In various embodiments, the present disclosure generally relates to novel compounds, compositions comprising the same, methods of preparing and methods of using the same, e.g., for inhibiting DGKs and/or for treating a number of diseases or disorders, such as cancers or infections.
Diacylglycerol kinases (DGKs) have been indicated as serving as intracellular checkpoints. Inhibition of DGKs are expected to enhance T cell signaling pathways and T cell activation. See e.g., Riese M. J. et al., Journal of Biological Chemistry, (2011) 7: 5254-5265; Zha Y et al., Nature Immunology, (2006) 12:1343; Olenchock B. A. et al., (2006) 11: 1174-81). DGKa (DGK alpha) and DGKz (DGK zeta) have been viewed as targets for cancer immunotherapy. There remains a need for compounds useful as inhibitors of one or both of DGKa and DGKz.
The present disclosure is based in part on Applicant's discovery of compounds that have activity as inhibitors of one or both of DGKa and DGKz. In various embodiments, the present disclosure provides novel compounds, pharmaceutical compositions, methods of preparing and using the same. Typically, the compounds herein are DGK inhibitors, such as DGKa and/or DGKz inhibitors. The compounds and compositions herein are useful for treating various diseases or disorders, such as cancer or viral infections.
In some embodiments, the present disclosure provides a compound of Formula III-1, III-2, III-3, III-4, III-5, III-6, III-7, III-8, III-9, III-10, III-11, or III-12, or a pharmaceutically acceptable salt thereof, as defined herein. In some embodiments, the compound of Formula III-2 can also be characterized as having a structure according to a subformula of Formula III-2-A, III-2-B, III-2-C, III-2-C-1, III-2-C-2, III-2-C-3, III-2-C-4, III-2-D, III-2-D-1, III-2-D-2, III-2-D-3, III-2-D-4, III-2-E-1, III-2-E-2, III-2-E-3, III-2-E-1a, III-2-E-2a, III-2-E-3a, III-2-E-1b, III-2-E-2b, III-2-E-3b, III-2-F-1a, III-2-F-1b, III-2-F-2a, III-2-F-2b, III-2-F-3a, or III-2-F-3b. In some embodiments, the compound of Formula III-3 can also be characterized as having a structure according to a subformula of Formula III-3-A. In some embodiments, the compound of Formula III-4 can also be characterized as having a structure according to a subformula of Formula III-4-A, III-4-B, III-4-C, III-4-D, III-4-C-1, III-4-C-2, III-4-C-3, III-4-C-4, III-4-D-1, III-4-D-2, III-4-C-4a, III-4-C-4b, III-4-C-4c, III-4-C-4d, III-4-C-4e, or III-4-C-4f. In some embodiments, the compound of Formula III-8 can also be characterized as having a structure according to a subformula of Formula III-8-A or III-8-B. In some embodiments, the present disclosure also provides a compound selected from the compounds shown in Table A herein, or a pharmaceutically acceptable salt thereof. In some embodiments, the present disclosure also provides a compound selected from Compound Nos. 1-347 herein, or a pharmaceutically acceptable salt thereof.
Certain embodiments of the present disclosure are directed to a pharmaceutical composition comprising one or more of the compounds of the present disclosure (e.g., a compound of Formula III-1, III-2 (e.g., III-2-A, III-2-B, III-2-C, III-2-C-1, III-2-C-2, III-2-C-3, III-2-C-4, III-2-D, III-2-D-1, III-2-D-2, III-2-D-3, III-2-D-4, III-2-E-1, III-2-E-2, III-2-E-3, III-2-E-1a, III-2-E-2a, III-2-E-3a, III-2-E-1b, III-2-E-2b, III-2-E-3b, III-2-F-1a, III-2-F-1b, III-2-F-2a, III-2-F-2b, III-2-F-3a, or III-2-F-3b), III-3 (e.g., III-3-A), III-4 (e.g., III-4-A, III-4-B, III-4-C, III-4-D, III-4-C-1, III-4-C-2, III-4-C-3, III-4-C-4, III-4-D-1, III-4-D-2, III-4-C-4a, III-4-C-4b, III-4-C-4c, III-4-C-4d, III-4-C-4e, or III-4-C-4f), III-5, III-6, III-7, III-8 (e.g., III-8-A or III-8-B), III-9, III-10, III-11, or III-12, any of Compound Nos. 1-347, any compound selected from the compounds shown in Table A herein, or a pharmaceutically acceptable salt thereof) and optionally a pharmaceutically acceptable excipient. The pharmaceutical composition described herein can be formulated for various routes of administration, such as oral administration, parenteral administration, or inhalation etc.
Certain embodiments are directed to a method of treating a disease or disorder associated with the activity of DGKa, DGKz, or both DGKa and DGKz. In some embodiments, the method comprises administering to a subject in need thereof a therapeutically effective amount of a compound of the present disclosure (e.g., a compound of Formula III-1, III-2 (e.g., III-2-A, III-2-B, III-2-C, III-2-C-1, III-2-C-2, III-2-C-3, III-2-C-4, III-2-D, III-2-D-1, III-2-D-2, III-2-D-3, III-2-D-4, III-2-E-1, III-2-E-2, III-2-E-3, III-2-E-1a, III-2-E-2a, III-2-E-3a, III-2-E-1b, III-2-E-2b, III-2-E-3b, III-2-F-1a, III-2-F-1b, III-2-F-2a, III-2-F-2b, III-2-F-3a, or III-2-F-3b), III-3 (e.g., III-3-A), III-4 (e.g., III-4-A, III-4-B, III-4-C, III-4-D, III-4-C-1, III-4-C-2, III-4-C-3, III-4-C-4, III-4-D-1, III-4-D-2, III-4-C-4a, III-4-C-4b, III-4-C-4c, III-4-C-4d, III-4-C-4e, or III-4-C-4f), III-5, III-6, III-7, III-8 (e.g., III-8-A or III-8-B), III-9, III-10, III-11, or III-12, any of Compound Nos. 1-347, any compound selected from the compounds shown in Table A herein, or a pharmaceutically acceptable salt thereof) or a therapeutically effective amount of a pharmaceutical composition described herein. Diseases or disorders associated with the activity of DGKa, DGKz, or both DGKa and DGKz suitable to be treated with the method include any of those described herein.
In some embodiments, a method of treating cancer is provided. In some embodiments, the method comprises administering to a subject in need thereof a therapeutically effective amount of a compound of the present disclosure (e.g., a compound of Formula III-1, III-2 (e.g., III-2-A, III-2-B, III-2-C, III-2-C-1, III-2-C-2, III-2-C-3, III-2-C-4, III-2-D, III-2-D-1, III-2-D-2, III-2-D-3, III-2-D-4, III-2-E-1, III-2-E-2, III-2-E-3, IT-2-E-1a, III-2-E-2a, III-2-E-3a, III-2-E-1b, III-2-E-2b, III-2-E-3b, III-2-F-1a, III-2-F-1b, III-2-F-2a, III-2-F-2b, III-2-F-3a, or III-2-F-3b), III-3 (e.g., III-3-A), III-4 (e.g., III-4-A, III-4-B, III-4-C, III-4-D, III-4-C-1, III-4-C-2, III-4-C-3, III-4-C-4, III-4-D-1, III-4-D-2, III-4-C-4a, III-4-C-4b, III-4-C-4c, III-4-C-4d, III-4-C-4e, or III-4-C-4f), III-5, III-6, III-7, III-8 (e.g., III-8-A or III-8-B), III-9, III-10, III-11, or III-12, any of Compound Nos. 1-347, any compound selected from the compounds shown in Table A herein, or a pharmaceutically acceptable salt thereof) or a therapeutically effective amount of a pharmaceutical composition described herein. In various embodiments, the cancer can be cancer of the colon, pancreatic cancer, breast cancer, prostate cancer, lung cancer, ovarian cancer, cervical cancer, renal cancer, cancer of the head and neck, lymphoma, leukemia, and/or melanoma.
In some embodiments, a method of treating viral infection is provided. In some embodiments, the method comprises administering to a subject in need thereof a therapeutically effective amount of a compound of the present disclosure (e.g., a compound of Formula III-1, III-2 (e.g., III-2-A, III-2-B, III-2-C, III-2-C-1, III-2-C-2, III-2-C-3, III-2-C-4, III-2-D, III-2-D-1, III-2-D-2, III-2-D-3, III-2-D-4, III-2-E-1, III-2-E-2, III-2-E-3, III-2-E-1a, III-2-E-2a, III-2-E-3a, III-2-E-1b, III-2-E-2b, III-2-E-3b, III-2-F-1a, III-2-F-1b, III-2-F-2a, III-2-F-2b, III-2-F-3a, or III-2-F-3b), III-3 (e.g., III-3-A), III-4 (e.g., III-4-A, III-4-B, III-4-C, III-4-D, III-4-C-1, III-4-C-2, III-4-C-3, III-4-C-4, III-4-D-1, III-4-D-2, III-4-C-4a, III-4-C-4b, III-4-C-4c, III-4-C-4d, III-4-C-4e, or III-4-C-4f), III-5, III-6, III-7, III-8 (e.g., III-8-A or III-8-B), III-9, III-10, III-11, or III-12, any of Compound Nos. 1-347, any compound selected from the compounds shown in Table A herein, or a pharmaceutically acceptable salt thereof) or a therapeutically effective amount of a pharmaceutical composition described herein.
The administering in the methods herein is not limited to any particular route of administration. For example, in some embodiments, the administering can be orally, nasally, transdermally, pulmonary, inhalationally, buccally, sublingually, intraperintoneally, subcutaneously, intramuscularly, intravenously, rectally, intrapleurally, intrathecally and parenterally.
The compounds of the present disclosure can be used as a monotherapy or in a combination therapy. In some embodiments, the combination therapy includes treating the subject with a targeted therapeutic agent, chemotherapeutic agent, therapeutic antibody, radiation, cell therapy, and/or immunotherapy. In some embodiments, the combination therapy includes treating the subject with an immune-oncology agent herein. In some embodiments, the combination therapy includes treating the subject with one or more additional antiviral agents.
In some embodiments, the present disclosure provides the following exemplary enumerated Embodiments 1-49:
Embodiment 1. A compound of Formula III-1, III-2, III-3, III-4, III-5, III-6, III-7, III-8, III-9, III-10, III-11, or III-12, or a pharmaceutically acceptable salt thereof:
wherein:
Embodiment 2. The compound of Embodiment 1, or a pharmaceutically acceptable salt thereof, wherein Y is N.
Embodiment 3. The compound of Embodiment 1, or a pharmaceutically acceptable salt thereof, wherein Y is CH.
Embodiment 4. The compound of any of Embodiments 1-3, or a pharmaceutically acceptable salt thereof, wherein U is N.
Embodiment 5. The compound of any of Embodiments 1-3, or a pharmaceutically acceptable salt thereof, wherein U is CR6.
Embodiment 6. The compound of Embodiment 5, or a pharmaceutically acceptable salt thereof, wherein Rb is hydrogen.
Embodiment 7. The compound of Embodiment 5, or a pharmaceutically acceptable salt thereof,
Embodiment 8. The compound of any of Embodiments 1-7, or a pharmaceutically acceptable salt thereof, wherein as applicable, R1 is CN.
Embodiment 9. The compound of any of Embodiments 1-7, or a pharmaceutically acceptable salt thereof, wherein as applicable, (i) R1 is halogen, e.g., F or Cl; (ii) R1 is
(iii) R1 is an optionally substituted 5 or 6 membered heteroaryl having 1-4 ring heteroatoms independently selected from O, S, and N, such as an optionally substituted pyrimidinyl or an optionally substituted thiazolyl, for example, R1 is
or (iv) R1 is SRA1, wherein RA1 is independently optionally substituted C1-4 alkyl, for example, R1 is SCH3.
Embodiment 10. The compound of Embodiment 1, or a pharmaceutically acceptable salt thereof, characterized as having a structure according to Formula III-4-C-1, III-4-C-2, III-4-C-3, III-4-C-4, III-4-D-1, or III-4-D-2
Embodiment 11. The compound of any of Embodiments 1-9, or a pharmaceutically acceptable salt thereof, wherein R2 is hydrogen.
Embodiment 12. The compound of any of Embodiments 1-11, or a pharmaceutically acceptable salt thereof, wherein (i) R3 is an optionally substituted C1-4 alkyl, such as CH3, CD3 etc; (ii) R3 is hydrogen; (iii) R3 is an optionally substituted C1-4 alkyl, such as CH(CH3)2, CH2CH3, CH2CHF2 etc; or (iv) R3 is an optionally substituted C3-6 cycloalkyl, such as cyclopropyl, cyclobutyl etc.
Embodiment 13. The compound of any of Embodiments 1-12, or a pharmaceutically acceptable salt thereof, wherein Z is C(O).
Embodiment 14. The compound of any of Embodiments 1-13, or a pharmaceutically acceptable salt thereof, wherein as applicable, R9 at each occurrence is independently an optionally substituted C1-4 alkyl or optionally substituted C3-6 cycloalkyl, preferably, CH3, CH2CH3, CH2CH2CH3, fluorine-substituted C1-3 alkyl (e.g., CF2H), cyclopropyl, cyclobutyl, CH2OH, CH2OCH3, CH2OCH2CH3, CH2NH2, CH2N3, or CH2NHC(O)OCH3.
Embodiment 15. The compound of any of Embodiments 1-14, or a pharmaceutically acceptable salt thereof, wherein as applicable, X is N.
Embodiment 16. The compound of any of Embodiments 1-14, or a pharmaceutically acceptable salt thereof, wherein as applicable, X is CR5, wherein R5 is CN.
Embodiment 17. The compound of any of Embodiments 1-14, or a pharmaceutically acceptable salt thereof, wherein as applicable, X is CR5, wherein R5 is CONH2, COOH, CONH(RC1) CON(RC1)2, or COORC1, wherein RC1 at each occurrence is independently a C1-4 alkyl, such as methyl.
Embodiment 18. The compound of any of Embodiments 1-17, or a pharmaceutically acceptable salt thereof, wherein as applicable, L1 is
or (either of the two attaching points of the piperidine, N or C, can be attached to L2), wherein:
Embodiment 19. The compound of Embodiment 18, or a pharmaceutically acceptable salt thereof, wherein R9A at each occurrence is independently CH3, CH2CH3, CH2CH2CH3, fluorine-substituted C1-3 alkyl (e.g., CF2H), cyclopropyl, cyclobutyl, CH2OH, CH2OCH3, CH2OCH2CH3, CH2NH2, CH2N3, or CH2NHC(O)OCH3.
Embodiment 20. The compound of any of Embodiments 1-17, or a pharmaceutically acceptable salt thereof, wherein as applicable, L1 is selected from the following (the bottom attaching point, N or C, is attached to L2):
Embodiment 21. The compound of any of Embodiments 1-17, or a pharmaceutically acceptable salt thereof, wherein as applicable, L1 is selected from the following (the bottom attaching point, N or C, is attached to L2):
Embodiment 22. The compound of any of Embodiments 1-17, or a pharmaceutically acceptable salt thereof, wherein as applicable, L1 is an optionally substituted 7-12 membered heterocyclylene having two or more rings and 1-4 ring heteroatoms each independently O, N, or S, when substituted, the substituent(s) can be attached to any one or more of the two or more rings.
Embodiment 23. The compound of any of Embodiments 1-17, or a pharmaceutically acceptable salt thereof, wherein as applicable, L1 is selected from the following bicyclic heterocyclylene (the bottom attaching point, N or C, is attached to L2
Embodiment 24. The compound of any of Embodiments 1-17, or a pharmaceutically acceptable salt thereof, wherein as applicable, L1 is selected from the following bicyclic heterocyclylene (the bottom attaching point, N or C, is attached to L2)
Embodiment 25. The compound of any of Embodiments 1-17, or a pharmaceutically acceptable salt thereof, wherein as applicable, L1 is —N(C1-4 alkyl)-, such as —N(CH3)—.
Embodiment 26. The compound of any of Embodiments 1-25, or a pharmaceutically acceptable salt thereof, wherein L2 is absent.
Embodiment 27. The compound of any of Embodiments 1-25, or a pharmaceutically acceptable salt thereof, wherein L2 is an optionally substituted C1-4 alkylene selected from the following:
wherein:
Embodiment 28. The compound of Embodiment 27, or a pharmaceutically acceptable salt thereof, wherein R13 is hydrogen.
Embodiment 29. The compound of Embodiment 27, or a pharmaceutically acceptable salt thereof, wherein R13 is CN, OH, COOH, CONH2, methoxy, ethoxy, cyclopropyl, cyclobutyl, optionally substituted phenyl, or optionally substituted 5 or 6 membered heteroaryl having 1-3 ring heteroatoms independently selected from N, O, and S, wherein, when substituted, the optionally substituted phenyl or 5 or 6 membered heteroaryl is substituted with 1-3 substituents independently selected from halogen (preferably F or Cl), OH, CN, C1-4 alkyl optionally substituted with 1-3 F, C1-4 heteroalkyl optionally substituted with 1-3 F, and 3-4 membered ring (including cyclopropyl, cyclobutyl, oxetanyl, azetidinyl, etc.) optionally substituted with F and/or methyl.
Embodiment 30. The compound of any of Embodiments 1-25, or a pharmaceutically acceptable salt thereof, wherein L2 is 0 or C(O).
Embodiment 31. The compound of any of Embodiments 1-25, or a pharmaceutically acceptable salt thereof, wherein L2 is an optionally substituted 5 or 6 membered heteroarylene, for example,
Embodiment 32. The compound of any of Embodiments 1-17, or a pharmaceutically acceptable salt thereof, wherein as applicable, L1 is —N(RE)—, L2 is an optionally substituted phenylene, e.g.,
and R30 is hydrogen.
Embodiment 33. The compound of any of Embodiments 1-17, or a pharmaceutically acceptable salt thereof, wherein as applicable, L1, L2, and R30 together are:
Embodiment 34. The compound of any of Embodiments 1-31, or a pharmaceutically acceptable salt thereof, wherein R30 is optionally substituted phenyl, such as a phenyl which is substituted with 1-3 (e.g., 1 or 2) R22, wherein R22 at each occurrence is independently halogen (e.g., F, Cl, or Br), CN, OH, NH2, RM, ORM, COOH, CONH2, COORM, CONH(RM), CON(RM)2, NHCO(RM), N(RM)CO(RM), SO2RM, SO2NH2, S(O)(NH)RM, S(O)(NRM)RM, SO2N(RM)2, NHSO2RM, N(RM)SO2RM, P(O)(RM)2, P(O)(ORM)2, NH(RM), N(RM)2, SRM, or SF5, wherein RM at each occurrence is independently an optionally substituted C1-4 alkyl, optionally substituted C2-4 alkenyl, optionally substituted C2-4 alkynyl, optionally substituted C3-6 cycloalkyl, optionally substituted phenyl, optionally substituted 5 or 6 membered heteroaryl having 1-4 ring heteroatoms independently selected from O, S, and N, or optionally substituted 4-7 membered heterocyclyl.
Embodiment 35. The compound of Embodiment 34, or a pharmaceutically acceptable salt thereof, wherein R30 is a phenyl which is substituted with 1-3 (e.g., 1 or 2) R22, wherein R22 at each occurrence is independently halogen (e.g., F, Cl, or Br), CN, OH, RM1, ORM1, SO2RM1, P(O)(RM1)2, SRM1, or SF5, wherein RM1 at each occurrence is independently an optionally substituted C1-4 alkyl or an optionally substituted 3-4 membered ring (including cyclopropyl, cyclobutyl, oxetanyl, azetidinyl, etc.), wherein when substituted, the optionally substituted C1-4 alkyl or 3-4 membered ring is substituted with 1-3 substituents independently selected from halogen (preferably F or Cl), OH, CN, C1-4 alkyl optionally substituted with 1-3 F, C1-4 heteroalkyl optionally substituted with 1-3 F, and 3-4 membered ring (including cyclopropyl, cyclobutyl, oxetanyl, azetidinyl, etc.) optionally substituted with F and/or methyl.
Embodiment 36. The compound of Embodiment 34, or a pharmaceutically acceptable salt thereof, wherein R30 is a phenyl which is substituted with 1-3 (e.g., 1 or 2) R22, wherein R22 at each occurrence is independently F, Cl, CN, RM2, ORM2, SRM2, or SF5, wherein RM2 at each occurrence is independently a C1-4 alkyl optionally substituted with 1-3 F, such as CF3.
Embodiment 37. The compound of any of Embodiments 1-31, or a pharmaceutically acceptable salt thereof, wherein R30 is optionally substituted 5 or 6-membered heteroaryl, such as a pyridyl
or pyrimidinyl
which is substituted with 1-3 (e.g., 1 or 2) R22, wherein R22 at each occurrence is independently halogen (e.g., F, Cl, or Br), CN, OH, NH2, RM, ORM, COOH, CONH2, COORM, CONH(RM), CON(RM)2, NHCO(RM), N(RM)CO(RM), SO2RM, SO2NH2, S(O)(NH)RM, S(O)(NRM)RM, SO2N(RM)2, NHSO2RM, N(RM)SO2RM, P(O)(RM)2, P(O)(ORM)2, NH(RM), N(RM)2, SRM, or SF5, wherein RM at each occurrence is independently an optionally substituted C1-4 alkyl, optionally substituted C2-4 alkenyl, optionally substituted C2-4 alkynyl, optionally substituted C3-6 cycloalkyl, optionally substituted phenyl, optionally substituted 5 or 6 membered heteroaryl having 1-4 ring heteroatoms independently selected from O, S, and N, or optionally substituted 4-7 membered heterocyclyl.
Embodiment 38. The compound of Embodiment 37, or a pharmaceutically acceptable salt thereof, wherein R30 is a 5-membered heteroaryl
or pyridyl
or pyrimidinyl
which is substituted with 1-3 (e.g., 1 or 2) R22, as valency permits, wherein R22 at each occurrence is independently halogen (e.g., F, Cl, or Br), CN, OH, RM1 ORM1, SO2RM1, P(O)(RM1)2, SRM1, or SF5, wherein RM1 at each occurrence is independently an optionally substituted C1-4 alkyl or an optionally substituted 3-4 membered ring (including cyclopropyl, cyclobutyl, oxetanyl, azetidinyl, etc.), wherein when substituted, the optionally substituted C1-4 alkyl or 3-4 membered ring is substituted with 1-3 substituents independently selected from halogen (preferably F or Cl), OH, CN, C1-4 alkyl optionally substituted with 1-3 F, C1-4 heteroalkyl optionally substituted with 1-3 F, and 3-4 membered ring (including cyclopropyl, cyclobutyl, oxetanyl, azetidinyl, etc.) optionally substituted with F and/or methyl.
Embodiment 39. The compound of Embodiment 37, or a pharmaceutically acceptable salt thereof, wherein R30 is a 5-membered heteroaryl
or pyridyl
or pyrimidinyl
which is substituted with 1-3 (e.g., 1 or 2) R22, as valency permits, wherein R22 at each occurrence is independently F, Cl, CN, RM2, ORM2, SRM2, or SF5, wherein RM2 at each occurrence is independently a C1-4 alkyl optionally substituted with 1-3 F, such as CF3.
Embodiment 40. The compound of any of Embodiments 1-31, or a pharmaceutically acceptable salt thereof, wherein R30 has a structure according to S-1-A, S-1-B, S-1-C, or S-1-D:
wherein R8 is C1-4 alkyl optionally substituted with 1-3 F (such as CH3, CF3, etc.), C1-4 alkoxy optionally substituted with 1-3 F (such as OCH3, OCF3, etc.), SCF3, SF5, cyclopropyl, cyclobutyl, or
Embodiment 41. The compound of any of Embodiments 1-17, or a pharmaceutically acceptable salt thereof, wherein L1, L2, and R30 together (i.e., L1-L2-R30) are selected from:
or L1-L2-R30 is selected from
Embodiment 42. The compound of any of Embodiments 1-17, or a pharmaceutically acceptable salt thereof, wherein L1, L2, and R30 together are selected from:
or L1-L2-R30 is selected from:
Embodiment 43. The compound of any of Embodiments 1-17, or a pharmaceutically acceptable salt thereof, wherein L1, L2, and R30 together are selected from:
or L1-L2-R30 is selected from:
or L1, L2, and R30 together are selected from:
or L1, L2, and R30 together are selected from:
Embodiment 44. The compound of any of Embodiments 1-31, or a pharmaceutically acceptable salt thereof, wherein R30 is selected from the following:
Embodiment 45. A compound selected from Compound Nos. 1-347 or any of the compounds disclosed in Table A, or a pharmaceutically acceptable salt thereof.
Embodiment 46. A pharmaceutical composition comprising the compound of any of Embodiments 1-45 or a pharmaceutically acceptable salt thereof and optionally a pharmaceutically acceptable excipient.
Embodiment 47. A method for treating a disease comprising the administration to a subject in need thereof a therapeutically-effective amount of at least one compound according to any one of Embodiments 1 to 45, wherein said disease is cancer or a viral infection.
Embodiment 48. The method according to Embodiment 47, wherein said cancer is selected from colon cancer, pancreatic cancer, breast cancer, prostate cancer, lung cancer, ovarian cancer, cervical cancer, renal cancer, cancer of the head and neck, lymphoma, leukemia and melanoma.
Embodiment 49. A method of inhibiting activity of at least one of diacylglycerol kinase selected from diacylglycerol kinase alpha (DGKa) and diacylglycerol kinase zeta (DGKz) comprising administering to a subject in need thereof a therapeutically effective amount of at least one compound according to any one of Embodiments 1 to 45.
It is to be understood that both the foregoing summary and the following detailed description are exemplary and explanatory only, and are not restrictive of the invention herein.
In a broad aspect, the present disclosure provides compounds and compositions that are useful for inhibiting DGKs (diacylglycerol kinases), such as DGKa and/or DGKz, and/or treating or preventing various diseases or disorders described herein, e.g., cancer or infectious diseases such as viral infections.
Some embodiments of the present disclosure are directed to novel compounds. The compounds herein typically can be a DGK inhibitor, and useful for treating various diseases or disorders, such as those described herein, e.g., cancer or viral infections.
In some embodiments, the present disclosure provides a compound of Formula III-1, III-2, III-3, III-4, III-5, III-6, III-7, III-8, III-9, III-10, III-11, or III-12, or a pharmaceutically acceptable salt thereof:
wherein:
As used herein, unless specified or otherwise obviously contrary from context, a “x-y membered ring” should be understood as encompassing any ring structure having the designated number of ring members, for example, such ring can be carbocyclic, heterocyclic, aryl or heteroaryl, which can be monocyclic, bicyclic, or having more than two rings, and each of the ring(s) can be saturated, partially unsaturated, or aromatic, and can optionally contain one or more ring heteroatoms. Further, when applicable, such “x-y membered ring” should be understood as attaching to the remainder of the molecule through one or more ring atoms.
To be clear, the term “substituted” or “unsubstituted” used in connection with a variable described herein should be understood as referring to whether the variable is substituted, without considering the required bonding of the variable with the remainder of the molecule. For example, an unsubstituted phenylene should be understood such that other than the two ring atoms of the phenylene that are attached to the remainder of the molecule, the other ring atoms of the phenylene are not substituted, whereas in a substituted phenylene, at least one ring atom, other than the two ring atoms of the phenylene that are attached to the remainder of the molecule, is substituted with a substituent described herein.
In some embodiments, the compound of Formula III-1 to III-12 can have stereoisomer(s). In such embodiments, the compound of Formula III-1 to III-12 can exist in the form of an individual enantiomer, diastereomer, atropisomer, and/or geometric isomer, as applicable, or a mixture of stereoisomers, including racemic mixtures and mixtures enriched in one or more stereoisomers. In some embodiments, when applicable, the compound of Formula III-1 to III-12 can exist as a mixture of a pair of enantiomers in any ratio, including a racemic mixture with a ratio of 1:1. In some embodiments, when applicable, the compound of Formula III-1 to III-12 can exist as an isolated or enriched individual enantiomer substantially free (e.g., with less than 20%, less than 10%, less than 5%, less than 1%, by weight, by HPLC or SFC area, or both, or with a non-detectable amount) of the other enantiomer.
In some embodiments, the present disclosure provides a compound having a structure according to Formula III-1, or a pharmaceutically acceptable salt thereof, with the variables defined and preferred herein. In some embodiments, the present disclosure provides a compound having a structure according to Formula III-2, or a pharmaceutically acceptable salt thereof, with the variables defined and preferred herein. In some embodiments, the present disclosure provides a compound having a structure according to Formula III-3, or a pharmaceutically acceptable salt thereof, with the variables defined and preferred herein. In some embodiments, the present disclosure provides a compound having a structure according to Formula III-4, or a pharmaceutically acceptable salt thereof, with the variables defined and preferred herein. In some embodiments, the present disclosure provides a compound having a structure according to Formula III-5, or a pharmaceutically acceptable salt thereof, with the variables defined and preferred herein. In some embodiments, the present disclosure provides a compound having a structure according to Formula III-6, or a pharmaceutically acceptable salt thereof, with the variables defined and preferred herein. In some embodiments, the present disclosure provides a compound having a structure according to Formula III-7, or a pharmaceutically acceptable salt thereof, with the variables defined and preferred herein. In some embodiments, the present disclosure provides a compound having a structure according to Formula III-8, or a pharmaceutically acceptable salt thereof, with the variables defined and preferred herein. In some embodiments, the compound of Formula III-8 can be characterized as having a structure according to Formula III-8-A or Formula III-8-B:
wherein the variables R1, R2, U, X, Y, L1, L2, and R30 are defined herein, such as those described herein below in connection with Formula III-2 in enumerated embodiments A2-A36. In some embodiments, the present disclosure provides a compound having a structure according to Formula III-9, or a pharmaceutically acceptable salt thereof, with the variables defined and preferred herein. In some embodiments, the present disclosure provides a compound having a structure according to Formula III-10, or a pharmaceutically acceptable salt thereof, with the variables defined and preferred herein. In some embodiments, the present disclosure provides a compound having a structure according to Formula III-11, or a pharmaceutically acceptable salt thereof, with the variables defined and preferred herein. In some embodiments, the present disclosure provides a compound having a structure according to Formula III-12, or a pharmaceutically acceptable salt thereof, with the variables defined and preferred herein.
It should be also understood that the definitions and preferred definitions described herein for a variable(s) in any of Formula III-1 to III-12 are also applicable for the respective variable(s) in any of its respective subformulae, unless specifically defined in such subformulae or otherwise contradictory. For example, the definitions and preferred definitions described herein for a variable(s) in Formula III-2, are also applicable to the respective variable(s) in a subformula herein, such as Formula III-2-A, III-2-B, III-2-C, III-2-C-1, III-2-C-2, III-2-C-3, III-2-C-4, III-2-D, III-2-D-1, III-2-D-2, III-2-D-3, III-2-D-4, III-2-E-1, III-2-E-2, III-2-E-3, III-2-E-1a, III-2-E-2a, III-2-E-3a, III-2-E-1b, III-2-E-2b, III-2-E-3b, III-2-F-1a, III-2-F-1b, III-2-F-2a, III-2-F-2b, III-2-F-3a, or III-2-F-3b, unless specifically defined in such subformula or otherwise contradictory. Similarly, the definitions and preferred definitions described herein for a variable(s) in Formula III-4, are also applicable to the respective variable(s) in a subformula herein, such as III-4-A, III-4-B, III-4-C, III-4-D, III-4-C-1, III-4-C-2, III-4-C-3, III-4-C-4, III-4-D-1, III-4-D-2, III-4-C-4a, III-4-C-4b, III-4-C-4c, III-4-C-4d, III-4-C-4e, or III-4-C-4f, unless specifically defined in such subformula or otherwise contradictory.
Typically, in Formula III-2, III-3, III-4, III-5, III-6, III-7, III-8, III-9, III-10, III-11, or III-12, Y is N.
In some embodiments, in Formula III-2, III-3, III-4, III-5, III-6, III-7, III-8, III-9, III-10, III-11, or III-12, Y is CR7, wherein R7 is hydrogen, F, Cl, CN, optionally substituted C1-4 alkyl, or optionally substituted C1-4 heteroalkyl. For example, in some embodiments, Y is CH.
