °; -(CH 2 ) 0–4 C(O)OSiR ° 3 ; –(CH 2 ) 0–4 OC(O)R °; –OC(O)(CH 2 ) 0–4 SR–, SC(S)SR°; –(CH 2 ) 0–4 SC(O)R °; –(CH 2 ) 0–4 C(O)NR °2; –C(S)NR °2; –C(S)SR°; –SC(S)SR°, -(CH 2 ) 0–4 OC(O)NR °2; -C(O)N(OR °)R °; –C(O)C(O)R °; –C(O)CH 2 C(O)R °; –C(NOR °)R °; -(CH 2 ) 0–4 SSR °; –(CH 2 ) 0– 4 S(O) 2 R °; –(CH 2 ) 0–4 S(O) 2 OR °; –(CH 2 ) 0–4 OS(O) 2 R °; –S(O) 2 NR °2; –S(O)(NR °)R °; – S(O) 2 N=C(NR ° 2 ) 2 ; -(CH 2 ) 0–4 S(O)R °; -N(R °)S(O) 2 NR °2; –N(R °)S(O) 2 R °; –N(OR °)R °; – C(NH)NR °2; –P(O) 2 R °; -P(O)R °2; -OP(O)R °2; –OP(O)(OR °) 2 ; SiR °3; –( C 1-4 straight or branched alkylene)O–N(R °) 2 ; or –(C 1-4 straight or branched alkylene)C(O)O–N(R °) 2 . [0049] Each R ° is independently hydrogen, C1–6 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, –(CH 2 ) 0–2 R , –(haloR ), –(CH 2 ) 0–2 OH, –(CH 2 ) 0–2 OR , – (CH 2 ) 0–2 CH(OR ) 2 ; -O(haloR ), –CN, –N3, –(CH 2 ) 0–2 C(O)R , –(CH 2 ) 0–2 C(O)OH, –(CH 2 )0– 2 C(O)OR , –(CH 2 ) 0–2 SR , –(CH 2 ) 0–2 SH, –(CH 2 ) 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 . [0050] 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) 2 R * , =NR * , =NOR * , –O(C(R * 2)) 2–3 O–, 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. [0051] When R * is C1–6 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. [0052] 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)CH 2 C(O)R , -S(O) 2 R , -S(O) 2 NR†2, –C(S)NR†2, – C(NH)NR 2, or –N(R )S(O) 2 R ; wherein each R is independently hydrogen, 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 C1–6 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. [0053] 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. [0054] 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. [0055] 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. [0056] 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. [0057] 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. [0058] 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. [0059] 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. [0060] The terms “a” and “an” as used herein mean “one or more” and include the plural unless the context is inappropriate. [0061] 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 C1-C12 alkyl, C1-C10 alkyl, and C1-C6 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. [0062] 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 “C 3 -C 6 cycloalkyl,” derived from a cycloalkane. Exemplary cycloalkyl groups include cyclohexyl, cyclopentyl, cyclobutyl, and cyclopropyl. The term “cycloalkylene” refers to a bivalent cycloalkyl group. [0063] 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. [0064] The term “hydroxyalkyl” refers to an alkyl group that is substituted with at least one hydroxyl. Exemplary hydroxyalkyl groups include -CH 2 CH 2 OH, -C(H)(OH)CH 3 , -CH 2 C(H)(OH)CH 2 CH 2 OH, and the like. [0065] 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. [0066] 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. [0067] The term “carbocyclylene” refers to a bivalent cycloaliphatic group. [0068] 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 -OCH 2 F, -OCHF 2 , -OCF 3 , -OCH 2 CF 3 , -OCF 2 CF 3 , and the like. [0069] The term “oxo” is art-recognized and refers to a “=O” substituent. For example, a cyclopentane susbsituted with an oxo group is cyclopentanone. [0070] 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: R 50 R 50 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. [0071] 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, -(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; and R 52 is an alkyl, an alkenyl, or -(CH 2 )m-R 61 . [0072] The symbol “ ” indicates a point of attachment. [0073] 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. [0074] 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. [0075] 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. [0076] 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. [0077] 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. [0078] 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. [0079] 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]. [0080] 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. [0081] 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. [0082] 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. [0083] As a general matter, compositions specifying a percentage are by weight unless otherwise specified. I. Heterobifunctional Compounds [0084] 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 [0085] One aspect of the invention provides a compound represented by Formula I: (I) or a pharmaceutically acceptable salt thereof; wherein: R 1 is phenyl substituted by cyano, halogen, and m occurrences of R 4 ; R 2 represents independently for each occurrence C 1-4 alkyl; R 3 is hydrogen or C 1-4 alkyl; R 4 is C 1-4 alkyl; R 5 represents independently for each occurrence C 1-4 alkyl or halogen; or one occurrence of R 5 is taken together with R 3 to form a C 1-3 alkylene; A 1 is a pyridazinylene, pyrimidinylene, pyrazinylene, or pyridinylene, each of which is substituted with n occurrences of R 5 ; A 2 is one of the following: , or ; R 1A is C 1-4 alkyl or C 3-4 cycloalkyl; R 2A represents independently for each occurrence C 1-4 alkyl or C 3-4 cycloalkyl; R 3A is phenyl substituted with 1, 2, or 3 substituents independently selected from halo, C 1 -C 4 alkyl, or C 1 -C 4 haloalkyl; R 4A is -(C 1-6 alkylene)-C(O)N(R 5A )(R 6A ), -(C 1-6 alkylene)-N(R 5A )C(O)R 7A , -(C 1-6 alkylene)-CO 2 R 8A , -(C 1-6 alkylene)-OC(O)R 7A , -(C 1-6 alkylene)-CN, -(C 1-6 alkylene)-O-(C 1-6 alkyl), C 1-6 alkyl, C 3-6 cycloalkyl, or -(C 0-6 alkylene)-(5-6 membered heteroaryl containing 1, 2, 3, or 4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, wherein the heteroaryl is substituted with 0, 1, or 2 occurrences of C 1-6 alkyl); R 5A and R 6A are independently hydrogen, C 1-6 alkyl, or C 3-6 cycloalkyl; or R 5A and R 6A are taken together with the nitrogen atom to which they are attached to form a 3-7 membered ring containing 1 nitrogen atom; R 7A is C 1-6 alkyl, -(C 1-6 alkylene)-(C 3-6 cycloalkyl), or C 3-6 cycloalkyl; R 8A is hydrogen, C 1-6 alkyl, -(C 1-6 alkylene)-(C 3-6 cycloalkyl), or C 3-6 cycloalkyl; R 9A is halo; R 10A represents independently for each occurrence C 1-4 alkyl or -N(R 11A ) 2 ; R 11A represents independently for each occurrence hydrogen or C 1-4 alkyl; R 12A is -(C 0-6 alkylene)-(5-6 membered heteroaryl containing 1, 2, 3, or 4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, wherein the heteroaryl is substituted with 0, 1, or 2 occurrences of C 1-6 alkyl); L is a linker; k is 1, 2, 3, or 4; m, n, p, and q are independently 0, 1, or 2; and s is 1 or 2. [0086] 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). [0087] In certain embodiments, the compound is a compound of Formula I. [0088] 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 phenyl substituted by cyano, chloro, 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 occu rrence o . In certain embodiments, R 1 is phenyl substituted by cyano, halogen, and 2 occurrences of R 4 . In certain embodiments, R 1 is selected from the groups depicted in the compounds in Table 1 below. [0089] As defined generally above, R 2 represents independently for each occurrence C 1-4 alkyl. In certain embodiments, R 2 is C1-2 alkyl. In certain embodiments, R 2 is methyl. In certain embodiments, R 2 is selected from the groups depicted in the compounds in Table 1 below. [0090] As defined generally above, R 3 is hydrogen or C 1-4 alkyl. In certain embodiments, R3 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. [0091] As defined generally above, R 4 is C 1-4 alkyl. In certain embodiments, R 4 is CH 3 . In certain embodiments, R 4 is selected from the groups depicted in the compounds in Table 1 below. [0092] As defined generally above, R 5 represents independently for each occurrence C 1-4 alkyl or halogen; or one occurrence of R 5 is taken together with R 3 to form a C 1-3 alkylene. In certain embodiments, R 5 represents independently for each occurrence C 1-4 alkyl or halogen. In certain embodiments, R 5 is C 1-4 alkyl. In certain embodiments, R 5 is halogen. In certain embodiments, one occurrence of R 5 is taken together with R 3 to form a C 1-3 alkylene. In certain embodiments, R 5 is selected from the groups depicted in the compounds in Table 1 below. [0093] As defined generally above, A 1 is a pyridazinylene, pyrimidinylene, pyrazinylene, or pyridinylene, each of which is substituted with n occurrences of R 5 . In certain embodments, 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 is . In certain embodiments, A 1 is pyridinylene substituted with n occurrences of R 5 . In certain embodiments, A 1 is pyridinylene substituted with 0 occurrences of R 5 . In certain embodiments, A 1 is , where ** is the point of attachment to L. In certain embodiments, A 1 is selected from the groups depicted in the compounds in Table 1 below. [0094] As defined generally above, A 2 is [0095] In certain embodiments, A 2 is . In certain embodiments, A 2 is . In certain embodiments, A 2 is . In certain embodiments, A 2 is . In certain embodiments, A 2 is . N In certain embodiments, A 2 is Cl . In certain embodiments, A 2 is . In certain embodiments, . In certain embodiments, A 2 is . [0096] In certain embodiments, A 2 is . In certain embodiments, A 2 is . In certain embodiments, A 2 is , wherein q is 1. In certain embodiments, A 2 is or . In certain embodiments, A 2 is . In certain embodiments, A 2 is . In certain embodiments, A 2 is one of the following:

. [0097] In certain embodiments, A 2 is selected from the groups depicted in the compounds in Table 1 below. [0098] As defined generally above, R 1A is C 1-4 alkyl or C 3-4 cycloalkyl. In certain embodiments, R 1A is C 1-4 alkyl. In certain embodiments, is methyl. In certain embodiments, R 1A is C 3-4 cycloalkyl. In certain embodiments, R 1A is selected from the groups depicted in the compounds in Table 1 below. [0099] As defined generally above, R 2A represents independently for each occurrence C 1-4 alkyl or C 3-4 cycloalkyl. In certain embodiments, R 2A is C 1-4 alkyl. In certain embodiments, R 2A is methyl. In certain embodiments, R 2A is C 3-4 cycloalkyl. In certain embodiments, R 2A is selected from the groups depicted in the compounds in Table 1 below. [0100] As defined generally above, R 3A is phenyl substituted with 1, 2, or 3 substituents independently selected from halo, C 1 -C 4 alkyl, or C 1 -C 4 haloalkyl. In certain embodiments, R 3A is phenyl substituted with halo. In certain embodiments, R 3A is phenyl substituted with 1 substituent selected from halo, C 1 -C 4 alkyl, or C 1 -C 4 haloalkyl. In certain embodiments, R 3A is phenyl substituted with 2 substituents independently selected from halo, C 1 -C 4 alkyl, or C 1 -C 4 haloalkyl. In certain embodiments, R 3A is phenyl substituted with 3 substituents independently selected from halo, C 1 -C 4 alkyl, or C 1 -C 4 haloalkyl. In certain embodiments, R 3A is selected from the groups depicted in the compounds in Table 1 below. [0101] As defined generally above, R 4A is -(C 1-6 alkylene)-C(O)N(R 5A )(R 6A ), -(C 1-6 alkylene)- N(R 5A )C(O)R 7A , -(C 1-6 alkylene)-CO 2 R 8A , -(C 1-6 alkylene)-OC(O)R 7A , -(C 1-6 alkylene)-CN, -(C1- 6 alkylene)-O-(C 1-6 alkyl), C 1-6 alkyl, C 3-6 cycloalkyl, or -(C 0-6 alkylene)-(5-6 membered heteroaryl containing 1, 2, 3, or 4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, wherein the heteroaryl is substituted with 0, 1, or 2 occurrences of C 1-6 alkyl). In certain embodiments, R 4A is -(C 1-6 alkylene)-C(O)N(R 5A )(R 6A ). In certain embodiments, R 4A is - (C 1-6 alkylene)-CO 2 R 8A . In certain embodiments, R 4A is -(C 1-6 alkylene)-N(R 5A )C(O)R 7A . In certain embodiments, R 4A is -(C 1-6 alkylene)-OC(O)R 7A . In certain embodiments, R 4A is -(C 1-6 alkylene)-CN. In certain embodiments, R 4A is -(C 1-6 alkylene)-O-(C 1-6 alkyl). In certain embodiments, R 4A is C 1-6 alkyl. In certain embodiments, R 4A is C 3-6 cycloalkyl. In certain embodiments, R 4A is -(C 0-6 alkylene)-(5-6 membered heteroaryl containing 1, 2, 3, or 4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, wherein the heteroaryl is substituted with 0, 1, or 2 occurrences of C 1-6 alkyl). In certain embodiments, R 4A is -(C 0-6 alkylene)-(5-6 membered heteroaryl containing 1, 2, 3, or 4 heteroatoms independently selected from nitrogen, oxygen, and sulfur). In certain embodiments, R 4A is -(C 1-3 alkylene)-(5-membered heteroaryl containing 1 or 2 heteroatoms independently selected from nitrogen, oxygen, and sulfur). In certain embodiments, R 4A is -(C 1-3 alkylene)-(oxazolyl). In certain embodiments, R4A is selected from the groups depicted in the compounds in Table 1 below. [0102] As defined generally above, R 5A and R 6A are independently hydrogen, C 1-6 alkyl, or C 3-6 cycloalkyl; or R 5A and R 6A are taken together with the nitrogen atom to which they are attached to form a 3-7 membered ring containing 1 nitrogen atom. In certain embodiments, R 5A and R 6A are independently hydrogen, C 1-6 alkyl, or C 3-6 cycloalkyl. In certain embodiments, R 5A is hydrogen. In certain embodiments, R 5A is C 1-6 alkyl. In certain embodiments, R 5A is C 3-6 cycloalkyl. In certain embodiments, R 6A is hydrogen. In certain embodiments, R 6A is C 1-6 alkyl. In certain embodiments, R 6A is C 3-6 cycloalkyl. In certain embodiments, R 5A and R 6A are taken together with the nitrogen atom to which they are attached to form a 3-7 membered ring containing 1 nitrogen atom. In certain embodiments, R 5A and R 6A are taken together with the nitrogen atom to which they are attached to form a 3-membered ring containing 1 nitrogen atom. In certain embodiments, R 5A and R 6A are taken together with the nitrogen atom to which they are attached to form a 4-membered ring containing 1 nitrogen atom. In certain embodiments, R 5A and R 6A are taken together with the nitrogen atom to which they are attached to form a 5- membered ring containing 1 nitrogen atom. In certain embodiments, R 5A and R 6A are taken together with the nitrogen atom to which they are attached to form a 6-membered ring containing 1 nitrogen atom. In certain embodiments, R 5A and R 6A are taken together with the nitrogen atom to which they are attached to form a 7-membered ring containing 1 nitrogen atom. In certain embodiments, R 5A is selected from the groups depicted in the compounds in Table 1 below. In certain embodiments, R 6A is selected from the groups depicted in the compounds in Table 1 below. [0103] As defined generally above, R 7A is C 1-6 alkyl, -(C 1-6 alkylene)-(C 3-6 cycloalkyl), or C 3-6 cycloalkyl. In certain embodiments, R 7A is C 1-6 alkyl. In certain embodiments, R 7A is -(C 1-6 alkylene)-(C 3-6 cycloalkyl). In certain embodiments, R 7A is C 3-6 cycloalkyl. In certain embodiments, R 7A is selected from the groups depicted in the compounds in Table 1 below. [0104] As defined generally above, R 8A is hydrogen, C 1-6 alkyl, -(C 1-6 alkylene)-(C 3-6 cycloalkyl), or C 3-6 cycloalkyl. In certain embodiments, R 8A is hydrogen. In certain embodiments, R 8A is C 1-6 alkyl. In certain embodiments, R 8A is -(C 1-6 alkylene)-(C 3-6 cycloalkyl). In certain embodiments, R 8A is C 3-6 cycloalkyl. In certain embodiments, R 8A is selected from the groups depicted in the compounds in Table 1 below. [0105] As defined generally above, R 9A is halo. In certain embodiments, R 9A is fluoro, chloro, or bromo. In certain embodiments, R 9A is fluoro. In certain embodiments, R 9A is selected from the groups depicted in the compounds in Table 1 below. [0106] As defined generally above, R 10A represents independently for each occurrence C 1-4 alkyl or -N(R 11A ) 2 . In certain embodiments, R 10A is C 1-4 alkyl. In certain embodiments, R 10A is N(R 11A ) 2 . In certain embodiments, R 10A is N(H)CH 3 . In certain embodiments, R 10A is selected from the groups depicted in the compounds in Table 1 below. [0107] As defined generally above, R 11A represents independently for each occurrence hydrogen or C 1-4 alkyl. In certain embodiments, R 11A represents independently for each occurrence C 1-4 alkyl. In certain embodiments, R 11A is hydrogen. In certain embodiments, R 11A is C 1-4 alkyl. In certain embodiments, R 11A represents independently for each occurrence hydrogen or methyl. In certain embodiments, R 11A is selected from the groups depicted in the compounds in Table 1 below. [0108] As defined generally above, R 12A is -(C 0-6 alkylene)-(5-6 membered heteroaryl containing 1, 2, 3, or 4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, wherein the heteroaryl is substituted with 0, 1, or 2 occurrences of C 1-6 alkyl). In certain embodiments, R 12A is -(C 0-6 alkylene)-(5-6 membered heteroaryl containing 1, 2, 3, or 4 heteroatoms independently selected from nitrogen, oxygen, and sulfur). In certain embodiments, R 12A is -(5-membered heteroaryl containing 1 or 2 heteroatoms independently selected from nitrogen, oxygen, and sulfur). In certain embodiments, R 12A is oxazolyl. In certain embodiments, R 12A is selected from the groups depicted in the compounds in Table 1 below. [0109] As defined generally above, k is 1, 2, 3, or 4. In certain embodiments, k is 1. In certain embodiments, k is 2. In certain embodiments, k is 3. In certain embodiments, k is 4. In certain embodiments, k is selected from the corresponding value in the groups depicted in the compounds in Table 1 below. [0110] As defined generally above, m, n, p, and q 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, 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, q is 0. In certain embodiments, q is 1. In certain embodiments, q is 2. In certain embodiments, s is 0. In certain embodiments, s is 1. In certain embodiments, s is 2. In certain embodiments, p 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. In certain embodiments, q is selected from the corresponding value in the groups depicted in the compounds in Table 1 below. [0111] As defined generally above, s is 1 or 2. In certain embodiments, s is 1. In certain embodiments, s is 2. In certain embodiments, s is selected from the corresponding value in the groups depicted in the compounds in Table 1 below. [0112] In certain embodiments, the compound of Formula I is further defined by Formula Ia or a pharmaceutically acceptable salt thereof: (1a). In certain embodiments, the definition of variables R 2 , R 3 , A 1 , and A 2 is one of the embodiments described above in connection with Formula I. [0113] In certain embodiments, the compound of Formula I is further defined by Formula Ib or a pharmaceutically acceptable salt thereof: (1b). In certain embodiments, the definition of variables R 2 , R 3 , A 1 , and A 2 is one of the embodiments described above in connection with Formula I. [0114] In certain embodiments, the compound of Formula I is further defined by Formula Ic or a pharmaceutically acceptable salt thereof: (1c). In certain embodiments, the definition of variables variables A 1 and A 2 is one of the embodiments described above in connection with Formula I. [0115] 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. [0116] In certain embodiments, the compound is a compound of Formula Ie or a pharmaceutically acceptable salt thereof: 1e wherein L is (i) Ψ-(8-10 membered spirocyclic saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen)-O-, where Ψ is the point of attachment to the pyrimidinylene in Formula Ie or (ii) a 9-11 membered spirocyclic saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen, wherein the heterocyclic ring is substituted with 0 or 1 occurrences of C 1-4 alkyl. [0117] In certain embodiments, L is Ψ-(8-10 membered spirocyclic saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen)-O-, where Ψ is the point of attachment to the pyrimidinylene in Formula Ie. In certain embodiments, L is Ψ-(9-membered spirocyclic saturated heterocyclic ring containing 1 heteroatom selected from nitrogen)-O-, where Ψ is the point of attachment to the pyrimidinylene in Formula Ie. [0118] In certain embodiments, L is a 9-11 membered spirocyclic saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen. In certain embodiments, L is a 10- membered spirocyclic saturated heterocyclic ring containing 1 heteroatom selected from nitrogen. [0119] In certain embodiments, the compound is a compound of Formula If or a pharmaceutically acceptable salt thereof: [0120] In certain embodiments, the compound is a compound of Formula Ig or a pharmaceutically acceptable salt thereof: 1g. [0121] In certain embodiments, the compound is a compound of Formula Ih or a pharmaceutically acceptable salt thereof: wherein L is (i) Ψ-(8-10 membered spirocyclic saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen)-O-, where Ψ is the point of attachment to the pyrimidinylene in Formula Ie or (ii) a 9-11 membered spirocyclic saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen, wherein the heterocyclic ring is substituted with 0 or 1 occurrences of C 1-4 alkyl. [0122] In certain embodiments, L is Ψ-(8-10 membered spirocyclic saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen)-O-, where Ψ is the point of attachment to the pyrimidinylene in Formula Ih. In certain embodiments, L is Ψ-(9-membered spirocyclic saturated heterocyclic ring containing 1 heteroatom selected from nitrogen)-O-, where Ψ is the point of attachment to the pyrimidinylene in Formula Ih. [0123] In certain embodiments, L is a 9-11 membered spirocyclic saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen. In certain embodiments, L is a 10- membered spirocyclic saturated heterocyclic ring containing 1 heteroatom selected from nitrogen. [0124] In certain embodiments, the compound is a compound of Formula Ii or a pharmaceutically acceptable salt thereof: [0125] In certain embodiments, the compound is a compound of Formula Ij or a pharmaceutically acceptable salt thereof:

1j. [0126] 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: Compounds of Formula I-1 [0127] One aspect of the invention provides a compound represented by Formula I-1: or a pharmaceutically ac ceptable salt thereof; wherein: R 1 is phenyl substituted by cyano, halogen, and m occurrences of R 4 ; R 2 represents independently for each occurrence C 1-4 alkyl; R 3 is hydrogen or C 1-4 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, or pyridinylene, each of which is substituted with n occurrences of R 5 ; A 2 is one of the following:

R 1A is C 1-4 alkyl or C 3-4 cycloalkyl; R 2A represents independently for each occurrence C 1-4 alkyl or C 3-4 cycloalkyl; R 3A is phenyl substituted with 1, 2, or 3 substituents independently selected from halo, C 1 -C 4 alkyl, or C 1 -C 4 haloalkyl; R 4A is -(C 1-6 alkylene)-C(O)N(R 5A )(R 6A ), -(C 1-6 alkylene)-N(R 5A )C(O)R 7A , -(C 1-6 alkylene)-CO 2 R 8A , -(C 1-6 alkylene)-OC(O)R 7A , or -(C 0-6 alkylene)-(5-6 membered heteroaryl containing 1, 2, or 3 heteroatoms independently selected from nitrogen, oxygen, and sulfur); R 5A and R 6A are independently hydrogen, C 1-6 alkyl, or C 3-6 cycloalkyl; or R 5A and R 6A are taken together with the nitrogen atom to which they are attached to form a 3-7 membered ring containing 1 nitrogen atom; R 7A is C 1-6 alkyl, -(C 1-6 alkylene)-(C 3-6 cycloalkyl), or C 3-6 cycloalkyl; R 8A is hydrogen, C 1-6 alkyl, -(C 1-6 alkylene)-(C 3-6 cycloalkyl), or C 3-6 cycloalkyl; L is a linker; k is 1, 2, 3, or 4; and m, n, and p are independently 0, 1, or 2. [0128] 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). [0129] In certain embodiments, the compound is a compound of Formula I. [0130] 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 phenyl substituted by cyano, chloro, 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 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 selected from the groups depicted in the compounds in Table 1 below. [0131] As defined generally above, R 2 represents independently for each occurrence C 1-4 alkyl. In certain embodiments, R 2 is C1-2 alkyl. In certain embodiments, R 2 is methyl. In certain embodiments, R 2 is selected from the groups depicted in the compounds in Table 1 below. [0132] As defined generally above, R 3 is hydrogen or C 1-4 alkyl. In certain embodiments, R3 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. [0133] As defined generally above, R 4 is C 1-4 alkyl. In certain embodiments, R 4 is CH 3 . In certain embodiments, R 4 is selected from the groups depicted in the compounds in Table 1 below. [0134] As defined generally above, R 5 represents independently for each occurrence C 1-4 alkyl or halogen. In certain embodiments, R 5 is C 1-4 alkyl. In certain embodiments, R 5 is halogen. In certain embodiments, R 5 is selected from the groups depicted in the compounds in Table 1 below. [0135] As defined generally above, A 1 is a pyridazinylene, pyrimidinylene, pyrazinylene, or pyridinylene, each of which is substituted with n occurrences of R 5 . In certain embodments, 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 is . In certain embodiments, A 1 is pyridinylene substituted with n occurrences of R 5 . In certain embodiments, A 1 is pyridinylene substituted with 0 occurrences of R 5 . In certain embodiments, A 1 is , where ** is the point of attachment to L. In certain embodiments, A 1 is selected from the groups depicted in the compounds in Table 1 below. [0136] As defined generally above, A 2 is [0137] In certain embodiments, A 2 is . In certain embodiments, A 2 is 1 A N N R . In certain embodiments, A 2 is . In certain embodiments, A s . n certain embodiments, A 2 is , , , or . In certain embodiments, A 2 is . [0138] In certain embodiments, A 2 is selected from the groups depicted in the compounds in Table 1 below. [0139] As defined generally above, R 1A is C 1-4 alkyl or C 3-4 cycloalkyl. In certain embodiments, R 1A is C 1-4 alkyl. In certain embodiments, is methyl. In certain embodiments, R 1A is C 3-4 cycloalkyl. In certain embodiments, R 1A is selected from the groups depicted in the compounds in Table 1 below. [0140] As defined generally above, R 2A represents independently for each occurrence C 1-4 alkyl or C 3-4 cycloalkyl. In certain embodiments, R 2A is C 1-4 alkyl. In certain embodiments, R 2A is methyl. In certain embodiments, R 2A is C 3-4 cycloalkyl. In certain embodiments, R 2A is selected from the groups depicted in the compounds in Table 1 below. [0141] As defined generally above, R 3A is phenyl substituted with 1, 2, or 3 substituents independently selected from halo, C 1 -C 4 alkyl, or C 1 -C 4 haloalkyl. In certain embodiments, R 3A is phenyl substituted with halo. In certain embodiments, R 3A is phenyl substituted with 1 substituent selected from halo, C 1 -C 4 alkyl, or C 1 -C 4 haloalkyl. In certain embodiments, R 3A is phenyl substituted with 2 substituents independently selected from halo, C 1 -C 4 alkyl, or C 1 -C 4 haloalkyl. In certain embodiments, R 3A is phenyl substituted with 3 substituents independently selected from halo, C 1 -C 4 alkyl, or C 1 -C 4 haloalkyl. In certain embodiments, R 3A is selected from the groups depicted in the compounds in Table 1 below. [0142] As defined generally above, R 4A is -(C 1-6 alkylene)-C(O)N(R 5A )(R 6A ), -(C 1-6 alkylene)- N(R 5A )C(O)R 7A , -(C 1-6 alkylene)-CO 2 R 8A , -(C 1-6 alkylene)-OC(O)R 7A , or -(C 0-6 alkylene)-(5-6 membered heteroaryl containing 1, 2, or 3 heteroatoms independently selected from nitrogen, oxygen, and sulfur). In certain embodiments, R 4A is -(C 1-6 alkylene)-C(O)N(R 5A )(R 6A ). In certain embodiments, R 4A is -(C 1-6 alkylene)-CO 2 R 8A . In certain embodiments, R 4A is -(C 1-6 alkylene)-N(R 5A )C(O)R 7A . In certain embodiments, R 4A is -(C 1-6 alkylene)-OC(O)R 7A . In certain embodiments, R 4A is -(C 0-6 alkylene)-(5-6 membered heteroaryl containing 1, 2, or 3 heteroatoms independently selected from nitrogen, oxygen, and sulfur). In certain embodiments, R 4A is -(C 1-3 alkylene)-(5-membered heteroaryl containing 1 or 2 heteroatoms independently selected from nitrogen, oxygen, and sulfur). In certain embodiments, R 4A is -(C 1-3 alkylene)-(oxazolyl). In certain embodiments, R 4A is selected from the groups depicted in the compounds in Table 1 below. [0143] As defined generally above, R 5A and R 6A are independently hydrogen, C 1-6 alkyl, or C 3-6 cycloalkyl; or R 5A and R 6A are taken together with the nitrogen atom to which they are attached to form a 3-7 membered ring containing 1 nitrogen atom. In certain embodiments, R 5A and R 6A are independently hydrogen, C 1-6 alkyl, or C 3-6 cycloalkyl. In certain embodiments, R 5A is hydrogen. In certain embodiments, R 5A is C 1-6 alkyl. In certain embodiments, R 5A is C 3-6 cycloalkyl. In certain embodiments, R 6A is hydrogen. In certain embodiments, R 6A is C 1-6 alkyl. In certain embodiments, R 6A is C 3-6 cycloalkyl. In certain embodiments, R 5A and R 6A are taken together with the nitrogen atom to which they are attached to form a 3-7 membered ring containing 1 nitrogen atom. In certain embodiments, R 5A and R 6A are taken together with the nitrogen atom to which they are attached to form a 3-membered ring containing 1 nitrogen atom. In certain embodiments, R 5A and R 6A are taken together with the nitrogen atom to which they are attached to form a 4-membered ring containing 1 nitrogen atom. In certain embodiments, R 5A and R 6A are taken together with the nitrogen atom to which they are attached to form a 5- membered ring containing 1 nitrogen atom. In certain embodiments, R 5A and R 6A are taken together with the nitrogen atom to which they are attached to form a 6-membered ring containing 1 nitrogen atom. In certain embodiments, R 5A and R 6A are taken together with the nitrogen atom to which they are attached to form a 7-membered ring containing 1 nitrogen atom. In certain embodiments, R 5A is selected from the groups depicted in the compounds in Table 1 below. In certain embodiments, R 6A is selected from the groups depicted in the compounds in Table 1 below. [0144] As defined generally above, R 7A is C 1-6 alkyl, -(C 1-6 alkylene)-(C 3-6 cycloalkyl), or C 3-6 cycloalkyl. In certain embodiments, R 7A is C 1-6 alkyl. In certain embodiments, R 7A is -(C 1-6 alkylene)-(C 3-6 cycloalkyl). In certain embodiments, R 7A is C 3-6 cycloalkyl. In certain embodiments, R 7A is selected from the groups depicted in the compounds in Table 1 below. [0145] As defined generally above, R 8A is hydrogen, C 1-6 alkyl, -(C 1-6 alkylene)-(C 3-6 cycloalkyl), or C 3-6 cycloalkyl. In certain embodiments, R 8A is hydrogen. In certain embodiments, R 8A is C 1-6 alkyl. In certain embodiments, R 8A is -(C 1-6 alkylene)-(C 3-6 cycloalkyl). In certain embodiments, R 8A is C 3-6 cycloalkyl. In certain embodiments, R 8A is selected from the groups depicted in the compounds in Table 1 below. [0146] As defined generally above, k is 1, 2, 3, or 4. In certain embodiments, k is 1. In certain embodiments, k is 2. In certain embodiments, k is 3. In certain embodiments, k is 4. In certain embodiments, k is selected from the corresponding value in the groups depicted in the compounds in Table 1 below. [0147] As defined generally above, m, n, and p 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, p 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. [0148] In certain embodiments, the compound of Formula I-1 is further defined by Formula Ia-1 or a pharmaceutically acceptable salt thereof: (1a-1). In certain embodiments, the definition of variables R 2 , R 3 , A 1 , and A 2 is one of the embodiments described above in connection with Formula I. [0149] In certain embodiments, the compound of Formula I is further defined by Formula Ib- 1 or a pharmaceutically acceptable salt thereof: (1b-1). In certain embodiments, the definition of variables R 2 , R 3 , A 1 , and A 2 is one of the embodiments described above in connection with Formula I-1. [0150] In certain embodiments, the compound of Formula I is further defined by Formula Ic or a pharmaceutically acceptable salt thereof: In certain embodiments, the definition of variables variables A 1 and A 2 is one of the embodiments described above in connection with Formula I-1. [0151] In certain embodiments, the compound of Formula I is further defined by Formula 1d- 1 or a pharmaceutically acceptable salt thereof: (1d-1). 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. [0152] 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* [0153] 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 represents independently for each occurrence C 1-4 alkyl; R 3 is hydrogen or C 1-4 alkyl; R 4 is C 1-4 alkyl; R 5 represents independently for each occurrence C 1-4 alkyl or halogen; or one occurrence of R 5 is taken together with R 3 to form a C 1-3 alkylene; A 1 is a pyridazinylene, pyrimidinylene, pyrazinylene, or pyridinylene, each of which is substituted with n occurrences of R 5 ; A 2 is ; R 1A is C 1-4 alkyl or C 3-4 cycloalkyl; R 2A represents independently for each occurrence C 1-4 alkyl or C 3-4 cycloalkyl; R 3A is phenyl substituted with 1, 2, or 3 substituents independently selected from halo, C 1 -C 4 alkyl, or C 1 -C 4 haloalkyl; R 4A is C 1-6 hydroxyalkyl, C 1-4 haloalkyl, -(C 1-6 alkylene)-C(O)N(R 5A )(R 6A ), -(C 1-6 alkylene)-N(R 5A )C(O)R 7A , -(C 1-6 alkylene)-CO 2 R 8A , -(C 1-6 alkylene)-OC(O)R 7A , -(C 1-6 alkylene)-CN, -(C 1-6 alkylene)-O-(C 1-6 alkyl), C 1-6 alkyl, C 3-6 cycloalkyl, or -(C 0-6 alkylene)- (5-6 membered heteroaryl containing 1, 2, 3, or 4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, wherein the heteroaryl is substituted with 0, 1, or 2 occurrences of C 1-6 alkyl); R 5A and R 6A are independently hydrogen, C 1-6 alkyl, or C 3-6 cycloalkyl; or R 5A and R 6A are taken together with the nitrogen atom to which they are attached to form a 3-7 membered ring containing 1 nitrogen atom; R 7A is C 1-6 alkyl, -(C 1-6 alkylene)-(C 3-6 cycloalkyl), or C 3-6 cycloalkyl; R 8A is hydrogen, C 1-6 alkyl, -(C 1-6 alkylene)-(C 3-6 cycloalkyl), or C 3-6 cycloalkyl; R 9A is halo; R 10A is hydrogen or C 1-4 alkyl; or R 4A and R 10A are taken together with the carbon atom to which they are attached to form a 3-5 membered saturated carbocylic ring; L is a linker; k is 1, 2, 3, or 4; and m, n, p, and q are independently 0, 1, or 2. [0154] 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). [0155] In certain embodiments, the compound is a compound of Formula I*. [0156] 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 phenyl substituted by cyano, chloro, 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 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 selected from the groups depicted in the compounds in Table 1 below. [0157] As defined generally above, R 2 represents independently for each occurrence C 1-4 alkyl. In certain embodiments, R 2 is C 1-2 alkyl. In certain embodiments, R 2 is methyl. In certain embodiments, R 2 is selected from the groups depicted in the compounds in Table 1 below. [0158] As defined generally above, R 3 is hydrogen or C 1-4 alkyl. In certain embodiments, R3 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. [0159] As defined generally above, R 4 is C 1-4 alkyl. In certain embodiments, R 4 is CH 3 . In certain embodiments, R 4 is selected from the groups depicted in the compounds in Table 1 below. [0160] As defined generally above, R 5 represents independently for each occurrence C 1-4 alkyl or halogen; or one occurrence of R 5 is taken together with R 3 to form a C 1-3 alkylene. In certain embodiments, R 5 represents independently for each occurrence C 1-4 alkyl or halogen. In certain embodiments, R 5 is C 1-4 alkyl. In certain embodiments, R 5 is halogen. In certain embodiments, one occurrence of R 5 is taken together with R 3 to form a C 1-3 alkylene. In certain embodiments, R 5 is selected from the groups depicted in the compounds in Table 1 below. [0161] As defined generally above, A 1 is a pyridazinylene, pyrimidinylene, pyrazinylene, or pyridinylene, each of which is substituted with n occurrences of R 5 . In certain embodments, A 1 is pyridazinylene substituted with n occurrences of R 5 . In certain embodiments, A 1 is . 1 5 In certain embodiments, A is pyrimidinylene substituted with n occurrences of R . 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 pyraz ny ene 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 pyridinylene substituted with 0 occurrences of R 5 . In certain embodiments, A 1 is , where ** is the point of attachment to L. In certain embodiments, A 1 is selected from the groups depicted in the compounds in Table 1 below. [0162] In certain embodiments, A 2 is one of the following: [0163] As defined generally above, R 1A is C 1-4 alkyl or C 3-4 cycloalkyl. In certain embodiments, R 1A is C 1-4 alkyl. In certain embodiments, is methyl. In certain embodiments, R 1A is C 3-4 cycloalkyl. In certain embodiments, R 1A is selected from the groups depicted in the compounds in Table 1 below. [0164] As defined generally above, R 2A represents independently for each occurrence C 1-4 alkyl or C 3-4 cycloalkyl. In certain embodiments, R 2A is C 1-4 alkyl. In certain embodiments, R 2A is methyl. In certain embodiments, R 2A is C 3-4 cycloalkyl. In certain embodiments, R 2A is selected from the groups depicted in the compounds in Table 1 below. [0165] As defined generally above, R 3A is phenyl substituted with 1, 2, or 3 substituents independently selected from halo, C 1 -C 4 alkyl, or C 1 -C 4 haloalkyl. In certain embodiments, R 3A is phenyl substituted with halo. In certain embodiments, R 3A is phenyl substituted with 1 substituent selected from halo, C 1 -C 4 alkyl, or C 1 -C 4 haloalkyl. In certain embodiments, R 3A is phenyl substituted with 2 substituents independently selected from halo, C 1 -C 4 alkyl, or C 1 -C 4 haloalkyl. In certain embodiments, R 3A is phenyl substituted with 3 substituents independently selected from halo, C 1 -C 4 alkyl, or C 1 -C 4 haloalkyl. In certain embodiments, R 3A is selected from the groups depicted in the compounds in Table 1 below. [0166] As defined generally above, R 4A is C 1-6 hydroxyalkyl, C 1-4 haloalkyl, -(C 1-6 alkylene)- C(O)N(R 5A )(R 6A ), -(C 1-6 alkylene)-N(R 5A )C(O)R 7A , -(C 1-6 alkylene)-CO 2 R 8A , -(C 1-6 alkylene)- OC(O)R 7A , -(C 1-6 alkylene)-CN, -(C 1-6 alkylene)-O-(C 1-6 alkyl), C 1-6 alkyl, C 3-6 cycloalkyl, or - (C 0-6 alkylene)-(5-6 membered heteroaryl containing 1, 2, 3, or 4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, wherein the heteroaryl is substituted with 0, 1, or 2 occurrences of C 1-6 alkyl). In certain embodiments, R 4A is -(C 1-6 alkylene)-C(O)N(R 5A )(R 6A ). In certain embodiments, R 4A is -(C 1-6 alkylene)-CO 2 R 8A . In certain embodiments, R 4A is -(C 1-6 alkylene)-N(R 5A )C(O)R 7A . In certain embodiments, R 4A is -(C 1-6 alkylene)-OC(O)R 7A . In certain embodiments, R 4A is -(C 1-6 alkylene)-CN. In certain embodiments, R 4A is -(C 1-6 alkylene)-O-(C 1- 6 alkyl). In certain embodiments, R 4A is C 1-6 alkyl. In certain embodiments, R 4A is C 3-6 cycloalkyl. In certain embodiments, R 4A is -(C 0-6 alkylene)-(5-6 membered heteroaryl containing 1, 2, 3, or 4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, wherein the heteroaryl is substituted with 0, 1, or 2 occurrences of C 1-6 alkyl). In certain embodiments, R4A is -(C 0-6 alkylene)-(5-6 membered heteroaryl containing 1, 2, 3, or 4 heteroatoms independently selected from nitrogen, oxygen, and sulfur). In certain embodiments, R 4A is -(C 1-3 alkylene)-(5- membered heteroaryl containing 1 or 2 heteroatoms independently selected from nitrogen, oxygen, and sulfur). In certain embodiments, R 4A is -(C 1-3 alkylene)-(oxazolyl). In certain embodiments, R 4A is C 1-6 hydroxyalkyl. In certain embodiments, R 4A is C 1-4 haloalkyl. In certain embodiments, R 4A is selected from the groups depicted in the compounds in Table 1 below. [0167] As defined generally above, R 5A and R 6A are independently hydrogen, C 1-6 alkyl, or C 3-6 cycloalkyl; or R 5A and R 6A are taken together with the nitrogen atom to which they are attached to form a 3-7 membered ring containing 1 nitrogen atom. In certain embodiments, R 5A and R 6A are independently hydrogen, C 1-6 alkyl, or C 3-6 cycloalkyl. In certain embodiments, R 5A is hydrogen. In certain embodiments, R 5A is C 1-6 alkyl. In certain embodiments, R 5A is C 3-6 cycloalkyl. In certain embodiments, R 6A is hydrogen. In certain embodiments, R 6A is C 1-6 alkyl. In certain embodiments, R 6A is C 3-6 cycloalkyl. In certain embodiments, R 5A and R 6A are taken together with the nitrogen atom to which they are attached to form a 3-7 membered ring containing 1 nitrogen atom. In certain embodiments, R 5A and R 6A are taken together with the nitrogen atom to which they are attached to form a 3-membered ring containing 1 nitrogen atom. In certain embodiments, R 5A and R 6A are taken together with the nitrogen atom to which they are attached to form a 4-membered ring containing 1 nitrogen atom. In certain embodiments, R 5A and R 6A are taken together with the nitrogen atom to which they are attached to form a 5- membered ring containing 1 nitrogen atom. In certain embodiments, R 5A and R 6A are taken together with the nitrogen atom to which they are attached to form a 6-membered ring containing 1 nitrogen atom. In certain embodiments, R 5A and R 6A are taken together with the nitrogen atom to which they are attached to form a 7-membered ring containing 1 nitrogen atom. In certain embodiments, R 5A is selected from the groups depicted in the compounds in Table 1 below. In certain embodiments, R 6A is selected from the groups depicted in the compounds in Table 1 below. [0168] As defined generally above, R 7A is C 1-6 alkyl, -(C 1-6 alkylene)-(C 3-6 cycloalkyl), or C 3-6 cycloalkyl. In certain embodiments, R 7A is C 1-6 alkyl. In certain embodiments, R 7A is -(C 1-6 alkylene)-(C 3-6 cycloalkyl). In certain embodiments, R 7A is C 3-6 cycloalkyl. In certain embodiments, R 7A is selected from the groups depicted in the compounds in Table 1 below. [0169] As defined generally above, R 8A is hydrogen, C 1-6 alkyl, -(C 1-6 alkylene)-(C 3-6 cycloalkyl), or C 3-6 cycloalkyl. In certain embodiments, R 8A is hydrogen. In certain embodiments, R 8A is C 1-6 alkyl. In certain embodiments, R 8A is -(C 1-6 alkylene)-(C 3-6 cycloalkyl). In certain embodiments, R 8A is C 3-6 cycloalkyl. In certain embodiments, R 8A is selected from the groups depicted in the compounds in Table 1 below. [0170] As defined generally above, R 9A is halo. In certain embodiments, R 9A is fluoro, chloro, or bromo. In certain embodiments, R 9A is fluoro. In certain embodiments, R 9A is selected from the groups depicted in the compounds in Table 1 below. [0171] As defined generally above, R 10A is hydrogen or C 1-4 alkyl; or R 4A and R 10A are taken together with the carbon atom to which they are attached to form a 3-5 membered saturated carbocylic ring. In certain embodiments, R 10A is hydrogen. In certain embodiments, R 10A is C 1-4 alkyl. In certain embodiments, R 4A and R 10A are taken together with the carbon atom to which they are attached to form a 3-5 membered saturated carbocylic ring. In certain embodiments, R 4A and R 10A are taken together with the carbon atom to which they are attached to form a 3- membered saturated carbocylic ring. In certain embodiments, R 10A is selected from the groups depicted in the compounds in Table 1 below. [0172] As defined generally above, k is 1, 2, 3, or 4. In certain embodiments, k is 1. In certain embodiments, k is 2. In certain embodiments, k is 3. In certain embodiments, k is 4. In certain embodiments, k is selected from the corresponding value in the groups depicted in the compounds in Table 1 below. [0173] As defined generally above, m, n, p, and q 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, 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, q is 0. In certain embodiments, q is 1. In certain embodiments, q is 2. In certain embodiments, p 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. In certain embodiments, q is selected from the corresponding value in the groups depicted in the compounds in Table 1 below. [0174] 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 B: Compound of Formula II [0175] Another aspect of the invention provides a compound represented by Formula II: (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: 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 represents independently for each occurrence C 1-4 alkyl; R 3 is hydrogen or C 1-4 alkyl; R 4 is C 1-4 alkyl; R 5 represents independently for each occurrence C 1-4 alkyl or halogen; or one occurrence of R 5 is taken together with R 3 to form a C 1-3 alkylene; A 1 is a pyridazinylene, pyrimidinylene, pyrazinylene, or pyridinylene, each of which is substituted with n occurrences of R 5 ; L is a linker; EPL is a moiety that binds to BRD4; k is 1, 2, 3, or 4; and m and n are independently 0, 1, or 2. [0176] 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). [0177] In certain embodiments, the compound is a compound of Formula II. [0178] 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. [0179] As defined generally above, R 2 represents independently for each occurrence C 1-4 alkyl. In certain embodiments, R 2 is C1-2 alkyl. In certain embodiments, R 2 is methyl. In certain embodiments, R 2 is selected from the groups depicted in the compounds in Table 1 below. [0180] As defined generally above, R 3 is hydrogen or C 1-4 alkyl. In certain embodiments, R3 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. [0181] As defined generally above, R 5 represents independently for each occurrence C 1-4 alkyl or halogen; or one occurrence of R 5 is taken together with R 3 to form a C 1-3 alkylene. In certain embodiments, R 5 represents independently for each occurrence C 1-4 alkyl or halogen. In certain embodiments, R 5 is C 1-4 alkyl. In certain embodiments, R 5 is halogen. In certain embodiments, one occurrence of R 5 is taken together with R 3 to form a C 1-3 alkylene. In certain embodiments, R 5 is selected from the groups depicted in the compounds in Table 1 below. [0182] As defined generally above, A 1 is a pyridazinyl, pyrimidinyl, pyrazinyl, or pyridinyl, 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 certain embodiments, A 1 is selected from the groups depicted in the compounds in Table 1 below. [0183] As defined generally above, k is 1, 2, 3, or 4. In certain embodiments, k is 1. In certain embodiments, k is 2. In certain embodiments, k is 3. In certain embodiments, k is 4. In certain embodiments, k is selected from the corresponding value in the groups depicted in the compounds in Table 1 below. [0184] As defined generally above, m and n are independently 0, 1, or 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, 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. [0185] In certain embodiments, the TPL is that is substituted by one occurrence of R II-1A . [0186] In certain embodiments, the TPL is that is substituted by one occurrence of R II-1A . In certain embodiments, the TPL is . In certain embodiments, the TPL is . [0187] 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: or ; wherein R II-2A is a bond to L; R 1A is C 1-4 alkyl or C 3-4 cycloalkyl; R 2A represents independently for each occurrence C 1-4 alkyl or C 3-4 cycloalkyl; R 3A is phenyl substituted with 1, 2, or 3 substituents independently selected from halo, C 1 -C 4 alkyl, or C 1 -C 4 haloalkyl; R 4A is -(C 1-6 alkylene)-C(O)N(R 5A )(R 6A ), -(C 1-6 alkylene)-N(R 5A )C(O)R 7A , -(C 1-6 alkylene)-CO 2 R 8A , -(C 1-6 alkylene)-OC(O)R 7A , or -(C 0-6 alkylene)-(5-6 membered heteroaryl containing 1, 2, or 3 heteroatoms independently selected from nitrogen, oxygen, and sulfur); R 5A and R 6A are independently hydrogen, C 1-6 alkyl, or C 3-6 cycloalkyl; or R 5A and R 6A are taken together with the nitrogen atom to which they are attached to form a 3-7 membered ring containing 1 nitrogen atom; R 7A is C 1-6 alkyl, -(C 1-6 alkylene)-(C 3-6 cycloalkyl), or C 3-6 cycloalkyl; R 8A is hydrogen, C 1-6 alkyl, -(C 1-6 alkylene)-(C 3-6 cycloalkyl), or C 3-6 cycloalkyl; and p is 0, 1, or 2. [0188] 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; R 1A is C 1-4 alkyl or C 3-4 cycloalkyl; R 2A represents independently for each occurrence C 1-4 alkyl or C 3-4 cycloalkyl; R 3A is phenyl substituted with 1, 2, or 3 substituents independently selected from halo, C 1 -C 4 alkyl, or C 1 -C 4 haloalkyl; R 4A is -(C 1-6 alkylene)-C(O)N(R 5A )(R 6A ), -(C 1-6 alkylene)-N(R 5A )C(O)R 7A , -(C 1-6 alkylene)-CO 2 R 8A , -(C 1-6 alkylene)-OC(O)R 7A , -(C 1-6 alkylene)-CN, -(C 1-6 alkylene)-O-(C 1-6 alkyl), C 1-6 alkyl, C 3-6 cycloalkyl, or -(C 0-6 alkylene)-(5-6 membered heteroaryl containing 1, 2, 3, or 4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, wherein the heteroaryl is substituted with 0, 1, or 2 occurrences of C 1-6 alkyl); R 5A and R 6A are independently hydrogen, C 1-6 alkyl, or C 3-6 cycloalkyl; or R 5A and R 6A are taken together with the nitrogen atom to which they are attached to form a 3-7 membered ring containing 1 nitrogen atom; R 7A is C 1-6 alkyl, -(C 1-6 alkylene)-(C 3-6 cycloalkyl), or C 3-6 cycloalkyl; R 8A is hydrogen, C 1-6 alkyl, -(C 1-6 alkylene)-(C 3-6 cycloalkyl), or C 3-6 cycloalkyl; R 9A is halo; R 10A represents independently for each occurrence C 1-4 alkyl or -N(R 11A ) 2 ; R 11A represents independently for each occurrence hydrogen or C 1-4 alkyl; R 12A is -(C 0-6 alkylene)-(5-6 membered heteroaryl containing 1, 2, 3, or 4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, wherein the heteroaryl is substituted with 0, 1, or 2 occurrences of C 1-6 alkyl); p and q are independently is 0, 1, or 2; and s is 1 or 2. [0189] In certain embodiments, the EPL is , , , or , each of which is substituted by one occurrence of R II-2A , wherein R II-2A is a bond to L.