Typically, U in Formula III-1, III-2, III-3, III-4, III-5, III-6, III-7, III-8, III-9, III-10, III-11, or III-12, is CR6, wherein R6 is hydrogen, halogen (e.g., F, Cl, or Br), CN, OH, NH2, RD, ORD, NH(RD), N(RD)2, COOH, CONH2, COORD, CONH(RD), CON(RD)2, SRD, SORD, SO2RD or P(O)(RD)2, wherein RD at each occurrence is independently an optionally substituted C1-4 alkyl, optionally substituted C2-4 alkenyl, optionally substituted C2-4 alkynyl, optionally substituted C3-6 cycloalkyl, optionally substituted 5 or 6 membered heteroaryl having 1-4 ring heteroatoms independently selected from O, S, and N, or optionally substituted 4-7 membered heterocyclyl. In some preferred embodiments, R6 is hydrogen, i.e., U is CH. In some embodiments, R6 can also be halogen, such as F or Cl. In some embodiments, R6 can also be ORD, wherein RD is defined herein. In some embodiments, R6 can be ORD1, wherein RD1 is optionally substituted C1-4 alkyl, optionally substituted C3-6 cycloalkyl, or optionally substituted 4-7 membered heterocyclyl having 1 or 2 ring heteroatoms independently selected from O, N, and S; wherein, when substituted, the optionally substituted C1-4 alkyl, C3-6 cycloalkyl, or 4-7 membered heterocyclyl can be substituted with one or more (e.g., 1-3) substituents, for example, each substituent can be independently selected from halogen (e.g., F), OH, CN, C1-4 alkyl or C1-4 alkoxy. For example, in some embodiments, R6 can be C1-4 alkoxy. In some specific embodiments, R6 can be
In some specific embodiments, R6 can be
In some specific embodiments, R6 can be
In some embodiments, in Formula III-1, III-2, III-3, III-4, III-5, III-6, III-7, III-8, III-9, III-10, III-11, or III-12, U is N.
Various groups are suitable as R1 for Formula III-2, III-3, III-4, III-5, III-6, III-7, III-8, III-9, III-10, III-11, or III-12. For example, in some embodiments, R1 can be OH, COOH, CONH2, NH2, RA, ORA, COORA, NH(RA), N(RA)2, CONH(RA), CON(RA)2, SRA, SORA, SO2RA, or P(O)(RA)2, wherein RA is defined herein. In some embodiments, RA at each occurrence can be optionally substituted C1-4 alkyl.
In some preferred embodiments, in Formula III-2, III-3, III-4, III-5, III-6, III-7, III-8, III-9, III-10, III-11, or III-12, R1 is CN.
In some preferred embodiments, R1 in Formula III-2, III-3, III-4, III-5, III-6, III-7, III-8, III-9, III-10, III-11, or III-12, is halogen, preferably F or Cl.
In some embodiments, R1 in Formula III-2, III-3, III-4, III-5, III-6, III-7, III-8, III-9, III-10, III-11, or III-12, is a C2-3 alkynyl, such as
In some embodiments, R1 in in Formula III-2, III-3, III-4, III-5, III-6, III-7, III-8, III-9, III-10, I-11, or III-12, is optionally substituted 5 or 6 membered heteroaryl having 1-4 ring heteroatoms independently selected from O, S, and N. For example, in some embodiments, R1 can be an optionally substituted pyrimidinyl or an optionally substituted thiazolyl, for example, R1 is
In some embodiments, R1 in Formula III-2, III-3, III-4, III-5, III-6, III-7, III-8, III-9, III-10, III-11, or III-12, is SRA, wherein RA is defined herein. For example, in some embodiments, R1 can be SRA1, wherein RA1 is independently optionally substituted C1-4 alkyl, for example, R1 is SCH3. Other suitable R1 are described herein.
In some embodiments, R1 in Formula III-2, III-3, III-4, III-5, III-6, III-7, III-8, III-9, III-10, III-1, or III-12, can be characterized in that (i) R1 is halogen, e.g., F or Cl; (ii) R1 is ; (iii) R1 is an optionally substituted 5 or 6 membered heteroaryl having 1-4 ring heteroatoms independently selected from O, S, and N, such as an optionally substituted pyrimidinyl or an
optionally substituted thiazolyl, for example, R1 is N or N; or (iv) R1 is SRA1, wherein RA1 is independently optionally substituted C1-4 alkyl, for example, R1 is SCH3.
Typically, R2 in Formula III-1, III-2, III-3, III-4, III-6, III-7, or III-8, is hydrogen. In some embodiments, R2 can be halogen (e.g., F, Cl, or Br), CN, OH, NH2, RB, ORB, NH(RB), or N(RB)2, wherein RB is defined herein. In some embodiments, RB can be an optionally substituted C1-4 alkyl or optionally substituted 3-4 membered cycloalkyl.
Typically, R3 in Formula III-1, III-3, III-4, III-6, or III-7, is an optionally substituted C1-4 alkyl, such as CH3, CD3 etc. In some preferred embodiments, R3 is CH3. In some preferred embodiments, R3 is CD3. In some preferred embodiments, R3 is CH2CH3. In some preferred embodiments, R3 is CH(CH3)2. In some preferred embodiments, R3 is CH2CHF2. In some embodiments, R3 can also be an optionally substituted C3-6 cycloalkyl. In some preferred embodiments, R3 is cyclopropyl. In some preferred embodiments, R3 is cyclobutyl. In some preferred embodiments, R3 is hydrogen.
Typically, Z in Formula III-1, III-3, III-4, III-5, III-6, III-7, III-9, III-11, or III-12, is C(O).
In some embodiments, Z in Formula III-1, III-3, III-4, III-5, III-6, III-7, III-9, 11-11, or III-12, can also be S(O)2.
In some typical embodiments, X in Formula III-1, III-2, III-5, III-6, III-7, III-8, or III-9, is N.
In some embodiments, X in Formula III-1, III-2, III-5, III-8, or III-9, is CR5, wherein R5 is defined herein. In some embodiments, X is CR5, wherein R5 is CN. In some embodiments, X is CR5, wherein R5 is CONH2, COOH, CONH(RC1), CON(RC1)2, or COORC1, wherein RC1 at each occurrence is independently a C1-4 alkyl, such as methyl.
In some embodiments, n1 is 0. In some embodiments, n1 is 1. In some embodiments, R9 at each occurrence is independently CH3, CH2CH3, CH2CH2CH3, fluorine-substituted C1-3 alkyl (e.g., CF2H), cyclopropyl, or cyclobutyl, CH2OH, CH2OCH3, CH2OCH2CH3, CH2NH2, CH2N3, or CH2NHC(O)OCH3.
In some embodiments, in Formula III-6 or III-7, J is NR15, wherein R15 is defined herein. For example, in some embodiments, J is N(C1-4 alkyl), such as NCH3.
In some embodiments, in Formula III-6 or III-7, J is an optionally substituted methylene. For example, in some embodiments, J is CH2. In some embodiments, J is CF2, CHCH3, C(CH3)2, CHOH, etc.
In some embodiments, in Formula III-6 or III-7, J is O. In some embodiments, in Formula III-6 or III-7, J is NH or NCH3.
In some embodiments, in Formula III-6 or III-7, W is absent. Thus, the ring containing J is a 5 membered ring.
In some embodiments, in Formula III-6 or III-7, p is 1 and W is W1, wherein W1 is defined herein. For example, in some embodiments, W1 is C(O), SO2, CH2, CF2, CHCH3, C(CH3)2, CHOH, etc. In such embodiments, the ring containing J and W is a 6-membered ring.
In some embodiments, in Formula III-6 or III-7, p is 2 and W is —W1—W1—, wherein each W1 is defined herein. For example, in some embodiments, each W1 is independently C(O), SO2, CH2, CF2, CHCH3, C(CH3)2, CHOH, etc. In some embodiments, both W1 can be CH2. In some embodiments, one W1 is O, and the other W1 is CH2. In some embodiments, one W1 is C(O), and the other W1 is O, NH, NCH3, or CH2. In such embodiments, the ring containing J and W is a 7-membered ring.
Typically, in Formula III-1, III-2, III-5, III-8, or III-9, L1 is an optionally substituted 4-12 membered heterocyclylene herein. In some embodiments, L1 in Formula III-1, III-2, III-5, III-8, or III-9, is an optionally substituted 4-12 membered heterocyclylene having one or more rings and 1-4 ring heteroatoms each independently O, N, or S. In some embodiments, L1 in Formula III-1, III-2, III-5, III-8, or III-9, can be —N(RE)—, wherein RE is optionally substituted C1-4 alkyl or optionally substituted 3-7 membered ring. For example, in some embodiments, L1 is —N(C1-4 alkyl)-, such as —N(CH3)—.
In some embodiments, L1 in Formula III-1, III-2, III-5, III-8, or III-9, is an optionally substituted 4-7 membered monocyclic heterocyclylene having 1 or 2 ring heteroatoms each independently O, N, or S. In some embodiments, the 4-7 membered monocyclic heterocyclylene is a saturated heterocyclylene having 1 or 2 ring heteroatoms, such as 1 or 2 ring nitrogen atoms, such as a piperazine or piperidine ring. The heterocyclylene is typically attached to the remainder of the molecule through two ring nitrogen atoms or one ring nitrogen and one ring carbon atom.
In some embodiments, in Formula III-1, III-2, III-5, III-8, or III-9, L1 can be
(either of the two attaching points of the piperidine, N or C, can be attached to L2),
In some embodiments, R9A at each occurrence is independently RN. In some embodiments, R9A at each occurrence is independently an optionally substituted C1-4 alkyl, and when substituted, the substituents can be independently selected from F, OH, C1-4 heteroalkyl having 1 or 2 heteroatoms, which is optionally substituted with F, or 3-4 membered ring (e.g., cyclopropyl or cyclobutyl, etc.). In some embodiments, R9A at each occurrence is independently CH3, CH2CH3, CH2CH2CH3, fluorine-substituted C1-3 alkyl (e.g., CF2H), cyclopropyl, or cyclobutyl, CH2OH, CH2OCH3, CH2OCH2CH3, CH2NH2, CH2N3, or CH2NHC(O)OCH3. Preferably, R9A at each occurrence is independently CH3, CH2CH3, or CH2CH2CH3, more preferably, R9A at each occurrence is CH2CH3. Typically, in Formula III-1, III-2, III-5, III-8, III-9, III-10, III-11, or III-12, n is 0, 1, or 2. For example, L1 can be selected from:
wherein either of the two attaching points can be attached to L2.
In some preferred embodiments, in Formula III-1, III-2, III-5, III-8, or III-9, L1 can be selected from the following (the bottom attaching point, N or C, is attached to L2):
In some preferred embodiments, in Formula III-1, III-2, III-5, III-8, or III-9, L1 can be selected from the following (the bottom attaching point, N or C, is attached to L2):
In some embodiments, L1 in Formula III-1, III-2, III-5, III-8, or III-9 can also be an optionally substituted 7-12 membered heterocyclylene having two or more rings and 1-4 ring heteroatoms each independently O, N, or S, when substituted, the substituent(s) can be attached to any one or more of the two or more rings. For example, in some embodiments, L1 can be an optionally substituted 8-12 membered fused, spiro, or bridged bicyclic heterocyclylene having 1-4 ring heteroatoms each independent O, N, or S, wherein each ring of the bicyclic heterocyclylene can be saturated, partially unsaturated, or aromatic, and each ring of the bicyclic heterocyclylene can have 0, 1, 2, or 3 ring heteroatoms, provided that the bicyclic heterocyclylene as a whole is not fully aromatic and the total number of heteroatoms in the bicyclic heterocyclylene does not exceed 4. In some embodiments, L1 can be an optionally substituted 8-11 membered fused bicyclic heterocyclylene having 1-3 ring heteroatoms each independent O, N, or S, wherein (1) one of the two fused rings is phenyl or 5 or 6 membered heteroaryl, and (2) the other of the two fused rings is a 5-7 membered heterocycle having one or two ring heteroatoms each independently O, N, or S, preferably, the 5-7 membered heterocycle has at least one ring nitrogen atom. In some embodiments, L1 can be an optionally substituted 8-11 (e.g., 8, 9, or 10) membered spiro bicyclic heterocyclylene having 1-4 ring heteroatoms each independent O, N, or S, wherein (1) one of the two spiro rings is a 5-7 membered heterocycle having one or two ring heteroatoms each independently O, N, or S, preferably, the 5-7 membered heterocycle has one ring nitrogen atom, and (2) the other of the two spiro rings is a 4-6 membered heterocycle having 1-3 ring heteroatoms each independently O, N, or S, for example, one of the two spiro rings is a pyrrolidine, piperidine, azepane ring and the other of the two spiro rings is azetidine, pyrrolidine, pyrrolidinone, piperidinone, oxazoline, isoxazoline, thiazoline, isothiazoline, etc.
For example, in some preferred embodiments, L1 in Formula III-1, III-2, III-5, III-8, or III-9 can be selected from the following (the bottom attaching point, N or C, is attached to L2):
wherein in each of the bicyclic heterocyclylene, each of the two rings can be optionally substituted with 1-3 R10, wherein R10 at each occurrence is independently halogen, OH, NH2, oxo (as applicable), RJ, ORJ, CN, NH(R1), or N(R1)2, wherein R1 at each occurrence is independently an optionally substituted C1-4 alkyl, optionally substituted C2-4 alkenyl, optionally substituted C2-4 alkynyl, or optionally substituted 3-4 membered ring.
In some preferred embodiments, L1 in Formula III-1, III-2, III-5, III-8, or III-9 can be selected from the following (the bottom attaching point, N or C, is attached to L2):
In some embodiments, L1 in Formula III-1, III-2, III-5, III-8, or III-9 can also be —N(C1-4 alkyl)-, such as —N(CH3)—.
In some embodiments, in Formula III-1, III-2, III-3, III-4, III-5, III-6, III-7, III-8, III-9, III-10, III-11, or III-12, L2 can be absent.
In some embodiments, L2 in Formula III-1, III-2, III-3, III-4, III-5, III-6, III-7, III-8, III-9, III-10, III-11, or III-12 can be an optionally substituted C1-4 alkylene, such as those substituted with one or more R3 groups defined herein. In some embodiments, R13 at each occurrence is independently selected from halogen (e.g., F, Cl, or Br), CN, OH, NH2, COOH, CONH2, SO2NH2, CORK, COORK, CONH(RK), CON(RK)2, SO2RK, SO2NH(RK), SO2N(RK)2, RK, ORK, NH(RK), N(RK)2, SRK, SORK, SO2RK, or P(O)(RK)2, wherein RK at each occurrence is independently optionally substituted C1-4 alkyl, optionally substituted C2-4 alkenyl, optionally substituted C2-4 alkynyl, optionally substituted C1-4 heteroalkyl, optionally substituted C3-6 cycloalkyl, optionally substituted phenyl, optionally substituted 5 or 6 membered heteroaryl having 1-4 ring heteroatoms independently selected from O, S, and N, or optionally substituted 4-7 membered heterocyclyl.
In some embodiments, L2 in Formula III-1, III-2, III-3, III-4, III-5, III-6, III-7, III-8, III-9, III-10, III-11, or III-12 can be an unsubstituted C1-4 alkylene such as CH2, CH(CH3), CH(C2H5), CH(C3H7), etc.
In some embodiments, L2 in Formula III-1, III-2, III-3, III-4, III-5, III-6, III-7, III-8, III-9, III-10, III-11, or III-12 can be an unsubstituted C1-4 alkylene selected from: CH2, or
In some embodiments, L2 in Formula III-1, III-2, III-3, III-4, III-5, III-6, III-7, III-8, III-9, III-10, III-11, or III-12 can be an optionally substituted C1-4 alkylene selected from:
wherein:
In some embodiments, R13 is OH, methoxy, cyclopropyl, phenyl,
In some embodiments, in Formula III-1, III-2, III-3, III-4, III-5, III-6, III-7, III-8, III-9, III-10, III-11, or III-12, L2 can be C(O). In some embodiments, in Formula III-1, III-2, III-3, III-4, III-5, III-6, III-7, III-8, III-9, III-10, III-11, or III-12, L2 can be O, preferably, in Formula III-4, L2 is not O.
In some embodiments, in Formula III-1, III-2, III-3, III-4, III-5, III-6, III-7, III-8, III-9, III-10, III-11, or III-12, L2 can be an optionally substituted phenylene, e.g.,
In some embodiments, in Formula III-1, III-2, III-3, III-4, III-5, III-6, III-7, III-8, III-9, III-10, III-11, or III-12, L2 can be an optionally substituted 5 or 6 membered heteroarylene, for example,
In some embodiments, in Formula III-1, III-2, III-5, III-8, or III-9, L1 can be —N(RE)—, L2 is an optionally substituted phenylene, e.g.,
and R30 is hydrogen. For example, in some embodiments, L1, L2, and R30 together are:
In some embodiments, in Formula III-1, III-2, III-3, III-4, III-5, III-6, III-7, III-8, III-9, III-10, III-11, or III-12, R30 is optionally substituted phenyl, such as a phenyl which is substituted with 1-3 (e.g., 1 or 2) R22, wherein R22 at each occurrence is independently halogen (e.g., F, Cl, or Br), CN, OH, NH2, RM, ORM, COOH, CONH2, COORM, CONH(RM), CON(RM)2, NHCO(RM), N(RM)CO(RM), SO2RM, SO2NH2, S(O)(NH)RM, S(O)(NRM)RM, SO2N(RM)2, NHSO2RM, N(RM)SO2RM, P(O)(RM)2, P(O)(ORM)2, NH(RM), N(RM)2, SRM, or SF5, wherein RM at each occurrence is independently an optionally substituted C1-4 alkyl, optionally substituted C2. 4 alkenyl, optionally substituted C2-4 alkynyl, optionally substituted C3-6 cycloalkyl, optionally substituted phenyl, optionally substituted 5 or 6 membered heteroaryl having 1-4 ring heteroatoms independently selected from O, S, and N, or optionally substituted 4-7 membered heterocyclyl. In some embodiments, one instance of R22 is halogen, such as F, Cl, or Br, and the remaining instance(s) of R22, if any, are defined herein. In some embodiments, two or three instances of R22 are halogen, each independently F, Cl, or Br, and the remaining instance(s) of R22, if any, are defined herein. In some embodiments, one instance of R22 is CN, and the remaining instance(s) of R22, if any, are defined herein. In some embodiments, one instance of R22 is RM, ORM, SRM, SF5, or SO2RM, wherein RM is defined herein, such as a C1-3 alkyl optionally substituted with F (e.g., CF3, CF2CH3, CHF2, etc.) or cyclopropyl, and the remaining instance(s) of R22, if any, are defined herein. For example, in some embodiments, R30 is a phenyl which is substituted with 1-3 (e.g., 1 or 2) R22, wherein R22 at each occurrence is independently halogen (e.g., F, Cl, or Br), CN, OH, RM1, ORM1, SO2RM1, P(O)(RM1)2, SRM1, or SF5, wherein RM1 at each occurrence is independently an optionally substituted C1-4 alkyl or an optionally substituted 3-4 membered ring (including cyclopropyl, cyclobutyl, oxetanyl, azetidinyl, etc.), wherein when substituted, the optionally substituted C1-4 alkyl or 3-4 membered ring is substituted with 1-3 substituents independently selected from halogen (preferably F or Cl), OH, CN, C1-4 alkyl optionally substituted with 1-3 F, C1-4 heteroalkyl optionally substituted with 1-3 F, and 3-4 membered ring (including cyclopropyl, cyclobutyl, oxetanyl, azetidinyl, etc.) optionally substituted with F and/or methyl. More preferably, R30 is a phenyl which is substituted with 1-3 (e.g., 1 or 2) R22, wherein R22 at each occurrence is independently F, Cl, CN, RM2, ORM2, SRM2, or SF5, wherein RM2 at each occurrence is independently a C1-4 alkyl optionally substituted with 1-3 F, such as CF3. In some embodiments, R22 at each occurrence can be independently halogen, CN, C1-4 alkyl optionally substituted with 1-3 F (such as CH3, CF3, etc.), C1-4 alkoxy optionally substituted with 1-3 F (such as OCH3, OCF3, etc.), SCF3, SF5,
cyclopropyl, cyclobutyl, or. In some preferred embodiments, R22 at each occurrence is independently F, Cl, Br, CF3, OCF3, SCF3, SF5, OMe, CN, methyl, CHF2, CF2CH3, cyclopropyl, CH3SO2, and OCH2-(cyclopropyl).
In some embodiments, in Formula III-1, III-2, III-3, III-4, III-5, III-6, III-7, III-8, III-9, III-10, III-11, or III-12, R30 is optionally substituted 5 or 6-membered heteroaryl, such as a pyridyl
or pyrimidinyl
which is substituted with 1-3 (e.g., 1 or 2) R22, wherein R22 at each occurrence is independently halogen (e.g., F, Cl, or Br), CN, OH, NH2, RM, ORM, COOH, CONH2, COORM, CONH(RM), CON(RM)2, NHCO(RM), N(RM)CO(RM), SO2RM, SO2NH2, S(O)(NH)RM, S(O)(NRM)RM, SO2N(RM)2, NHSO2RM, N(RM)SO2RM, P(O)(RM)2, P(O)(ORM)2, NH(RM), N(RM)2, SRM, or SF5, wherein RM at each occurrence is independently an optionally substituted C1-4 alkyl, optionally substituted C2-4 alkenyl, optionally substituted C2-4 alkynyl, optionally substituted C3-6 cycloalkyl, optionally substituted phenyl, optionally substituted 5 or 6 membered heteroaryl having 1-4 ring heteroatoms independently selected from O, S, and N, or optionally substituted 4-7 membered heterocyclyl. In some embodiments, one instance of R22 is halogen, such as F, Cl, or Br, and the remaining instance(s) of R22, if any, are defined herein. In some embodiments, two or three instances of R22 are halogen, each independently F, Cl, or Br, and the remaining instance(s) of R22, if any, are defined herein. In some embodiments, one instance of R22 is CN, and the remaining instance(s) of R22, if any, are defined herein. In some embodiments, one instance of R22 is RM, ORM, SRM, SF5, or SO2RM, wherein RM is defined herein, such as a C1-3 alkyl optionally substituted with F (e.g., CF3, CF2CH3, CHF2, etc.) or cyclopropyl, and the remaining instance(s) of R22, if an are defined herein. For example, in some embodiments, R30 is a 5-membered heteroaryl
or pyridyl
or pyrimidinyl
which is substituted with 1-3 (e.g., 1 or 2) R22, as valency permits, wherein R22 at each occurrence is independently halogen (e.g., F, Cl, or Br), CN, OH, RM1, ORM1, SO2RM1, P(O)(RM1)2, SRM1, or SF5, wherein RM1 at each occurrence is independently an optionally substituted C1-4 alkyl or an optionally substituted 3-4 membered ring (including cyclopropyl, cyclobutyl, oxetanyl, azetidinyl, etc.), wherein when substituted, the optionally substituted C1-4 alkyl or 3-4 membered ring is substituted with 1-3 substituents independently selected from halogen (preferably F or Cl), OH, CN, C1-4 alkyl optionally substituted with 1-3 F, C1-4 heteroalkyl optionally substituted with 1-3 F, and 3-4 membered ring (including cyclopropyl, cyclobutyl, oxetanyl, azetidinyl, etc.) optionally substituted with F and/or methyl. More preferably, R30 is a 5-membered heteroaryl
or pyridyl
or pyrimidinyl
which is substituted with 1-3 (e.g., 1 or 2) R22, as valency permits, wherein R22 at each occurrence is independently F, Cl, CN, RM2, ORM2, SRM2, or SF5, wherein RM2 at each occurrence is independently a C1-4 alkyl optionally substituted with 1-3 F, such as CF3. In some embodiments, R22 at each occurrence can be independently halogen, CN, C1-4 alkyl optionally substituted with 1-3 F (such as CH3, CF3, etc.), C1-4 alkoxy optionally substituted with 1-3 F (such as OCH3, OCF3, etc.), SCF3, SF5, cyclopropyl, cyclobutyl, or
In some preferred embodiments, R22 at each occurrence is independently F, Cl, Br, CF3, OCF3, SCF3, SF5, OMe, CN, methyl, CHF2, CF2CH3, cyclopropyl, CH3SO2, and OCH2-(cyclopropyl).
In some embodiments, in Formula III-1, III-2, III-3, III-4, III-5, III-6, III-7, III-8, III-9, III-10, III-11, or III-12, R30 can have a structure of formula S-1, S-2, S-3, or S-4 below:
wherein:
In some embodiments, Ring B in S-1, S-2, S-3, or S-4 contains no ring heteroatoms. In some embodiments, Ring B in S-1, S-2, S-3, or S-4 contains one ring heteroatom, such as O, N, or S. In some embodiments, Ring B in S-1, S-2, S-3, or S-4 contains no ring heteroatoms and is not substituted. In some embodiments, Ring B in S-1, S-2, S-3, or S-4 contains no ring heteroatoms and is optionally substituted with one or more substituents described herein, for example, in some embodiments, Ring B is optionally substituted with 1-3 RG1, wherein RG1 at each occurrence is independently F, OH, C1-4 alkyl optionally substituted with 1-3 F, or C1-4 alkoxy optionally substituted with 1-3 F.
The phenyl, pyridyl, or pyrimidyl portion of S-1, S-2, S-3, or S-4 is typically substituted with 1 R8, i.e., q is 1, wherein R8 is defined herein. In some embodiments, the phenyl, pyridyl, or pyrimidyl portion of S-1, S-2, S-3, or S-4 can be substituted with 2 R8, wherein R8 is defined herein. In some embodiments, R8 at each occurrence is independently RH1, ORH1, SRH1, SF5, or optionally substituted 5-8 membered carbocyclic having two or more rings, wherein RH1 is C1-4 alkyl optionally substituted with 1-3 F, C1-4 alkoxy optionally substituted with 1-3 F, or C3-6 cycloalkyl optionally substituted with 1-3 substituents independently selected from F, OH, methyl, and methoxy, and wherein when substituted, the optionally substituted 5-8 membered carbocyclic is substituted with 1-3 substituents each independently selected from halogen (e.g., F), OH, CN, C1-4 alkyl and C1-4 alkoxy. In some embodiments, R8 can be optionally substituted 5-8 membered carbocyclic having two or more rings, such as a bicyclic carbocyclic ring. For example, in some embodiments, R8 can be a 5-8 membered bicyclic carbocyclic, such as a bridged bicyclic carbocyclic, such as
which is optionally substituted, wherein, when substituted, the bicyclic carbocyclic can be substituted with one or more (e.g., 1-3) substituents, for example, each substituent can be independently selected from halogen (e.g., F), OH, CN, C1-4 alkyl or C1-4 alkoxy. In some embodiments, R8 can be
In some embodiments, R8 at each occurrence can be independently a C1-4 alkyl optionally substituted with 1-3 F (such as CH3, CF3, etc.), C1-4 alkoxy optionally substituted with 1-3 F (such as OCH3, OCF3, etc.), SCF3, SF5, cyclopropyl, cyclobutyl, or
For example, in some embodiments, R30 can have a structure according to S-1-A, S-1-B, S-1-C, or S-1-D:
wherein R8 is C1-4 alkyl optionally substitute with 1-3 F (such as CH3, CF3, etc.), C1-4 alkoxy optionally substituted with 1-3 F (such as OCH3, OCF3, etc.), SCF3, SF5, cyclopropyl, cyclobutyl, or
In some preferred embodiments, R30 in Formula III-1 to III-12 can be selected from the following:
In some preferred embodiments, R30 in Formula III-1 to III-12 can be selected from the following:
In some preferred embodiments, R30 in Formula III-1 to III-12 can be selected from the following:
In some preferred embodiments, R30 in Formula III-1 to III-12 can be selected from the following:
In some preferred embodiments, R30 in Formula III-1 to III-12 can be selected from the following:
In some preferred embodiments, R30 in Formula III-1 to III-12 can be selected from the following:
In some preferred embodiments, R30 in Formula III-1 to III-12 can be selected from the following:
In some preferred embodiments, R30 in Formula III-1 to III-12 can be selected from the following:
In some preferred embodiments, R30 in Formula III-1 to III-12 is not selected from the following:
In some preferred embodiments, R30 in Formula III-1 to III-12 can be selected from the following:
In some embodiments, in Formula III-1 to III-12, -L2-R30 can be:
In some embodiments, in Formula III-1 to III-12, -L2-R30 is not
In some embodiments, in Formula III-1 to III-12, -L2-R30 can be selected from:
In some embodiments, in Formula III-1 to III-12, -L2-R30 can be selected from:
In some embodiments, in Formula III-1 to III-12, -L2-R30 can be selected from:
In some embodiments, in Formula III-1 to III-12, -L2-R30 can be selected from:
In some embodiments, in Formula III-1 to III-12, -L2-R30 can be:
In some embodiments, in Formula III-1 to III-12, -L2-R30 is not
In some embodiments, in Formula III-1, III-2, III-5, III-8, or III-9, L1, L2, and R30 (i.e., L1-L2-R30) together can be selected from:
In some embodiments, in Formula III-1, III-2, III-5, III-8, or III-9, L1, L2, and R30 together can be selected from:
In some embodiments, in Formula III-1, III-2, III-5, III-8, or III-9, L1, L2, and R30 together can be selected from:
In some embodiments, in Formula III-1, III-2, III-5, III-8, or III-9, L1-L2-R30 is selected from the following:
In some embodiments, in Formula III-1, III-2, III-5, III-8, or III-9, L1, L2, and R30 together can be selected from:
In some embodiments, in Formula III-1, III-2, III-5, III-8, or III-9, L1, L2, and R30 together can be selected from:
In some embodiments, in Formula III-1, III-2, III-5, III-8, or III-9, L1, L2, and R30 together can be selected from:
In some embodiments, the present disclosure provides the following enumerated exemplified Embodiments A1-A38:
Embodiment A1. A compound of Formula III-2, or a pharmaceutically acceptable salt thereof,
Embodiment A2. The compound of Embodiment A1, or a pharmaceutically acceptable salt thereof, characterized as having a structure according to Formula III-2-A:
Embodiment A3. The compound of Embodiment A1, or a pharmaceutically acceptable salt thereof, characterized as having a structure according to Formula III-2-B:
Embodiment A4. The compound of any of Embodiments A1-A3, or a pharmaceutically acceptable salt thereof, wherein (i) R1 is CN; (ii) R1 is halogen, e.g., F or Cl; (iii) R1 is
(iv) R1 is an optionally substituted 5 or 6 membered heteroaryl having 1-4 ring heteroatoms independently selected from O, S, and N, such as an optionally substituted pyrimidinyl or an optionally substituted thiazolyl, for example, R1 is
or (v) R1 is SRA1, wherein RA1 is independently optionally substituted C1-4 alkyl, for example, R1 is SCH3.
Embodiment A5. The compound of any of Embodiments A1-A4, or a pharmaceutically acceptable salt thereof, wherein L1 is selected from the following (the bottom attaching point, N or C, is attached to L2)
Embodiment A6. The compound of any of Embodiments A1-A4, or a pharmaceutically acceptable salt thereof, wherein L1 is selected from the following (the bottom attaching point, N or C, is attached to L2):
Embodiment A7. The compound of any of Embodiments A1-A4, or a pharmaceutically acceptable salt thereof, characterized as having a structure according to Formula II-2-C:
wherein n is 0, 1, 2, or 3, and R9A is defined herein.
Embodiment A8. The compound of Embodiment A7, or a pharmaceutically acceptable salt thereof, characterized as having a structure according to Formula III-2-C-1, III-2-C-2, III-2-C-3, or III-2-C-4:
wherein the two R9A in Formula III-2-C-3 are independently selected.
Embodiment A9. The compound of Embodiment A7 or A8, wherein R9A at each occurrence is independently CH3, CH2CH3, CH2CH2CH3, fluorine-substituted C1-3 alkyl (e.g., CF2H), cyclopropyl, cyclobutyl, CH2OH, CH2OCH3, CH2OCH2CH3, CH2NH2, CH2N3, or CH2NHC(O)OCH3.
Embodiment A10. The compound of any of Embodiments A1-A4, or a pharmaceutically acceptable salt thereof, characterized as having a structure according to Formula III-2-D:
wherein n is 0, 1, 2, or 3, and R9A is defined herein.
Embodiment A11. The compound of Embodiment A10, or a pharmaceutically acceptable salt thereof, characterized as having a structure according to Formula III-2-D-1, III-2-D-2, III-2-D-3, or III-2-D-4:
wherein the two R9A in Formula III-2-D-3 are independently selected.
Embodiment A12. The compound of Embodiment A10 or A11, or a pharmaceutically acceptable salt thereof, wherein R9A at each occurrence is independently CH3, CH2CH3, CH2CH2CH3, fluorine-substituted C1-3 alkyl (e.g., CF2H), cyclopropyl, cyclobutyl, CH2OH, CH2OCH3, CH2OCH2CH3, CH2NH2, CH2N3, or CH2NHC(O)OCH3.
Embodiment A13. The compound of any of Embodiments A1-A12, or a pharmaceutically acceptable salt thereof, characterized as having a structure according to Formula III-2-E-1, III-2-E-2, or III-2-E-3:
wherein R13 is defined herein.
Embodiment A14. The compound of any of Embodiments A1-A12, or a pharmaceutically acceptable salt thereof, characterized as having a structure according to Formula III-2-E-1a, III-2-E-2a, II-2-E-3a, III-2-E-1b, III-2-E-2b, or III-2-E-3b:
wherein R13 is e me herein.
Embodiment A15. The compound of Embodiment A13, or a pharmaceutically acceptable salt thereof, characterized as having a structure according to Formula III-2-F-1a, III-2-F-1b, III-2-F-2a, III-2-F-2b, III-2-F-3a, or II-2-F-3b:
Embodiment A16. The compound of any of Embodiments A13-A15, or a pharmaceutically acceptable salt thereof, wherein R13 is hydrogen, C1-4 alkyl, e.g., methyl, isopropyl, etc., CN, OH, COOH, CONH2, methoxy, ethoxy, cyclopropyl, cyclobutyl, optionally substituted phenyl, or optionally substituted 5 or 6 membered heteroaryl having 1-3 ring heteroatoms independently selected from N, O, and S, wherein, when substituted, the optionally substituted phenyl or 5 or 6 membered heteroaryl is substituted with 1-3 substituents independently selected from halogen (preferably F or Cl), OH, CN, C1-4 alkyl optionally substituted with 1-3 F, C1-4 heteroalkyl optionally substituted with 1-3 F, and 3-4 membered ring (including cyclopropyl, cyclobutyl, oxetanyl, azetidinyl, etc.) optionally substituted with F and/or methyl, more preferably, R13 is hydrogen.
Embodiment A17. The compound of any of Embodiments A1-A12, or a pharmaceutically acceptable salt thereof, wherein L2 is absent, O, or C(O).
Embodiment A18. The compound of any of Embodiments A1-A17, or a pharmaceutically acceptable salt thereof, wherein R30 is an optionally substituted phenyl or 5 or 6-membered heteroaryl (e.g., such as oxadiazolyl
pyridyl
or pyrimidinyl
for example, when substituted, the phenyl or 5 or 6-membered heteroaryl can be substituted with 1-3 (e.g., 1 or 2) R22, wherein R22 at each occurrence is halogen (e.g., F, Cl, or Br), CN, C1-4 alkyl optionally substituted with 1-3 F (such as CH3, CF3, etc.), C1-4 alkoxy optionally substituted with 1-3 F (such as OCH3, OCF3, etc.), SCF3, SF5, cyclopropyl, cyclobutyl, or
Embodiment A19. The compound of any of Embodiments A1-A17, or a pharmaceutically acceptable salt thereof, wherein R30 is a phenyl which is substituted with 1-3 (e.g., 1 or 2) R22, wherein R22 at each occurrence is independently halogen (e.g., F, Cl, or Br), CN, OH, RM1, ORM1, SO2RM1, P(O)(RM1)2, SRM1, or SF5, wherein RM1 at each occurrence is independently an optionally substituted C1-4 alkyl or an optionally substituted 3-4 membered ring (including cyclopropyl, cyclobutyl, oxetanyl, azetidinyl, etc.), wherein when substituted, the optionally substituted C1-4 alkyl or 3-4 membered ring is substituted with 1-3 substituents independently selected from halogen (preferably F or Cl), OH, CN, C1-4 alkyl optionally substituted with 1-3 F, C1-4 heteroalkyl optionally substituted with 1-3 F, and 3-4 membered ring (including cyclopropyl, cyclobutyl, oxetanyl, azetidinyl, etc.) optionally substituted with F and/or methyl, preferably, R22 at each occurrence is independently F, Cl, Br, CF3, OCF3, SCF3, SF5, OMe, CN, methyl, CHF2, CF2CH3, cyclopropyl, CH3SO2, and OCH2-(cyclopropyl).
Embodiment A20. The compound of any of Embodiments A1-A17, or a pharmaceutically acceptable salt thereof, wherein R30 is a phenyl which is substituted with 1-3 (e.g., 1 or 2) R22, wherein R22 at each occurrence is independently F, Cl, CN, RM2, ORM2, SRM2, or SF5, wherein RM2 at each occurrence is independently a C1-4 alkyl optionally substituted with 1-3 F, such as CF3.
Embodiment A21. The compound of any of Embodiments A1-A17, or a pharmaceutically acceptable salt thereof, wherein R30 is a 5-membered heteroaryl
or pyridyl
or pyrimidinyl
which is substituted with 1-3 (e.g., 1 or 2) R22, as valency permits, wherein R22 at each occurrence is independently halogen (e.g., F, Cl, or Br), CN, OH, RM1, ORM1, SO2RM1, P(O)(RM1)2, SRM1, or SF5, wherein RM1 at each occurrence is independently an optionally substituted C1-4 alkyl or an optionally substituted 3-4 membered ring (including cyclopropyl, cyclobutyl, oxetanyl, azetidinyl, etc.), wherein when substituted, the optionally substituted C1-4 alkyl or 3-4 membered ring is substituted with 1-3 substituents independently selected from halogen (preferably F or Cl), OH, CN, C1-4 alkyl optionally substituted with 1-3 F, C1-4 heteroalkyl optionally substituted with 1-3 F, and 3-4 membered ring (including cyclopropyl, cyclobutyl, oxetanyl, azetidinyl, etc.) optionally substituted with F and/or methyl, preferably, R22 at each occurrence is independently F, Cl, Br, CF3, OCF3, SCF3, SF5, OMe, CN, methyl, CHF2, CF2CH3, cyclopropyl, CH3SO2, and OCH2-(cyclopropyl).
Embodiment A22. The compound of any of Embodiments A1-A17, or a pharmaceutically acceptable salt thereof, wherein R30 is a 5-membered heteroaryl
or pyridyl
or pyrimidinyl
which is substituted with 1-3 (e.g., 1 or 2) R22, as valency permits, wherein R22 at each occurrence is independently F, Cl, CN, RM2, ORM2, SRM2, or SF5, wherein RM2 at each occurrence is independently a C1-4 alkyl optionally substituted with 1-3 F, such as CF3.
Embodiment A23. The compound of any of Embodiments A1-A17, or a pharmaceutically acceptable salt thereof, wherein R30 has a structure according to S-1-A, S-1-B, S-1-C, or S-1-D:
wherein R8 is C1-4 alkyl optionally substituted with 1-3 F (such as CH3, CF3, etc.), C1-4 alkoxy optionally substituted with 1-3 F (such as OCH3, OCF3, etc.), SCF3, SF5, cyclopropyl, cyclobutyl, or
Embodiment A24. The compound of any of Embodiments A1-A17, or a pharmaceutically acceptable salt thereof, wherein R30 is selected from:
Embodiment A25. The compound of any of Embodiments A1-A17, or a pharmaceutically acceptable salt thereof, wherein R30 is selected from:
Embodiment A26. The compound of any of Embodiments A1-A17, or a pharmaceutically acceptable salt thereof, R30 is selected from the following:
Embodiment A27. The compound of any of Embodiments A1-A17, or a pharmaceutically acceptable salt thereof, wherein R30 is selected from:
Embodiment A28. The compound of any of Embodiments A1-A17, or a pharmaceutically acceptable salt thereof, wherein R30 is selected from:
Embodiment A29. The compound of any of Embodiments A1-A17, or a pharmaceutically acceptable salt thereof, wherein R30 is selected from:
Embodiment A30. The compound of any of Embodiments A1-A17, or a pharmaceutically acceptable salt thereof, wherein R30 is selected from:
Embodiment A31. The compound of any of Embodiments A1-A4, or a pharmaceutically acceptable salt thereof, wherein L1-L2-R30 is selected from the following:
Embodiment A32. The compound ofany of Embodiments A1-A4, or a pharmaceutically acceptable salt thereof, wherein L1-L2-R30 is selected from the following:
Embodiment A33. The compound of any of Embodiments A1-A4, or a pharmaceutically acceptable salt thereof, wherein L-L2-R30 is selected from the following:
Embodiment A34. The compound of any of Embodiments A1-A4, or a pharmaceutically acceptable salt thereof, wherein L1-L2-R30 is selected from the following:
Embodiment A35. The compound of any of Embodiments A1-A12, or a pharmaceutically acceptable salt thereof, wherein L2-R30 is selected from:
Embodiment A36. The compound of any of Embodiments A1-A12, or a pharmaceutically acceptable salt thereof, wherein L2-R30 is selected from:
Embodiment A37. A compound of Formula III-8, or a pharmaceutically acceptable salt thereof, wherein Ring C is an imidazole or triazole ring, and wherein the variables X, Y, U, R1, R2, L1, L2, and R30 are defined herein, including any of those shown in Embodiments A2-A36 as defined in connection with Formula III-2.
Embodiment A38. The compound of Embodiment A37, or a pharmaceutically acceptable salt thereof, characterized as having a formula according to III-8-A or III-8-B:
wherein the variables X, Y, U, R1, R2, L1, L2, and R30 are defined herein, including any of those shown in Embodiments A2-A36 as defined in connection with Formula III-2.
In some embodiments, the present disclosure provides the following enumerated exemplified Embodiments B1-B45:
Embodiment B1. A compound of Formula III-3, or a pharmaceutically acceptable salt thereof,
Embodiment B2. The compound of Embodiment B1, or a pharmaceutically acceptable salt thereof, characterized as having a structure according to Formula III-3-A:
Embodiment B3. A compound of Formula III-4, or a pharmaceutically acceptable salt thereof,
Embodiment B4. The compound of Embodiment B3, or a pharmaceutically acceptable salt thereof, characterized as having a structure according to Formula III-4-A:
Embodiment B5. The compound of Embodiment B3, or a pharmaceutically acceptable salt thereof, characterized as having a structure according to Formula III-4-B:
Embodiment B6. The compound of Embodiment B5, or a pharmaceutically acceptable salt thereof, characterized as having a structure according to Formula III-4-C or III-4-D:
Embodiment B7. The compound of Embodiment B6, or a pharmaceutically acceptable salt thereof, characterized as having a structure according to Formula III-4-C-1, III-4-C-2, III-4-C-3, III-4-C-4, III-4-D-1, or III-4-D-2:
Embodiment B8. The compound of any of Embodiments B1-B7, or a pharmaceutically acceptable salt thereof, wherein (i) R3 is an optionally substituted C1-4 alkyl, such as CH3, CD3, ethyl, isopropyl, CH2CHF2, etc. or R3 is an optionally substituted 3-4 membered ring, such as cyclopropyl or cyclobutyl, etc., when substituted, the C1-4 alkyl or 3-4 membered ring is typically substituted with 1-3 substituents independently deuterium, F, or methyl, preferably, R3 is methyl or CD3; (11) R3 is hydrogen.
Embodiment B9. The compound of any of Embodiments B1-B8, or a pharmaceutically acceptable salt thereof, wherein (i) R1 is CN; (ii) R1 is halogen, e.g., F or Cl; (iii) R1 is
(iv) R1 is an optionally substituted 5 or 6 membered heteroaryl having 1-4 ring heteroatoms independently selected from O, S, and N, such as an optionally substituted pyrimidinyl or an optionally substituted thiazolyl, for example, R1 is
or (v) R1 is SRA1, wherein RA1 is independently optionally substituted C1-4 alkyl, for example, R1 is SCH3.
Embodiment B10. The compound of any of Embodiments B1-B9, or a pharmaceutically acceptable salt thereof, wherein R9 at each occurrence is independently CH3, CH2CH3, CH2CH2CH3, fluorine-substituted C1-3 alkyl (e.g., CF2H), cyclopropyl, cyclobutyl, CH2OH, CH2OCH3, CH2OCH2CH3, CH2NH2, CH2N3, or CH2NHC(O)OCH3, preferably, R9 at each occurrence is independently CH3, CH2CH3, or CH2CH2CH3, more preferably, R9 at each occurrence is CH2CH3.
Embodiment B11. The compound of any of Embodiments B1-B10, or a pharmaceutically acceptable salt thereof, wherein L2 is an optionally substituted C1-4 alkylene.
Embodiment B12. The compound of any of Embodiments B1-B10, or a pharmaceutically acceptable salt thereof, wherein L2 is selected from the following:
wherein R13 is defined herein.
Embodiment B13. The compound of any of Embodiments B1-B10, or a pharmaceutically acceptable salt thereof, characterized as having a structure according to Formula III-4-C-4a, III-4-C-4b, III-4-C-4c, III-4-C-4d, III-4-C-4e, or III-4-C-4f:
wherein R13 is defined herein.
Embodiment B14. The compound of any of Embodiments B1-B10, or a pharmaceutically acceptable salt thereof, characterized as having a structure according to Formula III-4-C-4a:
wherein R13 is defined herein.
Embodiment B15. The compound of Embodiment B14, or a pharmaceutically acceptable salt thereof, which is substantially stereoisomerically pure, for example, with an enantiomeric excess (“ee”) of 80% or above, such as with 90% ee, 95% ee, 98% ee, 99% ee, or above, and is substantially free of the corresponding stereoisomer according to Formula III-4-C-4b,
for example, with less than 5%, such as with less than 2%, less than 1%, less than 0.5%, or less than 0.1%, of the corresponding stereoisomer according to Formula III-4-C-4b, as determined by HPLC or SFC area % and/or by weight.
Embodiment B16. The compound of any of Embodiments B1-B10, or a pharmaceutically acceptable salt thereof, characterized as having a structure according to Formula III-4-C-4c:
wherein R13 is defined herein.
Embodiment B17. The compound of Embodiment B16, or a pharmaceutically acceptable salt thereof, which is substantially stereoisomerically pure, for example, with an enantiomeric excess (“ee”) of 80% or above, such as with 90% ee, 95% ee, 98% ee, 99% ee, or above, and is substantially free of the corresponding stereoisomer according to Formula III-4-C-4d,
for example, with less than 5%, such as with less than 2%, less than 1%, less than 0.5%, or less than 0.1%, of the corresponding stereoisomer according to Formula III-4-C-4d, as determined by HPLC or SFC area % and/or by weight.
Embodiment B18. The compound of any of Embodiments B1-B10, or a pharmaceutically acceptable salt thereof, characterized as having a structure according to Formula III-4-C-4e:
wherein R13 is defined herein.
Embodiment B19. The compound of Embodiment B18, or a pharmaceutically acceptable salt thereof, which is substantially stereoisomerically pure, for example, with an enantiomeric excess (“ee”) of 80% or above, such as with 90% ee, 95% ee, 98% ee, 99% ee, or above, and is substantially free of the corresponding stereoisomer according to Formula III-4-C-4f,
f for example, with less than 5%, such as with less than 2%, less than 1%, less than 0.5%, or less than 0.1%, of the corresponding stereoisomer according to Formula III-4-C-4f, as determined by HPLC or SFC area % and/or by weight.
Embodiment B20. The compound of any of Embodiments B12-B19, or a pharmaceutically acceptable salt thereof, wherein R13 is hydrogen, CN, OH, COOH, CONH2, methoxy, ethoxy, cyclopropyl, cyclobutyl, optionally substituted phenyl, or optionally substituted 5 or 6 membered heteroaryl having 1-3 ring heteroatoms independently selected from N, O, and S, wherein, when substituted, the optionally substituted phenyl or 5 or 6 membered heteroaryl is substituted with 1-3 substituents independently selected from halogen (preferably F or Cl), OH, CN, C1-4 alkyl optionally substituted with 1-3 F, C1-4 heteroalkyl optionally substituted with 1-3 F, and 3-4 membered ring (including cyclopropyl, cyclobutyl, oxetanyl, azetidinyl, etc.) optionally substituted with F and/or methyl, more preferably, R13 is hydrogen.
Embodiment B21. The compound of any of Embodiments B12-B19, or a pharmaceutically acceptable salt thereof, wherein R13 is a C1-4 alkyl optionally substituted with one or more substituents independently selected from F, OH, or C1-4 alkoxy, for example, R13 is methyl, ethyl, isopropyl, CH2OH, CH2OMe, etc.
Embodiment B22. The compound of any of Embodiments B12-B19, or a pharmaceutically acceptable salt thereof, wherein R13 is phenyl, which is optionally substituted with 1-3 substituents independently selected from halogen (preferably F or Cl), OH, CN, C1-4 alkyl optionally substituted with 1-3 F, C1-4 heteroalkyl optionally substituted with 1-3 F, and 3-4 membered ring (including cyclopropyl, cyclobutyl, oxetanyl, azetidinyl, etc.) optionally substituted with F and/or methyl, for example, R13 is
Embodiment B23. The compound of any of Embodiments B12-B19, or a pharmaceutically acceptable salt thereof, wherein R13 is a 5 or 6 membered heteroaryl, such as
which is optionally substituted with 1-3 substituents independently selected from halogen (preferably F or Cl), OH, CN, C1-4 alkyl optionally substituted with 1-3 F, C1-4 heteroalkyl optionally substituted with 1-3 F, and 3-4 membered ring (including cyclopropyl, cyclobutyl, oxetanyl, azetidinyl, etc.) optionally substituted with F and/or methyl, for example, R13 is
Embodiment B24. The compound of any of Embodiments B1-B10, or a pharmaceutically acceptable salt thereof, wherein L2 is absent, O, or C(O).
Embodiment B25. The compound of any of Embodiments B1-B24, or a pharmaceutically acceptable salt thereof, wherein R30 is an optionally substituted phenyl or 5 or 6-membered heteroaryl (e.g., such as oxadiazoyl
pyridyl
or pyrimidinyl
for example, when substituted, the phenyl or 5 or 6-membered heteroaryl can be substituted with 1-3 (e.g., 1 or 2) R22, wherein R22 at each occurrence is halogen (e.g., F, Cl, or Br), CN, C1-4 alkyl optionally substituted with 1-3 F (such as CH3, CF3, etc.), C1-4 alkoxy optionally substituted with 1-3 F (such as OCH3, OCF3, etc.), SCF3, SF5, cyclopropyl, cyclobutyl, or
Embodiment B26. The compound of any of Embodiments B1-B24, or a pharmaceutically acceptable salt thereof, wherein R30 is a phenyl which is substituted with 1-3 (e.g., 1 or 2) R22, wherein R22 at each occurrence is independently halogen (e.g., F, Cl, or Br), CN, OH, RM1 ORM1, SO2RM1, P(O)(RM1)2, SRM1, or SF5, wherein RM1 at each occurrence is independently an optionally substituted C1-4 alkyl or an optionally substituted 3-4 membered ring (including cyclopropyl, cyclobutyl, oxetanyl, azetidinyl, etc.), wherein when substituted, the optionally substituted C1-4 alkyl or 3-4 membered ring is substituted with 1-3 substituents independently selected from halogen (preferably F or Cl), OH, CN, C1-4 alkyl optionally substituted with 1-3 F, C1-4 heteroalkyl optionally substituted with 1-3 F, and 3-4 membered ring (including cyclopropyl, cyclobutyl, oxetanyl, azetidinyl, etc.) optionally substituted with F and/or methyl, preferably, R22 at each occurrence is independently F, Cl, Br, CF3, OCF3, SCF3, SF5, OMe, CN, methyl, CHF2, CF2CH3, cyclopropyl, CH3SO2, and OCH2-(cyclopropyl).
Embodiment B27. The compound of any of Embodiments B1-B24, or a pharmaceutically acceptable salt thereof, wherein R30 is a phenyl which is substituted with 1-3 (e.g., 1 or 2) R22, wherein R22 at each occurrence is independently F, Cl, CN, RM2, ORM2, SRM2, or SF5, wherein RM2 at each occurrence is independently a C1-4 alkyl optionally substituted with 1-3 F, such as CF3.
Embodiment B28. The compound of any of Embodiments B1-B24, or a pharmaceutically acceptable salt thereof, wherein R30 is a 5-membered heteroaryl
or pyridyl
or pyrimidinyl
which is substituted with 1-3 (e.g., 1 or 2) R22, as valency permits, wherein R22 at each occurrence is independently halogen (e.g., F, Cl, or Br), CN, OH, RM1, ORM1, SO2RM1, P(O)(RM1)2, SRMI, or SF5, wherein RM1 at each occurrence is independently an optionally substituted C1-4 alkyl or an optionally substituted 3-4 membered ring (including cyclopropyl, cyclobutyl, oxetanyl, azetidinyl, etc.), wherein when substituted, the optionally substituted C1-4 alkyl or 3-4 membered ring is substituted with 1-3 substituents independently selected from halogen (preferably F or Cl), OH, CN, C1-4 alkyl optionally substituted with 1-3 F, C1-4 heteroalkyl optionally substituted with 1-3 F, and 3-4 membered ring (including cyclopropyl, cyclobutyl, oxetanyl, azetidinyl, etc.) optionally substituted with F and/or methyl, preferably, R22 at each occurrence is independently F, Cl, Br, CF3, OCF3, SCF3, SF5, OMe, CN, methyl, CHF2, CF2CH3, cyclopropyl, CH3SO2, and OCH2-(cyclopropyl).
Embodiment B29. The compound of any of Embodiments B1-B24, or a pharmaceutically acceptable salt thereof, wherein R30 is a 5-membered heteroaryl
or pyridyl
or pyrimidinyl
which is substituted with 1-3 (e.g., 1 or 2) R22, as valency permits, wherein R22 at each occurrence is independently F, Cl, CN, RM2, ORM2, SRM2, or SF5, wherein RM2 at each occurrence is independently a C1-4 alkyl optionally substituted with 1-3 F, such as CF3.
Embodiment B30. The compound of any of Embodiments B1-B24, or a pharmaceutically acceptable salt thereof, wherein R30 has a structure according to S-1-A, S-1-B, S-1-C, or S-1-D:
wherein R1 is C1-4 alkyl optionally substituted with 1-3 F (such as CH3, CF3, etc.), C1-4 alkoxy optionally substituted with 1-3 F (such as OCH3, OCF3, etc.), SCF3, SF5, cyclopropyl, cyclobutyl, or
Embodiment B31. The compound of any of Embodiments B1-B24, or a pharmaceutically acceptable salt thereof, wherein R30 is selected from:
Embodiment B32. The compound of any of Embodiments B1-B24, or a pharmaceutically acceptable salt thereof, wherein R30 is selected from:
Embodiment B33. The compound of any of Embodiments B1-B24, or a pharmaceutically acceptable salt thereof, wherein R30 is selected from:
Embodiment B34. The compound of any of Embodiments B1-B24, or a pharmaceutically acceptable salt thereof, wherein R30 is selected from:
Embodiment B35. The compound of any of Embodiments B1-B24, or a pharmaceutically acceptable salt thereof, wherein R30 is selected from:
Embodiment B36. The compound of any of Embodiments B1-B24, or a pharmaceutically acceptable salt thereof, wherein R30 is selected from:
Embodiment B37. The compound of any of Embodiments B1-B24, or a pharmaceutically acceptable salt thereof, wherein R30 is selected from:
Embodiment B38. The compound of any of Embodiments B1-B10, or a pharmaceutically acceptable salt thereof, wherein L2-R30 is selected from:
Embodiment B39. The compound of any of Embodiments B1-B10, or a pharmaceutically acceptable salt thereof, wherein L2-R30 is selected from:
Embodiment B40. The compound of any of Embodiments B1-B10, or a pharmaceutically acceptable salt thereof, wherein L2-R30 is selected from:
Embodiment B41. The compound of any of Embodiments B1-B10, or a pharmaceutically acceptable salt thereof, wherein L2-R30 is selected from:
Embodiment B42. The compound of any of Embodiments B1-B124, or a pharmaceutically acceptable salt thereof, wherein R30 is selected from:
Embodiment B43. The compound of any of Embodiments B1-B10, or a pharmaceutically acceptable salt thereof, wherein L2-R30 is selected from:
Embodiment B44. The compound of any of Embodiments B1-B10, or a pharmaceutically acceptable salt thereof, wherein L2-R30 is selected from:
Embodiment B45. A compound of Formula III-6 or III-7, or a pharmaceutically acceptable salt thereof, wherein -J-W-represents —CH2—O—CH2, —O—CH2—CH2—, —N(CH3)—C(O)—, —NH—C(O)—, —N(CH3)—CH2, or —NH—CH2, (from left to right, i.e., J is the first group from left, such as CH2, O, NH, or NCH3), and wherein the variables Z, Y, U, R1, R2, R3, n1, R9, L2, and R30 are defined herein, including any of those shown in Embodiments B1-B44 as defined in connection with Formula III-3 or III-4.
In some embodiments, the present disclosure also provides a compound selected from Compound Nos. 1-347, or a pharmaceutically acceptable salt thereof.
In some embodiments, the present disclosure also provides a compound selected from the compounds shown in Table A below, or a pharmaceutically acceptable salt thereof:
Exemplary synthesis and characterization of the above compounds are shown in the Examples section. The compounds may be prepared in a racemic form, with respect to one or more of the chiral centers, which can be separated into two enantiomers, including the as-drawn enantiomer, or be prepared through chiral synthesis, in view of the present disclosure.
In some embodiments, to the extent applicable, the genus of compounds in the present disclosure also excludes any of the compounds specifically prepared and disclosed prior to this disclosure. In other words, the genus of compounds in the present disclosure can optionally have a negative limitation to exclude one or more applicable compound(s) selected from any of the compounds specifically prepared prior to this disclosure.
In some embodiments, the genus of compounds in the present disclosure, includes any of those shown in original claims 1-63 herein, Embodiments 1-49, Embodiments A1-A38, and Embodiments B1-B45.
The compounds of the present disclosure can be readily synthesized by one of ordinary skilled in the art in view of the present disclosure. Exemplified syntheses are also shown in the Examples section.
As will be apparent to those having ordinary skill in the art, conventional protecting groups may be necessary to prevent certain functional groups from undergoing undesired reactions. Suitable protecting groups for various functional groups as well as suitable conditions for protecting and deprotecting particular functional groups are well known in the art. For example, numerous protecting groups are described in “Protective Groups in Organic Synthesis”, 4th ed. P. G. M. Wuts; T. W. Greene, John Wiley, 2007, and references cited therein. The reagents for the reactions described herein are generally known compounds or can be prepared by known procedures or obvious modifications thereof. For example, many of the reagents are available from commercial suppliers such as Aldrich Chemical Co. (Milwaukee, Wisconsin, USA), Sigma (St. Louis, Missouri, USA). Others may be prepared by procedures, or obvious modifications thereof, described in standard reference texts such as Fieser and Fieser's Reagents for Organic Synthesis, Volumes 1-15 (John Wiley and Sons, 1991), Rodd's Chemistry of Carbon Compounds, Volumes 1-5 and Supplemental (Elsevier Science Publishers, 1989), Organic Reactions, Volumes 1-40 (John Wiley and Sons, 1991), March's Advanced Organic Chemistry, (Wiley, 7th Edition), and Larock's Comprehensive Organic Transformations (Wiley-VCH, 1999), and any of available updates as of this filing.
Certain embodiments are directed to a pharmaceutical composition comprising one or more of the compounds of the present disclosure.
The pharmaceutical composition can optionally contain a pharmaceutically acceptable excipient. In some embodiments, the pharmaceutical composition comprises a compound of the present disclosure (e.g., a compound of Formula III-1, III-2 (e.g., III-2-A, III-2-B, III-2-C, III-2-C-1, III-2-C-2, III-2-C-3, III-2-C-4, III-2-D, III-2-D-1, III-2-D-2, III-2-D-3, III-2-D-4, III-2-E-1, III-2-E-2, III-2-E-3, III-2-E-1a, III-2-E-2a, III-2-E-3a, III-2-E-1b, III-2-E-2b, III-2-E-3b, III-2-F-1a, III-2-F-1b, III-2-F-2a, III-2-F-2b, III-2-F-3a, or III-2-F-3b), III-3 (e.g., III-3-A), III-4 (e.g., III-4-A, III-4-B, III-4-C, III-4-D, III-4-C-1, III-4-C-2, III-4-C-3, III-4-C-4, III-4-D-1, III-4-D-2, III-4-C-4a, III-4-C-4b, III-4-C-4c, III-4-C-4d, III-4-C-4e, or III-4-C-4f), III-5, III-6, III-7, III-8 (e.g., III-8-A or III-8-B), III-9, III-10, III-11, or III-12, any of Compound Nos. 1-347, any compound selected from the compounds shown in Table A herein, or a pharmaceutically acceptable salt thereof) and a pharmaceutically acceptable excipient. Pharmaceutically acceptable excipients are known in the art. Non-limiting suitable excipients include, for example, encapsulating materials or additives such as absorption accelerators, antioxidants, binders, buffers, carriers, coating agents, coloring agents, diluents, disintegrating agents, emulsifiers, extenders, fillers, flavoring agents, humectants, lubricants, perfumes, preservatives, propellants, releasing agents, sterilizing agents, sweeteners, solubilizers, wetting agents and mixtures thereof. See also Remington's The Science and Practice of Pharmacy, 21st Edition, A. R. Gennaro (Lippincott, Williams & Wilkins, Baltimore, Md., 2005; incorporated herein by reference), which discloses various excipients used in formulating pharmaceutical compositions and known techniques for the preparation thereof.
The pharmaceutical composition can include any one or more of the compounds of the present disclosure. For example, in some embodiments, the pharmaceutical composition comprises a compound of Formula III-1, III-2 (e.g., III-2-A, III-2-B, III-2-C, III-2-C-1, III-2-C-2, III-2-C-3, III-2-C-4, III-2-D, III-2-D-1, III-2-D-2, III-2-D-3, III-2-D-4, III-2-E-1, III-2-E-2, III-2-E-3, III-2-E-1a, III-2-E-2a, III-2-E-3a, III-2-E-1b, III-2-E-2b, III-2-E-3b, III-2-F-1a, III-2-F-1b, III-2-F-2a, III-2-F-2b, III-2-F-3a, or III-2-F-3b), III-3 (e.g., III-3-A), III-4 (e.g., III-4-A, III-4-B, III-4-C, III-4-D, III-4-C-1, III-4-C-2, III-4-C-3, III-4-C-4, III-4-D-1, III-4-D-2, III-4-C-4a, III-4-C-4b, III-4-C-4c, III-4-C-4d, III-4-C-4e, or III-4-C-4f), III-5, III-6, III-7, III-8 (e.g., III-8-A or III-8-B), III-9, III-10, III-11, or III-12, any of Compound Nos. 1-347, any compound selected from the compounds shown in Table A herein, or a pharmaceutically acceptable salt thereof, e.g., in a therapeutically effective amount. In any of the embodiments described herein, the pharmaceutical composition can comprise a therapeutically effective amount of a compound selected from Compound Nos. 1-347, or a pharmaceutically acceptable salt thereof. In any of the embodiments described herein, the pharmaceutical composition can comprise a therapeutically effective amount of a compound selected from the compounds shown in Table A herein, or a pharmaceutically acceptable salt thereof.
The pharmaceutical composition can also be formulated for delivery via any of the known routes of delivery, which include but are not limited to oral, parenteral, inhalation, etc.