[0190] In certain embodiments, the EPL is , each of which is substituted by one occurrence of R II-2A , wherein R II-2A is a bond to L. [0191] In certain embodiments, the EPL is one of the following: N R 1A R 1A is C 1-4 alkyl or C 3-4 cycloalkyl; R 2A represents independently for each occurrence C 1-4 alkyl or C 3-4 cycloalkyl; R 3A is phenyl substituted with 1, 2, or 3 substituents independently selected from halo, C 1 -C 4 alkyl, or C 1 -C 4 haloalkyl; R 4A is -(C 1-6 alkylene)-C(O)N(R 5A )(R 6A ), -(C 1-6 alkylene)-N(R 5A )C(O)R 7A , -(C 1-6 alkylene)-CO 2 R 8A , or -(C 1-6 alkylene)-OC(O)R 7A ; R 5A and R 6A are independently hydrogen, C 1-6 alkyl, or C 3-6 cycloalkyl; or R 5A and R 6A are taken together with the nitrogen atom to which they are attached to form a 3-7 membered ring containing 1 nitrogen atom; R 7A is C 1-6 alkyl, -(C 1-6 alkylene)-(C 3-6 cycloalkyl), or C 3-6 cycloalkyl; R 8A is hydrogen, C 1-6 alkyl, -(C 1-6 alkylene)-(C 3-6 cycloalkyl), or C 3-6 cycloalkyl; and p is 0, 1, or 2 [0192] In certain embodiments, the EPL is . In certain embodiments, the EPL is . [0193] In certain embodiments, the EPL is . In certain embodiments, the EPL is , , or . [0194] In certain embodiments, the EPL is , , or . In certain embodiments, the EPL is , O N N N N N S , , or . [0195] As defined generally above, R 1A is C 1-4 alkyl or C 3-4 cycloalkyl. In certain embodiments, R 1A is C 1-4 alkyl. In certain embodiments, is methyl. In certain embodiments, R 1A is C 3-4 cycloalkyl. In certain embodiments, R 1A is selected from the groups depicted in the compounds in Table 1 below. [0196] As defined generally above, R 2A represents independently for each occurrence C 1-4 alkyl or C 3-4 cycloalkyl. In certain embodiments, R 2A is C 1-4 alkyl. In certain embodiments, R 2A is methyl. In certain embodiments, R 2A is C 3-4 cycloalkyl. In certain embodiments, R 2A is selected from the groups depicted in the compounds in Table 1 below. [0197] As defined generally above, R 3A is phenyl substituted with 1, 2, or 3 substituents independently selected from halo, C 1 -C 4 alkyl, or C 1 -C 4 haloalkyl. In certain embodiments, R 3A is phenyl substituted with halo. In certain embodiments, R 3A is phenyl substituted with 1 substituent selected from halo, C 1 -C 4 alkyl, or C 1 -C 4 haloalkyl. In certain embodiments, R 3A is phenyl substituted with 2 substituents independently selected from halo, C 1 -C 4 alkyl, or C 1 -C 4 haloalkyl. In certain embodiments, R 3A is phenyl substituted with 3 substituents independently selected from halo, C 1 -C 4 alkyl, or C 1 -C 4 haloalkyl. In certain embodiments, R 3A is selected from the groups depicted in the compounds in Table 1 below. [0198] As defined generally above, R 4A is -(C 1-6 alkylene)-C(O)N(R 5A )(R 6A ), -(C 1-6 alkylene)- N(R 5A )C(O)R 7A , -(C 1-6 alkylene)-CO 2 R 8A , -(C 1-6 alkylene)-OC(O)R 7A , -(C 1-6 alkylene)-CN, -(C 1- 6 alkylene)-O-(C 1-6 alkyl), C 1-6 alkyl, C 3-6 cycloalkyl, or -(C 0-6 alkylene)-(5-6 membered heteroaryl containing 1, 2, 3, or 4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, wherein the heteroaryl is substituted with 0, 1, or 2 occurrences of C 1-6 alkyl). In certain embodiments, R 4A is -(C 1-6 alkylene)-C(O)N(R 5A )(R 6A ). In certain embodiments, R 4A is - (C 1-6 alkylene)-CO 2 R 8A . In certain embodiments, R 4A is -(C 1-6 alkylene)-N(R 5A )C(O)R 7A . In certain embodiments, R 4A is -(C 1-6 alkylene)-OC(O)R 7A . In certain embodiments, R 4A is -(C 1-6 alkylene)-CN. In certain embodiments, R 4A is -(C 1-6 alkylene)-O-(C 1-6 alkyl). In certain embodiments, R 4A is C 1-6 alkyl. In certain embodiments, R 4A is C 3-6 cycloalkyl. In certain embodiments, R 4A is -(C 0-6 alkylene)-(5-6 membered heteroaryl containing 1, 2, 3, or 4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, wherein the heteroaryl is substituted with 0, 1, or 2 occurrences of C 1-6 alkyl). In certain embodiments, R 4A is -(C 0-6 alkylene)-(5-6 membered heteroaryl containing 1, 2, 3, or 4 heteroatoms independently selected from nitrogen, oxygen, and sulfur). In certain embodiments, R 4A is -(C 1-3 alkylene)-(5-membered heteroaryl containing 1 or 2 heteroatoms independently selected from nitrogen, oxygen, and sulfur). In certain embodiments, R 4A is -(C 1-3 alkylene)-(oxazolyl).. In certain embodiments, R4A is selected from the groups depicted in the compounds in Table 1 below. [0199] As defined generally above, R 5A and R 6A are independently hydrogen, C 1-6 alkyl, or C 3-6 cycloalkyl; or R 5A and R 6A are taken together with the nitrogen atom to which they are attached to form a 3-7 membered ring containing 1 nitrogen atom. In certain embodiments, R 5A and R 6A are independently hydrogen, C 1-6 alkyl, or C 3-6 cycloalkyl. In certain embodiments, R 5A is hydrogen. In certain embodiments, R 5A is C 1-6 alkyl. In certain embodiments, R 5A is C 3-6 cycloalkyl. In certain embodiments, R 6A is hydrogen. In certain embodiments, R 6A is C 1-6 alkyl. In certain embodiments, R 6A is C 3-6 cycloalkyl. In certain embodiments, R 5A and R 6A are taken together with the nitrogen atom to which they are attached to form a 3-7 membered ring containing 1 nitrogen atom. In certain embodiments, R 5A and R 6A are taken together with the nitrogen atom to which they are attached to form a 3-membered ring containing 1 nitrogen atom. In certain embodiments, R 5A and R 6A are taken together with the nitrogen atom to which they are attached to form a 4-membered ring containing 1 nitrogen atom. In certain embodiments, R 5A and R 6A are taken together with the nitrogen atom to which they are attached to form a 5- membered ring containing 1 nitrogen atom. In certain embodiments, R 5A and R 6A are taken together with the nitrogen atom to which they are attached to form a 6-membered ring containing 1 nitrogen atom. In certain embodiments, R 5A and R 6A are taken together with the nitrogen atom to which they are attached to form a 7-membered ring containing 1 nitrogen atom. In certain embodiments, R 5A is selected from the groups depicted in the compounds in Table 1 below. In certain embodiments, R 6A is selected from the groups depicted in the compounds in Table 1 below. [0200] As defined generally above, R 7A is C 1-6 alkyl, -(C 1-6 alkylene)-(C 3-6 cycloalkyl), or C 3-6 cycloalkyl. In certain embodiments, R 7A is C 1-6 alkyl. In certain embodiments, R 7A is -(C 1-6 alkylene)-(C 3-6 cycloalkyl). In certain embodiments, R 7A is C 3-6 cycloalkyl. In certain embodiments, R 7A is selected from the groups depicted in the compounds in Table 1 below. [0201] As defined generally above, R 8A is hydrogen, C 1-6 alkyl, -(C 1-6 alkylene)-(C 3-6 cycloalkyl), or C 3-6 cycloalkyl. In certain embodiments, R 8A is hydrogen. In certain embodiments, R 8A is C 1-6 alkyl. In certain embodiments, R 8A is -(C 1-6 alkylene)-(C 3-6 cycloalkyl). In certain embodiments, R 8A is C 3-6 cycloalkyl. In certain embodiments, R 8A is selected from the groups depicted in the compounds in Table 1 below. [0202] As defined generally above, R 9A is halo. In certain embodiments, R 9A is fluoro, chloro, or bromo. In certain embodiments, R 9A is fluoro. In certain embodiments, R 9A is selected from the groups depicted in the compounds in Table 1 below. [0203] As defined generally above, R 10A represents independently for each occurrence C 1-4 alkyl or -N(R 11A ) 2 . In certain embodiments, R 10A is C 1-4 alkyl. In certain embodiments, R 10A is N(R 11A ) 2 . In certain embodiments, R 10A is N(H)CH 3 . In certain embodiments, R 10A is selected from the groups depicted in the compounds in Table 1 below. [0204] As defined generally above, R 11A represents independently for each occurrence hydrogen or C 1-4 alkyl. In certain embodiments, R 11A represents independently for each occurrence C 1-4 alkyl. In certain embodiments, R 11A is hydrogen. In certain embodiments, R 11A is C 1-4 alkyl. In certain embodiments, R 11A represents independently for each occurrence hydrogen or methyl. In certain embodiments, R 11A is selected from the groups depicted in the compounds in Table 1 below. [0205] As defined generally above, R 12A is -(C 0-6 alkylene)-(5-6 membered heteroaryl containing 1, 2, 3, or 4 heteroatoms independently selected from nitrogen, oxygen, and sulfur, wherein the heteroaryl is substituted with 0, 1, or 2 occurrences of C 1-6 alkyl). In certain embodiments, R 12A is -(C 0-6 alkylene)-(5-6 membered heteroaryl containing 1, 2, 3, or 4 heteroatoms independently selected from nitrogen, oxygen, and sulfur). In certain embodiments, R 12A is -(5-membered heteroaryl containing 1 or 2 heteroatoms independently selected from nitrogen, oxygen, and sulfur). In certain embodiments, R 12A is oxazolyl. In certain embodiments, R 12A is selected from the groups depicted in the compounds in Table 1 below. [0206] In certain embodiments, p is 2. In certain embodiments, p is 1. In certain embodiments, p is 0. 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 0. In certain embodiments, q is 1. In certain embodiments, q is 2. In certain embodiments, q is selected from the corresponding value in the groups depicted in the compounds in Table 1 below. [0207] In certain embodiments, s is 1. In certain embodiments, s is 2. In certain embodiments, s is selected from the corresponding value in the groups depicted in the compounds in Table 1 below. [0208] 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. [0209] 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 of Formula I and II [0210] Compounds 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. [0211] In some embodiments, L is symmetrical. In some embodiments, L is asymmetric. In certain embodiments, L is a bond. [0212] 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(C 1-4 alkyl)-, -N(C3-5 cycloalkyl)-, -O-, -C(O)-, -OC(O)-, -C(O)O-, -S-, -S(O)-, -S(O) 2 -, -S(O) 2 N(H)-, -S(O) 2 N(C 1-4 alkyl)-, -S(O) 2 N(C 3-5 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(C3-5 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. [0213] In certain embodiments, L is a bivalent, saturated or unsaturated, straight or branched C 1-6 0 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. [0214] 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(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. [0215] In certain embodiments, L is a bivalent, saturated or unsaturated, straight or branched C 1-6 0 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(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-, -N(C3-7 cycloalkyl)-, 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. [0216] 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(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. [0217] 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(C 1-6 alkyl)-. [0218] 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. [0219] 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. [0220] 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. [0221] In certain embodiments, L is a bivalent, saturated or unsaturated, straight or branched C 1-4 5 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. [0222] 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 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. [0223] In certain embodiments, L has the formula -N(R)-(optionally substituted 3-20 membered heteroalkylene)p-CH 2 -C(O)-, wherein R is hydrogen or optionally substituted C1-C6 alkyl, and p is 0 or 1. [0224] In certain embodiments, L has the formula -N(R)-(3-20 membered heteroalkylene) p - CH 2 -C(O)-; wherein the 3-20 membered heteroalkylene is optionally substituted with 1, 2, 3, or 4 substituents independently selected from halogen, C1-C6 haloalkyl, C3-C6 cycloalkyl, hydroxyl, and cyano; R is hydrogen or optionally substituted C 1 -C 6 alkyl; and p is 0 or 1. [0225] In certain embodiments, L has the formula -N(R)-(3-20 membered heteroalkylene)p- CH 2 -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. [0226] 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(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. [0227] 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)-, -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. [0228] 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(C 1-6 alkyl)-. [0229] 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(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. [0230] 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 . [0231] In certain embodiments, L is -(3-7 membered, monocyclic, saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen)-(OCH 2 CH 2 ) 1-5 -O-***, wherein *** is the point of attachment to A 2 . [0232] In certain embodiments, 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 . [0233] In certain embodiments, L is -piperidinylene-(OCH 2 CH 2 ) 1-15 -O-***, wherein *** is the point of attachment to A 2 . [0234] 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 embodim e s, s , wherein *** is the point of attachment to A 2 . [0235] 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)-(OCH 2 CH 2 ) 1-15 -N(C 1-4 alkyl)C(O)-C 1-10 alkylene-***, -(3-7 membered, monocyclic, saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen)- (OCH 2 CH 2 ) 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)-C1-10 alkylene-***, wherein *** is the point of attachment to A 2 . [0236] 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 -N(H)C(O)-C 1-5 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)- (OCH 2 CH 2 ) 1-10 -C(O)N(H)-C 1-5 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)-C 1-5 alkylene-***, wherein *** is the point of attachment to A 2 . [0237] In certain embodiments, L is -piperidinylene-(OCH 2 CH 2 ) 1-5 -N(H)C(O)-C 1-5 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)-C 1-5 alkylene-***, wherein *** is the point of attachment to A 2 . [0238] 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)- (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 . [0239] In certain embodiments, L is -piperidinylene-(OCH 2 CH 2 ) 1-5 -***, -piperidinylene-(C 0-5 alkylene)-O-***, or -piperidinylene-(C 1-5 alkylene)-***, wherein *** is the point of attachment to A 2 . [0240] 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) C1-10 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)-. [0241] In certain embodiments, 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(C 1-4 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)-. [0242] 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-, -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)-. [0243] In certain embodiments, L is -(piperazinylene)-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-, (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)-. [0244] In certain embodiments, L is , wherein *** is the point of attachment to A 2 , and X 1 is (i) C1-10 alky ene w ere or 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)-(C 1-10 alkylene)-. [0245] 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(C 1-6 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-. [0246] 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 -O-. [0247] 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(C 1-6 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-. [0248] 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-. [0249] 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-. [0250] 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 -(OCH 2 CH 2 )1-10 where 1 CH 2 group is optionally replaced with - C(H)(C 3-6 cycloalkyl)-. [0251] 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. [0252] In certain embodiments, L is a -(8-10 membered spirocyclic saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen)-O-***, where *** is the point of attachment to A 2 . [0253] In certain embodiments, L is a -(9-membered spirocyclic saturated heterocyclic ring containing 1 heteroatom selected from nitrogen)-O-***, where *** is the point of attachment to A 2 . [0254] 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 C 1-10 alkylene, -O-, -N(H)-, -N(C 1-4 alkyl)-, or a bond. [0255] In certain embodiments, L is -(piperidinylene)-(C 1-5 alkylene)-(piperazinylene)-***, wherein *** is the point of attachment to A 2 . [0256] In certain embodiments, L is -(piperazinylene)-(azetidinylene)-*** or (azetidinylene)- (piperazinylene)-***, wherein *** is the point of attachment to A 2 . [0257] 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)-(C 2-6 alkylene)- N(H)C(O)-(C 1-6 alkylene)-***, -(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)-(C 2-6 alkylene)-N(C 1-6 alkyl)C(O)-(C 1-6 alkylene)-***, 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)-(C 2-6 alkylene)- C(O)N(H)-(C 1-6 alkylene)-***, -(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)-(C 2-6 alkylene)- C(O)N(C 1-6 alkyl)-(C 1-6 alkylene)-***, wherein X 3 is C1-10 alkylene, -O-, -N(H)-, -N(C 1-4 alkyl)-, or a bond. [0258] In certain embodiments, L is -(piperidinylene)-(C 1-5 alkylene)-(piperazinylene)-(C2-5 alkylene)-N(H)C(O)-(C 1-6 alkylene)-***, wherein *** is the point of attachment to A 2 . [0259] In certain embodiments, 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 -(C 3-6 cycloalkylene)- N(C 1-4 alkyl)-***, wherein *** is the point of attachment to A 2 , and X 3 is C1-10 alkylene, -O-, - N(H)-, -N(C 1-4 alkyl)-, or a bond. [0260] In certain embodiments, 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(C 1-4 alkyl)-, or a bond. [0261] In certain embodiments, L is -N(C 1-3 alkyl)-(C 2-7 alkylene)-N(H)C(O)-(C 1-6 alkylene)- ***, -N(C 1-3 alkyl)-(C2-7 alkylene)-N(C 1-3 alkyl)C(O)-(C 1-6 alkylene)-***, -N(H)-(C2-7 alkylene)- N(H)C(O)-(C 1-6 alkylene)-***, -N(H)-(C2-7 alkylene)-N(C 1-3 alkyl)C(O)-(C 1-6 alkylene)-***, - N(C 1-3 alkyl)-(C 2-7 alkylene)-C(O)N(H)-(C 1-6 alkylene)-***, -N(C 1-3 alkyl)-(C 2-7 alkylene)- C(O)N(C 1-3 alkyl)-(C 1-6 alkylene)-***, -N(H)-(C2-7 alkylene)-C(O)N(H)-(C 1-6 alkylene)-***, or - N(H)-(C2-7 alkylene)-C(O)N(C 1-3 alkyl)-(C 1-6 alkylene)-***, where *** is a point of attachment to A 2 . [0262] In certain embodiments, L is -N(C 1-3 alkyl)-[(C2-4 alkylene)-O-]2-8-(C 2-6 alkylene)- N(H)C(O)-(C 1-6 alkylene)-, -N(C 1-3 alkyl)-[(C2-4 alkylene)-O-]2-8-(C 2-6 alkylene)-N(C 1-3 alkyl)C(O)-(C 1-6 alkylene)-, -N(H)-[(C 2-4 alkylene)-O-] 2-8 -(C 2-6 alkylene)-N(H)C(O)-(C 1-6 alkylene)-, -N(H)-[(C 2-4 alkylene)-O-] 2-8 -(C 2-6 alkylene)-N(C 1-3 alkyl)C(O)-(C 1-6 alkylene)-, - N(C 1-3 alkyl)-[(C2-4 alkylene)-O-]2-8-(C 2-6 alkylene)-C(O)N(H)-(C 1-6 alkylene)-, -N(C 1-3 alkyl)- [(C2-4 alkylene)-O-]2-8-(C 2-6 alkylene)-C(O)N(C 1-3 alkyl)-(C 1-6 alkylene)-, or -N(H)-[(C2-4 alkylene)-O-] 2-8 -(C 2-6 alkylene)-C(O)N(H)-(C 1-6 alkylene)-, -N(H)-[(C 2-4 alkylene)-O-] 2-8 -(C 2-6 alkylene)-C(O)N(C 1-3 alkyl)-(C 1-6 alkylene)-, where *** is a point of attachment to A 2 . [0263] In certain embodiments, L is -N(CH 3 )-[(CH 2 CH 2 )-O-]2-8-(C 2-6 alkylene)-N(H)C(O)- (C 1-6 alkylene)-***, -N(CH 3 )-[(CH 2 CH 2 )-O-] 2-8 -(C 2-6 alkylene)-N(CH 3 )C(O)-(C 1-6 alkylene)-***, -N(H)-[(CH 2 CH 2 )-O-]2-8-(C 2-6 alkylene)-N(H)C(O)-(C 1-6 alkylene)-***, or -N(H)-[(CH 2 CH 2 )-O- ]2-8-(C 2-6 alkylene)-N(H)C(O)-(C 1-6 alkylene)-***, where *** is a point of attachment to A 2 . [0264] In certain embodiments, L has the formula –(C 0-12 alkylene)-(optionally substituted 3- 40 membered heteroalkylene)-(C 0-12 alkylene)-. [0265] In certain embodiments, L is one of the following: wherein *** is the point of attachment to A 2 . [0266] In certain embodiments, L is one of the following:

wherein *** is the point of attachment to A 2 . [0267] In certain embodiments, L is wherein *** is the point of attachment to A 2 . [0268] In certain embodiments, L is wherein *** is the point of attachment to A 2 . [0269] In certain embodiments, wherein L is -(8-12 membered spirocyclic heterocyclyl containing 1, 2 or 3 heteroatoms independently selected from nitrogen and oxygen)-(C 3-4 cycloalkylene)-***, -(8-12 membered spirocyclic heterocyclyl containing 1, 2 or 3 heteroatoms independently selected from nitrogen and oxygen)-(C 0-4 alkylene)-***, -(8-12 membered spirocyclic heterocyclyl containing 1, 2 or 3 heteroatoms independently selected from nitrogen and oxygen)-C(O)-(C 1-4 alkylene)-***, -(5-6 membered saturated monocyclic heterocyclylene containing 1 or 2 heteroatoms independently selected from nitrogen and oxygen)-(3-5 membered saturated monocyclic heterocyclylene containing 1 or 2 heteroatoms independently selected from nitrogen and oxygen)-***, -(5-6 membered saturated monocyclic heterocyclylene containing 1 or 2 heteroatoms independently selected from nitrogen and oxygen)-O-***, -(5-6 membered saturated monocyclic heterocyclylene containing 1 or 2 heteroatoms independently selected from nitrogen and oxygen)-(C 0–4 alkylene)-***, -(5-6 membered saturated monocyclic heterocyclylene containing 1 or 2 heteroatoms )-(C 0–4 alkylene)-O-***, -(8-12 membered spirocyclic heterocyclyl containing 1, 2 or 3 heteroatoms independently selected from nitrogen and oxygen)-C(O))-***, - (8-12 membered spirocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)- O-(C 0-6 alkylene)-***, -(8-12 membered spirocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-(C 1-6 alkylene)-O-(C 0-6 alkylene)-***, -(5-6 membered saturated monocyclic heterocyclylene containing 1 or 2 heteroatoms )-C(O)N(H)-(C 0-6 alkylene)-***, - (N(C 1-6 alkyl)-(C 0-6 alkylene)-C(O)N(H)-(C 0-6 alkylene)-***, -(8-12 membered spirocyclic heterocyclyl containing 1, 2 or 3 heteroatoms independently selected from nitrogen and oxygen)- (C 2-4 alkynylene)-***, -(5-6 membered saturated monocyclic heterocyclylene containing 1 or 2 heteroatoms independently selected from nitrogen and oxygen)-(C 1-6 alkyl)-(3-5 membered saturated monocyclic heterocyclylene containing 1 or 2 heteroatoms independently selected from nitrogen and oxygen)-***, -(5-6 membered saturated monocyclic heterocyclylene containing 1 or 2 heteroatoms selected from nitrogen)-C(O)N(H)-(C 0-6 alkylene)-N(H))-***, -(C(O)N(H)-(C0- 6 alkylene)-(5-6 membered saturated monocyclic heterocyclylene containing 1 or 2 heteroatoms )-***, -(5-6 membered saturated monocyclic heterocyclylene containing 1 or 2 heteroatoms independently selected from nitrogen and oxygen)-N(C 1-6 alkyl)-(5-6 membered saturated monocyclic heterocyclylene containing 1 or 2 heteroatoms independently selected from nitrogen and oxygen)-O-***, -C(O)-(5-6 membered saturated monocyclic heterocyclylene containing 1 or 2 heteroatoms selected from nitrogen)-C(O)-***, -(C 0-6 alkylene)-(5-6 membered saturated monocyclic heterocyclylene containing 1 or 2 heteroatoms selected from nitrogen)-C(O)-***, - (5-6 membered saturated monocyclic heterocyclylene containing 1 or 2 heteroatoms selected from nitrogen)-C(O)-(C 0-6 alkylene)***, -C(O)-(5-6 membered saturated monocyclic heterocyclylene containing 1 or 2 heteroatoms selected from nitrogen)-(C 1-6 alkylene)-***, - (C(O)N(H)-(C 1-6 alkylene)-C(O)N(H)-(C 0-6 alkylene)-***, or -(8-11 membered fused bicyclic heterocyclyl containing 1, 2 or 3 heteroatoms independently selected from nitrogen and oxygen)- (C 1-4 alkylene)-***, wherein *** is the point of attachment to A 2 . [0270] In certain embodiments, L is -(8-12 membered spirocyclic heterocyclyl containing 1, 2 or 3 heteroatoms independently selected from nitrogen and oxygen)-(C 3-4 cycloalkylene)-***. In certain embodiments, L is -(8-12 membered spirocyclic heterocyclyl containing 1, 2 or 3 heteroatoms independently selected from nitrogen and oxygen)-(C 1-4 alkylene)-***. In certain embodiments, L is -(8-12 membered spirocyclic heterocyclyl containing 1, 2 or 3 heteroatoms independently selected from nitrogen and oxygen)-C(O)-(C 1-4 alkylene)-***. In certain embodiments, L is -(5-6 membered saturated monocyclic heterocyclylene containing 1 or 2 heteroatoms independently selected from nitrogen and oxygen)-(3-5 membered saturated monocyclic heterocyclylene containing 1 or 2 heteroatoms independently selected from nitrogen and oxygen)-***. In certain embodiments, L is -(5-6 membered saturated monocyclic heterocyclylene containing 1 or 2 heteroatoms independently selected from nitrogen and oxygen)-O-***. In certain embodiments, L is -(5-6 membered saturated monocyclic heterocyclylene containing 1 or 2 heteroatoms independently selected from nitrogen and oxygen)-(C 0-4 alkylene)-***. In certain embodiments, L is -(5-6 membered saturated monocyclic heterocyclylene containing 1 or 2 heteroatoms )-(C 0–4 alkylene)-O-***. In certain embodiments, L is -(8-12 membered spirocyclic heterocyclyl containing 1, 2 or 3 heteroatoms independently selected from nitrogen and oxygen)-C(O))-***. In certain embodiments, L is -(8- 12 membered spirocyclic heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-O- (C 0-6 alkylene)-***. In certain embodiments, L is -(5-6 membered saturated monocyclic heterocyclylene containing 1 or 2 heteroatoms )-C(O)N(H)-(C 0-6 alkylene)-***. In certain embodiments, L is -(N(C 1-6 alkyl)-(C 0-6 alkylene)-C(O)N(H)-(C 0-6 alkylene)-***. In certain embodiments, L is -(8-12 membered spirocyclic heterocyclyl containing 1, 2 or 3 heteroatoms independently selected from nitrogen and oxygen)-(C 2-4 alkynylene)-***. In certain embodiments, L is -(5-6 membered saturated monocyclic heterocyclylene containing 1 or 2 heteroatoms independently selected from nitrogen and oxygen)-(C 1-6 alkyl)-(3-5 membered saturated monocyclic heterocyclylene containing 1 or 2 heteroatoms independently selected from nitrogen and oxygen)-***. In certain embodiments, L is -(5-6 membered saturated monocyclic heterocyclylene containing 1 or 2 heteroatoms selected from nitrogen)-C(O)N(H)-(C 0-6 alkylene)-N(H))-***. In certain embodiments, L is -(C(O)N(H)-(C 0-6 alkylene)-(5-6 membered saturated monocyclic heterocyclylene containing 1 or 2 heteroatoms )-***. In certain embodiments, L is -(5-6 membered saturated monocyclic heterocyclylene containing 1 or 2 heteroatoms independently selected from nitrogen and oxygen)-N(C 1-6 alkyl)-(5-6 membered saturated monocyclic heterocyclylene containing 1 or 2 heteroatoms independently selected from nitrogen and oxygen)-O-***. In certain embodiments, L is -C(O)-(5-6 membered saturated monocyclic heterocyclylene containing 1 or 2 heteroatoms selected from nitrogen)-C(O)-***. In certain embodiments, L is -(C 0-6 alkylene)-(5-6 membered saturated monocyclic heterocyclylene containing 1 or 2 heteroatoms selected from nitrogen)-C(O)-***. In certain embodiments, L is - (5-6 membered saturated monocyclic heterocyclylene containing 1 or 2 heteroatoms selected from nitrogen)-C(O)-(C 0-6 alkylene)***. In certain embodiments, L is -C(O)-(5-6 membered saturated monocyclic heterocyclylene containing 1 or 2 heteroatoms selected from nitrogen)-(C1- 6 alkylene)-***, -(C(O)N(H)-(C 1-6 alkylene)-C(O)N(H)-(C 0-6 alkylene)-***. In certain embodiments, L is -(8-11 membered fused bicyclic heterocyclyl containing 1, 2 or 3 heteroatoms independently selected from nitrogen and oxygen)-(C 1-4 alkylene)-***. [0271] In certain embodiments, -N(C 1-6 alkyl)-(5-6 membered saturated monocyclic heterocyclylene containing 1 or 2 heteroatoms selected from nitrogen)-C(O)-(C 0-6 alkylene)-***, -(5-6 membered saturated monocyclic heterocyclylene containing 1 or 2 heteroatoms selected from nitrogen)-(5-6 membered saturated monocyclic heterocyclylene containing 1 or 2 heteroatoms selected from nitrogen)-(C 0-4 alkylene)-O-***, -(C 0-6 alkylene)-N(H)C(O)N(H)-(C 0-6 alkylene)-***, -N(H)-(C 0-6 alkylene)-(5-6 membered saturated monocyclic heterocyclylene containing 1 or 2 heteroatoms selected from nitrogen)-(C 0-6 alkylene)-***, -(C 0-6 alkylene)-C(O)- (5-6 membered saturated monocyclic heterocyclylene containing 1 or 2 heteroatoms selected from nitrogen)-(C 0-6 alkylene)-***, -(C 0-6 alkylene)-(5-6 membered saturated monocyclic heterocyclylene containing 1 or 2 heteroatoms selected from nitrogen)-C(O)N(C 1-6 alkyl)-(C 0-6 alkylene)-***, -(C 0-6 alkylene)-(5-6 membered saturated monocyclic heterocyclylene containing 1 or 2 heteroatoms selected from nitrogen)-(C 0-6 alkylene)-N(C 1-6 alkyl)-(3-6 membered saturated monocyclic heterocyclylene containing 1 or 2 heteroatoms selected from nitrogen)-***, -(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)-(C 0-6 alkylene)-(5-6 membered heteroaryl containing 1 or 2 heteroatoms selected from nitrogen)-***, -(8-12 membered spirocyclic heterocyclyl substituted with 1 or 2 fluoro containing 1, 2 or 3 heteroatoms independently selected from nitrogen and oxygen)-(C 1-4 alkylene)-***, -(5-6 membered saturated monocyclic heterocyclylene containing 1 or 2 heteroatoms selected from nitrogen)-(C 0-4 alkylene)-(C 3-6 cycloalkylene)-(C 0-4 alkylene)-O-***, - (5-6 membered saturated monocyclic heterocyclylene containing 1 or 2 heteroatoms selected from nitrogen)-(C 0–4 alkylene)-(C 3-6 cycloalkylene)-(C 0–4 alkylene)-***, -(C 0–4 alkylene)-(8-12 membered spirocyclic heterocyclyl containing 1, 2 or 3 heteroatoms independently selected from nitrogen and oxygen)-***, -(C 0–4 alkylene)-(C 3-6 cycloalkylene)-(C2-4 alkynylene)-***, -(C 0–4 alkylene)-(8-10 membered fused bicyclic heterocyclyl substituted with 1 or two fluoro containing 1 or 2 heteroatoms selected from nitrogen)-(C 0-4 alkylene)-***, -(8-12 membered spirocyclic heterocyclyl containing 1, 2 or 3 heteroatoms independently selected from nitrogen and oxygen)-O-***, -(8-12 membered spirocyclic heterocyclyl containing 1, 2 or 3 heteroatoms independently selected from nitrogen and oxygen)-(C 0-4 alkylene)-(5-6 membered saturated monocyclic heterocyclylene containing 1 or 2 heteroatoms selected from nitrogen)-***, -(C 0-4 alkylene)-(4-6 membered saturated heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-(phenylene substituted with trifluoromethyl)-(C 0–4 alkylene)-N(H)-***, or is -(5-6 membered saturated monocyclic heterocyclylene containing 1 or 2 heteroatoms selected from nitrogen)-C(O)-(5-6 membered saturated monocyclic heterocyclylene containing 1 or 2 heteroatoms selected from nitrogen)-(C 0-6 alkylene)-***, -(C 3-6 cycloalkylene)-C(O)N(C 1-6 alkyl)(C 0-6 alkylene)-***, -(5-6 membered saturated monocyclic heterocyclylene containing 1 or 2 heteroatoms selected from nitrogen)-(phenylene substituted with 0 or 1 occurrence of methyl or halo)-(C 0-6 alkylene)-***, -(5-6 membered saturated monocyclic heterocyclylene containing 1 or 2 heteroatoms selected from nitrogen)-(C 0-6 alkylene)-(5-6 membered saturated monocyclic oxo-substituted heterocyclylene containing 1 or 2 heteroatoms selected from nitrogen)-(C 0-6 alkylene)-***, -(8-12 membered spirocyclic C 1-4 alkyl substituted heterocyclyl containing 1, 2 or 3 heteroatoms independently selected from nitrogen and oxygen)-(C 0-6 alkylene)-(O)0-1***, -(C2- 4 alkynylene)-(8-12 membered spirocyclic heterocyclyl containing 1, 2 or 3 heteroatoms independently selected from nitrogen and oxygen)-(C 0-4 alkylene)-***, -(C 0-4 alkylene)-(C 3-7 cycloalkylene)-(C2-4 alkynylene)-***, -(C 1-4 alkylene)-(8-12 membered spirocyclic heterocyclyl containing 1, 2 or 3 heteroatoms independently selected from nitrogen and oxygen)-(C 0–4 alkylene)-***, -(C 1-4 alkylene)-(5-7 membered saturated heterocyclyl containing 1, 2 or 3 heteroatoms independently selected from nitrogen and oxygen)-(C 0–4 alkylene)-***, -(C 0–4 alkylene)-(5-7 membered saturated heterocyclyl containing 1 or 2 heteroatoms independently selected from nitrogen and oxygen)-(C 2-4 alkenylene)-***, or -(C 0-4 alkylene)-(6-8 membered saturated heterocyclyl substituted with 1 or 2 fluoro containing 1 or 2 heteroatoms independently selected from nitrogen and oxygen)-(C 0–4 alkylene)-***, wherein *** is the point of attachment to A 2 . [0272] In certain embodiments, L is -N(C 1-6 alkyl)-(5-6 membered saturated monocyclic heterocyclylene containing 1 or 2 heteroatoms selected from nitrogen)-C(O)-(C 0-6 alkylene)-***. In certain embodiments, L is -(5-6 membered saturated monocyclic heterocyclylene containing 1 or 2 heteroatoms selected from nitrogen)-(5-6 membered saturated monocyclic heterocyclylene containing 1 or 2 heteroatoms selected from nitrogen)-(C 0–4 alkylene)-O-***. In certain embodiments, L is -(C 0-6 alkylene)-N(H)C(O)N(H)-(C 0-6 alkylene)-***. In certain embodiments, L is -N(H)-(C 0-6 alkylene)-(5-6 membered saturated monocyclic heterocyclylene containing 1 or 2 heteroatoms selected from nitrogen)-(C 0-6 alkylene)-***. In certain embodiments, L is -(C 0-6 alkylene)-C(O)-(5-6 membered saturated monocyclic heterocyclylene containing 1 or 2 heteroatoms selected from nitrogen)-(C 0-6 alkylene)-***. In certain embodiments, L is -(C 0-6 alkylene)-(5-6 membered saturated monocyclic heterocyclylene containing 1 or 2 heteroatoms selected from nitrogen)-C(O)N(C 1-6 alkyl)-(C 0-6 alkylene)-***. In certain embodiments, L is - (C 0-6 alkylene)-(5-6 membered saturated monocyclic heterocyclylene containing 1 or 2 heteroatoms selected from nitrogen)-(C 0-6 alkylene)-N(C 1-6 alkyl)-(3-6 membered saturated monocyclic heterocyclylene containing 1 or 2 heteroatoms selected from nitrogen)-***. In certain embodiments, L is -(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-***. In certain embodiments, L is -(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-***. In certain embodiments, L is -(8-12 membered spirocyclic heterocyclyl substituted with 1 or 2 fluoro containing 1, 2 or 3 heteroatoms independently selected from nitrogen and oxygen)-(C 1-4 alkylene)-***. In certain embodiments, L is -(5-6 membered saturated monocyclic heterocyclylene containing 1 or 2 heteroatoms selected from nitrogen)-(C 0–4 alkylene)-(C 3-6 cycloalkylene)-(C 0-4 alkylene)-O-***. In certain embodiments, L is -(C 0-4 alkylene)-(8-12 membered spirocyclic heterocyclyl containing 1, 2 or 3 heteroatoms independently selected from nitrogen and oxygen)-***. In certain embodiments, L is -(C 0–4 alkylene)-(C 3-6 cycloalkylene)- (C 2-4 alkynylene)-***. In certain embodiments, L is -(C 0-4 alkylene)-(8-10 membered fused bicyclic heterocyclyl substituted with 1 or two fluoro containing 1 or 2 heteroatoms selected from nitrogen)--(C 0–4 alkylene)-***. In certain embodiments, L is -(8-12 membered spirocyclic heterocyclyl containing 1, 2 or 3 heteroatoms independently selected from nitrogen and oxygen)- O-***. In certain embodiments, L is -(C 0-4 alkylene)-(4-6 membered saturated heterocyclyl containing 1 or 2 heteroatoms selected from nitrogen)-(phenylene substituted with trifluoromethyl)-(C 0–4 alkylene)-N(H)-***. Additional Exemplary Embodiments for L [0273] In certain embodiments, L is -N(H)-(C2-9 alkylene)-O-(C 1-6 alkylene)-C(O)-***, - N(H)-(C10-20 alkylene)-O-(C 1-6 alkylene)-C(O)-***, -N(H)-[(C2-4 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)-(C 7-15 alkylene)-C(O)-***, -N(H)-[(C 2-4 alkylene)-O-] 2-6 -(C 1-6 alkylene)-***, -N(H)- [(C2-4 alkylene)-O-]7-15-(C 1-6 alkylene)-***, -N(H)-(C2-9 alkylene)-O-(C 1-6 alkylene)-C(O)N(C 1-6 alkyl)-(C 1-6 alkylene)-***, -N(H)-(C2-9 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)-(C 1-6 alkylene)-***, -N(H)-[(C2-4 alkylene)-O-] 2-6 -(C 1-6 alkylene)-N(C 1-6 alkyl)-(C 1-6 alkylene)-***, or -N(H)-[(C2-4 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 . [0274] In certain embodiments, L is -N(H)-(C2-9 alkylene)-O-(C 1-6 alkylene)-C(O)-***, - N(H)-(C10-20 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)-[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)-(C2-9 alkylene)-O-(C 1-6 alkylene)-C(O)N(C 1-6 alkyl)-(C 1-6 alkylene)- ***, -N(H)-(C2-9 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-(C 1-6 alkylene)-N(C 1-6 alkyl)-(C 1-6 alkylene)-***, where *** is a point of attachment to A 2 . [0275] In certain embodiments, L is -N(H)-[(C2-4 alkylene)-O-] 2-6 -(C 1-6 alkylene)-C(O)-***, - N(H)-[(C2-4 alkylene)-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)-(C 7-15 alkylene)-***, -N(C 1-6 alkyl)-(C 2-6 alkylene)-***, -N(C 1-6 alkyl)-(C 7-15 alkylene)-***, -N(H)-[(C2-4 alkylene)-O-] 2-6 -(C 1-6 alkylene)-***, -N(H)-[(C2-4 alkylene)-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)-(C 1-6 alkylene)-(3-6 membered heterocycloalkylene)- (C 1-6 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 . [0276] In certain embodiments, L is -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)-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)-(C 7-15 alkylene)-***, -N(C 1-6 alkyl)-(C 2-6 alkylene)-***, -N(C 1-6 alkyl)-(C 7-15 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)-(C 1-6 alkylene)-(3-6 membered heterocycloalkylene)-(C 1-6 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 . [0277] In certain embodiments, L is -[(C2-4 alkylene)-O-] 2-6 -(C 1-6 alkylene)-***, -[(C2-4 alkylene)-O-] 7-15 -(C 1-6 alkylene)-***, -[(C 2-4 alkylene)-O-] 2-6 -(C 1-6 alkylene)-N(C 1-6 alkyl)(C 1-6 alkylene)-***, -[(C2-4 alkylene)-O-]7-15-(C 1-6 alkylene)-N(C 1-6 alkyl)(C 1-6 alkylene)-***, -[(C2-4 alkylene)-O-] 2-6 -(C 1-6 alkylene)-N(H)(C 1-6 alkylene)-***, -[(C2-4 alkylene)-O-]7-15-(C 1-6 alkylene)- N(H)(C 1-6 alkylene)-***, -(C 1-9 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)-[(C 2-4 alkylene)-O-] 2-6 -(C 1-6 alkylene)- ***, -(C1-9 alkylene)-N(H)C(O)-[(C2-4 alkylene)-O-] 2-6 -(C 1-6 alkylene)-***, -(C1-9 alkylene)- C(O)N(H)-[(C2-4 alkylene)-O-]7-15-(C 1-6 alkylene)-***, -(C1-9 alkylene)-N(H)C(O)-[(C2-4 alkylene)-O-] 7-15 -(C 1-6 alkylene)-***, -(C 1-9 alkylene)-C(O)N(H)-[(C 2-4 alkylene)-O-] 2-6 -(C 1-6 alkylene)-N(C 1-6 alkyl)-(C 1-6 alkylene)-***, -(C1-9 alkylene)-N(H)C(O)-[(C2-4 alkylene)-O-] 2-6 - (C 1-6 alkylene)-N(C 1-6 alkyl)-(C 1-6 alkylene)-***, -(C1-9 alkylene)-C(O)N(H)-[(C2-4 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(C 1-6 alkyl)-(C 1-6 alkylene)-***, where *** is a point of attachment to A 2 . [0278] 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-(C 1-6 alkylene)-N(C 1-6 alkyl)(C 1-6 alkylene)-***, -[CH 2 CH 2 -O-] 2-6 -(C 1-6 alkylene)- N(H)(C 1-6 alkylene)-***, -[CH 2 CH 2 -O-]7-15-(C 1-6 alkylene)-N(H)(C 1-6 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)-[CH 2 CH 2 -O-] 2-6 -(C 1-6 alkylene)-***, -(C1-9 alkylene)-N(H)C(O)-[CH 2 CH 2 - O-] 2-6 -(C 1-6 alkylene)-***, -(C1-9 alkylene)-C(O)N(H)-[CH 2 CH 2 -O-]7-15-(C 1-6 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)-***, -(C1-9 alkylene)-N(H)C(O)- [CH 2 CH 2 -O-] 2-6 -(C 1-6 alkylene)-N(C 1-6 alkyl)-(C 1-6 alkylene)-***, -(C1-9 alkylene)-C(O)N(H)- [CH 2 CH 2 -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)- [(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 . [0279] In certain embodiments, L is -N(H)-[(C2-4 alkylene)-O-] 2-6 -(C 1-6 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)-[(C2-4 alkylene)-O-]7-15-(C 1-6 alkylene)-N(H)-***, -N(C 1-6 alkyl)-[(C2-4 alkylene)-O-] 2-6 -(C 1-6 alkylene)-N(C 1-6 alkyl)-***, or -N(C 1-6 alkyl)-[(C2-4 alkylene)- O-] 7-15 -(C 1-6 alkylene)-N(C 1-6 alkyl)-***, where *** is a point of attachment to A 2 . [0280] 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(C 1-6 alkyl)-[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(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 . [0281] In some embodiments, L is one of the following:

wh erein a dashed bond indicates a point of attachment. [0282] In certain embodiments, L is -C(H)(R 100 )-, -C(R 100 ) 2 -, O, -N(R 101 )-, -S(O) 2 -, an optionally substituted C3-7 cycloalkylene, an optionally substituted C4-7 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. [0283] 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 C 3-7 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 C3-7 cycloalkylene)-, -Y 20 -(optionally substituted C4-7 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. [0284] In certain embodiments, L is one of the following: [0285] 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. [0286] In certain embodiments, L is one of the following: 20 20 20 [0287] 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, 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. [0288] 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. [0289] 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 -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, 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, R100 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. [0290] 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 C3-7 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. [0291] In certain embodiments, L is one of the following: [0292] 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 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, 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. [0293] In certain embodiments, L is one of the following:

[0294] 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 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. [0295] In certain embodiments, L is one of the following: [0296] 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. [0297] In certain embodiments, L is one of the following: [0298] 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. [0299] In certain embodiments, L is one of the following:

[0300] wherein U, V, W, X, Y, and Z 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, 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. [0301] In certain embodiments, L is one of the following: [0302] wherein X, Y, and Z are independently -C(R 100 )- or -N-; V and W 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 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. [0303] In certain embodiments, L is one of the following: [0304] 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 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. [0305] In certain embodiments, L is one of the following:

[0306] wherein T, U, V, W, X, Y, and Z 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 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. [0307] In certain embodiments, L is one of the following: [0308] wherein W, X, Y, and Z are independently -C(R 100 )- or -N-; U and V 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; 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. [0309] In certain embodiments, L is one of the following: [0310] wherein X, Y, and Z are independently -C(R 100 )- or -N-; U, V, and W 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; 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.

[0311] In certain embodiments, L is one of the following:

[0 ] vara es m, n, o, p, an q are n epen en y , , , 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20.

[0313] 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. [0314] In certain embodiments, L is one of the following:

wherein any m or n are independently 0, 1, 2, 3, 4, 5, or 6. [0315] In certain embodiments, L is one of the following:

[0316] wherein any m or n are independently 0, 1, 2, 3, 4, 5, or 6.

[0317] In certain embodiments, L is one of the following: [0318] In certain embodiments, L is one of the following:

[0319] In certain embodiments, L has the formula –(C 0-12 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-. [0320] In certain embodiments, L is -CH 2 CH 2 (OCH 2 CH 2 )-***, -CH 2 CH 2 (OCH 2 CH 2 ) 2 -***, - CH 2 CH 2 (OCH 2 CH 2 )3-***, -CH 2 CH 2 (OCH 2 CH 2 )4-***, -CH 2 CH 2 (OCH 2 CH 2 )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 -***, - CH 2 CH 2 (OCH 2 CH 2 )12-***, -CH 2 CH 2 (OCH 2 CH 2 )13-***, -CH 2 CH 2 (OCH 2 CH 2 )14-***, - CH 2 CH 2 (OCH 2 CH 2 )15-***, or -CH 2 CH 2 (OCH 2 CH 2 )16-20-***, where *** is a point of attachment to A 2 . [0321] In certain embodiments, L is -(C 2-20 alkylene)-(OCH 2 CH 2 ) 2-4 -(C 0-4 alkylene)-***, -(C 2- 20 alkylene)-(OCH 2 CH 2 )5-7-(C 0–4 alkylene)-***, -(C2-20 alkylene)-(OCH 2 CH 2 )8-10-(C 0–4 alkylene)- ***, -(C2-20 alkylene)-(OCH 2 CH 2 )11-13-(C 0–4 alkylene)-***, -(C2-20 alkylene)-(OCH 2 CH 2 )14-16-(C 0–4 alkylene)-***, -(C 2-20 alkylene)-(OCH 2 CH 2 ) 17-20 -(C 0-4 alkylene)-***, -(C 1-20 alkylene)- (OCH 2 CH 2 )1-10-(C 0–4 alkylene)-C(O)-***, or -(C1-20 alkylene)-(OCH 2 CH 2 )11-20-(C 0–4 alkylene)- C(O)-***, where *** is a point of attachment to A 2 . [0322] 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 - (C 0–4 alkylene)-***, -O(CH 2 CH 2 O)8-10-(C 0–4 alkylene)-***, -O(CH 2 CH 2 O)11-13-(C 0–4 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 . [0323] In certain embodiments, L is -(C0-20 alkylene)-(OCH 2 CH 2 )1-10-(N(C 1-4 alkyl))-***, - (C 0-20 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(C 1-4 alkyl))-(C0-10 alkylene)-***, or -(C0-20 alkylene)-(CH 2 CH 2 O)11-20-(C2-10 alkylene)-(N(C 1-4 alkyl))-(C0-10 alkylene)-***, where *** is a point of attachment to A 2 . [0324] In certain embodiments, L is selected from those depicted in the compounds in Table 1, below. Exemplary Specific Compounds [0325] 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 any one of compounds I-1 to I-48 in Table 1, or a pharmaceutically acceptable salt thereof. In certain embodiments, the compound is any one of compounds I-1 to I-48 in Table 1. In certain embodiments, the compound is any one of compounds I-1 to I-52 in Table 1, or a pharmaceutically acceptable salt thereof. In certain embodiments, the compound is any one of compounds I-1 to I-52 in Table 1. In certain embodiments, the compound is any one of compounds I-1 to I-236 in Table 1, or a pharmaceutically acceptable salt thereof. In certain embodiments, the compound is any one of compounds I-1 to I-236 in Table 1.

TABLE 1.

Synthetic Methods [0326] 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. [0327] 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. [0328] The synthetic route illustrated in Scheme 1 is a general method for preparing compounds of Formula F. Reaction of carboxylic acid A and amine B under amide coupling conditions provides amide C. Removal of the protection group (Pg) from compound C provides compound D. The Pg may be, for example, a Boc protecting group that can be removed by treating the compound with trifluoroacetic acid. Coupling of compound D with compound E (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 F. SCHEME 1. [0329] 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 [0330] 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 [0331] 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. [0332] 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. [0333] 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. [0334] 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. [0335] 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. [0336] 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. [0337] 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. [0338] 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. [0339] 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. [0340] 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. [0341] 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. [0342] 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 [0343] 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 compounds 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. [0344] 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 prostate cancer cell. Combination Therapies [0345] 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. [0346] Accordingly, in certain embodiments, the method further comprises administering to the subject an additional therapeutic agent that treats the disease contemplated herein. [0347] 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. [0348] 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. [0349] 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. [0350] 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. [0351] 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). [0352] 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.). [0353] 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). [0354] 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). [0355] 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). [0356] 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). [0357] 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). [0358] 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). [0359] 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). [0360] 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. [0361] 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). [0362] 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). [0363] 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). [0364] 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). [0365] 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). [0366] 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). [0367] 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). [0368] 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). [0369] 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). [0370] 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). [0371] 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). [0372] 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). [0373] 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). [0374] 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.” [0375] 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). [0376] 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). [0377] 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. [0378] 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 [0379] 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 μL complete medium. Following seeding, plates are spun at 300 × g^for three minutes and cultured at 37 ℃ 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 μL aliquot of compound/media mixture is added to cells to bring total volume in well to 50 μL. DMSO alone is used as a negative control. Plates are then spun at 300×g^for three minutes and stored at 37 ℃ with 5% CO 2 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 μL. 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 [0380] 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. [0381] 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. [0382] 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. [0383] 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. [0384] 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. [0385] 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. [0386] 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. [0387] 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. [0388] 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. [0389] 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. [0390] 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. [0391] 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. [0392] 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. [0393] 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. [0394] 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. [0395] 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. [0396] 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. [0397] 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. [0398] 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. [0399] 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. [0400] 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. [0401] Ophthalmic formulations, eye ointments, powders, solutions and the like, are also contemplated as being within the scope of this invention. [0402] 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. [0403] 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. [0404] 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. [0405] 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. [0406] 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. [0407] 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. [0408] 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. [0409] 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. [0410] 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. [0411] 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. [0412] 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. [0413] 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. [0414] 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. [0415] 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. [0416] 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. [0417] 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. [0418] 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 [0419] 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 [0420] 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 represents independently for each occurrence C 1-4 alkyl; R 3 is hydrogen or C 1-4 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, or pyridinylene, each of which is substituted with n occurrences of R 5 ; A 2 is one of the following: or ; R 1A is C 1 4 alkyl or C 3 4 cycloalkyl; R 2A represents independently for each occurrence C 1-4 alkyl or C 3-4 cycloalkyl; R 3A is phenyl substituted with 1, 2, or 3 substituents independently selected from halo, C 1 -C 4 alkyl, or C 1 -C 4 haloalkyl; R 4A is -(C 1-6 alkylene)-C(O)N(R 5A )(R 6A ), -(C 1-6 alkylene)-N(R 5A )C(O)R 7A , -(C 1-6 alkylene)-CO 2 R 8A , -(C 1-6 alkylene)-OC(O)R 7A , or -(C 0-6 alkylene)-(5-6 membered heteroaryl containing 1, 2, or 3 heteroatoms independently selected from nitrogen, oxygen, and sulfur); R 5A and R 6A are independently hydrogen, C 1-6 alkyl, or C 3-6 cycloalkyl; or R 5A and R 6A are taken together with the nitrogen atom to which they are attached to form a 3-7 membered ring containing 1 nitrogen atom; R 7A is C 1-6 alkyl, -(C 1-6 alkylene)-(C 3-6 cycloalkyl), or C 3-6 cycloalkyl; R 8A is hydrogen, C 1-6 alkyl, -(C 1-6 alkylene)-(C 3-6 cycloalkyl), or C 3-6 cycloalkyl; L is a linker; k is 1, 2, 3, or 4; and m, n, and p are independently 0, 1, or 2. Embodiment No.2. The compound of embodiment 1, wherein R 2 is methyl Embodiment No.3. The compound of embodiment 1 or 2, wherein R 3 is hydrogen. Embodiment No.4. The compound of any one of embodiments 1-3, wherein m is 0. Embodiment No.5. The compound of any one of embodiments 1-3, wherein R 1 is . Embodiment No.6. The compound of any one of embodiments 1-5, wherein k is 4. Embodiment No.7. The compound of any one of embodiments 1-6, wherein the compound is a compound of Formula I. Embodiment No.8. The compound of embodiment 1 or 2, wherein the compound is a compound of Formula Ia or a pharmaceutically acceptable salt thereof: Embodiment No.9. The compound of embodiment 1 or 2, wherein the compound is a compound of Formula Ib or a pharmaceutically acceptable salt thereof: . Embodiment No.10. The compound of embodiment 1, wherein the compound is a compound of Formula Ic or a pharmaceutically acceptable salt thereof: . Embodiment No.11. The compound of embodiment 1, wherein the compound is a compound of Formula Id or a pharmaceutically acceptable salt thereof: . Embodiment No.12. The compound of any one of embodiments 1-11, wherein A 1 is pyridazinylene substituted with n occurrences of R 5 . Embodiment No.13. The compound of any one of embodiments 1-11, wherein A 1 is . Embodiment No.14. The compound of any one of embodiments 1-11, wherein A 1 is pyrimidinylene substituted with n occurrences of R 5 . Embodiment No.15. The compound of any one of embodiments 1-11, wherein A 1 is , where ** is the point of attachment to L. Embodiment No.16. The compound of any one of embodiments 1-11, wherein A 1 is , where ** is the point of attachment to L. Embodiment No.17. The compound of any one of embodiments 1-11, wherein A 1 is pyrazinylene substituted with n occurrences of R 5 . Embodiment No.18. The compound of any one of embodiments 1-11, wherein A 1 is . Embodiment No.19. The compound of any one of embodiments 1-11, wherein A 1 is pyridinylene substituted with n occurrences of R 5 . Embodiment No.20. The compound of any one of embodiments 1-11, wherein A 1 is , where ** is the point of attachment to L. Embodiment No.21. The compound of any one of embodiments 1-12, 14, 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 R 3A is phenyl substituted with halo. 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 or 24, wherein R 4A is - (C 1-6 alkylene)-C(O)N(R 5A )(R 6A ). Embodiment No.26. The compound of any one of embodiments 1-21 or 24, wherein R 4A is - (C 1-6 alkylene)-CO 2 R 8A . Embodiment No.27. The compound of any one of embodiments 1-21 or 24, wherein R 4A is - (C 0-6 alkylene)-(5-6 membered heteroaryl containing 1, 2, or 3 heteroatoms independently selected from nitrogen, oxygen, and sulfur). Embodiment No.28. The compound of any one of embodiments 1-21 or 24, wherein R 4A is - (C 1-3 alkylene)-(5-membered heteroaryl containing 1 or 2 heteroatoms independently selected from nitrogen, oxygen, and sulfur). Embodiment No.29. The compound of any one of embodiments 1-21 or 24, wherein R 4A is - (C 1-3 alkylene)-(oxazolyl). Embodiment No.30. The compound of any one of embodiments 1-29, wherein R 1A is C 1-4 alkyl. Embodiment No.31. The compound of any one of embodiments 1-29, wherein R 1A is methyl. Embodiment No.32. The compound of any one of embodiments 1-31, wherein R 2A is C 1-4 alkyl. Embodiment No.33. The compound of any one of embodiments 1-31, wherein R 2A is methyl. Embodiment No.34. The compound of any one of embodiments 1-33, wherein p is 2. Embodiment No.35. The compound of any one of embodiments 1-21, wherein A 2 is . Embodiment No.36. The compound of any one of embodiments 1-21, wherein A 2 is . Embodiment No.37. The compound of any one of embodiments 1-21, wherein A 2 is . Embodiment No.38. The compound of any one of embodiments 1-21, wherein A 2 is . Embodiment No.39. The compound of any one of embodiments 1-38, 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(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. Embodiment No.40. The compound of any one of embodiments 1-38, 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(C 1-6 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. Embodiment No.41. The compound of any one of embodiments 1-38, 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(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(C 1-6 alkyl)-. Embodiment No.42. The compound of any one of embodiments 1-38, 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.43. The compound of any one of embodiments 1-38, 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.44. The compound of any one of embodiments 1-38, 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.45. The compound of any one of embodiments 1-38, 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.46. The compound of any one of embodiments 1-38, wherein L is - piperidinylene-(OCH 2 CH 2 ) 1-15 -O-***, wherein *** is the point of attachment to A 2 . Embodiment No.47. The compound of any one of embodiments 1-38, wherein L is , wherein *** is the point of attachment to A 2 . Embodiment No.48. The compound of any one of embodiments 1-38, wherein L is , wherein *** is the point of attachment to A 2 . Embodiment No.49. The compound of any one of embodiments 1-38, wherein L is , wherein *** is the point of attachment to A 2 . Embodiment No.50. The compound of any one of embodiments 1-38, 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)-(OCH 2 CH 2 ) 1-15 -N(C 1-4 alkyl)C(O)-C1-10 alkylene-***, -(3-7 membered, monocyclic, saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen)-(OCH 2 CH 2 ) 1-15 -C(O)N(H)-C 1-10 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.51. The compound of any one of embodiments 1-38, wherein L is - piperidinylene-(OCH 2 CH 2 ) 1-5 -N(H)C(O)-C 1-5 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)-C 1-5 alkylene-***, wherein *** is the point of attachment to A 2 . Embodiment No.52. The compound of any one of embodiments 1-38, wherein 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)-(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.53. The compound of any one of embodiments 1-38, wherein L is - piperidinylene-(OCH 2 CH 2 ) 1-5 -***, -piperidinylene-(C 0-5 alkylene)-O-***, or - piperidinylene-(C 1-5 alkylene)-***, wherein *** is the point of attachment to A 2 . Embodiment No.54. The compound of any one of embodiments 1-38, 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(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)-. Embodiment No.55. The compound of any one of embodiments 1-38, 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(C 1-4 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.56. The compound of any one of embodiments 1-38, 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(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)-(C 1-10 alkylene)-. Embodiment No.57. The compound of any one of embodiments 1-38, wherein L is - (piperazinylene)-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-, (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.58. The compound of any one of embodiments 1-38, 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.59. The compound of any one of embodiments 1-38, 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(C 1-6 alkyl)-. Embodiment No.60. The compound of any one of embodiments 1-38, 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(C 1-6 alkyl)-. Embodiment No.61. The compound of any one of embodiments 1-38, 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(C 1-6 alkyl)-. Embodiment No.62. The compound of any one of embodiments 1-38, wherein L is , wherein *** i 2 2 s the point of attachment to A , and X is -O-, -N(H)-, or -N(C 1-6 alkyl)-. Embodiment No.63. The compound of any one of embodiments 1-38, wherein L is , wherein *** is the point of attachment to A 2 , and X 2 is -O-, -N(H)-, or -N(C 1-6 alkyl)-. Embodiment No.64. The compound of any one of embodiments 59-63, wherein X 2 is -O-. Embodiment No.65. The compound of any one of embodiments 1-38, 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.66. The compound of any one of embodiments 1-38, 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.67. The compound of any one of embodiments 1-38, wherein L is a 7-8 membered spirocyclic or fused bicyclic saturated heterocyclic ring containing 2 heteroatoms selected from nitrogen. Embodiment No.68. The compound of any one of embodiments 1-38, wherein L is a -(8-10 membered spirocyclic saturated heterocyclic ring containing 1 or 2 heteroatoms selected from nitrogen)-O-***, where *** is the point of attachment to A 2 . Embodiment No.69. The compound of any one of embodiments 1-38, wherein L is a -(9- membered spirocyclic saturated heterocyclic ring containing 1 heteroatom selected from nitrogen)-O-***, where *** is the point of attachment to A 2 . Embodiment No.70. The compound of any one of embodiments 1-38, 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.71. The compound of any one of embodiments 1-38, wherein L is - (piperidinylene)-(C 1-5 alkylene)-(piperazinylene)-***, wherein *** is the point of attachment to A 2 . Embodiment No.72. The compound of any one of embodiments 1-38, wherein L is - (piperazinylene)-(azetidinylene)-*** or (azetidinylene)-(piperazinylene)-***, wherein *** is the point of attachment to A 2 . Embodiment No.73. The compound of any one of embodiments 1-38, 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)-(C 2-6 alkylene)-N(H)C(O)-(C 1-6 alkylene)-***, -(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)-(C 2-6 alkylene)-N(C 1-6 alkyl)C(O)-(C 1-6 alkylene)- ***, 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)-(C 2-6 alkylene)-C(O)N(H)-(C 1-6 alkylene)-***, -(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)-(C 2-6 alkylene)-C(O)N(C 1-6 alkyl)-(C 1-6 alkylene)-***, wherein X 3 is C 1-10 alkylene, -O-, -N(H)-, -N(C 1-4 alkyl)-, or a bond. Embodiment No.74. The compound of any one of embodiments 1-38, wherein L is - (piperidinylene)-(C 1-5 alkylene)-(piperazinylene)-(C 2-5 alkylene)-N(H)C(O)-(C 1-6 alkylene)-***, wherein *** is the point of attachment to A 2 . Embodiment No.75. The compound of any one of embodiments 1-38, 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 -(C 3-6 cycloalkylene)-N(C 1-4 alkyl)-***, wherein *** is the point of attachment to A 2 , and X 3 is C1-10 alkylene, -O-, - N(H)-, -N(C 1-4 alkyl)-, or a bond. Embodiment No.76. The compound of any one of embodiments 1-38, 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 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 1-38, wherein L is -N(C 1-3 alkyl)-(C2-7 alkylene)-N(H)C(O)-(C 1-6 alkylene)-***, -N(C 1-3 alkyl)-(C2-7 alkylene)-N(C 1-3 alkyl)C(O)-(C 1-6 alkylene)-***, -N(H)-(C 2-7 alkylene)-N(H)C(O)-(C 1-6 alkylene)-***, - N(H)-(C 2-7 alkylene)-N(C 1-3 alkyl)C(O)-(C 1-6 alkylene)-***, -N(C 1-3 alkyl)-(C 2-7 alkylene)- C(O)N(H)-(C 1-6 alkylene)-***, -N(C 1-3 alkyl)-(C2-7 alkylene)-C(O)N(C 1-3 alkyl)-(C 1-6 alkylene)-***, -N(H)-(C2-7 alkylene)-C(O)N(H)-(C 1-6 alkylene)-***, or -N(H)-(C2-7 alkylene)-C(O)N(C 1-3 alkyl)-(C 1-6 alkylene)-***, where *** is a point of attachment to A 2 . Embodiment No.78. The compound of any one of embodiments 1-38, wherein L is -N(C 1-3 alkyl)-[(C2-4 alkylene)-O-]2-8-(C 2-6 alkylene)-N(H)C(O)-(C 1-6 alkylene)-, -N(C 1-3 alkyl)- [(C 2-4 alkylene)-O-] 2-8 -(C 2-6 alkylene)-N(C 1-3 alkyl)C(O)-(C 1-6 alkylene)-, -N(H)-[(C 2-4 alkylene)-O-]2-8-(C 2-6 alkylene)-N(H)C(O)-(C 1-6 alkylene)-, -N(H)-[(C2-4 alkylene)-O-]2-8- (C 2-6 alkylene)-N(C 1-3 alkyl)C(O)-(C 1-6 alkylene)-, -N(C 1-3 alkyl)-[(C2-4 alkylene)-O-]2-8- (C 2-6 alkylene)-C(O)N(H)-(C 1-6 alkylene)-, -N(C 1-3 alkyl)-[(C 2-4 alkylene)-O-] 2-8 -(C 2-6 alkylene)-C(O)N(C 1-3 alkyl)-(C 1-6 alkylene)-, or -N(H)-[(C 2-4 alkylene)-O-] 2-8 -(C 2-6 alkylene)-C(O)N(H)-(C 1-6 alkylene)-, -N(H)-[(C 2-4 alkylene)-O-] 2-8 -(C 2-6 alkylene)- C(O)N(C 1-3 alkyl)-(C 1-6 alkylene)-, where *** is a point of attachment to A 2 . Embodiment No.79. The compound of any one of embodiments 1-38, wherein L is -N(CH 3 )- [(CH 2 CH 2 )-O-] 2-8 -(C 2-6 alkylene)-N(H)C(O)-(C 1-6 alkylene)-***, -N(CH 3 )-[(CH 2 CH 2 )-O- ]2-8-(C 2-6 alkylene)-N(CH 3 )C(O)-(C 1-6 alkylene)-***, -N(H)-[(CH 2 CH 2 )-O-]2-8-(C 2-6 alkylene)-N(H)C(O)-(C 1-6 alkylene)-***, or -N(H)-[(CH 2 CH 2 )-O-]2-8-(C 2-6 alkylene)- N(H)C(O)-(C 1-6 alkylene)-***, where *** is a point of attachment to A 2 . Embodiment No.80. The compound of any one of embodiments 1-38, wherein L has the formula –(C0-12 alkylene)-(optionally substituted 3-40 membered heteroalkylene)-(C0-12 alkylene)-. Embodiment No.81. The compound of any one of embodiments 1-38, wherein L is one of the following: wherein *** is the point of attachment to A 2 . Embodiment No.82. The compound of any one of embodiments 1-38, 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 1-38, wherein L is wherein *** is the point of attachment to A 2 . EXAMPLES [0421] 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 are not intended to limit the invention. General Methods [0322] 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 (CH 2 Cl2), 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. [0323] 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 DD2600 (600 MHz 1 H; 150 MHz 13 C) or Agilent DD2400 (400 MHz 1 H; 100 MHz 13 C) spectrometer at room temperature. Chemical shifts were reported in ppm relative to the residual CDCl 3 (δ 7.26 ppm 1 H; δ 77.0 ppm 13 C), CD 3 OD (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. [0324] 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. [0325] 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.