In some embodiments, the pharmaceutical composition can be formulated for oral administration. The oral formulations can be presented in discrete units, such as capsules, pills, cachets, lozenges, or tablets, each containing a predetermined amount of the active compound; as a powder or granules; as a solution or a suspension in an aqueous or non-aqueous liquid; or as an oil-in-water or water-in-oil emulsion. Excipients for the preparation of compositions for oral administration are known in the art. Non-limiting suitable excipients include, for example, agar, alginic acid, aluminum hydroxide, benzyl alcohol, benzyl benzoate, 1,3-butylene glycol, carbomers, castor oil, cellulose, cellulose acetate, cocoa butter, corn starch, corn oil, cottonseed oil, cross-povidone, diglycerides, ethanol, ethyl cellulose, ethyl laureate, ethyl oleate, fatty acid esters, gelatin, germ oil, glucose, glycerol, groundnut oil, hydroxypropylmethyl cellulose, isopropanol, isotonic saline, lactose, magnesium hydroxide, magnesium stearate, malt, mannitol, monoglycerides, olive oil, peanut oil, potassium phosphate salts, potato starch, povidone, propylene glycol, Ringer's solution, safflower oil, sesame oil, sodium carboxymethyl cellulose, sodium phosphate salts, sodium lauryl sulfate, sodium sorbitol, soybean oil, stearic acids, stearyl fumarate, sucrose, surfactants, talc, tragacanth, tetrahydrofurfuryl alcohol, triglycerides, water, and mixtures thereof.
In some embodiments, the pharmaceutical composition is formulated for parenteral administration (such as intravenous injection or infusion, subcutaneous or intramuscular injection). The parenteral formulations can be, for example, an aqueous solution, a suspension, or an emulsion. Excipients for the preparation of parenteral formulations are known in the art. Non-limiting suitable excipients include, for example, 1,3-butanediol, castor oil, corn oil, cottonseed oil, dextrose, germ oil, groundnut oil, liposomes, oleic acid, olive oil, peanut oil, Ringer's solution, safflower oil, sesame oil, soybean oil, U.S.P. or isotonic sodium chloride solution, water and mixtures thereof.
In some embodiments, the pharmaceutical composition is formulated for inhalation. The inhalable formulations can be, for example, formulated as a nasal spray, dry powder, or an aerosol administrable through a metered-dose inhaler. Excipients for preparing formulations for inhalation are known in the art. Non-limiting suitable excipients include, for example, lactose, talc, silicic acid, aluminum hydroxide, calcium silicates and polyamide powder, and mixtures of these substances. Sprays can additionally contain propellants, such as chlorofluorohydrocarbons and volatile unsubstituted hydrocarbons, such as butane and propane.
The pharmaceutical composition can include various amounts of the compounds of the present disclosure, depending on various factors such as the intended use and potency and selectivity of the compounds. In some embodiments, the pharmaceutical composition comprises a therapeutically effective amount of a compound of the present disclosure (e.g., a compound of Formula III-1, III-2 (e.g., III-2-A, III-2-B, III-2-C, III-2-C-1, III-2-C-2, III-2-C-3, III-2-C-4, III-2-D, III-2-D-1, III-2-D-2, III-2-D-3, III-2-D-4, III-2-E-1, III-2-E-2, III-2-E-3, III-2-E-1a, III-2-E-2a, III-2-E-3a, III-2-E-1b, III-2-E-2b, III-2-E-3b, III-2-F-1a, III-2-F-1b, III-2-F-2a, III-2-F-2b, III-2-F-3a, or III-2-F-3b), III-3 (e.g., III-3-A), III-4 (e.g., III-4-A, III-4-B, III-4-C, III-4-D, III-4-C-1, III-4-C-2, III-4-C-3, III-4-C-4, III-4-D-1, III-4-D-2, III-4-C-4a, III-4-C-4b, III-4-C-4c, III-4-C-4d, III-4-C-4e, or III-4-C-4f), III-5, III-6, III-7, III-8 (e.g., III-8-A or III-8-B), III-9, III-10, III-11, or III-12, any of Compound Nos. 1-347, any compound selected from the compounds shown in Table A herein, or a pharmaceutically acceptable salt thereof). In some embodiments, the pharmaceutical composition comprises a therapeutically effective amount of the compound of the present disclosure and a pharmaceutically acceptable excipient. As used herein, a therapeutically effective amount of a compound of the present disclosure is an amount effective to treat a disease or disorder as described herein, which can depend on the recipient of the treatment, the disease or disorder being treated and the severity thereof, the composition containing the compound, the time of administration, the route of administration, the duration of treatment, the compound potency (e.g., for inhibiting DGKa and/or DGKz), its rate of clearance and whether or not another drug is co-administered.
For veterinary use, a compound of the present disclosure can be administered as a suitably acceptable formulation in accordance with normal veterinary practice. The veterinarian can readily determine the dosing regimen and route of administration that is most appropriate for a particular animal.
In some embodiments, all the necessary components for the treatment of DGKa and/or DGKz associated diseases or disorders using a compound of the present disclosure either alone or in combination with another agent or intervention traditionally used for the treatment of such disease can be packaged into a kit. Specifically, in some embodiments, the present invention provides a kit for use in the therapeutic intervention of the disease comprising a packaged set of medicaments that include the compound disclosed herein as well as buffers and other components for preparing deliverable forms of said medicaments, and/or devices for delivering such medicaments, and/or any agents that are used in combination therapy (e.g., any of those described herein, such as an immuno-oncology agent described herein, including for example an antagonist of a protein that inhibits T cell activation, an agonist of a protein that stimulates T cell activation, etc., in particular, one or more antibodies selected from anti-PD-1, anti-PD-L1, anti-CTLA-4, and combinations thereof) with the compound of the present disclosure, and/or instructions for the treatment of the disease packaged with the medicaments. The instructions may be fixed in any tangible medium, such as printed paper, or a computer readable magnetic or optical medium, or instructions to reference a remote computer data source such as a world wide web page accessible via the internet.
Compounds of the present disclosure are useful as therapeutic active substances for the treatment and/or prophylaxis of diseases or disorders that are associated with the activity of DGKa, DGKz, or both DGKa and DGKz, such as DGK target inhibition in T cells. Such diseases or disorders include viral and other infections (e.g., skin infections, GI infection, urinary tract infections, genito-urinary infections, systemic infections), and proliferative diseases (e.g., cancer).
In some embodiments, the present disclosure provides a method of inhibiting the activity of diacylglycerol kinase alpha and/or zeta (DGKa/z) in a cell comprising contacting a cell with an effective amount of one or more compounds of the present disclosure (e.g., a compound of Formula III-1, III-2 (e.g., III-2-A, III-2-B, III-2-C, III-2-C-1, III-2-C-2, III-2-C-3, III-2-C-4, III-2-D, III-2-D-1, III-2-D-2, III-2-D-3, III-2-D-4, III-2-E-1, III-2-E-2, III-2-E-3, III-2-E-1a, III-2-E-2a, III-2-E-3a, III-2-E-1b, III-2-E-2b, III-2-E-3b, III-2-F-1a, III-2-F-1b, III-2-F-2a, III-2-F-2b, III-2-F-3a, or III-2-F-3b), III-3 (e.g., III-3-A), III-4 (e.g., III-4-A, III-4-B, III-4-C, III-4-D, III-4-C-1, III-4-C-2, III-4-C-3, III-4-C-4, III-4-D-1, III-4-D-2, III-4-C-4a, III-4-C-4b, III-4-C-4c, III-4-C-4d, III-4-C-4e, or III-4-C-4f), III-5, III-6, III-7, III-8 (e.g., III-8-A or III-8-B), III-9, III-10, III-11, or III-12, any of Compound Nos. 1-347, any compound selected from the compounds shown in Table A herein, or a pharmaceutically acceptable salt thereof). As used herein, the term “cell” is meant to refer to a cell that is in vitro, ex vivo or in vivo. In some embodiments, an ex vivo cell can be part of a tissue sample excised from an organism such as a mammal. In some embodiments, an in vitro cell can be a cell in a cell culture. In some embodiments, an in vivo cell is a cell living in an organism such as a mammal. As used herein, the term “contacting” refers to the bringing together of indicated moieties in an in vitro system or an in vivo system. For example, “contacting” the DGKa and DGKz enzyme with a compound of the present disclosure includes the administration of a compound of the present disclosure to a subject, such as a human, having DGKa and DGKz, as well as, for example, introducing a compound of the present disclosure into a sample containing a cellular or purified preparation containing DGKa and DGKz enzyme. The term “DGK inhibitor” such as a DGKa and/or DGKz inhibitor refers to an agent capable of inhibiting the activity of diacylglycerol kinase alpha and/or diacylglycerol kinase zeta (DGKa and/or DGKz), such as in T cells resulting in T cell stimulation.
In some embodiments, the present disclosure provides a method of treating a disease associated with activity or expression, including abnormal activity and/or overexpression, of DGKa and/or DGKz in a subject in need thereof, the method comprising administering to the subject an effective amount of one or more compounds of the present disclosure (e.g., a compound of Formula III-1, III-2 (e.g., III-2-A, III-2-B, III-2-C, III-2-C-1, III-2-C-2, III-2-C-3, III-2-C-4, III-2-D, III-2-D-1, III-2-D-2, III-2-D-3, III-2-D-4, III-2-E-1, II-2-E-2, III-2-E-3, III-2-E-1a, III-2-E-2a, III-2-E-3a, III-2-E-1b, III-2-E-2b, III-2-E-3b, III-2-F-1a, III-2-F-1b, III-2-F-2a, III-2-F-2b, III-2-F-3a, or III-2-F-3b), III-3 (e.g., III-3-A), III-4 (e.g., III-4-A, III-4-B, III-4-C, III-4-D, III-4-C-1, III-4-C-2, III-4-C-3, III-4-C-4, III-4-D-1, III-4-D-2, III-4-C-4a, III-4-C-4b, III-4-C-4c, III-4-C-4d, III-4-C-4e, or III-4-C-4f), III-5, III-6, III-7, III-8 (e.g., III-8-A or III-8-B), III-9, III-10, III-11, or III-12, any of Compound Nos. 1-347, any compound selected from the compounds shown in Table A herein, or a pharmaceutically acceptable salt thereof). Examples of diseases can include any disease, disorder or condition that is directly or indirectly linked to expression or activity of DGKa and/or DGKz enzyme, such as over expression or abnormal activity. A DGKa and/or DGKz-associated disease can also include any disease, disorder or condition that can be prevented, ameliorated, or cured by modulating DGKa and/or DGKz enzyme activity. Examples of DGKa and/or DGKz associated diseases include cancer and viral infections such as HIV infection, hepatitis B, and hepatitis C. Examples of cancer include cancer of the colon, pancreatic cancer, breast cancer, prostate cancer, lung cancer, ovarian cancer, cervical cancer, renal cancer, cancer of the head and neck, lymphoma, leukemia, and/or melanoma.
In some embodiments, the present disclosure provides a method of treating cancer in a subject, the method comprising administering to the subject a therapeutically effective amount of one or more compounds of the present disclosure (e.g., a compound of Formula III-1, III-2 (e.g., III-2-A, III-2-B, III-2-C, III-2-C-1, III-2-C-2, III-2-C-3, III-2-C-4, III-2-D, III-2-D-1, III-2-D-2, III-2-D-3, III-2-D-4, III-2-E-1, III-2-E-2, III-2-E-3, III-2-E-1a, III-2-E-2a, III-2-E-3a, III-2-E-1b, III-2-E-2b, III-2-E-3b, III-2-F-1a, III-2-F-1b, III-2-F-2a, III-2-F-2b, III-2-F-3a, or III-2-F-3b), III-3 (e.g., III-3-A), III-4 (e.g., III-4-A, III-4-B, III-4-C, III-4-D, III-4-C-1, III-4-C-2, III-4-C-3, III-4-C-4, III-4-D-1, III-4-D-2, III-4-C-4a, III-4-C-4b, III-4-C-4c, III-4-C-4d, III-4-C-4e, or III-4-C-4f), III-5, III-6, III-7, III-8 (e.g., III-8-A or III-8-B), III-9, III-10, III-11, or III-12, any of Compound Nos. 1-347, any compound selected from the compounds shown in Table A herein, or a pharmaceutically acceptable salt thereof) or a therapeutically effective amount of a pharmaceutical composition described herein. In some embodiments, the cancer is cancer of the colon, pancreatic cancer, breast cancer, prostate cancer, lung cancer, ovarian cancer, cervical cancer, renal cancer, cancer of the head and neck, lymphoma, leukemia, and/or melanoma. Types of cancers that may be treated with the compound of the present disclosure include, but are not limited to, brain cancers, skin cancers, bladder cancers, ovarian cancers, breast cancers, gastric cancers, pancreatic cancers, prostate cancers, colon cancers, blood cancers, lung cancers and bone cancers. Examples of such cancer types include neuroblastoma, intestine carcinoma such as rectum carcinoma, colon carcinoma, familiar adenomatous polyposis carcinoma and hereditary non-polyposis colorectal cancer, esophageal carcinoma, labial carcinoma, larynx carcinoma, hypopharynx carcinoma, tongue carcinoma, salivary gland carcinoma, gastric carcinoma, adenocarcinoma, medullary thyroid carcinoma, papillary thyroid carcinoma, renal carcinoma, kidney parenchymal carcinoma, ovarian carcinoma, cervix carcinoma, uterine corpus carcinoma, endometrium carcinoma, chorion carcinoma, pancreatic carcinoma, prostate carcinoma, testis carcinoma, breast carcinoma, urinary carcinoma, melanoma, brain tumors such as glioblastoma, astrocytoma, meningioma, medulloblastoma and peripheral neuroectodermal tumors, Hodgkin lymphoma, non-Hodgkin lymphoma, Burkitt lymphoma, acute lymphatic leukemia (ALL), chronic lymphatic leukemia (CLL), acute myeloid leukemia (AML), chronic myeloid leukemia (CML), adult T-cell leukemia lymphoma, diffuse large B-cell lymphoma (DLBCL), hepatocellular carcinoma, gall bladder carcinoma, bronchial carcinoma, small cell lung carcinoma, non-small cell lung carcinoma, multiple myeloma, basalioma, teratoma, retinoblastoma, choroid melanoma, seminoma, rhabdomyosarcoma, craniopharyngioma, osteosarcoma, chondrosarcoma, myosarcoma, liposarcoma, fibrosarcoma, Ewing sarcoma and plasmocytoma.
In some embodiments, the present disclosure provides a method of treating viral infection in a subject, the method comprising administering to the subject a therapeutically effective amount of one or more compounds of the present disclosure (e.g., a compound of Formula III-1, III-2 (e.g., III-2-A, III-2-B, III-2-C, III-2-C-1, III-2-C-2, III-2-C-3, III-2-C-4, III-2-D, III-2-D-1, III-2-D-2, III-2-D-3, III-2-D-4, III-2-E-1, III-2-E-2, III-2-E-3, III-2-E-1a, III-2-E-2a, III-2-E-3a, III-2-E-1b, III-2-E-2b, III-2-E-3b, III-2-F-1a, III-2-F-1b, III-2-F-2a, III-2-F-2b, III-2-F-3a, or III-2-F-3b), III-3 (e.g., III-3-A), III-4 (e.g., III-4-A, III-4-B, III-4-C, III-4-D, III-4-C-1, III-4-C-2, III-4-C-3, III-4-C-4, III-4-D-1, III-4-D-2, III-4-C-4a, III-4-C-4b, III-4-C-4c, III-4-C-4d, III-4-C-4e, or III-4-C-4f), III-5, III-6, III-7, III-8 (e.g., III-8-A or III-8-B), III-9, III-10, III-11, or III-12, any of Compound Nos. 1-347, any compound selected from the compounds shown in Table A herein, or a pharmaceutically acceptable salt thereof) or a therapeutically effective amount of a pharmaceutical composition described herein. Viral infections that may be treated include, but are not limited to, diseases caused by: hepatitis C virus (HCV), human papilloma virus (HPV), cytomegalovirus (CMV), herpes simplex virus (HSV), Epstein-Barr virus (EBV), varicella zoster virus, coxsackie virus, human immunodeficiency virus (HIV). In some embodiments, parasitic infections (e.g., malaria) may also be treated by the above methods.
In some embodiments, the present disclosure provides a method of treating a disease or disorder, e.g., a cancer associated with DGKa and/or DGKz in a subject in need thereof. In some embodiments, the method comprises administering to the subject a therapeutically effective amount of a compound of the present disclosure (e.g., a compound of Formula III-1, III-2 (e.g., III-2-A, III-2-B, III-2-C, III-2-C-1, III-2-C-2, III-2-C-3, III-2-C-4, III-2-D, III-2-D-1, III-2-D-2, III-2-D-3, III-2-D-4, III-2-E-1, III-2-E-2, III-2-E-3, III-2-E-1a, III-2-E-2a, III-2-E-3a, III-2-E-1b, III-2-E-2b, III-2-E-3b, III-2-F-1a, III-2-F-1b, III-2-F-2a, III-2-F-2b, III-2-F-3a, or III-2-F-3b), III-3 (e.g., III-3-A), III-4 (e.g., III-4-A, III-4-B, III-4-C, III-4-D, III-4-C-1, III-4-C-2, III-4-C-3, III-4-C-4, III-4-D-1, III-4-D-2, III-4-C-4a, III-4-C-4b, III-4-C-4c, III-4-C-4d, III-4-C-4e, or III-4-C-4f), III-5, III-6, III-7, III-8 (e.g., III-8-A or III-8-B), III-9, III-10, III-11, or III-12, any of Compound Nos. 1-347, any compound selected from the compounds shown in Table A herein, or a pharmaceutically acceptable salt thereof) or a therapeutically effective amount of a pharmaceutical composition described herein. In some embodiments, the cancer is cancer of the colon, pancreatic cancer, breast cancer, prostate cancer, lung cancer, ovarian cancer, cervical cancer, renal cancer, cancer of the head and neck, lymphoma, leukemia, and/or melanoma. Other types of cancer suitable to be treated by the method include those described herein.
Compounds of the present disclosure can be used as a monotherapy or in a combination therapy. In some embodiments, the combination therapy includes treating the subject with a targeted therapeutic agent, chemotherapeutic agent, therapeutic antibody, radiation, cell therapy, and/or immunotherapy. In some embodiments, compounds of the present disclosure can also be co-administered with an additional pharmaceutically active compound, either concurrently or sequentially in any order, to a subject in need thereof. In some embodiments, the combination therapy includes treating the subject with one or more additional therapies such as anti-viral agents, chemotherapeutics or other anti-cancer agents, immune enhancers, immunosuppressants, radiation, anti-tumor and anti-viral vaccines, cytokine therapy (e.g., IL2 and GM-CSF), and/or tyrosine kinase inhibitors.
In some embodiments, compounds of the present disclosure can be administered concurrently or sequentially in any order with an immuno-oncology agent. Immuno-oncology agents include, for example, a small molecule drug, antibody, or other biologic or small molecule. Examples of biologic immuno-oncology agents include, but are not limited to, cancer vaccines, antibodies, and cytokines. In some embodiments, the immuno-oncology agent is (i) an agonist of a stimulatory (including a co-stimulatory) receptor or (ii) an antagonist of an inhibitory (including a co-inhibitory) signal on T cells, both of which result in amplifying antigen-specific T cell responses (often referred to as immune checkpoint regulators). Certain of the stimulatory and inhibitory molecules are members of the immunoglobulin super family (IgSF). One important family of membrane-bound ligands that bind to co-stimulatory or co-inhibitory receptors is the B7 family, which includes B7-1, B7-2, B7-H1 (PD-L1), B7-DC (PD-L2), B7-H2 (ICOS-L), B7-H3, B7-H4, B7-H5 (VISTA), and B7-H6. Another family of membrane bound ligands that bind to co-stimulatory or co-inhibitory receptors is the TNF family of molecules that bind to cognate TNF receptor family members, which includes CD40 and CD40L, OX-40, OX-40L, CD70, CD27L, CD30, CD30L, 4-1BBL, CD137 (4-1BB), TRAIL/Apo2-L, TRAILR1/DR4, TRAILR2/DR5, TRAILR3, TRAILR4, OPG, RANK, RANKL, TWEAKR/Fn14, TWEAK, BAFFR, EDAR, XEDAR, TACl, APRIL, BCMA, LTbR, LIGHT, DcR3, HVEM, VEGI/TL1A, TRAMP/DR3, EDAR, EDA1, XEDAR, EDA2, TNFR1, Lymphotoxin a/TNFb, TNFR2, TNFa, LTbR, Lymphotoxin a 1b2, FAS, FASL, RELT, DR6, TROY, NGFR. T cell responses can be stimulated by a combination of a compound of the present disclosure and one or more of (i) an antagonist of a protein that inhibits T cell activation (e.g., immune checkpoint inhibitors) such as CTLA-4, PD-1, PD-L1, PD-L2, LAG-3, TIM-3, Galectin 9, CEACAM-1, BTLA, CD69, Galectin-1, TIGIT, CD113, GPR56, VISTA, 2B4, CD48, GARP, PD1H, LAIR1, TIM-1, and TIM-4, and (ii) an agonist of a protein that stimulates T cell activation such as B7-1, B7-2, CD28, 4-1BB (CD137), 4-1BBL, ICOS, ICOS-L, OX40, OX40L, GITR, GITRL, CD70, CD27, CD40, DR3 and CD28H. Other agents that can be combined with compounds of the present disclosure for the treatment of cancer include antagonists of inhibitory receptors on NK cells or agonists of activating receptors on NK cells. For example, a compound of the present disclosure can be combined with antagonists of KIR, such as lirilumab. Other agents for combination therapies include agents that inhibit or deplete macrophages or monocytes, including but not limited to CSF-1R antagonists such as CSF-1R antagonist antibodies including RG7155 (WO11/70024, WO11/107553, WO11/131407, WO13/87699, WO13/119716, WO13/132044) or FPA-008 (WO11/140249; WO13169264; WO14/036357). In some embodiments, a compound of the present disclosure can be used with one or more of agonistic agents that ligate positive costimulatory receptors, blocking agents that attenuate signaling through inhibitory receptors, antagonists, and one or more agents that increase systemically the frequency of anti-tumor T cells, agents that overcome distinct immune suppressive pathways within the tumor microenvironment (e.g., block inhibitory receptor engagement (e.g., PD-L1/PD-1 interactions), deplete or inhibit Tregs (e.g., using an anti-CD25 monoclonal antibody (e.g., daclizumab) or by ex vivo anti-CD25 bead depletion), inhibit metabolic enzymes such as IDO, or reverse/prevent T cell anergy or exhaustion) and agents that trigger innate immune activation and/or inflammation at tumor sites. In some embodiments, the immuno-oncology agent is a CTLA-4 antagonist, such as an antagonistic CTLA-4 antibody. Suitable CTLA-4 antibodies include, for example, YERVOY (ipilimumab) or tremelimumab. In some embodiments, the immuno-oncology agent is a PD-1 antagonist, such as an antagonistic PD-1 antibody. Suitable PD-1 antibodies include, for example, OPDIVO (nivolumab), KEYTRUDA (pembrolizumab), or MEDI-0680 (AMP-514; WO2012/145493). The immuno-oncology agent may also include pidilizumab (CT-011), though its specificity for PD-1 binding has been questioned. Another approach to target the PD-1 receptor is the recombinant protein composed of the extracellular domain of PD-L2 (B7-DC) fused to the Fe portion of IgG1, called AMP-224. In some embodiments, the immuno-oncology agent is a PD-L1 antagonist, such as an antagonistic PD-L1 antibody. Suitable PD-L1 antibodies include, for example, MPDL3280A (RG7446; WO2010/077634), durvalumab (MEDI4736), BMS-936559 (WO2007/005874), and MSB0010718C (WO2013/79174). In some embodiments, the immuno-oncology agent is a LAG-3 antagonist, such as an antagonistic LAG-3 antibody. Suitable LAG3 antibodies include, for example, BMS-986016 (WO10/19570, WO14/08218), or IMP-731 or IMP-321 (WO08/132601, WO09/44273). In some embodiments, the immuno-oncology agent is a CD137 (4-1BB) agonist, such as an agonistic CD137 antibody. Suitable CD137 antibodies include, for example, urelumab and PF-05082566 (WO12/32433). In some embodiments, the immuno-oncology agent is a GITR agonist, such as an agonistic GITR antibody. Suitable GITR antibodies include, for example, BMS-986153, BMS-986156, TRX-518 (WO06/105021, WO09/009116) and MK-4166 (WO11/028683). In some embodiments, the immuno-oncology agent is an IDO antagonist. Suitable IDO antagonists include, for example, INCB-024360 (WO2006/122150, WO07/75598, WO08/36653, WO08/36642), indoximod, BMS-986205, orNLG-919 (WO09/73620, WO09/1156652, WO11/56652, WO12/142237). In some embodiments, the immuno-oncology agent is an OX40 agonist, such as an agonistic OX40 antibody. Suitable OX40 antibodies include, for example, MEDI-6383 or MEDI-6469. In another aspect, the immuno-oncology agent is an OX40L antagonist, such as an antagonistic OX40 antibody. Suitable OX40L antagonists include, for example, RG-7888 (WO06/029879). In some embodiments, the immuno-oncology agent is a CD40 agonist, such as an agonistic CD40 antibody. In yet another embodiment, the immuno-oncology agent is a CD40 antagonist, such as an antagonistic CD40 antibody. Suitable CD40 antibodies include, for example, lucatumumab or dacetuzumab. In some embodiments, the immuno-oncology agent is a CD27 agonist, such as an agonistic CD27 antibody. Suitable CD27 antibodies include, for example, varlilumab. In some embodiments, the immuno-oncology agent is MGA271 (to B7H3) (WO11/109400).
Combination therapy also can include the administration of the therapeutic agents as described above in further combination with other biologically active ingredients and/or non-drug therapies (e.g., surgery or radiation treatment.)
Suitable antiviral agents contemplated for use in combination with the compound of the present disclosure can comprise nucleoside and nucleotide reverse transcriptase inhibitors (NRTIs), non-nucleoside reverse transcriptase inhibitors (NNRTIs), protease inhibitors and other antiviral drugs. Examples of suitable NRTIs include zidovudine (AZT); didanosine (ddl); zalcitabine (ddC); stavudine (d4T); lamivudine (3TC); abacavir (1592U89); adefovir dipivoxil [bis(POM)-PMEA]; lobucavir; BCH-I0652; emitricitabine [(−)-FTC]; beta-L-FD4 (also called beta-L-D4C and named beta-L-2¢,3¢-dicleoxy-5-fluoro-cytidene); DAPD, ((−)-beta-D-2,6-diamino-purine dioxolane); and lodenosine (FddA). Typical suitable NNRTIs include nevirapine (BI-RG-587); delaviradine (BHAP, U-90152); efavirenz (DMP-266); PNU-142721; AG-1549; MKC-442 (1-(ethoxy-methyl)-5-(1-methylethyl)-6-(phenylmethyl)-(2,4(1H,3H)-pyrimidinedione); and (+)-calanolide A (NSC-675451) and B. Typical suitable protease inhibitors include saquinavir (Ro 31-8959); ritonavir (ABT-538); indinavir (MK-639); nelfnavir (AG-1343); amprenavir (141W94); lasinavir; DMP-450; BMS-2322623; ABT-378; and AG-1549. Other antiviral agents include hydroxyurea, ribavirin, IL-2, IL-12, pentafuside and Yissum Project No. 11607.
Additional combination therapies such as additional immune-oncology agents also include any of those described as suitable for combination with DGK inhibitors in any of the following published patent applications: WO2019005883; WO2020006016; WO2020006018; WO2021041588; WO2021105115; WO2021105116; WO2021105117; WO2021127554; WO2021130638; WO2021132422; WO2021133748; WO2021133749; WO2021133750; WO2021133751; WO2021133752; WO2021214019; or WO2021214020.
The administering herein is not limited to any particular route of administration. For example, in some embodiments, the administering can be orally, nasally, transdermally, pulmonary, inhalationally, buccally, sublingually, intraperintoneally, subcutaneously, intramuscularly, intravenously, rectally, intrapleurally, intrathecally and parenterally. In some embodiments, the administering is orally.
Dosing regimen including doses can vary and can be adjusted, which can depend on the recipient of the treatment, the disease or disorder being treated and the severity thereof, the composition containing the compound, the time of administration, the route of administration, the duration of treatment, the compound potency, its rate of clearance and whether or not another drug is co-administered.
It is meant to be understood that proper valences are maintained for all moieties and combinations thereof.
It is also meant to be understood that a specific embodiment of a variable moiety herein can be the same or different as another specific embodiment having the same identifier.
The present disclosure encompasses all combinations of the aspects and/or embodiments of the disclosure herein. It is understood that any and all embodiments of the present disclosure may be taken in conjunction with any other embodiment or embodiments to describe additional embodiments. It is also to be understood that each individual element of the embodiments is meant to be combined with any and all other elements from any embodiment to describe an additional embodiment.
Suitable atoms or groups for the variables herein are independently selected. The definitions of the variables can be combined. Using Formula III-4 as an example, any of the definitions of one of R1, R2, R3, R9, R30, U, Y, Z, n1, and L2, in Formula III-4 can be combined with any of the definitions of the others of R1, R2, R3, R9, R30, U, Y, Z, n1, and L2, in Formula III-4. Such combination is contemplated and within the scope of the present disclosure. Non-limiting useful groups for the variables in compounds of Formula III-1 to III-12, or a subformula thereof, as applicable, include any of the respective groups, individually or in any combination, as shown in the Examples or in the specific compounds described in Table A herein.
Although well understood in the field, to be clear, when a formula herein is said to be a sub-formula of a broader formula, it should be understood that for a variable(s) that is defined in connection with the broader formula, such variable(s) for the sub-formula can have the same definition as any of those defined for the broader formula, and the preferred definition of such variable(s) for the sub-formula can also include the same preferred definition as any of those described for the broader formula, unless obviously contrary from context. Similarly, for a variable(s) that is defined in connection with the sub-formula, such variable(s) for the broader formula or a different sub-formula of the broader formula can have the same definition as any of those defined for the sub-formula, unless obviously contrary from context.
The symbol, when displayed perpendicular to (or otherwise crossing) a bond, indicates the point at which the displayed moiety is attached to the remainder of the molecule. It should be noted that for a divalent structure (or multivalent structure), the immediately connected group or groups or appropriate variable(s) shown in a formula maybe shown in the divalent structure (or multivalent structure) beyond the symbol,
, to indicate direction of attachment. When the immediately connected group(s) or variable is not shown for either of the two attaching points of a divalent structure, it should mean that either direction of attachment to the remainder of the molecule is allowed, unless otherwise specified or obviously contrary from context. Using a structure of “X-A-G-B” to illustrate, for example, if G is defined as
i.e., the immediately connected group(s) or variable(s) is not shown, then the structure of “X-A-G-B” can be either
on the other hand, if G is defined as
then the structure of “X-A-G-B” should be understood as
Definitions of specific functional groups and chemical terms are described in more detail below. The chemical elements are identified in accordance with the Periodic Table of the Elements, CAS version, Handbook of Chemistry and Physics, 75th Ed., inside cover, and specific functional groups are generally defined as described therein. Additionally, general principles of organic chemistry, as well as specific functional moieties and reactivity, are described in Thomas Sorrell, Organic Chemistry, University Science Books, Sausalito, 1999; Smith and March, March's Advanced Organic Chemistry, 5th Edition, John Wiley & Sons, Inc., New York, 2001; Larock, Comprehensive Organic Transformations, VCH Publishers, Inc., New York, 1989; and Carruthers, Some Modern Methods of Organic Synthesis, 3rd Edition, Cambridge University Press, Cambridge, 1987. The disclosure is not intended to be limited in any manner by the exemplary listing of substituents described herein.