LC-MS Method 10

L M M h 2 LC-MS METHOD 40

LC-MS METHOD 50 [0326] 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 hexafluorophosphate; 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-[3-(3-chloro-4-cyano-phenoxy)-2,2,4,4-tetramethyl- cyclobutyl]-2-[2-[[2-[(9S)-7-(4-chlorophenyl)-4,5,13-trimeth yl-3-thia-1,8,11,12- tetrazatricyclo[8.3.0.02,6]trideca-2(6),4,7,10,12-pentaen-9- yl]acetyl]amino]ethyl-methyl- amino]pyrimidine-5-carboxamide (I-1) N 1.1 Synthesis of compound 3 [0327] To a solution of tert-butyl N-[2-(methylamino) ethyl] carbamate (0.50 g, 2.8 μmol, 1.0 equiv), ethyl 2-chloropyrimidine-5-carboxylate (535 mg, 2.80 μmol, 1.0 equiv) in MeCN (3 mL) was added K 2 CO 3 (396 mg, 2.80 μmol, 1.0 equiv). The mixture was stirred at 80 ℃ for 12 hours. After filtration, the filtrate was purified by prep-HPLC (column: Waters Xbridge C18150 x 50 mm, 10μm; mobile phase: [water (NH4HCO3)-ACN]; B%:35%-65%, 11 min) to afford ethyl 2- [2-(tert-butoxycarbonylamino)ethyl-methyl-amino]pyrimidine-5 -carboxylate (0.45 g, 48% yield) as a white solid. 1.2 Synthesis of compound 4 [0328] To a solution of ethyl 2-[2-(tert-butoxycarbonylamino) ethyl-methyl-amino] pyrimidine-5-carboxylate (300 mg, 924 μmol, 1 equiv) in THF (2 mL) and H 2 O (2 mL) was added LiOH.H 2 O (155 mg, 3.70 μmol, 4 equiv). The mixture was stirred at 25 ℃ for 0.5 hour. The pH was adjusted to 5~6 by HCl (1M). The resulting solution was concentrated to afford compound 2-[2-(tert-butoxycarbonylamino) ethyl-methyl-amino] pyrimidine-5-carboxylic acid (0.25 g, 91% yield) as a white solid. 1.3 Synthesis of compound 6 [0329] To a solution of 2-[2-(tert-butoxycarbonylamino)ethyl-methyl-amino]pyrimidine -5- carboxylic acid (120 mg, 405 μmol, 1.0 equiv) and 4-(3-amino-2,2,4,4-tetramethyl-cyclobutoxy)- 2-chloro-benzonitrile (238 mg, 607 μmol, 1.5 equiv, TFA salt) in DMF (1 mL) was added DIEA (157 mg, 1.20 μmol, 211 μL, 3.0 equiv) and HATU (169 mg, 445 μmol, 1.1 equiv). The mixture was stirred at 25 ℃ for 0.5 hour. The residue was purified by prep-HPLC (column: Waters Xbridge C18150 x 50 mm, 10 μm; mobile phase: [water(NH4HCO3)-ACN]; B%: 48%-78%, 11 min) to afford compound tert-butyl N-[2-[[5-[[3-(3-chloro-4-cyano-phenoxy)-2,2,4,4-tetramethyl -cyclobutyl]carbamoyl]pyrimidin-2-yl]-methyl-amino]ethyl]car bamate (0.18 g, 80% yield) as a white solid. 1.4 Synthesis of compound 7 [0330] To a solution of tert-butyl N-[2-[[5-[[3-(3-chloro-4-cyano-phenoxy)-2, 2, 4, 4- tetramethyl-cyclobutyl] carbamoyl] pyrimidin-2-yl]-methyl-amino] ethyl] carbamate (70 mg, 0.13μmol, 1.0 equiv) in DCM (2 mL) was added TFA (1 mL). The mixture was stirred at 25 ℃ for 0.5 hour. The reaction mixture was concentrated to afford compound 2-[2-aminoethyl (methyl)amino]-N-[3-(3-chloro-4-cyano-phenoxy)-2, 2, 4, 4-tetramethyl-cyclobutyl] pyrimidine- 5-carboxamide (71 mg, 98% yield, TFA salt) as a white solid. 1.5 Synthesis of compound I-1

[0331] To a solution of 2-[2-aminoethyl(methyl)amino]-N-[3-(3-chloro-4-cyano-phenoxy )- 2,2,4,4-tetramethyl-cyclobutyl]pyrimidine-5-carboxamide (65 mg, 0.11 μmol, 1.0 equiv, TFA salt) and 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]acetic acid (70 mg, 0.14 μmol, 1.2 equiv, TFA salt) in DMF (1 mL) was added HATU (47 mg, 0.13 μmol, 1.1 equiv) and DIEA (44 mg, 0.34 μmol, 60 μL, 3.0 equiv). The mixture was stirred at 25 ℃ for 0.5 hour. The residue was purified by prep- HPLC (column: Waters Xbridge 150 x 25 mm, 5 μm; mobile phase: [water(NH4HCO3)-ACN]; B%: 56%-86%, 9 min) to afford compound N-[3-(3-chloro-4-cyano-phenoxy)-2,2,4,4- tetramethyl-cyclobutyl]-2-[2-[[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]acetyl]amino ]ethyl-methyl- amino]pyrimidine-5-carboxamide (26 mg, 27% yield) as an off-white solid. 1 H NMR (400 MHz, CD 3 OD): 8.74 (s, 2 H), 7.72 (d, J = 8.63 Hz, 1 H), 7.48 - 7.38 (m, 4 H), 7.12 (d, J = 2.38 Hz, 1 H), 6.98 (m, 1 H), 4.68 - 4.68 (m, 4 H), 4.26 (s, 1 H), 4.12 (s, 1 H), 3.90 - 3.68 (m, 2 H), 3.65 - 3.52 (m, 2 H), 3.28 (s, 2 H), 2.69 (s, 3 H), 2.45 (s, 3 H), 1.70 (s, 3 H), 1.25 (s, 6 H), 1.20 (s, 6 H).

EXAMPLE 2 – Synthesis of N-[3-(3-chloro-4-cyano-phenoxy)-2,2,4,4-tetramethyl- cyclobutyl]-2-[2-[2-[2-[[2-[(9S)-7-(4-chlorophenyl)-4,5,13-t rimethyl-3-thia-1,8,11,12- tetrazatricyclo[8.3.0.02,6]trideca-2(6),4,7,10,12-pentaen-9- yl]acetyl]amino] ethoxy]ethoxy] ethyl-methylamino]pyrimidine-5-carboxamide (I-2) [0332] To a solution of 2-chloro-N-[3-(3-chloro-4-cyano-phenoxy)-2,2,4,4-tetramethyl - cyclobutyl]pyrimidine-5-carboxamide (200 mg, 476 μmol, 1.0 equiv) in NMP (3 mL) was added K 2 CO 3 (131 mg, 953 μmol, 2.0 equiv) and tert-butyl N-tert-butoxycarbonyl-N-[2-[2-[2- (methylamino)ethoxy]ethoxy]ethyl]carbamate (173 mg, 477 μmol, 1.0 equiv). The mixture was stirred at 50 ℃ for 2 hours and purified by prep-HPLC (column: Waters Xbridge C18150 x 50 mm, 10 μm; mobile phase: [water(NH 4 HCO 3 )-ACN]; B%: 56%-86%, 11 min) to give tert-butyl N-tert-butoxycarbonyl-N-[2-[2-[2-[[5-[[3-(3-chloro-4-cyano-p henoxy)-2,2,4,4- tetramethylcyclobutyl]carbamoyl]pyrimidin-2-yl]-methyl-amino ]ethoxy]ethoxy] ethyl]carbamate (280 mg, 79% yield) as a black brown gum. 1.2 Synthesis of compound 4 [0333] To a solution of tert-butyl N-tert-butoxycarbonyl-N-[2-[2-[2-[[5-[[3-(3-chloro-4-cyano -phenoxy)-2,2,4,4-tetramethylcyclobutyl]carbamoyl]pyrimidin- 2-yl]-methyl-amino]ethoxy] ethoxy]ethyl]carbamate (100 mg, 134 μmol, 1.0 equiv) in DCM (3 mL) was added TFA (500 μL). The mixture was stirred at 20 ℃ for 0.3 hour and concentrated to give 2-[2-[2-(2- aminoethoxy)ethoxy]ethyl-methyl-amino]-N-[3-(3-chloro-4-cyan o-phenoxy)-2,2,4,4- tetramethylcyclobutyl]pyrimidine-5-carboxamide (85 mg, 96% yield, TFA salt) as a yellow gum.

1.3 Synthesis of compound I-2 [0334] To a solution of 2-[2-[2-(2-aminoethoxy)ethoxy]ethyl-methyl-amino]-N-[3-(3-ch loro- 4-cyano-phenoxy)-2,2,4,4-tetramethylcyclobutyl]pyrimidine-5- carboxamide (85 mg, 129 μmol, 1.0 equiv, TFA salt) and 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]acetic acid (52 mg, 129 μmol, 1.0 equiv) in DMF (3 mL) was added HATU (59 mg, 154 μmol, 1.2 equiv) and DIEA (50 mg, 387 μmol, 3.0 equiv). The mixture was stirred at 20 ℃ for 0.5 hour and purified by prep-HPLC (column: Phenomenex luna C18250 x 50 mm, 15 μm; mobile phase: [water(FA)-ACN]; B%: 80%-100%, 10 min) to give N-[3-(3-chloro-4-cyano-phenoxy)-2,2,4,4-tetramethyl-cyclobut yl]-2- [2-[2-[2-[[2-[(9S)-7-(4-chlorophenyl)-4,5,13-trimethyl-3-thi a-1,8,11,12-tetrazatricyclo [8.3.0.02,6]trideca-2(6),4,7,10,12-pentaen-9-yl]acetyl]amino ]ethoxy]ethoxy]ethyl-methylamino] pyrimidine-5-carboxamide (60 mg, 49% yield) as an off-white solid. 1 H NMR (400 MHz, MeOD): δ 8.74 (s, 2H), 8.45-8.37 (m, 1H), 7.77 (d, J = 8.8 Hz, 1H), 7.73 (d, J = 8.8 Hz, 1H), 7.49-7.41 (m, 4H), 7.13 (d, J = 2.4 Hz, 1H), 7.00-6.98 (m, 1H), 4.67-4.65 (m, 1H), 4.27 (s, 1H), 4.14 (d, J = 8.8 Hz, 1H), 3.95-3.89 (m, 2H), 3.78-3.73 (m, 2H), 3.67-3.63 (m, 4H), 3.61-3.57 (m, 2H), 3.49-3.38 (m, 4H), 3.28 (s, 3H), 2.71 (s, 3H), 2.46 (s, 3H), 1.71 (s, 3H), 1.27 (s, 6H), 1.22 (s, 6H). EXAMPLE 3 – Synthesis of N-((1r,3r)-3-(3-chloro-4-cyanophenoxy)-2,2,4,4- tetramethylcyclobutyl)-2-((1-((S)-4-(4-chlorophenyl)-2,3,9-t rimethyl-6H-thieno[3,2-f] [1,2,4]triazolo[4,3-a][1,4]diazepin-6-yl)-2-oxo-6,9,12,15-te traoxa-3-azaheptadecan-17- yl)(methyl)amino)pyrimidine-5-carboxamide (I-3) [0335] To a solution of compound 1 (1.90 g, 4.34 μmol, 1.0 equiv) and TEA (878 mg, 8.69 μmol, 1.2 mL, 2.0 equiv) in DCM (25 mL) was added TosCl (827 mg, 4.34 μmol, 1.0 equiv). The mixture was stirred at 20 ℃ for 12 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO 2 , petroleum ether/ethyl acetate = 5/1 to 2/1) to afford compound 2 (1.88 g, 3.18 μmol, 73% yield) as a yellow oil. 1 H NMR (400 MHz, CDCl3): 7.81 (d, J = 8.3 Hz, 2H), 7.35 (d, J = 8.0 Hz, 2H), 4.19 - 4.14 (m, 2H), 3.81 - 3.77 (m, 2H), 3.71 - 3.67 (m, 2H), 3.63 - 3.58 (m, 14H), 2.45 (s, 3H), 1.50 (s, 18H). 1.2 Synthesis of compound 4 [0336] To a solution of compound 2 (1.80 g, 3.04 μmol, 1.0 equiv) and compound 3 (736 mg, 6.08 μmol, 784 μL, 2.0 equiv) in CH 3 CN (25 mL) was added K2CO3 (1.26 g, 9.12 μmol, 3.0 equiv). The mixture was stirred at 80 ℃ for 12 hours. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (column: Waters X bridge C18150 x 50 mm, 10 μm; mobile phase: [water(NH4HCO3)-ACN]; B%: 54%-84%, 11 min) to afford desired compound 4 (1.43 g, 2.64 μmol, 87% yield) as a yellow oil. 1 H NMR (400 MHz, CDCl 3 ): 7.31 (d, J = 4.2 Hz, 4H), 7.26 - 7.21 (m, 1H), 3.83 - 3.74 (m, 2H), 3.65 - 3.59 (m, 16H), 3.56 (s, 2H), 2.63 (t, J = 6.1 Hz, 2H), 2.26 (s, 3H), 1.50 (s, 18H). 1.3 Synthesis of compound 5 [0337] To a solution of compound 4 (600 mg, 1.11 μmol, 1.0 equiv) in TFE (8 mL) was added Pd (OH) 2 (155 mg) under N2 atmosphere. The suspension was degassed and purged with H 2 three times. The mixture was stirred under H 2 (15 psi) at 20 ℃ for 12 hours. The reaction mixture was filtered and concentrated under reduced pressure to afford compound 5 (480 mg, 90% yield) as a yellow oil, which was used directly in the next step. 1 H NMR (400 MHz, CDCl3): 3.81 - 3.77 (m, 2H), 3.65 - 3.57 (m, 18H), 2.75 (t, J = 5.2 Hz, 2H), 2.44 (s, 3H), 1.51 (s, 18H). 1.4 Synthesis of compound 7 [0338] To a solution of compound 5 (240 mg, 532 μmol, 1.0 equiv) and compound 6 (99 mg, 0.53 μmol, 1.0 equiv) in CH 3 CN (4 mL) was added DIEA (206 mg, 1.60 μmol, 278 μL, 3.0 equiv). The mixture was stirred at 20 ℃ for 12 hours. The residue was purified by prep-HPLC (column: Phenomenex luna C18150 x 40 mm, 15 μm; mobile phase: [water (TFA)-ACN]; B%: 55%-85%, 11 min) to afford desired compound 7 (277 mg, 461 μmol, 86% yield) as a brown oil. 1 H NMR (400 MHz, CDCl3): 8.90 (m, 2H), 4.41 - 4.29 (m, 2H), 3.98 - 3.91 (m, 2H), 3.82 - 3.77 (m, 2H), 3.74 (t, J = 5.5 Hz, 2H), 3.65 - 3.59 (m, 13H), 3.37 (s, 3H), 1.50 (s, 18H), 1.37 - 1.37 (m, 1H), 1.38 (t, J = 7.1 Hz, 2H). 1.5 Synthesis of compound 8 [0339] To a solution of compound 7 (150 mg, 249 μmol, 1.0 equiv) in THF (2 mL) was added LiOH.H 2 O (1 M, 2.0 mL, 6.0 equiv). The mixture was stirred at 20 ℃ for 12 hours. The pH was adjusted to 6-7 by 1N HCl. After extraction with DCM/MeOH (10:1, 30 mL x 3), the organic layers were dried over anhydrous Na2SO4, filtered, and concentrated to afford compound 8 (142 mg, 87% yield) as a colorless oil, which was used directly in the next step.

1.6 Synthesis of compound 10 [0340] To a solution of compound 8 (105 mg, 268 μmol, 1.1 equiv, TFA salt) and compound 9 (140 mg, 244 μmol, 1.0 equiv) in DMF (1 mL) was added DIEA (94 mg, 0.73 μmol, 127 μL, 3.0 equiv) and HATU (139 mg, 366 μmol, 1.5 equiv). The mixture was stirred at 20 ℃ for 0.5 hour. The residue was purified by prep-HPLC (column: 3_Phenomenex Luna C1875 mm x 30 mm, 3 μm; mobile phase: [water(TFA)-ACN]; B%:63%-93%, 7 min) to afford compound 10 (35 mg, 27 μmol, 11% yield) as a yellow gum. 1.7 Synthesis of compound 11 [03 41] To a solution of compound 10 (110 mg, 150 μmol, 1.0 equiv) in DCM (2 mL) was added TFA (1 mL). The mixture was stirred at 25 ℃ for 0.5 hour. The reaction mixture was concentrated under reduced pressure to afford compound 10 (112 mg, 95% yield, TFA salt) as a yellow oil, which was used directly in the next step. 1.8 Synthesis of compound I-3 [0342] To a solution of compound 11 (112 mg, 149 μmol, 1.0 equiv, TFA salt) and compound 12 (60 mg, 0.15μmol, 1.0 equiv, TFA salt) in DMF (2 mL) was added DIEA (58 mg, 0.45 μmol, 78 μL, 3.0 equiv) and HATU (85 mg, 0.23 μmol, 1.5 equiv). The mixture was stirred at 20 ℃ for 0.5 hour. The residue was purified by prep-HPLC (column: Phenomenex C 1875 mm x 30 mm, 3 μm; mobile phase: [water(FA)-ACN]; B%:58%-88%, 7 min) to afford Compound N-((1r,3r)- 3-(3-chloro-4-cyanophenoxy)-2,2,4,4-tetramethylcyclobutyl)-2 -((1-((S)-4-(4-chlorophenyl)- 2,3,9-trimethyl-6H-thieno[3,2-f][1,2,4]triazolo[4,3-a][1,4]d iazepin-6-yl)-2-oxo-6,9,12,15- tetraoxa-3-azaheptad ecan-17-yl)(methyl)amino)pyrimidine-5-carboxamide (49 mg, 48 μmol, 32% yield) as an off-white solid. 1 H NMR (400 MHz, CD 3 OD): 8.74 (s, 2H), 7.73 (d, J = 8.7 Hz, 1H), 7.48 - 7.40 (m, 4H), 7.13 (d, J = 2.3 Hz, 1H), 6.99 (m, 1H), 4.79 - 4.73 (m, 3H), 4.30 - 4.25 (m, 1H), 4.17 - 4.12 (m, 1H), 3.99 - 3.87 (m, 1H), 3.92 - 3.86 (m, 2H), 3.71 (t, J = 5.6 Hz, 2H), 3.66 (s, 4H), 3.64 - 3.61 (m, 9H), 3.49 - 3.44 (m, 3H), 3.26 (s, 3H), 2.71 (s, 3H), 2.46 (s, 3H), 1.71 (s, 3H), 1.28 (d, J = 2.9 Hz, 6H), 1.22 (s, 6H). EXAMPLE 4 – Synthesis of N-((1r,3r)-3-(3-chloro-4-cyanophenoxy)-2,2,4,4- tetramethylcyclobutyl)-2-((3-(4-(3-(2-((S)-4-(4-chlorophenyl )-2,3,9-trimethyl-6H-thieno[3,2- f][1,2,4]triazolo[4,3-a][1,4]diazepin-6-yl)acetamido)propyl) piperazin-1-yl)propyl)(methyl) amino)pyrimidine-5-carboxamide (I-5) [0343] To a solution of tert-butyl N-(3-hydroxypropyl)-N-methyl-carbamate (8.0 g, 42 μmol, 1.0 equiv) in DCM (80 mL) was added TosCl (16 g, 84 μmol, 2.0 equiv) and TEA (16.0 g, 158 μmol, 22.0 mL, 3.7 equiv). The mixture was stirred at 25 ℃ for 12 hours. The reaction mixture was concentrated in vacuo to give the crude product. The crude product was purified by column chromatography on silica gel (petroleum ether/ethyl acetate = 10/1 to 5/1) to give compound 3- [tert-butoxycarbonyl(methyl)amino]propyl 4-methylbenzenesulfonate (7.0 g, 20 μmol, 48% yield) as a colorless oil. 1.2 Synthesis of compound 4 [0344] To a solution of 3-[tert-butoxycarbonyl(methyl)amino]propyl 4- methylbenzenesulfonate (7.0 g, 20 μmol, 1.0 equiv) in DMF (70 mL) was added K2CO3 (5.6 g, 41 μmol, 2.0 equiv) and benzyl piperazine-1-carboxylate (4.49 g, 20.4 μmol, 3.9 mL, 1.0 equiv). The mixture was stirred at 50 ℃ for 12 hours. The reaction mixture was concentrated in vacuo to give the crude product. The crude product was purified by prep-HPLC (column: Phenomenex luna C18150 mm x 40 mm, 15 μm; mobile phase: [water(FA)-ACN]; B%: 12%-42%, 10 mins additive) to yield compound benzyl 4-[3-[tert-butoxycarbonyl(methyl)amino]propyl]piperazine- 1-carboxylate (1.2 g, 3.1 μmol, 15% yield) as a yellow oil. 1.3 Synthesis of compound 5 [0345] To a solution of benzyl 4-[3-[tert-butoxycarbonyl(methyl)amino]propyl]piperazine-1- carboxylate (1.2 g, 3.1 μmol, 1.0 equiv) in TFE (12 mL) was added Pd/C (300 mg, 10% purity) under N 2 . The suspension was degassed under vacuum and purged with H 2 several times. The mixture was stirred under H 2 (15 psi) at 25 ℃ for 12 hours. The reaction was filtered, and the filtrate was concentrated in vacuo to give the crude product. Compound tert-butyl N-methyl-N- (3-piperazin-1-ylpropyl)carbamate (780 mg, crude) was obtained as a white solid. 1.4 Synthesis of compound 7 [0346] To a solution of tert-butyl N-methyl-N-(3-piperazin-1-ylpropyl)carbamate (680 mg, 2.64 μmol, 1.2 equiv) in DMF (20 mL) was added K 2 CO 3 (609 mg, 4.41 μmol, 2.0 equiv) and benzyl N-(3-bromopropyl)carbamate (600 mg, 2.20 μmol, 1.0 equiv). The mixture was stirred at 25 ℃ for 12 hours. 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. The residue was purified by prep-HPLC (column: Waters Xbridge C18150 mm x 50 mm, 10 μm; mobile phase: [water(NH4HCO3)-ACN]; B%: 37%-67%, 11 min). Compound tert-butyl N-[3-[4-[3- (benzyloxycarbonylamino)propyl]piperazin-1-yl]propyl]-N-meth yl-carbamate (680 mg, 1.52 μmol, 69% yield) was obtained as a colorless oil. 1.5 Synthesis of compound 8 [0347] To a solution of tert-butyl N-[3-[4-[3-(benzyloxycarbonylamino)propyl]piperazin-1- yl]propyl]-N-methyl-carbamate (680 mg, 1.52 μmol, 1.0 equiv) in TFE (10 mL) was added Pd/C (1.52 μmol, 10% purity, 1.0 equiv) under N 2 . The suspension was degassed under vacuum and purged with H 2 several times. The mixture was stirred under H 2 (15 psi) at 25 ℃ for 1 hour. The reaction was filtered, and the filtrate was concentrated in vacuo to give the crude product. Compound tert-butyl N-[3-[4-(3-aminopropyl)piperazin-1-yl]propyl]-N-methyl-carba mate (400 mg, crude) was obtained as a white solid. 1.6 Synthesis of compound 10 [0348] To a solution of tert-butyl N-[3-[4-(3-aminopropyl)piperazin-1-yl]propyl]-N-methyl- carbamate (75 mg, 0.24 ^mol, 1.0 equiv) and 2-[(9S)-7-(4-chlorophenyl)-4,5,13-trimethyl-3-thia- 1,8,11,12-tetrazatricyclo[8.3.0.0 2,6 ]trideca-2(6),4,7,10,12-pentaen-9-yl]acetic acid (95.61 mg, 238.5 μmol, 1.0 equiv) in DMF (2 mL) was added HATU (181 mg, 0.477 μmol, 2 equiv) and DIEA (154 mg, 1.19 μmol, 0.21 mL, 5.0 equiv). The mixture was stirred at 25 ℃ for 1 hour. The reaction mixture was concentrated in vacuo to give the crude product. The residue was purified by column chromatography (SiO 2 , dichloromethane/methyl alcohol = 30/1 to 3/1). Compound tert-butyl N-[3-[4-[3-[[2-[(9S)-7-(4-chlorophenyl)-4,5,13-trimethyl-3-t hia-1,8,11,12- tetrazatricyclo[8.3.0.0 2,6 ]trideca-2(6),4,7,10,12-pentaen-9-yl]acetyl]amino]prop yl]piperazin-1- yl]propyl]-N-methyl-carbamate (150 mg, 0.215 μmol, 90% yield) was obtained as a white solid. Synthesis of compound 11 [0349] To a solution of tert-butyl N-[3-[4-[3-[[2-[(9S)-7-(4-chlorophenyl)-4,5,13-trimethyl-3- thia-1,8,11,12-tetrazatricyclo[8.3.0.0 2,6 ]trideca-2(6),4,7,10,12-pentaen-9-yl]acetyl]amino] propyl]piperazin-1-yl]propyl]-N-methyl-carbamate (150 mg, 0.215 μmol, 1.0 equiv) in DCM (5 mL) was added TFA (3.08 g, 27.0 μmol, 2 mL, 126 equiv). The mixture was stirred at 25 ℃ for 1 hour. The reaction mixture was concentrated in vacuo to give the crude product. Compound 2- [(9S)-7-(4-chlorophenyl)-4,5,13-trimethyl-3-thia-1,8,11,12-t etrazatricyclo[8.3.0.0 2,6 ]trideca- 2(6),4,7,10,12-pentaen-9-yl]-N-[3-[4-[3-(methylamino)propyl] piperazin-1-yl]propyl]acetamide (128 mg, crude) was obtained as a white solid. 1.7 Synthesis of I-5 [0350] To a solution of 2-chloro-N-[3-(3-chloro-4-cyano-phenoxy)-2,2,4,4-tetramethyl - cyclobutyl]pyrimidine-5-carboxamide (84 mg, 0.20 μmol, 1 equiv) in NMP (3 mL) was added K 2 CO 3 (56 mg, 0.40 μmol, 2.0 equiv) and 2-[(9S)-7-(4-chlorophenyl)-4,5,13-trimethyl-3-thia- 1,8,11,12-tetrazatricyclo[8.3.0.0 2,6 ]trideca-2(6),4,7,10,12-pentaen-9-yl]-N-[3-[4-[3-(meth ylamino )propyl]piperazin-1-yl]propyl]acetamide (120 mg, 0.201 μmol, 1.0 equiv). The mixture was stirred at 50 ℃ for 0.5 hour. 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. The residue was purified by prep-HPLC (column: Waters Xbridge C18150 mm x 50 mm, 10 μm; mobile phase: [water(NH4HCO3)-ACN]; B%:48%-78%, 11 min). Compound N-[3-(3-chloro-4-cyano- phenoxy)-2,2,4,4-tetramethyl-cyclobutyl]-2-[3-[4-[3-[[2-[(9S )-7-(4-chlorophenyl)-4,5,13- trimethyl-3-thia-1,8,11,12-tetrazatricyclo[8.3.0.02,6]tridec a-2(6),4,7,10,12-pentaen-9-yl]acetyl] amino]propyl]piperazin-1-yl]propyl-methyl-amino]pyrimidine-5 -carboxamide (36 mg, 35 μmol, 17% yield, 95% purity) was obtained as a yellow solid. 1 H NMR (CD 3 OD ,400 MHz): δ 8.75 (s, 2H), 7.74 (d, J = 8.8 Hz, 1H), 7.49 - 7.38 (m, 4H), 7.14 (d, J = 2.4 Hz, 1H), 7.00 (m, 1H), 4.65 (m, 3H), 4.29 (s, 1H), 4.15 (s, 1H), 3.78 (t, J = 7.2 Hz, 2H), 3.46 - 3.38 (m, 1H), 3.31 - 3.27 (m, 3H), 3.24 (s, 3H), 2.71 (s, 3H), 2.64 - 2.51 (m, 4H), 2.50 - 2.41 (m, 8H), 1.92 - 1.84 (m, 2H), 1.83 - 1.75 (m, 2H), 1.72 (s, 3H), 1.30 (s, 6H), 1.23 - 1.23 (m, 1H), 1.23 (s, 6H). EXAMPLE 5 – Synthesis of N-((1r,3r)-3-(3-chloro-4-cyanophenoxy)-2,2,4,4- tetramethylcyclobutyl)-2-(4-((5-(2-((S)-4-(4-chlorophenyl)-2 ,3,9-trimethyl-6H-thieno[3,2-f] [1,2,4]triazolo[4,3-a][1,4]diazepin-6-yl)acetamido)pentyl)ox y)piperidin-1-yl)pyrimidine-5- carboxamide (I-22) 1.1 Synthesis of compound 3 [0351] To a solution of tert-butyl 4-hydroxypiperidine-1-carboxylate (2.89 g, 14.4 μmol, 1.0 equiv) in THF (50 mL) was added NaH (861 mg, 21.5 μmol, 60% purity, 1.5 equiv) and 5- benzyloxypentyl 4-methylbenzenesulfonate (5.0 g, 14.4 μmol, 1.0 equiv) at 0 ℃. The mixture was stirred at 25 ℃ for 12 hours. To the reaction mixture was added water (100 mL), and the mixture was extracted with ethyl acetate (100 mL x 3). The combined organic phase was washed with brine (100 mL), dried over anhydrous Na2SO4, filtered, and concentrated in vacuo. The residue was purified by column chromatography (SiO 2 , petroleum ether/ethyl acetate = 20/1 to 3/1). Compound tert-butyl 4-(5-benzyloxypentoxy)piperidine-1-carboxylate (3.3 g, 8.7 μmol, 61% yield) was obtained as a white solid. 1.2 Synthesis of compound 4 [0352] To a solution of tert-butyl 4-(5-benzyloxypentoxy)piperidine-1-carboxylate (3.3 g, 8.7 μmol, 1.0 equiv) in TFE (30 mL) was added Pd/C (2.0 g, 10% purity) under N2. The suspension was degassed under vacuum and purged with H 2 several times. The mixture was stirred under H 2 (15 psi) at 25 ℃ for 2 hours. The reaction was filtered, and the filtrate was concentrated in vacuo to give crude tert-butyl 4-(5-hydroxypentoxy)piperidine-1-carboxylate (2.4 g, crude) as a white solid. 1.3 Synthesis of compound 5 [0353] To a solution of tert-butyl 4-(5-hydroxypentoxy)piperidine-1-carboxylate (2.4 g, 8.4 μmol, 1.0 equiv) in DCM (30 mL) was added TosCl (3.18 g, 16.7 μmol, 2.0 equiv) and TEA (3.38 g, 33.4 μmol, 4.7mL, 4.0 equiv). The mixture was stirred at 25 ℃ for 2 hours. The reaction mixture was concentrated in vacuo to give the crude product. The residue was purified by column chromatography (SiO 2 , petroleum ether/ethyl acetate = 20/1 to 3/1). Compound tert- butyl 4-[5-(p-tolylsulfonyloxy)pentoxy]piperidine-1-carboxylate (3.3 g, 7.5 μmol, 89% yield) was obtained as a white solid. 1.4 Synthesis of compound 7 [0354] To a solution of tert-butyl 4-[5-(p-tolylsulfonyloxy)pentoxy]piperidine-1-carboxylate (1.6 g, 3.6 μmol, 1.0 equiv) in DMSO (15 mL) was added DIEA (2.34 g, 18.1 μmol, 3.16 mL, 5.0 equiv) and N-benzyl-1-phenyl-methanamine (715 mg, 3.62 μmol, 694 μL, 1.0 equiv). The mixture was stirred at 80 ℃ for 2 hours. 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 (30 mL), dried over anhydrous Na 2 SO 4 , filtered, and concentrated in vacuo. The residue was purified by column chromatography (SiO 2 , petroleum ether/ethyl acetate = 20/1 to 1/1). Compound tert-butyl 4-[5-(dibenzylamino)pentoxy]piperidine-1-carboxylate (200 mg, 429 μmol, 12% yield) was obtained as a white solid. 1.5 Synthesis of compound 8 [0355] To a solution of tert-butyl 4-[5-(dibenzylamino)pentoxy]piperidine-1-carboxylate (200 mg, 429 μmol, 1.0 equiv) in TFE (10 mL) was added Pd/C (100 mg, 10% purity) under N 2 . The suspension was degassed under vacuum and purged with H 2 several times. The mixture was stirred under H 2 (15psi) at 25 ℃ for 2 hours. The reaction was filtered, and the filtrate was concentrated in vacuo to give crude product tert-butyl 4-(5-aminopentoxy)piperidine-1- carboxylate (120 mg, crude) as a white solid. 1.6 Synthesis of compound 10 [0356] To a solution of tert-butyl 4-(5-aminopentoxy)piperidine-1-carboxylate (80 mg, 0.28 μmol, 1.0 equiv), 2-[(9S)-7-(4-chlorophenyl)-4,5,13-trimethyl-3-thia-1,8,11,12 -tetrazatricyclo [8.3.0.0 2,6 ]trideca-2(6),4,7,10,12-pentaen-9-yl]acetic acid (112 mg, 279 μmol, 1.0 equiv) in DMF (0.5 mL) was added HATU (212 mg, 559 μmol, 2.0 equiv) and DIEA (181 mg, 1.40 μmol, 243 μL, 5.0 equiv). The mixture was stirred at 25 ℃ for 0.5 hour. The reaction mixture was concentrated in vacuo to give the crude product. The crude product was purified by preparative HPLC (column: 3_Phenomenex Luna C1875 mm x 30 mm, 3 ^m; mobile phase: [water(TFA)- ACN]; B%: 65%-85%, 7 min as additive) to yield tert-butyl 4-[5-[[2-[(9S)-7-(4-chlorophenyl)- 4,5,13-trimethyl-3-thia-1,8,11,12-tetrazatricyclo[8.3.0.0 2,6 ]trideca-2(6),4,7,10,12-pentaen-9- yl]acetyl]amino]pentoxy]piperidine-1-carboxylate (120 mg, 179 μmol, 64% yield) as a white solid. 1.7 Synthesis of compound 11 [0357] To a solution of tert-butyl 4-[5-[[2-[(9S)-7-(4-chlorophenyl)-4,5,13-trimethyl-3-thia- 1,8,11,12-tetrazatricyclo[8.3.0.0 2,6 ]trideca-2(6),4,7,10,12-pentaen-9-yl]acetyl]amino]pent oxy] piperidine-1-carboxylate (60 mg, 90 μmol, 1.0 equiv) in DCM (2 mL) was added TFA (1.85 g, 16.2 μmol, 1.2 mL, 181 equiv). The mixture was stirred at 25 ℃ for 0.5 hour. The reaction mixture was concentrated in vacuo to give crude 2-[(9S)-7-(4-chlorophenyl)-4,5,13-trimethyl-3- thia-1,8,11,12-tetrazatricyclo[8.3.0.0 2,6 ]trideca-2(6),4,7,10,12-pentaen-9-yl]-N-[5-(4- piperidyloxy)pentyl]acetamide (50 mg, crude) as a white solid. 1.8 Synthesis of I-22 [0358] To a solution of 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]-N-[5-(4-piperidyloxy)pentyl] acetamide (45 mg, 79 μmol, 1.0 equiv) in NMP (1 mL) was added K2CO3 (55 mg, 0.40 μmol, 1.1 mL, 5.0 equiv) and 2-chloro-N-[3-(3-chloro-4-cyano-phenoxy)-2,2,4,4-tetramethyl -cyclobutyl] pyrimidine-5-carboxamide (33 mg, 79 μmol, 1.0 equiv). The mixture was stirred at 50 ℃ for 12 hours. The reaction mixture was concentrated in vacuo to give the crude product. The crude product was purified by preparative HPLC (column: Unisil 3-100 C18 Ultra 150 mm x 50 mm, 3 ^m; mobile phase: [water(FA)-ACN]; B%: 60%-90%, 10 min as additive) to yield N-[3-(3- chloro-4-cyano-phenoxy)-2,2,4,4-tetramethyl-cyclobutyl]-2-[4 -[5-[[2-[(9S)-7-(4-chlorophenyl)- 4,5,13-trimethyl-3-thia-1,8,11,12-tetrazatricyclo[8.3.0.0 2,6 ]trideca-2(6),4,7,10,12-pentaen-9-yl] acetyl]amino]pentoxy]-1-piperidyl]pyrimidine-5-carboxamide (35 mg, 37 μmol, 47% yield, 100% purity) as a yellow solid. 1 H NMR (400 MHz, MeOD): δ 8.69-8.74 (m, 2 H), 7.72 (d, J = 8.8 Hz, 1 H), 7.39-7.48 (m, 4 H), 7.12 (d, J = 2.4 Hz, 1 H), 6.98 (m, 1 H), 4.52-4.65 (m, 2 H), 4.23-4.35 (m, 3 H), 3.61 (m, 1 H), 3.51-3.58 (m, 4 H), 3.42 (m, 1 H), 3.26 (m, 3 H), 2.70 (s, 3 H), 2.45 (s, 3 H), 1.92 (m, 2 H), 1.70 (s, 3 H), 1.59-1.67 (m, 4 H), 1.45-1.56 (m, 4 H), 1.28 (s, 6 H), 1.21 (s, 6 H). EXAMPLE 6 – Synthesis of N-((1r,3r)-3-(3-chloro-4-cyanophenoxy)-2,2,4,4- tetramethylcyclobutyl)-2-(4-((4-(2-(2-((S)-4-(4-chlorophenyl )-2,3,9-trimethyl-6H-thieno[3,2- f][1,2,4]triazolo[4,3-a][1,4]diazepin-6-yl)acetamido)ethyl)p iperazin-1-yl)methyl)piperidin- 1-yl)pyrimidine-5-carboxamide (I-35) 1.1 Synthesis of compound 2 [0359] To a solution of benzyl piperazine-1-carboxylate (10 g, 45.4 μmol, 1.0 equiv) and tert- butyl 4-formylpiperidine-1-carboxylate (9.68 g, 45.4 μmol, 1.0 equiv) in DCE (30 mL) was added AcOH (272 mg, 4.54 μmol, 0.1 equiv). The mixture was stirred at 25 ℃ for 1 hour and NaBH(OAc) 3 (28.9 g, 136 μmol, 3.0 equiv) was added at 0 ℃. The mixture was stirred at 25 ℃ for 12 hours. The residue was purified by column chromatography (SiO 2 , petroleum ether/ethyl acetate = 5/1 to 0/1). Compound benzyl 4-[(1-tert-butoxycarbonyl-4-piperidyl)methyl] piperazine-1-carboxylate was obtained (18 g, 95% yield). 1 H NMR (400 MHz, CDCl 3 ): δ 7.41 - 7.31 (m, 5H), 5.15 (s, 2H), 4.19 - 4.11 (m, 2H), 3.52 (s, 4H), 2.70 (J = 11.4 Hz, 2H), 2.38 (s, 4H), 2.25 - 2.14 (m, 2H), 1.74 (J = 10.9 Hz, 2H), 1.70 - 1.62 (m, 1H), 1.48 - 1.46 (m, 9H), 1.16 - 0.98 (m, 2H). 1.2 Synthesis of compound 3 [0360] To a solution of benzyl 4-[(1-tert-butoxycarbonyl-4-piperidyl) methyl] piperazine-1- carboxylate (5 g, 11.97 μmol, 1.0 equiv) in THF (150 mL) was added Pd/C (500 mg, 10% purity). The mixture was stirred under H 2 (15 psi) at 20 ℃ for 1 hour. The mixture was filtered and concentrated to give tert-butyl 4-(piperazin-1-ylmethyl)piperidine-1-carboxylate (3.1 g, 91% yield). 1.3 Synthesis of compound 4 [0361] To a solution of tert-butyl 4-(piperazin-1-ylmethyl)piperidine-1-carboxylate (1 g, 3.53 μmol, 1 equiv) and 2-(1,3-dioxoisoindolin-2-yl)acetaldehyde (667 mg, 3.53 μmol, 1.0 equiv) in DCE (10 mL) was added AcOH (21 mg, 353 μmol, 0.1 equiv). The mixture was stirred at 25 ℃ for 1 hour and NaBH(OAc) 3 (2.24 g, 10.59 μmol, 3.0 equiv) was added at 0 ℃. The mixture was stirred at 25 ℃ for 12 hours. The mixture was partitioned between H 2 O (30 mL) and DCM (30 mL). The organic phase was separated, filtered, and concentrated. The residue was purified by column chromatography (SiO 2 , petroleum ether/ethyl acetate = 5/1 to 0/1) to give tert-butyl 4- [[4-[2-(1,3-dioxoisoindolin-2-yl)ethyl]piperazin-1-yl]methyl ]piperidine-1-carboxylate (1.6 g, 99% yield). 1 H NMR (400 MHz, CDCl3): δ 7.82 - 7.73 (m, 2H), 7.68 - 7.61 (m, 2H), 4.03 - 3.91 (m, 2H), 3.76 (t, J = 6.6 Hz, 2H), 2.70 - 2.47 (m, 8H), 2.40 (s, 3H), 2.14 (d, J = 6.8 Hz, 2H), 1.87 (s, 2H), 1.63 (d, J = 13.2 Hz, 2H), 1.38 (s, 8H), 1.12 - 0.87 (m, 2H). 1.4 Synthesis of compound 5 [0362] To a solution of tert-butyl 4-[[4-[2-(1,3-dioxoisoindolin-2-yl)ethyl]piperazin-1- yl]methyl]piperidine-1-carboxylate (1.2 g, 2.63 μmol, 1.0 equiv) in EtOH (50 mL) was added N 2 H 4 .H 2 O (1.34 g, 26.3 μmol, 98% purity, 10 equiv). The mixture was stirred at 70 ℃ for 2 hours. The mixture was concentrated and purified by prep-HPLC (column: Waters Xbridge 150 mm x 25 mm, 5 ^m; mobile phase: [water(NH4HCO3)-ACN]; B%: 18%-48%, 10 min) to give tert-butyl 4-[[4-(2-aminoethyl)piperazin-1-yl]methyl]piperidine-1-carbo xylate (480 mg, 56% yield) as a white oil. 1 H NMR (400 MHz, CDCl 3 ): δ 4.13 (s, 4H), 2.84 (t, J = 6.1 Hz, 2H), 2.68 (t, J = 12.0 Hz, 2H), 2.55 - 2.40 (m, 8H), 2.17 (d, J = 6.8 Hz, 2H), 1.71 (d, J = 12.8 Hz, 2H), 1.66 - 1.56 (m, 1H), 1.45 (s, 9H), 1.06 (J = 4.3, 12.2 Hz, 2H). 1.5 Synthesis of compound 6 [0363] To a solution of tert-butyl 4-[[4-(2-aminoethyl)piperazin-1-yl]methyl]piperidine-1- carboxylate (50 mg, 153 μmol, 1.0 equiv) in DMF (2 mL) was added DIEA (99 mg, 766 μmol, 5.0 equiv), 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]acetic acid (61 mg, 153 μmol, 1.0 equiv), and HATU (69 mg, 183 μmol, 1.2 equiv). The mixture was stirred at 20 ℃ for 0.5 hour. The mixture was concentrated and purified by prep-TLC (SiO 2 , DCM/MeOH = 10/1) to give tert-butyl 4-[[4- [2-[[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]acetyl]amino]ethyl]piper azin-1-yl]methyl] piperidine-1- carboxylate (40 mg, 37% yield). 1.6 Synthesis of compound 7 [0364] To a solution of tert-butyl 4-[[4-[2-[[2-[(9S)-7-(4-chlorophenyl)-4,5,13-trimethyl-3- thia-1,8,11,12-tetrazatricyclo[8.3.0.02,6]trideca-2(6),4,7,1 0,12-pentaen-9-yl]acetyl]amino]ethyl] piperazin-1-yl]methyl]piperidine-1-carboxylate (80 mg, 113 μmol, 1.0 equiv) in DCM (2 mL) was added TFA (0.5 mL). The mixture was stirred at 20 ℃ for 0.5 hour. The crude product was used directly in the next step. 1.7 Synthesis of I-35 [0365] To a solution of 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]-N-[2-[4-(4-piperidylmethyl) piperazin-1-yl]ethyl]acetamide (60 mg, 98 μmol, 1.0 equiv) in NMP (3 mL) was added K 2 CO 3 (136 mg, 985 μmol, 10 equiv) and 2-chloro-N-[3-(3-chloro-4-cyano-phenoxy)-2,2,4,4- tetramethyl-cyclobutyl]pyrimidine-5-carboxamide (45 mg, 108 μmol, 1.1 equiv). The mixture was stirred at 50 ℃ for 2 hours and concentrated. The residue was purified by prep-HPLC (column: Waters Xbridge 150 mm x 25 mm, 5 ^m; mobile phase: [water(NH4HCO3)-ACN]; B%: 55%-85%, 9 min) to give N-[3-(3-chloro-4-cyano-phenoxy)-2,2,4,4-tetramethyl-cyclobut yl] -2-[4-[[4-[2-[[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]acetyl]amino]ethyl]piper azin-1-yl]methyl]-1- piperidyl]pyrimidine-5-carboxamide (31.5 mg, 31% yield). 1 H NMR (400 MHz, METHANOL- d4): δ 8.74 (s, 2H), 7.74 (d, J = 8.8 Hz, 1H), 7.59 - 7.35 (m, 4H), 7.15 (d, J = 2 Hz, 1H), 7.00 (d, J = 2.4, 8.7 Hz, 1H), 4.71 - 4.53 (m, 2H), 4.29 (s, 1H), 4.15 (s, 1H), 3.50 - 3.40 (m, 3H), 3.38 - 3.35 (m, 1H), 3.00 (t, J = 12.2 Hz, 2H), 2.72 (s, 3H), 2.64 - 2.41 (m, 12H), 2.25 (d, J = 6.8 Hz, 2H), 1.95 - 1.85 (m, 3H), 1.73 (s, 3H), 1.33 - 1.26 (m, 7H), 1.23 (s, 6H), 1.19 - 1.05 (m, 2H). EXAMPLE 7 – Synthesis of N-((1r,3r)-3-(3-chloro-4-cyanophenoxy)-2,2,4,4- tetramethylcyclobutyl)-2-(4-((4-(3-(2-((S)-4-(4-chlorophenyl )-2,3,9-trimethyl-6H-thieno[3,2- f][1,2,4]triazolo[4,3-a][1,4]diazepin-6-yl)acetamido)propyl) piperazin-1-yl)methyl)piperidin- 1-yl)pyrimidine-5-carboxamide (I-36) 1.1 Synthesis of compound 2 [0366] To a solution of tert-butyl 4-(piperazin-1-ylmethyl)piperidine-1-carboxylate (200 mg, 705 μmol, 1.0 equiv) in DCE (2 mL) was added NaBH(OAc)3 (299 mg, 1.41 μmol, 2.0 equiv) and TEA (214 mg, 2.12 μmol, 3.0 equiv) 3-(1,3-dioxoisoindolin-2-yl)propanal (157 mg, 776 μmol, 1.1 equiv). The mixture was stirred at 25 ℃ for 12 hours and concentrated. The residue was purified by prep-HPLC (column: Waters Xbridge 150 mm x 25 mm, 5 ^m; mobile phase: [water(NH4HCO3)-ACN]; B%:53%-83%, 8 min) to give tert-butyl 4-[[4-[3-(1,3-dioxoisoindolin- 2-yl) propyl]piperazin-1-yl]methyl ]piperidine-1-carboxylate (230 mg, 69% yield) as a white solid. 1 H NMR (400 MHz, CDCl 3 ): δ 7.89-7.81 (m, 2H), 7.75-7.69 (m, 2H), 4.06 (m, 2H), 3.76 (t, J = 6.8 Hz, 2H), 2.66 (m, 3H), 2.55-2.15 (m, 9H), 2.06 (m, 3H), 2.01-1.79 (m, 5H), 1.68 (m, 2H), 1.63-1.52 (m, 1H), 1.45 (s, 10H), 1.35-1.19 (m, 1H), 1.03 (m, 2H). 1.2 Synthesis of compound 3 [0367] To a solution of tert-butyl 4-[[4-[3-(1,3-dioxoisoindolin-2-yl)propyl]piperazin-1-yl] methyl]piperid ine-1-carboxylate (230 mg, 488 μmol, 1.0 equiv) in EtOH (3 mL) was added N 2 H 4 .H 2 O (325 mg, 6.36 μmol, 98% purity, 13 equiv). The mixture was stirred at 70 ℃ for 2 hours and concentrated. The residue was purified by prep-HPLC(column: Waters Xbridge 150 mm x 25 mm, 5 ^m; mobile phase: [water(NH 4 HCO 3 )-ACN]; B%: 18%-48%, 10 min)to give tert-butyl 4-[[4-(3-aminopropyl)piperazin-1-yl]methyl]piperidine-1-carb oxylate (68 mg, 40% yield). 1 H NMR (400 MHz, CDCl 3 ): δ 4.09 (s, 1H), 2.79-2.56 (m, 2H), 2.56-2.33 (m, 4H), 2.18 (d, J = 7.2 Hz, 1H), 1.76-1.60 (m, 2H), 1.47 (s, 4H), 1.08 (m, 1H).

1.3 Synthesis of compound 4 [0368] To a solution of tert-butyl 4-[[4-(3-aminopropyl)piperazin-1-yl]methyl]piperidine-1- carboxylate (68 mg, 199 μmol, 1 equiv) in DMF (1 mL) was added 2-[(9S)-7-(4-chlorophenyl)- 4,5,13- trimethyl-3-thia-1,8,11,12- tetrazatricyclo[8.3.0.0 2,6 ]trideca-2(6),4,7,10,12-pentaen-9- yl]acetic acid (102 mg, 199 μmol, 1.0 equiv, TFA salt), HATU (98 mg, 259 μmol, 1.3 equiv), and DIEA (258 mg, 2.0 μmol, 10.0 equiv). The mixture was stirred at 20 ℃ for 1 hour and concentrated. The residue was purified by prep-TLC (SiO 2 , DCM/MeOH = 10/1) to give tert- butyl4-[[4-[3-[[2-[(9S)-7-(4-chlorophenyl)-4,5,13-trimethyl- 3-thia-1,8,11,12-tetrazatricyclo [8.3.0.0 2,6 ]trideca-2(6),4,7,10,12-pentaen-9-yl]acetyl]amino]prop yl]piperazin-1-yl]methyl] piperidine-1-carboxylate(140 mg, 96% yield). 1.4 Synthesis of compound 5 [0369] To a solution of tert-butyl 4-[[4-[3-[[2-[(9S)-7-(4-chlorophenyl)-4,5,13-trimethy l-3- thia-1,8,11,12-tetraza tricyclo[8.3.0.0 2,6 ]trideca-2(6),4,7,10,12-pentaen-9-yl]acetyl]amino] propyl]piperazin-1-yl]methyl]piperidine-1-carboxylate (140 mg, 193 μmol, 1.0 equiv) in DCM (3 mL) was added TFA (1 mL). The mixture was stirred at 25 ℃ for 1 hour. The mixture was used directly in the next step without further purification. 1.5. Synthesis of I-36 [0370] To a solution of 2-[(9S)-7-(4-chlorophenyl)-4,5,13-trimethyl-3-thia-1,8,11,12 -tetra zatricyclo[8.3.0.0 2,6 ]trideca-2(6),4,7,10,12-pentaen-9-yl]-N-[3-[4-(4-piper idylmethyl)piperazin- 1-yl]propyl]acetamide (140 mg, 189 μmol, 1.0 equiv, TFA salt) in NMP (1.5 mL) was added K2CO3 (78 mg, 569 μmol, 3.0 equiv) and 2-chloro-N-3-(3-chloro-4-cyano-phenoxy)-2,2,4,4- tetramethyl-cyclobutyl]pyrimidine-5-carboxamide (87 mg, 208 μmol, 1.1 equiv). The mixture was stirred at 50 ℃ for 12 hours and concentrated. The residue was purified by prep-HPLC (column: Waters Xbridge 150 mm x 25 mm, 5 ^m; mobile phase: [water(NH4HCO3)-ACN]; B%: 57%-87%, 9 min) to give N-[3-(3-chloro-4-cyano-phenoxy)-2,2,4,4-tetramethyl-cyclobut y l]-2-[4-[[4-[3-[[2-[(9S)-7-(4-chlorophenyl)-4,5,13-trimethyl -3-thia-1,8,11,12-tetrazatricyclo [8.3.0.0 2,6 ] trideca-2(6),4,7,10,12-pentaen-9-yl]acetyl]amino]propyl]pipe razin-1-yl]methyl]-1- piperidyl]pyrimidine-5-carboxamide (68 mg, 35% yield). 1 H NMR (400 MHz, DMSO-d6): δ 8.74 (s, 2H), 8.20 (t, J = 4.8 Hz, 1H), 7.90 (d, J = 8.8 Hz, 1H), 7.74-7.66 (m, 1H), 7.52-7.39 (m, 4H), 7.21 (d, J = 2.4 Hz, 1H), 7.03 (s, 1H), 4.76-4.69 (m, 2H), 4.54-4.46 (m, 1H), 4.28 (s, 1H), 4.03 (d, J = 9.2 Hz, 1H), 3.00-2.89 (m, 2H), 2.68-2.66 (m, 1H), 2.59 (s, 3H), 2.52 (m, 10H), 2.41 (s, 4H), 2.33 (m, 4H), 2.12 (m, 2H), 1.82-1.73 (m, 2H), 1.62 (s, 3H), 1.61-1.54 (m, 2H), 1.21 (s, 6H), 1.11 (s, 6H), 1.01 (m, 2H). EXAMPLE 8 – Synthesis of N-((1r,3r)-3-(3-chloro-4-cyanophenoxy)-2,2,4,4- tetramethylcyclobutyl)-2-(4-((4-(4-(2-((S)-4-(4-chlorophenyl )-2,3,9-trimethyl-6H-thieno[3,2- f][1,2,4]triazolo[4,3-a][1,4]diazepin-6-yl)acetamido)butyl)p iperazin-1-yl)methyl)piperidin- 1-yl)pyrimidine-5-carboxamide (I-37) 1.1 Synthesis of compound 2 [0371] To a solution of 4-bromobutan-1-ol (2 g, 13 μmol, 1.0 equiv) in DMF (20 mL) was added (1,3-dioxoisoindolin-2-yl)potassium (4.84 g, 26 μmol, 2.0 equiv). The mixture was stirred at 50 ℃ for 12 hours and concentrated. The residue was purified by prep-HPLC (column: Shim- pack C18150 mm x 25 mm, 10 ^m; mobile phase: [water(TFA)-ACN]; B%: 18%-48%, 10 min) to give 2-(4-hydroxybutyl) isoindoline-1,3-dione (700 mg, 24% yield) as a white solid. 1 H NMR (400 MHz, CDCl3): δ 7.89-7.81 (m, 2H), 7.76-7.69 (m, 2H), 3.80-3.66 (m, 4H), 1.85-1.73 (m, 2H), 1.67-1.62 (m, 2H). 1.2 Synthesis of compound 3 [0372] To a solution of 2-(4-hydroxybutyl)isoindoline-1,3-dione (270 mg, 1.23 μmol, 1.0 equiv) in DMF (2 mL) was added (1,1-diacetoxy-3-oxo-1λ 5 ,2-benziodoxol-1-yl) acetate (1.31 g, 3.08 μmol, 2.5 equiv). The mixture was stirred at 20 ℃ for 1 hour. The mixture was used directly in the next step without further purification. 1.3 Synthesis of compound 4 [0373] To a solution of tert-butyl 4-(piperazin-1-ylmethyl)piperidine-1-carboxylate (129 mg, 455 μmol, 1.5 equiv), TEA (768 mg, 7.6 μmol, 25 equiv), and NaBH(OAc) 3 (322 mg, 1.52 μmol, 5.0 equiv) in DCM (2 mL) was added 4-(1,3-dioxoisoindolin-2-yl)butanal (66 mg, 303 μmol, 1.0 equiv) at 0 ℃. The mixture was stirred at 20 ℃ for 1 hour. The mixture was diluted with H 2 O (4 mL) and extracted with EtOAc (3 x 5 mL). The combined organic layers were dried over anhydrous sodium sulfate, filtered, and concentrated. The residue was purified by prep-TLC (SiO 2 , DCM/MeOH = 10/1) to give tert-butyl 4-[[4-[4-(1,3-dioxoisoindolin-2-yl)butyl]piperazin- 1-yl]methyl]piperidine -1-carboxylate (60 mg, 40% yield) as a brown oil. 1 H NMR (400 MHz, MeOD): δ 7.87-7.82 (m, 2H), 7.75-7.69 (m, 2H), 4.11-3.97 (m, 2H), 3.75-3.65 (m, 2H), 2.77- 2.60 (m, 2H), 2.54-2.30 (m, 9H), 2.20-2.14 (m, 2H), 1.77-1.68 (m, 4H), 1.63 (s, 4H), 1.46 (s, 9H), 1.29-1.24 (m, 2H). 1.4 Synthesis of compound 5 [0374] To a solution of tert-butyl 4-[[4-[4-(1,3-dioxoisoindolin-2-yl)butyl]piperazin-1-yl] methyl]piperidine -1-carboxylate (380 mg, 784 μmol, 1.0 equiv) in EtOH (4 mL) was added N 2 H 4 . H 2 O (6.66 g, 130 μmol, 166.31 equiv). The mixture was stirred at 70 ℃ for 2 hours and concentrated. The residue was purified by prep-HPLC (column: Waters Xbridge 150 mm x 25 mm, 5 ^m; mobile phase: [water(NH 4 HCO 3 )-ACN]; B%: 10%-40%, 10 min) to give tert-butyl 4-[[4-(4-aminobutyl)piperazin- 1-yl]methyl]piperidine-1-carboxylate (150 mg, 54% yield) as a brown oil. 1 NMR (400 MHz, MeOD): δ 4.30-3.90 (m, 2H), 2.78-2.61 (m, 4H), 2.60-2.29 (m, 9H), 2.20-2.15 (m, 2H), 2.13-1.79 (m, 4H), 1.76-1.68 (m, 2H), 1.59-1.47 (m, 4H), 1.46 (s, 9H), 1.14-0.97 (m, 2H). 1.5 Synthesis of compound 6 [0375] To a solution of tert-butyl 4-[[4-(4-aminobutyl)piperazin-1-yl]methyl]piperidine-1- carboxylate (75 mg, 211 μmol, 1.0 equiv) in DMF (2 mL) was added DIEA (136 mg, 1.06 μmol, 5.0 equiv) and HATU (104 mg, 275 μmol, 1.3 equiv), and 2-[(9S)-7-(4-chlorophenyl)-4,5,13- trimethyl-3-thia-1,8,11,12-tetrazatricyclo [8.3.0.0 2,6 ]trideca-2(6),4,7,10,12-pentaen-9-yl]acetic acid (109 mg, 211 μmol, 1.0 equiv, TFA salt). The mixture was stirred at 25 ℃ for 0.5 hour. The mixture was diluted with H 2 O (10 mL) and extracted with EtOAc (3 x 5 mL). The combined organic layers were washed with H 2 O, dried over anhydrous sodium sulfate, filtered, and concentrated. The residue was purified by prep-TLC (SiO 2 , DCM/MeOH = 10/1) to give tert- butyl 4-[[4-[4-[[2-[(9S)-7-(4-chlorophenyl) -4,5,13-trimethyl-3-thia-1,8,11,12-tetrazatricyclo [8.3.0.0 2,6 ]trideca-2(6),4,7,10,12-pentaen-9-yl]acetyl]amino]buty l]piperazin-1-yl]methyl] piperidine-1-carboxylate (70 mg, 44% yield) as a brown oil. 1 H NMR (400 MHz, CDCl 3 ): δ 7.49-7.43 (m, 2H), 7.40-7.35 (m, 2H), 3.50 (s, 6H), 3.44-3.38 (m, 3H), 3.25-3.17 (m, 2H), 3.11- 2.79 (m, 4H), 2.74-2.65 (m, 6H), 2.42 (s, 4H), 2.31-2.25 (m, 2H), 1.73-1.66 (m, 6H), 1.47 (s, 9H), 1.20-1.14 (d, J = 6.4 Hz, 3H), 1.12-0.98 (m, 2H), 0.96-0.86 (m, 2H). 1.6 Synthesis of compound 7 [0376] A mixture of tert-butyl 4-[[4-[4-[[2-[(9S)-7-(4-chlorophenyl)-4,5,13-trimethyl-3-thi a- 1,8,11,12- tetrazatricyclo[8.3.0.0 2,6 ]trideca-2(6),4,7,10,12-pentaen-9-yl]acetyl]amino] butyl]piperazin-1-yl]methyl]piperidine-1-carboxylate (70 mg, 95 μmol, 1.0 equiv) in TFA (0.5 mL) and DCM (1.5 mL) was stirred at 20 ℃ for 0.5 hour. The solution was concentrated under reduced pressure to give 2-[(9S)-7-(4-chlorophenyl)-4,5,13-trimethyl-3-thia-1,8,11,12 - tetrazatricyclo[8.3.0.0 2,6 ]trideca-2(6),4,7,10,12-pentaen-9-yl]-N-[4-[4-(4-piper idylmethyl) piperazin-1-yl]butyl]acetamide (70 mg, crude, TFA salt) as a brown oil. 1.7 Synthesis of I-37 [0377] To a solution of 2-[(9S)-7-(4-chlorophenyl)-4,5,13-trimethyl-3-thia-1,8,11,12 - tetrazatricyclo[8.3.0.0 2,6 ] trideca-2(6),4,7,10,12-pentaen-9-yl]-N-[4-[4-(4-piperidylmet hyl) piperazin-1-yl]butyl]acetamide (70 mg, 93 μmol, 1.0 equiv, TFA salt) in NMP (1 mL) was added K2CO3 (39 mg, 279 μmol, 3.0 equiv) and 2-chloro-N-[3-(3-chloro-4-cyano-phenoxy)-2,2,4,4- tetramethyl-cyclobutyl]pyrimidine-5-carboxamide (39 mg, 93 μmol, 1.0 equiv). The mixture was stirred at 50 ℃ for 4 hours and concentrated. The residue was purified by prep-HPLC (column: Unisil 3-100 C18 Ultra 150 mm x 50 mm, 3 ^m; mobile phase: [water(FA)-ACN]; B%: 30%- 60%, 7 min) to give N-[3-(3-chloro-4-cyano-phenoxy)-2,2,4,4-tetramethyl- cyclobutyl]-2-[4-[[4- [4-[[2-[(9S)-7-(4-chlorophenyl)-4,5,13-trimethyl-3-thia-1,8, 11,12-tetrazatricyclo [8.3.0.0 2,6 ]trideca-2(6),4,7,10,12-pentaen-9-yl]acetyl]amino]buty l]piperazin-1-yl]methyl]-1- piperidyl]pyrimidine-5-carboxamide (25 mg, 24% yield). 1 H NMR (400 MHz, DMSO-d 6 ): δ 8.74 (s, 2H), 8.22-8.14 (m, 1H), 7.92-7.87 (m, 1H), 7.73-7.66 (m, 1H), 7.52-7.46 (m, 2H), 7.45- 7.39 (m, 2H), 7.21 (s, 1H), 7.04-6.91 (m, 1H), 4.76-4.67 (m, 2H), 4.53-4.46 (m, 1H), 4.29 (s, 1H), 4.05-4.01 (m, 1H), 3.00-2.90 (m, 4H), 2.67 (s, 2H), 2.33 (s, 6H), 2.29-2.18 (m, 5H), 2.15- 2.05 (m, 3H), 1.87-1.70 (m, 4H), 1.63 (s, 3H), 1.44 (s, 5H), 1.24-1.22 (m, 1H), 1.21 (s, 6H), 1.11 (s, 6H), 1.05-0.94 (m, 3H). EXAMPLE 9 – Synthesis of N-((1r,3r)-3-(3-chloro-4-cyanophenoxy)-2,2,4,4- tetramethylcyclobutyl)-2-(4-(2-(2-(2-(4-((S)-6-(2-(ethylamin o)-2-oxoethyl)-2,3,9-trimethyl- 6H-thieno[3,2-f][1,2,4]triazolo[4,3-a][1,4]diazepin-4-yl)phe noxy)ethoxy)ethoxy) ethoxy) piperidin-1-yl)pyrimidine-5-carboxamide (I-45) [0378] To a solution of 2-[(9S)-7-[4-[2-[2-[2-[[1-[5-[[3-(3-chloro-4-cyano-phenoxy)- 2,2,4,4- tetramethyl-cyclobutyl]carbamoyl]pyrimidin-2-yl]-4-piperidyl ]oxy]ethoxy]ethoxy]ethoxy] phenyl]-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]acetic acid (30 mg, 31 μmol, 1.0 equiv) and ethanamine (1 mg, 31 μmol, 2.00 μL, 1.0 equiv) in DMF (1.5 mL) was added DIEA (8 mg, 61 μmol, 2.0 equiv) and HATU (17 mg, 46 μmol, 1.5 equiv). The mixture was stirred at 25 ℃ for 1 hour. The residue was purified by prep- HPLC (column: Unisil 3-100 C18 Ultra 150 mm x 50 mm, 3 ^m; mobile phase: [water(FA)- ACN]; B%: 48%-78%, 7 min) to afford N-[3-(3-chloro-4-cyano-phenoxy)-2,2,4,4-tetramethyl- cyclobutyl]-2-[4-[2-[2-[2-[4-[(9S)-9-[2-(ethylamino)-2-oxo-e thyl]-4,5,13-trimethyl-3-thia- 1,8,11,12-tetrazatricyclo[8.3.0.02,6]trideca-2(6),4,7,10,12- pentaen-7-yl]phenoxy]ethoxy] ethoxy]ethoxy]-1-piperidyl]pyrimidine-5-carboxamide (7.0 mg, 6.4 μmol, 21% yield, 91% purity) as a white solid. 1 H NMR (CD3OD, 400 MHz): 8.72 (s, 2H), 8.3-8.4 (m, 1H), 7.72 (d, 1H, J = 8.8 Hz), 7.39 (m, 2H), 7.12 (d, 1H, J = 2.4 Hz), 6.9-7.0 (m, 3H), 4.59 (m, 1H), 4.3-4.3 (m, 3H), 4.1-4.2 (m, 3H), 3.8-3.9 (m, 2H), 3.6-3.7 (m, 11H), 3.53 (m, 2H), 3.4-3.4 (m, 1H), 3.35 (s, 1H), 2.69 (s, 3H), 2.44 (s, 3H), 1.9-2.0 (m, 2H), 1.70 (s, 3H), 1.5-1.6 (m, 2H), 1.3-1.3 (m, 2H), 1.2-1.3 (m, 12H). EXAMPLE 10 – Synthesis of N-((1r,3r)-3-(3-chloro-4-cyanophenoxy)-2,2,4,4- tetramethylcyclobutyl)-2-(4-(2-(3-((S)-6-(2-(ethylamino)-2-o xoethyl)-2,3,9-trimethyl-6H- thieno[3,2-f][1,2,4]triazolo[4,3-a][1,4]diazepin-4-yl)phenox y)ethoxy)piperidin-1- yl)pyrimidine-5-carboxamide (I-46) [0379] To a solution of 2-[(9S)-7-[3-[2-[[1-[5-[[3-(3-chloro-4-cyano-phenoxy)-2,2,4, 4- tetramethyl-cyclobutyl]carbamoyl]pyrimidin-2-yl]-4-piperidyl ]oxy]ethoxy]phenyl]-4,5,13- trimethyl-3-thia-1,8,11,12-tetrazatricyclo[8.3.0.0 2,6 ]trideca-2(6),4,7,10,12-pentaen-9-yl]acetic acid (100mg 112μmol, 1.0 equiv) in DMF (1.5 mL) was added DIEA (43.5 mg, 336 μmol, 3.0 equiv) and HATU (64 mg, 0.17 μmol, 1.5 equiv). The mixture was stirred at 25 ℃ for 1 hour. The reaction mixture was filtered to give a residue. The residue was purified by prep-HPLC (column: Phenomenex C1875 mm x 30 mm, 3 ^m; mobile phase: [water(FA)-ACN]; B%: 65%- 95%, 7 min) to afford N-[3-(3-chloro-4-cyano-phenoxy)-2,2,4,4-tetramethyl-cyclobut yl]-2-[4-[2- [3-[(9S)-9-[2-(ethylamino)-2-oxo-ethyl]-4,5,13-trimethyl-3-t hia-1,8,11,12-tetrazatricyclo [8.3.0.0 2,6 ]trideca-2(6),4,7,10,12-pentaen-7-yl]phenoxy]ethoxy]-1 -piperidyl]pyrimidine-5- carboxamide (30 mg, 33 μmol, 29% yield) as a white solid. 1 H NMR (400 MHz, CD3OD): δ 8.73 (s, 3H), 7.78 - 7.69 (m, 3H), 7.30 (t, J = 7.9 Hz, 2H), 7.17 - 7.06 (m, 5H), 7.02 - 6.93 (m, 3H), 4.65 - 4.59 (m, 3H), 4.32 - 4.22 (m, 5H), 4.22 - 4.08 (m, 5H), 3.88 (t, J = 4.5 Hz, 4H), 3.80 - 3.69 (m, 2H), 3.66 - 3.55 (m, 4H), 3.51 - 3.33 (m, 4H), 3.29 - 3.13 (m, 4H), 2.74 - 2.67 (m, 5H), 2.44 (s, 5H), 2.02 - 1.86 (m, 4H), 1.69 (s, 5H), 1.59 (m, 4H), 1.28 (d, J = 3.8 Hz, 11H), 1.24 - 1.17 (m, 15H). EXAMPLE 11 – Synthesis of N-((1r,3r)-3-(3-chloro-4-cyanophenoxy)-2,2,4,4- tetramethylcyclobutyl)-2-(4-(2-(2-(3-((S)-6-(2-(ethylamino)- 2-oxoethyl)-2,3,9-trimethyl-6H- thieno[3,2-f][1,2,4]triazolo[4,3-a][1,4]diazepin-4-yl)phenox y)ethoxy)ethoxy)piperidin-1- yl)pyrimidine-5-carboxamide (I-47) [0380] To a solution of 2-[(9S)-7-[3-[2-[2-[[1-[5-[[3-(3-chloro-4-cyano-phenoxy)-2,2 ,4,4- tetramethyl-cyclobutyl]carbamoyl]pyrimidin-2-yl]-4-piperidyl ]oxy]ethoxy]ethoxy]phenyl]- 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]acetic acid (240 mg, 256 μmol, 1.0 equiv) in DMF (0.5 mL) was added EtNH 2 (256 μmol, 0.1 mL, 1 equiv), DIEA (712 mg, 5.5 μmol, 960 μL, 21.5 equiv), and HATU (146 mg, 384 μmol, 1.5 equiv). The mixture was stirred at 25 ℃ for 0.5 hour. The residue was purified by prep-HPLC (column: Waters Xbridge 150 mm x 25 μm, 5 ^m; mobile phase: [water (NH 4 HCO 3 )-ACN]; B%: 54%-84%, 9 min) to give compound N-[3-(3-chloro-4-cyano-phenoxy)-2, 2, 4, 4-tetramethyl- cyclobutyl]-2-[4-[2-[2-[3-[(9S)-9-[2-(ethylamino)-2-oxo-ethy l]-4,5,13-trimethyl-3-thia-1,8,11,12 -tetrazatricyclo[8.3.0.02,6]trideca-2(6),4,7,10,12-pentaen-7 -yl]phenoxy]ethoxy]ethoxy]-1- piperidyl]pyrimidine-5-carboxamide (151 mg, 61% yield) as a white solid. 1 H NMR (400 MHz, CD3OD): δ 8.74 (s, 2 H), 8.18 - 8.21 (m, 1 H), 7.90 (d, J = 8.80 Hz, 1 H), 7.71 (d, J = 9.17 Hz, 1 H), 7.27 - 7.34 (m, 1 H), 7.21 (d, J = 2.45 Hz, 1 H), 6.84 - 7.11 (m, 4 H), 4.43 - 4.51 (m, 1 H), 4.18 - 4.34 (m, 3 H), 4.00 - 4.13 (m, 3 H), 3.73 - 3.76 (m, 2 H), 3.59 (s, 5 H), 3.40 - 3.52 (m, 2 H), 3.06 - 3.25 (m, 4 H), 2.62 - 2.68 (m, 1 H), 2.59 (s, 3 H), 2.40 (s, 3 H), 1.83 - 1.92 (m, 2 H), 1.62 (s, 2 H), 1.33 - 1.48 (m, 2 H), 1.21 (d, J = 4.40 Hz, 6 H), 1.02 - 1.14 (m, 9 H). EXAMPLE 12 – Synthesis of N-((1r,3r)-3-(3-chloro-4-cyanophenoxy)-2,2,4,4- tetramethylcyclobutyl)-2-(4-(2-(2-(2-(3-((S)-6-(2-(ethylamin o)-2-oxoethyl)-2,3,9-trimethyl- 6H-thieno[3,2-f][1,2,4]triazolo[4,3-a][1,4]diazepin-4-yl)phe noxy)ethoxy)ethoxy)ethoxy) piperidin-1-yl)pyrimidine-5-carboxamide (I-48) [0381] To a solution of 2-[(9S)-7-[3-[2-[2-[2-[[1-[5-[[3-(3-chloro-4-cyano-phenoxy)- 2,2,4,4- tetramethyl-cyclobutyl]carbamoyl]pyrimidin-2-yl]-4-piperidyl ]oxy]ethoxy] ethoxy]ethoxy] phenyl]-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]acetic acid (160 mg, 163 μmol, 1.0 equiv) in DMF (0.5 mL) was added EtNH 2 (163 μmol, 0.1 mL, 1.0 equiv), DIEA (453 mg, 3.50 μmol, 611 μL, 21 equiv), and HATU (93 mg, 0.24 μmol, 1.5 equiv). The mixture was stirred at 25 ℃ for 0.5 hour. The residue was purified by prep-HPLC (column: Waters Xbridge 150 mm x 25 mm, 5 ^m; mobile phase: [water(NH4HCO3)-ACN]; B%: 54%-84%, 9 min) to afford compound N-[3-(3-chloro-4-cyano- phenoxy)-2, 2, 4, 4-tetramethyl-cyclobutyl]-2-[4-[2-[2-[2-[3-[(9S)-9-[2-(ethyl amino)-2-oxo- ethyl]-4, 5, 13-trimethyl-3-thia-1,8,11,12-tetrazatricyclo[8.3.0.02,6]tri deca-2(6),4,7,10,12- pentaen-7-yl]phenoxy]ethoxy] ethoxy]ethoxy]-1-piperidyl]pyrimidine-5-carboxamide (105 mg, 63% yield) as a white solid. 1 H NMR (400 MHz, CD 3 OD): δ 8.74 (s, 2 H), 8.18 - 8.21 (m, 1 H), 7.90 (d, J = 8.68 Hz, 1 H), 7.71 (d, J = 9.16 Hz, 1 H), 7.27 - 7.32(m, 1 H), 7.21 (d, J = 2.32 Hz, 1 H), 6.91 - 7.10 (m, 4 H), 4.46 - 4.49 (m, 1 H), 4.23 - 4.28 (m, 3 H), 4.04 - 4.06 (m, 3 H), 3.72 - 3.74 (m, 2 H), 3.51 - 3.64 (m, 10 H), 3.39 - 3.48 (m, 2 H), 3.09 - 3.25 (m, 4 H), 2.60 (s, 3 H), 2.40 (s, 3 H), 1.81 - 1.91 (m, 2 H), 1.62 (s, 2 H), 1.33 - 1.46 (m, 2 H), 1.21 (d, J=2.32 Hz, 6 H), 1.05 - 1.14 (m, 9 H). EXAMPLE 13 – Synthesis of N-[3-(3-chloro-4-cyano-phenoxy)-2,2,4,4-tetramethyl- cyclobutyl]-2-[2-[4-[(9S)-4,5,13-trimethyl-9-(oxazol-2-ylmet hyl)-3-thia-1,8,11,12- tetrazatricyclo[8.3.0.02,6]trideca-2(6),4,7,10,12-pentaen-7- yl]phenoxy]-7- azaspiro[3.5]nonan-7-yl]pyrimidine-5-carboxamide (I-49) , and N-[3-(3-chloro-4-cyano- phenoxy)-2,2,4,4-tetramethyl-cyclobutyl]-2-[2-[4-[(9R)-4,5,1 3-trimethyl-9-(oxazol-2- ylmethyl)-3-thia-1,8,11,12-tetrazatricyclo[8.3.0.02,6]tridec a-2(6),4,7,10,12-pentaen-7- yl]phenoxy]-7-azaspiro[3.5]nonan-7-yl]pyrimidine-5-carboxami de (I-50) .

[0382] Step 1: Preparation of 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]acetic acid. 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 (1.0 g, 2.1 mmol, 1.0 equiv) in DCM (10 mL) was added TFA (7.7 g, 67 mmol, 5.0 mL, 30 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 comprising the compound 2-[(9S)-7-(4-chlorophenyl)-4,5,13- trimethyl-3-thia-1,8,11,12-tetrazatricyclo[8.3.0.02,6]tridec a-2(6),4,7,10,12-pentaen-9-yl]acetic acid (877 mg, 99% yield). The crude residue was used for the next step. [0383] Step 2: Preparation of 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]acetamide. To a solution of 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]acetic acid (877 mg, 2.19 mmol, 1.0 equiv) and NH4Cl (351 mg, 6.56 mmol, 3.0 equiv) in DMF (8 mL) was added HATU (915 mg, 2.41 mmol, 1.1 equiv) and DIEA (848 mg, 6.56 mmol, 3.0 equiv). The mixture was stirred at 25 °C for 1 h. The mixture was filtered and concentrated. The residue was purified by prep-HPLC (column: Waters Xbridge C18150*50mm* 10μm;mobile phase: [water( NH4HCO3)-ACN];B%: 24%- 54%,10 min) to give 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]acetamide (740 mg, 84% yield). [0384] Step 3: Preparation of 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]methyl]oxazole. A solution of 1,3-dioxol-2-one (103 mg, 1.20 mmol, 1.2 equiv) and 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]acetamide (400 mg, 1.00 mmol, 1.0 equiv) in PPA (4 mL) was prepared. This mixture was stirred at 160 °C for 3 h. The mixture was quenched by addition of H 2 O 50 mL at 0°C, and then diluted with EA 200 mL. The mixture was extracted with H 2 O (15 mL * 2). The combined organic layers were washed with H 2 O (10 mL * 3), dried over Na2SO4, filtered and concentrated. The residue was purified by column chromatography (SiO 2 , Petroleum ether/Ethyl acetate=2/1 to 0/1) to give compound 2-[[(9S)-7-(4-chlorophenyl)-4,5,13-trimethyl-3-thia-1,8,11,1 2- tetrazatricyclo [8.3.0.02,6]trideca-2(6),4,7,10,12-pentaen-9-yl]methyl]oxazo le (260 mg, 61% yield). [0385] Step 4: Preparation of 4-[(9S)-4,5,13-trimethyl-9-(oxazol-2-ylmethyl)-3-thia- 1,8,11,12-tetrazatricyclo[8.3.0.02,6]trideca-2(6),4,7,10,12- pentaen-7-yl]phenol. A mixture of 2-[[(9S)-7-(4-chlorophenyl)-4,5,13-trimethyl-3-thia-1,8,11,1 2-tetrazatricyclo[8.3.0.02,6]trideca- 2(6),4,7,10,12-pentaen-9-yl]methyl]oxazole (5.8 g, 13.7 mmol, 1.0 equiv), BrettPhos Pd G3 (1.2 g, 1.4 mmol, 0.1 equiv), KOH (3.8 g, 68.4 mmol, 5.0 equiv) in dioxane (58 mL) and H 2 O (11.6 mL) was degassed and purged with N2 3 times, and then the mixture was stirred at 90 °C for 3 h under N 2 atmosphere. The reaction mixture was partitioned between EA (2000 mL) and water (500 mL). The organic phase was separated, washed with brine (500 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 , Dichloromethane : Methanol=40/1) to give the 4- [(9S)-4,5,13-trimethyl-9-(oxazol-2-ylmethyl)-3-thia-1,8,11,1 2-tetrazatricyclo[8.3.0.02,6] trideca- 2(6),4,7,10,12-pentaen-7-yl]phenol (3.5 g, 8.6 mmol, 63% yield) as a yellow solid. [0386] Step 5: Preparation of tert-butyl 2-[4-[(9S)-4,5,13-trimethyl-9-(oxazol-2- ylmethyl)-3-thia-1,8,11,12-tetrazatricyclo[8.3.0.02,6]tridec a-2(6),4,7,10,12-pentaen-7- yl]phenoxy]-7-azaspiro[3.5]nonane-7-carboxylate. To a 250 mL well equipped 3 neck flask was added 4-[(9S)-4,5,13-trimethyl-9-(oxazol-2-ylmethyl)-3-thia-1,8,11 ,12-tetrazatricyclo [8.3.0.02,6]trideca-2(6),4,7,10,12-pentaen-7-yl]phenol (4.0 g, 9.9 mmol, 1.0 equiv), tert-butyl 2- hydroxy-7-azaspiro[3.5]nonane-7-carboxylate (4.8 g, 19.7 mmol, 2.0 equiv) in THF (10 mL) and stirred at 25 °C under N 2 protection. In a separate flask, PPh3 (5.4 g, 20.7 mmol, 2.1 equiv) in THF (56 mL) was cooled to 0 °C under N2 protection, and DIAD (4.0 g, 19.7 mmol, 3.8 mL, 2.0 equiv) was added to this mixture and stirred for 0.5 h to create the betaine intermediate. Betain was added to the mixture of two reactants in the three neck flask at 25 °C, allowed to warm up to 50 °C, and stirred for 12 h. The mixture was concentrated to give a residue. The residue was purified by column chromatography (SiO 2 , DCM: MeOH = 30:1) to give the tert-butyl 2-[4- [(9S)-4,5,13-trimethyl-9-(oxazol-2-ylmethyl)-3-thia-1,8,11,1 2-tetrazatricyclo[8.3.0.02,6]trideca- 2(6),4,7,10,12-pentaen-7-yl]phenoxy]-7-azaspiro[3.5]nonane-7 -carboxylate (4.8 g, 7.6 mmol, 94.8% ee, 77% yield) as a yellow solid. [0387] Step 6: Preparation of 2-[[(9S)-7-[4-(7-azaspiro[3.5]nonan-2-yloxy)phenyl]-4,5,13- trimethyl-3-thia-1,8,11,12-tetrazatricyclo[8.3.0.02,6]tridec a-2(6),4,7,10,12-pentaen-9- yl]methyl]oxazole. A mixture of tert-butyl 2-[4-[(9S)-4,5,13-trimethyl-9-(oxazol-2-ylmethyl)-3- thia-1,8,11,12-tetrazatricyclo[8.3.0.02,6]trideca-2(6),4,7,1 0,12-pentaen-7-yl]phenoxy]-7- azaspiro[3.5]nonane-7-carboxylate (3.5 g, 5.6 mmol, 1.0 equiv) in TFA (10 mL), DCM (30 mL) was degassed and purged with N 2 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 residue was dissolved in EA (100 mL) and washed with NaHCO3 (100 mL) and brine(100 mL), dried over Na2SO4, filtered and concentrated to give 2-[[(9S)-7-[4-(7-azaspiro[3.5]nonan-2-yloxy)phenyl]-4,5,13- trimethyl-3-thia-1,8,11,12-tetrazatricyclo[8.3.0.02,6]tridec a-2(6),4,7,10,12-pentaen-9- yl]methyl]oxazole (2.9 g, 5.5 mmol, 98% yield) as a yellow solid. [0388] Step 7: Preparation of N-((1r,3r)-3-(3-chloro-4-cyanophenoxy)-2,2,4,4- tetramethylcyclobutyl)-2-(2-(4-((S)-2,3,9-trimethyl-6-(oxazo l-2-ylmethyl)-6H-thieno[3,2- f][1,2,4]triazolo[4,3-a][1,4]diazepin-4-yl)phenoxy)-7-azaspi ro[3.5]nonan-7-yl)pyrimidine-5- carboxamide (I-49), and N-((1r,3r)-3-(3-chloro-4-cyanophenoxy)-2,2,4,4- tetramethylcyclobutyl)-2-(2-(4-((R)-2,3,9-trimethyl-6-(oxazo l-2-ylmethyl)-6H-thieno[3,2- f][1,2,4]triazolo[4,3-a][1,4]diazepin-4-yl)phenoxy)-7-azaspi ro[3.5]nonan-7-yl)pyrimidine-5- carboxamide (I-50). A mixture of 22-chloro-N-((1r,3r)-3-(3-chloro-4-cyanophenoxy)-2,2,4,4- tetramethylcyclobutyl)pyrimidine-5-carboxamide (1.7 g, 4.0 mmol, 1.0 equiv), (S)-2-((4-(4-((7- azaspiro[3.5]nonan-2-yl)oxy)phenyl)-2,3,9-trimethyl-6H-thien o[3,2-f][1,2,4]triazolo[4,3- a][1,4]diazepin-6-yl)methyl)oxazole (2.1 g, 4.0 mmol, 1.0 equiv), DIEA (1.0 g, 7.9 mmol, 1.4 mL, 2.0 equiv) in NMP (20 mL) was degassed and purged with N23 times, and then the mixture was stirred at 25 °C for 12 h under N 2 atmosphere. The mixture was poured into H 2 O (100 mL) and extracted with EA (200 mL x 2), the organic layer was washed with brine (200 mL), and dried over Na2SO4. The organic layer was concentrated to give a residue and purified by column chromatography (SiO 2 , DCM/MeOH=50/1 to 30/1). The compound was purified again by the SFC-HPLC(column: REGIS (R,R)WHELK-O1(250mm*25mm, 10 um);mobile phase: [CO 2 -i- PrOH/ACN];B%: 70%-70%,6.8min ) to give N-((1r,3r)-3-(3-chloro-4-cyanophenoxy)-2,2,4,4- tetramethylcyclobutyl)-2-(2-(4-((S)-2,3,9-trimethyl-6-(oxazo l-2-ylmethyl)-6H-thieno[3,2- f][1,2,4]triazolo[4,3-a][1,4]diazepin-4-yl)phenoxy)-7-azaspi ro[3.5]nonan-7-yl)pyrimidine-5- carboxamide (1.7 g, 1.8 mmol, 46 % yield, 98% purity, 100% ee) and N-((1r,3r)-3-(3-chloro-4- cyanophenoxy)-2,2,4,4-tetramethylcyclobutyl)-2-(2-(4-((R)-2, 3,9-trimethyl-6-(oxazol-2- ylmethyl)-6H-thieno[3,2-f][1,2,4]triazolo[4,3-a][1,4]diazepi n-4-yl)phenoxy)-7- azaspiro[3.5]nonan-7-yl)pyrimidine-5-carboxamide (0.9 g, 977 μmol, 24 % yield, 99% purity, 100% ee) as a white solid. [0389] I-49: 1 H NMR (400 MHz, MeOD): δ = 8.74 (s, 2H), 7.91 (d, J = 0.8 Hz, 1H), 7.74 (d, J = 8.8 Hz, 1H), 7.34 (d, J = 8.8 Hz, 2H), 7.21 - 7.12 (m, 2H), 7.05 - 6.96 (m, 1H), 6.85 (d, J = 8.8 Hz, 2H), 4.82 (d, J = 6.8 Hz, 1H), 4.78 - 4.70 (m, 1H), 4.28 (s, 1H), 4.18 - 4.10 (m, 1H), 4.02 - 3.81 (m, 6H), 2.72 (s, 3H), 2.61 - 2.50 (m, 2H), 2.47 (s, 3H), 2.04 - 1.92 (m, 2H), 1.78 - 1.63 (m, 7H), 1.29 (s, 6H), 1.23 (s, 6H). [0390] I-50: 1 H NMR (400 MHz, MeOD): δ = 8.72 (s, 2H), 7.89 (d, J = 0.8 Hz, 1H), 7.72 (d, J = 8.8 Hz, 1H), 7.32 (d, J = 8.8 Hz, 2H), 7.17 - 7.07 (m, 2H), 7.04 - 6.92 (m, 1H), 6.83 (d, J = 8.8 Hz, 2H), 4.83 (s, 1H), 4.76 - 4.71 (m, 1H), 4.26 (s, 1H), 4.18 - 4.08 (m, 1H), 4.03 - 3.78 (m, 6H), 2.70 (s, 3H), 2.57 - 2.49 (m, 2H), 2.46 (s, 3H), 2.02 - 1.92 (m, 2H), 1.81 - 1.60 (m, 7H), 1.28 (s, 6H), 1.21 (s, 6H). EXAMPLE 14 – Synthesis of Additional Compounds [0422] The following additional compounds were prepared based on procedures described herein: compounds I-4, I-6, I-7, I-8, I-9, I-10, I-11, I-12, I-13, I-14, I-15, I-16, I-17, I-18, I-19, I- 20, I-21, 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-38, I-39, I-40, I-41, I- 42, I-43, and I-44. Starting materials used when synthesizing these compounds are listed in Table 2. TABLE 2. C

EXAMPLE 15 – LC-MS Physical Characterization Data [0391] Exemplary compounds were analyzed by LC-MS. Results are provided in Table 3 below. TABLE 3. Compound Observed Mass Retention Time LCMS EXAMPLE 16 - Synthesis of tert-butyl 2-((S)-4-(4-(2-((1-(5-(((1r,3r)-3-(3-chloro-4- cyanophenoxy)-2,2,4,4-tetramethylcyclobutyl)carbamoyl)pyrimi din-2-yl)piperidin-4- yl)oxy)ethoxy)phenyl)-2,3,9-trimethyl-6H-thieno[3,2-f][1,2,4 ]triazolo[4,3-a][1,4]diazepin-6- yl)acetate (I-235) [0423] Step 1: Preparation of tert-butyl 4-[2-[4-[(9S)-9-(2-tert-butoxy-2-oxo-ethyl)- 4,5,13-trimethyl-3-thia-1,8,11,12-tetrazatricyclo[8.3.0.02,6 ]trideca-2(6),4,7,10,12-pentaen-7- yl]phenoxy]ethoxy]piperidine-1-carboxylate. A mixture of tert-butyl 4-(2- hydroxyethoxy)piperidine-1-carboxylate (403 mg, 1.64 mmol, 1.5 equiv), tert-butyl2-[(9S)-7-(4- chlorophenyl)-4,5,13-trimethyl-3-thia-1,8,11,12-tetrazatricy clo[8.3.0.02,6]trideca-2(6),4,7,10,12- pentaen-9-yl]acetate (500.00 mg, 1.09 mmol, 1.0 equiv), Cs2CO3 (1.07 g, 3.28 mmol, 3.0 equiv) and [2-(2-aminophenyl)phenyl]-methylsulfonyloxy-palladium;ditert -butyl-[2-(2,4,6- triisopropylphenyl)phenyl]phosphane (86.9 mg, 109.41 μmol, 0.1 equiv) in dioxane (5 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 prep-HPLC (column: Waters Xbridge C18 150*50mm* 10um;mobile phase: [water( NH4HCO3)-ACN];B%: 61%-91%,10min).Compound tert-butyl 4-[2-[4-[(9S)-9-(2-tert-butoxy-2-oxo-ethyl)-4,5,13-trimethyl -3-thia-1,8,11,12- tetrazatricyclo[8.3.0.02,6]trideca-2(6),4,7,10,12-pentaen-7- yl]phenoxy]ethoxy]piperidine-1- carboxylate (400 mg, 600.74 μmol, 54.91% yield) was obtained as a yellow solid. [0424] Step 2: Preparation of tert-butyl (S)-2-(2,3,9-trimethyl-4-(4-(2-(piperidin-4- yloxy)ethoxy)phenyl-6H-thieno[3,2-f][1,2,4]triazolo[4,3-a][1 ,4]diazepin-6-yl)acetate. To a solution of tert-butyl 4-[2-[4-[(9S)-9-(2-tert-butoxy-2-oxo-ethyl)-4,5,13-trimethyl -3-thia- 1,8,11,12-tetrazatricyclo[8.3.0.02,6]trideca-2(6),4,7,10,12- pentaen-7- yl]phenoxy]ethoxy]piperidine-1-carboxylate (700 mg, 1.05 mmol, 1.0 equiv) in DCM (2 mL) was added TFA (2 mL). The mixture was stirred at 20 °C for 1 h. The reaction mixture was concentrated under reduced pressure to give tert-butyl (S)-2-(2,3,9-trimethyl-4-(4-(2-(piperidin- 4-yloxy)ethoxy)phenyl-6H-thieno[3,2-f][1,2,4]triazolo[4,3-a] [1,4]diazepin-6-yl)acetate (700 mg, 20% purity, TFA salt) as a yellow oil. [0425] Step 3: Preparation of tert-butyl 2-((S)-4-(4-(2-((1-(5-(((1r,3r)-3-(3-chloro-4- cyanophenoxy)-2,2,4,4-tetramethylcyclobutyl)carbamoyl)pyrimi din-2-yl)piperidin-4- yl)oxy)ethoxy)phenyl)-2,3,9-trimethyl-6H-thieno[3,2-f][1,2,4 ]triazolo[4,3-a][1,4]diazepin-6- yl)acetate (I-51). To a solution of tert-butyl (S)-2-(2,3,9-trimethyl-4-(4-(2-(piperidin-4- yloxy)ethoxy)phenyl-6H-thieno[3,2-f][1,2,4]triazolo[4,3-a][1 ,4]diazepin-6-yl)acetate (700 mg, 20% purity, TFA salt) in NMP (5 mL) was added K 2 CO 3 (609 mg, 4.41 mmol, 5.0 equiv) and 2- chloro-N-((1r,3r)-3-(3-chloro-4-cyanophenoxy)2,2,4,4-tetrame thylcyclobutyl)pyrimidine-5- carboxamide (443 mg, 1.06 mmol, 1.2 equiv). The mixture 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: Waters Xbridge 150*25mm* 5um; mobile phase: [water (NH4HCO3)-ACN]; B%: 40%-70%, 10 min) to give desired compound tert-butyl 2-((S)- 4-(4-(2-((1-(5-(((1r,3r)-3-(3-chloro-4-cyanophenoxy)-2,2,4,4 -tetramethylcyclobutyl) carbamoyl)pyrimidin-2-yl)piperidin-4-yl)oxy)ethoxy)phenyl)-2 ,3,9-trimethyl-6H-thieno[3,2- f][1,2,4]triazolo[4,3-a][1,4]diazepin-6-yl)acetate (82 mg, 86 μmol, 9% yield, 100% purity) was obtained as a white solid. 1 H NMR (400 MHz, CD3OD): δ 8.72 (s, 2H), 7.72 (d, J = 8.8 Hz, 1H), 7.40 (d, J = 8.7 Hz, 2H), 7.13 (d, J = 2.4 Hz, 1H), 7.03 - 6.93 (m, 3H), 4.51 (dd, J = 6.1, 8.5 Hz, 1H), 4.32 - 4.23 (m, 3H), 4.22 - 4.17 (m, 2H), 4.13 (s, 1H), 3.93 - 3.85 (m, 2H), 3.75 (m, 1H), 3.66 - 3.56 (m, 2H), 3.49 - 3.35 (m, 2H), 2.70 (s, 3H), 2.45 (s, 3H), 1.95 (m, 2H), 1.71 (s, 3H), 1.66 - 1.54 (m, 2H), 1.49 (s, 9H), 1.28 (s, 6H), 1.21 (s, 6H) LC-MS: MS (ES + ): RT = 3.131 min, m/z = =948.3 [M + H + ]; LCMS method: 25. EXAMPLE 17 – Synthesis of tert-butyl 2-[(9S)-7-[4-[4-[1-[5-[[3-(3-chloro-4-cyano- phenoxy)-2,2,4,4-tetramethyl-cyclobutyl]carbamoyl]pyrimidin- 2-yl]azetidin-3-yl]piperazin- 1-yl]phenyl]-4,5,13-trimethyl-3-thia-1,8,11,12-tetrazatricyc lo[8.3.0.02,6]trideca- 2(6),4,7,10,12-pentaen-9-yl]acetate (I-236) [0 426] Step 1: Preparation of tert-butyl 3-[4-[4-[(9S)-9-(2-tert-butoxy-2-oxo-ethyl)- 4,5,13-trimethyl-3-thia-1,8,11,12-tetrazatricyclo[8.3.0.0 2,6 ]trideca-2(6),4,7,10,12-pentaen-7- yl]phenyl]piperazin-1-yl]azetidine-1-carboxylate. To a solution of tert-butyl 2-[(9S)-7-(4- chlorophenyl)-4,5,13-trimethyl-3-thia-1,8,11,12-tetrazatricy clo[8.3.0.0 2,6 ]trideca-2(6),4,7,10,12- pentaen-9-yl]acetate (800 mg, 1.75 mmol, 1.0 equiv) and tert-butyl 3-piperazin-1-ylazetidine-1- carboxylate (633 mg, 2.63 mmol, 1.5 equiv) in dioxane (9 mL) was added Cs 2 CO 3 (1.71 g, 5.25 mmol, 3.0 equiv) and SPhos Pd G3 (136 mg, 175 μmol, 0.1 equiv). The mixture was stirred at 90 °C for 12 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 Na2SO4, filtered and concentrated in vacuum. The residue was purified by prep- HPLC (column: Phenomenex luna C18150*25mm*10um; mobile phase: [water(FA)-ACN];B%: 34%-64%,10min) to give tert-butyl 3-[4-[4-[(9S)-9-(2-tert-butoxy-2-oxo-ethyl)-4,5,13-trimethyl - 3-thia-1,8,11,12-tetrazatricyclo[8.3.0.0 2,6 ]trideca-2(6),4,7,10,12-pentaen-7-yl]phenyl]piperazin- 1-yl]azetidine-1-carboxylate (610 mg, 921 μmol, 53% yield) as a yellow solid. [0427] Step 2: Preparation of tert-butyl 2-[(9S)-7-[4-[4-(azetidin-3-yl)piperazin-1- yl]phenyl]-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. To a solution of tert-butyl 3-[4-[4-[(9S)-9-(2-tert-butoxy-2-oxo-ethyl)- 4,5,13-trimethyl-3-thia-1,8,11,12-tetrazatricyclo[8.3.0.0 2,6 ]trideca-2(6),4,7,10,12-pentaen-7- yl]phenyl]piperazin-1-yl]azetidine-1-carboxylate (610 mg, 921 μmol, 1.0 equiv) in DCM (3 mL) was added TFA (1 mL). The mixture was stirred at 20 °C for 2 h. After concentrated in vacuum, tert-butyl 2-[(9S)-7-[4-[4-(azetidin-3-yl)piperazin-1-yl]phenyl]-4,5,13 -trimethyl-3-thia- 1,8,11,12-tetrazatricyclo[8.3.0.0 2,6 ]trideca-2(6),4,7,10,12-pentaen-9-yl]acetate (207 mg, 368 μmol, crude) was obtained as a yellow oil and used for next step directly. [0428] Step 3: Preparation of tert-butyl 2-[(9S)-7-[4-[4-[1-[5-[[3-(3-chloro-4-cyano- phenoxy)-2,2,4,4-tetramethyl-cyclobutyl]carbamoyl]pyrimidin- 2-yl]azetidin-3-yl]piperazin- 1-yl]phenyl]-4,5,13-trimethyl-3-thia-1,8,11,12-tetrazatricyc lo[8.3.0.02,6]trideca- 2(6),4,7,10,12-pentaen-9-yl]acetate (I-52). To a solution of tert-butyl 2-[(9S)-7-[4-[4-(azetidin- 3-yl)piperazin-1-yl]phenyl]-4,5,13-trimethyl-3-thia-1,8,11,1 2-tetrazatricyclo[8.3.0.02,6]trideca- 2(6),4,7,10,12-pentaen-9-yl]acetate (103 mg, 183 μmol, 1.0 equiv), 2-chloro-N-[3-(3-chloro-4- cyano-phenoxy)-2,2,4,4-tetramethyl-cyclobutyl]pyrimidine-5-c arboxamide (92 mg, 0.22 mmol, 1.2 equiv) in NMP (2 mL) was added K2CO3 (76 mg, 0.55 mmol, 3.0 equiv). The mixture was stirred at 50 °C for 1 h. After filtration, the filtrate was concentrated to afford crude product. The residue was purified by prep-HPLC (column: Waters Xbridge 150*25mm*5um; mobile phase: [water(NH 4 HCO 3 )-ACN]; B%: 63%-93%,10min). Compound tert-butyl 2-[(9S)-7-[4-[4-[1-[5- [[3-(3-chloro-4-cyano-phenoxy)-2,2,4,4-tetramethyl-cyclobuty l]carbamoyl]pyrimidin-2- yl]azetidin-3-yl]piperazin-1-yl]phenyl]-4,5,13-trimethyl-3-t hia-1,8,11,12- tetrazatricyclo[8.3.0.02,6]trideca-2(6),4,7,10,12-pentaen-9- yl]acetate (19 mg, 20 μmol, 11% yield) was obtained as a yellow solid. 1 H NMR: (CD3OD, 400 MHz) 8.78 (m, 2H), 7.74-7.71(m, 1H), 7.38-7.36 (m, 2H), 7.15(m, 1H), 7.00-6.98(m,3H), 4.53(m, 1H), 4.37-4.32 (m, 3H), 4.17- 4.15 (m,3H), 3.44-3.12(m, 8H),2.74-2.70(m, 6H), 2.48(m, 3H),1.77(m,3H), 1.51 (m, 9H), 1.30 (m,6H), 1.21(m, 6H). LC-MS: MS (ES + ): RT = 2.315min, m/z = 944.3 [M + H] + LCMS: Method: 25. EXAMPLE 18 – Synthesis of N-[3-(3-chloro-4-cyano-phenoxy)-2,2,4,4-tetramethyl- cyclobutyl]-2-[4-[3-[4-[(9S)-9-[2-(ethylamino)-2-oxo-ethyl]- 4,5,13-trimethyl-3-thia- 1,8,11,12-tetrazatricyclo[8.3.0.02,6]trideca-2(6),4,7,10,12- pentaen-7-yl]phenyl]prop-2- ynyl]piperazin-1-yl]pyrimidine-5-carboxamide (I-54)

[0429] Step 1: Preparation of tert-butyl 4-[3-[4-[(9S)-9-(2-tert-butoxy-2-oxo-ethyl)- 4,5,13-trimethyl-3-thia-1,8,11,12-tetrazatricyclo[8.3.0.02,6 ]trideca-2(6),4,7,10,12-pentaen-7- yl]phenyl]prop-2-ynyl]piperazine-1-carboxylate. A mixture of tert-butyl 4-prop-2- ynylpiperazine-1-carboxylate (53 mg, 236 μmol, 1.8 equiv), tert-butyl 2-[(9S)-7-(4- chlorophenyl)-4,5,13-trimethyl-3-thia-1,8,11,12-tetrazatricy clo[8.3.0.0 2,6 ]trideca-2(6),4,7,10,12- pentaen-9-yl]acetate (60 mg, 131 μmol, 1.0 equiv), [2-(2-aminophenyl)phenyl]palladium(1+) ;bis(1-adamantyl)-butyl-phosphane;methanesulfonate (10 mg, 13 μmol, 0.1 equiv) and Cs2CO3(111 mg, 341 μmol, 2.6 equiv) in ACN (1 mL) and DMF (3 mL) was degassed and purged with N2 for 3 times. The mixture was stirred at 90 °C for 2 h under N2 atmosphere. The mixture was filtered and concentrated. The residue was purified by prep-HPLC (column: Waters Xbridge C18150*50 mm* 10 μm; mobile phase: [water(NH4HCO3)-ACN]; B%: 51%-81%, 10min) to give tert-butyl 4-[3-[4-[(9S)-9-(2-tert-butoxy-2-oxo-ethyl)-4,5,13-trimethyl -3-thia- 1,8,11,12-tetrazatricyclo[8.3.0.0 2,6 ]trideca-2(6),4,7,10,12-pentaen-7-yl]phenyl]prop-2- ynyl]piperazine-1-carboxylate (50 mg, 59% yield). [0430] Step 2: Preparation of (S)-2-(2,3,9-trimethyl-4-(4-(3-(piperazin-1-yl)prop-1-yn-1- yl)phenyl)-6H-thieno[3,2-f][1,2,4]triazolo[4,3-a][1,4]diazep in-6-yl)acetic acid. To a solution of tert-butyl 4-[3-[4-[(9S)-9-(2-tert-butoxy-2-oxo-ethyl)-4,5,13-trimethyl -3-thia-1,8,11,12- tetrazatricyclo[8.3.0.0 2,6 ]trideca-2(6),4,7,10,12-pentaen-7-yl]phenyl]prop-2-yny l]piperazine-1- carboxylate (300 mg, 465 μmol, 1.0 equiv) in DCM (5 mL) was added TFA (0.5 mL). The mixture was stirred at 25 °C for 1 h. The reaction mixture was concentrated under reduced pressure to give a crude product. [0431] Step 3: Preparation of 2-[(9S)-7-[4-[3-[4-[5-[[3-(3-chloro-4-cyano-phenoxy)- 2,2,4,4-tetramethyl-cyclobutyl]carbamoyl]pyrimidin-2-yl]pipe razin-1-yl]prop-1- ynyl]phenyl]-4,5,13-trimethyl-3-thia-1,8,11,12-tetrazatricyc lo[8.3.0.0 2,6 ]trideca- 2(6),4,7,10,12-pentaen-9-yl]acetic acid. To a solution of 2-[(9S)-4,5,13-trimethyl-7-[4-(3- piperazin-1-ylprop-1-ynyl)phenyl]-3-thia-1,8,11,12-tetrazatr icyclo[8.3.0.0 2,6 ]trideca- 2(6),4,7,10,12-pentaen-9-yl]acetic acid (140 mg, 232 μmol, 1.0 equiv, TFA salt) in NMP (1 mL) was added K 2 CO 3 (96 mg, 697 μmol, 3.0 equiv) and 2-chloro-N-[3-(3-chloro-4-cyano-phenoxy)- 2,2,4,4-tetramethyl-cyclobutyl]pyrimidine-5-carboxamide (97 mg, 232 μmol, 1.0 equiv). The mixture was stirred at 50 °C for 2 h and concentrated. The residue was purified by prep-HPLC (column: Waters Xbridge 150*25 mm*5 μm; mobile phase: [water(NH 4 HCO 3 )-ACN]; B%: 30%-60%, 8min) to give 2-[(9S)-7-[4-[3-[4-[5-[[3-(3-chloro-4-cyano-phenoxy)-2,2,4,4 - tetramethyl-cyclobutyl]carbamoyl]pyrimidin-2-yl]piperazin-1- yl]prop-1-ynyl]phenyl]-4,5,13- trimethyl-3-thia-1,8,11,12-tetrazatricyclo[8.3.0.0 2,6 ]trideca-2(6),4,7,10,12-pentaen-9-yl]acetic acid (100 mg, 49% yield). [0432] Step 4: Preparation of N-[3-(3-chloro-4-cyano-phenoxy)-2,2,4,4-tetramethyl- cyclobutyl]-2-[4-[3-[4-[(9S)-9-[2-(ethylamino)-2-oxo-ethyl]- 4,5,13-trimethyl-3-thia- 1,8,11,12-tetrazatricyclo[8.3.0.02,6]trideca-2(6),4,7,10,12- pentaen-7-yl]phenyl]prop-2- ynyl]piperazin-1-yl]pyrimidine-5-carboxamide (I-54). To a solution of 2-[(9S)-7-[4-[3-[4-[5- [[3-(3-chloro-4-cyano-phenoxy)-2,2,4,4-tetramethyl-cyclobuty l]carbamoyl]pyrimidin-2- yl]piperazin-1-yl]prop-1-ynyl]phenyl]-4,5,13-trimethyl-3-thi a-1,8,11,12-tetrazatricyclo [8.3.0.02,6]trideca-2(6),4,7,10,12-pentaen-9-yl]acetic acid (100 mg, 115 μmol, 1.0 equiv) in DMF (1 mL) was added ethanamine (16 mg, 344 μmol, 3.0 equiv), DIEA (74 mg, 574 μmol, 5.0 equiv) and HATU (57 mg, 149 μmol, 1.3 equiv). The mixture was stirred at 25 °C for 0.5 h and concentrated. The residue was purified by prep-HPLC (column: Waters Xbridge 150*25 mm*5 μm; mobile phase: [water(NH 4 HCO 3 )-ACN]; B%: 53%-83%, 8min) to give N-[3-(3-chloro-4- cyano-phenoxy)-2,2,4,4-tetramethyl-cyclobutyl]-2-[4-[3-[4-[( 9S)-9-[2-(ethylamino)-2-oxo- ethyl]-4,5,13-trimethyl-3-thia-1,8,11,12-tetrazatricyclo[8.3 .0.0 2,6 ]trideca-2(6),4,7,10,12-pentaen- 7-yl]phenyl]prop-2-ynyl]piperazin-1-yl]pyrimidine-5-carboxam ide (66 mg, 64% yield). 1 H NMR (400 MHz, CDCl 3 ): δ 8.71 (s, 2 H), 7.58 (d, J = 8.8 Hz, 1 H), 7.37-7.44 (m, 4 H), 6.97 (d, J = 2.6 Hz, 1 H), 6.79-6.82 (m, 1 H), 6.40 (t, J = 5.2 Hz, 1 H), 5.94 (d, J = 8.0 Hz, 1 H), 4.61-4.64 (m, 1 H), 4.13 (d, J = 8.2 Hz, 1 H), 3.96-4.07 (m, 5 H), 3.51-3.64 (m, 3 H), 3.20-3.44 (m, 3 H), 2.71 (t, J = 5.0 Hz, 4 H), 2.67 (s, 3 H), 2.40 (s, 3 H), 1.24 (d, J = 12.2 Hz, 12 H), 1.18 (t, J = 7.4 Hz, 3 H). LC-MS: MS (ES + ): RT = 2.051 min, m/z = 898.5 [M+H] + . LCMS Method: 25. EXAMPLE 19 – Synthesis of N-((1r,3r)-3-(3-chloro-4-cyanophenoxy)-2,2,4,4- tetramethylcyclobutyl)-2-(4-(2-(4-((S)-6-(2-(ethylamino)-2-o xoethyl)-2,3,9-trimethyl-6H- thieno[3,2-f][1,2,4]triazolo[4,3-a][1,4]diazepin-4-yl)phenox y)ethoxy)piperidin-1- yl)pyrimidine-5-carboxamide (I-60) [ 0 33] Step : reparat on o tert-buty (S)- -( -( -(6-( -(tert-butoxy)- -oxoet y )- ,3,9- trimethyl-6H-thieno[3,2-f][1,2,4]triazolo[4,3-a][1,4]diazepi n-4-yl)phenoxy)ethoxy) piperidine-1-carboxylate. A mixture of tert-butyl (S)-2-(4-(4-chlorophenyl)-2,3,9-trimethyl-6H- thieno[3,2-f][1,2,4]triazolo[4,3-a][1,4]diazepin-6-yl)acetat e (200 mg, 437 μmol, 1.0 equiv), tert- butyl 4-(2-hydroxyethoxy)piperidine-1-carboxylate (161 mg, 656 μmol, 1.5 equiv), Cs2CO3 (427 mg, 1.31 mmol, 3.0 equiv) and [2-(2-aminophenyl)phenyl]-methylsulfonyloxy-palladium; ditert- butyl-[2-(2,4,6-triisopropylphenyl)phenyl]phosphane (34 mg, 43 μmol, 0.1 equiv) in dioxane (2 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 90 °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 prep-HPLC (column: Waters Xbridge C18150*50mm*10um; mobile phase: [water (NH4HCO3)-ACN]; B%: 53%-83%, 10 min). tert- butyl (S)-4-(2-(4-(6-(2-(tert-butoxy)-2-oxoethyl)-2,3,9-trimethyl- 6H-thieno[3,2- f][1,2,4]triazolo[4,3-a][1,4]diazepin-4-yl)phenoxy)ethoxy)pi peridine-1-carboxylate (150 mg, 225 μmol, 51% yield) was obtained as a white solid. [0434] Step 2: Preparation of (S)-2-(2,3,9-trimethyl-4-(4-(2-(piperidin-4-yloxy)ethoxy) phenyl)-6H-thieno[3,2-f][1,2,4]triazolo[4,3-a][1,4]diazepin- 6-yl)acetic acid. To a solution of tert-butyl (S)-4-(2-(4-(6-(2-(tert-butoxy)-2-oxoethyl)-2,3,9-trimethyl- 6H-thieno[3,2- f][1,2,4]triazolo[4,3-a][1,4]diazepin-4-yl)phenoxy)ethoxy)pi peridine-1-carboxylate (100 mg, 150 μmol, 1.0 equiv) in DCM (1 mL) was added TFA (0.5 mL). The mixture was stirred at 20 °C for 1 h. The reaction mixture was concentrated under reduced pressure to give (S)-2-(2,3,9- trimethyl-4-(4-(2-(piperidin-4-yloxy)ethoxy)phenyl)-6H-thien o[3,2-f][1,2,4]triazolo[4,3- a][1,4]diazepin-6-yl)acetic acid (94 mg, TFA salt) as a yellow oil. [0435] Step 3: Preparation of 2-((S)-4-(4-(2-((1-(5-(((1r,3r)-3-(3-chloro-4-cyanophenoxy)- 2,2,4,4-tetramethylcyclobutyl)carbamoyl)pyrimidin-2-yl)piper idin-4-yl)oxy)ethoxy)phenyl) -2,3,9-trimethyl-6H-thieno[3,2-f][1,2,4]triazolo[4,3-a][1,4] diazepin-6-yl)acetic acid. To a solution of S)-2-(2,3,9-trimethyl-4-(4-(2-(piperidin-4-yloxy)ethoxy)phen yl)-6H-thieno[3,2- f][1,2,4]triazolo[4,3-a][1,4]diazepin-6-yl)acetic acid (550 mg, 882 μmol, 1.0 equiv, TFA salt) in NMP (5 mL) was added K2CO3 (609 mg, 4.41 mmol, 5.0 equiv) and 2-chloro-N-((1r,3r)-3-(3- chloro-4-cyanophenoxy)-2,2,4,4-tetramethylcyclobutyl)pyrimid ine-5-carboxamide (443 mg, 1.06 mmol, 1.2 equiv). The mixture 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: Waters Xbridge 150*25mm* 5um; mobile phase: [water (NH 4 HCO 3 )-ACN]; B%: 40%-70%, 10 min) to give desired 2-((S)-4-(4-(2-((1-(5-(((1r,3r)-3-(3-chloro-4- cyanophenoxy)-2,2,4,4-tetramethylcyclobutyl)carbamoyl)pyrimi din-2-yl)piperidin-4-yl)oxy) ethoxy)phenyl)-2,3,9-trimethyl-6H-thieno[3,2-f][1,2,4]triazo lo[4,3-a][1,4]diazepin-6-yl)acetic acid (600 mg, 672 μmol, 76% yield, 100% purity) was obtained as a white solid. [0436] Step 4: Preparation of N-((1r,3r)-3-(3-chloro-4-cyanophenoxy)-2,2,4,4- tetramethylcyclobutyl)-2-(4-(2-(4-((S)-6-(2-(ethylamino)-2-o xoethyl)-2,3,9-trimethyl-6H- thieno[3,2-f][1,2,4]triazolo[4,3-a][1,4]diazepin-4-yl)phenox y)ethoxy)piperidin-1- yl)pyrimidine-5-carboxamide (I-60). To a solution of 2-((S)-4-(4-(2-((1-(5-(((1r,3r)-3-(3- chloro-4-cyanophenoxy)-2,2,4,4-tetramethylcyclobutyl)carbamo yl)pyrimidin-2-yl)piperidin-4- yl)oxy)ethoxy)phenyl)-2,3,9-trimethyl-6H-thieno[3,2-f][1,2,4 ]triazolo[4,3-a][1,4]diazepin-6- yl)acetic acid (300 mg, 336 μmol, 1.0 equiv) and ethanamine (75 mg, 1.68 mmol, 5.0 equiv) in DMF (4 mL) was added HATU (639 mg, 1.68 mmol, 5.0 equiv) and DIEA (217 mg, 1.68 mmol, 5.0 equiv) .The mixture was stirred at 20 °C for 0.5 h. The reaction mixture was 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( NH4HCO3)-ACN];B%: 48%- 78%, 10 min) to give compound N-((1r,3r)-3-(3-chloro-4-cyanophenoxy)-2,2,4,4- tetramethylcyclobutyl)-2-(4-(2-(4-((S)-6-(2-(ethylamino)-2-o xoethyl)-2,3,9-trimethyl-6H- thieno[3,2-f][1,2,4]triazolo[4,3-a][1,4]diazepin-4-yl)phenox y)ethoxy)piperidin-1-yl)pyrimidine- 5-carboxamide (178 mg, 194 μmol, 57% yield, 100% purity) as a yellow solid. 1 H NMR (400 MHz, CD 3 OD):δ 8.72 (s, 2H), 7.72 (d, J = 8.7 Hz, 1H), 7.39 (d, J = 8.7 Hz, 2H), 7.12 (d, J = 2.4 Hz, 1H), 7.01 - 6.93 (m, 3H), 4.63 - 4.56 (m, 1H), 4.32 - 4.23 (m, 3H), 4.19 (dd, J = 3.7, 5.3 Hz, 2H), 4.12 (s, 1H), 3.94 - 3.86 (m, 2H), 3.75 (tt, J = 3.5, 7.6 Hz, 1H), 3.66 - 3.55 (m, 2H), 3.41 - 3.34 (m, 1H), 3.29 - 3.22 (m, 3H), 2.69 (s, 3H), 2.44 (s, 3H), 1.94 (ddd, J = 3.1, 6.3, 9.7 Hz, 2H), 1.70 (s, 3H), 1.65 - 1.52 (m, 2H), 1.27 (s, 6H), 1.22 - 1.16 (m, 9H). LC-MS: MS (ES + ): RT = 2.773 min, m/z = 919.2 [M + H + ]; LCMS method: 25.