Compounds of the present disclosure can comprise one or more asymmetric centers and/or axial chirality, and thus can exist in various isomeric forms, e.g., enantiomers and/or diastereomers. For example, the compounds described herein can be in the form of an individual enantiomer, diastereomer, atropisomer, or geometric isomer, or can be in the form of a mixture of stereoisomers, including racemic mixtures and mixtures enriched in one or more stereoisomer. Isomers can be isolated from mixtures by methods known to those having ordinary skill in the art, including chiral high performance liquid chromatography (HPLC) and the formation and crystallization of chiral salts; or preferred isomers can be prepared by asymmetric syntheses. See, for example, Jacques et al., Enantiomers, Racemates and Resolutions (Wiley Interscience, New York, 1981); Wilen et al., Tetrahedron 33:2725 (1977); Eliel, Stereochemistry of Carbon Compounds (McGraw Hill, NY, 1962); and Wilen, Tables of Resolving Agents and Optical Resolutions p. 268 (E. L. Eliel, Ed., Univ. of Notre Dame Press, Notre Dame, IN 1972). The disclosure additionally encompasses compounds described herein as individual isomers substantially free of other isomers, and alternatively, as mixtures of various isomers including racemic mixtures. In embodiments herein, unless otherwise obviously contrary from context, when a stereochemistiy is specifically drawn, it should be understood that with respect to that particular chiral center or axial chirality, the compound can exist predominantly as the as-drawn stereoisomer, such as with less than 20%, less than 10%, less than 5%, less than 1%, by weight, by HPLC or SFC area, or both, or with a non-detectable amount of the other stereoisomer(s), for example, the compound can be characterized as having an enantiomeric excess (“ee”) of greater than 60%, such as greater than 80% ee, greater than 90% ee, greater than 95% ee, greater than 98% ee, or greater than 99% ee. The presence and/or amounts of stereoisomers can be determined by those having ordinary skill in the art in view of the present disclosure, including through the use of chiral HPLC or SFC. When the stereochemistry of a chiral center is not specifically drawn, it should be understood that the structure or a fragment thereof is intended to encompass all possible stereoisomers with respect to that particular chiral center.
When a range of values is listed, it is intended to encompass each value and sub-range within the range. For example “C1-6” is intended to encompass, C1, C2, C3, C4, C5, C6, C1-6, C1-5, C1-4, C1-3, C1-2, C2-6, C2-5, C2-4, C2-3, C3-6, C3-5, C3-4, C4-6, C4-5, and C5-6.
As used herein, the term “compound(s) of the present disclosure” or “compound(s) of the present invention” refers to any of the compounds described herein according to III-1, III-2 (e.g., III-2-A, III-2-B, III-2-C, III-2-C-1, III-2-C-2, III-2-C-3, III-2-C-4, III-2-D, III-2-D-1, III-2-D-2, III-2-D-3, III-2-D-4, III-2-E-1, III-2-E-2, III-2-E-3, III-2-E-1a, III-2-E-2a, III-2-E-3a, III-2-E-1b, III-2-E-2b, III-2-E-3b, III-2-F-1a, III-2-F-1b, III-2-F-2a, III-2-F-2b, III-2-F-3a, or III-2-F-3b), III-3 (e.g., III-3-A), III-4 (e.g., III-4-A, III-4-B, III-4-C, III-4-D, III-4-C-1, III-4-C-2, III-4-C-3, III-4-C-4, III-4-D-1, III-4-D-2, III-4-C-4a, III-4-C-4b, III-4-C-4c, III-4-C-4d, III-4-C-4e, or III-4-C-4f), III-5, III-6, III-7, III-8 (e.g., III-8-A or III-8-B), III-9, III-10, 11-11, or III-12, any of Compound Nos. 1-347, any compound selected from the compounds shown in Table A herein, isotopically labeled compound(s) thereof (such as a deuterated analog wherein one or more of the hydrogen atoms is substituted with a deuterium atom with an abundance above its natural abundance), possible stereoisomers thereof (including diastereoisomers, enantiomers, and racemic mixtures), geometric isomers thereof, atropisomers thereof, tautomers thereof, conformational isomers thereof, and/or pharmaceutically acceptable salts thereof (e.g., acid addition salt such as HCl salt or base addition salt such as Na salt). For the avoidance of doubt, Compound Nos. 1-347 or Compounds 1-347 refers to the compounds described herein labeled as integers 1, 2, 3, . . . , 347, see for example the title compounds of Examples and Table 1. For ease of description, synthetic starting materials or intermediates may be labeled with an integer (compound number) followed by a “−” and additional numeric values, such as 2-1, 2-2, etc., see examples for details. The labeling of such synthetic starting materials or intermediates should not be confused with the compounds labeled with an integer only without the “−” and additional numeric value. Hydrates and solvates of the compounds of the present disclosure are considered compositions of the present disclosure, wherein the compound(s) is in association with water or solvent, respectively. In some embodiments, the compound of the present disclosure can be any of those defined in original claims 1-63 herein. In some embodiments, the compound of the present disclosure can be any of those defined in Embodiments 1-49 herein. In some embodiments, the compound of the present disclosure can be any of those defined in any of the enumerated embodiments A1-A38 and B1-B45.
Compounds of the present disclosure can exist in isotope-labeled or -enriched form containing one or more atoms having an atomic mass or mass number different from the atomic mass or mass number most abundantly found in nature. Isotopes can be radioactive or non-radioactive isotopes. Isotopes of atoms such as hydrogen, carbon, phosphorous, sulfur, fluorine, chlorine, and iodine include, but are not limited to 2H, 3H, 13C, 14C, 15N, 18O, 32P, 35S, 18F, 36Cl, and 25I. Compounds that contain other isotopes of these and/or other atoms are within the scope of this invention.
As used herein, the phrase “administration” of a compound, “administering” a compound, or other variants thereof means providing the compound or a prodrug of the compound to the individual in need of treatment.
As used herein, the term “alkyl” as used by itself or as part of another group refers to a straight- or branched-chain aliphatic saturated hydrocarbon. In some embodiments, the alkyl which can include one to twelve carbon atoms (i.e., C1-12 alkyl) or the number of carbon atoms designated (i.e., a C1 alkyl such as methyl, a C2 alkyl such as ethyl, a C3 alkyl such as propyl or isopropyl, etc.). In one embodiment, the alkyl group is a straight chain C1-10 alkyl group. In another embodiment, the alkyl group is a branched chain C3-10 alkyl group. In another embodiment, the alkyl group is a straight chain C1-6 alkyl group. In another embodiment, the alkyl group is a branched chain C3-6 alkyl group. In another embodiment, the alkyl group is a straight chain C1-4 alkyl group. In one embodiment, the alkyl group is a C1-4 alkyl group selected from methyl, ethyl, propyl (n-propyl), isopropyl, butyl (n-butyl), sec-butyl, tert-butyl, and iso-butyl. As used herein, the term “alkylene” as used by itself or as part of another group refers to a divalent radical derived from an alkyl group. For example, non-limiting straight chain alkylene groups include —CH2—CH2—CH2—CH2—, —CH2—CH2—CH2—, —CH2—CH2—, and the like.
As used herein, the term “alkenyl” as used by itself or as part of another group refers to a straight- or branched-chain aliphatic hydrocarbon containing one or more, such as one, two or three carbon-to-carbon double bonds. In one embodiment, the alkenyl group is a C2-6 alkenyl group. In another embodiment, the alkenyl group is a C2-4 alkenyl group. Non-limiting exemplary alkenyl groups include ethenyl, propenyl, isopropenyl, butenyl, sec-butenyl, pentenyl, and hexenyl.
As used herein, the term “alkynyl” as used by itself or as part of another group refers to a straight- or branched-chain aliphatic hydrocarbon containing one or more, such as one to three carbon-to-carbon triple bonds. In one embodiment, the alkynyl has one carbon-carbon triple bond. In one embodiment, the alkynyl group is a C2-6 alkynyl group. In another embodiment, the alkynyl group is a C2-4 alkynyl group. Non-limiting exemplary alkynyl groups include ethynyl, propynyl, butynyl, 2-butynyl, pentynyl, and hexynyl groups.
As used herein, the term “alkoxy” as used by itself or as part of another group refers to a radical of the formula ORa1, wherein Rai is an alkyl. As used herein, the term “cycloalkoxy” as used by itself or as part of another group refers to a radical of the formula ORa1, wherein Ra1 is a cycloalkyl.
As used herein, the term “haloalkyl” as used by itself or as part of another group refers to an alkyl substituted with one or more fluorine, chlorine, bromine and/or iodine atoms. In preferred embodiments, the haloalkyl is an alkyl group substituted with one or more fluorine atoms, alternatively referred to herein as fluorine-substituted alkyl, such as with one, two, or three fluorine atoms. In one embodiment, the haloalkyl group is a C1-4 haloalkyl group. In one embodiment, the haloalkyl group is a fluorine-substituted C1-4 alkyl group.
As used herein, the term “heteroalkyl,” by itself or in combination with another term, means, unless otherwise stated, a stable straight or branched-chain alkyl group, e.g., having from 2 to 14 carbons, such as 2 to 10 carbons in the chain, one or more of the carbons has been replaced by a heteroatom selected from S, O, P and N, and wherein the nitrogen, phosphine, and sulfur atoms can optionally be oxidized and the nitrogen heteroatom can optionally be quaternized. The heteroatom(s) S, O, P and N may be placed at any interior position of the heteroalkyl group or at the position at which the alkyl group is attached to the remainder of the molecule. When the heteroalkyl is said to be substituted, the substituent(s) can replace one or more hydrogen atoms attached to the carbon atom(s) and/or the heteroatom(s) of the heteroalkyl. In some embodiments, the heteroalkyl is a C1-4 heteroalkyl, which refers to the heteroalkyl defined herein having 1-4 carbon atoms. Examples of C1-4 heteroalkyl include, but are not limited to, C4 heteroalkyl such as —CH2—CH2—N(CH3)—CH3, C3 heteroalkyl such as —CH2—CH2—O—CH3, —CH2—CH2—NH—CH3, —CH2—S—CH2—CH3, —CH2—CH2—S(O)—CH3, —CH2—CH2—S(O)2—CH3, C2 heteroalkyl such as —CH2—CH2—OH, —CH2—CH2—NH2, —CH2—NH(CH3), —O—CH2—CH3 and C1 heteroalkyl such as, —CH2—OH, —CH2—NH2, —O—CH3. Preferably, the C1-4 heteroalkyl (or C1-4 heteroalkylene) herein contains 1 or 2 heteroatoms, such as one oxygen, one nitrogen, two oxygens, two nitrogens, or one oxygen and one nitrogen. Similarly, the term “heteroalkylene” by itself or as part of another substituent means a divalent radical derived from heteroalkyl, as exemplified, but not limited by, —CH2—CH2—O—CH2—CH2— and —O—CH2—CH2—NH—CH2—. For heteroalkylene groups, heteroatoms can also occupy either or both of the chain termini (e.g., alkyleneoxy, alkylenedioxy, alkyleneamino, alkylenediamino, and the like). Still further, for alkylene and heteroalkylene linking groups, no orientation of the linking group is implied by the direction in which the formula of the linking group is written. Where “heteroalkyl” is recited, followed by recitations of specific heteroalkyl groups, such as —NR′R″ or the like, it will be understood that the terms heteroalkyl and —NR′R″ are not redundant or mutually exclusive. Rather, the specific heteroalkyl groups are recited to add clarity. Thus, the term “heteroalkyl” should not be interpreted herein as excluding specific heteroalkyl groups, such as —NR′R″ or the like.
“Carbocyclyl” or “carbocyclic” as used by itself or as part of another group refers to a radical of a non-aromatic cyclic hydrocarbon group having from 3 to 10 ring carbon atoms (“C3-10 carbocyclyl”) and zero heteroatoms in the non-aromatic ring system. The carbocyclyl group can be either monocyclic (“monocyclic carbocyclyl”) or contain a fused, bridged or spiro ring system such as a bicyclic system (“bicyclic carbocyclyl”) and can be saturated or can be partially unsaturated. “Carbocyclyl” also includes ring systems wherein the carbocyclic ring, as defined above, is fused with one or more aryl or heteroaryl groups wherein the point of attachment is on the carbocyclic ring, and in such instances, the number of carbons continue to designate the number of carbons in the carbocyclic ring system. Non-limiting exemplary carbocyclyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, norbornyl, decalin, adamantyl, cyclopentenyl, and cyclohexenyl.
In some embodiments, “carbocyclyl” is fully saturated, which is also referred to as cycloalkyl. In some embodiments, the cycloalkyl can have from 3 to 10 ring carbon atoms (“C3-10 cycloalkyl”). In preferred embodiments, the cycloalkyl is a monocyclic ring.
“Heterocyclyl” or “heterocyclic” as used by itself or as part of another group refers to a radical of a 3- to 10-membered non-aromatic ring system having ring carbon atoms and 1 to 4 ring heteroatoms, wherein each heteroatom is independently selected from nitrogen, oxygen, sulfur, boron, phosphorus, and silicon (“3-10 membered heterocyclyl”). Heterocyclyl or heterocyclic ring that has a ring size different from the 3-10 membered heterocyclyl is specified with a different ring size designation when applicable. Those having ordinary skill in the art would understand that such different ring-sized heterocyclyl is also a non-aromatic ring system having ring carbon atoms and 1 to 4 ring heteroatoms, wherein each heteroatom is independently selected from nitrogen, oxygen, sulfur, boron, phosphorus, and silicon. In heterocyclyl groups that contain one or more nitrogen atoms, the point of attachment can be a carbon or nitrogen atom, as valency permits. A heterocyclyl group can either be monocyclic (“monocyclic heterocyclyl”) or a fused, bridged, or spiro ring system, such as a bicyclic system (“bicyclic heterocyclyl”), and can be saturated or can be partially unsaturated. Heterocyclyl bicyclic ring systems can include one or more heteroatoms in one or both rings. “Heterocyclyl” also includes ring systems wherein the heterocyclic ring, as defined above, is fused with one or more carbocyclyl groups wherein the point of attachment is either on the carbocyclyl or heterocyclic ring, or ring systems wherein the heterocyclic ring, as defined above, is fused with one or more aryl or heteroaryl groups, wherein the point of attachment is on the heterocyclic ring, and in such instances, the number of ring members continue to designate the number of ring members in the heterocyclic ring system.
Exemplary 3-membered heterocyclyl groups containing one heteroatom include, without limitation, azirdinyl, oxiranyl, thiiranyl. Exemplary 4-membered heterocyclyl groups containing one heteroatom include, without limitation, azetidinyl, oxetanyl and thietanyl. Exemplary 5-membered heterocyclyl groups containing one heteroatom include, without limitation, tetrahydrofuranyl, dihydrofuranyl, tetrahydrothiophenyl, dihydrothiophenyl, pyrrolidinyl, dihydropyrrolyl, and pyrrolyl-2,5-dione. Exemplary 5-membered heterocyclyl groups containing two heteroatoms include, without limitation, dioxolanyl, oxasulfuranyl, disulfuranyl, and oxazolidin-2-one. Exemplary 5-membered heterocyclyl groups containing three heteroatoms include, without limitation, triazolinyl, oxadiazolinyl, and thiadiazolinyl. Exemplary 6-membered heterocyclyl groups containing one heteroatom include, without limitation, piperidinyl, tetrahydropyranyl, dihydropyridinyl, and thianyl. Exemplary 6-membered heterocyclyl groups containing two heteroatoms include, without limitation, piperazinyl, morpholinyl, dithianyl, and dioxanyl. Exemplary 6-membered heterocyclyl groups containing three heteroatoms include, without limitation, triazinanyl. Exemplary 7-membered heterocyclyl groups containing one heteroatom include, without limitation, azepanyl, oxepanyl and thiepanyl. Exemplary 8-membered heterocyclyl groups containing one heteroatom include, without limitation, azocanyl, oxecanyl and thiocanyl. Exemplary 5-membered heterocyclyl groups fused to a C6 aryl ring (also referred to herein as a 5,6-bicyclic heterocyclic ring) include, without limitation, indolinyl, isoindolinyl, dihydrobenzofuranyl, dihydrobenzothienyl, benzoxazolinonyl, and the like. Exemplary 6-membered heterocyclyl groups fused to an aryl ring (also referred to herein as a 6,6-bicyclic heterocyclic ring) include, without limitation, tetrahydroquinolinyl, tetrahydroisoquinolinyl, and the like.
“Aryl” as used by itself or as part of another group refers to a radical of a monocyclic or polycyclic (e.g., bicyclic or tricyclic) 4n+2 aromatic ring system (e.g., having 6, 10, or 14 pi electrons shared in a cyclic array) having 6-14 ring carbon atoms and zero heteroatoms provided in the aromatic ring system (“C6-14 aryl”). In some embodiments, an aryl group has six ring carbon atoms (“C6 aryl”; e.g., phenyl). In some embodiments, an aryl group has ten ring carbon atoms (“C10 aryl”; e.g., naphthyl such as 1-naphthyl and 2-naphthyl). In some embodiments, an aryl group has fourteen ring carbon atoms (“C1-4 aryl”; e.g., anthracyl). “Aryl” also includes ring systems wherein the aryl ring, as defined above, is fused with one or more carbocyclyl or heterocyclyl groups wherein the radical or point of attachment is on the aryl ring, and in such instances, the number of carbon atoms continue to designate the number of carbon atoms in the aryl ring system.
“Aralkyl” as used by itself or as part of another group refers to an alkyl substituted with one or more aryl groups, preferably, substituted with one aryl group. Examples of aralkyl include benzyl, phenethyl, etc. When an aralkyl is said to be optionally substituted, either the alkyl portion or the aryl portion of the aralkyl can be optionally substituted.
“Heteroaryl” as used by itself or as part of another group refers to a radical of a 5-10 membered monocyclic or bicyclic 4n+2 aromatic ring system (e.g., having 6 or 10 pi electrons shared in a cyclic array) having ring carbon atoms and 1-4 ring heteroatoms provided in the aromatic ring system, wherein each heteroatom is independently selected from nitrogen, oxygen and sulfur (“5-10 membered heteroaryl”). Heteroaryl that has a ring size different from the 5-10 membered heteroaryl is specified with a different ring size designation when applicable. Those having ordinary skill in the art would understand that such different ring-sized heteroaryl is also a 4n+2 aromatic ring system (e.g., having 6 or 10 pi electrons shared in a cyclic array) having ring carbon atoms and 1-4 ring heteroatoms provided in the aromatic ring system, wherein each heteroatom is independently selected from nitrogen, oxygen and sulfur. In heteroaryl groups that contain one or more nitrogen atoms, the point of attachment can be a carbon or nitrogen atom, as valency permits. Heteroaryl bicyclic ring systems can include one or more heteroatoms in one or both rings. “Heteroaryl” includes ring systems wherein the heteroaryl ring, as defined above, is fused with one or more carbocyclyl or heterocyclyl groups wherein the point of attachment is on the heteroaryl ring, and in such instances, the number of ring members continue to designate the number of ring members in the heteroaryl ring system. “Heteroaryl” also includes ring systems wherein the heteroaryl ring, as defined above, is fused with one or more aryl groups wherein the point of attachment is either on the aryl or heteroaryl ring, and in such instances, the number of ring members designates the number of ring members in the fused (aryl/heteroaryl) ring system. Bicyclic heteroaryl groups wherein one ring does not contain a heteroatom (e.g., indolyl, quinolinyl, and the like) the point of attachment can be on either ring, i.e., either the ring bearing a heteroatom (e.g., 2-indolyl) or the ring that does not contain a heteroatom (e.g., 5-indolyl).
Exemplary 5-membered heteroaryl groups containing one heteroatom include, without limitation, pyrrolyl, furanyl, and thiophenyl. Exemplary 5-membered heteroaryl groups containing two heteroatoms include, without limitation, imidazolyl, pyrazolyl, oxazolyl, isoxazolyl, thiazolyl, and isothiazolyl. Exemplary 5-membered heteroaryl groups containing three heteroatoms include, without limitation, triazolyl, oxadiazolyl, and thiadiazolyl. Exemplary 5-membered heteroaryl groups containing four heteroatoms include, without limitation, tetrazolyl. Exemplary 6-membered heteroaryl groups containing one heteroatom include, without limitation, pyridinyl. Exemplary 6-membered heteroaryl groups containing two heteroatoms include, without limitation, pyridazinyl, pyrimidinyl, and pyrazinyl. Exemplary 6-membered heteroaryl groups containing three or four heteroatoms include, without limitation, triazinyl and tetrazinyl, respectively. Exemplary 7-membered heteroaryl groups containing one heteroatom include, without limitation, azepinyl, oxepinyl, and thiepinyl. Exemplary 5,6-bicyclic heteroaryl groups include, without limitation, indolyl, isoindolyl, indazolyl, benzotriazolyl, benzothiophenyl, isobenzothiophenyl, benzofuranyl, benzoisofuranyl, benzimidazolyl, benzoxazolyl, benzisoxazolyl, benzoxadiazolyl, benzothiazolyl, benzisothiazolyl, benzothiadiazolyl, indolizinyl, and purinyl. Exemplary 6,6-bicyclic heteroaryl groups include, without limitation, naphthyridinyl, pteridinyl, quinolinyl, isoquinolinyl, cinnolinyl, quinoxalinyl, phthalazinyl, and quinazolinyl.
“Heteroaralkyl” as used by itself or as part of another group refers to an alkyl substituted with one or more heteroaryl groups, preferably, substituted with one heteroaryl group. When a heteroaralkyl is said to be optionally substituted, either the alkyl portion or the heteroaryl portion of the heteroaralkyl can be optionally substituted.
As commonly understood in the art, alkylene, alkenylene, alkynylene, heteroalkylene, carbocyclylene, heterocyclylene, arylene, and heteroarylene refer to the corresponding divalent radicals of alkyl, alkenyl, alkynyl, heteroalkyl, carbocyclyl, heterocyclyl, aryl, and heteroaryl groups, respectively.
An “optionally substituted” group, such as an optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted heteroalkyl, optionally substituted carbocyclyl, optionally substituted heterocyclyl, optionally substituted aryl, and optionally substituted heteroaryl groups, refers to the respective group that is unsubstituted or substituted. In general, the term “substituted”, whether preceded by the term “optionally” or not, means that at least one hydrogen present on a group (e.g., a carbon or nitrogen atom) is replaced with a permissible substituent, e.g., a substituent which upon substitution results in a stable compound, e.g., a compound which does not spontaneously undergo transformation such as by rearrangement, cyclization, elimination, or other reaction. Unless otherwise indicated, a “substituted” group has a substituent at one or more substitutable positions of the group, and when more than one position in any given structure is substituted, the substituent can be the same or different at each position. Typically, when substituted, the optionally substituted groups herein can be substituted with 1-5 substituents. Substituents can be a carbon atom substituent, a nitrogen atom substituent, an oxygen atom substituent or a sulfur atom substituent, as applicable.
Unless expressly stated to the contrary, combinations of substituents and/or variables are allowable only if such combinations are chemically allowed and result in a stable compound. A “stable” compound is a compound that can be prepared and isolated and whose structure and properties remain or can be caused to remain essentially unchanged for a period of time sufficient to allow use of the compound for the purposes described herein (e.g., therapeutic administration to a subject).
In some embodiments, the “optionally substituted” alkyl, alkenyl, alkynyl, heteroalkyl, carbocyclic, cycloalkyl, alkoxy, cycloalkoxy, or heterocyclic group herein can be unsubstituted or substituted with 1, 2, 3, or 4 substituents independently selected from F, Cl, —OH, protected hydroxyl, oxo (as applicable), NH2, protected amino, NH(C1-4 alkyl) or a protected derivative thereof, N(C1-4 alkyl((C1-4 alkyl), C1-4 alkyl, C2-4 alkenyl, C2-4 alkynyl, C1-4 alkoxy, C3-6 cycloalkyl, C3-6 cycloalkoxy, phenyl, 5 or 6 membered heteroaryl containing 1, 2, or 3 ring heteroatoms independently selected from O, S, and N, 3-7 membered heterocyclyl containing 1 or 2 ring heteroatoms independently selected from O, S, and N, wherein each of the alkyl, alkenyl, alkynyl, alkoxy, cycloalkyl, cycloalkoxy phenyl, heteroaryl, and heterocyclyl, is optionally substituted with 1, 2, or 3 substituents independently selected from F, —OH, oxo (as applicable), C1-4 alkyl, fluoro-substituted C1-4 alkyl (e.g., CF3), C1-4 alkoxy and fluoro-substituted C1-4 alkoxy. In some embodiments, the “optionally substituted” aryl or heteroaryl group herein can be unsubstituted or substituted with 1, 2, 3, or 4 substituents independently selected from F, Cl, —OH, —CN, NH2, protected amino, NH(C1-4 alkyl) or a protected derivative thereof, N(C1-4 alkyl((C1-4 alkyl), —S(═O)(C1-4 alkyl), —SO2(C1-4 alkyl), C1-4 alkyl, C2-4 alkenyl, C2-4 alkynyl, C1-4 alkoxy, C3-6 cycloalkyl, C3-6 cycloalkoxy, phenyl, 5 or 6 membered heteroaryl containing 1, 2 or 3 ring heteroatoms independently selected from O, S, and N, 3-7 membered heterocyclyl containing 1 or 2 ring heteroatoms independently selected from O, S, and N, wherein each of the alkyl, alkenyl, alkynyl, alkoxy, cycloalkyl, cycloalkoxy, phenyl, heteroaryl, and heterocyclyl, is optionally substituted with 1, 2, or 3 substituents independently selected from F, —OH, oxo (as applicable), C1-4 alkyl, fluoro-substituted C1-4 alkyl, C1-4 alkoxy and fluoro-substituted C1-4 alkoxy.
Exemplary carbon atom substituents include, but are not limited to, halogen, —CN, —NO2, —N3, —SO2H, —SO3H, —OH, —ORaa, —ON(Rbb)2, —N(Rbb)2, —N(Rbb)3+X−, —N(ORcc)Rbb, —SH, —SRaa, —SSRcc, —C(═O)Raa, —CO2H, —CHO, —C(ORcc)2, —CO2Raa, —OC(═O)Raa, —OCO2Raa, —C(═O)N(Rbb)2, —OC(═O)N(Rbb)2, —NRbbC(═O)Raa, —NRbbCO2Raa, —NRbbC(═O)N(Rbb)2, —C(═NRbb)Raa, —C(═NRbb)ORaa, —OC(═NRbb)Raa, —OC(═NRbb)ORaa, —C(═NRbb)N(Rbb)2, —OC(═NRbb)N(Rbb)2, —NRbbC(═NRbb)N(Rbb)2, —C(═O)NRbbSO2Raa, —NRbbSO2Raa, —SO2N(Rbb)2, SO2Raa, —SO2ORaa, —OSO2Raa, —S(═O)Raa, —OS(═O)Raa, —Si(Raa)3, —OSi(Raa)3—C(═S)N(Rbb)2, C(═O)SRaa, —C(═S)SRaa, —SC(═S)SRaa, —SC(═O)SRaa, —OC(═O)SRaa, —SC(═O)ORaa, —SC(═O)Raa, —P(═O)(Raa)2, —P(═O)(ORcc)2, —OP(═O)(Raa)2, —OP(═O)(ORcc)2, —P(═O)(N(Rbb)2)2, —OP(═O)(N(Rbb)2)2, —NRbbP(═O)(Raa)2, —NRbbP(═O)(OR″ °)2, —NRbbP(═O)(N(Rbb)2)2, —P(Rcc)2, —P(ORcc)2, —P(RC)3+X−, —P(ORcc)3+X−, —P(Rcc)4, —P(ORcc)4, —OP(Rcc)2, —OP(Rcc)3+X−, —OP(ORcc)2, —OP(ORcc)3X—, —OP(Rcc)4, —OP(ORcc)4, —B(Raa)2, —B(ORcc)2, —BRaa(ORcc), C1-10 alkyl, C1-10 haloalkyl, C2-10 alkenyl, C2-10 alkynyl, C3-10 carbocyclyl, 3-14 membered heterocyclyl, C6-14 aryl, and 5-14 membered heteroaryl, wherein each alkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl, and heteroaryl is independently substituted with 0, 1, 2, 3, 4, or 5 Rdd groups; wherein X− is a counterion;
A “counterion” or “anionic counterion” is a negatively charged group associated with a positively charged group in order to maintain electronic neutrality. An anionic counterion may be monovalent (i.e., including one formal negative charge). An anionic counterion may also be multivalent (i.e., including more than one formal negative charge), such as divalent or trivalent. Exemplary counterions include halide ions (e.g., F−, Cl−, Br−, I−), NO3−, ClO4−, OH−, H2PO4−, HSO4−, sulfonate ions (e.g., methansulfonate, trifluoromethanesulfonate, p-toluenesulfonate, benzenesulfonate, 10-camphor sulfonate, naphthalene-2-sulfonate, naphthalene-1-sulfonic acid-5-sulfonate, ethan-1-sulfonic acid-2-sulfonate, and the like), carboxylate ions (e.g., acetate, propanoate, benzoate, glycerate, lactate, tartrate, glycolate, gluconate, and the like), BF4−, PF4−, PF6−, AsF6−, SbF6−, B[3,5-(CF3)2C6H3]4]−, BPh4−, Al(OC(CF3)3)4−, and a carborane anion (e.g., CB11H12− or (HCB11Me5Br6)−). Exemplary counterions which may be multivalent include CO32−, HPO42−, PO43−, B4O72−, S42−, S2O32−, carboxylate anions (e.g., tartrate, citrate, fumarate, maleate, malate, malonate, gluconate, succinate, glutarate, adipate, pimelate, suberate, azelate, sebacate, Sali cylate, phthalates, aspartate, glutamate, and the like), and carboranes.
“Halo” or “halogen” refers to fluorine (fluoro, —F), chlorine (chloro, —Cl), bromine (bromo, —Br), or iodine (iodo, —I).
“Acyl” refers to a moiety selected from the group consisting of —C(═O)Raa, —CHO, —CO2Raa, —C(═O)N(Rbb)2, —C(═NRbb)Raa, —C(═NRbb)ORaa, —C(═NRbb)N(Rbb)2, —C(═O)NRbbSO2Raa, —C(═S)N(Rbb)2, —C(═O)SRaa, or —C(═S)SRaa, wherein Raa and Rbb are as defined herein.
Nitrogen atoms can be substituted or unsubstituted as valency permits, and include primary, secondary, tertiary, and quaternary nitrogen atoms. Exemplary nitrogen atom substituents include, but are not limited to, hydrogen, —OH, —ORaa, —N(RC)2, —CN, —C(═O)Raa, —C(═O)N(Rcc)2, CO2Raa, SO2Raa, —C(═NRbb)Raa, —C(═NRcc)ORaa, —C(═NRcc)N(Rcc)2, SO2N(Rcc)2, —SO2Rcc, —SO2ORcc, —SORaa, —C(═S)N(Rcc)2, —C(═O)SRcc, —C(═S)SRcc, —P(═O)(ORCc)2, —P(═O)(Raa)2, —P(═O)(N(Rcc)2)2, C1-10 alkyl, C1-10 haloalkyl, C2-10 alkenyl, C2-10 alkynyl, C3-10 carbocyclyl, 3-14 membered heterocyclyl, C6-14 aryl, and 5-14 membered heteroaryl, or two Rcc groups attached to a nitrogen atom are joined to form a 3-14 membered heterocyclyl or 5-14 membered heteroaryl ring, wherein each alkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl, and heteroaryl is independently substituted with 0, 1, 2, 3, 4, or 5 Rdd groups, and wherein Raa, Rbb, Rcc, and Rdd are as defined above.
In certain embodiments, the substituent present on a nitrogen atom is a nitrogen protecting group (also referred to as an amino protecting group). Nitrogen protecting groups are well known in the art and include those described in detail in Protective Groups in Organic Synthesis, T. W. Greene and P. G. M. Wuts, 3rd edition, John Wiley & Sons, 1999, incorporated by reference herein. Exemplary nitrogen protecting groups include, but not limited to, those forming carbamates, such as Carbobenzyloxy (Cbz) group, p-Methoxybenzyl carbonyl (Moz or MeOZ) group, tert-Butyloxycarbonyl (BOC) group, Troc, 9-Fluorenylmethyloxycarbonyl (Fmoc) group, etc., those forming an amide, such as acetyl, benzoyl, etc., those forming a benzylic amine, such as benzyl, p-methoxybenzyl, 3,4-dimethoxybenzyl, etc., those forming a sulfonamide, such as tosyl, Nosyl, etc., and others such as p-methoxyphenyl.