EXAMPLE 20 – Synthesis of N-[3-(3-chloro-4-cyano-phenoxy)-2,2,4,4-tetramethyl- cyclobutyl]-2-[4-[3-[4-[(9S)-9-[2-(ethylamino)-2-oxo-ethyl]- 4,5,13-trimethyl-3-thia- 1,8,11,12-tetrazatricyclo[8.3.0.02,6]trideca-2(6),4,7,10,12- pentaen-7-yl]phenyl]propoxy]-1- piperidyl]pyrimidine-5-carboxamide (I-87) [ 0437] Step 1: Preparation of tert-butyl 4-[3-[4-[(9S)-9-(2-tert-butoxy-2-oxo-ethyl)- 4,5,13-trimethyl-3-thia-1,8,11,12-tetrazatricyclo[8.3.0.02,6 ]trideca-2(6),4,7,10,12-pentaen-7- yl]phenyl]prop-2-ynoxy]piperidine-1-carboxylate. A mixture of tert-butyl 2-[(9S)-7-(4- chlorophenyl)-4,5,13-trimethyl-3-thia-1,8,11,12-tetrazatricy clo[8.3.0.02,6]trideca-2(6),4,7,10,12- pentaen-9-yl]acetate (800 mg, 1.75 mmol, 1.0 equiv), tert-butyl 4-prop-2-ynoxypiperidine-1- carboxylate (649 mg, 2.71 mmol, 1.6 equiv), Ad2nBuP Pd G3(cataCXium® A Pd G3) (127 mg, 175 μmol, 0.1 equiv) and Cs2CO3 (1.48 g, 4.55 mmol, 2.6 equiv) in ACN (4 mL) and DMF (12 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 90 °C for 1 h under N2 atmosphere. The mixture was poured into water (80 mL). The aqueous phase was extracted with ethyl acetate (80 mL x 2). The combined organic phase was washed with brine (70 mL x 2), dried with anhydrous Na2SO4, filtered and concentrated in vacuum. The residue was purified by silica gel chromatography (0% - 100% ethyl acetate in petroleum ether) and further purified by prep-HPLC (column: Phenomenex luna C18150*40mm* 15um; mobile phase: [water (FA) - ACN]; B%: 62%-92%, 10min). Compound tert-butyl 4-[3-[4-[(9S)-9-(2- tert-butoxy-2-oxo-ethyl)-4,5,13-trimethyl-3-thia-1,8,11,12-t etrazatricyclo[8.3.0.02,6]trideca- 2(6),4,7,10,12-pentaen-7-yl]phenyl]prop-2-ynoxy]piperidine-1 -carboxylate (270 mg, 409 μmol, 23% yield) was obtained as a light yellow solid. [0438] Step 2: Preparation of tert-butyl 4-[3-[4-[(9S)-9-(2-tert-butoxy-2-oxo-ethyl)- 4,5,13-trimethyl-3-thia-1,8,11,12-tetrazatricyclo[8.3.0.02,6 ]trideca-2(6),4,7,10,12-pentaen-7- yl]phenyl]propoxy]piperidine-1-carboxylate. To a solution of tert-butyl 4-[3-[4-[(9S)-9-(2- tert-butoxy-2-oxo-ethyl)-4,5,13-trimethyl-3-thia-1,8,11,12-t etrazatricyclo[8.3.0.02,6]trideca- 2(6),4,7,10,12-pentaen-7-yl]phenyl]prop-2-ynoxy]piperidine-1 -carboxylate (260 mg, 394 μmol, 1.0 equiv) in CF 3 CH 2 OH (10 mL) was added Pd/C (220 mg, 10% purity) under N2 atmosphere. The suspension was degassed and purged with H 2 for 3 times. The mixture was stirred under H 2 (15 Psi) at 25 °C for 2 h. The mixture was filtered and concentrated under reduced pressure. Compound tert-butyl 4-[3-[4-[(9S)-9-(2-tert-butoxy-2-oxo-ethyl)-4,5,13-trimethyl -3-thia- 1,8,11,12-tetrazatricyclo[8.3.0.02,6]trideca-2(6),4,7,10,12- pentaen-7-yl]phenyl]propoxy] piperidine-1-carboxylate (260 mg, 392 μmol, 99% yield) was obtained as a yellow gum. [0439] Step 3: Preparation of 2-[(9S)-4,5,13-trimethyl-7-[4-[3-(4-piperidyloxy) propyl]phenyl]-3-thia-1,8,11,12-tetrazatricyclo[8.3.0.02,6]t rideca-2(6),4,7,10,12-pentaen-9- yl]acetic acid. To a solution of tert-butyl 4-[3-[4-[(9S)-9-(2-tert-butoxy-2-oxo-ethyl)-4,5,13- trimethyl-3-thia-1,8,11,12-tetrazatricyclo[8.3.0.02,6]tridec a-2(6),4,7,10,12-pentaen-7- yl]phenyl]propoxy]piperidine-1-carboxylate (270 mg, 406 μmol, 1.0 equiv) in DCM (3 mL) was added TFA (4.62 g, 40.5 mmol, 3 mL). The mixture was stirred at 25 °C for 3 h. The mixture was concentrated under reduced pressure. Compound 2-[(9S)-4,5,13-trimethyl-7-[4-[3-(4- piperidyloxy)propyl]phenyl]-3-thia-1,8,11,12-tetrazatricyclo [8.3.0.02,6] trideca-2(6),4,7,10,12- pentaen-9-yl]acetic acid (250 mg, 402 μmol, 99% yield, TFA salt) was obtained as a yellow gum. [0440] Step 4: Preparation of 2-[(9S)-7-[4-[3-[[1-[5-[[3-(3-chloro-4-cyano-phenoxy)- 2,2,4,4-tetramethyl-cyclobutyl]carbamoyl]pyrimidin-2-yl]-4-p iperidyl]oxy]propyl]phenyl]- 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]acetic acid. To a solution of 2-[(9S)-4,5,13-trimethyl-7-[4-[3-(4-piperidyloxy)propyl]phen yl]- 3-thia-1,8,11,12-tetrazatricyclo[8.3.0.02,6]trideca-2(6),4,7 ,10,12-pentaen-9-yl]acetic acid (125 mg, 201 μmol, 1.0 equiv, TFA salt) in NMP (2 mL) was added K2CO3 (278 mg, 2.01 mmol, 10 equiv) and 2-chloro-N-[3-(3-chloro-4-cyano-phenoxy)-2,2,4,4-tetramethyl -cyclobutyl] pyrimidine-5-carboxamide (84 mg, 0.20 mmol, 1.0 equiv). The mixture was stirred at 50 °C for 3 h. The mixture was filtered. The residue was purified by prep-HPLC (column: Phenomenex luna C18150*25mm* 10um; mobile phase: [water (FA) - ACN]; B%: 69%-99%, 2min). Compound 2-[(9S)-7-[4-[3-[[1-[5-[[3-(3-chloro-4-cyano-phenoxy)-2,2,4, 4-tetramethyl-cyclobutyl] carbamoyl]pyrimidin-2-yl]-4-piperidyl]oxy]propyl]phenyl]-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]acetic acid (120 mg, 135 μmol, 67% yield) was obtained as a pink solid. [0441] Step 5: Preparation of N-[3-(3-chloro-4-cyano-phenoxy)-2,2,4,4-tetramethyl- cyclobutyl]-2-[4-[3-[4-[(9S)-9-[2-(ethylamino)-2-oxo-ethyl]- 4,5,13-trimethyl-3-thia- 1,8,11,12-tetrazatricyclo[8.3.0.02,6]trideca-2(6),4,7,10,12- pentaen-7-yl]phenyl]propoxy]-1- piperidyl]pyrimidine-5-carboxamide (I-87). To a solution of 2-[(9S)-7-[4-[3-[[1-[5-[[3-(3- chloro-4-cyano-phenoxy)-2,2,4,4-tetramethyl-cyclobutyl]carba moyl]pyrimidin-2-yl]-4- piperidyl]oxy]propyl]phenyl]-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]acetic acid (120 mg, 135 μmol, 1.0 equiv) and ethanamine (122 mg, 2.70 mmol, 20 equiv) in DMF (2 mL) was added HATU (102 mg, 270 μmol, 2.0 equiv) and DIEA (87 mg, 0.67 mmol, 5.0 equiv). The mixture was stirred at 25 °C for 48 h. The mixture was filtered. The residue was purified by prep-HPLC (column: Waters Xbridge C18150*50mm* 10um; mobile phase: [water (NH4HCO3) - ACN]; B%: 58%-88%, 10min). Compound N-[3-(3- chloro-4-cyano-phenoxy)-2,2,4,4-tetramethyl-cyclobutyl]-2-[4 -[3-[4-[(9S)-9-[2-(ethylamino)-2- oxo-ethyl]-4,5,13-trimethyl-3-thia-1,8,11,12-tetrazatricyclo [8.3.0.02,6]trideca-2(6),4,7,10,12- pentaen-7-yl]phenyl]propoxy]-1-piperidyl]pyrimidine-5-carbox amide (87 mg, 92 μmol, 68% yield, 97% purity) was obtained as a white solid. 1 H NMR (400 MHz, MeOD): δ 8.73 (s, 2H), 7.73 (d, J = 8.8 Hz, 1H), 7.37 (d, J = 8.4 Hz, 2H), 7.26 (d, J = 8.4 Hz, 2H), 7.13 (d, J = 2.4 Hz, 1H), 6.98 (dd, J = 2.4, 8.8 Hz, 1H), 4.63 (t, J = 4.4 Hz, 1H), 4.35-4.28 (m, 2H), 4.27 (s, 1H), 4.13 (s, 1H), 3.63-3.49 (m, 5H), 3.44-3.37 (m, 1H), 3.36-3.33 (m, 1H), 3.30-3.24 (m, 2H), 2.76 (t, J = 7.6 Hz, 2H), 2.70 (s, 3H), 2.45 (s, 3H), 1.96-1.87 (m, 4H), 1.68 (s, 3H), 1.58-1.48 (m, 2H), 1.28 (s, 6H), 1.21 (s, 6H), 1.20-1.17 (m, 3H). LC-MS: MS (ES + ): RT = 2.995 min, m/z = 917.2 [M + H + ]; LCMS method: 25. EXAMPLE 21 – Synthesis of N-[3-(3-chloro-4-cyano-phenoxy)-2,2,4,4-tetramethyl- cyclobutyl]-2-[4-[[4-[4-[(9S)-9-[2-(ethylamino)-2-oxo-ethyl] -4,5,13-trimethyl-3-thia- 1,8,11,12-tetrazatricyclo[8.3.0.02,6]trideca-2(6),4,7,10,12- pentaen-7-yl]phenyl]piperazin-1- yl]methyl]-1-piperidyl]pyrimidine-5-carboxamide (I-105)

[0442] Step 1: Preparation of tert-butyl 4-[[4-[4-[(9S)-9-(2-tert-butoxy-2-oxo-ethyl)- 4,5,13-trimethyl-3-thia-1,8,11,12-tetrazatricyclo[8.3.0.02,6 ]trideca-2(6),4,7,10,12-pentaen-7- yl]phenyl]piperazin-1-yl]methyl]piperidine-1-carboxylate. To a solution of tert-butyl 2-[(9S)- 7-(4-chlorophenyl)-4,5,13-trimethyl-3-thia-1,8,11,12-tetraza tricyclo[8.3.0.02,6]trideca- 2(6),4,7,10,12-pentaen-9-yl]acetate (200 mg, 438 μmol, 1.00 equiv) in dioxane (2 mL) was added Cs2CO3 (428 mg, 1.31 mmol, 3.00 equiv) , tert-butyl 4-(piperazin-1-ylmethyl)piperidine- 1-carboxylate (186 mg, 656 μmol, 1.50 equiv) and SPhos Pd G3 (34.2 mg, 43.8 μmol, 0.10 equiv) .The mixture was stirred at 90 °C for 12 hours . The reaction mixture was filtered and the filter was concentrated. The residue was purified by prep-HPLC(column: Phenomenex luna C18 150*25mm* 10um;mobile phase: [water(FA)-ACN];B%: 18%-48%,2min) to give tert-butyl 4- [[4-[4-[(9S)-9-(2-tert-butoxy-2-oxo-ethyl)-4,5,13-trimethyl- 3-thia-1,8,11,12-tetrazatricyclo [8.3.0.02,6]trideca-2(6),4,7,10,12-pentaen-7-yl]phenyl]piper azin-1-yl]methyl]piperidine-1- carboxylate (190 mg, 270 μmol, 62% yield) as a yellow solid. [0443] Step 2: Preparation of crude 6-piperazin-1-ylpyridine-3-carbonitrile. To a solution of tert-butyl 4-[[4-[4-[(9S)-9-(2-tert-butoxy-2-oxo-ethyl)-4,5,13-trimethy l-3-thia- 1,8,11,12-tetrazatricyclo[8.3.0.02,6]trideca-2(6),4,7,10,12- pentaen-7-yl]phenyl]piperazin-1- yl]methyl]piperidine-1-carboxylate (190 mg, 270 μmol, 1.00 equiv) in DCM (2 mL) was added TFA (1.39 g, 12.2 mmol, 0.90 mL, 45.0 equiv). The mixture was stirred at 25 °C for 8 hours. The reaction mixture was concentrated to give crude 6-piperazin-1-ylpyridine-3-carbonitrile (11.7 g, crude, HCl salt). [0444] Step 3: Preparation of 2-[(9S)-7-[4-[4-[[1-[5-[[3-(3-chloro-4-cyano-phenoxy)- 2,2,4,4-tetramethyl-cyclobutyl]carbamoyl]pyrimidin-2-yl]-4-p iperidyl]methyl]piperazin-1- yl]phenyl]-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]acetic acid. To a solution of 2-[(9S)-4,5,13-trimethyl-7-[4-[4-(4-piperidylmethyl )piperazin-1-yl]phenyl]-3-thia-1,8,11,12-tetrazatricyclo[8.3 .0.02,6]trideca-2(6),4,7,10,12- pentaen-9-yl]acetic acid (140 mg, 256 μmol, 1.00 equiv) in NMP was added K2CO3 (35.3 mg, 256 μmol, 1.00 equiv) and 2-chloro-N-[3-(3-chloro-4-cyano-phenoxy)-2,2,4,4-tetramethyl - cyclobutyl]pyrimidine-5-carboxamide (107 mg, 256 μmol, 1.00 equiv). The mixture was stirred at 50 °C for 2 hours . The reaction mixture was filtered and purified by prep-HPLC (column: Phenomenex Synergi C18150*25mm* 10um;mobile phase: [water(FA)-ACN];B%: 23%- 53%,10min) to give 2-[(9S)-7-[4-[4-[[1-[5-[[3-(3-chloro-4-cyano-phenoxy)-2,2,4, 4-tetramethyl- cyclobutyl]carbamoyl]pyrimidin-2-yl]-4-piperidyl]methyl]pipe razin-1-yl]phenyl]-4,5,13- trimethyl-3-thia-1,8,11,12-tetrazatricyclo[8.3.0.02,6]tridec a-2(6),4,7,10,12-pentaen-9-yl]acetic acid (130 mg, 139.70 μmol, 55% yield) as a yellow solid. [0445] Step 4: Preparation of N-[3-(3-chloro-4-cyano-phenoxy)-2,2,4,4-tetramethyl- cyclobutyl]-2-[4-[[4-[4-[(9S)-9-[2-(ethylamino)-2-oxo-ethyl] -4,5,13-trimethyl-3-thia- 1,8,11,12-tetrazatricyclo[8.3.0.02,6]trideca-2(6),4,7,10,12- pentaen-7-yl]phenyl]piperazin-1- yl]methyl]-1-piperidyl]pyrimidine-5-carboxamide (I-105). To a solution of 2-[(9S)-7-[4-[4- [[1-[5-[[3-(3-chloro-4-cyano-phenoxy)-2,2,4,4-tetramethyl-cy clobutyl]carbamoyl]pyrimidin-2- yl]-4-piperidyl]methyl]piperazin-1-yl]phenyl]-4,5,13-trimeth yl-3-thia-1,8,11,12-tetrazatricyclo [8.3.0.02,6]trideca-2(6),4,7,10,12-pentaen-9-yl]acetic acid (130 mg, 140 μmol, 1.00 equiv) in DMF (1 mL) was added DIEA (90.3 mg, 699 μmol, 122 μL, 5.00 equiv), ethanamine (18.9 mg, 419 μmol, 27.4 μL, 3.00 equiv) and HATU (159 mg, 419 μmol, 3.00 equiv) .The mixture was stirred at 25 °C for 1 hour. The reaction mixture was filtered and purified by prep- HPLC(column: Waters Xbridge 150*25mm* 5um;mobile phase: [water( NH4HCO3)- ACN];B%: 57%-87%,8min) to give N-[3-(3-chloro-4-cyano-phenoxy)-2,2,4,4-tetramethyl- cyclobutyl]-2-[4-[[4-[4-[(9S)-9-[2-(ethylamino)-2-oxo-ethyl] -4,5,13-trimethyl-3-thia-1,8,11,12- tetrazatricyclo[8.3.0.02,6]trideca-2(6),4,7,10,12-pentaen-7- yl]phenyl]piperazin-1-yl]methyl]-1- piperidyl]pyrimidine-5-carboxamide (80.0 mg, 84 μmol, 60% yield) as a white solid. 1 H NMR (400 MHz, CD3OD): δ 8.74 (s, 2H), 7.74 (d, J = 8.7 Hz, 1H), 7.35 (d, J = 8.7 Hz, 2H), 7.14 (d, J = 2.4 Hz, 1H), 7.00 (m, 1H), 6.95 (d, J = 9.0 Hz, 2H), 4.62 - 4.55 (m, 2H), 4.29 (s, 1H), 4.15 (s, 1H), 3.43 - 3.34 (m, 3H), 3.31 - 3.23 (m, 7H), 3.07 - 2.97 (m, 2H), 2.71 (s, 3H), 2.61 (d, J = 4.6 Hz, 3H), 2.47 (s, 3H), 2.31 (d, J = 6.4 Hz, 2H), 2.05 (s, 1H), 1.93 (d, J = 12.5 Hz, 2H), 1.76 (s, 3H), 1.30 (m,5H), 1.25 - 1.15 (m, 12H). LC-MS: MS (ES + ): RT = 2.115 min, m/z = 957.3 [M + H + ]; LCMS method: 25. EXAMPLE 22 – Synthesis of N-[3-(3-chloro-4-cyano-phenoxy)-2,2,4,4-tetramethyl- cyclobutyl]-2-[4-[3-[4-[(9S)-4,5,13-trimethyl-9-(oxazol-2-yl methyl)-3-thia-1,8,11,12- tetrazatricyclo[8.3.0.02,6]trideca-2(6),4,7,10,12-pentaen-7- yl]phenyl]propoxy]-1- piperidyl]pyrimidine-5-carboxamide (I-131)

[0446] Step 1: Preparation of 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]acetic acid. 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 (1.0 g, 2.1 mmol, 1.0 equiv) in DCM (10 mL) was added TFA (7.7 g, 67 mmol, 5.0 mL, 30 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 2-[(9S)- 7-(4-chlorophenyl)-4,5,13-trimethyl-3-thia-1,8,11,12-tetraza tricyclo[8.3.0.02,6]trideca- 2(6),4,7,10,12-pentaen-9-yl]acetic acid (877 mg, 99% yield).

[0447] Step 2: Preparation of 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]acetamide. To a solution of 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]acetic acid (877 mg, 2.19 mmol, 1.0 equiv) and NH4Cl (351 mg, 6.56 mmol, 3.0 equiv) in DMF (8 mL) was added HATU (915 mg, 2.41 mmol, 1.1 equiv) and DIEA (848 mg, 6.56 mmol, 3.0 equiv). The mixture was stirred at 25 °C for 1 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%: 24%- 54%,10min) to give 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]acetamide (740 mg, 84% yield). [0448] Step 3: Preparation of 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]methyl]oxazole. To a solution of 1,3-dioxol-2-one (103 mg, 1.20 mmol, 1.2 equiv) and 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]acetamide (400 mg, 1.00 mmol, 1.0 equiv) in PPA (4 mL). The mixture was stirred at 160 °C for 3 h. The mixture was quenched by addition H 2 O 50 mL at 0°C, and then diluted with EA 200 mL. The mixture was extracted with H 2 O (15 mL * 2). The combined organic layers were washed with H 2 O (10 mL * 3), dried over [Na2SO4], filtered and concentrated. The residue was purified by column chromatography (SiO 2 , Petroleum ether/Ethyl acetate=2/1 to 0/1) to give compound 2-[[(9S)-7-(4-chlorophenyl)-4,5,13-trimethyl-3-thia-1,8,11,1 2-tetrazatricyclo [8.3.0.02,6]trideca-2(6),4,7,10,12-pentaen-9-yl]methyl]oxazo le (260 mg, 61% yield). [0449] Step 4: Preparation of tert-butyl 4-[3-[4-[(9S)-4,5,13-trimethyl-9-(oxazol-2- ylmethyl)-3-thia-1,8,11,12-tetrazatricyclo[8.3.0.02,6]tridec a-2(6),4,7,10,12-pentaen-7- yl]phenyl]prop-2-ynoxy]piperidine-1-carboxylate. A mixture of 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]methyl]oxazole (100 mg, 235 μmol, 1.0 equiv), tert-butyl 4-prop-2-ynoxypiperidine-1- carboxylate (169 mg, 707 μmol, 3.0 equiv), Cs 2 CO 3 (153 mg, 471 μmol, 2.0 equiv) , [2-(2- aminophenyl)phenyl]-methylsulfonyloxy-palladium;2-(2-dicyclo hexylphosphanylphenyl)-N,N- dimethyl-aniline (18 mg, 23.5 μmol, 0.1 equiv) in MeCN (3 mL) was degassed and purged with N2 for 3 times and the mixture was stirred at 90 °C for 2 h under N2 atmosphere. The mixture was filtered and concentrated. The residue was purified by prep-HPLC (column: Phenomenex luna C18150*25mm* 10um; mobile phase: [water (FA)-ACN]; B%: 61%-91%, 11min) to give compound tert-butyl 4-[3-[4-[(9S)-4,5,13-trimethyl-9-(oxazol-2-ylmethyl)-3-thia- 1,8,11,12- tetrazatricyclo[8.3.0.02,6]trideca-2(6),4,7,10,12-pentaen-7- yl]phenyl]prop-2-ynoxy]piperidine-1- carboxylate (74 mg, 50% yield). [0450] Step 5: Preparation of tert-butyl 4-[3-[4-[(9S)-4,5,13-trimethyl-9-(oxazol-2- ylmethyl)-3-thia-1,8,11,12-tetrazatricyclo[8.3.0.02,6]tridec a-2(6),4,7,10,12-pentaen-7- yl]phenyl]propoxy]piperidine-1-carboxylate. To a solution of tert-butyl 4-[3-[4-[(9S)-4,5,13- trimethyl-9-(oxazol-2-ylmethyl)-3-thia-1,8,11,12-tetrazatric yclo[8.3.0.02,6]trideca- 2(6),4,7,10,12-pentaen-7-yl]phenyl]prop-2-ynoxy]piperidine-1 -carboxylate (74 mg, 118 μmol, 1.0 equiv) in THF (3 mL) was added PtO2 (30 mg, 132 μmol, 1.1 equiv) under N2 atmosphere. The mixture was stirred under H 2 (15 Psi) at 25 °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: Waters Xbridge C18150*50mm* 10um; mobile phase: [water (NH4HCO3)-ACN]; B%: 52%-82%, 10min) to give compound tert-butyl 4-[3-[4-[(9S)-4,5,13-trimethyl-9-(oxazol-2- ylmethyl)-3-thia-1,8,11,12-tetrazatricyclo[8.3.0.02,6]tridec a-2(6),4,7,10,12-pentaen-7- yl]phenyl]propoxy]piperidine-1-carboxylate (30 mg, 40% yield). [0451] Step 6: Preparation of 2-[[(9S)-4,5,13-trimethyl-7-[4-[3-(4-piperidyloxy)propyl] phenyl]-3-thia-1,8,11,12-tetrazatricyclo[8.3.0.02,6]trideca- 2(6),4,7,10,12-pentaen-9- yl]methyl]oxazole. To a solution of tert-butyl 4-[3-[4-[(9S)-4,5,13-trimethyl-9-(oxazol-2- ylmethyl)-3-thia-1,8,11,12-tetrazatricyclo[8.3.0.02,6]tridec a-2(6),4,7,10,12-pentaen-7- yl]phenyl]propoxy]piperidine-1-carboxylate (30 mg, 47 μmol, 1.0 equiv) in DCM (2 mL) was added TFA (0.5 mL). The mixture was stirred at 25 °C for 0.5 h. The mixture was filtered and concentrated. The residue was used for next step directly to give 2-[[(9S)-4,5,13-trimethyl-7-[4- [3-(4-piperidyloxy)propyl]phenyl]-3-thia-1,8,11,12-tetrazatr icyclo [8.3.0.02,6]trideca- 2(6),4,7,10,12-pentaen-9-yl]methyl]oxazole (25 mg, 99% yield). [0452] Step 7: Preparation of N-[3-(3-chloro-4-cyano-phenoxy)-2,2,4,4-tetramethyl- cyclobutyl]-2-[4-[3-[4-[(9S)-4,5,13-trimethyl-9-(oxazol-2-yl methyl)-3-thia-1,8,11,12- tetrazatricyclo[8.3.0.02,6]trideca-2(6),4,7,10,12-pentaen-7- yl]phenyl]propoxy]-1-piperidyl] pyrimidine-5-carboxamide (I-131). To a solution of 2-[[(9S)-4,5,13-trimethyl-7-[4-[3-(4- piperidyloxy)propyl]phenyl]-3-thia-1,8,11,12-tetrazatricyclo [8.3.0.02,6]trideca-2(6),4,7,10,12- pentaen-9-yl]methyl]oxazole (25 mg, 47 μmol, 1.0 equiv) and 2-chloro-N-[3-(3-chloro-4-cyano- phenoxy)-2,2,4,4-tetramethyl-cyclobutyl]pyrimidine-5-carboxa mide (19 mg, 47 μmol, 1.0 equiv) in NMP (0.5 mL) was added K2CO3 (19 mg, 141 μmol, 3.0 equiv). The mixture was stirred at 50 °C for 2 h. The mixture was filtered and concentrated. The residue was purified by prep- HPLC (column: Unisil 3-100 C18 Ultra 150*50mm*3 μm; mobile phase: [water(FA)-ACN];B%: 70%-100%,7min) to give N-[3-(3-chloro-4-cyano-phenoxy)-2,2,4,4-tetramethyl-cyclobut yl]-2- [4-[3-[4-[(9S)-4,5,13-trimethyl-9-(oxazol-2-ylmethyl)-3-thia -1,8,11,12-tetrazatricyclo [8.3.0.02,6]trideca-2(6),4,7,10,12-pentaen-7-yl]phenyl]propo xy]-1-piperidyl]pyrimidine-5- carboxamide (18 mg, 41% yield). 1 H NMR (400 MHz, DMSO): δ 8.76 (s, 2H), 8.05 (s, 1H), 7.92 (d, J = 8.7 Hz, 1H), 7.74 (d, J = 9.3 Hz, 1H), 7.30 - 7.22 (m, 5H), 7.14 (s, 1H), 7.02 - 6.99 (m , 1H), 4.66 (t, J = 7.3 Hz, 1H), 4.30 - 4.20 (m, 3H), 4.05 (d, J = 9.2 Hz, 1H), 3.91 - 3.78 (m, 2H), 3.56 - 3.41 (m, 6H), 2.71 - 2.65 (m, 4H), 2.35 - 2.31 (m, 1H), 1.90 - 1.76 (m, 5H), 1.62 (s, 3H), 1.42 - 1.39 (m, 3H), 1.22 (s, 6H), 1.11 (s, 6H). LC-MS: MS (ES + ): RT = 3.275 min, m/z = 913.3 [M + H + ]; LCMS method: 25. EXAMPLE 23 – Synthesis of N-[3-(3-chloro-4-cyano-phenoxy)-2,2,4,4-tetramethyl- cyclobutyl]-2-[4-[1-[4-[(9S)-4,5,13-trimethyl-9-(oxazol-2-yl methyl)-3-thia-1,8,11,12- tetrazatricyclo[8.3.0.02,6]trideca-2(6),4,7,10,12-pentaen-7- yl]phenyl]azetidin-3-yl]oxy-1- piperidyl]pyrimidine-5-carboxamide (I-206)

[0453] Step 1: Preparation of tert-butyl 3-[[1-(2,2,2-trifluoroacetyl)-4-piperidyl]oxy] azetidine-1-carboxylate. To a stirred solution of tert-butyl 3-(4-piperidyloxy) azetidine-1- carboxylate (2.80 g, 10.9 mmol, 1.0 equiv) in DCM (100 mL) , DIEA (3.53 g, 27.3 mmol, 4.76 mL, 2.5 equiv) was added followed by the addition of TFAA (2.79 g, 13.3 mmol, 1.85 mL, 1.2 equiv) at 0°C and the reaction mixture was stirred at 25 °C for 2 h. The reaction mixture was diluted with DCM 50 mL and adjusted PH=7.0 with 10% NaHCO3 aq, then extracted withDCM (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 column chromatography (SiO2, Petroleum ether/Ethyl acetate=10/1 to 1/1). Compound tert-butyl 3-[[1-(2,2,2-trifluoroacetyl)-4-piperidyl]oxy]azetidine-1-ca rboxylate (2.00 g, 5.68 mmol, 52% yield) was obtained as a white solid. [0454] Step 2: Preparation of 1-[4-(azetidin-3-yloxy)-1-piperidyl]-2,2,2-trifluoro- ethanone. To a stirred solution of tert-butyl 3-[[1-(2,2,2-trifluoroacetyl)-4-piperidyl]oxy] azetidine-1-carboxylate (2.00 g, 5.68 mmol, 1.0 equiv) in DCM (10 mL) was added followed by the addition of TFA (15.4 g, 135 mmol, 10 mL, 23.8 equiv) and the reaction mixture was stirred at 25 °C for 2 h. The reaction mixture was concentrated under reduced pressure to give a residue. Compound 1-[4-(azetidin-3-yloxy)-1-piperidyl]-2,2,2-trifluoro-ethanon e (2.08 g, 5.68 mmol, 100% yield, TFA) as a Colorless gum was used into the next step without further purification. [0455] Step 3: Preparation of 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]acetic acid. 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 (1.0 g, 2.1 mmol, 1.0 equiv) in DCM (10 mL) was added TFA (7.7 g, 67 mmol, 5.0 mL, 30 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 2-[(9S)-7-(4- chlorophenyl)-4,5,13-trimethyl-3-thia-1,8,11,12-tetrazatricy clo[8.3.0.02,6]trideca-2(6),4,7,10,12- pentaen-9-yl]acetic acid (877 mg, 99% yield). [0456] Step 4: Preparation of 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]acetamide. To a solution of 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]acetic acid (877 mg, 2.19 mmol, 1.0 equiv) and NH4Cl (351 mg, 6.56 mmol, 3.0 equiv) in DMF (8 mL) was added HATU (915 mg, 2.41 mmol, 1.1 equiv) and DIEA (848 mg, 6.56 mmol, 3.0 equiv). The mixture was stirred at 25 °C for 1 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%: 24%- 54%,10min) to give 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]acetamide (740 mg, 84% yield). [0457] Step 5: Preparation of 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]methyl]oxazole. To a solution of 1,3-dioxol-2-one (103 mg, 1.20 mmol, 1.2 equiv) and 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]acetamide (400 mg, 1.00 mmol, 1.0 equiv) in PPA (4 mL). The mixture was stirred at 160 °C for 3 h. The mixture was quenched by addition H 2 O 50 mL at 0°C, and then diluted with EA 200 mL. The mixture was extracted with H 2 O (15 mL * 2). The combined organic layers were washed with H 2 O (10 mL * 3), dried over [Na2SO4], filtered and concentrated. The residue was purified by column chromatography (SiO2, Petroleum ether/Ethyl acetate=2/1 to 0/1) to give compound 2-[[(9S)-7-(4-chlorophenyl)-4,5,13-trimethyl-3-thia-1,8,11,1 2-tetrazatricyclo [8.3.0.02,6]trideca-2(6),4,7,10,12-pentaen-9-yl]methyl]oxazo le (260 mg, 61% yield). [0458] Step 6: Preparation of 2-[[(9S)-4,5,13-trimethyl-7-[4-[3-(4-piperidyloxy)azetidin- 1-yl]phenyl]-3-thia-1,8,11,12-tetrazatricyclo[8.3.0.02,6]tri deca-2(6),4,7,10,12-pentaen-9- yl]methyl]oxazole. A mixture of 2-[[(9S)-7-(4-chlorophenyl)-4,5,13-trimethyl-3-thia-1,8,11,1 2- tetrazatricyclo[8.3.0.02,6]trideca-2(6),4,7,10,12-pentaen-9- yl]methyl]oxazole (150 mg, 353 μmol, 1.0 equiv), 1-[4-(azetidin-3-yloxy)-1-piperidyl]-2,2,2-trifluoro-ethanon e (142 mg, 389 μmol, 1.1 equiv,), SPhos Pd G3 (28 mg, 36 μmol, 0.1 equiv), Cs2CO3 (347 mg, 1.1 mmol, 3.0 equiv) in dioxane (3 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 mixture was filtered and concentrated to give the residue. The residue was purified by the prep-TLC (Dichloromethane : Methanol = 10:1) to give the 2-[[(9S)-4,5,13-trimethyl-7-[4-[3-(4-piperidyloxy)azetidin-1 -yl]phenyl]-3-thia- 1,8,11,12-tetrazatricyclo[8.3.0.02,6]trideca-2(6),4,7,10,12- pentaen-9-yl]methyl]oxazole (100 mg, 184 μmol, 52% yield) as a yellow oil. [0459] Step 7: Preparation of N-[3-(3-chloro-4-cyano-phenoxy)-2,2,4,4-tetramethyl- cyclobutyl]-2-[4-[1-[4-[(9S)-4,5,13-trimethyl-9-(oxazol-2-yl methyl)-3-thia-1,8,11,12- tetrazatricyclo[8.3.0.02,6]trideca-2(6),4,7,10,12-pentaen-7- yl]phenyl]azetidin-3-yl]oxy-1- piperidyl]pyrimidine-5-carboxamide (I-206). A mixture of 2-[[(9S)-4,5,13-trimethyl-7-[4-[3- (4-piperidyloxy)azetidin-1-yl]phenyl]-3-thia-1,8,11,12-tetra zatricyclo[8.3.0.02,6]trideca- 2(6),4,7,10,12-pentaen-9-yl]methyl]oxazole (94 mg, 173 μmol, 1.0 equiv), 2-chloro-N-[3-(3- chloro-4-cyano-phenoxy)-2,2,4,4-tetramethyl-cyclobutyl]pyrim idine-5-carboxamide (87 mg, 207 μmol, 1.2 equiv), K 2 CO 3 (72 mg, 519 μmol, 3.0 equiv) in DMF (2.0 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 residue was filtered and concentrated to get the residue .The crude product was purified by reversed-phase HPLC(column: Phenomenex luna C18150*25mm* 10um;mobile phase: [water(FA)-ACN];B%: 53%-83%,15min) and reversed-phase HPLC(column: Phenomenex luna C18150*25mm* 10um;mobile phase: [water(FA)-ACN];B%: 54%- 84%,8min) to give the N-[3-(3-chloro-4-cyano-phenoxy)-2,2,4,4-tetramethyl-cyclobut yl]-2-[4- [1-[4-[(9S)-4,5,13-trimethyl-9-(oxazol-2-ylmethyl)-3-thia-1, 8,11,12-tetrazatricyclo[8.3.0.02,6] trideca-2(6),4,7,10,12-pentaen-7-yl]phenyl]azetidin-3-yl]oxy -1-piperidyl]pyrimidine-5- carboxamide (37.92 mg, 38.06 umol, 22% yield, 93% purity). 1 H NMR: (400 MHz, MeOD) δ = 8.73 (s, 2H), 7.93 - 7.84 (m, 1H), 7.72 (d, J = 8.8 Hz, 1H), 7.22 (d, J = 8.0 Hz, 2H), 7.12 (d, J = 2.4 Hz, 2H), 7.01 - 6.93 (m, 1H), 6.42 (d, J = 8.8 Hz, 2H), 4.70 - 4.64 (m, 2H), 4.44 - 4.34 (m, 2H), 4.31 - 4.24 (m, 1H), 4.24 - 4.17 (m, 2H), 4.15 - 4.10 (m, 1H), 3.90 - 3.86 (m, 1H), 3.98 - 3.85 (m, 1H), 3.79 - 3.69 (m, 3H), 3.60 - 3.50 (m, 2H), 2.69 (s, 3H), 2.45 (s, 3H), 1.99 - 1.90 (m, 2H), 1.73 (s, 3H), 1.62 - 1.52 (m, 2H), 1.28 (s, 6H), 1.23 - 1.20 (m, 6H). LC-MS: MS (ES + ): RT = 2.687min, m/z = 926.6 [M +H + ] EXAMPLE 24 – Synthesis of N-[3-(3-chloro-4-cyano-phenoxy)-2,2,4,4-tetramethyl- cyclobutyl]-2-[4-[3-[4-[(9S)-4,5,13-trimethyl-9-(2-pyridylme thyl)-3-thia-1,8,11,12- tetrazatricyclo[8.3.0.02,6]trideca-2(6),4,7,10,12-pentaen-7- yl]phenyl]propoxy]-1- piperidyl]pyrimidine-5-carboxamide (I-212)

[0 460] Step 1: Preparation of tert-butyl 4-[3-[4-[(9S)-9-(2-tert-butoxy-2-oxo-ethyl)- 4,5,13-trimethyl-3-thia-1,8,11,12-tetrazatricyclo[8.3.0.02,6 ]trideca-2(6),4,7,10,12-pentaen-7- yl]phenyl]prop-2-ynoxy]piperidine-1-carboxylate. A mixture of tert-butyl 2-[(9S)-7-(4- chlorophenyl)-4,5,13-trimethyl-3-thia-1,8,11,12-tetrazatricy clo[8.3.0.02,6]trideca-2(6),4,7,10,12- pentaen-9-yl]acetate (800 mg, 1.75 mmol, 1.0 equiv), tert-butyl 4-prop-2-ynoxypiperidine-1- carboxylate (649 mg, 2.71 mmol, 1.6 equiv), Ad2nBuP Pd G3(cataCXium® A Pd G3) (127 mg, 175 umol, 0.1 equiv) and Cs2CO3 (1.48 g, 4.55 mmol, 2.6 equiv) in ACN (4 mL) and DMF (12 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 90 °C for 1 h under N 2 atmosphere. The mixture was poured into water (80 mL). The aqueous phase was extracted with ethyl acetate (80 mL x 2). The combined organic phase was washed with brine (70 mL x 2), dried with anhydrous Na 2 SO 4 , filtered and concentrated in vacuum. The residue was purified by silica gel chromatography (0% - 100% ethyl acetate in petroleum ether) and further purified by prep-HPLC (column: Phenomenex luna C18150*40mm* 15um; mobile phase: [water (FA) - ACN]; B%: 62%-92%, 10min). Compound tert-butyl 4-[3-[4-[(9S)-9-(2- tert-butoxy-2-oxo-ethyl)-4,5,13-trimethyl-3-thia-1,8,11,12-t etrazatricyclo[8.3.0.02,6]trideca- 2(6),4,7,10,12-pentaen-7-yl]phenyl]prop-2-ynoxy]piperidine-1 -carboxylate (270 mg, 409 μmol, 23% yield) was obtained as a light yellow solid. [0461] Step 2: Preparation of tert-butyl 4-[3-[4-[(9S)-9-(2-tert-butoxy-2-oxo-ethyl)- 4,5,13-trimethyl-3-thia-1,8,11,12-tetrazatricyclo[8.3.0.02,6 ]trideca-2(6),4,7,10,12-pentaen-7- yl]phenyl]propoxy]piperidine-1-carboxylate. To a solution of tert-butyl 4-[3-[4-[(9S)-9-(2- tert-butoxy-2-oxo-ethyl)-4,5,13-trimethyl-3-thia-1,8,11,12-t etrazatricyclo[8.3.0.02,6]trideca- 2(6),4,7,10,12-pentaen-7-yl]phenyl]prop-2-ynoxy]piperidine-1 -carboxylate (260 mg, 394 μmol, 1.0 equiv) in CF 3 CH 2 OH (10 mL) was added Pd/C (220 mg, 10% purity) under N2 atmosphere. The suspension was degassed and purged with H 2 for 3 times. The mixture was stirred under H 2 (15 Psi) at 25 °C for 2 h. The mixture was filtered and concentrated under reduced pressure. Compound tert-butyl 4-[3-[4-[(9S)-9-(2-tert-butoxy-2-oxo-ethyl)-4,5,13-trimethyl -3-thia- 1,8,11,12-tetrazatricyclo[8.3.0.02,6]trideca-2(6),4,7,10,12- pentaen-7-yl]phenyl]propoxy] piperidine-1-carboxylate (260 mg, 392 μmol, 99% yield) was obtained as a yellow gum. [0462] Step 3: Preparation of 2-[(9S)-7-[4-[3-[(1-tert-butoxycarbonyl-4- piperidyl)oxy]propyl]phenyl]-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]acetic acid. To a solution of tert-butyl 4-[3-[4- [(9S)-9-(2-tert-butoxy-2-oxo-ethyl)-4,5,13-trimethyl-3-thia- 1,8,11,12-tetrazatricyclo[8.3.0.0 2,6 ] trideca-2(6),4,7,10,12-pentaen-7-yl]phenyl]propoxy]piperidin e-1-carboxylate (400 mg, 602 μmol, 1.0 equiv) in THF (5 mL) MeOH (5 mL) H 2 O (5 mL) was added LiOH.H 2 O (252 mg, 6.03 mmol, 10.0 equiv). The mixture was stirred at 25 °C for 12 h. The reaction mixture was quenched by addition water (50 mL), and extracted with ethyl acetate (30 mL x 3). The water- course by HCl (2 mol) adjust PH = 3, and extracted with ethyl acetate 30 mL (30 mL x 3). The combined organic dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. Compound 2-[(9S)-7-[4-[3-[(1-tert-butoxycarbonyl-4-piperidyl)oxy] propyl]phenyl]- 4,5,13-trimethyl-3-thia-1,8,11,12-tetrazatricyclo[8.3.0.0 2,6 ]trideca-2(6),4,7,10,12-pentaen-9- yl]acetic acid (400 mg, crude) was obtained as a white solid. [0463] Step 4: Preparation of tert-butyl 4-[3-[4-[(9S)-9-[2-(1,3-dioxoisoindolin-2-yl)oxy- 2-oxo-ethyl]-4,5,13-trimethyl-3-thia-1,8,11,12-tetrazatricyc lo[8.3.0.02,6]trideca-2(6),4,7, 10,12-pentaen-7-yl]phenyl]propoxy]piperidine-1-carboxylate. To a solution of 2-[(9S)-7-[4- [3-[(1-tert-butoxycarbonyl-4-piperidyl)oxy]propyl]phenyl]-4, 5,13-trimethyl-3-thia-1,8,11,12- tetrazatricyclo[8.3.0.0 2,6 ]trideca-2(6),4,7,10,12-pentaen-9-yl]acetic acid (400 mg, 658 μmol, 1 equiv) in DCM (5 mL) was added DMAP (8.04 mg, 65.8 μmol, 0.1 equiv), 2-hydroxy- isoindoline-1,3-dione (112 mg, 691 μmol, 1.0 equiv), DIC (91 mg, 0.72 mmol, 112 μL, 1.1 equiv). The mixture was stirred at 25 °C for 12 h. To the reaction mixture was added water (20 mL) and the mixture was extracted with dichloromethane (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-butyl 4-[3-[4-[(9S)-9-[2-(1,3-dioxoisoindolin-2-yl)oxy-2-oxo-ethyl ]- 4,5,13-trimethyl-3-thia-1,8,11,12-tetrazatricyclo[8.3.0.0 2,6 ]trideca-2(6),4,7,10,12-pentaen-7- yl]phenyl]propoxy]piperidine-1-carboxylate (350 mg, 70 % yield) was obtained as a white solid. [0464] Step 5: Preparation of tert-butyl 4-[3-[4-[(9S)-4,5,13-trimethyl-9-(2- pyridylmethyl)-3-thia-1,8,11,12-tetrazatricyclo[8.3.0.02,6]t rideca-2(6),4,7,10,12-pentaen-7- yl]phenyl]propoxy]piperidine-1-carboxylate. A mixture of tert-butyl 4-[3-[4-[(9S)-9-[2-(1,3- dioxoisoindolin-2-yl)oxy-2-oxo-ethyl]-4,5,13-trimethyl-3-thi a-1,8,11,12-tetrazatricyclo [8.3.0.02,6]trideca-2(6),4,7,10,12-pentaen-7-yl]phenyl]propo xy]piperidine-1-carboxylate (200 mg, 266 μmol, 1.0 equiv), 2-iodopyridine (54 mg, 0.27 mmol, 28 μL, 1.0 equiv), Zn (104 mg, 765 μmol, 2.9 equiv), 4-tert-butyl-2-(4-tert-butyl-2-pyridyl)pyridine; dichloronickel (21 mg, 53 μmol, 0.2 equiv) in DMF (2 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 45 °C for 12 h under N 2 atmosphere. To the reaction mixture was added water (20 mL) and the mixture was extracted with dichloromethane (20 mL x 3). The combined organic phase was washed with brine (20 mL), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. Compound tert-butyl 4-[3-[4-[(9S)-4,5,13-trimethyl-9-(2-pyridylmethyl)- 3-thia-1,8,11,12-tetrazatricyclo[8.3.0.02,6] trideca-2(6),4,7,10,12-pentaen-7-yl]phenyl]propoxy] piperidine-1-carboxylate (100 mg, 156 μmol, 59% yield) was obtained as a white solid. [0465] Step 6: Preparation of (9S)-4,5,13-trimethyl-7-[4-[3-(4-piperidyloxy) propyl]phenyl]-9-(2-pyridylmethyl)-3-thia-1,8,11,12-tetrazat ricyclo [8.3.0.02,6]trideca- 2(6),4,7,10,12-pentaene. To a solution of tert-butyl 4-[3-[4-[(9S)-4,5,13-trimethyl-9-(2- pyridylmethyl)-3-thia-1,8,11,12-tetrazatricyclo[8.3.0.02,6]t rideca-2(6),4,7,10,12-pentaen-7- yl]phenyl]propoxy]piperidine-1-carboxylate (100 mg, 156 μmol, 1.0 equiv) in DCM (1 mL) was added TFA (768 mg, 6.73 mmol, 0.5 mL, 43 equiv). The mixture was stirred at 25 °C for 0.5 h. The reaction mixture was concentrated in vacuo to give the crude product. Compound (9S)- 4,5,13-trimethyl-7-[4-[3-(4-piperidyloxy)propyl]phenyl]-9-(2 -pyridyl methyl)-3-thia-1,8,11,12- tetrazatricyclo[8.3.0.02,6]trideca-2(6),4,7,10,12-pentaene (70 mg, crude) was obtained as a brown oil. [0466] Step 7: Preparation of N-[3-(3-chloro-4-cyano-phenoxy)-2,2,4,4-tetramethyl- cyclobutyl]-2-[4-[3-[4-[(9S)-4,5,13-trimethyl-9-(2-pyridylme thyl)-3-thia-1,8,11,12- tetrazatricyclo [8.3.0.02,6]trideca-2(6),4,7,10,12-pentaen-7-yl]phenyl]propo xy]-1-piperidyl] pyrimidine-5-carboxamide (I-212). To a solution of (9S)-4,5,13-trimethyl-7-[4-[3-(4- piperidyloxy) propyl]phenyl]-9-(2-pyridylmethyl)-3-thia-1,8,11,12-tetrazat ricyclo[8.3.0.02,6] trideca-2(6),4,7,10,12-pentaene (70 mg, 0.13 mmol, 1.0 equiv) in NMP (2 mL) was added DIEA (84 mg, 0.65 mmol, 0.11 mL, 5.0 equiv) and 2-chloro-N-[3-(3-chloro-4-cyano-phenoxy)-2,2,4,4- tetramethyl-cyclobutyl]pyrimidine-5-carboxamide (54 mg, 0.13 mmol, 1.0 equiv). The mixture was stirred at 25 °C for 2 h. To the reaction mixture was added water (20 mL) and the mixture was extracted with dichloromethane (20 mL x 3). The combined organic phase was washed with brine (20 mL), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. The residue was purified by prep-SFC (column: DAICEL CHIRALPAK AD(250mm*30mm,10um);mobile phase: [CO 2 -ACN/i-PrOH(0.1% NH3H 2 O)];B%:60%, isocratic elution mode). Compound N-[3- (3-chloro-4-cyano-phenoxy)-2,2,4,4-tetramethyl-cyclobutyl]-2 -[4-[3-[4-[(9S)-4,5,13-trimethyl-9- (2-pyridylmethyl)-3-thia-1,8,11,12-tetrazatricyclo[8.3.0.02, 6]trideca-2(6),4,7,10,12-pentaen-7- yl]phenyl]propoxy]-1-piperidyl]pyrimidine-5-carboxamide (47 mg, 39% yield, 98% purity) was obtained as a white solid. 1 H NMR (400 MHz, METHANOL-d4) δ 8.73 (s, 2H), 8.44 (d, J = 4.8 Hz, 1H), 7.84 (s, 1H), 7.72 (d, J = 8.8 Hz, 1H), 7.64 (d, J = 7.6 Hz, 1H), 7.34-7.20 (m, 5H), 7.13 (d, J = 2.4 Hz, 1H), 6.98 (s, 1 H), 4.67 (s, 1H), 4.27 (s, 3H), 4.13 (s, 1H), 3.98-3.92 (m, 2H), 3.60-3.53 (m, 2H), 3.50 (t, J = 6.4 Hz, 2H), 2.75 (t, J = 7.2 Hz, 2H), 2.70 (s, 3H), 2.43 (s, 3H), 1.94-1.85 (m, 4H), 1.61 (s, 3H), 1.53 (s, 2H), 1.37-1.31 (m, 2H), 1.28 (s, 6H), 1.21 (s, 6H). LC- MS: MS (ES + ): RT = 2.501 min, m/z = 923.6 [M + H + ]; LCMS method: 25 EXAMPLE 25 – Synthesis of N-[3-(3-chloro-4-cyano-phenoxy)-2,2,4,4-tetramethyl- cyclobutyl]-2-[2-[4-[(9S)-4,5,13-trimethyl-9-[(2-methyltetra zol-5-yl)methyl]-3-thia- 1,8,11,12-tetrazatricyclo[8.3.0.02,6]trideca-2(6),4,7,10,12- pentaen-7-yl]phenoxy]-7- azaspiro[3.5]nonan-7-yl]pyrimidine-5-carboxamide (I-218)

[0467] Step 1: Preparation of 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]acetic acid. A mixture 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 (4.9 g, 10.7 mmol, 1.0 equiv) in TFA (30 mL) and DCM (100 mL) was degassed and purged with N 2 for 3 times, and then the mixture was stirred at 25 °C for 12 h under N2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give the 2-[(9S)-7-(4-chlorophenyl)-4,5,13- trimethyl-3-thia-1,8,11,12-tetrazatricyclo[8.3.0.02,6]tridec a-2(6),4,7,10,12-pentaen-9-yl]acetic acid (4.3 g, 10.7 mmol, 100% yield) as a brown oil.

[0468] Step 2: Preparation of 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]acetamide. A mixture of 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]acetic acid (4.3 g, 10.7 mmol, 1.0 equiv), TEA (10.85 g, 107.3 mmol, 15 mL, 10.0 equiv), NH4Cl (1.72 g, 32.2 mmol, 3.0 equiv) in DMF (50 mL) and the mixture was stirred at 25°C for 10 min.Then HATU (5.30 g, 13.9 mmol, 1.3 equiv) and then the mixture was stirred at 25 °C for 2 h under N 2 atmosphere. The reaction mixture was diluted with Ethyl acetate (100 mL) and washed with NaCl (210 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (column: Kromasil Eternity XT 250*80mm*10um;mobile phase: [water( NH 4 HCO 3 )-ACN];B%: 28%-58%,20min) to give the 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]acetamide (2.9 g, 3.4 mmol, 32% yield) as a yellow solid. [0469] Step 3: Preparation of 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]acetonitrile. To a mixture of (COCl) 2 (1.90 g, 15.0 mmol, 1.3 mL, 3.0 equiv), DCM (15 mL) stirred at -78 °C under N2 protection. The DMSO (1.56 g, 20.0 mmol, 1.6 mL, 4.0 equiv) was dropwised to the mixture and stirred for 30min. Then the 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]acetamide (2 g, 5.0 mmol, 1.0 equiv) was dissolved in the DCM (5 mL) and was added to the -78°C mixture and stirred for further 30 min. Finally, the TEA (4.05 g, 40.0 mmol, 5.6 mL, 8.0 equiv) was added to the mixture at -78°C under N 2 protection and allowed slowlly warmed up to 25 °C. The reaction mixture was diluted with DCM (100 mL) and washed by water (70 mL x 3), then the organic phase was dried over anhydrous Na2SO4, evaporated in vacuum. The residue was purified by column chromatography (SiO 2 , Petroleum ether/Ethyl acetate=0/1) to give the 2-[(9S)-7-(4- chlorophenyl)-4,5,13-trimethyl-3-thia-1,8,11,12-tetrazatricy clo[8.3.0.02,6]trideca-2(6),4,7,10,12- pentaen-9-yl]acetonitrile (500 mg, 1.3 mmol, 25% yield) as a white solid. [0470] Step 4: Preparation of (9S)-7-(4-chlorophenyl)-4,5,13-trimethyl-9-(2H-tetrazol-5- ylmethyl)-3-thia-1,8,11,12-tetrazatricyclo[8.3.0.02,6]tridec a-2(6),4,7,10,12-pentaene. A mixture of 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]acetonitrile (300 mg, 786 μmol, 1.0 equiv), TMSN 3 (272 mg, 2.4 mmol, 310 μL, 3.0 equiv), TBAF (248 mg, 786 μmol, 1.0 equiv) in Tol. (3 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 100 °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 prep-HPLC (column: Phenomenex Synergi Polar-RP 100*25mm*4um;mobile phase: [water(TFA)-ACN];B%: 46%-76%,9min) to give the (9S)-7-(4-chlorophenyl)-4,5,13-trimethyl-9-(2H-tetrazol-5-yl methyl)-3-thia-1,8,11,12- tetrazatricyclo[8.3.0.02,6]trideca-2(6),4,7,10,12-pentaene (260 mg, 587 μmol, 75% yield) as a yellow solid. [0471] Step 5: Preparation of (9S)-7-(4-chlorophenyl)-4,5,13-trimethyl-9-[(2- methyltetrazol-5-yl)methyl]-3-thia-1,8,11,12-tetrazatricyclo [8.3.0.02,6]trideca- 2(6),4,7,10,12-pentaene. To a solution of (9S)-7-(4-chlorophenyl)-4,5,13-trimethyl-9-(2H- tetrazol-5-ylmethyl)-3-thia-1,8,11,12-tetrazatricyclo[8.3.0. 02,6]trideca-2(6),4,7,10,12-pentaene (460 mg, 1.1 mmol, 1.0 equiv) in DMF (5 mL) was added K2CO3 (300 mg, 2.2 mmol, 2.0 equiv) and CH 3 I (154 mg, 1.1 mmol, 67 μL, 1.0 equiv). The mixture was stirred at 40 °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: Waters Xbridge 150*25mm* 5um;mobile phase: [water( NH4HCO3)-ACN];B%: 39%-69%,10min) to give the (9S)-7-(4-chlorophenyl)-4,5,13- trimethyl-9-[(2-methyltetrazol-5-yl)methyl]-3-thia-1,8,11,12 -tetrazatricyclo[8.3.0.02,6]trideca- 2(6),4,7,10,12-pentaene (100 mg, 226 μmol, 21% yield) as a white solid. [0472] Step 6: Preparation of 4-[(9S)-4,5,13-trimethyl-9-[(2-methyltetrazol-5-yl)methyl]- 3-thia-1,8,11,12-tetrazatricyclo[8.3.0.02,6]trideca-2(6),4,7 ,10,12-pentaen-7-yl]phenol. A mixture of (9S)-7-(4-chlorophenyl)-4,5,13-trimethyl-9-[(2-methyltetrazo l-5-yl)methyl]-3-thia- 1,8,11,12-tetrazatricyclo[8.3.0.02,6]trideca-2(6),4,7,10,12- pentaene (100 mg, 228 μmol, 1.0 equiv), BrettPhos Pd G3 (21 mg, 23 μmol, 0.1 equiv), KOH (64 mg, 1.1 mmol, 5.0 equiv) in dioxane (1 mL) and H 2 O (0.5 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 90 °C for 2 h under N2 atmosphere. The reaction mixture 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]; gradient: 23%-50% B over 9 min).Compound 4-[(9S)-4,5,13-trimethyl-9-[(2-methyltetrazol-5-yl)methyl]- 3-thia-1,8,11,12-tetrazatricyclo[8.3.0.02,6]trideca-2(6),4,7 ,10,12-pentaen-7-yl]phenol (80 mg, 188 μmol, 83% yield) as a white solid. [0473] Step 7: Preparation of tert-butyl 2-[4-[(9S)-4,5,13-trimethyl-9-[(2-methyltetrazol- 5-yl)methyl]-3-thia-1,8,11,12-tetrazatricyclo[8.3.0.02,6]tri deca-2(6),4,7,10,12-pentaen-7- yl]phenoxy]-7-azaspiro[3.5]nonane-7-carboxylate. A mixture of 4-[(9S)-4,5,13-trimethyl-9- [(2-methyltetrazol-5-yl)methyl]-3-thia-1,8,11,12-tetrazatric yclo[8.3.0.02,6]trideca-2(6),4,7,10,12 -pentaen-7-yl]phenol (70 mg, 166 μmol, 1.0 equiv), tert-butyl 2-hydroxy-7-azaspiro[3.5]nonane- 7-carboxylate (80 mg, 333 μmol, 2.0 equiv), PPh3 (92 mg, 350 μmol, 350 μL, 2.1 equiv), DIAD (67 mg, 333 μmol, 65 μL, 2.0 equiv) in THF (1 mL) was degassed and purged with N 2 for 3 times, and then the mixture was stirred at 50 °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 prep-HPLC (column: Phenomenex luna C18150*40mm* 15um;mobile phase: [water(TFA)-ACN];gradient:55%-85% B over 10 min) to give the tert-butyl 2-[4-[(9S)-4,5,13- trimethyl-9-[(2-methyltetrazol-5-yl)methyl]-3-thia-1,8,11,12 -tetrazatricyclo[8.3.0.02,6]trideca- 2(6),4,7,10,12-pentaen-7-yl]phenoxy]-7-azaspiro[3.5]nonane-7 -carboxylate (70 mg, 105 μmol, 63% yield) as a yellow solid. [0474] Step 8: Preparation of (9S)-7-[4-(7-azaspiro[3.5]nonan-2-yloxy)phenyl]-4,5,13- trimethyl-9-[(2-methyltetrazol-5-yl)methyl]-3-thia-1,8,11,12 -tetrazatricyclo[8.3.0.02,6] trideca-2(6),4,7,10,12-pentaene. A mixture of tert-butyl 2-[4-[(9S)-4,5,13-trimethyl-9-[(2- methyltetrazol-5-yl)methyl]-3-thia-1,8,11,12-tetrazatricyclo [8.3.0.02,6] trideca-2(6),4,7,10,12- pentaen-7-yl]phenoxy]-7-azaspiro[3.5]nonane-7-carboxylate (70 mg, 109 μmol, 1.0 equiv) in TFA (1 mL) and DCM (3 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 25 °C for 2 h under N2 atmosphere. The reaction mixture was quenched by addition NaHCO 3 (20 mL), and then diluted with DCM (40 mL) and extracted with NaCl (30 mL x 2), dried over Na2SO4, filtered and concentrated under reduced pressure to give the (9S)-7-[4- (7-azaspiro[3.5]nonan-2-yloxy)phenyl]-4,5,13-trimethyl-9-[(2 -methyltetrazol-5-yl)methyl]-3- thia-1,8,11,12-tetrazatricyclo[8.3.0.02,6]trideca-2(6),4,7,1 0,12-pentaene (59 mg, 109 μmol, 100% yield) as a white solid. [0475] Step 9: Preparation of N-[3-(3-chloro-4-cyano-phenoxy)-2,2,4,4-tetramethyl- cyclobutyl]-2-[2-[4-[(9S)-4,5,13-trimethyl-9-[(2-methyltetra zol-5-yl)methyl]-3-thia- 1,8,11,12-tetrazatricyclo[8.3.0.02,6]trideca-2(6),4,7,10,12- pentaen-7-yl]phenoxy]-7- azaspiro[3.5]nonan-7-yl]pyrimidine-5-carboxamide (I-218). A mixture of (9S)-7-[4-(7- azaspiro[3.5]nonan-2-yloxy)phenyl]-4,5,13-trimethyl-9-[(2-me thyltetrazol-5-yl)methyl]-3-thia- 1,8,11,12-tetrazatricyclo[8.3.0.02,6]trideca-2(6),4,7,10,12- pentaene (59 mg, 109 μmol, 1.0 equiv), 2-chloro-N-[3-(3-chloro-4-cyano-phenoxy)-2,2,4,4-tetramethyl -cyclobutyl]pyrimidine-5- carboxamide (50 mg, 119 μmol, 1.1 equiv), DIEA (28 mg, 217 μmol, 38 μL, 2.0 equiv) in NMP (2 mL) was degassed and purged with N 2 for 3 times, and then the mixture was stirred at 25 °C for 2 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: DAICEL CHIRALPAK AD(250mm*30mm,10um) ;mobile phase: [CO2-i-PrOH/ACN ];B%:60%, isocratic elution mode ). The residue was separated by SFC (Rt = 0.589 min, 29 mg) (detailed condition) to give the N-[3-(3-chloro-4-cyano-phenoxy)-2,2,4,4-tetramethyl-cyclobut yl]-2-[2-[4- [(9S)-4,5,13-trimethyl-9-[(2-methyltetrazol-5-yl)methyl]-3-t hia-1,8,11,12-tetrazatricyclo [8.3.0.02,6]trideca-2(6),4,7,10,12-pentaen-7-yl]phenoxy]-7-a zaspiro[3.5]nonan-7-yl]pyrimidine- 5-carboxamide (29 mg, 31 μmol, 29% yield) was obtained as a white solid. 1 H NMR: (400 MHz, MeOD) δ = 8.72 (s, 2H), 7.72 (d, J = 8.4 Hz, 1H), 7.29 (d, J = 7.6 Hz, 2H), 7.13 (s, 1H), 7.02 - 6.94 (m, 1H), 6.85 (d, J = 8.8 Hz, 2H), 4.58 (s, 3H), 4.26 (s, 3H), 4.13 (s, 1H), 4.05 (d, J = 5.6 Hz, 2H), 3.95 - 3.81 (m, 4H), 2.71 (s, 3H), 2.58 - 2.49 (m, 2H), 2.45 (s, 3H), 2.02 - 1.92 (m, 2H), 1.75 - 1.64 (m, 7H), 1.28 (s, 6H), 1.21 (s, 6H). LC-MS: MS (ES + ): RT = 2.795 min, m/z = 926.4 [M +H + ] EXAMPLE 26 – Synthesis of N-[3-(3-chloro-4-cyano-phenoxy)-2,2,4,4-tetramethyl- cyclobutyl]-2-[2-[4-[(4S)-1-methyl-4-(oxazol-2-ylmethyl)-4H- isoxazolo[5,4-d][2]benzazepin- 6-yl]phenoxy]-5-oxa-8-azaspiro[3.5]nonan-8-yl]pyrimidine-5-c arboxamide (I-221)

[0476] Step 1: Preparation of (2-bromophenyl)(4-chlorophenyl)methanol. To a solution of 1-bromo-4-chlorobenzene (20 g, 104.46 mmol, 20.0 mL, 1.0 equiv) in THF (200 mL) was added n-BuLi (2.5 M, 50.14 mL, 1.2 equiv) at -78 o C. The mixture was stirred at -78 o C for 1 h. The 2-bromobenzaldehyde (23.19 g, 125.36 mmol, 14.50 mL, 1.2 equiv) was added. The mixture was stirred at -78 o C for 1 h .The mixture was stirred at 25 o C for 10 h. The residue was diluted with NH 4 Cl (100 mL) and extracted with EA (300 mL) (3 x 100 mL). The combined organic layers were 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). (2- bromophenyl)(4-chlorophenyl)methanol (32 g, 107.54 mmol, 51% yield) was obtained as a white solid [0477] Step 2: Preparation of (2-bromophenyl)(4-chlorophenyl)methanone. To a solution of (2-bromophenyl)(4-chlorophenyl)methanol (15.0 g, 50.41 mmol, 1.0 equiv) in DCM (200 mL) was added DMP (64.14 g, 151.22 mmol, 46.82 mL, 3.0 equiv) .The mixture was stirred at 25 o C for 12 h. The residue was diluted with Na 2 SO 3 (100 mL) and extracted with EA (3 x 100 mL). The combined organic layers were filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO 2 , Petroleum ether/Ethyl acetate = 90/1 to 50/1). (2-bromophenyl)(4-chlorophenyl)methanone (28.0 g, 94.74 mmol, 93% yield) was obtained as a colourless oil. [0478] Step 3: Preparation of (4-chlorophenyl)(2-(4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2-yl)phenyl)methanone. To a solution of (2-bromophenyl)(4-chlorophenyl) methanone (5.0 g, 16.92 mmol, 1.0 equiv), 4,4,4’,4’,5,5,5’,5’-octamethyl-2,2’-bi(1,3,2- dioxaborolane) (8.59 g, 33.83 mmol, 2.0 equiv) in dioxane (40 mL) was added potassium;acetate (4.98 g, 50.75 mmol, 3.0 equiv) and Pd(dppf)Cl 2 (1.24 g, 1.69 mmol, 0.1 equiv) .The mixture was stirred at 110 o 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 = 90/1 to 80/1). (4-chlorophenyl)(2-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan- 2-yl)phenyl)methanone (2.0 g, 5.84 mmol, 34% yield) was obtained as a white solid. [0479] Step 4: Preparation of (4-iodo-3-methylisoxazol-5-yl)methanol. To a solution of (4- iodo-3-methylisoxazol-5-yl)methyl benzoate (24.3 g, 70.82 mmol, 1.0 equiv) in THF (260 mL) and MeOH (130 mL) was added NaOH (8.50 g, 212.46 mmol, 3.0 equiv) in H 2 O (140 mL). The mixture was stirred at 25 °C for 4 h. The reaction mixture was then diluted with H 2 O 100 mL, and pH was adjusted to 5~6 by HCl (1M), then extracted with ethyl acetate (500 mL). The combined organic layers were washed with H 2 O (200 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 = 10/1 to 5/1) to give a (4-iodo-3- methylisoxazol-5-yl)methanol (50 g, 209.20 mmol, 98% yield) as a white solid. [0480] Step 5: Preparation of 4-iodo-3-methylisoxazole-5-carbaldehyde. To a solution of (4-iodo-3-methylisoxazol-5-yl)methanol (25 g, 104.6 mmol, 1.0 equiv) in DCM (300 mL) was added DMP (66.55 g, 156.90 mmol, 48.61 mL, 1.5 equiv). The mixture was stirred at 25 °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 = 1/0 to 2/1).4-iodo-3-methylisoxazole-5-carbaldehyde (24.0 g, 101.27 mmol, 96% yield) was obtained as a yellow oil. [0481] Step 6: Preparation of (R,Z)-N-((4-iodo-3-methylisoxazol-5-yl)methylene)-2- methylpropane-2-sulfinamide. To a solution of 4-iodo-3-methylisoxazole-5-carbaldehyde (12.0 g, 50.63 mmol, 1.0 equiv) and (R)-2-methylpropane-2-sulfinamide (9.21 g, 75.95 mmol, 1.5 equiv) in THF (130 mL) was added Ti(OEt) 4 (28.88 g, 126.59 mmol, 26.25 mL, 2.5 equiv). The mixture was stirred at 30 o C for 2 h. The reaction mixture was washed with NH4Cl (200 mL), and then washed with H 2 O (100 mL) and EA (400 mL), 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 10/1). (R,Z)-N-((4-iodo-3-methylisoxazol-5-yl)methylene)-2- methylpropane-2-sulfinamide (31.0 g, 91.13 mmol, 89% yield) was obtained as a yellow solid. [0482] Step 7: Preparation of tert-butyl (3S)-3-((tert-butylsulfinyl)amino)-3-(4-iodo-3- methylisoxazol-5-yl)propanoate. To a solution of tert-butyl acetate (6.83 g, 58.79 mmol, 4.0 equiv) in THF (50 mL) was added dropwise LDA (2.0 M, 29.48 mL, 4.0 equiv) at -78 °C. After addition, the mixture was stirred at this temperature for 1 h, and then (R,Z)-N-((4-iodo-3- methylisoxazol-5-yl)methylene)-2-methylpropane-2-sulfinamide (5.0 g, 14.70 mmol, 1.0 equiv) in THF (10 mL) was added dropwise at -78 °C. The resulting mixture was stirred at -78 °C for 1 h. The reaction mixture was quenched by addition AcOH (8 mL) at -78 °C, and then diluted with H 2 O (50 mL) and extracted with EtOAc (50 mL). The combined organic layers were washed with brine (500 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (ISCO®; X g SepaFlash® Silica Flash Column, Eluent of 12-20% Ethylacetate/Petroleum ethergradient 100 mL/min). tert-butyl (3S)-3-((tert-butylsulfinyl)amino)-3-(4-iodo-3-methylisoxazo l-5- yl)propanoate (3.5 g, 7.67 mmol, 52% yield) was obtained as a yellow gum. [0483] Step 8: Preparation of tert-butyl (3S)-3-((tert-butylsulfinyl)amino)-3-(4-(2-(4- chlorobenzoyl)phenyl)-3-methylisoxazol-5-yl)propanoate. A mixture of tert-butyl (3S)-3- ((tert-butylsulfinyl)amino)-3-(4-iodo-3-methylisoxazol-5-yl) propanoate (19.18 g, 42.03 mmol, 1.2 equiv), (4-chlorophenyl)(2-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2 -yl)phenyl)methanone (12.0 g, 35.02 mmol, 1.0 equiv), Pd(dppf)Cl 2 (1.28 g, 1.75 mmol, 0.05 equiv) and Na 2 CO 3 (1.5 M in H 2 O, 71.78 mL, 3.07 equiv) in dioxane (300 mL) was degassed and purged with N 2 for 3 times, and then the mixture was stirred at 90 °C for 12 h under N2 atmosphere. The reaction mixture was partitioned between EtOAc (200mL) and H 2 O (200 mL). The organic phase was separated, 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: Welch Ultimate XB-CN 250*50*10um; mobile phase: [Hexane-EtOH (0.1% FA)]; gradient: 1%-10% B over 15 min). tert-butyl (3S)-3-((tert-butylsulfinyl)amino)-3-(4-(2-(4-chlorobenzoyl) phenyl)-3- methylisoxazol-5-yl)propanoate (27.0 g, 49.53 mmol, 70% yield) was obtained as a yellow gum. [0484] Step 9: Preparation of ethyl (S)-2-(6-(4-chlorophenyl)-1-methyl-4H-benzo[c] isoxazolo[4,5-e]azepin-4-yl)acetate. To a solution of tert-butyl (3S)-3-((tert-butylsulfinyl) amino)-3-(4-(2-(4-chlorobenzoyl)phenyl)-3-methylisoxazol-5-y l)propanoate (13.5 g, 24.77 mmol, 1.0 equiv) in EtOH (150 mL) was added acetyl chloride (11.0 g, 140.13 mmol, 10 mL, 5.6 equiv). The mixture was stirred at 60 °C for 12 h. The reaction mixture concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (ISCO®; X g SepaFlash® Silica Flash Column, Eluent of 50-100% Ethylacetate/Petroleum ethergradient 100 mL/min). ethyl (S)-2-(6-(4-chlorophenyl)-1-methyl-4H-benzo[c]isoxazolo[4,5- e]azepin-4-yl)acetate (18 g, 22.78mmol, 70% yield) was obtained as a yellow solid. [0485] Step 10: Preparation of (S)-2-(6-(4-chlorophenyl)-1-methyl-4H-benzo[c] isoxazolo[4,5-e]azepin-4-yl)acetic acid. To a solution of ethyl (S)-2-(6-(4-chlorophenyl)-1- methyl-4H-benzo[c]isoxazolo[4,5-e]azepin-4-yl)acetate (120 mg, 303 μmol, 1.0 equiv) in THF (2 mL) and MeOH (1 mL) was added LiOH.H 2 O (1.0 M, 911 μL, 3.0 equiv) in H 2 O (1 mL). The mixture was stirred at 20 °C for 2 h. The reaction mixture was quenched by addition 1 M HCl (2 mL) at 0 °C, and then extracted with EtOAc (2 x 2 mL). The combined organic layers were washed with brine (2 mL), dried over Na 2 SO 4 , filtered and concentrated under reduced pressure to give a residue. (S)-2-(6-(4-chlorophenyl)-1-methyl-4H-benzo[c] isoxazolo[4,5-e]azepin-4- yl)acetic acid (130 mg, crude) was obtained as a yellow solid. [0486] Step 11: Preparation of (S)-2-(6-(4-chlorophenyl)-1-methyl-4H-benzo[c] isoxazolo[4,5-e]azepin-4-yl)-N-(2,2-dimethoxyethyl)acetamide . To a solution of (S)-2-(6-(4- chlorophenyl)-1-methyl-4H-benzo[c]isoxazolo[4,5-e]azepin-4-y l)acetic acid (12.5 g, 34.08 mmol, 1.0 equiv), 2,2-dimethoxyethan-1-amine (10.75 g, 102.24 mmol, 11.14 mL, 3.0 equiv) in DMF (40 mL) was added DIEA (13.21 g, 102.24 mmol, 17.81 mL, 3.0 equiv) and HATU (19.44 g, 51.12 mmol, 1.5 eq) .The mixture was stirred at 25 o 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=5/1 to 2/1). (S)-2-(6-(4-chlorophenyl)-1- methyl-4H-benzo[c]isoxazolo[4,5-e]azepin-4-yl)-N-(2,2-dimeth oxyethyl) acetamide (13.0 g, 28.64 mmol, 84% yield) was obtained as a white solid. [0487] Step 12: Preparation of (S)-6-(4-chlorophenyl)-1-methyl-4-(oxazol-2-ylmethyl)- 4H-benzo[c]isoxazolo[4,5-e]azepine. A mixture of (S)-2-(6-(4-chlorophenyl)-1-methyl-4H- benzo[c]isoxazolo[4,5-e]azepin-4-yl)-N-(2,2-dimethoxyethyl)a cetamide (300 mg, 660 μmol, 1.0 equiv) in Eaton's reagent (22.72 g, 95.46 mmol, 15 mL, 144 equiv) was stirred at 100 o C for 3 h under N 2 atmosphere. The reaction mixture was poured into NaHCO 3 (500 mL) at 0 °C, and then extracted with EtOAc (2 x 300 mL). The combined organic layers were washed with brine (200 mL), dried over Na2SO4, filtered and then concentrated with (50 mL) NH3.H 2 O 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). (S)-6-(4-chlorophenyl)-1-methyl-4-(oxazol-2-ylmethyl)-4H- benzo[c]isoxazolo[4,5-e]azepine (3.3 g, 8.47 mmol, 64% yield) was obtained as a yellow solid. [0488] Step 13: Preparation of (S)-1-methyl-4-(oxazol-2-ylmethyl)-6-(4-(4,4,5,5- tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)-4H-benzo[c]isoxa zolo[4,5-e]azepine. A mixture of 4,4,5,5-tetramethyl-2-(4,4,5,5-tetramethyl-1,3,2-dioxaborola n-2-yl)-1,3,2-dioxaborolane (716 mg, 2.82 mmol, 2.2 equiv), (S)-6-(4-chlorophenyl)-1-methyl-4-(oxazol-2-ylmethyl)-4H- benzo[c]isoxazolo[4,5-e]azepine (500 mg, 1.28 mmol, 1.0 equiv), AcOK (415 mg, 4.23 mmol, 3.3 equiv) and dichloropalladium;tricyclohexylphosphane (189 mg, 256 μmol, 0.2 equiv) in THF (10 mL) was degassed and purged with N 2 for 3 times, and then the mixture was stirred at 75 °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=5/1 to 3/1). (S)-1-methyl-4-(oxazol-2-ylmethyl)-6-(4-(4,4,5,5-tetramethyl - 1,3,2-dioxaborolan-2-yl)phenyl)-4H-benzo[c]isoxazolo[4,5-e]a zepine (1.2 g, 2.49 mmol) was obtained as a yellow solid. [0489] Step 14: Preparation of 4-[(4S)-1-methyl-4-(oxazol-2-ylmethyl)-4H-isoxazolo [5, 4-d] [2] benzazepin-6-yl] phenol. To a solution of (S)-1-methyl-4-(oxazol-2-ylmethyl)-6-(4- (4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)-4H-benz o[c]isoxazolo[4,5-e]azepine (1.2 g, 2.49 mmol, 1.0 equiv) in THF (30 mL) was added H 2 O 2 (4.59 g, 47.23 mmol, 3.89 mL, 35% purity, 18.95 equiv) and NaOH (4.0 M, 1.87 mL, 3.0 equiv) in H 2 O. The mixture was stirred at 20 °C for 1 h. The reaction mixture was quenched by addition Na 2 SO 3 (10 mL) at 0 o C and stirred at 20 o C over 30 min. Then the mixture was diluted with H 2 O (10 mL) and extracted with EtOAc (2 x 10 mL). The combined organic layers were washed with brine (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 = 3/1 to 1/1). The residue was separated by SFC (column: DAICEL CHIRALPAK AS (250mm*30mm, 10um); mobile phase: [CO2-i- PrOH (0.1%NH3H 2 O)]; B%:25%, isocratic elution mode).4-[(4S)-1- methyl-4-(oxazol-2-ylmethyl)-4H-isoxazolo [5, 4-d] [2] benzazepin-6-yl] phenol (270 mg, 668 μmol, 26% yield, 92% purity) as obtained as a yellow solid. [0490] Step 15: Preparation of tert-butyl 2-[4-[(4S)-1-methyl-4-(oxazol-2-ylmethyl)-4H- isoxazolo[5,4-d][2]benzazepin-6-yl]phenoxy]-5-oxa-8-azaspiro [3.5]nonane-8-carboxylate. To a solution of PPh3 (211.8 mg, 807.8 μmol, 2.5 equiv) in THF (1 mL) was added DIAD (150.2 mg, 743.6 μmol, 144.0 μL, 2.3 equiv) at 0 °C. The mixture was stirred at 25 °C for 0.5 h. Then the mixture was added to 4-[(4S)-1-methyl-4-(oxazol-2-ylmethyl)-4H-isoxazolo [5, 4-d] [2] benzazepin-6-yl] phenol (120 mg, 323.1 μmol, 1.0 equiv), tert-butyl 2-hydroxy-5-oxa-8-azaspiro [3.5] nonane-8-carboxylate (157.2 mg, 646.2 μmol, 2.0 equiv) in THF (2 mL). The mixture was stirred at 25 °C for 0.5 h. The reaction mixture was concentrated under reduced pressure to remove solvent. The residue was purified by column chromatography (SiO 2 , Petroleum ether/ Ethyl acetate=20/1 to 0/1) to give a compound tert-butyl 2-[4-[(4S)-1-methyl-4-(oxazol-2- ylmethyl)-4H-isoxazolo[5,4-d][2]benzazepin-6-yl]phenoxy]-5-o xa-8-azaspiro[3.5]nonane-8- carboxylate (0.15 g, 78 % yield) as a white solid. [0491] Step 16: Preparation of (4S)-1-methyl-6-[4-(5-oxa-8-azaspiro [3.5] nonan-2-yloxy) phenyl]-4-(oxazol-2-ylmethyl)-4H- isoxazolo [5, 4-d] [2] benzazepine. To a solution of tert- butyl 2-[4-[(4S)-1-methyl-4-(oxazol-2-ylmethyl)-4H-isoxazolo[5,4-d ][2]benzazepin-6- yl]phenoxy]-5-oxa-8-azaspiro[3.5]nonane-8-carboxylate (0.1 g, 167.6 μmol, 1.0 equiv) in DCM (2 mL) was added TFA (1.5 g, 13.4 mmol, 1 mL, 80.3 equiv). The mixture was stirred at 25 °C for 1 h. The reaction mixture was adjusted to PH = 8 by aq.NaHCO320 mL and extracted with DCM 60 mL (3 x 20 mL). The combined organic layers were filtered and concentrated under reduced pressure to give a compound (4S)-1-methyl-6-[4-(5-oxa-8-azaspiro [3.5] nonan-2-yloxy) phenyl]-4-(oxazol-2-ylmethyl)-4H- isoxazolo [5, 4-d] [2] benzazepine (83 mg, 99 % yield) as a colourless oil. [0492] Step 17: Preparation of N-[3-(3-chloro-4-cyano-phenoxy)-2,2,4,4-tetramethyl- cyclobutyl]-2-[2-[4-[(4S)-1-methyl-4-(oxazol-2-ylmethyl)-4H- isoxazolo[5,4-d][2]benzazepin- 6-yl]phenoxy]-5-oxa-8-azaspiro[3.5]nonan-8-yl]pyrimidine-5-c arboxamide (I-221). To a solution of (4S)-1-methyl-6-[4-(5-oxa-8-azaspiro[3.5]nonan-2-yloxy)pheny l]-4-(oxazol-2- ylmethyl)-4H-isoxazolo[5,4-d][2]benzazepine (80 mg, 161 μmol, 1.0 equiv), 2-chloro-N-[3-(3- chloro-4-cyano-phenoxy)-2,2,4,4-tetramethyl-cyclobutyl]pyrim idine-5-carboxamide (74 mg, 177 μmol, 1.1 equiv) in NMP (1 mL) was added DIEA (62 mg, 483 μmol, 84 μL, 3.0 equiv). The mixture was stirred at 25 °C for 12 h. The residue was diluted with H 2 O 40 mL and extracted with DCM (20 mL X 3). The combined organic layers were 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]; gradient: 69%- 99% B over 10 min) to give a compound N-[3-(3-chloro-4-cyano-phenoxy)-2,2,4,4-tetramethyl- cyclobutyl]-2-[2-[4-[(4S)-1-methyl-4-(oxazol-2-ylmethyl)-4H- isoxazolo[5,4-d][2]benzazepin-6- yl]phenoxy]-5-oxa-8-azaspiro[3.5]nonan-8-yl]pyrimidine-5-car boxamide (50 mg, 99 % purity, 0.99 ee) as a white solid. 1 H NMR (400 MHz, MeOD-d6) δ 8.69 (s, 2 H), 7.85 - 7.95 (m, 1 H), 7.78 - 7.84 (m, 1 H), 7.63 - 7.76 (m, 2 H), 7.34 - 7.51 (m, 2 H), 7.29 (s, 2 H), 7.12 (s, 2 H), 6.98 (d, J = 9.17 Hz, 1 H), 6.80 (d, J = 7.95 Hz, 2 H), 4.44 - 4.67 (m, 2 H), 4.26 (s, 1 H), 4.07 - 4.17 (m, 1 H), 3.98 - 4.05 (m, 2 H), 3.79 - 3.94 (m, 4 H), 3.72 (s, 2 H), 2.48 - 2.63 (m, 5 H), 2.15 (d, J = 13.45 Hz, 2 H), 1.12 - 1.28 (m, 12 H). LC-MS: MS (ES + ): RT = 2.749 min, m/z = 879.4 [M + H + ]; LCMS method: 25 EXAMPLE 27 – Synthesis of N-[3-(3-chloro-4-cyano-phenoxy)-2,2,4,4-tetramethyl- cyclobutyl]-2-[2-[[4-[(9S)-4,5,13-trimethyl-9-(oxazol-2-ylme thyl)-3-thia-1,8,11,12- tetrazatricyclo[8.3.0.02,6]trideca-2(6),4,7,10,12-pentaen-7- yl]phenyl]methylene]-6- azaspiro[3.5]nonan-6-yl]pyrimidine-5-carboxamide (I-225)

[0493] Step 1: Preparation of 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]acetic acid. To a solution of tert-butyl 2-[(9S)-7-(4-chlorophenyl)-4,5,13-trimethyl-3-thia-1,8,11,12 - tetrazatricyclo[8.3.0.0 2,6 ]trideca-2(6),4,7,10,12-pentaen-9-yl]acetate (1.0 g, 2.1 mmol, 1.0 equiv) in DCM (10 mL) was added TFA (7.7 g, 67 mmol, 5.0 mL, 30 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 2-[(9S)- 7-(4-chlorophenyl)-4,5,13-trimethyl-3-thia-1,8,11,12-tetraza tricyclo[8.3.0.0 2,6 ]trideca- 2(6),4,7,10,12-pentaen-9-yl]acetic acid (877 mg, 99% yield). [0494] Step 2: Preparation of 2-[(9S)-7-(4-chlorophenyl)-4,5,13-trimethyl-3-thia- 1,8,11,12-tetrazatricyclo[8.3.0.0 2,6 ]trideca-2(6),4,7,10,12-pentaen-9-yl]acetamide. To a solution of 2-[(9S)-7-(4-chlorophenyl)-4,5,13-trimethyl-3-thia-1,8,11,12 -tetrazatricyclo [8.3.0.0 2,6 ]trideca-2(6),4,7,10,12-pentaen-9-yl]acetic acid (877 mg, 2.19 mmol, 1.0 equiv) and NH4Cl (351 mg, 6.56 mmol, 3.0 equiv) in DMF (8 mL) was added HATU (915 mg, 2.41 mmol, 1.1 equiv) and DIEA (848 mg, 6.56 mmol, 3.0 equiv). The mixture was stirred at 25 °C for 1 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%: 24%- 54%,10min) to give 2-[(9S)-7-(4-chlorophenyl)-4,5,13-trimethyl-3-thia-1,8,11,12 - tetrazatricyclo[8.3.0.0 2,6 ]trideca-2(6),4,7,10,12-pentaen-9-yl]acetamide (740 mg, 84% yield). [0495] Step 3: Preparation of 2-[[(9S)-7-(4-chlorophenyl)-4,5,13-trimethyl-3-thia- 1,8,11,12-tetrazatricyclo[8.3.0.0 2,6 ]trideca-2(6),4,7,10,12-pentaen-9-yl]methyl]oxazole. To a solution of 1,3-dioxol-2-one (103 mg, 1.20 mmol, 1.2 equiv) and 2-[(9S)-7-(4-chlorophenyl)- 4,5,13-trimethyl-3-thia-1,8,11,12-tetrazatricyclo[8.3.0.0 2,6 ]trideca-2(6),4,7,10,12-pentaen-9- yl]acetamide (400 mg, 1.00 mmol, 1.0 equiv) in PPA (4 mL). The mixture was stirred at 160 °C for 3 h. The mixture was quenched by addition H 2 O 50 mL at 0°C, and then diluted with EA 200 mL. The mixture was extracted with H 2 O (15 mL * 2). The combined organic layers were washed with H 2 O (10 mL * 3), dried over [Na2SO4], filtered and concentrated. The residue was purified by column chromatography (SiO 2 , Petroleum ether/Ethyl acetate=2/1 to 0/1) to give compound 2-[[(9S)-7-(4-chlorophenyl)-4,5,13-trimethyl-3-thia-1,8,11,1 2-tetrazatricyclo [8.3.0.0 2,6 ]trideca-2(6),4,7,10,12-pentaen-9-yl]methyl]oxazole (260 mg, 61% yield). [0496] Step 4: Preparation of tert-butyl 2-[(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2- yl)methylene]-6-azaspiro[3.5]nonane-6- carboxylate. To the solution of TMP (2.2 g, 16 mmol, 2.7 mL, 2.0 equiv) in THF (12 mL) was added n-BuLi (2.5 M, 6.4 mL, 2.0 equiv) at -30 °C, then the solution was stirred at -30 °C for 0.5 h. The solution of 4,4,5,5-tetramethyl-2-[(4,4,5,5- tetramethyl-1,3,2-dioxaborolan-2-yl)methyl]-1,3,2-dioxaborol ane (3.2 g, 12 mmol, 1.5 equiv) in THF (4 mL) was added to the solution at -70 °C. The mixture was stirred at -70 °C for 0.5 h. tert- butyl 2-oxo-6-azaspiro[3.5]nonane-6- carboxylate (1.9 g, 8.0 mmol, 1.0 equiv) in THF (4 mL) was added to the mixtue at -70 °C, the mixture was stirred at -70 °C for 2 h. Then stirred at 25 °C for 12 h and concentrated. The residue was purified by column chromatography (SiO 2 , Petroleum ether/Ethyl acetate = 20/1 to 10/1) to give tert-butyl 2-[(4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2-yl)methylene]-6-azaspiro[3.5]nonane-6- carboxylate (1.7 g, 59% yield) as colorless oil. [0497] Step 5: Preparation of tert-butyl 2-[[4-[(9S)-4,5,13-trimethyl-9-(oxazol-2- ylmethyl)-3-thia-1,8,11,12-tetrazatricyclo[8.3.0.02,6]tridec a-2(6),4,7,10,12-pentaen-7- yl]phenyl]methylene]-6-azaspiro[3.5]nonane-6-carboxylate. A mixture of 2-[[(9S)-7-(4- chlorophenyl)-4,5,13-trimethyl-3-thia-1,8,11,12-tetrazatricy clo[8.3.0.0 2,6 ]trideca-2(6),4,7,10,12- pentaen-9-yl]methyl]oxazole (600 mg, 1.4 mmol, 1.0 equiv), tert-butyl 2-[(4,4,5,5-tetramethyl- 1,3,2-dioxaborolan-2-yl)methylene]-6-azaspiro[3.5]nonane-6-c arboxylate (1 g, 2.8 mmol, 2.0 equiv), SPhos Pd G3 (110 mg, 142 μmol, 0.1 equiv) and Cs 2 CO 3 (461 mg, 1.4 mmol, 1.0 equiv) in DMF (10 mL) was degassed and purged with N2 for 3 times. The mixture was stirred at 90 °C for 12 h under N2 atmosphere and concentrated. The residue was purified by column chromatography (SiO 2 , Petroleum ether/Ethyl acetate = 0/1) to give tert-butyl 2-[[4-[(9S)-4,5,13- trimethyl-9-(oxazol-2-ylmethyl)-3-thia-1,8,11,12-tetrazatric yclo[8.3.0.02,6]trideca- 2(6),4,7,10,12-pentaen-7-yl]phenyl]methylene]-6-azaspiro[3.5 ]nonane-6-carboxylate (700 mg, 79% yield) as a yellow oil. [0498] Step 6: Preparation of 2-[[(9S)-7-[4-(6-azaspiro[3.5]nonan-2-ylidenemethyl) phenyl]-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]methyl]oxazole. To a solution of tert-butyl 2-[[4-[(9S)-4,5,13-trimethyl-9-(oxazol- 2-ylmethyl)-3-thia-1,8,11,12-tetrazatricyclo[8.3.0.0 2,6 ]trideca-2(6),4,7,10,12-pentaen-7- yl]phenyl]methylene]-6-azaspiro[3.5]nonane-6-carboxylate (700 mg, 1 mmol, 1.0 equiv) in DCM (3 mL) was added TFA (1 mL). The mixture was stirred at 25 °C for 1 h. The mixture was concentrated under reduced pressure to get the residue. The resiude was diltured with 10 mL DCM and adjusted the pH to 7~8 with saturated NaHCO3 aqueous, then extracted with DCM(20 mL * 3), the combined organic layer was concentrated under reduced pressure to give a crude product. [0499] Step 7: Preparation of N-[3-(3-chloro-4-cyano-phenoxy)-2,2,4,4-tetramethyl- cyclobutyl]-2-[2-[[4-[(9S)-4,5,13-trimethyl-9-(oxazol-2-ylme thyl)-3-thia-1,8,11,12- tetrazatricyclo[8.3.0.02,6]trideca-2(6),4,7,10,12-pentaen-7- yl]phenyl]methylene]-6- azaspiro[3.5]nonan-6-yl]pyrimidine-5-carboxamide (I-225). To a solution of 2-[[(9S)-7-[4-(6- azaspiro[3.5]nonan-2-ylidenemethyl)phenyl]-4,5,13-trimethyl- 3-thia-1,8,11,12- tetrazatricyclo[8.3.0.0 2,6 ]trideca-2(6),4,7,10,12-pentaen-9-yl]methyl]oxazole (280 mg, 534 μmol, 1.0 equiv) in NMP (3 mL) was added DIEA (138 mg, 1.1 mmol, 2.0 equiv) and 2-chloro-N-[3- (3-chloro-4-cyano-phenoxy)-2,2,4,4-tetramethyl-cyclobutyl]py rimidine-5-carboxamide (224 mg, 534 μmol, 1.0 equiv). The mixture was stirred at 25 °C for 12 h and concentrated. The residue was further by prep-TLC (SiO 2 , DCM/MeOH = 15/1) and prep-SFC (column: REGIS(S, S)WHELK-O1(250 mm * 25 mm, 10 μm); mobile phase: [CO 2 -MeOH/ACN]; B%: 70%, isocratic elution mode) to give N-[3-(3-chloro-4-cyano-phenoxy)-2,2,4,4-tetramethyl- cyclobutyl]-2-[2-[[4-[(9S)-4,5,13-trimethyl-9-(oxazol-2-ylme thyl)-3-thia-1,8,11,12- tetrazatricyclo[8.3.0.0 2,6 ]trideca-2(6),4,7,10,12-pentaen-7-yl]phenyl]methylene] -6-azaspiro [3.5]nonan-6-yl]pyrimidine-5-carboxamide (102 mg, 21% yield) as a white solid. 1 H NMR (400 MHz, CDCl 3 ): δ 8.67 (s, 2 H), 7.55-7.66 (m, 2 H), 7.33 (d, J = 7.6 Hz, 2 H), 7.16 (d, J = 8.2 Hz, 2 H), 7.05 (s, 1 H), 6.97 (s, 1 H), 6.81 (d, J = 8.6 Hz, 1 H), 6.22 (s, 1 H), 5.92-5.95 (m, 1 H), 4.73 (t, J = 7.2 Hz, 1 H), 4.03-4.18 (m, 4 H), 3.73-4.01 (m, 4 H), 2.77-2.86 (m, 1 H), 2.64-2.75 (m, 5 H), 2.52-2.60 (m, 1 H), 2.40 (s, 3 H), 1.79 (s, 2 H), 1.65-1.70 (m, 5 H), 1.17-1.30 (m, 12 H). LC- MS: MS (ES + ): RT = 2.910 min, m/z = 907.4 [M] + ; LCMS method: 25. EXAMPLE 28 – Synthesis of N-[3-(3-chloro-4-cyano-phenoxy)-2,2,4,4-tetramethyl- cyclobutyl]-2-[4-[3-[4-[(9S)-9-(2-methoxyethyl)-4,5,13-trime thyl-3-thia-1,8,11,12- tetrazatricyclo[8.3.0.02,6]trideca-2(6),4,7,10,12-pentaen-7- yl]phenyl]propoxy]-1- piperidyl]pyrimidine-5-carboxamide (I-227)

[0500] Step 1: Preparation of tert-butyl 4-[3-[4-[(9S)-9-(2-methoxyethyl)-4,5,13- trimethyl-3-thia-1,8,11,12-tetrazatricyclo[8.3.0.02,6]tridec a-2(6),4,7,10,12-pentaen-7- yl]phenyl]prop-2-ynoxy]piperidine-1-carboxylate. A mixture of(9S)-7-(4-chlorophenyl)-9-(2- methoxyethyl)-4,5,13-trimethyl-3-thia-1,8,11,12-tetrazatricy clo[8.3.0.02,6]trideca-2(6),4,7,10,12 -pentaene (285 mg, 711 μmol, 1.0 equiv), tert-butyl 4-prop-2-ynoxypiperidine-1-carboxylate (510 mg, 2.00mmol, 3.0 equiv), Cs2CO3 (463 mg, 1.00 mmol, 1.5 equiv), DavePhos Pd G3 (54 mg, 71 μmol, 0.1 equiv) in MeCN (3 mL) was degassed and purged with N 2 for 3 times, and the mixture was stirred at 90 °C for 2 h. The reaction mixture was filtered. The residue was purified by prep-HPLC(column: Phenomenex luna C18150*25mm*10um; mobile phase: [water(FA)- ACN];B%: 51%-81%,10min). Compound tert-butyl 4-[3-[4-[(9S)-9-(2-methoxyethyl)-4,5,13- trimethyl-3-thia-1,8,11,12-tetrazatricyclo[8.3.0.02,6]tridec a-2(6),4,7,10,12 -pentaen-7-yl]phenyl] prop-2-ynoxy]piperidine-1-carboxylate (85 mg, 0.14 mmol, 20% yield) was obtained as a white solid. [0501] Step 2: Preparation of tert-butyl 4-[3-[4-[(9S)-9-(2-methoxyethyl)-4,5,13- trimethyl-3-thia-1,8,11,12-tetrazatricyclo[8.3.0.02,6]tridec a-2(6),4,7,10,12-pentaen-7- yl]phenyl]propoxy]piperidine-1-carboxylate. To a solution of tert-butyl 4-[3-[4-[(9S)-9-(2- methoxyethyl)-4,5,13-trimethyl-3-thia-1,8,11,12-tetrazatricy clo[8.3.0.0 2,6 ]trideca-2(6),4,7,10,12- pentaen-7-yl]phenyl]prop-2-ynoxy]piperidine-1-carboxylate (85 mg, 0.14 mmol, 1.0 equiv) in TFE (5 mL) was added Pd/C (35 mg, 10% purity). The suspension was degassed and purged with H 2 . The mixture was stirred under H 2 (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-[3-[4- [(9S)-9-(2-methoxyethyl)-4,5,13-trimethyl-3-thia-1,8,11,12-t etrazatricyclo[8.3.0.0 2,6 ]trideca- 2(6),4,7,10,12-pentaen-7-yl]phenyl]propoxy]piperidine-1-carb oxylate (85 mg, 0.14 mmol, 99% yield) was obtained as a yellow oil. [0502] Step 3: Preparation of (9S)-9-(2-methoxyethyl)-4,5,13-trimethyl-7-[4-[3-(4- piperidyloxy)propyl]phenyl]-3-thia-1,8,11,12-tetrazatricyclo [8.3.0.02,6]trideca- 2(6),4,7,10,12-pentaene. To a solution of tert-butyl 4-[3-[4-[(9S)-9-(2-methoxyethyl)-4,5,13- trimethyl-3-thia-1,8,11,12-tetrazatricyclo[8.3.0.0 2,6 ]trideca-2(6),4,7,10,12-pentaen-7-yl]phenyl] propoxy]piperidine-1-carboxylate (85 mg, 0.14 mmol, 1.0 equiv) in DCM (1 mL) was added TFA (0.5 mL). The mixture was stirred at 25 °C for 2 h. The reaction mixture was concentrated to afford compound (9S)-9-(2-methoxyethyl)-4,5,13-trimethyl-7-[4-[3-(4-piperidy loxy)propyl] phenyl]-3-thia-1,8,11,12-tetrazatricyclo[8.3.0.0 2,6 ]trideca-2(6),4,7,10,12-pentaene (86 mg, 0.14 mmol, 99% yield, TFA salt) as a yellow oil. [0503] Step 4: Preparation of N-[3-(3-chloro-4-cyano-phenoxy)-2,2,4,4-tetramethyl- cyclobutyl]-2-[4-[3-[4-[(9S)-9-(2-methoxyethyl)-4,5,13-trime thyl-3-thia-1,8,11,12- tetrazatricyclo[8.3.0.02,6]trideca-2(6),4,7,10,12-pentaen-7- yl]phenyl]propoxy]-1- piperidyl]pyrimidine-5-carboxamide (I-227). To a solution of (9S)-9-(2-methoxyethyl)-4,5,13- trimethyl-7-[4-[3-(4-piperidyloxy)propyl]phenyl]-3-thia-1,8, 11,12-tetrazatricyclo [8.3.0.0 2,6 ]trideca-2(6),4,7,10,12-pentaene (85 mg, 0.14 mmol, 1.0 eqquiv, TFA salt), 2-chloro- N-[3-(3-chloro-4-cyano-phenoxy)-2,2,4,4-tetramethyl-cyclobut yl]pyrimidine-5-carboxamide (57 mg, 0.14 mmol, 1.0 equiv) in NMP (1 mL) was added K2CO3 (57 mg, 0.41 mmol, 3.0 equiv). The mixture was stirred at 50 °C for 1 h. The reaction mixture was filtered. The residue was purified by prep-HPLC (column: Phenomenex luna C18150*25mm*10um; mobile phase: [water(FA)-ACN]; B%: 82%-100%,10min). Compound N-[3-(3-chloro-4-cyano-phenoxy)- 2,2,4,4-tetramethyl-cyclobutyl]-2-[4-[3-[4-[(9S)-9-(2-methox yethyl)-4,5,13-trimethyl-3-thia- 1,8,11,12-tetrazatricyclo[8.3.0.0 2,6 ]trideca-2(6),4,7,10,12-pentaen-7-yl]phenyl]propoxy]-1 - piperidyl]pyrimidine-5-carboxamide (38 mg, 41 μmol, 30% yield, 98% purity) was obtained as a yellow solid. 1 H NMR: (CD3OD, 400 MHz) 8.73 (s, 2 H) 7.72 (s, 1 H) 7.40 (s, 2 H) 7.26 - 7.30 (s, 2 H) 7.13 (s, 1 H) 6.98 (s, 1 H) 4.59 (s, 2 H) 4.22 - 4.35 (s, 4 H) 4.13 (s, 1 H) 3.72 - 3.86 (s, 2 H) 3.48 - 3.61 (s, 5 H) 3.34 (s, 3 H) 2.77 (s, 2 H) 2.68 - 2.72 (s, 4 H) 2.45 (s, 3 H) 1.88 - 1.93 (s, 3 H) 1.69 - 1.70 (s, 1 H) 1.68 (s, 3 H) 1.49 - 1.58 (s, 2 H) 1.28 (s, 6 H) 1.21 (s, 6 H). LC-MS: MS (ES + ): RT =3.169min, m/z = 890.3[M+ H + ]; LCMS Method: 25 EXAMPLE 29 – Synthesis of N-[3-(3-chloro-4-cyano-phenoxy)-2,2,4,4-tetramethyl- cyclobutyl]-2-[4-[3-[4-[(9S)-4,5,9,13-tetramethyl-3-thia-1,8 ,11,12-tetrazatricyclo [8.3.0.02,6]trideca-2(6),4,7,10,12-pentaen-7-yl]phenyl]propo xy]-1-piperidyl]pyrimidine-5- carboxamide (I-228)

[0504] Step 1: Preparation of tert-butyl 4-[3-[4-[(9S)-9-(2-tert-butoxy-2-oxo-ethyl)- 4,5,13-trimethyl-3-thia-1,8,11,12-tetrazatricyclo[8.3.0.02,6 ]trideca-2(6),4,7,10,12-pentaen-7- yl]phenyl]prop-2-ynoxy]piperidine-1-carboxylate. A mixture of tert-butyl 2-[(9S)-7-(4- chlorophenyl)-4,5,13-trimethyl-3-thia-1,8,11,12-tetrazatricy clo[8.3.0.02,6]trideca-2(6),4,7,10,12- pentaen-9-yl]acetate (800 mg, 1.75 mmol, 1.0 equiv), tert-butyl 4-prop-2-ynoxypiperidine-1- carboxylate (649 mg, 2.71 mmol, 1.6 equiv), Ad2nBuP Pd G3(cataCXium® A Pd G3) (127 mg, 175 μmol, 0.1 equiv) and Cs2CO3 (1.48 g, 4.55 mmol, 2.6 equiv) in ACN (4 mL) and DMF (12 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 90 °C for 1 h under N2 atmosphere. The mixture was poured into water (80 mL). The aqueous phase was extracted with ethyl acetate (80 mL x 2). The combined organic phase was washed with brine (70 mL x 2), dried with anhydrous Na2SO4, filtered and concentrated in vacuum. The residue was purified by silica gel chromatography (0% - 100% ethyl acetate in petroleum ether) and further purified by prep-HPLC (column: Phenomenex luna C18150*40mm* 15um; mobile phase: [water (FA) - ACN]; B%: 62%-92%, 10min). Compound tert-butyl 4-[3-[4-[(9S)-9-(2- tert-butoxy-2-oxo-ethyl)-4,5,13-trimethyl-3-thia-1,8,11,12-t etrazatricyclo[8.3.0.02,6]trideca- 2(6),4,7,10,12-pentaen-7-yl]phenyl]prop-2-ynoxy]piperidine-1 -carboxylate (270 mg, 409 μmol, 23% yield) was obtained as a light yellow solid. [0505] Step 2: Preparation of tert-butyl 4-[3-[4-[(9S)-9-(2-tert-butoxy-2-oxo-ethyl)- 4,5,13-trimethyl-3-thia-1,8,11,12-tetrazatricyclo[8.3.0.02,6 ]trideca-2(6),4,7,10,12-pentaen-7- yl]phenyl]propoxy]piperidine-1-carboxylate. To a solution of tert-butyl 4-[3-[4-[(9S)-9-(2- tert-butoxy-2-oxo-ethyl)-4,5,13-trimethyl-3-thia-1,8,11,12-t etrazatricyclo[8.3.0.02,6]trideca- 2(6),4,7,10,12-pentaen-7-yl]phenyl]prop-2-ynoxy]piperidine-1 -carboxylate (260 mg, 394 μmol, 1.0 equiv) in CF 3 CH 2 OH (10 mL) was added Pd/C (220 mg, 10% purity) under N2 atmosphere. The suspension was degassed and purged with H 2 for 3 times. The mixture was stirred under H 2 (15 Psi) at 25 °C for 2 h. The mixture was filtered and concentrated under reduced pressure. Compound tert-butyl 4-[3-[4-[(9S)-9-(2-tert-butoxy-2-oxo-ethyl)-4,5,13-trimethyl -3-thia- 1,8,11,12-tetrazatricyclo[8.3.0.02,6]trideca-2(6),4,7,10,12- pentaen-7-yl]phenyl]propoxy] piperidine-1-carboxylate (260 mg, 392 μmol, 99% yield) was obtained as a yellow gum. [0506] Step 3: Preparation of 2-[(9S)-7-[4-[3-[(1-tert-butoxycarbonyl-4-piperidyl) oxy]propyl]phenyl]-4,5,13-trimethyl-3-thia-1,8,11,12-tetraza tricyclo[8.3.0.02,6] trideca- 2(6),4,7,10,12-pentaen-9-yl]acetic acid. To a solution of tert-butyl 4-[3-[4-[(9S)-9-(2-tert- butoxy-2-oxo-ethyl)-4,5,13-trimethyl-3-thia-1,8,11,12-tetraz atricyclo[8.3.0.0 2,6 ]trideca-2(6),4,7 ,10,12-pentaen-7-yl]phenyl]propoxy]piperidine-1-carboxylate (400 mg, 602 μmol, 1.0 equiv) in THF (5 mL) MeOH (5 mL) H 2 O (5 mL) was added LiOH.H 2 O (252 mg, 6.03 mmol, 10.0 equiv). The mixture was stirred at 25 °C for 12 h. The reaction mixture was quenched by addition water (50 mL), and extracted with ethyl acetate (30 mL x 3). The water-course by HCl (2 mol) adjust PH = 3, and extracted with ethyl acetate 30 mL (30 mL x 3). The combined organic dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. Compound 2-[(9S)- 7-[4-[3-[(1-tert-butoxycarbonyl-4-piperidyl)oxy]propyl]pheny l]-4,5,13-trimethyl-3-thia- 1,8,11,12-tetrazatricyclo[8.3.0.0 2,6 ]trideca-2(6),4,7,10,12-pentaen-9-yl]acetic acid (400 mg, crude) was obtained as a white solid. [0507] Step 4: Preparation of tert-butyl 4-[3-[4-[(9S)-9-[2-(1,3-dioxoisoindolin-2-yl)oxy- 2-oxo-ethyl]-4,5,13-trimethyl-3-thia-1,8,11,12-tetrazatricyc lo[8.3.0.02,6]trideca-2(6),4, 7,10,12-pentaen-7-yl]phenyl]propoxy]piperidine-1-carboxylate . To a solution of 2-[(9S)-7-[4- [3-[(1-tert-butoxycarbonyl-4-piperidyl)oxy]propyl]phenyl]-4, 5,13-trimethyl-3-thia-1,8,11,12- tetrazatricyclo[8.3.0.0 2,6 ]trideca-2(6),4,7,10,12-pentaen-9-yl]acetic acid (400 mg, 658 μmol, 1 equiv) in DCM (5 mL) was added DMAP (8.04 mg, 65.8 μmol, 0.1 equiv), 2-hydroxy- isoindoline-1,3-dione (112 mg, 691 μmol, 1.0 equiv), DIC (91 mg, 0.72 mmol, 112 μL, 1.1 equiv). The mixture was stirred at 25 °C for 12 h. To the reaction mixture was added water (20 mL) and the mixture was extracted with dichloromethane (20 mL x 3). The combined organic phase was washed with brine (20 mL), dried over anhydrous Na2SO4, filtered and concentrated in vacuo. Compound tert-butyl 4-[3-[4-[(9S)-9-[2-(1,3-dioxoisoindolin-2-yl)oxy-2-oxo-ethyl ]- 4,5,13-trimethyl-3-thia-1,8,11,12-tetrazatricyclo[8.3.0.0 2,6 ]trideca-2(6),4,7,10,12-pentaen-7- yl]phenyl]propoxy]piperidine-1-carboxylate (350 mg, 70 % yield) was obtained as a white solid. [0508] Step 5: Preparation of tert-butyl 4-[3-[4-[(9S)-4,5,9,13-tetramethyl-3-thia- 1,8,11,12-tetrazatricyclo[8.3.0.02,6]trideca-2(6),4,7,10,12- pentaen-7-yl]phenyl]propoxy] piperidine-1-carboxylate. A culture tube was charged with tert-butyl 4-[3-[4-[(9S)-9-[2-(1,3- dioxoisoindolin-2-yl)oxy-2-oxo-ethyl]-4,5,13-trimethyl-3-thi a-1,8,11,12-tetrazatricyclo [8.3.0.0 2,6 ]trideca-2(6),4,7,10,12-pentaen-7-yl]phenyl]propoxy]pi peridine-1-carboxylate (120 mg, 159 μmol, 1 equiv), Zn (410 mg, 6.27 mmol, 39.3 equiv) and a stir bar. The tube was then evacuated and backfilled with argon from a balloon. THF (10 mL) DMF (5 mL) and i-PrOH (628 mg, 10.4 mmol, 800 μL, 65.6 equiv) were added. A solution of dichloronickel; hexahydrate (120 mg, 505 μmol, 3.2 equiv), 4-tert-butyl-2-(4-tert-butyl-2-pyridyl)pyridine (270 mg, 1.01 mmol, 6.3 equiv) and phenylsilane (172 mg, 1.59 mmol, 197 μL, 10.0 equiv) were added in quick succession. The culture tube was then placed in a preheated 40 °C oil bath and stirred for 1 hour. The reaction mixture was quenched with saturated aqueous NH4Cl (5 mL) solution at 0 °C dropwise. And the resulting mixture was extracted with ethyl acetate (20 mL x 3). The combined organic layers were dried over anhydrous Na2SO4, filtered and concentrated in vacuum. The residue was purified by column chromatography (SiO 2 , petroleum ether/ethyl acetate=20/1 to 0/1). Compound tert-butyl 4-[3-[4-[(9S)-4,5,9,13-tetramethyl-3-thia-1,8,11,12-tetrazat ricyclo [8.3.0.0 2,6 ]trideca-2(6),4,7,10,12-pentaen-7-yl]phenyl]propoxy]pi peridine-1-carboxylate (25 mg, 28 % yield) was obtained as a white solid. [0509] Step 6: Preparation of (9S)-4,5,9,13-tetramethyl-7-[4-[3-(4-piperidyloxy) propyl]phenyl]-3-thia-1,8,11,12-tetrazatricyclo[8.3.0.02,6]t rideca-2(6),4,7,10,12-pentaene. To a solution of tert-butyl 4-[3-[4-[(9S)-4,5,9,13-tetramethyl-3-thia-1,8,11,12- tetrazatricyclo[8.3.0.0 2,6 ]trideca-2(6),4,7,10,12-pentaen-7-yl]phenyl]propoxy] piperidine-1- carboxylate (25 mg, 44 μmol, 1.0 equiv) in DCM (2 mL) was added TFA (1.54 g, 13.5 mmol, 1.0 mL, 304.6 equiv). The mixture was stirred at 25 °C for 0.5 h. The reaction mixture was concentrated in vacuo to give the crude product. Compound (9S)-4,5,9,13-tetramethyl-7-[4-[3- (4-piperidyloxy)propyl]phenyl]-3-thia-1,8,11,12-tetrazatricy clo [8.3.0.0 2,6 ]trideca-2(6),4,7,10,12- pentaene (20 mg, crude) was obtained as a white solid. [0510] Step 7: Preparation of N-[3-(3-chloro-4-cyano-phenoxy)-2,2,4,4-tetramethyl- cyclobutyl]-2-[4-[3-[4-[(9S)-4,5,9,13-tetramethyl-3-thia-1,8 ,11,12-tetrazatricyclo [8.3.0.02,6] trideca-2(6),4,7,10,12-pentaen-7-yl]phenyl]propoxy]-1-piperi dyl]pyrimidine-5-carboxamide (I-228). To a solution of (9S)-4,5,9,13-tetramethyl-7-[4-[3-(4-piperidyloxy)propyl]phe nyl]-3- thia-1,8,11,12-tetrazatricyclo[8.3.0.0 2,6 ]trideca-2(6),4,7,10,12-pentaene (20 mg, 43 μmol, 1 equiv) in NMP (0.5 mL) was added K2CO3 (30 mg, 0.21 mmol, 5.0 equiv) and2-chloro-N-[3-(3- chloro-4-cyano-phenoxy)-2,2,4,4-tetramethyl-cyclobutyl]pyrim idine-5-carboxamide (18 mg, 43 μmol, 1.0 equiv). The mixture was stirred at 50 °C for 0.5 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 C1875*30mm*3um;mobile phase: [water(FA)- ACN];B%: 65%-95%,7min as additive) to yield. Compound N-[3-(3-chloro-4-cyano-phenoxy)- 2,2,4,4-tetramethyl-cyclobutyl]-2-[4-[3-[4-[(9S)-4,5,9,13-te tramethyl-3-thia-1,8,11,12- tetrazatricyclo[8.3.0.0 2,6 ]trideca-2(6),4,7,10,12-pentaen-7-yl]phenyl]propoxy]-1 -piperidyl] pyrimidine-5-carboxamide (14 mg, 38 % yield, 100 % purity) was obtained as a white solid. 1 H NMR (400 MHz, DMSO-d6) δ 8.75 (s, 2 H), 7.90 (d, J = 8.8 Hz, 1 H), 7.71 (d, J = 9.2 Hz, 1 H), 7.36 (d, J = 8.4 Hz, 2 H), 7.20-7.27 (m, 3 H), 7.01 (s, 1 H), 4.29 (s, 1 H), 4.18-4.26 (m, 3 H), 4.03 (d, J = 9.2 Hz, 1 H), 3.54 (s, 1 H), 3.46-3.50 (m, 1 H), 3.41 (t, J = 6.4 Hz, 3 H), 2.67-2.70 (m, 2 H), 2.59 (s, 3 H), 2.40 (s, 3 H), 1.78-1.89 (m, 7 H), 1.62 (s, 3 H), 1.36-1.45 (m, 2 H), 1.21 (s, 6 H), 1.11 (s, 6 H). LC-MS: MS (ES + ): RT = 3.012 min, m/z = 846.3 [M + H + ]; LCMS method: 25 EXAMPLE 30 – Synthesis of N-[3-(3-chloro-4-cyano-phenoxy)-2,2,4,4-tetramethyl- cyclobutyl]-2-[2-[4-[(9S)-9-(cyanomethyl)-4,5,13-trimethyl-3 -thia-1,8,11,12- tetrazatricyclo[8.3.0.02,6]trideca-2(6),4,7,10,12-pentaen-7- yl]phenoxy]-7-azaspiro [3.5]nonan-7-yl]pyrimidine-5-carboxamide (I-229)

[0511] Step 1: Preparation of 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]acetic acid. A mixture 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 (4.9 g, 10.7 mmol, 1.0 equiv) in TFA (30 mL) and DCM (100 mL) was degassed and purged with N 2 for 3 times, and then the mixture was stirred at 25 °C for 12 h under N2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give the 2-[(9S)-7-(4-chlorophenyl)-4,5,13-trimethyl-3- thia-1,8,11,12-tetrazatricyclo[8.3.0.02,6]trideca-2(6),4,7,1 0,12-pentaen-9-yl]acetic acid (4.3 g, 10.7 mmol, 100% yield) as a brown oil. [0512] Step 2: Preparation of 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]acetamide. A mixture of 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]acetic acid (4.3 g, 10.7 mmol, 1.0 equiv), TEA (10.85 g, 107.3 mmol, 15 mL, 10.0 equiv), NH4Cl (1.72 g, 32.2 mmol, 3.0 equiv) in DMF (50 mL) and the mixture was stirred at 25°C for 10 min.Then HATU (5.30 g, 13.9 mmol, 1.3 equiv) and then the mixture was stirred at 25 °C for 2 h under N 2 atmosphere. The reaction mixture was diluted with Ethyl acetate (100 mL) and washed with NaCl (210 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (column: Kromasil Eternity XT 250*80mm*10um;mobile phase: [water( NH 4 HCO 3 )-ACN];B%: 28%-58%,20min) to give the 2-[(9S)-7-(4-chlorophenyl)-4,5,13- trimethyl-3-thia-1,8,11,12-tetrazatricyclo[8.3.0.02,6]tridec a-2(6),4,7,10,12-pentaen-9- yl]acetamide (2.9 g, 3.4 mmol, 32% yield) as a yellow solid. [0513] Step 3: Preparation of 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]acetonitrile. A mixture of DMSO (1.6 g, 20.0 mmol, 1.6 mL, 4.0 equiv) in DCM (15 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at -78 °C for 12 h under N2 atmosphere. Then the (COCl) 2 (1.9 g, 15,0 mmol, 1.3 mL, 3.0 equiv) was added to the mixture and stirred for 30min at -78°C under N 2 protection. Then the 2-[(9S)-7-(4-chlorophenyl)-4,5,13- trimethyl-3-thia-1,8,11,12-tetrazatricyclo[8.3.0.02,6]tridec a-2(6),4,7,10,12-pentaen-9- yl]acetamide (2.0 g, 5.0 mmol, 1.0 equiv) was added to the mixture and stirred for 30min at - 78°C under N 2. Then the TEA (4.0 g, 40.0 mmol, 5.6 mL, 8.0 equiv) was added to the mixture and stirred for 30min at -78°C. Reaction mixture was partitioned between Ethyl acetate (150 mL) and brine (210 mL). The organic phase was separated, washed with brine (70 mL x 3), dried over, filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO 2 , Petroleum ether/Ethyl acetate=0/1 to Dichloromethane : Methanol=10/1) to give the 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]acetonitrile (700 mg, 1.8 mmol, 36% yield) as a yellow solid. [0514] Step 4: Preparation of tert-butyl 2-[4-[(9S)-9-(cyanomethyl)-4,5,13-trimethyl-3- thia-1,8,11,12-tetrazatricyclo[8.3.0.02,6]trideca-2(6),4,7,1 0,12-pentaen-7-yl]phenoxy]-7- azaspiro[3.5]nonane-7-carboxylate. A mixture of 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]acetonitrile (500 mg, 1.3 mmol, 1.0 equiv) , tert-butyl 2-hydroxy-7-azaspiro[3.5]nonane-7-carboxylate (1.7 g, 6.6 mmol, 5.0 equiv), Cs2CO3 (2.1 g, 6.6 mmol, 5.0 equiv), [2-(2-aminophenyl)phenyl]- methylsulfonyloxy-palladium;ditert-butyl-[2-(2,4,6-triisopro pylphenyl)phenyl]phosphane (208 mg, 262 μmol, 0.2 equiv) in dioxane (5 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 reaction mixture was partitioned between Ethyl acetate 100 mL and brine (150 mL). The organic phase was separated, washed with brine (50 mL x 3), dried over, filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (column: Phenomenex luna C18 150*40mm* 15um;mobile phase: [water(FA)-ACN];B%: 55%-85%,15min) to give the tert-butyl 2-[4-[(9S)-9-(cyanomethyl)-4,5,13-trimethyl-3-thia-1,8,11,12 -tetrazatricyclo[8.3.0.02,6]trideca- 2(6),4,7,10,12-pentaen-7-yl]phenoxy]-7-azaspiro[3.5]nonane-7 -carboxylate (300 mg, 511 μmol, 39% yield) as a brown solid. [0515] Step 5: Preparation of 2-[(9S)-7-[4-(7-azaspiro[3.5]nonan-2-yloxy)phenyl]-4,5,13- trimethyl-3-thia-1,8,11,12-tetrazatricyclo[8.3.0.02,6]tridec a-2(6),4,7,10,12-pentaen-9- yl]acetonitrile. A mixture of tert-butyl 2-[4-[(9S)-9-(cyanomethyl)-4,5,13-trimethyl-3-thia- 1,8,11,12-tetrazatricyclo[8.3.0.02,6]trideca-2(6),4,7,10,12- pentaen-7-yl]phenoxy]-7-azaspiro [3.5]nonane-7-carboxylate (300 mg, 511 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 partitioned between Ethyl acetate (200 mL) and water (100 mL). The organic phase was separated, washed with brine 200 mL (100 mL x 2) [Drying agent], filtered and concentrated under reduced pressure to give the 2-[(9S)-7-[4-(7- azaspiro[3.5]nonan-2-yloxy)phenyl]-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]acetonitrile (240 mg, 493 μmol, 96% yield) as a yellow solid. [0516] Step 6: Preparation of N-[3-(3-chloro-4-cyano-phenoxy)-2,2,4,4-tetramethyl- cyclobutyl]-2-[2-[4-[(9S)-9-(cyanomethyl)-4,5,13-trimethyl-3 -thia-1,8,11,12- tetrazatricyclo[8.3.0.02,6]trideca-2(6),4,7,10,12-pentaen-7- yl]phenoxy]-7-azaspiro[3.5] nonan-7-yl]pyrimidine-5-carboxamide (I-229). A mixture of 2-[(9S)-7-[4-(7- azaspiro[3.5]nonan-2-yloxy)phenyl]-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]acetonitrile (120 mg, 246 μmol, 1.0 equiv), 2-chloro-N-[3-(3- chloro-4-cyano-phenoxy)-2,2,4,4-tetramethyl-cyclobutyl]pyrim idine-5-carboxamide (103 mg, 246 μmol, 1.0 equiv), DIEA (64 mg, 493 μmol, 86 μL, 2.0 equiv) in NMP (2 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 25 °C for 12 h under N2 atmosphere. The residue was purified by prep-HPLC (column: Phenomenex luna C18 150*25mm* 10um;mobile phase: [water(FA)-ACN];gradient:70%-90% B over 10 min) to give the N-[3-(3-chloro-4-cyano-phenoxy)-2,2,4,4-tetramethyl-cyclobut yl]-2-[2-[4-[(9S)-9- (cyanomethyl)-4,5,13-trimethyl-3-thia-1,8,11,12-tetrazatricy clo[8.3.0.02,6]trideca-2(6),4,7,10,12 -pentaen-7-yl]phenoxy]-7-azaspiro[3.5]nonan-7-yl]pyrimidine- 5-carboxamide (57 mg, 64 μmol, 26% yield) as a white solid. 1 H NMR: (400 MHz, MeOD) δ = 8.72 (s, 2H), 7.72 (d, J = 8.8 Hz, 1H), 7.47 (d, J = 8.4 Hz, 2H), 7.12 (d, J = 2.4 Hz, 1H), 6.98 (d, J = 2.4, 8.8 Hz, 1H), 6.88 (d, J = 8.8 Hz, 2H), 4.83 - 4.78 (m, 1H), 4.53 (d, J = 5.6, 8.0 Hz, 1H), 4.27 (s, 1H), 4.17 - 4.04 (m, 1H), 3.95 - 3.79 (m, 4H), 3.67 - 3.40 (m, 2H), 2.70 (s, 3H), 2.54 (d, J = 7.6, 12.5 Hz, 2H), 2.45 (s, 3H), 1.98 (d, J = 6.4, 12.8 Hz, 2H), 1.81 - 1.63 (m, 7H), 1.28 (s, 6H), 1.21 (s, 6H). LC-MS: MS (ES + ): RT = 2.844 min, m/z = 869.4 [M +H + ] EXAMPLE 31 – Synthesis of the N-[3-(3-chloro-4-cyano-phenoxy)-2,2,4,4-tetramethyl- cyclobutyl]-2-[2-[4-[(9E)-4,5,13-trimethyl-9-(oxazol-2-ylmet hylene)-3-thia-1,8,11,12- tetrazatricyclo[8.3.0.02,6]trideca-2(6),4,7,10,12-pentaen-7- yl]phenoxy]-7- azaspiro[3.5]nonan-7-yl]pyrimidine-5-carboxamide (I-230)