Exemplary oxygen atom substituents include, but are not limited to, —Raa, —C(═O)SRaa, —C(═O)Raa, —CO2Raa, —C(═O)N(Rbb)2, —C(═NRbb)Raa, —C(═NRbb)ORaa, —C(═NRbb)N(Rbb)2, —S(═O)Raa, —SO2Raa, —Si(Raa)3, —P(Rcc)2—P(Rcc)3+X−, —P(ORCc)2, —P(ORcc)3+X−, —P(═O)(Rcc)2, —P(═O)(ORcc)2, and —P(═O)(N(Rbb)2)2, wherein X−, Raa, Rbb, and Rcc are as defined herein. In certain embodiments, the oxygen atom substituent present on an oxygen atom is an oxygen protecting group (also referred to as a hydroxyl protecting group). Oxygen protecting groups are well known in the art and include those described in detail in Protective Groups in Organic Synthesis, T. W. Greene and P. G. M. Wuts, 3rd edition, John Wiley & Sons, 1999, incorporated herein by reference. Exemplary oxygen protecting groups include, but are not limited to, alkyl ethers or substituted alkyl ethers such as methyl, allyl, benzyl, substituted benzyls such as 4-methoxybenzyl, methoxymethyl (MOM), benzyloxymethyl (BOM), 2-methoxyethoxymethyl (MEM), etc., silyl ethers such as trymethylsilyl (TMS), triethylsilyl (TES), triisopropylsilyl (TIPS), t-butyldimethylsilyl (TBDMS), etc., acetals or ketals, such as tetrahydropyranyl (THP), esters such as formate, acetate, chloroacetate, dichloroacetate, trichloroacetate, trifluoroacetate, methoxyacetate, etc., carbonates, sulfonates such as methanesulfonate (mesylate), benzylsulfonate, and tosylate (Ts), etc.
The term “leaving group” is given its ordinary meaning in the art of synthetic organic chemistry, for example, it can refer to an atom or a group capable of being displaced by a nucleophile. See, for example, Smith, March Advanced Organic Chemistry 6th ed. (501-502). Examples of suitable leaving groups include, but are not limited to, halogen (such as F, Cl, Br, or I (iodine)), alkoxycarbonyloxy, aryloxycarbonyloxy, alkanesulfonyloxy, arenesulfonyloxy, alkyl-carbonyloxy (e.g., acetoxy), arylcarbonyloxy, aryloxy, methoxy, N,O-dimethylhydroxylamino, pixyl, and haloformates.
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.
The term “tautomers” or “tautomeric” refers to two or more interconvertible compounds resulting from at least one formal migration of a hydrogen atom and at least one change in valency (e.g., a single bond to a double bond, a triple bond to a single bond, or vice versa). The exact ratio of the tautomers depends on several factors, including temperature, solvent, and pH. Tautomerizations (i.e., the reaction providing a tautomeric pair) may catalyzed by acid or base. Exemplary tautomerizations include keto-to-enol, amide-to-imide, lactam-to-lactim, enamine-to-imine, and enamine-to-(a different enamine) tautomerizations.
The term “subject” (alternatively referred to herein as “patient”) as used herein, refers to an animal, preferably a mammal, most preferably a human, who has been the object of treatment, observation or experiment.
As used herein, the terms “treat,” “treating,” “treatment,” and the like refer to eliminating, reducing, or ameliorating a disease or condition, and/or symptoms associated therewith. Although not precluded, treating a disease or condition does not require that the disease, condition, or symptoms associated therewith be completely eliminated. As used herein, the terms “treat,” “treating,” “treatment,” and the like may include “prophylactic treatment,” which refers to reducing the probability of redeveloping a disease or condition, or of a recurrence of a previously-controlled disease or condition, in a subject who does not have, but is at risk of or is susceptible to, redeveloping a disease or condition or a recurrence of the disease or condition. The term “treat” and synonyms contemplate administering a therapeutically effective amount of a compound described herein to a subject in need of such treatment.
As used herein, the singular form “a”, “an”, and “the”, includes plural references unless it is expressly stated or is unambiguously clear from the context that such is not intended.
The term “and/or” as used in a phrase such as “A and/or B” herein is intended to include both A and B; A or B; A (alone); and B (alone). Likewise, the term “and/or” as used in a phrase such as “A, B, and/or C” is intended to encompass each of the following embodiments: A, B, and C; A, B, or C; A or C; A or B; B or C; A and C; A and B; B and C; A (alone); B (alone); and C (alone).
Headings and subheadings are used for convenience and/or formal compliance only, do not limit the subject technology, and are not referred to in connection with the interpretation of the description of the subject technology. Features described under one heading or one subheading of the subject disclosure may be combined, in various embodiments, with features described under other headings or subheadings. Further it is not necessarily the case that all features under a single heading or a single subheading are used together in embodiments.
The various starting materials, intermediates, and compounds of the preferred embodiments can be isolated and purified where appropriate using conventional techniques such as precipitation, filtration, crystallization, evaporation, distillation, and chromatography. Characterization of these compounds can be performed using conventional methods such as by melting point, mass spectrum, nuclear magnetic resonance, and various other spectroscopic analyses. The examples are illustrative only and do not limit the claimed invention in any way.
Exemplary embodiments of steps for performing the synthesis of products described herein are described in greater detail infra. Some of the Examples discussed herein can be prepared by separating the corresponding racemic mixtures. As would be understood by a person of ordinary skill in the art, the compounds described in the Examples section immediately prior to the chiral separation step, e.g., by supercritical fluid chromatography (SFC), exist in racemic and/or stereoisomeric mixture forms. It should be understood that the enantiomeric excesses (“ee”) and/or diastereomeric excesses (“de”) reported for these examples are only representative from the exemplified procedures herein and not limiting; those of ordinary skill in the art would understand that such enantiomers and/or diastereomers with a different ee and/or de, such as a higher ee and/or de, can be obtained in view of the present disclosure. Typically, a “de” value is reported herein when a pair of diastereomers, having only one of the chiral centers being different, are separated from a corresponding diastereomeric mixture. In such cases, the “de” value indicates the degree of enrichment of one of the diastereomers.
The abbreviations used in the Examples section should be understood as having their ordinary meanings in the art unless specifically indicated otherwise or obviously contrary from context. The following shows certain abbreviations used in the Examples section herein.
Step 1: To a solution of 1-1 (2.5 g, 10.4 mmol) in dioxane (20 mL) was added tert-butyl 2,8-diazaspiro[4.5]decane-8-carboxylate (3.0 g, 12.45 mmol), Cs2CO3 (6.76 g, 20.75 mmol) and RuPhos Pd G3 (867.6 mg, 1.4 mmol). The resulting mixture was degassed with N2 for 10 minutes, then stirred at 120° C. for 4 h. After cooled down to room temperature, the mixture was filtered and the filtrate was concentrated. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 3/1) to afford 1-2.
Step 2: To a solution of 1-2 (2.8 g, 7 mmol) in dioxane (20 mL) was added HCl (4.3 mL, 43 mmol). The reaction mixture was stirred at room temperature for 5 h. The pH was adjusted to 6 by addition of saturated NaHCO3. The mixture was extracted with ethyl acetate. The organic layer was dried over Na2SO4, filtered and the filtrate was concentrated. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 0/1) to afford 1-3.
Step 3: To a solution of 1-4 (24 g, 144.4 mmol) in acetonitrile (250 mL) was added NBS (28.3 g, 158.9 mmol) at room temperature. The mixture was stirred at room temperature for 1 h. Water was added, and the mixture was extracted with ethyl acetate. The combined organic layers were dried over Na2SO4, filtered and the filtrated was concentrated. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 0/1) to afford 1-5.
Step 4: To a solution of 2-cyanoacetic acid (16.6 g, 195.2 mmol) in DMF (400 mL) were added HATU (1.39 g, 3.7 mmol) and triethylamine (45.2 mL, 325.4 mmol) at room temperature. The mixture was stirred at room temperature for 15 minutes. Then 1-5 (31.9 g, 130.2 mmol) was added. The mixture was stirred at room temperature for 12 h. Water was added to the reaction mixture. The solid was collected by filtration to afford 1-6.
Step 5: To a solution of 1-6 (21.5 g, 68.9 mmol) in THF (300 mL) was added KHMDS (1M in THF, 89.6 mL, 89.6 mmol) at room temperature. The mixture was stirred at room temperature for 12 h. HCl (1 N) was added until the pH was 4. The solid was collected by filtration to afford 1-7.
Step 6: To a solution of 1-7 (15.8 g, 59.4 mmol) in DMF (200 mL) was added NaH (9.5 g, 60%, 237.6 mmol) in portions at 0. The mixture was stirred at 0
for 30 minutes. CH3I (14.8 mL, 237.6 mmol) was added. The reaction was stirred at room temperature for 12 h, then quenched with H2O. The pH of the reaction mixture was adjusted to 3 with HCl (1 N). The solid was collected by filtration to afford 1-8.
Step 7: To a solution of 1-8 (17.3 g, 61.8 mmol), triethylamine (25.8 mL, 185.3 mmol) and DMAP (0.8 g, 6.2 mmol) in dichloromethane (200 mL) was added Tf2O (34.9 g, 123.5 mmol) at 0 . The mixture was stirred at 0
for 0.5 h, then quenched with H2O and extracted with dichloromethane. The combined organic layers were dried over Na2SO4, filtered and the filtrate was concentrated. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 2/1) to afford 1-9.
Step 8: To a mixture of 1-9 (300 mg, 0.73 mmol) and 1-3 (327.9 mg, 1.1 mmol) in acetonitrile (10 mL) was added DIPEA (0.36 mL, 2.18 mmol). The mixture was stirred at 85 for 2 h. The reaction was quenched by H2O. The aqueous layer was extracted with ethyl acetate. The organic layers were combined and washed with brine, dried over anhydrous Na2SO4, filtered and the filtrate was concentrated. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/1) to afford 1-10.
Step 9: To a solution of 1-10 (210 mg, 0.37 mmol) in NMP (8 mL) were added Zn(CN)2 (87.7 mg, 0.75 mmol), Zn (4.9 mg, 0.075 mmol), Pd2(dba)3 (34.2 mg, 0.037 mmol) and dppf (12.6 mg, 0.022 mmol) under N2. The reaction mixture was stirred at 80 for 1 h. The reaction was quenched by H2O. The aqueous layer was extracted with ethyl acetate. The combined organic layers were washed with brine, dried over anhydrous Na2SO4, filtered and the filtrate was concentrated. The residue was purified by reverse phase HPLC (Reverse phase HPLC (acetonitrile/water: 5-95%) to afford 1. LCMS (ESI, m/z): [M+H]+=509.4; 1H NMR (400 MHz, DMSO-d6, ppm): δ 8.25 (d, J=8.8 Hz, 1H), 8.14 (d, J=8.8 Hz, 1H), 7.12 (d, J=8.4 Hz, 2H), 6.58 (d, J=9.2 Hz, 2H), 3.90-3.85 (m, 4H), 3.52 (s, 3H), 3.38-3.34 (m, 2H), 3.25 (s, 2H), 1.99 (t, J=6.8 Hz, 2H), 1.86-1.80 (m, 4H).
Step 1: To a solution of 2-1 (19 g, 106.7 mmol), tert-butyl 4-hydroxypiperidine-1-carboxylate (21.5 g, 106.7 mmol) and PPh3 (30.8 g, 117.3 mmol) in THF (85 mL) was added DEAD (18.6 mL, 117.3 mmol) at 0 under N2. The mixture was stirred at 20
under N2 for 16 h. The mixture was concentrated. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 4/1) to afford 2-2.
Step 2: To a solution of 2-2 (22.1 g, 61.2 mmol) in dichloromethane (20 mL) was added a solution of HCl in dioxane (4M, 61.2 mL, 244.8 mmol) at 20 . The mixture was stirred at 20
for 16 h. The mixture was concentrated under reduced pressure to afford 2-3.
Step 3: To a mixture of 1-9 (600 mg, 1.46 mmol) and 2-3 (456.4 mg, 1.75 mmol) in acetonitrile (10 mL) was added DIPEA (564.4 mg, 4.37 mmol) at room temperature. The mixture was stirred at 85 for 1 h. This mixture was concentrated and the residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 10/1) to afford 2-4.
Step 4: To a solution of 2-4 (300 mg, 0.57 mmol) and acetaldoxime (609.5 mg, 10.3 mmol) in toluene (15 mL) was added Rh(PPh3)3Cl (53.0 mg, 0.057 mmol) at room temperature. Then the mixture was heated at 110 for 12 h. The mixture was concentrated and the residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 10/1) to afford 2-5.
Step 5: To a mixture of 2-5 (250 mg, 0.46 mmol), Zn (6.0 mg, 0.09 mmol) and Zn(CN)2 (108.5 mg, 0.92 mmol) in NMP (4 mL) were added Pd2(dba)3 (42.3 mg, 0.046 mmol A pf (12.8 mg, 0.023 mmol) at room temperature. Then the mixture was stirred at 90 for 1 h. The reaction mixture was quenched with H2O and extracted with ethyl acetate. The combined organic layers were dried over Na2SO4, filtered and the filtrate was concentrated. The residue was purified by reverse phase HPLC (acetonitrile/0.05% TFA in water: 5%-80%) to afford 2 as 0.2 eq of TFA salt. LCMS (ESI, m/z): [M+H]+=488.4; 1H NMR (400 MHz, DMSO-d6, ppm): δ 8.17 (d, J=8.8 Hz, 1H), 8.08 (d, J=8.9 Hz, 1H), 7.65 (s, 1H), 7.50 (s, 1H), 7.27 (d, J=8.6 Hz, 2H), 7.13-7.08 (m, 2H), 4.73-4.65 (m, 1H), 3.63 (s, 2H), 3.55 (s, 3H), 3.34-3.29 (m, 2H), 2.13-2.10 (m, 2H), 1.86-1.83 (m, 2H). 19F NMR (376 MHz, DMSO-d6, ppm): δ −57.25 (3F), −74.58 (0.6F, TFA).
Step 1: 3-1 (10 g, 81.2 mmol) and piperidine-4-carboxylic acid (10.5 g, 81.2 mmol) were added to PPA (53.4 g, 81.2 mmol) and the mixture was stirred at 180 for 2 h. The reaction mixture was cooled to 90° C., then H2O was added to quench the reaction. The pH of the mixture was adjusted to 12 with 50% KOH. The mixture was extracted with CH2Cl2. The organic layer was dried over Na2SO4, filtered and concentrated to afford 3-2.
Step 2: Compound 3-3 was prepared from compound 3-2 following the procedure for the synthesis of compound 1-10 in example 1.
Step 3: Compound 3 was prepared from compound 3-3 following the procedure for the synthesis of compound 1 in example 1. LCMS (ESI, m/z): [M+H]+=425.2; 1H NMR (400 MHz, DMSO-d6, ppm) δ 8.27-8.24 (m, 1H), 8.16-8.13 (m, 1H), 7.54-7.56 (m, 1H), 7.51 (s, 1H), 7.17-7.15 (m, 1H), 4.28-4.24 (m, 2H), 3.67 (t, J=11.2 Hz, 2H), 3.53 (s, 3H), 3.50-3.46 (m, 1H), 2.40 (s, 3H), 2.30-2.28 (m, 2H), 2.16-2.08 (m, 2H).
Step 1: To a solution of 4-1 (2.5 g, 10.37 mmol) in dioxane (20 mL) was added Cs2CO3 (6.8 g, 20.75 mmol), Pd2(dba)3 (0.9 g, 1.04 mmol), BINAP (1.3 g, 2.08 mmol) and tert-butyl 2,6-diazaspiro[3.4]octane-6-carboxylate (2.64 g, 12.45 mmol). The resulting mixture was degassed for 10 minutes, then stirred for 4 h at 120° C. The mixture was filtered and the filtrate was concentrated. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 3/1) to afford 4-2.
Step 2: To a solution of 4-2 (2.9 g, 7.8 mmol) in dichloromethane (20 mL) was added TFA (5 mL, 805.6 mmol). Then the reaction mixture was stirred at 25° C. for 3 h. The pH was adjusted to 6 by aqueous solution of NaHCO3 and the mixture was extracted with ethyl acetate. The organic layer was dried over Na2SO4, filtered and concentrated to afford 4-3.
Step 3: Compound 4-4 was prepared from compound 4-3 following the procedure for the synthesis of compound 1-10 in example 1.
Step 4: Compound 4 was prepared as a 0.33 eq of TFA salt from compound 4-4 following the procedure for the synthesis of compound 1 in example 1. LCMS (ESI, m/z): [M+H]+=481.2; 1H NMR (400 MHz, DMSO-d6, ppm) δ 8.24-8.22 (m, 1H), 8.08-8.05 (m, 1H), 7.16-7.13 (m, 2H), 6.49-6.47 (m, 2H), 4.40 (s, 2H), 4.24 (t, J=6.7 Hz, 2H), 3.87-3.81 (m, 4H), 3.49 (s, 3H), 2.24 (t, J=6.9 Hz, 2H). 19F NMR (376 MHz, DMSO-d6, ppm) 6-57.34 (3F), −73.62 (1F, TFA).
Step 1: To a solution of 5-1 (46 g, 250.6 mmol) in con. HCl (170 mL) and EtOH (510 mL) was added Fe powder (42.0 g, 751.8 mmol) at 40 in portions. Then the mixture was stirred at 80
for 1 h. The mixture was filtered and the filtrate was concentrated. The pH of the mixture was adjusted to 8 with ammonia. Then the mixture was filtered and the filtrate was extracted with ethyl acetate. The combined organic layers were dried over Na2SO4, filtered and the filtrate was concentrated to afford 5-2.
Step 2: To a solution of 5-2 (36 g, crude) in THF (400 mL) was added pyridine (56.6 mL, 703.3 mmol), DMAP (2.9 g, 23.4 mmol) and TFAA (73.9 g, 351.6 mmol) at 0 slowly. Then the mixture was stirred at room temperature for 0.5 h. The mixture was concentrated. H2O was added to the residue. The mixture was extracted with ethyl acetate. The combined organic layers were dried over Na2SO4, filtered and the filtrate was concentrated. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 2/1) to afford 5-3.
Step 3: To a solution of 5-3 (47 g, 188.3 mmol) in DMF (450 mL) was added K2CO3 (78.1 g, 565 mmol) and CH3I (35.2 mL, 565 mmol) at room temperature. The mixture was stirred at room temperature for 12 h, then quenched with H2O. The mixture was extracted with ethyl acetate. The combined organic layers were dried over Na2SO4, filtered and the filtrate was concentrated. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 2/1) to afford 5-4.
Step 4: To a mixture of 5-4 (49.0 g, 175.4 mmol) and K2CO3 (72.7 g, 526.3 mmol) in DMSO (500 mL) was added H2O2 (30%, 59.7 g, 526.3 mmol) at 0 slowly. The mixture was stirred at room temperature for 2 h. H2O was added to the mixture, the mixture was extracted with ethyl acetate. The combined organic layers were dried over Na2SO4, filtered and the filtrate was concentrated. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 2/1) to afford 5-5.
Step 5: To a solution of 5-5 (27 g, 87.3 mmol) in DMF (300 mL) was added NaH (60%, 7.0 g, 174.6 mmol) at 0 in portions. After the mixture was stirred at 0
for 1 h, CDI (21.2 g, 130.9 mmol) was added to the reaction mixture. Then the mixture was stirred at 70
for 2 h. The solid was collected by filtration and washed with ethyl acetate and methanol to afford 5-6.
Step 6: To a solution of 5-6 (1.5 g, 7.09 mmol) in toluene (15 mL) were added POCl3 (5.27 mL, 56.71 mmol) and DIPEA (2.93 mL, 17.72 mmol) at room temperature. Then the mixture was stirred at 110 for 12 h. The mixture was concentrated to afford 5-7 which was used for the next step directly without further purification.
Step 7: To a solution of 5-7 (crude, 1.2 g) and DIPEA (2.5 mL, 15.1 mmol) in acetonitrile (10 mL) was added 2-3 (500 mg, 1.68 mmol) at room temperature. Then the mixture was stirred at 80 for 1 h. The mixture was concentrated. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 1/4) to afford 5-8.
Step 8: To a solution of 5-8 (98 mg, 0.22 mmol) in NMP (6 mL) were added dppf (11.9 mg, 0.022 mmol), Zn (5.6 mg, 0.088 mmol), Zn(CN)2 (50.6 mg, 0.43 mmol) and Pd2(dba)3 (19.7 mg, 0.022 mmol) at room temperature. Then the mixture was stirred at 90 for 3 h. The mixture was quenched with H2O and extracted with ethyl acetate. The combined organic layers were dried over Na2SO4, filtered and the filtrate was concentrated under vacuum. The residue was purified by reverse phase HPLC (acetonitrile/0.05% TFA in water: 5%-90%) to afford 5 as a 0.4 eq of TFA salt. LCMS (ESI, m/z): [M+H]+=446.4; 1H NMR (400 MHz, DMSO-d6, ppm): δ 8.26 (d, J=9.2 Hz, 1H), 8.02 (d, J=9.2 Hz, 1H), 7.35-7.26 (m, 2H), 7.18-7.06 (m, 2H), 4.82-4.71 (m, 1H), 3.96-3.79 (m, 4H), 3.46 (s, 3H), 2.20-2.05 (m, 2H), 1.86-1.70 (m, 2H). 19F NMR (376 MHz, DMSO-d6, ppm): δ −57.23 (3F), −74.35 (1.2F, TFA).
Step 1: To a solution of 12-1 (7.4 g, 34 mmol) in DMSO (100 mL) was added 4-bromo-1H-pyrazole (5.0 g, 34 mmol), K3PO4 (21.7 g, 102 mmol), CuO (0.2 g, 1.7 mmol) and N1,N2-bis(furan-2-ylmethyl)oxalamide (0.4 g, 1.7 mmol). The reaction mixture was degassed with nitrogen 3 times, then the reaction mixture was stirred for 14 h at 120 . The reaction mixture was diluted with water and extracted with ethyl acetate. The combined organic layers were washed with water and brine, dried and concentrated. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 5/1) to afford 12-2.
Step 2: To a solution of tert-butyl 4-(tetramethyl-1,3,2-dioxaborolan-2-yl)-1,2,3,6-tetrahydropyridine-1-carboxylate (4.7 g, 15.2 mmol) in dioxane (60 mL) was added 12-2 (3 g, 12.7 mmol) and K2CO3 (5.2 g, 38 mmol). The reaction mixture was degassed with nitrogen 3 times, then Pd(dppf)Cl2 (0.9 g, 1.27 mmol) and H2O (20 mL) were added to the reaction mixture. The reaction mixture was stirred for 14 h at 80 . The reaction mixture was diluted with water and extracted with ethyl acetate. The combined organic layers were washed with water and brine, dried over Na2SO4, and concentrated. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 5/1) to afford 12-3.
Step 3: To a solution of 12-3 (973 mg, 2.9 mmol) in dichloromethane (10 mL) was added Pd/C (10%, 97.6 mg). The reaction mixture was degassed with H2 3 times. The reaction mixture was stirred at room temperature for 14 h under H2 atmosphere. The reaction mixture was filtered and the filtrate was concentrated. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 5/11) to afford 12-4.
Step 4: To a solution of 12-4 (826 mg, 2.4 mmol) in dichloromethane (5 mL) was added HCl in ethyl acetate (4M, 5 mL, 20 mmol) and the reaction mixture was stirred at room temperature for 16 h. The reaction mixture was concentrated under reduced pressure to afford 12-5 which was used for the next step directly without further purification.
Compound 12 was prepared from compound 12-5 following the procedure for the synthesis of compound 5 in example 5. LCMS (ESI, m/z): [M+H]+=426.2; 1H NMR (400 MHz, methanol-d4, ppm) δ 8.11 (d, J=8.9 Hz, 1H), 8.03 (d, J=8.9 Hz, 1H), 7.74 (s, 1H), 7.68 (s, 1H), 7.39-7.36 (m, 2H), 7.33-7.30 (m, 2H), 3.61 (s, 3H), 3.36-3.34 (m, 2H), 3.32-3.29 (m, 2H), 3.17-3.11 (m, 1H), 2.28-2.21 (m, 2H), 2.20 (s, 3H), 1.94-1.81 (m, 2H).
Step 1: To a solution of 16-1 (1.9 g, 8.36 mmol) in methanol (12 mL) was added NaBH4 (0.2 g, 10.87 mmol) at 0 . The mixture was stirred at 20
for 16 h. The mixture was quenched with water and concentrated. The reside was diluted with H2O and extracted with ethyl acetate. The combined organic layers were dried over Na2SO4, filtered and concentrated. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 2/1) to afford 16-2 and 16-3.
Step 2: To a solution of 16-2 (604 mg, 2.63 mmol), 16-4 (563 mg, 3.16 mmol) and PPh3 (1.04 g, 3.95 mmol) in toluene (6 mL) was added DIAD (0.78 mL, 3.95 mmol) at 0 under N2. The mixture was stirred at 110
under N2 for 5 h. The mixture was concentrated. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 5/1) to afford 16-5.
Step 3: To a solution of 16-5 (923 mg, 2.37 mmol) in dichloromethane (5 mL) was added HCl in ethyl acetate (4M, 2.37 mL, 9.5 mmol) at 25. The mixture was stirred at 25
for 3 h. The mixture was concentrated to afford 16-6 which was used for the next step directly without further purification.
Step 4: Compound 16-7 was prepared from compound 16-6 following the procedure for the synthesis of compound 5 in example 5.
Step 5: 16-7 (70 mg) was purified by SFC (column: DAICELCHIRALCEL®OZ, methanol (+0.10% 7.0 mol/1 Ammonia in methanol)/Supercritical CO2=40/60) to afford 16 (31.8 mg) and 17 (7.02 mg). respectively. 16: SFC analysis: 99.52% ee; retention time: 1.87 min; column: DAICELCHIRALPAK®OZ, methanol (0.1% DEA) in CO2, 40%; pressure: 1800 psi; flow rate: 1.5 mL/min. LCMS (ESI, m/z): [M+H]+=474.2; 1H NMR (400 MHz, DMSO-d6, ppm): δ 8.28-8.23 (m, 1H), 8.04-7.99 (m, 1H), 7.33-7.29 (m, 2H), 7.11-7.07 (m, 2H), 4.85-4.77 (m, 1H), 3.47-3.43 (m, 4H), 3.37-3.29 (m, 2H), 2.17-2.02 (m, 3H), 2.01-1.89 (m, 3H), 0.87-0.79 (m, 3H). 19F NMR (376 MHz, DMSO-d6, ppm): δ −57.23 (3F). 17: SFC analysis: 96.6% ee; retention time: 2.14 min; column: DAICELCHIRALPAK®OZ, methanol (0.1% DEA) in CO2, 40%; pressure: 1800 psi; flow rate: 1.5 mL/min. LCMS (ESI, m/z): [M+H]+=474.2; 1H NMR (400 MHz, DMSO-d6, ppm): δ 8.23-8.17 (m, 1H), 7.99-7.92 (m, 1H), 7.30 7.22 (m, 2H), 7.10-6.98 (m, 2H), 4.81-4.71 (m, 1H), 3.40 (s, 3H), 3.34-3.32 (m, 3H), 2.13-1.83 (m, 6H1), 0.83-0.73 (m, 3H1). 19F NMR (376 MHz, DMSO-d6, ppm): δ −57.23 (3F).
Step 1: Compound 21-1 was prepared from compound 2-3 following the procedure for the synthesis of compound 5-8 in example 5.
Step 2: To a mixture of 21-1 (730 mg, 1.61 mmol) and (tert-butoxy)carbohydrazide (640 mg, 4.8 mmol) in dioxane (10 mL) were added Pd2(dba)3 (147.0 mg, 0.16 mmol), Cs2CO3 (1.57 g, 4.8 mmol) and t-BuBrettPhos (77.8 mg, 0.16 mmol) at room temperature. Then the mixture was stirred at 80 for 1 h. This mixture was filtered and the filtrate was concentrated. The residue was purified by column chromatography on silica gel (dichloromethane/methanol=1/0 to 10/1) to afford 21-2.
Step 3: To a solution of 21-2 (310 mg, 0.56 mmol) in methanol (5 mL) was added HCl in ethyl acetate (4M, 3 mL) at room temperature. Then the mixture was stirred at room temperature for 0.5 h. The mixture was concentrated to afford 21-3.
Step 4: To a solution of 21-3 (250 mg, crude) in methanol (6 mL) was added CH(OCH3)3 (3 mL, 0.56 mmol) at room temperature. Then the mixture was heated at 100 for 12 h. The mixture was concentrated. The residue was purified by column chromatography on silica gel (dichloromethane/methanol=1/0 to 10/1) to afford 21. LCMS (ESI, m/z): [M+H]+=461.4; 1H NMR (400 MHz, DMSO-d6, ppm) δ 9.49 (d, J=3.3 Hz, 1H), 8.26 (d, J=10.1 Hz, 1H), 7.77 (d, J=10.1 Hz, 1H), 7.33-7.26 (m, 2H), 7.15-7.08 (m, 2H), 4.73-4.65 (m, 1H), 3.68-3.50 (m, 5H), 3.44-3.36 (m, 1H), 3.29-3.22 (m, 1H), 2.17-1.87 (m, 3H), 1.78-1.65 (m, 1H). 19F NMR (376 MHz, DMSO-d6, ppm): δ −57.23 (3F).
Step 1: To a solution of LDA (2M in THF, 1.57 mL, 3.15 mmol) in THF (20 mL) was added dropwise 23-1 (550 mg, 2.42 mmol) at −68° C. After 30 minutes, a solution of 1,1,1-trifluoro-N-phenyl-N-trifluoromethanesulfonylmethanesulfonamide (864.44 mg, 2.42 mmol) in THF (5 mL) was added. The reaction mixture was stirred at room temperature for 16 h. The mixture was quenched with saturated NH4Cl and extracted with ethyl acetate. The combined organic layers were dried over Na2SO4, filtered and the filtrate was concentrated. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 1/1) to afford 23-2.
Step 2: To a solution of 12-2 (1.0 g, 4.22 mmol), 4,4,5,5-tetramethyl-2-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1,3,2-dioxaborolane (1.39 g, 5.48 mmol) in dioxane (20 mL) was added KOAc (0.8 g, 8.44 mmol) and Pd(dppf)Cl2 (0.3 g, 0.42 mmol). The mixture was stirred at 100° C. for 16 h under N2 atmosphere. The mixture was quenched with H2O and extracted with ethyl acetate. The combined organic layers were dried over Na2SO4, filtered and the filtrate was concentrated. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 1/1) to afford 23-3.
Step 3: To a solution of 23-2 (500 mg, 1.39 mmol), 23-3 (395.4 mg, 1.39 mmol) in dioxane (20 mL) and H2O (5 mL) was added Pd(dppf)Cl2 (113.6 mg, 0.14 mmol) and Na2CO3 (295 mg, 2.78 mmol). The mixture was stirred at 90° C. for 16 h under N2 atmosphere. The mixture was concentrated. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 1/1) to afford 23-4.
Step 4: To a solution of 23-4 (310 mg, 0.84 mmol) in methanol (10 mL) was added 10% Pd/C (179.5 mg). The mixture was degassed with H2 several times. Then the mixture was stirred at 25° C. under H2 atmosphere (1 atm) for 16 h. The mixture was filtered and the filtrate was concentrated to afford 23-5 which was used for the next step directly without further purification.
Step 5: To a solution of 23-5 (240 mg, crude) in dichloromethane (4 mL) was added TFA (1 mL). The mixture was stirred at 25° C. for 1 h. The mixture was concentrated to afford 23-6 which was used for the next step directly without further purification.
Step 6: Compound 23-7 was prepared from compound 23-6 following the procedure for the synthesis of compound 5 in example 5.
Step 7: 23-7 (120 mg, 0.26 mmol) was purified by prep-SFC (column: DAICELCHIRALCEL®AS, methanol (+0.1% 7.0 mol/1 Ammonia in methanol)/Supercritical CO2=40/60) to afford 23 (25.05 mg), 24 (3.11 mg) and 25 (9.99 mg). 23: SFC analysis: retention time: 1.795 min; column: DAICELCHIRALPAK®AS, methanol (0.1% of DEA) in CO2, 40%; pressure: 1800 psi; flow rate: 1.5 mL/min; LCMS (ESI, m/z): [M+H]+=454.4, 1H NMR (400 MHz, methanol-d4, ppm): δ 8.13-8.06 (m, 1H), 8.06-7.98 (m, 1H), 7.76-7.69 (m, 1H), 7.69-7.63 (m, 1H), 7.39-7.33 (m, 2H), 7.33-7.25 (m, 2H), 6.12-5.00 (m, 2H), 3.72-3.35 (m, 4H), 3.00-2.48 (m, 1H), 2.35-1.69 (m, 9H), 1.16-0.94 (m, 3H). 24: SFC analysis: 99.76% de; retention time: 2.300 min; column: DAICELCHIRALPAK®AS, methanol (0.1% of DEA) in CO2, 40%; pressure: 1800 psi; flow rate: 1.5 mL/min; LCMS (ESI, m/z): [M+H]+=454.4, 1H NMR (400 MHz, methanol-d4, ppm): δ 8.16-8.05 (m, 1H), 8.05-7.96 (m, 1H), 7.70 (s, 1H), 7.65 (s, 1H), 7.39-7.33 (m, 2H), 7.33-7.25 (m, 2H), 6.05-5.03 (m, 2H), 3.58 (s, 3H), 3.36 (s, 1H), 2.28-2.10 (m, 6H), 2.11-1.99 (m, 1H), 1.95-1.86 (m, 1H), 1.85-1.72 (m, 1H), 1.36-1.27 (m, 1H), 1.11-0.97 (m, 3H). 25: SFC analysis: 100% de; retention time: 2.422 min; column: DAICELCHIRALPAK®AS, methanol (0.1% of DEA) in CO2, 40%; pressure: 1800 psi; flow rate: 1.5 mL/min; LCMS (ESI, m/z): [M+H]+=454.4, 1H NMR (400 MHz, methanol-d4, ppm): δ 8.19-8.06 (m, 1H), 8.05-7.96 (m, 1H), 7.75-7.70 (m, 1H), 7.67 (s, 1H), 7.40-7.34 (m, 2H), 7.34-7.27 (m, 2H), 5.17 (s, 2H), 3.61-3.55 (m, 3H), 3.55-3.44 (m, 1H), 3.00-2.81 (m, 1H), 2.65-2.51 (m, 1H), 2.36-2.22 (m, 1H), 2.22-2.16 (m, 3H), 2.14-1.98 (m, 1H), 2.01-1.85 (m, 2H), 1.78 (s, 1H), 1.13-0.97 (m, 3H).
Step 1: To a mixture of 26-1 (10 g, 46.29 mmol) and potassium trifluoro(vinyl)borate (7.44 g, 55.55 mmol) in THF (160 mL) and H2O (40 mL) was added 1,1′-bis (di-t-butylphosphino)ferrocene palladium dichloride (3.0 g, 4.63 mmol) and K3PO4 (24.6 g, 115.73 mmol) under N2. Then the mixture was stirred at room temperature for 16 h.
The mixture quenched by addition of water. The aqueous layer was extracted with ethyl acetate. The organic layers were combined, washed with brine, dried over anhydrous Na2SO4 and concentrated. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=5/1) to afford 26-2.
Step 2: To a mixture of methyl 26-2 (7.8 g, 47.8 mmol) in AcOH (100 mL) was added PtO2 (2.2 g, 9.56 mmol). The mixture was degassed with H2 several times, then the mixture was stirred at 80 for 16 h under H2 (1 atm) atmosphere. The mixture was filtered and the filtrate was concentrated to afford 26-3 which was used for the next step directly without further purification.
Step 3: To a mixture of 26-3 (8 g, 46.72 mmol) in THF (80 mL) and H2O (80 mL) was added NaHCO3 (39.2 g, 467.18 mmol) and (Boc)2O (21.47 mL, 93.44 mmol). Then the mixture was stirred at room temperature for 3 h. The mixture quenched by addition of water, and the aqueous layer was extracted with ethyl acetate. The organic layers were combined, washed with brine, dried over anhydrous Na2SO4 and concentrated. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=5/1) to afford 26-4.
Step 4: To a mixture of 26-4 (9.6 g, 35.38 mmol) in THF (60 mL) and H2O (60 mL) was added LiOH (7.4 g, 176.89 mmol). Then the mixture was stirred at room temperature for 16 h. The pH of mixture was adjusted to 4 with 1N HCl and the aqueous layer was extracted with ethyl acetate. The organic extracts were combined, washed with brine, dried over anhydrous Na2SO4 and concentrated to afford 26-5 which was used for the next step directly without further purification.
Step 5: A mixture of 26-5 (4.55 g, crude) and PPA (100 mL) was stirred at 180 for 2 h. After being cooled to 90° C., the reaction was quenched by water. The filtrate was adjusted pH to about 12 with 50% potassium hydroxide aqueous solution and then extracted with methylene chloride. The organic layer was dried by Na2SO4, filtered and concentrated to afford 26-6 which was used for the next step directly without further purification.
Step 6: Compound 26-7 was prepared from compound 26-6 following the procedure for the synthesis of compound 5 in example 5.
Step 7: 26-7 (150 mg, 0.35 mmol) was separated by reverse phase HPLC (acetonitrile/H2O, 5-95%) to afford 26 (24.47 mg) and 27 (54.71 mg). 26: Analytical HPLC: retention time: 1.85 min; column: Waters ACQUITY BEH C18 2.1*50 mm, 1.7 um; Mobile phase A: H2O (0.05% TFA); Mobile phase B: Acetonitrile (0.05% TFA); flow rate: 1.0 mL/min; Run time: 5 min; 95 to 5% A in 3 min, 5% A for 2 min. LCMS (ESI, m/z): [M+H]+=429.4; 1H NMR (400 MHz, DMSO-d6, ppm): δ 8.27-8.23 (m, 1H), 8.01 (d, J=9.2 Hz, 1H), 7.54 (d, J=8.4 Hz, 1H), 7.47 (s, 1H), 7.18-7.15 (m, 1H), 5.77-5.44 (m, 1H), 5.34-4.80 (m, 1H), 3.68-3.61 (m, 2H), 3.45 (s, 3H), 2.40 (s, 3H), 2.29-2.16 (m, 2H), 2.05-1.78 (m, 4H), 1.00-0.89 (m, 3H). 27: Analytical HPLC: retention time: 1.85 min; column: Waters ACQUITY BEH C18 2.1*50 mm, 1.7 um; Mobile phase A: H2O (0.05% TFA); Mobile phase B: Acetonitrile (0.05% TFA); flow rate: 1.0 mL/min; Run time: 5 min; 95 to 5% A in 3 min, 5% A for 2 min. LCMS (ESI, m/z): [M+H]+=429.4; 1H NMR (400 MHz, DMSO-d(, ppm): δ 8.26 (d, J=8.8 Hz, 1H), 8.00 (d, J=9.2 Hz, 1H), 7.57 (d, J=8.4 Hz, 1H), 7.47 (s, 1H), 7.18-7.15 (m, 1H), 5.37-4.81 (m, 2H), 3.49-3.46 (m, 2H), 3.45 (s, 3H), 2.42 (s, 3H), 2.37-2.25 (m, 3H), 2.24-2.15 (m, 1H), 1.79-1.64 (m, 11H), 1.53-1.45 (m, 1H), 0.92-0.81 (m, 3H).
Step 1: A solution of 28-1 (4.5 g, 19.38 mmol) in THF (30 mL) at −30 was added C2H5MgBr (29.07 mL, 29.07 mmol, 1M in THF). The mixture was stirred at room temperature for 2 h. The reaction was quenched with saturated aqueous ammonium chloride. The organic phase was separated, dried over Na2SO4, filtered and concentrated. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=4/1) to afford 28-2.
Step 2: To a solution of 28-2 (1 g, 3.81 mmol) in dichloromethane (10 mL) at 0 was added DMF (0.029 mL, 0.38 mmol) and SOCl2 (0.42 mL, 5.72 mmol). The reaction was stirred at room temperature for 18 h. The reaction mixture was concentrated, and the residue was purified by column chromatography on silica gel (petroleum ether) to afford 28-3.
Step 3: To a stirred solution of 28-4 (100 g, 651 mmol) in dry CH2Cl2 (1.5 L) were added benzaldehyde (67.5 mL, 664 mmol), K2CO3 (90.0 g, 651 mmol) and Na2SO4 (92.5 g, 651 mmol). The reaction mixture was stirred at room temperature for 16 h. The reaction mixture was cooled with an ice water bath and NaBH(OAc)3 (207.0 g, 976 mmol) was added in portions over 30 minutes. Then the mixture was stirred at room temperature for 16 h. The solid was filtered and the filtration was concentrated. The residue was washed with 1 N HCl and extracted with ethyl acetate. The aqueous layer was adjusted to pH=8.0 with NaHCO3. Then the mixture was extracted with ethyl acetate. The combined organic layers were washed with brine, dried over anhydrous Na2SO4, filtered and concentrated to afford 28-5 which was used in the next step directly without further purification.
Step 4: To a solution of (2R)-2-{[(tert-butoxy)carbonyl]amino}butanoic acid (124 g, crude) in DMF (1 L) were added DIPEA (137 mL, 832 mmol) and HATU (253 g, 666 mmol). The reaction was stirred at 0 for 10 minutes, then 28-5 (115 g, 555 mmol) was added to the mixture. The resulting mixture was stirred for 16 h at room temperature. The reaction mixture was diluted with ethyl acetate and washed with water. The organic layer was dried over anhydrous Na2SO4, filtered and evaporated to afford 28-6 which was used for next step directly without further purification.
Step 5: To a solution of 28-6 (300 g, crude) in dichloromethane (1 L) was added TFA (500 mL, 764 mmol), the reaction was stirred at room temperature for 16 h. The solvent was removed to afford 28-7 which was used for next step directly without further purification.
Step 6: 28-7 (500 g, crude) was dissolved in methanol (1.5 L) and the reaction mixture was heated at 70 for 16 h. The solvent was removed, the residue was dissolved in dichloromethane and washed with saturated aqueous NaHCO3 solution. The combined organic layer was dried over anhydrous Na2SO4, filtered and concentrated. The residue was dissolved in propan-2-ol (500 mL) and heated at 70
for 1 h. The mixture was filtered, the solution was cooled to −10
, the solid was filtered and dried to afford 28-8.
Step 7: To a stirred solution of 28-8 (20 g, 76.82 mmol) in dry THF (500 mL) was slowly added BH3·THF (1M, 768 mL, 768 mmol) at 0 . The reaction mixture heated at 70
for 16 h. The reaction was quenched with methanol and 1.5 N HCl. The solvent was removed, the residue was dissolved in dichloromethane and washed with saturated aqueous NaHCO3 solution. The organic layer was dried over anhydrous Na2SO4, filtered and concentrated to afford 28-9 which was used in the next step directly.
Step 8: To a stirred solution of 28-9 (23 g, crude) in THF (100 mL) and H2O (100 mL) was added NaHCO3 (16.6 g, 198 mmol) and (Boc)2O (34.1 mL, 148.5 mmol). The reaction mixture was stirred at room temperature for 4 h. The mixture was diluted with ethyl acetate and washed with water and brine, dried over anhydrous Na2SO4 and concentrated. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=20/1 to 10/1) to afford 28-10.
Step 9: To a stirred solution of 28-10 (20 g, 60.15 mmol) in ethanol (200 mL) was added Pd/C (10%, 10 g). The reaction mixture was stirred under H2 atmosphere for 18 h at room temperature. The reaction mixture was filtered, the solvent was removed. The residue was purified by column chromatography on silica gel (dichloromethane/methanol=1/0 to 10/1) to afford 28-11.
Step 10: To a solution of 5-7 (1.63 g, crude) and DIPEA (7 mL, 42.5 mmol) in acetonitrile (40 mL) was added 28-11 (1.37 g, 5.66 mmol) at room temperature and the mixture was stirred at 80 for 1 h. Then the mixture was concentrated, the residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 1/4) to afford 28-12.
Step 11: To a solution of 28-12 (800 mg, 1.84 mmol) in NMP (8 mL) was added dppf (207.2 mg, 0.37 mmol), Zn (120.0 mg, 1.84 mmol), Zn(CN)2 (431.0 mg, 3.67 mmol) and Pd2(dba)3 (168.0 mg, 0.18 mmol) at room temperature. Then the mixture was heated at 90 for 12 h. The mixture was quenched with H2O and extracted with ethyl acetate. The combined organic layers were dried over Na2SO4, filtered and the filtrate was concentrated. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 1/4) to afford 28-13.
Step 12: To a solution of 28-13 (570 mg, 1.34 mmol) in dichloromethane (5 mL) was added TFA (2 mL, 1.34 mmol) at room temperature. Then the mixture was stirred at room temperature for 1 h. The mixture was concentrated to 28-14 which was used in the next step directly without further purification.
Step 13: The mixture of 28-14 (220 mg, 0.67 mmol), 28-3 (283.8 mg, 1.01 mmol), DIPEA (1.11 mL, 6.74 mmol) and NaI (151.5 mg, 1.01 mmol) in acetonitrile (10 mL) was stirred at 90° C. for 24 h. The solvent was removed, and the residue was purified by Reverse phase HPLC (acetonitrile with 0.05% of TFA in water: 10% to 70%) to afford 28-15.
Step 14: 28-15 (170 mg) was purified by SFC (column: DAICELCHIRALPAK®IG, methanol (+0.10% 7.0 mol/l Ammonia in methanol)/Supercritical CO2=40/60) to afford 28 (28 mg) and 29 (18 mg) respectively. 28: SFC analysis: 96.44% de; retention time: 4.29 min; column: DAICELCHIRALPAK®IB, methanol (0.1% of DEA) in CO2, 5% to 40%; pressure: 1800 psi; flow rate: 1.5 mL/min. LCMS (ESI, m/z): [M+H]+=571.5. 1H NMR (400 MHz, DMSO-d6, ppm) δ 8.23 (d, J=8.32 Hz, 1H), 7.98 (d, J=8.86 Hz, 1H), 7.89 (d, J=8.52 Hz, 2H), 7.56 (d, J=6.04 Hz, 2H), 5.85-5.47 (m, 1H), 4.97-4.85 (m, 1H), 3.71-3.62 (m, 1H), 3.66 (s, 3H), 3.09 (d, J=12.24 Hz, 1H), 2.93-2.76 (m, 2H), 2.44-2.24 (m, 1H), 2.22-2.06 (m, 1H), 2.05-1.80 (m, 3H), 1.62-1.54 (m, 1H), 1.53-1.39 (m, 1H), 0.97-0.91 (m, 3H), 0.70-0.52 (m, 6H); 19F NMR (376 MHz, DMSO-de, ppm): δ 87.99 (1F), 64.42 (4F) 29: SFC analysis: 97.86% de; retention time: 4.44 min; column: DAICELCHIRALPAK®IB, methanol (0.1% of DEA) in CO2, 5% to 40%; pressure: 1800 psi; flow rate: 1.5 mL/min. LCMS (ESI, m/z): [M+H]+=571.5. 1H NMR (400 MHz, DMSO-de, ppm): δ 8.27-8.19 (m, 1H), 8.01-7.95 (m, 1H), 7.87 (d, J=7.88 Hz, 2H), 7.58 (d, J=8.28 Hz, 2H), 6.05-5.28 (m, 1H), 5.09-4.75 (m, 1H), 3.64-3.50 (m, 2H), 3.43 (s, 3H), 3.18-3.07 (m, 1H), 2.21-2.12 (m, 1H), 2.04-1.95 (m, 1H), 1.83-1.39 (m, 6H), 1.00-0.91 (m, 3H), 0.65-0.54 (m, 6H). 19F NMR (376 MHz, DMSO-d6, ppm): δ 88.00 (1F), 64.37 (4F).
Step 1: To a solution of 30-1 (5 g, 26.3 mmol) and NaOH (1.6 g, 39.5 mmol) in EtOH (30 mL) and H2O (15 mL) was added hydroxylamine hydrochloride (2.2 g, 31.6 mmol) at 25. The mixture was stirred at 25
for 16 h. Ice was added to the reaction mixture, the solid was filtered, the filter cake was washed with H2O and dried to afford 30-2.
Step 2: A solution of 30-2 (1.6 g, 7.8 mmol) and NCS (1.1 g, 8.6 mmol) in DMF (15 mL) was stirred at 25 for 16 h. The mixture was concentrated, the residue was diluted with ethyl acetate, washed with H2O. The organic layer was dried over Na2SO4, filtered and concentrated to afford 30-3.
Step 3: To a solution of 30-3 (800 mg, 3.3 mmol) and tert-butyl 3-ethylidenepyrrolidine-1-carboxylate (795.5 mg, 4.3 mmol) in THF (20 mL) was added triethylamine (0.93 mL, 6.7 mmol) at 25 . The mixture was stirred at 25
for 4 h. The mixture was concentrated, the residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 4/1) to afford 30-4.
Step 4: To a solution of 30-4 (300 mg, 0.78 mmol) in dichloromethane (5 mL) was added HCl (4 M in ethyl acetate, 2.0 mL) at 25. The mixture was stirred at 25
for 2 h. The mixture was concentrated to afford 30-5.
Step 5: Compound 30 was prepared from compound 30-5 following the procedure for the synthesis of compound 5 in example 5. LCMS (ESI, m/z): [M+H]+=471.2; 1H NMR (400 MHz, DMSO-d6, ppm): δ 8.33-8.20 (m, 1H), 8.06-7.94 (m, 1H), 7.88-7.76 (m, 2H), 7.55-7.43 (m, 2H), 4.69-4.49 (m, 1H), 4.44-4.19 (m, 1H), 4.12-3.93 (m, 1H), 3.90-3.68 (m, 2H), 3.64-3.52 (m, 1H), 3.46 (s, 3H), 2.45-2.15 (m, 2H). 19F NMR (376 MHz, DMSO-d6, ppm): δ −56.74 (3F).
Step 1: To a solution of 32-1 (15 g, 99.25 mmol) in acetonitrile (100 mL) was added CH3NH2 (200 mL, 3.56 mmol, 40% in H2O). Then the reaction mixture was stirred at 80° C. for 16 h. The reaction mixture was quenched with H2O. The mixture was extracted with ethyl acetate. The combined organic layers were washed with H2O. The organic layer was dried over Na2SO4, filtered and concentrated to afford 32-2.
Step 2: To a solution of 32-2 (8.2 g, 50.56 mmol) in acetonitrile (150 mL) was added NCS (7.4 g, 55.61 mmol), then the reaction mixture was stirred at 60° C. for 70 minutes. The mixture was quenched by addition of water. The aqueous layer was extracted with ethyl acetate. The organic layer was combined, washed with brine, dried over anhydrous Na2SO4, filtered and concentrated. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 5/1) to afford 32-3.
Step 3: To a solution of 32-3 (5.0 g, 25.43 mmol) in DMSO (60 mL) were added K2CO3 (5.3 g, 38.14 mmol) and H2O2 (17.3 g, 30%, 152.6 mmol). Then the reaction mixture was stirred at 25° C. for 16 h. The reaction mixture was quenched with H2O. The mixture was extracted with ethyl acetate. The combined organic layers were washed with H2O. The organic layer was dried over Na2SO4, filtered and the filtrate was concentrated to afford 32-4.
Step 4: To a stirred 0° C. solution of 32-4 (5 g, 23.29 mmol) in DMF (70 mL) was in portions added NaH (1.9 g, 46.59 mmol). After stirred at 0° C. for 0.5 h, CDI (5.7 g, 34.94 mmol) was added to the mixture, then the mixture was stirred at 60° C. for 16 h. The reaction mixture was quenched with H2O. The mixture was extracted with ethyl acetate. The combined organic layer was washed with H2O. The organic layer was dried by Na2SO4, filtered and the filtrate was concentrated to afford 32-5.
Step 5: To a solution of 32-5 (1 g, 4.16 mmol) in toluene (10 mL) was added DIPEA (1.2 g, 9.14 mmol) and POCl3 (3.2 g, 20.78 mmol). The mixture was stirred at 90° C. for 3 h. The reaction mixture was concentrated to afford 32-6.
Step 6: A mixture of 32-6 (1.0 g, 3.86 mmol), 3-2 (1.00 g, 4.63 mmol) and DIPEA (7.5 g, 57.9 mmol) in propan-2-ol (15 mL) was stirred at 90° C. for 5 h. The mixture was cooled to room temperature. The precipitate was collected by filtration to afford 32-7.
Step 7: To a solution of 32-7 (1.0 g, 2.28 mmol) in dichloromethane (10 mL) under N2 at −78 was added dropwise BBr3 (5.7 g, 22.8 mmol). After the addition was completed, the mixture was stirred at room temperature for 16 h. Ice water was added to the reaction mixture and the precipitate was collected by filtration, washed with H2O and dried to afford 32-8.
Step 8: To a mixture of 32-8 (300 mg, 0.71 mmol) in DMF (10 mL) were added Cs2CO3 (690 mg, 2.12 mmol) and 3-bromooxolane (213.2 mg, 1.41 mmol). Then the mixture was stirred at 85° C. for 1 h. The solvent was removed under vacuum. The residue was purified by reverse phase HPLC (acetonitrile/H2O: 5%-42%) to afford 32-9.
Step 9: 32-9 (100 mg) was purified by SFC (column: DAICELCHIRALPAK®IC, ETOH (+0.10% 7.0 mol/1 Ammonia in methanol) to afford 32 (14.80 mg) and 33 (15.23 mg) respectively. 32: SFC analysis: 100% ee; retention time: 9.164 min; column: DAICELCHIRALPAK®IC, ETOH (+0.1% 7.0 mol/l Ammonia in methanol) in CO2, 40% to 60%; pressure: 100 bar; flow rate: 1.0 mL/min; LCMS (ESI, m/z): [M+H]+=495.4; 1H NMR (400 MHz, DMSO-d6, ppm): δ 7.79 (s, 1H), 7.56 (d, J=8.0 Hz, 1H), 7.50 (s, 1H), 7.20-7.16 (d, J=8.0 Hz, 1H), 6.91 (s, 1H), 5.44-5.42 (m, 1H), 4.22-4.15 (m, 2H), 4.05-3.95 (m, 1H), 3.95-3.78 (m, 3H), 3.51-3.49 (m, 3H), 3.49-3.38 (m, 2H), 2.41 (s, 3H), 2.39-2.30 (m, 2H), 2.22-2.18 (m, 2H), 2.09-1.94 (m, 3H). 33: SFC analysis: 98.89% cc; retention time: 10.858 min; column: DAICELCHIRALPAK®IC, ETOH (+0.1% 7.0 mol/l Ammonia in methanol) in CO2, 40% to 60%; pressure: 100 bar; flow rate: 1.0 mL/min; LCMS (ESI, m/z): [M+H]+=495.4; 1H NMR (400 MHz, DMSO-d6, ppm): δ 7.79 (s, 1H), 7.56 (d, J=8.0 Hz, 1H), 7.50 (s, 1H), 7.20-7.16 (d, J=8.0 Hz, 1H), 6.91 (s, 1H), 5.44-5.41 (m, 1H), 4.23-4.15 (m, 2H), 4.01-3.95 (m, 1H), 3.91-3.86 (m, 2H), 3.83-3.78 (m, 1H), 3.51-3.49 (m, 3H), 3.41-3.95 (m, 2H), 2.41 (s, 3H), 2.39-2.30 (m, 2H), 2.22-2.18 (m, 2H), 2.07-1.96 (m, 3H).
Step 1: Compound 34-2 was prepared from compound 34-1 following the procedure for the synthesis of compound 28-3 in example 11.
Step 2: A mixture of 34-2 (3.2 g, 14.3 mmol), 28-9 (4.0 g, 17.2 mmol), K2CO3 (5.9 g, 43.0 mmol), NaI (0.2 g, 1.4 mmol) and TBAI (0.5 g, 1.4 mmol) in acetonitrile (80 mL) was stirred at 90 for 66 h. The reaction mixture was filtered and concentrated. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=3/1) to afford 34-3.
Step 3: To a stirred solution of 34-3 (450 mg, 1.1 mmol) in isopropanol (10 mL) was added Pd/C (10%, 45 mg) at 25. The mixture was degassed several times with H2, then the mixture was stirred at 25
under H2 for 21 h. The reaction mixture was filtered and the filtrate was concentrated to afford 34-4.
Step 4: To a mixture of 5-2 (1.0 g, 6.54 mmol) and K2CO3 (2.7 g, 19.54 mmol) in DMSO (10 mL) was added H2O2 (2.2 g, 19.54 mmol) at 0 slowly. Then the mixture was stirred at room temperature for 2 h. The reaction mixture was diluted with water and extracted with ethyl acetate. The combined organic layers were washed with water and brine, dried and concentrated under reduced pressure. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=10/1 to 2/1) to afford 34-5.
Step 5: To a solution of 34-5 (5.0 g, 29.2 mmol) in NMP (100 mL) were added Zn (0.4 g, 5.84 mmol), Zn(CN)2 (6.9 g, 58.4 mmol), dppf (1.0 g, 1.75 mmol) and Pd2(dba)3 (2.7 g, 2.92 mmol) under N2. The reaction mixture was stirred at 120 for 2 h. The reaction mixture was diluted with water and extracted with ethyl acetate. The combined organic layers were washed with water and brine, dried and concentrated under reduced pressure. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=10/1 to 0/1) to afford 34-6.
Step 6: To a solution of 34-6 (0.5 g, 3.08 mmol) in dioxane (20 mL) was added triphosgene (457.5 mg, 1.54 mmol) under N2. The reaction mixture was stirred at 100 for 3 h. The resulting mixture was cooled to room temperature, filtered and dried to afford 34-7.
Step 7: To a solution of 34-7 (255 mg, 1.36 mmol) in toluene (10 mL) were added POCl3 (1.26 mL, 13.6 mmol) and DIPEA (0.67 mL, 4.06 mmol). The reaction mixture was stirred at 100 for 16 h. The reaction mixture was concentrated to afford 34-8 which was used for the next step directly without further purification.
Step 8: To a solution of 34-8 (300 mg, crude) in THF (10 mL) were added DIPEA (2.2 mL, 13.3 mmol) and 34-4 (437.8 mg, 1.33 mmol). The reaction mixture was stirred at 50 for 4 h. The reaction mixture was diluted with water and extracted with ethyl acetate. The combined organic layers were washed with water and brine, dried and concentrated under reduced pressure. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=10/1 to 0/1) to afford 34-9.
Step 9: To a solution of 34-9 (240 mg, 0.46 mmol) in dioxane (15 mL) were added (tert-butoxy)carbohydrazide (184 mg, 1.39 mmol), Cs2CO3 (453.8 mg, 1.39 mmol), tBubrettphos (22.5 mg, 0.046 mmol) and Pd2(dba)3 (42.5 mg, 0.046 mmol). The reaction mixture was stirred at 80 for 2 h. The reaction mixture was diluted with water and extracted with ethyl acetate. The combined organic layers were washed with water and brine, dried and concentrated under reduced pressure. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=10/1 to 0/1) to afford 34-10.
Step 10: To a solution of 34-10 (240 mg, 0.39 mmol) in dichloromethane (6 mL) was added TFA (3 mL). The reaction mixture was stirred at 25 for 1 h. The mixture was concentrated to afford 34-11 which was used in the next step directly without further purification.
Step 11: A mixture of 34-11 (180 mg, crude) and CH(OCH3)3 (5 mL) was stirred at 80 for 1 h. The mixture was concentrated. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=2/1 to 0/1) to afford 34-12.
Step 12: 34-12 (90 mg) was purified by SFC (column: DAICELCHIRALCEL®AD, EtOH (+0.10% 7.0 mol/l Ammonia in EtOH)/Supercritical CO2=40/60) to afford 34 (31.64 mg) and 35 (8.09 mg) respectively. 34: SFC analysis: 100% de; retention time: 4.35 min; column: DAICELCHIRALPAK®AD, EtOH (0.1% DEA) in CO2, 5% to 40%; pressure: 1800 psi; flow rate: 1.5 mL/min. LCMS (ESI, m/z): [M+H]+=523.5. 1H NMR (400 MHz, methanol-d4, ppm): δ 9.32 (s, 1H), 8.77-8.75 (m, 1H), 8.31-8.29 (m, 1H), 7.69-7.67 (m, 2H), 7.67-7.55 (m, 2H), 5.96-5.53 (m, 1H), 3.68-3.65 (m, 1H), 3.48-3.39 (m, 2H), 3.12-3.05 (m, 1H), 2.96-2.92 (m, 1H), 2.46 (s, 1H), 2.23 (s, 1H), 2.06-2.00 (m, 2H), 1.73-1.65 (m, 1H), 1.58-1.53 (m, 2H), 1.09-1.06 (m, 3H), 0.78-0.69 (m, 6H). 19F NMR (376 MHz, methanol-d4, ppm): δ −63.83 (3F). 35: SFC analysis: 97.18% de; retention time: 4.94 min; column: DAICELCHIRALPAK®AD, EtOH (0.1% DEA) in CO2, 5% to 40%; pressure: 1800 psi; flow rate: 1.5 mL/min. LCMS (ESI, m/z): [M+H]+=523.5. 1H NMR (400 MHz, methanol-d4, ppm): δ 9.32 (s, 1H), 8.77-8.75 (m, 1H), 8.30-8.28 (m, 1H), 7.65-7.63 (m, 2H), 7.60-7.58 (m, 2H), 5.32 (s, 1H), 3.66-3.47 (m, 3H), 2.70-2.66 (m, 1H), 2.37-2.33 (m, 1H), 2.20-2.16 (m, 1H), 2.07-1.90 (m, 3H), 1.70-1.56 (m, 3H), 1.13-1.03 (m, 3H1), 0.71-0.65 (m, 6H1). 19F NMR (376 MHz, methanol-d4, ppm): δ −63.82 (3F).
Step 1: A mixture of LiAlH4 (0.30 g, 8.2 mmol) in THF (20 mL) was stirred at 0 under N2 for 0.5 h. Then 36-1 (1 g, 4.1 mmol) was added and the mixture was stirred at 25
for 16 h. The mixture was diluted with THF, quenched with saturated Na2SO4 (1.5 mL) and filtered. The filtrate was concentrated to afford 36-2 which was used for the next step directly without further purification.
Step 2: A solution of 36-2 (700 mg, crude), 4-(trifluoromethoxy)benzoic acid (626.5 mg, 3.04 mmol), HATU (1733.5 mg, 4.56 mmol) and DIPEA(0.75 mL, 4.56 mmol) in DMF (12 mL) was stirred at 25 for 3 h. The mixture was diluted with H2O and extracted with ethyl acetate. The organic phase was washed with H2O and concentrated. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 2/1) to afford 36-3.
Step 3: To a solution of 36-3 (580 mg, 1.39 mmol) in dichloromethane (8 mL) was added DAST (0.28 mL, 2.08 mmol) at 0. Then the mixture was stirred at 20
for 1 h. The mixture was quenched with NaHCO3 aqueous, extracted with dichloromethane. The organic phase was concentrated, the residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 3/1) to afford 36-4.
Step 4: To a solution of 36-4 (588 mg, 1.47 mmol) in dichloromethane (6 mL) was added HCl in ethyl acetate (4M, 2.5 mL) at 20. The mixture was stirred at 20
for 1.5 h. The mixture was concentrated under reduced pressure to afford 36-5.
Step 5: A solution of 5-7 (250 mg, 0.87 mmol), 36-5 (208.9 mg, 0.67 mmol) and DIPEA (1.50 mL, 9.1 mmol) in acetonitrile (10 mL) was stirred at 85 for 3 h under N2. The mixture was concentrated. The residue was dissolved in ethyl acetate and washed with H2O. The organic phase was concentrated. The residue was purified by reverse phase HPLC (acetonitrile/H2O: 5%-80%) to afford 37. LCMS (ESI, m/z): [M+H]+=494.2. 1H NMR (400 MHz, DMSO-d6, ppm): δ 8.28-8.23 (m, 1H), 8.09-7.92 (m, 3H), 7.55-7.42 (m, 2H), 4.75-3.90 (m, 6H), 3.46 (s, 3H), 1.98-1.78 (m, 4H). 19F NMR (376 MHz, DMSO-d6, ppm): δ −56.68 (3F).