[0517] Step 1: Preparation of 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]acetic acid. To a solution of tert-butyl 2-[(9S)-7-(4-chlorophenyl)-4,5,13-trimethyl-3-thia-1,8,11,12 - tetrazatricyclo[8.3.0.0 2,6 ]trideca-2(6),4,7,10,12-pentaen-9-yl]acetate (1.0 g, 2.1 mmol, 1.0 equiv) in DCM (10 mL) was added TFA (7.7 g, 67 mmol, 5.0 mL, 30 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 2-[(9S)- 7-(4-chlorophenyl)-4,5,13-trimethyl-3-thia-1,8,11,12-tetraza tricyclo[8.3.0.0 2,6 ]trideca- 2(6),4,7,10,12-pentaen-9-yl]acetic acid (877 mg, 99% yield). [0518] Step 2: Preparation of 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]acetamide. To a solution of 2-[(9S)-7-(4-chlorophenyl)-4,5,13-trimethyl-3-thia-1,8,11,12 -tetrazatricyclo [8.3.0.0 2,6 ]trideca-2(6),4,7,10,12-pentaen-9-yl]acetic acid (877 mg, 2.19 mmol, 1.0 equiv) and NH 4 Cl (351 mg, 6.56 mmol, 3.0 equiv) in DMF (8 mL) was added HATU (915 mg, 2.41 mmol, 1.1 equiv) and DIEA (848 mg, 6.56 mmol, 3.0 equiv). The mixture was stirred at 25 °C for 1 h. The mixture was 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%: 24%- 54%,10min) to give 2-[(9S)-7-(4-chlorophenyl)-4,5,13-trimethyl-3-thia-1,8,11,12 - tetrazatricyclo[8.3.0.0 2,6 ]trideca-2(6),4,7,10,12-pentaen-9-yl]acetamide (740 mg, 84% yield). [0519] Step 3: Preparation of 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]methyl]oxazole. To a solution of 1,3-dioxol-2-one (103 mg, 1.20 mmol, 1.2 equiv) and 2-[(9S)-7-(4-chlorophenyl)- 4,5,13-trimethyl-3-thia-1,8,11,12-tetrazatricyclo[8.3.0.0 2,6 ]trideca-2(6),4,7,10,12-pentaen-9- yl]acetamide (400 mg, 1.00 mmol, 1.0 equiv) in PPA (4 mL). The mixture was stirred at 160 °C for 3 h. The mixture was quenched by addition H 2 O 50 mL at 0°C, and then diluted with EA 200 mL. The mixture was extracted with H 2 O (15 mL * 2). The combined organic layers were washed with H 2 O (10 mL * 3), dried over [Na2SO4], filtered and concentrated. The residue was purified by column chromatography (SiO 2 , Petroleum ether/Ethyl acetate=2/1 to 0/1) to give compound 2-[[(9S)-7-(4-chlorophenyl)-4,5,13-trimethyl-3-thia-1,8,11,1 2-tetrazatricyclo [8.3.0.0 2,6 ]trideca-2(6),4,7,10,12-pentaen-9-yl]methyl]oxazole (260 mg, 61% yield). [0520] Step 4: Preparation of tert-butyl 2-[4-[(9S)-4,5,13-trimethyl-9-(oxazol-2- ylmethyl)-3-thia-1,8,11,12-tetrazatricyclo[8.3.0.02,6]tridec a-2(6),4,7,10,12-pentaen-7- yl]phenoxy]-7-azaspiro[3.5]nonane-7-carboxylate. To a solution of 2-[[(9S)-7-(4- chlorophenyl)-4,5,13-trimethyl-3-thia-1,8,11,12-tetrazatricy clo[8.3.0.02,6]trideca-2(6),4,7,10,12- pentaen-9-yl]methyl]oxazole (300 mg, 707 μmol, 1.0 equiv), tert-butyl 2-hydroxy-7- azaspiro[3.5]nonane-7-carboxylate (853 mg, 3.54 mmol, 5.0 equiv) in dioxane (5 mL) was added tBuXPhos Pd G3 (112 mg, 141 μmol, 0.2 equiv) and Cs 2 CO 3 (1.1 g, 3.5 mmol, 5.0 equiv). The mixture was degassed and purged with N2 for 3 times, and then the mixture was stirred at 90 °C for 2 h under N2 atmosphere. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by flash silica gel chromatography (ISCO®; 2 g SepaFlash® Silica Flash Column, Eluent of 0~3% Methanol/Dichloromethanegradient @ 18 mL/min) to give the tert-butyl 2-[4-[(9S)-4,5,13-trimethyl-9-(oxazol-2-ylmethyl)-3-thia- 1,8,11,12-tetrazatricyclo[8.3.0.02,6]trideca-2(6),4,7,10,12- pentaen-7-yl]phenoxy]-7- azaspiro[3.5]nonane-7-carboxylate (210 mg, 333 μmol, 47% yield) as a yellow solid. [0521] Step 5: Preparation of tert-butyl 2-[4-[(9Z)-4,5,13-trimethyl-9-(oxazol-2- ylmethylene)-3-thia-1,8,11,12-tetrazatricyclo[8.3.0.02,6]tri deca-2(6),4,7,10,12-pentaen-7- yl]phenoxy]-7-azaspiro[3.5]nonane-7-carboxylate. A mixture of tert-butyl 2-[4-[(9S)-4,5,13- trimethyl-9-(oxazol-2-ylmethyl)-3-thia-1,8,11,12-tetrazatric yclo[8.3.0.02,6]trideca- 2(6),4,7,10,12-pentaen-7-yl]phenoxy]-7-azaspiro[3.5]nonane-7 -carboxylate (200 mg, 318 μmol, 1.0 equiv), Pd(OAc) 2 (22 mg, 96 μmol, 0.3 equiv), K 2 CO 3 (66 mg, 477 μmol, 1.5 equiv) and dcpp (83 mg, 191 μmol, 0.6 equiv) in DMSO (2 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 120 °C for 12 h under N2 atmosphere. The reaction mixture 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];gradient:69%-99% B over 10 min) to give the tert-butyl 2-[4-[(9Z)-4,5,13- trimethyl-9-(oxazol-2-ylmethylene)-3-thia-1,8,11,12-tetrazat ricyclo[8.3.0.02,6]trideca- 2(6),4,7,10,12-pentaen-7-yl]phenoxy]-7-azaspiro[3.5]nonane-7 -carboxylate (60 mg, 96 μmol, 30% yield) as a yellow solid. [0522] Step 6: Preparation of 2-[(E)-[7-[4-(7-azaspiro[3.5]nonan-2-yloxy)phenyl]-4,5,13- trimethyl-3-thia-1,8,11,12-tetrazatricyclo[8.3.0.02,6]tridec a-2(6),4,7,10,12-pentaen-9- ylidene]methyl]oxazole. A mixture of tert-butyl 2-[4-[(9E)-4,5,13-trimethyl-9-(oxazol-2- ylmethylene)-3-thia-1,8,11,12-tetrazatricyclo[8.3.0.02,6]tri deca-2(6),4,7,10,12-pentaen-7- yl]phenoxy]-7-azaspiro[3.5]nonane-7-carboxylate (50 mg, 80 μmol, 1.0 equiv) in DCM (3 mL) and TFA (1 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 25 °C for 2 h under N 2 atmosphere. The reaction mixture was concentrated under reduced pressure to give the 2-[(E)-[7-[4-(7-azaspiro[3.5]nonan-2-yloxy)phenyl]-4,5,13-tr imethyl-3-thia- 1,8,11,12-tetrazatricyclo[8.3.0.02,6]trideca-2(6),4,7,10,12- pentaen-9-ylidene]methyl]oxazole (51 mg, 80 μmol, 100% yield, TFA) as a colorless oil. [0523] Step 7: Preparation of the N-[3-(3-chloro-4-cyano-phenoxy)-2,2,4,4-tetramethyl- cyclobutyl]-2-[2-[4-[(9E)-4,5,13-trimethyl-9-(oxazol-2-ylmet hylene)-3-thia-1,8,11,12- tetrazatricyclo[8.3.0.02,6]trideca-2(6),4,7,10,12-pentaen-7- yl]phenoxy]-7-azaspiro[3.5] nonan-7-yl]pyrimidine-5-carboxamide (I-230). To a solution of 2-[(E)-[7-[4-(7- azaspiro[3.5]nonan-2-yloxy)phenyl]-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-ylidene]methyl] oxazole (50 mg, 78 μmol, 1.0 equiv TFA) in NMP (2 mL) was added 2-chloro-N-[3-(3-chloro-4-cyano-phenoxy)-2,2,4,4- tetramethyl-cyclobutyl]pyrimidine-5-carboxamide (36 mg, 86 μmol, 1.1 equiv) and K 2 CO 3 (108 mg, 780 μmol, 10.0 equiv). The mixture was stirred at 50 °C for 1 h. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (column: Waters xbridge 150*25mm 10um;mobile phase: [water( NH4HCO3)- ACN];gradient:60%-90% B over 10 min) to give the N-[3-(3-chloro-4-cyano-phenoxy)-2,2,4,4- tetramethyl-cyclobutyl]-2-[2-[4-[(9E)-4,5,13-trimethyl-9-(ox azol-2-ylmethylene)-3-thia- 1,8,11,12-tetrazatricyclo[8.3.0.02,6]trideca-2(6),4,7,10,12- pentaen-7-yl]phenoxy]-7- azaspiro[3.5]nonan-7-yl]pyrimidine-5-carboxamide (18 mg, 20 μmol, 25% yield) as a white solid. 1 H NMR: (400 MHz, MeOD) δ = 8.73 (s, 2H), 8.07 (s, 1H), 7.82 - 7.67 (m, 3H), 7.35 (s, 1H), 7.13 (d, J = 2.4 Hz, 1H), 7.01 - 6.88 (m, 3H), 6.44 (s, 1H), 4.58 (s, 2H), 4.27 (s, 1H), 4.13 (s, 1H), 3.98 - 3.82 (m, 4H), 2.77 (s, 3H), 2.61 - 2.52 (m, 2H), 2.41 (s, 3H), 2.07 - 1.96 (m, 2H), 1.80 - 1.64 (m, 7H), 1.29 - 1.26 (m, 6H), 1.21 (s, 6H). LC-MS: MS (ES + ): RT = 3.092 min, m/z = 909.2 [M +H + ] EXAMPLE 32 – Synthesis of N-[3-(3-chloro-4-cyano-phenoxy)-2,2,4,4-tetramethyl- cyclobutyl]-2-[2-[4-[3-methyl-12-(methylamino)-5-oxa-4,8,11- triazatricyclo [8.4.0.02,6]tetradeca-1(10),2(6),3,8,11,13-hexaen-9-yl]pheno xy]-6-azaspiro[3.5]nonan-6- yl]pyrimidine-5-carboxamide (I-231)

[0524] Step 1: Preparation of (4-iodo-3-methyl-isoxazol-5-yl)methyl benzoate. To a solution of (3-methylisoxazol-5-yl) methyl benzoate (500 mg, 2.30 mmol, 1.0 equiv) in TFA (5 mL) was added NIS (1.04 g, 4.60 mmol, 2.0 equiv) .The mixture was stirred at 90 °C for 12 h .The residue was quenched with NaHCO 3 (10 ML), then diluted with H 2 O (50 mL) and extracted with EA (20 mL x 3), the organic phase was concentrated to afford crude product. The residue was purified by silica chromatography (PE:EA=1:0-30:1) to afford (4-iodo-3-methyl-isoxazol-5- yl)methyl benzoate (690 mg, 2.01 mmol, 87% yield) as a yellow oil . [0525] Step 2: Preparation of [3-methyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2- yl)isoxazol-5-yl]methyl benzoate. To a solution of (4-iodo-3-methyl-isoxazol-5-yl)methyl benzoate (32.0 g, 93.3 mmol, 1.0 equiv) and 4,4,5,5-tetramethyl-1,3,2-dioxaborolane (35.8 g, 280 mmol, 41 mL, 3.0 equiv) in dioxane (300 mL) was added Pd(PPh3) 2 Cl2 (3.27 g, 4.66 mmol, 0.1 equiv) and TEA (28.3 g, 280 mmol, 39 mL, 3.0 equiv) . The suspension was degassed and purged with N 2 for 3 times, the mixture was stirred under 80 °C for 12 h, the reaction mixture was concentrated to afford crude product. The residue was purified by prep-HPLC (column: Phenomenex luna C18250*80mm*10 um;mobile phase: [water(FA)-ACN];gradient:65%-95% B over 20 min) to afford [3-methyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)iso xazol-5- yl]methyl benzoate (17 g, 50.0 mmol, 53% yield) as a yellow solid. [0526] Step 3: Preparation of methyl 6-[bis(tert-butoxycarbonyl)amino]-3-bromo- pyridine-2-carboxylate. To a solution of methyl 6-amino-3-bromo-pyridine-2-carboxylate (25.0 g, 108 mmol, 1.0 equiv) and tert-butoxycarbonyl tertbutylcarbonate (70.8 g, 325 mmol, 3.0 equiv) in THF (200 mL) was added DMAP (2.64 g, 21.6 mmol, 0.2 equiv) at 0 °C. The mixture was stirred at 60 °C for 12 h. The reaction mixture was filtered to afford crude product. The residue was purified by column chromatography (SiO 2 , Petroleum ether/Ethyl acetate = 50/1 to 20/1) to afford methyl 6-[bis(tert-butoxycarbonyl)amino]-3-bromo-pyridine-2-carboxy late (34.5 g, 80.0 mmol, 74%) as a yellow solid . [0527] Step 4: Preparation of methyl 3-bromo-6-(tert-butoxycarbonylamino)pyridine-2- carboxylate. To a solution of methyl 6-[bis(tert-butoxycarbonyl)amino]-3-bromo-pyridine-2- carboxylate (24.0 g, 55.6 mmol, 1.0 equiv) in DCM (209 mL) was added TFA (19.0 g, 167 mmol, 12.3 mL, 3.0 equiv) .The mixture was stirred at 0 °C for 12 h .The residue was added NaHCO 3 until pH>7, diluted with H 2 O (200 mL), then extracted with DCM(300 mL*3) and washed with H 2 O (2 x 100 mL). The organic phase was concentrated to afford crude product. The crude product methyl 3-bromo-6-(tert-butoxycarbonylamino) pyridine-2-carboxylate (18.5 g, crude) was used into the next step without further purification. The product was a yellow oil. [0528] Step 5: Preparation of methyl 3-bromo-6-[tert-butoxycarbonyl(methyl) amino]pyridine-2-carboxylate. To a solution of methyl 3-bromo-6-(tert-butoxycarbonylamino) pyridine-2-carboxylate (18.5 g, 55.9 mmol, 1.0 equiv) in DMF (180 mL) was added MeI (9.52 g, 67.0 mmol, 1.2 equiv) and Cs 2 CO 3 (54.6 g, 167 mmol, 3.0 equiv) at 0°C. The mixture was stirred at 25 °C for 12 h. The residue was quenched by NH4Cl (50ml) , then the residue was diluted with H 2 O (900 mL) and extracted with EA(3 x 300 mL), and the organic phase was concentrated to afford crude product. The residue was purified by silica chromatography (PE:EA=1:0-40:1) to afford methyl 3-bromo-6-[tert-butoxycarbonyl(methyl)amino]pyridine-2-carbo xylate (16.2 g, 46.9 mmol, 84% yield) as a yellow oil. [0529] Step 6: Preparation of 3-bromo-6-[tert-butoxycarbonyl(methyl)amino]pyridine- 2-carboxylic acid. To a solution of methyl 3-bromo-6-[tert-butoxycarbonyl(methyl)amino] pyridine-2-carboxylate (19.0 g, 55.0 mmol, 1.0 equiv) in THF (80 mL) and H 2 O (40 mL) was added LiOH (2.64 g, 110 mmol, 2.0 equiv). The mixture was stirred at 25 °C for 12 h. The reaction mixture was added CH 3 COOH until pH < 7, then diluted with H 2 O (100 mL) and extracted with EA(3 x 200mL). The organic phase was concentrated to afford crude product. The crude product 3-bromo-6-[tert-butoxycarbonyl(methyl)amino]pyridine-2-carbo xylic acid (18.3 g, crude) was used into the next step without further purification. The product was a yellow oil. [0530] Step 7: Preparation of tert-butyl N-[5-bromo-6-[methoxy(methyl)carbamoyl]-2- pyridyl]-N-methyl-carbamate. To a solution of 3-bromo-6-[tert-butoxycarbonyl(methyl)amino] pyridine-2-carboxylic acid (16.7 g, 50.4 mmol, 1.0 equiv) and N-methoxymethanamine (5.90 g, 60.5 mmol, 1.2 equiv, HCl salt) in DMF (160 mL) was added HATU (28.8 g, 75.6 mmol, 1.5 equiv) and DIEA (13.0 g, 101 mmol, 2.0 equiv) .The mixture was stirred at 25 °C for 1 h. The residue was diluted with H 2 O (500 mL) and extracted with EA (300 mL x 3).The organic phase was concentrated to afford crude product. the residue was purified by silica chromatography (PE:EA = 10:1-4:1) and prep-HPLC (column: Kromasil Eternity XT 250*80mm*10um;mobile phase: [water (ammonia hydroxide v/v)-ACN];gradient:38%-68% B over 20 min) to afford tert- butyl N-[5-bromo-6-[methoxy(methyl)carbamoyl]-2-pyridyl]-N-methyl- carbamate (15.0 g, 39.3 mmol, 78% yield, 98% purity) as a yellow solid. [0531] Step 8: Preparation of tert-butyl N-[5-bromo-6-(4-chlorobenzoyl)-2-pyridyl]-N- methyl-carbamate. To a solution of tert-butyl N-[5-bromo-6-[methoxy(methyl)carbamoyl]-2- pyridyl]-N-methyl-carbamate (6.70 g, 17.9 mmol, 1.0 equiv) in THF (100 mL) was added bromo-(4-chlorophenyl)magnesium (1 M, 89.5 mL, 5.0 equiv) at -78 °C for 0.5 h. The reaction mixture was allowed to slowly warm to 25 °C for 1 h. The residue was quenched by NH4Cl (50ml), then diluted with H 2 O (300 mL) and extracted with EA (3 x 300 mL). The organic phase was concentrated to afford crude product. The residue was purified by silica chromatography (PE:EA = 1:0-50:1) to afford tert-butyl N-[5-bromo-6-(4-chlorobenzoyl)-2-pyridyl]-N-methyl- carbamate (6.30 g, 14.80 mmol, 83% yield) as a yellow solid. [0532] Step 9: Preparation of [4-[6-[tert-butoxycarbonyl(methyl)amino]-2-(4- chlorobenzoyl)-3-pyridyl]-3-methyl-isoxazol-5-yl]methyl benzoate. To a solution of tert-butyl N-[5-bromo-6-(4-chlorobenzoyl)-2-pyridyl]-N-methyl-carbamate (8.50 g, 20.0 mmol, 1.0 equiv) and [3-methyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)iso xazol-5-yl]methyl benzoate (7.54 g, 22.0 mmol, 1.1 equiv) and[3-methyl-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2- yl)isoxazol-5-yl]methyl benzoate (7.54 g, 22.0 mmol, 1.1 equiv) in dioxane (85 mL) and H 2 O (8.5 mL) was added Pd(dppf)Cl 2 (730 mg, 999 μmol, 0.1 equiv) and K 2 CO 3 (5.52 g, 39.9 mmol, 2.0 equiv). The mixture was stirred at 60 °C for 3 h.the reaction mixture was concentrated to afford crude product and purified by silica chromatography (PE:EA = 30:1-10:1) to afford [4-[6- [tert-butoxycarbonyl(methyl)amino]-2-(4-chlorobenzoyl)-3-pyr idyl]-3-methyl-isoxazol-5- yl]methyl benzoate (9.0 g, 16.0 mmol, 80% yield) as a yellow oil. [0533] Step 10: Preparation of tert-butyl N-[6-(4-chlorobenzoyl)-5-[5-(hydroxymethyl)- 3-methyl-isoxazol-4-yl]-2-pyridyl]-N-methyl-carbamate. To a solution of [4-[6-[tert- butoxycarbonyl(methyl)amino]-2-(4-chlorobenzoyl)-3-pyridyl]- 3-methyl-isoxazol-5-yl]methyl benzoate (1.50 g, 2.67 mmol, 1.0 equiv) in THF (6 mL) and H 2 O (3 mL) was added LiOH (128 mg, 5.34 mmol, 2.0 equiv) .The mixture was stirred at 25 °C for 12 h. The residue was diluted with H 2 O (20 mL) and extracted with DCM (3 x 20 mL). The organic phase was concentrated to afford crude product. The residue was purified by silica chromatography (PE:EA = 10:1-3:1) to afford tert-butyl N-[6-(4-chlorobenzoyl)-5-[5-(hydroxymethyl)-3-methyl-isoxazo l-4-yl]-2- pyridyl]-N-methyl-carbamate (0.90 g, 1.97 mmol, 74% yield) as a yellow oil. [0534] Step 11: Preparation of ([4-[6-[tert-butoxycarbonyl(methyl)amino]-2-(4- chlorobenzoyl)-3-pyridyl]-3-methyl-isoxazol-5-yl]methyl methanesulfonate. To a solution of tert-butyl N-[6-(4-chlorobenzoyl)-5-[5-(hydroxymethyl)-3-methyl-isoxazo l-4-yl]-2-pyridyl]-N- methyl-carbamate (13.2 g, 28.8 mmol, 1.0 equiv) in DCM (130 mL) was added MsCl (10.7 g, 93.3 mmol, 7.22 mL, 3.2 equiv) and TEA (14.6 g, 144 mmol, 5.0 equiv). The mixture was stirred at 25 °C for 0.5 h .The reaction was quenched by cold H 2 O (50 mL) , then diluted with H 2 O (100 mL) and extracted with DCM (80 mL x 3).the organic phase was concentrated to afford crude product, the crude product ([4-[6-[tert-butoxycarbonyl(methyl)amino]-2-(4-chlorobenzoyl )-3- pyridyl]-3-methyl-isoxazol-5-yl]methyl methanesulfonate (15.5 g, crude) ) was used into the next step without further purification. The product was a yellow oil. [0535] Step 12: Preparation of tert-butyl N-[5-[5-(azidomethyl)-3-methyl-isoxazol-4-yl]- 6-(4-chlorobenzoyl)-2-pyridyl]-N-methyl-carbamate. To a solution of [4-[6-[tert- butoxycarbonyl(methyl)amino]-2-(4-chlorobenzoyl)-3-pyridyl]- 3-methyl-isoxazol-5-yl]methyl methanesulfonate (15.0 g, 28.0 mmol, 1.0 equiv) in DMF (150 mL) was added NaN3 (5.58 g, 85.8 mmol, 3.1 equiv). The mixture was stirred at 25 °C for 12 h. The reaction mixture was diluted with H 2 O (500 mL) and extracted with EA (3 x 300 mL).the organic phase was concentrated to afford crude product, The residue was purified by silica chromatography (PE:EA=30:1-10:1) to afford tert-butyl N-[5-[5-(azidomethyl)-3-methyl-isoxazol-4-yl]-6-(4- chlorobenzoyl)-2-pyridyl]-N-methyl-carbamate (12.0 g, 22.36 mmol, 80% yield, 90% purity) as a yellow oil. [0536] Step 13: Preparation of tert-butyl N-[9-(4-chlorophenyl)-3-methyl-5-oxa-4,8,11- triazatricyclo[8.4.0.02,6]tetradeca-1(10),2(6),3,8,11,13-hex aen-12-yl]-N-methyl-carbamate. To a solution of tert-butyl N-[5-[5-(azidomethyl)-3-methyl-isoxazol-4-yl]-6-(4-chloroben zoyl)- 2-pyridyl]-N-methyl-carbamate (9.70 g, 20.1 mmol, 1.0 equiv) in THF (100 mL) and H 2 O (20 mL) was added PPh3 (15.8 g, 60.1 mmol, 3.0 equiv). The mixture was stirred at 25 °C for 12 h. The residue was diluted with H 2 O (300 mL) and extracted with EA (3 x 200 mL).the organic phase was concentrated to afford crude product. The residue was purified by silica chromatography (PE:EA = 1:0-40:1) to afford tert-butyl N-[9-(4-chlorophenyl)-3-methyl-5-oxa- 4,8,11-triazatricyclo[8.4.0.02,6]tetradeca-1(10),2(6),3,8,11 ,13-hexaen-12-yl]-N-methyl- carbamate (8.00 g, 16.4 mmol, 82% yield, 90% purity) as a yellow oil. [0537] Step 14: Preparation of tert-butyl N-methyl-N-[3-methyl-9-[4-(4,4,5,5- tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl]-5-oxa-4,8,11-tri azatricyclo[8.4.0.02,6] tetradeca-1(10),2(6),3,8,11,13-hexaen-12-yl]carbamate. To a solution of tert-butyl N-[9-(4- chlorophenyl)-3-methyl-5-oxa-4,8,11-triazatricyclo[8.4.0.02, 6]tetradeca-1(10),2(6),3,8,11,13- hexaen-12-yl]-N-methyl-carbamate (4.00 g, 9.11 mmol, 1.0 equiv) and 4,4,5,5-tetramethyl-2- (4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1,3,2-dioxabor olane (5.09 g, 20.1 mmol, 2.2 equiv) in dioxane (60 mL) was added AcOK (2.95 g, 30.1 mmol, 3.3 equiv) and dichloropalladium; tricyclohexylphosphane (874 mg, 1.18 mmol, 0.1 equiv) .The mixture was stirred at 80 °C for 12 h. The residue was diluted with H 2 O (80 mL) and extracted with EA (3 x 80 mL).the organic phase was concentrated to afford crude product. The residue was purified by silica chromatography (PE:EA = 30:1-5:1) to afford tert-butyl N-methyl-N-[3-methyl-9-[4-(4,4,5,5- tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl]-5-oxa-4,8,11-tri azatricyclo[8.4.0.02,6]tetradeca- 1(10),2(6),3,8,11,13-hexaen-12-yl]carbamate (4.50 g, 5.94 mmol, 65% yield, 70% purity) as a yellow oil. [0538] Step 15: Preparation of tert-butyl N-[9-(4-hydroxyphenyl)-3-methyl-5-oxa-4,8,11- triazatricyclo[8.4.0.02,6]tetradeca-1(10),2(6),3,8,11,13-hex aen-12-yl]-N-methyl-carbamate. To a solution of tert-butyl N-methyl-N-[3-methyl-9-[4-(4,4,5,5-tetramethyl-1,3,2-dioxabo rolan- 2-yl)phenyl]-5-oxa-4,8,11-triazatricyclo[8.4.0.02,6]tetradec a-1(10),2(6),3,8,11,13-hexaen-12- yl]carbamate (4.50 g, 8.48 mmol, 1.0 equiv) in THF (60 mL) was added H 2 O 2 (2.05 g, 18.1 mmol, 1.74 mL, 30% purity, 2.1 equiv) and NaOH (4 M, 11 mL, 5.0 equiv). The mixture was stirred at 25 °C for 2 h .The reaction mixture was quenched by addition Na2SO3 (15 mL) at 0 °C. Then the mixture was diluted with H 2 O (50 mL), AcOH (15 mL) and extracted with EA (50 mL x 2), and concentrated under reduced pressure to give a residue. The residue was purified by silica chromatography (PE:EA = 10:1-3:1) to afford tert-butyl N-[9-(4-hydroxyphenyl)-3- methyl-5-oxa-4,8,11-triazatricyclo[8.4.0.02,6]tetradeca-1(10 ),2(6),3,8,11,13-hexaen-12-yl]-N- methyl-carbamate (1.80 g, 3.85 mmol, 45% yield, 90% purity) as a yellow oil. [0539] Step 16: Preparation of tert-butyl 2-[4-[12-[tert-butoxycarbonyl(methyl)amino]- 3-methyl-5-oxa-4,8,11-triazatricyclo[8.4.0.02,6]tetradeca-1( 10),2(6),3,8,11,13-hexaen-9- yl]phenoxy]-6-azaspiro[3.5]nonane-6-carboxylate. To a solution of PPh3 (780 mg, 2.97 mmol, 2.5 equiv) in THF (3 mL) was added DIAD (481 mg, 2.38 mmol, 2.0 equiv) at 0 °C, and then it was stirred for 0.5 h. The resulting solution was added to a solution of tert-butyl N-[9-(4- hydroxyphenyl)-3-methyl-5-oxa-4,8,11-triazatricyclo[8.4.0.02 ,6]tetradeca-1(10),2(6),3,8,11,13- hexaen-12-yl]-N-methyl-carbamate (500 mg, 1.19 mmol, 1.0 equiv) and tert-butyl 2-hydroxy-6- azaspiro[3.5]nonane-6-carboxylate (574 mg, 2.38 mmol, 2.0 equiv) in THF (1 mL), and then it was stirred at 50 °C for 12 h. The reaction mixture was concentrated to afford crude product. The residue was purified by silica chromatography (PE:EA = 10:1-4:1) and prep-HPLC (column: Waters Xbridge C18150*50mm* 10um;mobile phase: [water( NH4HCO3)-ACN];gradient:59%- 89% B over 10 min) to afford tert-butyl 2-[4-[12-[tert-butoxycarbonyl(methyl)amino]-3-methyl- 5-oxa-4,8,11-triazatricyclo[8.4.0.02,6]tetradeca-1(10),2(6), 3,8,11,13-hexaen-9-yl]phenoxy]-6- azaspiro[3.5]nonane-6-carboxylate (450 mg, 699 μmol, 59% yield) as a yellow oil. [0540] Step 17: Preparation of 9-[4-(6-azaspiro[3.5]nonan-2-yloxy)phenyl]-N,3- dimethyl-5-oxa-4,8,11-triazatricyclo[8.4.0.02,6]tetradeca-1( 10),2(6),3,8,11,13-hexaen-12- amine. To a solution of tert-butyl 2-[4-[12-[tert-butoxycarbonyl(methyl)amino]-3-methyl-5-oxa- 4,8,11-triazatricyclo[8.4.0.02,6]tetradeca-1(10),2(6),3,8,11 ,13-hexaen-9-yl]phenoxy]-6- azaspiro[3.5]nonane-6-carboxylate (225 mg, 350 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 to afford crude product, the crude product 9-[4-(6-azaspiro[3.5]nonan-2-yloxy)phenyl]-N,3- dimethyl-5-oxa-4,8,11-triazatricyclo[8.4.0.02,6]tetradeca-1( 10),2(6),3,8,11,13-hexaen-12-amine (195 mg, crude, TFA salt) was used into the next step without further purification, the product was a yellow oil. [0541] Step 18: Preparation of N-[3-(3-chloro-4-cyano-phenoxy)-2,2,4,4-tetramethyl- cyclobutyl]-2-[2-[4-[3-methyl-12-(methylamino)-5-oxa-4,8,11- triazatricyclo[8.4.0.02,6] tetradeca-1(10),2(6),3,8,11,13-hexaen-9-yl]phenoxy]-6-azaspi ro[3.5]nonan-6-yl]pyrimidine- 5-carboxamide (I-231). To a solution of 9-[4-(6-azaspiro[3.5]nonan-2-yloxy)phenyl]-N,3- dimethyl-5-oxa-4,8,11-triazatricyclo[8.4.0.02,6]tetradeca-1( 10),2(6),3,8,11,13-hexaen-12-amine (61 mg, 109 μmol, 1.0 equiv, TFA salt) and 2-chloro-N-[3-(3-chloro-4-cyano-phenoxy)-2,2,4,4- tetramethyl-cyclobutyl]pyrimidine-5-carboxamide (45 mg, 109 μmol, 1.0 equiv) in NMP (2 mL) was added K 2 CO 3 (15 mg, 109 μmol, 1.0 equiv) .The mixture was stirred at 50 °C for 2 h. The reaction mixture was filtered to remove K2CO3 and concentrated to afford crude product. The residue was purified by prep-HPLC(column: Waters xbridge 150*25mm 10um;mobile phase: [water(NH 4 HCO 3 )-ACN];gradient:64%-94% B over 10 min) to afford N-[3-(3-chloro-4-cyano- phenoxy)-2,2,4,4-tetramethyl-cyclobutyl]-2-[2-[4-[3-methyl-1 2-(methylamino)-5-oxa-4,8,11- triazatricyclo[8.4.0.02,6]tetradeca-1(10),2(6),3,8,11,13-hex aen-9-yl]phenoxy]-6-azaspiro[3.5] nonan-6-yl]pyrimidine-5-carboxamide (12 mg, 13.50 μmol, 12% yield, 93% purity) as a yellow solid. 1 H NMR (400 MHz, CD3OD) δ 8.77 - 8.70 (m, 2H), 7.85 - 7.80 (m, 1H), 7.75 - 7.69 (m, 1H), 7.29 (d, J = 8.8 Hz, 2H), 7.15 - 7.10 (m, 1H), 6.98 (dd, J = 2.4, 8.8 Hz, 1H), 6.75 (dd, J = 8.8, 17.8 Hz, 3H), 4.83 - 4.78 (m, 3H), 4.27 (s, 1H), 4.15 - 4.11 (m, 1H), 3.93 (s, 2H), 3.89 - 3.81 (m, 2H), 2.78 - 2.73 (m, 3H), 2.53 - 2.48 (m, 3H), 2.47 - 2.38 (m, 2H), 1.86 - 1.73 (m, 4H), 1.64 - 1.55 (m, 2H), 1.28 (s, 6H), 1.21 (s, 6H). LC-MS: MS (ES + ): RT = 2.445 min, m/z = 826.5 [M+H + ]; LCMS method 25. EXAMPLE 33 – Synthesis of Additional Compounds [0542] The following compounds were synthesized using procedures analogous to those described above: I-53, I-55, I-56, I-57, I-58, I-59, 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- 88, I-89, I-90, I-91, I-92, I-93, I-94, I-95, I-96, I-97, I-98, I-99, I-100, I-101, I-102, I-103, I-104, 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-132, I-133, I-134, I- 135, I-136, I-137, 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-151, I-152, I-153, I-154, I-155, I-156, I-157, I-158, 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- 205, 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-222, I- 223, I-224, I-226, I-232, I-233, and I-234. Physical characterization data for exemplary compounds is provided in Table 4. TABLE 4. Com ound No MW Exact Observed Mass Percent Purity Measured

EXAMPLE 34 – Synthesis of 4-chloro-N-((1r,3r)-3-(3-chloro-4-cyanophenoxy)-2,2,4,4- tetramethylcyclobutyl)-2-(2-(4-((S)-2,3,9-trimethyl-6-(oxazo l-2-ylmethyl)-6H-thieno[3,2- f][1,2,4]triazolo[4,3-a][1,4]diazepin-4-yl)phenoxy)-9-azadis piro[3.1.56.14]dodecan-9- yl)pyrimidine-5-carboxamide (I-672) [0543] Step 1: Preparation of tert-butyl 2-[4-[(9S)-4,5,13-trimethyl-9-(oxazol-2- ylmethyl)-3-thia-1,8,11,12-tetrazatricyclo[8.3.0.02,6]tridec a-2(6),4,7,10,12-pentaen-7- yl]phenoxy]-9-azadispiro[3.1.56.14]dodecane-9-carboxylate. To a solution of PPh3 (323 mg, 1.2 mmol, 2.5 equiv) in THF (2 mL) was added DIAD (200 mg, 986 μmol, 0.2 mL, 2.0 equiv) at 0°C. The mixture was stirred at 25°C for 30 min. And then 4-[(9S)-4,5,13-trimethyl-9-(oxazol-2- ylmethyl)-3-thia-1,8,11,12-tetrazatricyclo[8.3.0.02,6]tridec a-2(6),4,7,10,12-pentaen-7-yl]phenol (200 mg, 493 μmol, 1.0 equiv) and tert-butyl 2-hydroxy-9-azadispiro[3.1.56.14]dodecane-9- carboxylate (277 mg, 986 μmol, 2.0 equiv) was added in THF (2 mL).The mixture was stirred at 50 °C for 8h. The reaction mixture was filtered and concentrated to give a residue. The residue was purified by column chromatography (SiO 2 , Petroleum ether/Ethyl acetate to Dichloromethane/Methanol=1:1 to 10:1) to give Compound tert-butyl 2-[4-[(9S)-4,5,13- trimethyl-9-(oxazol-2-ylmethyl)-3-thia-1,8,11,12-tetrazatric yclo[8.3.0.02,6]trideca-2(6),4, 7,10,12-pentaen-7-yl]phenoxy]-9-azadispiro[3.1.56.14]dodecan e-9-carboxylate (250 mg, 373 μmol, 75% yield) was obtained as a yellow solid. [0544] Step 2: Preparation of (9S)-7-[4-(2-azaspiro[4.5]decan-8-yl)phenyl]-9-(2- methoxyethyl)-4,5,13-trimethyl-3-thia-1,8,11,12-tetrazatricy clo[8.3.0.02,6]trideca- 2(6),4,7,10,12-pentaene. To a solution of tert-butyl 2-[4-[(9S)-4,5,13-trimethyl-9-(oxazol-2- ylmethyl)-3-thia-1,8,11,12-tetrazatricyclo[8.3.0.02,6]tridec a-2(6),4,7,10,12-pentaen-7- yl]phenoxy]-9-azadispiro[3.1.56.14]dodecane-9-carboxylate (250 mg, 373 μmol, 1.0 equiv) in DCM (2 mL) was added TFA (1 mL). The mixture was stirred at 25 °C for 1h. The reaction mixture was concentrated to afford crude product. Then diluted with DCM (3 mL), after added it to NaHCO 3 (20 mL) and extracted with DCM/MeOH (10:1, 10 mL x 3), the organic phase was concentrated to give Compound (9S)-7-[4-(2-azaspiro[4.5]decan-8-yl)phenyl]-9-(2-methoxy ethyl)-4,5,13-trimethyl-3-thia-1,8,11,12-tetrazatricyclo[8.3 .0.02,6]trideca-2(6),4,7,10,12- pentaene (120 mg, 238. μmol) as a yellow solid. [0545] Step 3: Preparation of 2,4-dichloro-N-[3-(3-chloro-4-cyano-phenoxy)-2,2,4,4- tetramethyl-cyclobutyl]pyrimidine-5-carboxamide. To a solution of 4-(3-amino-2,2,4,4- tetramethyl-cyclobutoxy)-2-chloro-benzonitrile (700 mg, 1.78 mmol, 1.0 equiv, TFA salt) in DCM (3 mL) was added TEA (1.80 g, 17.8 mmol, 2.5 mL, 10.0 equiv) and 2,4-dichloro- pyrimidine-5-carbonyl chloride (565 mg, 2.67 mmol, 1.5 equiv). The mixture was stirred at 25 °C for 1 h. The reaction mixture was filtered and concentrated to give a residue. The residue was purified by column chromatography (SiO 2 , Petroleum ether/Ethyl acetate=1:0 to 3:1) to give Compound 2,4-dichloro-N-[3-(3-chloro-4-cyano-phenoxy)-2,2,4,4-tetrame thyl-cyclobutyl] pyrimidine-5-carboxamide (280 mg, 617 μmol, 34% yield) as a white solid. [0546] Step 4: Preparation of 4-chloro-N-((1r,3r)-3-(3-chloro-4-cyanophenoxy)-2,2,4,4- tetramethylcyclobutyl)-2-(2-(4-((S)-2,3,9-trimethyl-6-(oxazo l-2-ylmethyl)-6H-thieno[3,2- f][1,2,4]triazolo[4,3-a][1,4]diazepin-4-yl)phenoxy)-9-azadis piro[3.1.56.14]dodecan-9- yl)pyrimidine-5-carboxamide (I-672). To a solution of 2,4-dichloro-N-[3-(3-chloro-4-cyano- phenoxy)-2,2,4,4-tetramethyl-cyclobutyl]pyrimidine-5-carboxa mide (50 mg, 110 μmol, 1.0 equiv) and 2-[[(9S)-7-[4-(9-azadispiro[3.1.56.14]dodecan-2-yloxy)phenyl ]-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]methyl]oxazole (70 mg, 123 μmol, 1.1 equiv) in NMP (0.5 mL) was added DIEA (42 mg, 330 μmol, 0.1 mL, 3.0 equiv). The mixture was stirred at 25 °C for 8h. The reaction mixture was filtered and concentrated to give a residue. The residue was purified by prep-HPLC (column: Phenomenex luna C18150*25mm* 10um; mobile phase: [water(FA)-ACN];gradient:70%-100% B over 10 min) to give Compound 4- chloro-N-((1r,3r)-3-(3-chloro-4-cyanophenoxy)-2,2,4,4-tetram ethylcyclobutyl)-2-(2-(4-((S)- 2,3,9-trimethyl-6-(oxazol-2-ylmethyl)-6H-thieno[3,2-f][1,2,4 ]triazolo[4,3-a][1,4]diazepin-4- yl)phenoxy)-9-azadispiro[3.1.56.14]dodecan-9-yl)pyrimidine-5 -carboxamide (24.68 mg, 25.03 μmol, 22% yield, 97.77% purity) as a off-white solid. 1 H NMR (400 MHz, CD3OD) δ 8.06 (s, 1H), 7.91 (d, J = 0.9 Hz, 1H), 7.74 (d, J = 8.7 Hz, 1H), 7.31 (d, J = 8.7 Hz, 2H), 7.19 - 7.11 (m, 2H), 7.00 (dd, J = 2.4, 8.8 Hz, 1H), 6.81 (d, J = 8.9 Hz, 2H), 4.27 (s, 1H), 4.15 (s, 1H), 4.03 - 3.92 (m, 2H), 3.63 (d, J = 7.1 Hz, 2H), 3.56 - 3.56 (m, 1H), 3.61 - 3.50 (m, 3H), 2.72 (s, 3H), 2.66 - 2.57 (m, 2H), 2.47 (s, 3H), 2.22 - 2.14 (m, 2H), 2.08 (s, 2H), 1.99 (s, 2H), 1.71 (s, 3H), 1.64 (br s, 4H), 1.30 (s, 6H), 1.23 - 1.19 (m, 8H). LC-MS: MS (ES + ): RT = 2.858 min, m/z = 985.6 [M+H + ].

EXAMPLE 35 – Synthesis of N-[3-(3-chloro-4-cyano-phenoxy)-2,2,4,4-tetramethyl- cyclobutyl]-4-methyl-2-[2-[4-[(9S)-4,5,13-trimethyl-9-(oxazo l-2-ylmethyl)-3-thia-1,8,11,12- tetrazatricyclo[8.3.0.02,6]trideca-2(6),4,7,10,12-pentaen-7- yl]phenoxy]-9-azadispiro [3.1.56.14]dodecan-9-yl]pyrimidine-5-carboxamide (I-673)

[0547] Step 1: Preparation of tert-butyl N-[3-(3-chloro-4-cyano-phenoxy)-2,2,4,4- tetramethyl-cyclobutyl]carbamate. To a solution of tert-butyl N-(3-hydroxy-2,2,4,4- tetramethyl-cyclobutyl)carbamate (840 mg, 3.45 mmol, 1.0 equiv) in DMF (15 mL) was added dropwise NaH (207 mg, 5.18 mmol, 60% purity, 1.5 equiv) at 0 °C . After addition, the mixture was stirred at this temperature for 1.0 h, and then 2-chloro-4-fluoro-benzonitrile (537 mg, 3.45 mmol, 1.0 equiv) was added dropwise at 0 °C. The resulting mixture was stirred at 25 °C for 1h. The reaction mixture was quenched by addition NH 4 Cl 20mL at 0 °C, and then diluted with Ethyl acetate 20 mL and extracted with Ethyl acetate (10 mL × 2). The residue was purified by flash silica gel chromatography (ISCO®; 12g SepaFlash® Silica Flash Column, Eluent of 0~10% Ethyl acetate/Petroleum ethergradient) to afford tert-butyl N-[3-(3-chloro-4-cyano- phenoxy)-2,2,4,4-tetramethyl-cyclobutyl]carbamate (1.19 g, 3.14 mmol, 91% yield) as a white solid. [0548] Step 2: Preparation of 4-(3-amino-2,2,4,4-tetramethyl-cyclobutoxy)-2-chloro- benzonitrile. To a solution of tert-butyl N-[3-(3-chloro-4-cyano-phenoxy)-2,2,4,4-tetramethyl- cyclobutyl]carbamate (1.19 g, 3.14 mmol, 1.0 equiv) in DCM (9.0 mL) was added TFA (3.79 g, 33.3 mmol, 2.5 mL, 10.6 equiv). The mixture was stirred at 25 °C for 2 h. The reaction mixture was concentrated under reduced pressure to remove solvent to give 4-(3-amino-2,2,4,4- tetramethyl-cyclobutoxy)-2-chloro-benzonitrile (1.15 g, 2.9 mmol, 92% yield, 99% purity, TFA) as a white solid. [0549] Step 3: Preparation of tert-butyl 2-[4-[(9S)-4,5,13-trimethyl-9-(oxazol-2- ylmethyl)-3-thia-1,8,11,12-tetrazatricyclo[8.3.0.02,6]tridec a-2(6),4,7,10,12-pentaen-7- yl]phenoxy]-9-azadispiro[3.1.56.14]dodecane-9-carboxylate. To a solution of PPh3 (323 mg, 1.2 mmol, 2.5 equiv) in THF (2 mL) was added DIAD (200 mg, 986 μmol, 0.2 mL, 2.0 equiv) at 0°C. The mixture was stirred at 25°C for 30 min. And then 4-[(9S)-4,5,13-trimethyl-9-(oxazol-2- ylmethyl)-3-thia-1,8,11,12-tetrazatricyclo[8.3.0.02,6]tridec a-2(6),4,7,10,12-pentaen-7-yl]phenol (200 mg, 493 μmol, 1.0 equiv) and tert-butyl 2-hydroxy-9-azadispiro[3.1.56.14]dodecane-9- carboxylate (277 mg, 986 μmol, 2.0 equiv) was added in THF (2 mL).The mixture was stirred at 50 °C for 8h. The reaction mixture was filtered and concentrated to give a residue. The residue was purified by column chromatography (SiO 2 , Petroleum ether/Ethyl acetate to Dichloromethane/Methanol=1:1 to 10:1) to give Compound tert-butyl 2-[4-[(9S)-4,5,13- trimethyl-9-(oxazol-2-ylmethyl)-3-thia-1,8,11,12-tetrazatric yclo[8.3.0.02,6]trideca- 2(6),4,7,10,12-pentaen-7-yl]phenoxy]-9-azadispiro[3.1.56.14] dodecane-9-carboxylate (250 mg, 373 μmol, 75% yield) was obtained as a yellow solid. [0550] Step 4: Preparation of (9S)-7-[4-(2-azaspiro[4.5]decan-8-yl)phenyl]-9-(2- methoxyethyl)-4,5,13-trimethyl-3-thia-1,8,11,12-tetrazatricy clo[8.3.0.02,6]trideca- 2(6),4,7,10,12-pentaene. To a solution of tert-butyl 2-[4-[(9S)-4,5,13-trimethyl-9-(oxazol-2- ylmethyl)-3-thia-1,8,11,12-tetrazatricyclo[8.3.0.02,6]tridec a-2(6),4,7,10,12-pentaen-7-yl] phenoxy]-9-azadispiro[3.1.56.14]dodecane-9-carboxylate (250 mg, 373 μmol, 1.0 equiv) in DCM (2 mL) was added TFA (1 mL). The mixture was stirred at 25 °C for 1h. The reaction mixture was concentrated to afford crude product. Then diluted with DCM (3 mL), after added it to NaHCO 3 (20 mL) and extracted with DCM/MeOH (10:1, 10 mL x 3), the organic phase was concentrated to give Compound (9S)-7-[4-(2-azaspiro[4.5]decan-8-yl)phenyl]-9-(2- methoxyethyl)-4,5,13-trimethyl-3-thia-1,8,11,12-tetrazatricy clo[8.3.0.02,6]trideca- 2(6),4,7,10,12-pentaene (120 mg, 238. μmol) as a yellow solid. [0551] Step 5: Preparation of 2-chloro-N-[3-(3-chloro-4-cyano-phenoxy)-2,2,4,4- tetramethyl-cyclobutyl]-4-methyl-pyrimidine-5-carboxamide. To a solution of 4-(3-amino- 2,2,4,4-tetramethyl-cyclobutoxy)-2-chloro-benzonitrile (228 mg, 579 μmol, 1.0 equiv, TFA salt) in DMF (2.0 mL) was added TEA (293 mg, 2.90 mmol, 403 μL, 5.0 equiv) and 2-chloro-4- methyl-pyrimidine-5-carboxylic acid (100 mg, 579 μmol, 1.0 equiv), followed by T 4 P (1.25 g, 1.74 mmol, 50% purity, 3.0 equiv) was added. The reaction mixture was stirred at 25°C for 1 h. The residue was purified by prep-HPLC (column: Phenomenex luna C18150*25mm* 10um;mobile phase: [water(FA)-ACN]; gradient:56%-76% B over 10 min). Compound 2-chloro- N-[3-(3-chloro-4-cyano-phenoxy)-2,2,4,4-tetramethyl-cyclobut yl]-4-methyl-pyrimidine-5- carboxamide (100 mg, 231 μmol, 40% yield) was obtained as a white solid. [0552] Step 6: Preparation of N-[3-(3-chloro-4-cyano-phenoxy)-2,2,4,4-tetramethyl- cyclobutyl]-4-methyl-2-[2-[4-[(9S)-4,5,13-trimethyl-9-(oxazo l-2-ylmethyl)-3-thia-1,8,11,12- tetrazatricyclo[8.3.0.02,6]trideca-2(6),4,7,10,12-pentaen-7- yl]phenoxy]-9- azadispiro[3.1.56.14]dodecan-9-yl]pyrimidine-5-carboxamide (I-673). To a solution of 2- chloro-N-[3-(3-chloro-4-cyano-phenoxy)-2,2,4,4-tetramethyl-c yclobutyl]-4-methyl-pyrimidine- 5-carboxamide (53.3 mg, 123 μmol, 1.0 equiv) and 2-[[(9S)-7-[4-(9-azadispiro[3.1.5 6 .1 4 ] dodecan-2-yloxy)phenyl]-4,5,13-trimethyl-3-thia-1,8,11,12-te trazatricyclo[8.3.0.0 2,6 ]trideca- 2(6),4,7,10,12-pentaen-9-yl]methyl]oxazole (70.0 mg, 123 μmol, 1.0 equiv) in NMP (0.5 mL) was added DIEA (15.9 mg, 123 μmol, 21.4 μL, 1.0 equiv). The reaction mixture was stirred at 25 °C for 12 h. The residue was purified by prep-HPLC (column: Phenomenex luna C18 150*25mm* 10um; mobile phase: [water (FA)-ACN]; gradient:80%-100% B over 10 min). Compound N-[3-(3-chloro-4-cyano-phenoxy)-2,2,4,4-tetramethyl-cyclobut yl]-4-methyl-2-[2-[4- [(9S)-4,5,13-trimethyl-9-(oxazol-2-ylmethyl)-3-thia-1,8,11,1 2-tetrazatricyclo[8.3.0.0 2,6 ]trideca- 2(6),4,7,10,12-pentaen-7-yl]phenoxy]-9-azadispiro[3.1.5 6 .1 4 ]dodecan-9-yl]pyrimidine-5- carboxamide (33.7 mg, 33.9 μmol, 28% yield, 97% purity) was obtained as an off-white solid. 1 H NMR (400 MHz, CD3OD) δ 8.31 (s, 1H), 7.90 (s, 1H), 7.72 (d, J = 8.8 Hz, 1H), 7.30 (d, J = 8.7 Hz, 2H), 7.12 (s, 2H), 6.99 - 6.96 (m, 1H), 6.80 (d, J = 8.9 Hz, 2H), 4.91 - 4.90 (m, 7H), 4.82 - 4.81 (m, 1H), 4.74 (d, J = 6.5 Hz, 1H), 4.65 (s, 1H), 4.58 (s, 16H), 4.24 (s, 1H), 4.10 (s, 1H), 3.98 - 3.94 (m, 2H), 3.79 (br d, J = 5.5 Hz, 4H), 2.70 (s, 3H), 2.61 (br s, 2H), 2.45 (d, J = 4.0 Hz, 6H), 2.18 (br d, J = 6.6 Hz, 2H), 2.06 (s, 2H), 1.97 (s, 2H), 1.70 (s, 3H), 1.61 - 1.53 (m, 4H), 1.31 - 1.16 (m, 14H). LC-MS: MS (ES + ): RT = 2.992 min, m/z = 965.6 [M +H + ].

EXAMPLE 36 – Synthesis of N-[3-(3-chloro-4-cyano-phenoxy)-2,2,4,4-tetramethyl- cyclobutyl]-2-[8-[4-[(9S)-4,5,13-trimethyl-9-(oxazol-2-ylmet hyl)-3-thia-1,8,11,12- tetrazatricyclo[8.3.0.02,6]trideca-2(6),4,7,10,12-pentaen-7- yl]phenyl]-2-azaspiro[4.5]decan- 2-yl]pyrimidine-5-carboxamide (I-671) [055 ep : epaa o o e - uy - y oy--aasp o . eca e-- carboxylate. To a solution of tert-butyl 8-oxo-2-azaspiro[4.5]decane-2-carboxylate (2 g, 7.8 mmol, 1 equiv) in DCM (20 mL) was added NaBH4 (400 mg, 10 mmol, 1.3 equiv). The mixture was stirred at 20 °C for 12 h. The reaction mixture was filtered and concentrated under reduced pressure to give a residue.After filtration, the filtrate was concentrated to afford crude product. [0554] Step 2: Preparation of tert-butyl 8-iodo-2-azaspiro[4.5]decane-2-carboxylate. To a solution of tert-butyl 8-hydroxy-2-azaspiro[4.5]decane-2-carboxylate (2 g, 7.8 mmol, 1 equiv) in DCM (20 mL) was added PPh3 (2.8 g, 10 mmol, 1.4 equiv) and imidazole (1.6 g, 23 mmol, 3 equiv) and I 2 (2.9 g, 11 mmol, 2 mL, 1.5 equiv). The mixture was stirred at 40 °C for 12 h. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. Then purified by prep-TLC (SiO 2 , EA: PE = 5: 1) to afford compound tert-butyl 8-iodo-2- azaspiro[4.5]decane-2-carboxylate (2 g, 69 % yield) as a white oil. [0555] Step 3: Preparation of (2-tert-butoxycarbonyl-2-azaspiro[4.5]decan-8-yl)-iodo- zinc. To a stirring solution of Zn (830 mg, 12 mmol, 5.8 equiv) in DMAC (2 mL) was added TMSCl (88 mg, 810 μmol, 102 μL, 0.37 equiv) and 1,2-dibromoethane (152 mg, 810 μmol, 61 μL, 0.37 equiv) in DMAC (1 mL) at 40 °C. After stirring for 30 min, tert-butyl 8-iodo- 2-azaspiro[4.5]decane-2-carboxylate (800 mg, 2 mmol, 1 equiv) was added, The mixture was stirred at 40 °C for another 1 h, after filtration, the filtrate was concentrated to afford crude product. [0556] Step 4: Preparation of racemic tert-butyl (S)-8-(4-(2,3,9-trimethyl-6-(oxazol-2- ylmethyl)-6H-thieno[3,2-f][1,2,4]triazolo[4,3-a][1,4]diazepi n-4-yl)phenyl)-2-azaspiro[4.5] decane-2-carboxylate. To a solution of (2-tert-butoxycarbonyl-2-azaspiro[4.5]decan-8-yl)- iodo-zinc (3.50 g, 8 mmol, 3.5 equiv), (S)-2-((4-(4-chlorophenyl)-2,3,9-trimethyl-6H-thieno[3,2- f][1,2,4]triazolo[4,3-a][1,4]diazepin-6-yl)methyl)oxazole (984 mg, 2 mmol, 1 equiv) in THF (10 mL) was added dicyclohexyl-[2-(2,6-dimethoxyphenyl)phenyl]phosphane;methan esulfonate;(2- phenylanilino)palladium(1+) (362 mg, 464 μmol, 0.2 equiv). The mixture was stirred at 70 °C for 3 h . The reaction mixture was partitioned between EA 20 mL. The organic phase was separated, washed with H 2 O 5 mL (5 mL * 3), 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 * 25 mm* 10 um; mobile phase: [water ( FA )- ACN]; gradient: 63 %-93 % B over 10 min ) to give racemic tert-butyl (S)-8-(4-(2,3,9-trimethyl-6- (oxazol-2-ylmethyl)-6H-thieno[3,2-f][1,2,4]triazolo[4,3-a][1 ,4]diazepin-4-yl)phenyl)-2- azaspiro[4.5]decane-2-carboxylate (1 g, 68.70% yield). [0557] Step 5: Preparation of enantiopure tert-butyl (S)-8-(4-(2,3,9-trimethyl-6-(oxazol- 2-ylmethyl)-6H-thieno[3,2-f][1,2,4]triazolo[4,3-a][1,4]diaze pin-4-yl)phenyl)-2-azaspiro[4.5] decane-2-carboxylate. The racemic tert-butyl (S)-8-(4-(2,3,9-trimethyl-6-(oxazol-2-ylmethyl)- 6H-thieno[3,2-f][1,2,4]triazolo[4,3-a][1,4]diazepin-4-yl)phe nyl)-2-azaspiro[4.5]decane-2- carboxylate was by SFC. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. The residue was purified by SFC (column: DAICEL CHIRALPAK IK (250 mm* 30 mm, 10 um); mobile phase: [CO 2 - ACN/ EtOH (0.1 % NH3H 2 O)]; B %:55 %, isocratic elution mode) and was purified by SFC (column: DAICEL CHIRALPAK AD (250 mm* 30 mm, 10 um); mobile phase: [CO 2 –CAN/ i-PrOH (0.1 % NH 3 H 2 O)]; B %: 60 %, isocratic elution mode) to give enantiopure tert-butyl (S)-8-(4-(2,3,9-trimethyl-6-(oxazol-2- ylmethyl)-6H-thieno[3,2-f][1,2,4]triazolo[4,3-a][1,4]diazepi n-4-yl)phenyl)-2- azaspiro[4.5]decane-2-carboxylate. [0558] Step 6: Preparation of N-[3-(3-chloro-4-cyano-phenoxy)-2,2,4,4-tetramethyl- cyclobutyl]-2-[8-[4-[(9S)-4,5,13-trimethyl-9-(oxazol-2-ylmet hyl)-3-thia-1,8,11,12- tetrazatricyclo[8.3.0.02,6]trideca-2(6),4,7,10,12-pentaen-7- yl]phenyl]-2-azaspiro[4.5]decan- 2-yl]pyrimidine-5-carboxamide. To a solution of 2-chloro-N-[3-(3-chloro-4-cyano-phenoxy)- 2,2,4,4-tetramethyl-cyclobutyl]pyrimidine-5-carboxamide (61 mg, 146 μmol, 1.1 equiv) in NMP (0.5 mL) was added DIEA (51 mg, 398 μmol, 69 μL, 3 equiv) and2-[[(9S)-7-[4-(2- azaspiro[4.5]decan-8-yl)phenyl]-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]methyl]oxazole (70 mg, 132 μmol, 1 equiv). The mixture was stirred at 20 °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*25 mm* 10 um; mobile phase: [water (FA)-ACN]; gradient: 68 % - 98 % B over 10 min) to give N-[3-(3-chloro-4-cyano-phenoxy)-2,2,4,4-tetramethyl-cyclobut yl]-2-[8-[4-[(9S)- 4,5,13-trimethyl-9-(oxazol-2-ylmethyl)-3-thia-1,8,11,12-tetr azatricyclo[8.3.0.02,6]trideca- 2(6),4,7,10,12-pentaen-7-yl]phenyl]-2-azaspiro[4.5]decan-2-y l]pyrimidine-5-carboxamide (52 mg, 43 % yield) as a yellow solid. 1 H NMR (400 MHz, MeOD) δ = 8.78 (s, 2H), 7.92 (d, J = 0.8 Hz, 1H), 7.74 (d, J = 8.8 Hz, 1H), 7.33 (q, J = 8.4 Hz, 4H), 7.15 (s, 2H), 7.07-6.94 (m, 1H), 4.78 (s, 1H), 4.29 (s, 1H), 4.16 (s, 1H), 4.07-3.93 (m, 2H), 3.71 (t, J = 7.2 Hz, 2H), 3.66 (s, 2H), 2.73 (s, 3H), 2.69-2.62 (m, 1H), 2.47 (s, 3H), 1.95-1.80 (m, 6H), 1.68 (s, 3H), 1.66-1.57 (m, 4H), 1.31 (s, 6H), 1.23 (s, 6H). LC-MS: MS (ES + ): RT = 2.881 min, m/z = 909.6 [M + H + ]; LCMS Method: 25. EXAMPLE 37 – Synthesis of N-[3-(3-chloro-4-cyano-phenoxy)-2,2,4,4-tetramethyl- cyclobutyl]-2-[2-[1-methyl-1-[4-[(9S)-4,5,9,13-tetramethyl-3 -thia-1,8,11,12-tetraza- tricyclo[8.3.0.02,6]trideca-2(6),4,7,10,12-pentaen-7-yl]phen yl]ethyl]-2,8-diazaspiro [3.5]nonan-8-yl]pyrimidine-5-carboxamide (I-456)

[0559] Step 1: Preparation of [(2R)-2-(tert-butoxycarbonylamino)-3-methoxy-3-oxo- propyl]-iodo-zinc. To a solution of tert-butyl N-[2-(methylamino)ethyl]carbamate (5.00 g, 28.7 mmol, 1.0 equivuiv) and 1- A 100 mL oven-dried Schlenk tube equivuipped with a magnetic stirrer bar was charged with Zn (60.2 g, 921 mmol, 6.0 equiv). The tube was evacuated and backfilled with argon for three times. Dry DMF (75 mL) was then added to the tube via a syringe.1,2-DIBROMOETHANE (17.3 g, 92.1 mmol, 6.95 mL, 0.60 equiv) was added next to the stirred suspension. The reaction mixtures were then stirred at 60 °C in an oil bath for 45 min. The mixture was cooled to 25°C. Chlorotrimethyl silane (3.33 g, 30.7 mmol, 3.89 mL, 0.20 equiv) was added via a syringe to the slurry, which is stirred for 45 min at 25 °C. A solution of methyl (2R)-2-(tert-butoxycarbonylamino)-3-iodo-propanoate (50.5 g, 153 mmol, 1.0 equiv) in dry DMF (75 mL) was added via a syringe to the mixture of activated zinc at 25 °C, which is then stirred at 35 °C in an oil bath for 60 min. The crude product was used into the next step without further purification. Compound [(2R)-2-(tert-butoxycarbonylamino)-3-methoxy-3-oxo- propyl]-iodo-zinc (60.0 g, 152 mmol, 99% yield) was obtained as a colorless oil. [0560] Step 2: Preparation of 2-bromooxazole. To a solution of oxazole (20.0 g, 290 mmol, 18.5 mL, 1.0 equiv) in THF (300 mL) was added dropwise n-BuLi (2.5 M, 128 mL, 1.1 equiv) at -78 °C. After addition, the mixture was stirred at -78 °C for 1 h. Then 1,2-dibromo- 1,1,2,2-tetrafluoro-ethane (79.0 g, 304 mmol, 1.1 equiv) was added dropwise at -78 °C. The resulting mixture was stirred at 25 °C for 12 h. The reaction mixture was quenched by addition 10 % NH 4 Cl aq (200 mL) at 0 °C, and then extracted with DCM (200 mL x 3). The combined organic layers were washed with brine 100 mL, dried over Na2SO4, filtered and concentrated under reduced pressure to give a residue. The crude product was distilled in vacuum (22 °C, 0.2 Mpa, oil pump). Compound 2-bromooxazole (19.0 g, 128 mmol, 44% yield) was obtained as a colorless oil. [0561] Step 3: Preparation of methyl (2S)-2-(tert-butoxycarbonylamino)-3-oxazol-2-yl- propanoate. To a solution of 2-bromooxazole (30.0 g, 89.2 mmol, 1.0 equiv), [(2R)-2-(tert- butoxycarbonylamino)-3-methoxy-3-oxopropyl]-iodo-zinc (60.0 g, 152 mmol, 1.7 equiv) in THF (240 mL) was added Xphos (4.25 g, 8.92 mmol, 0.10 equiv) and Pd 2 (dba) 3 (4.08 g, 4.46 mmol, 0.050 equiv) under N2. The mixture was stirred at 60 °C for 12h. The reaction mixture was quenched by addition 10 % NH4Cl aq (20 mL) at 0 °C, and then extracted with EtOAc (200 mL x 3). 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=10/1to3/1). Compound methyl (2S)-2- (tert-butoxycarbonylamino)-3-oxazol-2-yl-propanoate (17.5 g, 64.8 mmol, 73% yield) was obtained as a colorless oil. [0562] Step 4: Preparation of (2S)-2-(tert-butoxycarbonylamino)-3-oxazol-2-yl- propanoic acid. To a solution ofmethyl (2S)-2-(tert-butoxycarbonylamino)-3-oxazol-2-yl- propanoate (17.3 g, 64.0 mmol, 1.0 equiv) in THF (180 mL) and H 2 O (90 mL) was added LiOH·H 2 O (5.37 g, 128 mmol, 2.0 equiv) under N 2 . The mixture was stirred at 25 °C for 12 h. The reaction mixture was quenched by addition water (50 mL) at 0 °C, and then concentrated under reduced pressure to remove THF. Diluted with water (50 mL) and extracted with Ethyl acetate (20 mL x 3). The water layers were adjusted to a ph=4.0 with 1M HCl. then extracted with Ethyl acetate (50mL x 3). The combined organic layers were dried over Na 2 SO 4 , filtered, and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (SiO 2 , Petroleum ether/Ethyl acetate=1/1 to 0/1). Compound (2S)-2-(tert- butoxycarbonylamino)-3-oxazol-2-yl-propanoic acid (16.0 g, 62.4 mmol, 98% yield) was obtained as a yellow oil. [0563] Step 5: Preparation of isopropoxycarbonyl (2S)-2-(tert-butoxycarbonylamino)-3- oxazol-2-yl-propanoate. To a solution of (2S)-2-(tert-butoxycarbonylamino)-3-oxazol-2-yl- propanoic acid (14.0 g, 54.6 mmol, 1.0 equiv) and NMM (16.6 g, 164 mmol, 18.0 mL, 3.0 equiv) in THF (200 mL) was added isobutyl carbonochloridate (9.70 g, 71.0 mmol, 9.29 mL, 1.3 equiv) at 0 °C and stirred at 0 °C for 2 h. The residue was used for the next step without further purification. Compound isopropoxycarbonyl (2S)-2-(tert-butoxycarbonylamino)-3-oxazol-2-yl- propanoate (19 g, crude) was obtained as a white solid. [0564] Step 6: Preparation of tert-butyl 2-(ethanehydrazonoylamino)-4,5-dimethyl- thiophene-3-carboxylate. To a solution of tert-butyl 2-(ethanethioylamino)-4,5-dimethyl- thiophene-3-carboxylate (20.0 g, 70.1 mmol, 1.0 equiv) in THF (250 mL) was added NH 2 NH 2 ·H 2 O (18.6 g, 364 mmol, 18.0 mL, 98% purity, 5.2 equiv) and stirred at 25 °C for 1 h. The mixture was poured into ice-water (400 mL). The aqueous phase was extracted with ethyl acetate (100 mL × 3). The combined organic phase was washed with brine (100 mL × 3), dried with anhydrous sodium sulfate, filtered, and concentrated under vacuum. The residue was used for the next step without further purification. Compound tert-butyl 2-(ethanehydrazonoylamino)- 4,5-dimethyl-thiophene-3-carboxylate (18.0 g, 63.5 mmol, 90% yield) was obtained as a light- yellow solid. [0565] Step 7: Preparation of tert-butyl 2-[[(E)-N-[[(2S)-2-(tert-butoxycarbonylamino)- 3-oxazol-2-yl-propanoyl]amino]-C-methyl-carbonimidoyl]amino] -4,5-dimethyl-thiophene- 3-carboxylate. To a solution of tert-butyl 2-(ethanehydrazonoylamino)-4,5-dimethyl-thiophene- 3-carboxylate (14.0 g, 49.4 mmol, 1.0 equiv) and NMM (9.99 g, 98.8 mmol, 10.9 mL, 2.0 equiv) in THF (150 mL) was added isopropoxycarbonyl (2S)-2-(tert-butoxycarbonylamino)-3-oxazol-2- yl-propanoate (18.6 g, 54.3 mmol, 1.1 equiv) and stirred at 25 °C for 2 h. The mixture was poured into ice-water (500 mL). The aqueous phase was extracted with ethyl acetate (150 mL × 3). The combined organic phase was washed with brine (150 mL × 2), dried with anhydrous sodium sulfate, filtered, and concentrated under vacuum. The residue was used for the next step without further purification. Compound tert-butyl 2-[[(E)-N-[[(2S)-2-(tert-butoxycarbonylamino) -3-oxazol-2-yl-propanoyl]amino]-C-methyl-carbonimidoyl]amino ]-4,5-dimethyl-thiophene-3- carboxylate (25.0 g, crude) was obtained as a light-yellow gum. [0566] Step 8: Preparation of tert-butyl 2-[3-[(1S)-1-(tert-butoxycarbonylamino)-2- oxazol-2-yl-ethyl]-5-methyl-1,2,4-triazol-4-yl]-4,5-dimethyl -thiophene-3-carboxylate. To a solution of tert-butyl 2-[[(E)-N-[[(2S)-2-(tert-butoxycarbonylamino)-3-oxazol-2-yl- propanoyl]amino]-C-methyl-carbonimidoyl]amino]-4,5-dimethyl- thiophene-3-carboxylate (25.0 g, 47.9 mmol, 1.0 equiv) in i-PrOH (200 mL) was added AcOH (14.4 g, 240 mmol, 13.7 mL, 5.0 equiv) and stirred at 80 °C for 3 h. After cooled to RT, the mixture was concentrated to give a residue. The residue was diluted with EtOAc (300 mL), adjusted to pH = 8 with saturated sodium bicarbonate at 0 °C, extracted with ethyl acetate (100 mL x 3). The combined organic phase was washed with brine (100 mL × 2), dried with anhydrous sodium sulfate, filtered, and concentrated under vacuum. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=0/1 to THF/ethyl acetate=1/2). Compound tert-butyl 2-[3-[(1S)-1- (tert-butoxycarbonylamino)-2-oxazol-2-yl-ethyl]-5-methyl-1,2 ,4-triazol-4-yl]-4,5-dimethyl- thiophene-3-carboxylate (18.0 g, crude) was obtained as a light brown gum. [0567] Step 9: Preparation of 2-[3-[(1S)-1-amino-2-oxazol-2-yl-ethyl]-5-methyl-1,2,4- triazol-4-yl]-4,5-dimethyl-thiophene-3-carboxylic acid. To a solution of tert-butyl 2-[3-[(1S)- 1-(tert-butoxycarbonylamino)-2-oxazol-2-yl-ethyl]-5-methyl-1 ,2,4-triazol-4-yl]-4,5-dimethyl- thiophene-3-carboxylate (18.0 g, 35.7 mmol, 1.0 equiv) in DCM (50 mL) was added TFA (92.1 g, 808 mmol, 60.0 mL, 22.6 equiv) and stirred at 45 °C for 3 h. The mixture was concentrated to give a residue. The residue was used for the next step without further purification. Compound 2- [3-[(1S)-1-amino-2-oxazol-2-yl-ethyl]-5-methyl-1,2,4-triazol -4-yl]-4,5-dimethyl-thiophene-3- carboxylic acid (12.4 g, crude) was obtained as a brown gum. [0568] Step 10: Preparation of (9S)-4,5,13-trimethyl-9-(oxazol-2-ylmethyl)-3-thia- 1,8,11,12-tetrazatricyclo[8.3.0.02,6]trideca-2(6),4,10,12-te traen-7-one. To a solution of 2-[3- [(1S)-1-amino-2-oxazol-2-yl-ethyl]-5-methyl-1,2,4-triazol-4- yl]-4,5-dimethyl-thiophene-3- carboxylic acid (12.4 g, 26.9 mmol, 1.0 equiv, TFA) in DCM (150 mL) was added DIEA (17.4 g, 134 mmol, 23.4 mL, 5.0 equiv) and HATU (20.4 g, 53.7 mmol, 2.0 equiv) and stirred at 25 °C for 12 h. The mixture was poured into ice-water (300 mL). The aqueous phase was extracted with DCM (80 mL × 3). The combined organic phase was washed with brine (100 mL × 2), dried with anhydrous sodium sulfate, filtered, and concentrated under vacuum. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=0/1 to THF/ethyl acetate =1/5 to 1/3). Compound (9S)-4,5,13-trimethyl-9-(oxazol-2-ylmethyl)-3-thia-1,8,11,12 - tetrazatricyclo[8.3.0.02,6]trideca-2(6),4,10,12-tetraen-7-on e (13.0 g, crude) was obtained as a light yellow solid. [0569] Step 11: Preparation of 2-[[(9S)-7-chloro-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]methyl]oxazole. To a solution of (9S)-4,5,13-trimethyl-9-(oxazol-2-ylmethyl)-3-thia-1,8,11,12 -tetrazatricyclo[8.3.0.02,6] trideca-2(6),4,10,12-tetraen-7-one (13.0 g, 39.5 mmol, 1.0 equiv) in CHCl3 (65 mL) was added PCl 5 (12.3 g, 59.2 mmol, 1.5 equiv) at 25 °C and stirred at 70 °C for 3.5 h. After cooled to RT, the reaction mixture was slowly poured into ice cold sat. NaHCO3 (250 mL) slowly and then extracted with DCM (60 mL x 3). The combined organic layers were dried, filtered and concentrated under reduced pressure to give a residue. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate=0/1 to THF/ethyl acetate =1/5 to 1/3). Compound 2-[[(9S)-7-chloro-4,5,13-trimethyl-3-thia-1,8,11,12-tetrazat ricyclo[8.3.0.02,6]trideca- 2(6),4,7,10,12-pentaen-9-yl]methyl]oxazole (3.50 g, 8.25 mmol, 21% yield) was obtained as a light yellow solid. [0570] Step 12: Preparation of tert-butyl 2-(4-bromo-2-fluoro-phenoxy)-7-azaspiro [3.5]nonane-7-carboxylate. A mixture of 4-bromo-2-fluoro-phenol (500 mg, 2.6 mmol, 1.0 equiv), tert-butyl 2-(p-tolylsulfonyloxy)-7-azaspiro[3.5]nonane-7-carboxylate (1.0 g, 2.6 mmol, 1.0 equiv), Cs2CO3 (1.7 g, 5.2 mmol, 2.0 equiv) in DMF (5 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 100 °C for 12 h under N2 atmosphere. The reaction mixture was partitioned between ethyl acetate 300 mL and water 200 mL. The organic phase was separated, washed with brine 200 mL (100 mL x 2), dried over Na2SO4, filtered, and concentrated under reduced pressure to give a residue. The crude product tert-butyl 2-(4-bromo- 2-fluoro-phenoxy)-7-azaspiro[3.5]nonane-7-carboxylate (1.0 g, 2.4 mmol, 92% yield) was used into the next step without further purification. [0571] Step 13: Preparation of tert-butyl 2-[2-fluoro-4-(4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2-yl)phenoxy]-7-azaspiro[3.5]nonane-7-carboxyla te. A mixture of tert-butyl 2- (4-bromo-2-fluoro-phenoxy)-7-azaspiro[3.5]nonane-7-carboxyla te (1.0 g, 2.4 mmol, 1.0 equiv), BPD (1.2 g, 4.8 mmol, 2.0 equiv), Pd(dppf)Cl 2 (177 mg, 241 μmol, 0.1 equiv), AcOK (474 mg, 4.8 mmol, 2.0 equiv) in dioxane (10 mL) was degassed and purged with N 2 for 3 times, and then the mixture was stirred at 90 °C for 12 h under N2 atmosphere. The reaction mixture was partitioned between ethyl acetate 300 mL and water 200 mL. The organic phase was separated, washed with brine 300 mL (150 mL x 2), 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 x 40mm x 15um; mobile phase: [water (FA) – CAN]; gradient : 70%- 100% B over 15 min) to give the product tert-butyl 2-[2-fluoro-4-(4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2-yl)phenoxy]-7-azaspiro[3.5]nonane-7-carboxyla te (400 mg, 867 μmol, 35% yield) obtained as a white solid. [0572] Step 14: Preparation of tert-butyl 2-[2-fluoro-4-[(9S)-4,5,13-trimethyl-9-(oxazol- 2-ylmethyl)-3-thia-1,8,11,12-tetrazatricyclo[8.3.0.02,6]trid eca-2(6),4,7,10,12-pentaen-7- yl]phenoxy]-7-azaspiro[3.5]nonane-7-carboxylate. A mixture of tert-butyl 2-[2-fluoro-4- (4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenoxy]-7-azas piro[3.5]nonane-7-carboxylate (144 mg, 312 μmol, 1.3 equiv), 2-[[(9S)-7-chloro-4,5,13-trimethyl-3-thia-1,8,11,12-tetraza- tricyclo[8.3.0.02,6]trideca-2(6),4,7,10,12-pentaen-9-yl]meth yl]oxazole (83 mg, 239 μmol, 1.0 equiv), Pd 2 (dba) 3 (11 mg, 12 μmol, 0.1 equiv), PCy 3 (7 mg, 24 μmol, 8 μL, 0.1 equiv) and K 3 PO 4 (153 mg, 719 μmol, 3.0 equiv) in dioxane (2 mL), H 2 O (0.4 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 100 °C for 3 h under N2 atmosphere. The reaction mixture was partitioned between ethyl acetate 200 mL and water 150 mL. The organic phase was separated, washed with brine 200 mL (100 mL x 2), 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 x 25mm x 10um; mobile phase: [ water (FA) – CAN ]; gradient: 69%-99% B over 10 min) to give the tert-butyl 2-[2-fluoro-4-[(9S)-4,5,13-trimethyl-9- (oxazol-2-ylmethyl)-3-thia-1,8,11,12-tetrazatricyclo[8.3.0.0 2,6]trideca-2(6),4,7,10,12-pentaen-7- yl]phenoxy]-7-azaspiro[3.5]nonane-7-carboxylate (145 mg, 224 μmol, 93% yield) obtained as a white solid. [0573] Step 15: Preparation of 2-[[(9S)-7-[4-(7-azaspiro[3.5]nonan-2-yloxy)-3-fluoro- phenyl]-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]methyl]oxazole. A mixture of tert-butyl 2-[2-fluoro-4-[(9S)-4,5,13-trimethyl-9- (oxazol-2-ylmethyl)-3-thia-1,8,11,12-tetrazatricyclo[8.3.0.0 2,6]trideca-2(6),4,7,10,12-pentaen-7- yl]phenoxy]-7-azaspiro[3.5]nonane-7-carboxylate (145 mg, 224 μmol, 1.0 equiv) in DCM (3 mL), TFA (1 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 25 °C for 1 h under N2 atmosphere. The reaction mixture was concentrated under reduced pressure to remove solvent. The crude product 2-[[(9S)-7-[4-(7-azaspiro[3.5]nonan-2-yloxy)-3- fluoro-phenyl]-4,5,13-trimethyl-3-thia-1,8,11,12-tetrazatric yclo[8.3.0.02,6]trideca-2(6) ,4,7,10,12-pentaen-9-yl]methyl]oxazole (120 mg, 219 μmol, 97 % yield) was used into the next step without further purification. [0574] Step 16: Preparation of N-[3-(3-chloro-4-cyano-phenoxy)-2,2,4,4-tetramethyl- cyclobutyl]-2-[2-[1-methyl-1-[4-[(9S)-4,5,9,13-tetramethyl-3 -thia-1,8,11,12-tetraza- tricyclo[8.3.0.02,6]trideca-2(6),4,7,10,12-pentaen-7-yl]phen yl]ethyl]-2,8-diazaspiro [3.5]nonan-8-yl]pyrimidine-5-carboxamide (I-456). A mixture of 2-[[(9S)-7-[4-(7- azaspiro[3.5]nonan-2-yloxy)-3-fluoro-phenyl]-4,5,13-trimethy l-3-thia-1,8,11,12- tetrazatricyclo[8.3.0.02,6]trideca-2(6),4,7,10,12-pentaen-9- yl]methyl]oxazole (120 mg, 219 μmol, 1.0 equiv), 2-chloro-N-[3-(3-chloro-4-cyano-phenoxy)-2,2,4,4-tetramethyl -cyclobutyl] pyrimidine-5-carboxamide (92 mg, 219 μmol, 1.0 equiv), DIEA (28 mg, 219 μmol, 38 μL, 1.0 equiv) in NMP (2 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 25 °C for 12 h under N 2 atmosphere. The residue was purified by prep-HPLC (column: Waters x bridge 150 x 25mm 10um; mobile phase: [water (NH4HCO3) -ACN]; gradient: 70%- 100% B over 10 min). The residue was separated by SFC (column: REGIS(S, S)WHELK-O1 (250mm x 25mm,10um); mobile phase: [CO 2 -i-PrOH/ACN]; B%:70%, isocratic elution mode) to give the N-[3-(3-chloro-4-cyano-phenoxy)-2,2,4,4-tetramethyl-cyclobut yl]-2-[2-[1-methyl-1- [4-[(9S)-4,5,9,13-tetramethyl-3-thia-1,8,11,12-tetrazatricyc lo[8.3.0.02,6]trideca-2(6),4,7,10,12- pentaen-7-yl]phenyl]ethyl]-2,8-diazaspiro[3.5]nonan-8-yl]pyr imidine-5-carboxamide(230 mg, 262 μmol, 85% yield) obtained as a white solid. 1 H NMR: (400 MHz, DMSO-d6) δ = 8.74 (s, 2H), 8.04 (s, 1H), 7.90 (d, J = 8.8 Hz, 1H), 7.70 (d, J = 9.6 Hz, 1H), 7.23 - 7.17 (m, 2H), 7.13 (s, 1H), 7.11 - 7.07 (m, 1H), 7.06 - 6.98 (m, 2H), 4.89 (d, J = 6.8 Hz, 1H), 4.63 (d, J = 7.2 Hz, 1H), 4.29 (s, 1H), 4.03 (d, J = 9.2 Hz, 1H), 3.90 - 3.80 (m, 4H), 3.77 (d, J = 5.2 Hz, 2H), 2.69 - 2.55 (m, 6H), 2.42 (s, 4H), 1.68 - 1.57 (m, 7H), 1.21 (s, 6H), 1.11 (s, 6H). LC-MS: MS (ES + ): RT = 2.937 min, m/z = 929.2 [M +H + ]. EXAMPLE 38 – Synthesis of N-[3-(3-chloro-4-cyano-phenoxy)-2,2,4,4-tetramethyl- cyclobutyl]-2-[2-[3-fluoro-4-[(9S)-4,5,13-trimethyl-9-(oxazo l-2-ylmethyl)-3-thia-1,8,11,12- tetrazatricyclo[8.3.0.02,6]trideca-2(6),4,7,10,12-pentaen-7- yl]phenoxy]-7-azaspiro [3.5]nonan-7-yl]pyrimidine-5-carboxamide (I-457)

[ 0575] Step 1: Preparation of tert-butyl 2-(4-bromo-3-fluoro-phenoxy)-7-azaspiro [3.5]nonane-7-carboxylate. A mixture of 4-bromo-3-fluoro-phenol (500 mg, 2.6 mmol, 1.0 equiv), tert-butyl 2-(p-tolylsulfonyloxy)-7-azaspiro[3.5]nonane-7-carboxylate (1.04 g, 2.6 mmol, 1.0 equiv), K 2 CO 3 (724 mg, 5.2 mmol, 2.0 equiv) in DMF (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 partitioned between Ethyl acetate (100 mL) and H 2 O (100 mL x 2). The organic phase was separated, washed with brine (100 mL), dried over Na2SO4, filtered, and concentrated under reduced pressure to give the tert-butyl 2-(4-bromo-3-fluoro-phenoxy)-7- azaspiro[3.5]nonane-7-carboxylate (1.0 g, 2.4 mmol, 92% yield) as a white solid. [0576] Step 2: Preparation of tert-butyl 2-[3-fluoro-4-(4,4,5,5-tetramethyl-1,3,2- dioxaborolan-2-yl)phenoxy]-7-azaspiro[3.5]nonane-7-carboxyla te. A mixture of tert-butyl 2- (4-bromo-3-fluoro-phenoxy)-7-azaspiro[3.5]nonane-7-carboxyla te (900 mg, 2.2 mmol, 1.0 equiv), BPD (1.10 g, 4.3 mmol, 2.0 equiv), Pd(dppf)Cl 2 (159 mg, 217 μmol, 0.1 equiv), KOAc (427 mg, 4.3 mmol, 2.0 equiv) in dioxane (9.0 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 90 °C for 12 h under N2 atmosphere. The reaction mixture was partitioned between Ethyl acetate (100 mL) and (100 mL x 2). The organic phase was separated, washed with brine (100 mL), dried over Na2SO4, 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];gradient:70%-100% B over 15 min) to give the tert-butyl 2-[3-fluoro-4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2- yl)phenoxy]-7-azaspiro[3.5]nonane-7-carboxylate (800 mg, 1.7 mmol, 80% yield) as a white solid. [0577] Step 3: Preparation of tert-butyl 2-[3-fluoro-4-[(9S)-4,5,13-trimethyl-9-(oxazol-2- ylmethyl)-3-thia-1,8,11,12-tetrazatricyclo[8.3.0.02,6]tridec a-2(6),4,7,10,12-pentaen-7- yl]phenoxy]-7-azaspiro[3.5]nonane-7-carboxylate. A mixture of tert-butyl 2-[3-fluoro-4- (4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenoxy]-7-azas piro[3.5]nonane-7-carboxylate (138 mg, 299 μmol, 1.3 equiv), 2-[[(9S)-7-chloro-4,5,13-trimethyl-3-thia-1,8,11,12-tetraza- tricyclo[8.3.0.02,6]trideca-2(6),4,7,10,12-pentaen-9-yl]meth yl]oxazole (80 mg, 230 μmol, 1.0 equiv), Pd2(dba)3 (11 mg, 12 μmol, 0.05 equiv), PCy3 (6 mg, 23 μmol, 7 μL, 0.1 equiv) and K3PO4 (146 mg, 690 μmol, 3.0 equiv) in dioxane (2 mL) and H 2 O (0.4 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 100 °C for 3 h under N 2 atmosphere. The reaction mixture was partitioned between Ethyl acetate 100 mL and H 2 O 200 mL (100 mL x 2). The organic phase was separated, 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 prep-HPLC (column: Phenomenex luna C18150*25mm* 10um;mobile phase: [water(FA)-ACN];gradient:62%-89% B over 9 min) to give the tert-butyl 2-[3-fluoro-4-[(9S)- 4,5,13-trimethyl-9-(oxazol-2-ylmethyl)-3-thia-1,8,11,12-tetr azatricyclo[8.3.0.02,6]trideca- 2(6),4,7,10,12-pentaen-7-yl]phenoxy]-7-azaspiro[3.5]nonane-7 -carboxylate (60 mg, 92 μmol, 40% yield) as a white solid. [0578] Step 4: Preparation of 2-[[(9S)-7-[4-(7-azaspiro[3.5]nonan-2-yloxy)-2-fluoro- phenyl]-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]methyl]oxazole. A mixture of tert-butyl 2-[3-fluoro-4-[(9S)-4,5,13-trimethyl-9- (oxazol-2-ylmethyl)-3-thia-1,8,11,12-tetrazatricyclo[8.3.0.0 2,6]trideca-2(6),4,7,10,12-pentaen-7- yl]phenoxy]-7-azaspiro[3.5]nonane-7-carboxylate (60 mg, 92 μmol, 1.0 equiv) in DCM (3 mL) and TFA (1 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 25 °C for 1 h under N2 atmosphere. The reaction mixture was concentrated under reduced pressure and the residue was added DCM (30 mL) and washed with NaHCO 3 (30 mL) and brine (60ml), dried over Na2SO4, concentrated to get the residue, used for next step without further purification. The 2-[[(9S)-7-[4-(7-azaspiro[3.5]nonan-2-yloxy)-2-fluoro-phenyl ]-4,5,13- trimethyl-3-thia-1,8,11,12-tetrazatricyclo[8.3.0.02,6]tridec a-2(6),4,7,10,12-pentaen-9-yl] methyl]oxazole (51 mg, 93 μmol, 100% yield) was obtained as a yellow oil. [0579] Step 5: Preparation of N-[3-(3-chloro-4-cyano-phenoxy)-2,2,4,4-tetramethyl- cyclobutyl]-2-[2-[3-fluoro-4-[(9S)-4,5,13-trimethyl-9-(oxazo l-2-ylmethyl)-3-thia-1,8,11,12- tetrazatricyclo[8.3.0.02,6]trideca-2(6),4,7,10,12-pentaen-7- yl]phenoxy]-7-azaspiro[3.5] nonan-7-yl]pyrimidine-5-carboxamide. A mixture of 2-[[(9S)-7-[4-(7-azaspiro[3.5]nonan-2- yloxy)-2-fluoro-phenyl]-4,5,13-trimethyl-3-thia-1,8,11,12-te trazatricyclo[8.3.0.02,6]trideca- 2(6),4,7,10,12-pentaen-9-yl]methyl]oxazole (51 mg, 93 μmol, 1.0 equiv), 2-chloro-N-[3-(3- chloro-4-cyano-phenoxy)-2,2,4,4-tetramethyl-cyclobutyl] pyrimidine-5-carboxamide (47 mg, 111 μmol, 1.2 equiv), DIEA (24 mg, 186 μmol, 32 μL, 2.0 equiv) in NMP (2 mL) was degassed and purged with N2 for 3 times, and then the mixture was stirred at 25 °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 prep-HPLC (column: Phenomenex Luna C8150*25mm* 10um;mobile phase: [water(FA)-ACN];gradient:80%-98% B over 9 min) to give the N-[3-(3- chloro-4-cyano-phenoxy)-2,2,4,4-tetramethyl-cyclobutyl]-2-[2 -[3-fluoro-4-[(9S)-4,5,13- trimethyl-9-(oxazol-2-ylmethyl)-3-thia-1,8,11,12-tetrazatric yclo[8.3.0.02,6]trideca- 2(6),4,7,10,12-pentaen-7-yl]phenoxy]-7-azaspiro[3.5]nonan-7- yl]pyrimidine-5-carboxamide (50 mg, 54 μmol, 58% yield) as a white solid. Step 6: Preparation of N-[3-(3-chloro-4-cyano-phenoxy)-2,2,4,4-tetramethyl-cyclobut yl]-2- [2-[3-fluoro-4-[(9S)-4,5,13-trimethyl-9-(oxazol-2-ylmethyl)- 3-thia-1,8,11,12- tetrazatricyclo[8.3.0.02,6]trideca-2(6),4,7,10,12-pentaen-7- yl]phenoxy]-7-azaspiro [3.5]nonan-7-yl]pyrimidine-5-carboxamide (I-457). The residue was purified by SFC (P1 = 50 mg, Rt = 1.542 min) (column: REGIS(S,S)WHELK-O1(250mm*25mm,10um);mobile phase: [CO 2 -i-PrOH/ACN];B%:70%, isocratic elution mode) to give the N-[3-(3-chloro-4-cyano- phenoxy)-2,2,4,4-tetramethyl-cyclobutyl]-2-[2-[3-fluoro-4-[( 9S)-4,5,13-trimethyl-9-(oxazol-2- ylmethyl)-3-thia-1,8,11,12-tetrazatricyclo[8.3.0.02,6]tridec a-2(6),4,7,10,12-pentaen-7-yl] phenoxy]-7-azaspiro[3.5]nonan-7-yl]pyrimidine-5-carboxamide (50 mg, 54 μmol, 58% yield) as a white solid. 1 H NMR: (400 MHz, MeOD) δ = 8.72 (s, 2H), 7.90 (s, 1H), 7.72 (d, J = 8.8 Hz, 1H), 7.37 - 7.23 (m, 1H), 7.13 (s, 2H), 7.03 - 6.89 (m, 1H), 6.79 - 6.66 (m, 1H), 6.63 - 6.49 (m, 1H), 4.85 - 4.73 (m, 2H), 4.60 (s, 1H), 4.27 (s, 1H), 4.13 (s, 1H), 4.05 - 3.94 (m, 2H), 3.93 - 3.88 (m, 2H), 3.88 - 3.82 (m, 2H), 2.70 (s, 3H), 2.60 - 2.48 (m, 2H), 2.43 (s, 3H), 2.03 - 1.91 (m, 2H), 1.75 - 1.62 (m, 7H), 1.28 (s, 6H), 1.21 (s, 6H). LC-MS: MS (ES + ): RT = 2.936 min, m/z = 929.2 [M + H + ]. EXAMPLE 39 – Synthesis of Additional Compounds [0580] The following compounds were synthesized using procedures analogous to those described above: I-237 to I-670. Physical characterization data for exemplary compounds is provided in Table 4A. TABLE 4A. Observed Percent Purity Measured

EXAMPLE 40– Assay for Binding Affinity to Androgen Receptor [0392] Exemplary compounds were tested for ability to bind to the androgen receptor. Experimental procedures and results are provided below. Part I – Experimental Procedure [0393] Fractions of cell cytosol (106 cell/point) were incubated for 24 hours at 4 ℃ 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 [0394] Results showing ability of exemplary compounds to bind to the androgen receptor are provided in Table 5 below. The symbol “++++” indicates a Kd less than 0.05 μM. The symbol “+++” indicates an Kd in the range of 0.05 μM to 0.5 μM. The symbol “++” indicates a Kd in the range of greater than 0.5 μM to 2.5 μM. The symbol “+” indicates a Kd greater than 2.5 μM. TABLE 5. Compound No Kd Compound No Kd EXAMPLE 41 – Assay for Binding Affinity to BRD4-BD1 [0395] Exemplary compounds were tested for ability to bind to BRD4-BD1. Experimental procedures and results are provided below. Part I – Experimental Procedure [0396] 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 ℃ 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 [0397] Results showing ability of exemplary compounds to bind to BRD4-BD1 are provided in Table 6 below. The symbol “++++” indicates a Kd less than 0.05 μM. The symbol “+++” indicates a Kd in the range of 0.05 μM to 0.5 μM. The symbol “++” indicates a Kd in the range of greater than 0.5 μM to 2.5 μM. The symbol “+” indicates a Kd greater than 2.5 μM. TABLE 6. EXAMPLE 42 – Assay for Binding Affinity to BRD4-BD2 [0398] Exemplary compounds were tested for ability to bind to BRD4-BD2. Experimental procedures and results are provided below. Part I – Experimental Procedure [0399] 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 ℃ until lysis (90-150 minutes). The lysates were centrifuged (5,000 x g) and filtered (0.2 μm) 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 [0400] Results showing ability of exemplary compounds to bind to BRD4-BD2 are provided in Table 7 below. The symbol “++++” indicates a Kd less than 0.05 μM. The symbol “+++” indicates a Kd in the range of 0.05 μM to 0.5 μM. The symbol “++” indicates a Kd in the range of greater than 0.5 μM to 2.5 μM. The symbol “+” indicates a Kd greater than 2.5 μM. TABLE 7. Compound No. Kd Compound No. Kd

EXAMPLE 43 – Cellular Growth Inhibition Assay Using T-Rex 293 Cells [0401] 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 [0402] 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. [0403] 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. [0404] The SC3 cells were seeded on poly-D-lysine coated, black clear-bottom 384- well plates at 2500/well, in 25 μL 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 . [0405] 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 μL 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. 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. 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 [0406] The half-maximal growth inhibitory concentration (GI 50 ) results are provided in Tables 8 and 9 below for exemplary compounds. Table 8 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 GI 50 in the range of greater than 1.5 μM to 3 μM. The symbol “+” indicates a GI 50 greater than 3 μM. TABLE 8. [0407] Table 9 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 GI 50 less than 0.5 μM. The symbol “+++” indicates an GI 50 in the range of 0.5 μM to 1.5 μM. The symbol “++” indicates a GI 50 in the range of greater than 1.5 μM to 3 μM. The symbol “+” indicates a GI 50 greater than 3 μM. TABLE 9. Compound Compound Compound EXAMPLE 44 – Cellular Growth Inhibition Assay for VCaP Cells [0408] 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 [0409] 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 μL 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% CO 2 . [0410] 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 μL 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. [0411] 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. [0412] 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 [0413] The half-maximal growth inhibitory concentration (GI 50 ) results are provided in Table 10 below for exemplary compounds. The symbol “++++” indicates a GI 50 less than 0.5 μM. The symbol “+++” indicates an GI 50 in the range of 0.5 μM to 1.5 μM. The symbol “++” indicates a GI 50 in the range of greater than 1.5 μM to 3 μM. The symbol “+” indicates a GI 50 greater than 3 μM. TABLE 10. Compound Compound Compound EXAMPLE 45 – Cellular Growth Inhibition Assay Using T-Rex 293 Cells [0581] 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 [0582] 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. [0583] 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. [0584] The SC cells were seeded on poly-D-lysine coated, black clear-bottom 384-well plates at 2500/well, in 25 μL 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 . [0585] 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 μL 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. [0586] 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). [0587] 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 [0588] The half-maximal growth inhibitory concentration (GI 50 ) results are provided in Tables 11 and 12 below for exemplary compounds. Table 11 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 GI 50 less than 0.5 μM. The symbol “+++” indicates an GI 50 in the range of 0.5 μM to 1.5 μM. The symbol “++” indicates a GI 50 in the range of greater than 1.5 μM to 3 μM. The symbol “+” indicates a GI 50 greater than 3 μM.

TABLE 11. [0589] Table 12 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 GI 50 less than 0.5 μM. The symbol “+++” indicates an GI 50 in the range of 0.5 μM to 1.5 μM. The symbol “++” indicates a GI 50 in the range of greater than 1.5 μM to 3 μM. The symbol “+” indicates a GI 50 greater than 3 μM. TABLE 12. Compound No. GI 50 Compound No. GI 50 EXAMPLE 45 – Cellular Growth Inhibition Assay for T47D Cells [0590] 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 [0591] 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 μL 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 . [0592] 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 μL 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. [0593] 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). [0594] 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 [0595] The half-maximal growth inhibitory concentration (GI 50 ) results are provided in Table 13 below for exemplary compounds. The symbol “++++” indicates a GI 50 less than 0.5 μM. The symbol “+++” indicates an GI 50 in the range of 0.5 μM to 1.5 μM. The symbol “++” indicates a GI 50 in the range of greater than 1.5 μM to 3 μM. The symbol “+” indicates a GI 50 greater than 3 μM. TABLE 13. Compound Compound Compound

EXAMPLE 47– Cellular Growth Inhibition Assay Using T-Rex 293 Cells [0596] 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 [0597] 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. [0598] 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. [0599] The SC cells were seeded on poly-D-lysine coated, black clear-bottom 384-well plates at 2500/well, in 25 μL 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 . [0600] 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 μL 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. [0601] 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). [0602] 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 [0603] The half-maximal growth inhibitory concentration (GI 50 ) results are provided in Tables 14 and 15 below for exemplary compounds. Table 14 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 GI 50 in the range of greater than 1.5 μM to 3 μM. The symbol “+” indicates a GI 50 greater than 3 μM. TABLE 14. [0604] Table 15 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 GI 50 less than 0.5 μM. The symbol “+++” indicates an GI 50 in the range of 0.5 μM to 1.5 μM. The symbol “++” indicates a GI 50 in the range of greater than 1.5 μM to 3 μM. The symbol “+” indicates a GI 50 greater than 3 μM. TABLE 15. EXAMPLE 48 – Cellular Growth Inhibition Assay for LnCaP95 Cells [0605] Exemplary compounds were tested for ability to inhibit the proliferation of LnCaP95 Parental and LnCaP95 AR-FL KO cells. Experimental procedures and results are provided below. Part I – Experimental Procedure [0606] T47D LnCap95 Parental and T47D LnCap95 AR-FL KO cells were seeded in 25 μL growth media in poly-d lysine coated black clear-bottom 384-well plates at 2500 cells/well with Phenol Red Free RPMI containing 10% charcoal stripped serum and 1% pen strep. Following seeding, plates were spun at 300 × g for 30 seconds, then equilibrated to room temperature for 30 minutes, and moved into the incubator.24 hours after seeding, compounds were titrated in 100% DMSO and diluted in growth medium.25 μL of the compound/medium mixture was added to cells, bringing the total volume in each well to 50 μL. DMSO was used as a negative control. [0607] After treatment, plates were spun at 300 × g for 30 seconds, then cultured at 37°C with 5% CO 2 for 6 days in a humidified tissue culture incubator. On Day 6 of treatment, cell viability was quantified with CellTiter-Glo 2.0 reagent (Promega). Plates were equilibrated to room temperature for 30 minutes, then 25 μL of CellTiter-Glo 2.0 reagent was added to cells, bringing the total volume in each well to 75 μL. After reagent was added, plates were spun at 300 x g for 30 seconds then mixed on a shaker for two minutes at 500 rpm and then incubated in dark at room temperature for 10 minutes. Following incubation, plates were spun at 300 x g for 30 seconds, sealed with an optical adhesive cover, and luminescence readings were measured with an EnVision Plate Reader. [0608] Data was normalized to DMSO 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). Part II – Results [0609] Compound I-49 showed enhanced activity in parental LnCaP95 cells (GI 50 = 37 nM) compared to AR knockout cells in the CellTiterGlo assay. The half-maximal growth inhibitory concentration (GI 50 ) results are provided in Figure 1 and Figure 2. Figure 1 shows the cell viability dose-response curve for Compound I-49 in two cell lines, LnCap95 Parental cells and LnCap95AR-FL KO cells. Figure 2 shows the cell viability dose-response curves for Compound I-49 and Compound I-50 in LnCap95 Parental cells. EXAMPLE 49 – 22RV1 Apoptosis Assay [0610] Exemplary compounds were tested for ability to cause apoptosis in 22RV1 cell lines. Experimental procedures and results are provided below. Part I – Experimental Procedure [0611] 22RV1 Parental and 22RV1/AR cells were seeded in 25 μL growth media in poly-d lysine coated black clear-bottom 384-well plates at 5000 cells/well with Phenol Red Free RPMI containing 10% charcoal stripped serum and 1% PenStrep. Following seeding, plates were spun at 300 × g for 30 seconds, then equilibrated to room temperature for 30 minutes, and moved into the incubator.24 hours after seeding, compounds were titrated in 100% DMSO and diluted in growth medium.25 μL of the compound/medium mixture was added to cells, bringing the total volume in each well to 50 μL. DMSO was used as a negative control. [0612] After treatment, plates were spun at 300 × g for 30 seconds, then cultured at 37°C with 5% CO 2 for 24 or 48 hours in a humidified tissue culture incubator. After 24 or 48 hours of treatment, Caspase 3/7 activity was quantified with Caspase-Glo 3/7 reagent (Promega). Plates were equilibrated to room temperature for 30 minutes, then 25 μL of Caspase-Glo 3/7 reagent was added to cells, bringing the total volume in each well to 75 μL. After reagent was added, plates were spun at 300 x g for 30 seconds then mixed on a shaker for two minutes at 500 rpm and then incubated in the dark at room temperature for one hour. Following incubation, plates were spun at 300 x g for 30 seconds, sealed with an optical adhesive cover, and luminescence reads were measured with an EnVision Plate Reader. [0613] Data was normalized to DMSO for baseline. A four-parameter non-linear regression curve fit was applied to normalized data in GraphPad Prism data analysis software. Part II – Results [0614] Compound I-49 was found to be highly apoptotic in 22RV1 cells overexpressing FL-AR (22RV1 Parental(FL-AR hi )) (EC50 = 126 nM) as compared to the parental 22RV1 cell line that expressed low FL-AR levels (22RV1 Parental(FL-AR low )), as determined by the Caspase 3/7 Glo assay. The results are provided in Figure 3 and Figure 4. Figure 3 shows the relative Caspase 3/7 activity dose response curve for Compound I-49 in two cell lines, 22RV1 Parental(FL-AR hi ) and 22RV1 Parental(FL-AR low ). Figure 4 shows the relative Caspase 3/7 activity dose response curves of Compound I-49 and Compound I-50 in 22RV1 Parental(FL-AR hi ) cells. EXAMPLE 50 – TRex qPCR [0615] Exemplary compounds were tested for effector inhibition in androgen receptor (AR) expressing cells via quantitative RT-PCR of Gene X, a PD biomarker for essential protein (EP) inhibition. Experimental procedures and results are provided below. Part I – Experimental Procedure [0616] TReX293 cells were seeded in 2 mL medium in poly-d lysine coated 6-well plate at 500K cells/well with or without 10 ng/mL doxycycline Phenol Red Free DMEM containing 10% charcoal stripped serum and 1% pen strep and moved into the incubator.24 hours after seeding, compounds were titrated in 100% DMSO and diluted in growth medium. A 0.5 mL aliquot of the compound/medium mixture was added to cells, bringing the total volume in each well to 2.5 mL. DMSO was used as a negative control.16 hours after treatment, cells were collected and pelleted down. [0617] Total RNA was isolated using RNeasy Mini kit (#74104, Qiagen) according to the manufacturers’ instructions. After quantification, 1 ^g RNA of each sample was processed to first strand C-DNA synthesis with Verso cDNA synthesis kit (#AB1453B, ThermoFisher) and following qRT-PCR reaction was performed using Power SYBR Green Master mix (#4367659, ThermoFisher) with CFX Opus 384 Real-Time PCR System (#12011452, Biorad) according to the manufacturers’ instructions. The gene-specific primers used in qRT-PCR are HEXIM1, forward 5′-GGTGGCAATCGAGAGCGTT-3′ (SEQ ID 1) and reverse 5′- GGAGGTGGTTGGGATTCCA-3′ (SEQ ID 2); HPRT, forward 5′- CCTGGCGTCGTGATTAGTGAT-3′ (SEQ ID 3) and reverse 5′- AGACGTTCAGTCCTGTCCATA-3′ (SEQ ID 4). All PCR reactions were performed in biological duplicates and technical triplicates. Part II – Results [0618] The relative total Gene X mRNA dose response curve results are provided in Figure 5 Figure 5 shows the relative total Gene X mRNA dose response curve for Compound I-49 in TReX293 cells in the presence and absence of doxycycline. EXAMPLE 51 – Castrate VCaP Tumor Xenograft Model [0619] Exemplary compounds were tested for oral in-vivo efficacy in mice in an Ar amp , V7 + Castrate VcaP Model. Experimental procedures and results are provided below. Part I – Experimental Procedure [0620] 5M VCaP cells were subcutaneously implanted into CB17.SCID mice in 50% Matrigel (Corning). Mice were castrated under anesthesia once tumor volumes reached 100 mm 3 . Tumors were then allowed to reach an average of approximately 150 mm 3 before randomization into efficacy arms (n=10/12/15) as indicated. Animals were dosed daily via oral gavage with the indicated compounds for varying treatment durations. In Arm 1of the study mice were dosed daily via oral gavage with indicated compounds starting on treatment day 0. In Arm 2, mice were dosed daily via oral gavage with Enzalutamide starting on treatment day 0, then at treatment day 21 mice were randomized into treatment groups and subsequently dosed daily via oral gavage with indicated compounds. For the PK/PD study, tumors were allowed to reach an average of 300 mm 3 , before dosing the mice with indicated compounds by oral gavage (n=5) for three days. Mice were sacrificed 24 hours following the final dose, and tumors were harvested for downstream analysis. Plasma samples were collected from all dosed mice 2 hours following the final dose to determine drug exposures by LC-MS/MS. Part II – Results [0621] The Castrate VCaP tumor xenograft growth curves are provided in Figure 6 and Figure 7. Figure 6 shows tumor growth curves for Compound I-49 treated mice, Enzalutimide treated mice, and vehicle treated mice in Arm 1 of the Ar amp , V7 + Castrate VcaP Model. Figure 7 shows tumor growth curves for Compound I-49 treated mice, Enzalutimide treated mice, and vehicle treated mice in Arm 2 of the Ar amp , V7 + Castrate VcaP Model. EXAMPLE 52 – Tumor qPCR [0622] Exemplary compounds were tested for oral in-vivo efficacy via quantitative RT-PCR of Gene X, a PD biomarker for EP inhibition, in tumor xenografts from the above described Ar amp , V7 + Castrate VcaP Model. Experimental procedures and results are provided below. Part I – Experimental Procedure [0623] Tumors collected from mice at the end of the PK/PD study were lysed in 1.5 mL buffer RLT from the RNeasy Mini Kit (#74104, Qiagen) and pelleted. Total RNA was isolated using RNeasy Mini kit according to the manufacturers’ instructions. After quantification, 1 ^g RNA of each sample was used as a template for first strand C-DNA synthesis with Verso cDNA synthesis kit (#AB1453B, ThermoFisher) and the qRT-PCR reaction was performed using Power SYBR Green Master mix (#4367659, ThermoFisher) with CFX Opus 384 Real-Time PCR System (#12011452, Biorad) according to the manufacturers’ instructions. The gene-specific primers used in qRT-PCR are HEXIM1, forward 5′-GGTGGCAATCGAGAGCGTT-3′ (SEQ ID 1) and reverse 5′-GGAGGTGGTTGGGATTCCA-3′ (SEQ ID 2); HPRT, forward 5′- CCTGGCGTCGTGATTAGTGAT-3′ (SEQ ID 3) and reverse 5′- AGACGTTCAGTCCTGTCCATA-3′ (SEQ ID 4). All PCR reactions were performed in technical triplicates. Part II – Results [0624] The relative quantitaive Gene X mRNA results are provided in Figure 8 and Figure 9. Figure 8 shows the relative total Gene X mRNA present in tumors collected at the end of the PK/PD Ar amp , V7 + Castrate VcaP Model described above, for varying doses of Compound I-49. Figure 9 shows the relative total Gene X mRNA present in tumors collected at the end of the PK/PD Ar amp , V7 + Castrate VcaP Model described above, for treatment with Compound I-49 and Compound I-50. EXAMPLE 53 – Plasma PSA Concentration [0625] Exemplary compounds were tested for oral in-vivo efficacy in mice via plasma Prostate Specific Antigen (PSA) reduction in the Castrate VCaP Tumor Xenograft Model desribed above. Experimental procedures and results are provided below. Part I – Experimental Procedure [0626] All assay reagents were provided in the Human Prostate Specific Antigen (PSA) ELISA Kit (ab113327, Abcam). Plasma samples were collected at the end of the above described Castrate VCaP Tumor Xenograft Model and diluted in assay diluent A for the absorbance signal to be in the linear range of the recombinant human PSA standard curve. Diluted samples and PSA standard were loaded into a pre-coated assay plate and incubated for 2.5 hours at ambient temperature. Samples and standards were flicked out, plate was washed, and biotinylated PSA detection antibody was added and incubated for 1 hour. Detection antibody was flicked out, plate was washed, and HRP-Streptavidin was added and incubated for 45 minutes. HRP-Streptavidin was flicked out, plate was washed, and TMB Substrate was added and incubated for 30 minutes while reading absorbance at 620 nm on the SpectraMax M5e, for up to 30 minutes. Stop solution was added directly on top of TMB Substrate. Endpoint read was obtained at an absorbance of 450 nm. During analysis, sample absorbances were multiplied by the dilution factor to calculate the concentration of PSA. Part II – Results [0627] The are provided in Figure 10 and Figure 11. Figure 10 shows the plasma PSA concentration at varying Compound I-49 doses for samples collected from Arm 1 of the Castrate VCaP Tumor Xenograft Model described above. Figure 11 shows the plasma PSA concentration at varying Compound I-49 doses, for samples collected from Arm 2 of the Castrate VCaP Tumor Xenograft Model described above. EXAMPLE 54 – In-vitro Ternary Complex Formation via ELISA [0628] Exemplary compounds were tested for ability to induce in VCaP cells the formation of a ternary complex comprising AR, test compond, and EP. Experimental procedures and results are provided below. Part I – Experimental Procedure [0629] VCaP cells were cultured in ATCC DMEM medium supplemented with 10% ATCC fetal bovine serum, and 1% pen-strep (culture medium).5000 cells were seeded in 100 µLof culture medium, per well in a clear 96-well poly-d-lysine coated plate. Plates were equilibrated to room temperature for 30 minutes. After equilibration, plates were incubated at 37°C with 5% CO 2 ^in a humidified tissue culture incubator. Black, opaque 96-well MaxiSorp plates were coated with 50 µL of a solution of androgen receptor antibody diluted in phosphate buffered saline (PBS). The coated plates were then quick-spun for 30 seconds before being sealed and placed in a fridge at 4 °C overnight. Compounds were titrated in 100% DMSO the day before the experiment was performed and were left sealed in the desiccator overnight. QuantaRed™ Enhanced Chemifluorescent HRP Substrate Kit was taken out from 4 °C to warm up to room temperature. Plates coated with androgen receptor antibody were washed three times with 200 µL 1x Tris-Buffered Saline plus 0.1% Tween20 (TBST), then blocked with 200 µL 3% bovine serum albumin in TBST for approximately 2 hours on a room temperature plate shaker at 500 rpm for 1 hour. Compound titrations were diluted in culture medium.100 µL of the compound/media mixture was added to cells, bringing the total volume to 200 µL per well. DMSO was used as a negative control. [0630] After treatment, plates were incubated at 37°C with 5% CO 2 in a humidified tissue culture incubator for 1 hour. Compound/media was flicked off cells into sink, then cells were washed once with 250 µL 1x PBS.50 µL of 1x cell signaling technology lysis buffer, phosphatase/protease inhibitor, and nuclease was added to each cell well, then put on a plate shaker in a 4 °C Fridge at 400 rpm for 20 minutes. Plate was washed three times with TBST while waiting for the incubation to be complete. Lysates were quick-spun for 30 seconds, then lysate was transferred from clear 96-well plate into black, MaxiSorp plate. Plate was covered with aluminum foil seal and placed onto a room temperature plate shaker at 500 rpm for 1 hour. Lysates were flicked from plate and washed three time with TBST.50 µL HRP-BRD4 antibody diluted in TBST was added to the black MaxiSorp plate. Plate was covered with aluminum foil seal. This was put onto a room temperature plate shaker at 500 rpm for 1 hour. HRP-BRD4 antibody solution was flicked and washed three time with TBST. It was then washed three times with PBS. QuantaRed™ Enhanced Chemifluorescent HRP Substrate Kit solution was made and 100 µL was added to each well. Plate was covered with aluminum foil seal and incubated in drawer for 1 hour. Fluorescent endpoint read was conducted on the SpectraMax. Part II – Results [0631] Intracellular ternary complexes can be detected following lysis using a sandwich ELISA. Endogenous AR is immunoprecipitated and the presence of EP in the complex is detected using an anti-EP antibody. Neither the AR ligand nor the EP ligand by itself induces ternary complex formation. Results are provided in Figure 12 and Figure 13. Figure 12 shows the in-vitro relative ternary complex formation dose response curve for Compound I-49 versus free AR ligand and free EP ligand. Figure 13 shows the in-vitro relative ternary complex formation dose response curve for Compound I-49 and Compound I-50. EXAMPLE 55 – In-vivo TR-FRET [0632] Exemplary compounds were tested for ability to induce cellular ternary complex formation in-vivo in the above described Ar amp , V7 + Castrate VcaP Model. Experimental procedures and results are provided below. Part I – Experimental Procedure [0633] VCaP tumors obtained in the above described Ar amp , V7 + Castrate VcaP Model were lysed in Cell Signaling Technology lysis buffer supplemented with protease/phosphatase inhibitors and nuclease. Each tumor was lysed in 10 µL of lysis buffer per mg of tumor and placed in a TissueLyser for 4 minutes at 20 Hz. Each sample was spun down at 16000 x g for 10 minutes, the supernatant was removed, and a BCA assay was run to quantify the total protein concentration in each sample. In a white opaque 384 well plate each sample was diluted in supplemented CST lysis buffer to 48 and 24 µg of protein in a total volume of 16 µL.4 µL of a 1:1 mixture of anti-AR and anti-BRD4 antibodies with donor and acceptor fluorophores were mixed into each well containing tumor samples. The plate was spun down at 300 x g for one minute, placed on a plate shaker at 400 RPM for 10 minutes, spun down at 300 x g for one minute, then sealed with a plate seal and stored overnight. Approximately 16 hours after adding antibodies, the plate was read on an Envision plate reader. The ratiometric value of each well was calculated by dividing the acceptor signal by the donor signal and multiplying by 10,000. Part II – Results The presence of the ternary complex comprising androgen receptor, test compound, and EP in tumor cells collected in the above described Ar amp , V7 + Castrate Model was evaluated using anti- AR and anti-BRD4 antibodies with donor and acceptor fluorophores. The FRET signal results are provided in Figures 14, 15, and 16. Figure 14 shows the relative ternary complex formation present with treatment of Compound I-49 at varying doses in samples collected at the end of the PK/PD Castrate VCaP Tumor Xenograft Model study described above. Figure 15 shows the relative ternary complex formation present with treatment of Compound I-49 in samples collected at the end of Arm 2 of the Castrate VCaP Tumor Xenograft Model described above. Figure 16 shows the relative ternary complex formation present with treatment of Compound I- 49 and Compund I-50 in samples collected at the end of the PK/PD Castrate VCaP Tumor Xenograft Model described above. EXAMPLE 56 – Assay for Binding Affinity to Progesterone Receptor [0634] Exemplary compounds were tested for ability to bind to the progesterone receptor. Experimental procedures and results are provided below. Part I – Experimental Procedure [0635] 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 [0636] Results showing ability of exemplary compounds to bind to the progesterone receptor are provided in Table 16 below. The symbol “++++” indicates a Ki less than 0.05 μM. The symbol “+++” indicates an Ki in the range of 0.05 μM to 0.5 μM. The symbol “++” indicates a Ki in the range of greater than 0.5 μM to 2.5 μM. The symbol “+” indicates a Ki greater than 2.5 μM. TABLE 16. EXAMPLE 57 – FP Assay for Binding Affinity to Progesterone Receptor [0637] 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 [0638] 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 [0639] The half-maximal inhibitory concentration (IC 50 ) results are provided in Table 17 below for exemplary compounds. The symbol “++++” indicates a IC 50 less than 0.05 μM. The symbol “+++” indicates an IC 50 in the range of 0.05 μM to 0.5 μM. The symbol “++” indicates a IC 50 in the range of greater than 0.5 μM to 2.5 μM. The symbol “+” indicates a IC 50 greater than 2.5 μM. TABLE 17. INCORPORATION BY REFERENCE [0414] The entire disclosure of each of the patent documents and scientific articles referred to herein is incorporated by reference for all purposes. EQUIVALENTS [0415] 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.