Step 6: A solution of 37 (100 mg, 0.20 mmol), dppf (22.5 mg, 0.040 mmol), Zn (13.2 mg, 0.20 mmol), Zn(CN)2 (47.6 mg, 0.41 mmol) and Pd2(dba)3 (18.5 mg, 0.02 mmol) in NMP (8 mL) was stirred at 90 for 16 h under N2. The mixture was diluted with H2O and extracted with ethyl acetate. The organic phase was washed with H2O and concentrated. The residue was purified by reverse phase HPLC (acetonitrile/H2O: 5%-80%) to afford 36. LCMS (ESI, m/z): [M+H]+=485.2. 1H NMR (400 MHz, DMSO-d6, ppm): δ 8.13-7.89 (m, 3H), 7.85-7.78 (m, 1H), 7.54-7.43 (m, 2H), 4.80-3.90 (m, 6H), 3.45 (s, 3H), 1.94-1.75 (m, 4H). 19F NMR (376 MHz, DMSO-d6, ppm): δ −56.68 (3F).
Step 1: To a stirred solution of 39-1 (1 g, 4.71 mmol) in NMP (10 mL) were added ZnBr2 (10.6 g, 47.15 mmol), Pd(dppf)Cl2 (0.3 g, 0.47 mmol), DIPEA (15.59 mL, 94.3 mmol) and ethynylcyclopropane (3.12 g, 47.15 mmol) under N2. The reaction mixture was stirred at 120° C. for 2 h. The reaction mixture was quenched with H2O and extracted with ethyl acetate. The combined organic layer was washed with brine. The organic layer was dried over Na2SO4, filtered and concentrated. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 20/1) to afford 39-2.
Step 2: To a solution of 5-7 (583 mg, 2.53 mmol) in acetonitrile (5 mL) were added 39-2 (500 mg, 2.53 mmol) and DIPEA (983 mg, 7.6 mmol). The reaction was stirred at room temperature for 3 h under N2. The reaction mixture was concentrated, the residue was purified by reverse phase HPLC (acetonitrile/H2O, 5%-50%) to afford 39. LCMS (ESI, m/z): [M+H]+=391.2; 1H NMR (400 MHz, DMSO-d6, ppm): δ 7.98 (d, J=8.0 Hz, 1H), 7.75 (d, J=8.0 Hz, 1H), 7.14-7.10 (m, 1H), 6.97-6.90 (m, 2H), 4.06-4.02 (m, 2H), 3.50 (s, 3H), 2.84-2.80 (m, 2H), 1.99-1.93 (m, 2H), 1.63-1.56 (m, 1H), 0.96-0.94 (m, 2H), 0.93-0.91 (m, 2H).
Step 3: To a solution of 39 (140 mg, 0.36 mmol) in DMF (1.5 mL) were added Zn(CN)2 (420 mg, 3.58 mmol) and Pd(PPh3)4 (206.8 mg, 0.18 mmol). Then the reaction mixture was stirred at 110° C. for 1 h under N2. The reaction mixture was concentrated, the residue was purified by reverse phase HPLC (acetonitrile/H2O: 5%˜50%) to afford 42. LCMS (ESI, m/z): [M+H]+=382.2; 1H NMR (400 MHz, DMSO-d6, ppm): δ 8.19 (d, J=8.0 Hz, 1H), 8.05 (d, J=8.0 Hz, 1H), 7.17-7.13 (m, 1H), 6.96-6.94 (m, 2H), 4.08-4.03 (m, 2H), 3.50 (s, 3H), 2.85-2.82 (m, 2H), 2.01-1.95 (m, 2H), 1.62-1.58 (m, 1H), 0.95-0.93 (m, 2H), 0.92-0.91 (m, 2H).
Step 1: To a solution of N1,N1,N2,N2-tetramethylethane-1,2-diamine (5.8 g, 56.28 mmol) in THF (80 mL) was added n-BuLi (40 mL, 64 mmol, 1.6 mol/L in THF) dropwise at −40 under N2. The reaction was stirred at −40
for 40 minutes. Then 44-1 (7 g, 40.2 mmol) was added to the mixture over 5 minutes. The mixture was stirred at −40
for 30 minutes. The second batch of n-BuLi (25.328 mL, 40.525 mmol, 1.6 mol/L in THF) was added dropwise over 5 minutes, then the reaction mixture was stirred at −40
for 2 h. Remove the cold bath, CuBr (7.5 g, 52.26 mmol) was added to the mixture, and the reaction mixture was stirred at 25
for another 1 h. The reaction was cooled to −30
and a solution of 3-bromoprop-1-ene (9.73 g, 80.4 mmol) in THF (10 mL) was added dropwise over 5 minutes, then the reaction mixture was stirred at −40
for 2 h. The reaction was quenched by addition of methanol. The pH of the mixture was adjusted to 6 with HCl (6M). The reaction mixture was filtered and the filter cake was washed with ethyl acetate. The mixture was extracted with ethyl acetate. The combined organic layer was washed with brine. The combine organic layer was concentrated. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 3/1) to afford 44-2.
Step 2: To a solution of 44-2 (5 g, 23.34 mmol) in THF (50 mL) was added allylmagnesium bromide (25.68 mL, 25.68 mmol), and the reaction was stirred at −40 for 1 h. The reaction was quenched with saturated aqueous NH4Cl. The reaction mixture was extracted with ethyl acetate. The combined organic layers were washed with brine and concentrated. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 2/1) to afford 44-3.
Step 3: To a solution of 44-3 (2.0 g, 7.8 mmol) in dichloromethane (160 mL) was added Grubbs 2nd catalyst (331 mg, 0.39 mmol). The reaction was stirred at 20 for 18 h under N2. The reaction mixture was diluted with water. The organic layer was separated, washed with brine and concentrated. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 2/1) to afford 44-4.
Step 4: To a solution of 44-4 (1.36 g, 5.96 mmol) in EtOH (25 mL) was added 10% Pd/C (0.1 g). The suspension was degassed under vacuum and purged with H2 3 times, then the mixture was stirred at 25 for 1 h under H2. The suspension was filtered, and the filter cake was washed with EtOH (20 mL). The combined filtrates were concentrated to afford 44-5.
Step 5: To a solution of 44-5 (1 g, 4.34 mmol) in dichloromethane (25 mL) was added SOCl2 (0.47 mL, 6.51 mmol), and the reaction was stirred at 0 for 1.5 h. The reaction was quenched with water. The aqueous layer was extracted with dichloromethane. The combined organic layers were washed with brine. The organic layer was dried over Na2SO4, filtered and concentrated. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 10/1) to afford 44-6.
Step 6: To a solution of 28-14 (250 mg, 0.77 mmol) in acetonitrile (10 mL) were added DIPEA (0.25 mL, 1.53 mmol), NaI (344 mg, 2.3 mmol) and 44-6 (380 mg, 1.53 mmol). The reaction mixture was stirred at 80 for 24 h. Then another batch of 44-6 (190 mg, 0.76 mmol) was added to the reaction mixture, and reaction was stirred at 80
for another 24 h. The reaction mixture was concentrated in vacuo. The residue was purified by column chromatography on silica gel (dichloromethane/methanol=1/0 to 10/1) to afford 44-7.
Step 7: 44-7 (150 mg, 0.28 mmol) was purified by reverse phase HPLC (acetonitrile/0.1% TFA in water: 75%) to afford 44 (10.43 mg) and 45 (4.01 mg). 44: Analytical HPLC: retention time: 1.85 min; column: Waters ACQUITY BEH C18 2.1*50 mm, 1.7 um; Mobile phase A: H2O (0.05% TFA); Mobile phase B: Acetonitrile (0.05% TFA); flow rate: 1.0 mL/min; Run time: 5 min; 95 to 5% A in 3 min, 5% A for 2 min. LCMS (ESI, m/z): [M+H]+=539.4; 1H NMR (400 MHz, CDCl3): δ 7.89-7.77 (m, 1H), 7.63-7.54 (m, 1H), 7.40-7.30 (m, 2H), 7.25-7.20 (m, 1H), 6.16-5.54 (m, 1H), 5.34-5.09 (m, 1H), 3.75-3.67 (m, 1H), 3.64-3.34 (m, 5H), 3.12-2.97 (m, 1H), 2.74-2.63 (m, 1H), 2.63-2.52 (m, 1H), 2.18-2.11 (m, 1H), 2.06-1.90 (m, 4H), 1.87-1.75 (m, 2H), 1.73-1.63 (m, 1H), 1.49-1.36 (m, 2H), 1.31-1.25 (m, 1H), 1.09-0.90 (m, 3H), 0.74-0.60 (m, 3H). 19F NMR (376 MHz, CDCl3, ppm): δ −62.33 (3F). 45: Analytical HPLC: retention time: 1.85 min; column: Waters ACQUITY BEH C18 2.1*50 mm, 1.7 um; Mobile phase A: H2O (0.05% TFA); Mobile phase B: Acetonitrile (0.05% TFA); flow rate: 1.0 mL/min; Run time: 5 min; 95 to 5% A in 3 min, 5% A for 2 min. LCMS (EST, m/z): [M+H]+=539.4; 1H NMR (400 MHz, CDCl3): δ 7.89-7.77 (m, 1H), 7.64-7.54 (m, 1H), 7.46-7.29 (m, 2H), 7.24-7.16 (m, 1H), 6.03-4.80 (m, 2H), 3.77-3.07 (m, 6H), 2.95-2.80 (m, 1H), 2.78-2.53 (m, 2H), 2.30-2.10 (m, 4H), 2.08-1.90 (m, 2H), 1.74-1.67 (m, 2H), 1.41-1.29 (m, 3H), 1.05-0.96 (m, 3H), 0.69-0.37 (m, 3H). 19F NMR (376 MHz, CDCl3, ppm): δ −62.34 (3F).
Step 1: To a solution of methyltriphenylphosphonium bromide (4.4 g, 12.19 mmol) in THF (25 mL) was added potassium tert-butoxide (12.19 mL, 12.19 mmol) at 20 The mixture was stirred at 55
under N2 for 2 h. Then 50-1 (1.3 g, 6.09 mmol) was added and the mixture was stirred at 55
for 16 h. The mixture was concentrated. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 10/1) to afford 50-2.
Step 2: Compound 50 was prepared from 50-2 following the procedure for the synthesis of compound 30 in example 12. LCMS (ESI, m/z): [M+H]+=499.2; 1H NMR (400 MHz, DMSO-d6, ppm): δ 8.30-8.23 (m, 1H), 8.06-7.98 (m, 1H), 7.88-7.72 (m, 2H), 7.53-7.42 (m, 2H), 5.60-5.35 (m, 1H), 5.25-4.80 (m, 1H), 3.70-3.60 (m, 1H), 3.46 (s, 3H), 3.42-3.35 (m, 1H), 3.34-3.30 (m, 1H), 2.24-2.10 (m, 1H), 2.07-1.79 (m, 3H), 1.56-1.36 (m, 3H). 19F NMR (376 MHz, DMSO-d6, ppm): δ −56.75 (3F).
Step 1: To a stirred solution of 57-1 (45 g, 276 mmol) in EtOH (450 mL) was added thiourea (22.06 g, 290 mmol) at room temperature. The resulting mixture was stirred at 80 for 15 h under N2. The solvent was removed under reduced pressure. The residue was diluted with H2O and neutralized with NaHCO3 until the pH is 8-9, then dichloromethane was added to the mixture. The mixture was extracted with dichloromethane. The combined organic layers were washed with saturated NaHCO3 and brine, dried over anhydrous Na2SO4 and concentrated to afford 57-2.
Step 2: To a stirred solution of 57-2 (6 g, 19.9 mmol) and CuBr (4.3 g, 29.9 mmol) in acetonitrile (80 mL) was added t-BuONO (3.59 mL, 29.9 mmol) at 0-5 . The resulting mixture was stirred at room temperature for 30 h. The reaction mixture was filtered and the filtrate was concentrated. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 6/1) to afford 57-3.
Step 3: To a stirred solution of 57-3 (1.0 g, 4.9 mmol) in THF (20 mL) was added n-BuLi (2.55 mL, 6.37 mmol) below −60 under N2. The resulting mixture was stirred at this temperature for 1 h. A solution of 4-((trifluoromethyl)thio)benzaldehyde (1313.4 mg, 6.37 mmol) in THF (20 mL) was added into the above reaction mixture below −60
. The resulting mixture was stirred at room temperature for 30 minutes. The reaction mixture was quenched with saturated NH4Cl and diluted with H2O. The mixture was extracted with ethyl acetate. The organic layer was combined, washed with brine, dried over anhydrous Na2SO4 and concentrated. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 5/1) to afford 57-4.
Step 4: To a stirred solution of 57-4 (450 mg, 1.36 mmol) in dichloromethane (6 mL) was added SOCl2 (0.15 mL, 2.04 mmol) at 25. The resulting mixture was stirred at 25
for 15 h. The reaction mixture was concentrated to afford 57-5 which was used for the next step directly without further purification.
Step 5: To a stirred solution of 57-5 (1.15 g, 2.63 mmol) and 28-14 (0.86 g, 2.63 mmol) in acetonitrile (10 mL) was added DIPEA (1.83 mL, 10.52 mmol) at 25 . The resulting mixture was stirred at 90
for 15 h. The reaction mixture was concentrated. The residue was purified by reverse phase HPLC (acetonitrile/0.05% TFA in H2O, 0-60%) to afford 57 (11.65 mg) and 58 (6.24 mg). 57: Analytical HPLC: retention time: 1.85 min; column: Waters ACQUITY BEH C18 2.1*50 mm, 1.7 um; Mobile phase A: H2O (0.05% TFA); Mobile phase B: Acetonitrile (0.05% TFA); flow rate: 1.0 mL/min; Run time: 5 min; 95 to 5% A in 3 min, 5% A for 2 min. LCMS (ESI, m/z): [M+H]+=640.2; 1H NMR (400 MHz, methanol-d4, ppm): δ 8.09-8.03 (m, 1H), 8.02-7.95 (m, 1H), 7.75-7.66 (m, 4H), 7.06 (s, 1H), 6.19-5.51 (m, 1H), 5.22-5.12 (m, 1H), 3.68-3.33 (m, 1H), 3.56 (s, 3H), 2.93-2.82 (m, 1H), 2.76-2.64 (m, 2H), 2.46-2.31 (m, 1H), 2.12-1.97 (m, 3H), 1.55-1.40 (m, 2H), 0.98-0.84 (m, 6H), 0.80-0.73 (m, 2H), 0.69-0.65 (m, 1H), 0.58-0.54 (m, 1H). 19F NMR (376 MHz, methanol-d4, ppm): δ −44.64 (3F). 58: Analytical HPLC: retention time: 1.85 min; column: Waters ACQUITY BEH C18 2.1*50 mm, 1.7 um; Mobile phase A: H2O (0.05% TFA); Mobile phase B: Acetonitrile (0.05% TFA); flow rate: 1.0 mL/min; Run time: 5 min; 95 to 5% A in 3 min, 5% A for 2 min. LCMS (ESI, m/z): [M+H]+=640.2; 1H NMR (400 MHz, CDCl3, ppm): δ 7.86-7.81 (m, 1H), 7.65-7.57 (m, 5H), 6.85 (s, 1H), 6.07-5.62 (m, 1H), 5.68-5.56 (m, 1H), 3.73-3.41 (m, 1H), 3.58 (s, 3H), 2.76-2.55 (m, 2H), 2.52-2.42 (m, 1H), 2.16-1.93 (m, 4H), 1.50-1.39 (m, 2H), 0.93-0.90 (m, 2H), 0.86-0.81 (m, 6H), 0.78-0.67 (m, 2H). 19F NMR (376 MHz, CDCl3, ppm): δ −42.58 (3F).
Step 1: Compound 59-2 was prepared from 59-1 following the procedure for the synthesis of compound 30-5 in example 12.
Step 2: To a mixture of 59-3 (4.9 g, 25.4 mmol) and methyl 2-hydroxyacetate (2.74 g, 30.4 mmol) in DMF (50 mL) was added NaH (1.22 g, 60%, 30.5 mmol) at 0. Then the mixture was stirred at room temperature for 1 h. H2O was added to the reaction mixture and the mixture was extracted with ethyl acetate. The combined organic layers were dried over Na2SO4, filtered and the filtrate was concentrated. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 2/1) to afford 59-4.
Step 3: To a solution of 59-4 (4.9 g, 19.87 mmol) in EtOH (50 mL) and HOAc (20 mL) was added Fe (5.6 g, 100 mmol) at room temperature. Then the mixture was stirred at 80 for 2 h. The mixture was filtered and the filtrate was concentrated. The residue was purified by column chromatography on silica gel (dichloromethane/methanol=1/0 to 20/1) to afford 59-5.
Step 4: To a solution of 59-5 (2.6 g, 14.09 mmol) in THF (20 mL) was added BH3-THF (1M, 65 mL, 65 mmol) at room temperature. Then the mixture was stirred at 80 for 2 h. The mixture was quenched with methanol and stirred at 80
for 30 minutes. Then the mixture was concentrated to afford 59-6 which was used for the next step directly without further purification.
Step 5: To a solution of 59-6 (1.0 g, crude) and (Boc)2O (4.04 mL, 17.59 mmol) in THF (15 mL) was added DMAP (1.43 g, 11.72 mmol) and triethylamine (0.82 mL, 5.86 mmol) at room temperature. Then the mixture was stirred at 80 for 12 h. The mixture was concentrated, the residue was purified by column chromatography on silica gel (dichloromethane/methanol=1/0 to 20/1) to afford 59-7.
Step 6: To a solution of 59-7 (1.5 g, 5.54 mmol) in CHCl3 (20 mL) was added mCPBA (2.25 g, 11.08 mmol) at room temperature. Then the mixture was stirred at 80 for 12 h. This mixture was concentrated and the residue was purified by column chromatography on silica gel (dichloromethane/methanol=1/0 to 10/1) to afford 59-8.
Step 7: To a solution of 59-8 (1.2 g, 4.19 mmol) in acetonitrile (20 mL) were added TMSCN (1.25 g, 12.56 mmol) and triethylamine (1.75 mL, 12.56 mmol) at room temperature. Then the mixture was stirred at 80 for 12 h. The mixture was concentrated and the residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 4/1) to afford 59-9.
Step 8: To a solution of 59-9 (620 mg, 2.1 mmol) and K2CO3 (870 mg, 6.29 mmol) in DMSO (10 mL) was added H2O2 (30%, 713.0 mg, 6.29 mmol) at 0 . Then the mixture was stirred at room temperature for 1 h. The solid was collected by filtration and washed with dichloromethane. The solid was suspended in H2O, then cone. HCl was added until the pH was 4-5. The solid was collected by filtration to afford 59-10.
Step 9: Compound 59 was prepared from 59-10 following the procedure for the synthesis of compound 5 in example 5. LCMS (ESI, m/z): [M+H]+=513.2; 1H NMR (400 MHz, DMSO-d6, ppm): δ 7.89 (s, 1H), 7.84-7.76 (m, 2H), 7.53-7.44 (m, 2H), 5.06-4.58 (m, 1H), 4.55-4.46 (m, 2H), 4.48-4.06 (m, 2H), 4.05-3.68 (m, 3H), 3.35 (s, 2H), 2.06-1.82 (m, 4H). 19F NMR (376 MHz, DMSO-d6, ppm): δ −56.75 (3F).
Step 1: To a suspension of 64-1 (950 mg, 3.62 mmol) in CCl4 (20 mL) was added NBS (709.4 mg, 3.99 mmol) and AIBN (89.2 mg, 0.54 mmol). Then the mixture was stirred at 85 for 3 h. The mixture was concentrated and the residue was purified by column chromatography on silica gel (dichloromethane/methanol=1/0 to 10/1) to afford 64-2.
Step 2: To a mixture of 64-2 (500 mg, 1.17 mmol) and 28-14 (421.05 mg, 1.29 mmol) in acetonitrile (15 mL) was added DIPEA (0.97 mL, 5.86 mmol) and NaI (351.6 mg, 2.35 mmol). Then the mixture was stirred at 85 for 3 h. The mixture was extracted with ethyl acetate. The organic layer was washed with H2O and brine (20 mL), dried over Na2SO4, filtered and concentrated. The residue was purified by reverse phase HPLC (acetonitrile/0.05% TFA in water: 0-60%) to afford 64-3 and 64-4.
Step 3: To a mixture of 64-3 (75 mg, 0.13 mmol) and N-[azanylidene (cyclopropyl)methyl]hydroxylamine (14.08 mg, 0.14 mmol) in DMSO (2 mL) was added DIPEA (58.3 mg, 0.15 mmol) and HATU (58.3 mg, 0.15 mmol). The mixture was stirred at room temperature for 30 minutes, then the mixture was heated to 90 and stirred for 30 minutes. The mixture was cooled and extracted with ethyl acetate. The organic layer was washed with H2O and brine, dried over anhydrous Na2SO4 and concentrated. The residue was purified by reverse phase HPLC (acetonitrile/0.05% TFA in water: 0-70%) to afford 64 as 3.3 eq of TFA salt. LCMS (ESI, m/z): [M+H]+=651.4; 1H NMR (400 MHz, DMSO-d6, ppm): δ 8.24 (d, J=8.8 Hz, 1H), 8.02-7.98 (m, 3H), 7.88-7.81 (m, 2H), 6.00-5.37 (m, 2H), 4.98-4.91 (m, 1H), 3.65-3.56 (m, 0.5 H), 3.44 (s, 3H), 3.30-3.18 (m, 1H), 2.90-2.82 (m, 0.5 H), 2.2-2.57 (m, 1H), 2.40-2.21 (m, 2H), 2.17-2.12 (m, 1H), 2.04-1.78 (m, 2H), 1.63-1.39 (m, 2H), 1.11-1.04 (m, 2H), 0.90-0.69 (m, 7H). 19F NMR (376 MHz, DMSO-d6, ppm): δ 87.41-86.60 (1F), 64.20-63.80 (4F), −73.59 (9.9F, TFA).
Step 1: To a stirred solution of 67-1 (25 g, 150.4 mmol) in acetonitrile (250 mL) was added NBS (28.1 g, 158 mmol) in portions at 25-40. The reaction mixture was stirred at 25-40
for 2 h. The precipitate was filtered and the cake was washed with acetonitrile. The cake was collected and dried to afford 67-2.
Step 2: To a stirred solution of 67-2 (20 g, 81.6 mmol) in THF (100 mL) was added dropwise acetyl acetate (24.14 mL, 257.06 mmol) at 25. The reaction mixture was stirred at 75
for 19 h. The reaction mixture was concentrated. The residue was diluted with H2O and extracted with ethyl acetate. The organic layer was combined, dried over anhydrous Na2SO4 and concentrated to afford 67-3.
Step 3: To a solution of 67-3 (20.9 g, 72.8 mmol) and Cs2CO3 (52.2 g, 160.15 mmol) in DMF (150 mL) was added dropwise CH3I (7.7 mL, 123.7 mmol) at 5-15 . The reaction mixture was stirred at 25
for 67 h. The reaction mixture was diluted with H2O and extracted with ethyl acetate. The organic layer was combined, washed with H2O and brine, dried over anhydrous Na2SO4 and concentrated. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 1/4) to afford 67-4.
Step 4: KHMDS in THF (1M, 54.6 mL, 54.6 mmol) was added to THF (200 mL) at −10 under N2, then 67-4 (13.7 g, 45.5 mmol) in THF (400 mL) was added to the solution at −60° C. to −50
under N2. Then the reaction mixture was stirred at 20
for 1.5 h. The reaction was quenched with H2O, and the mixture was extracted with ethyl acetate. The aqueous layer was acidified with HCl (2 N) to change the pH to 3-4. The precipitate was filtered, washed with H2O and dried to afford 67-5.
Step 5: To a stirred solution of 67-5 (2 g, 7.84 mmol) in AcOH (20 mL) was added HNO3 (1.3 mL, 31.74 mmol) at 25 . The reaction mixture was stirred at 80
for 15 h. The reaction mixture was cooled to 25
and diluted with H2O. The mixture was stirred for 10 minutes and the precipitation was filtered and dried to afford 67-6.
Step 6: To a stirred solution of 67-6 (1.4 g, 4.67 mmol) in toluene (28 mL) was added DIPEA (4.06 mL, 23.33 mmol) and POCl3 (2.17 mL, 23.33 mmol) at 25 . The reaction mixture was stirred at 110
for 3 h under N2. The reaction mixture was concentrated to afford 67-7 which was used for the next step directly without further purification.
Step 7: To a stirred solution of 67-8 (34.9 g, 292.99 mmol) in methanol (250 mL) was added triethylamine (42.76 mL, 307.63 mmol) and benzaldehyde (31.27 mL, 307.63 mmol) at 5-10 . The reaction mixture was stirred at this temperature for 3 h. Then the reaction mixture was cooled to 0° C. and NaBH4 (22.2 g, 585.964 mmol) was added into the reaction mixture in portions. The reaction mixture was stirred at 0-20
for 2.5 h. The reaction mixture was quenched with HCl (3 N) and extracted with ethyl acetate. The organic layer was washed with HCl (3 N). The aqueous layer was combined and neutralized with NaHCO3 powder. The aqueous layer was concentrated. The residue was dissolved in dichloromethane/methanol (10:1) and filtered. The organic layer was concentrated, the residue was purified by column chromatography on silica gel (dichloromethane/methanol=1/0 to 10/1) to afford 67-9.
Step 8: To a solution of 67-9 (32.86 g, 157.04 mmol) and (2R)-2-{[(tert-butoxy)carbonyl]amino}butanoic acid (31.92 g, 157.04 mmol) in DMF (350 mL) was added dropwise DIPEA (82.06 mL, 471.11 mmol) at 25 , then HATU (89.6 g, 235.56 mmol) was added to the solution at 5-15
. The reaction mixture was stirred at 25
for 48 h. The reaction mixture was diluted with H2O and extracted with ethyl acetate. The organic layer was combined, washed with H2O and brine, dried over anhydrous Na2SO4 and concentrated. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 0/1) to afford 67-10.
Step 9: To a stirred solution of 67-10 (28.9 g, 73.26 mmol) in methanol (280 mL) was added HCl in ethyl acetate (4M, 50 mL) at 25-35 . The reaction mixture was stirred at room temperature for 16 h. The reaction mixture was concentrated to afford 67-11.
Step 10: A solution of 67-11 (25.4 g, 69.14 mmol) in DIPEA (35 mL, 200.93 mmol) and methanol (300 mL) was stirred at 70 for 15 h. The reaction mixture was concentrated. The residue was purified by column chromatography on silica gel (dichloromethane/methanol=1/0 to 10/1) to afford 67-12.
Step 11: To a stirred solution of 67-12 (21 g, 80.06 mmol) in THF (100 mL) was added BH3-THF (1M in THF, 22.9 mL, 22.9 mmol) at 0 C under N2. The reaction mixture was stirred at 70
for 18 h. The reaction mixture was cooled to 0-10
, then quenched with methanol and HCl (2 N). The resulting mixture was stirred at 70
for 2 h. The reaction mixture was concentrated. The residue was diluted with H2O (100 mL) and basified with NaHCO3 to pH=8-9. The mixture was concentrated to dryness. The residue was dissolved in a mixture of 10% methanol in dichloromethane and the suspension was filtered. The filtrate was concentrated to afford 67-13 which was used for the next step directly without further purification.
Step 12: To a stirred solution of 67-13 (27.7 g, crude), DMAP (0.9 g, 7.09 mmol) and triethylamine (19.72 mL, 141.85 mmol) in dichloromethane (280 mL) was added (Boc)2O (14.66 mL, 63.83 mmol) at 0-10 . The reaction mixture was stirred at room temperature for 15 h. The reaction mixture was concentrated. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 2/1) to afford 67-14.
Step 13: To a stirred solution of 67-14 (5 g, 14.95 mmol) in methanol (30 mL) was added HCl in ethyl acetate (4M, 30 mL) at 25 , the reaction mixture was stirred at 25
for 15 h. The reaction mixture was concentrated to afford 67-15 which was used for the next step directly without further purification.
Step 14: A mixture of 67-15 (4.5 g, 14.61 mmol), 34-2 (8.13 g, 36.53 mmol), DIPEA (10.18 mL, 58.44 mmol), K2CO3 (8.1 g, 58.44 mmol) and KI (4.9 g, 29.22 mmol) in CH3CN (100 mL) was stirred at 90 for 48 h. The reaction mixture was concentrated. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 2/3) to afford 67-16 and 67-17.
Step 15: To a stirred solution of 67-16 (1.93 g, 4.59 mmol) in isopropanol (60 mL) was added Pd/C (5%, 450 mg) at 25 . The reaction mixture was degassed with H2 several times, then the mixture was stirred at 60
for 15 h under H2. The reaction mixture was filtered and the filtrate was concentrated to afford 67-18.
Step 16: A solution of 67-18 (1.6 g, 4.84 mmol), 67-7 (1.46 g, crude) and DIPEA (8.44 mL, 48.43 mmol) in CH3CN (30 mL) was stirred at 90 for 15 h. The reaction mixture was concentrated. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 1/4) to afford 67-19.
Step 17: To a stirred solution of 67-19 (700 mg, 1.14 mmol), Zn(CN)2 (268.5 mg, 2.29 mmol), Zn (15.0 mg, 0.23 mmol) and dppf (38.0 mg, 0.069 mmol) in NMP (7 mL) was added Pd2(dba)3 (104.7 mg, 0.114 mmol) at 25 . The reaction mixture was degassed through N2 for a while and stirred at 90
for 15 h. The reaction mixture was diluted with H2O and extracted with ethyl acetate. The organic layer was combined, washed with H2O and brine, dried over anhydrous Na2SO4 and concentrated. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 1/4) to afford 67-20.
Step 18: To a stirred solution of 67-20 (180 mg, 0.31 mmol) in DMF (1.8 mL) was added DBU (139.8 mg, 0.92 mmol) at 25 . The reaction mixture was stirred at 130
for 15 h. The reaction mixture was diluted with H2O and extracted with ethyl acetate. The organic layer was combined, washed with brine, dried over anhydrous Na2SO4 and concentrated. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 0/1) and then by reverse phase HPLC (MeCN/0.1% TFA in water: 0-60%) to afford 67. Analytical HPLC: retention time: 1.69 min; column: Waters ACQUITY BEH C18 2.1*50 mm, 1.7 um; Mobile phase A: H2O (0.05% TFA); Mobile phase B: Acetonitrile (0.05% TFA); flow rate: 1.0 mL/min; Run time: 5 min; 95 to 5% A in 3 min, 5% A for 2 min. LCMS (ESI, m/z): [M+H]+=512.2; 1H NMR (400 MHz, methanol-d4, ppm): δ 8.06-7.99 (m, 1H), 7.89-7.85 (m, 1H), 7.68-7.62 (m, 2H), 7.59-7.53 (m, 2H), 5.13-5.08 (m, 1H), 4.41-4.35 (m, 1H), 4.08-4.04 (m, 1H), 3.82-3.74 (m, 1H), 3.71 (s, 3H), 3.63-3.52 (m, 1H), 3.16-3.09 (m, 1H), 3.09-3.02 (m, 1H), 2.74-2.65 (m, 1H), 2.52-2.43 (m, 1H), 2.12-2.01 (m, 1H), 1.81-1.59 (m, 3H), 0.81 (t, J=7.4 Hz, 3H), 0.69 (t, J=7.3 Hz, 3H). 19F NMR (376 MHz, methanol-d4, ppm): δ −63.86 (3F).
Step 1: To a mixture of methyl 72-1 (500 mg, 2.31 mmol) in 2-methoxyethyl ether (10 mL) was added 2,2′-bipyridine (542 mg, 3.47 mmol), AgSCF3 (725.4 mg, 3.47 mmol) and CuI (661 mg, 3.47 mmol). Then the mixture was stirred at 130° C. for 24 h under N2. The mixture was quenched by addition of water, the aqueous layer was extracted with ethyl acetate. The organic extracts were combined, washed with brine, dried over anhydrous Na2SO4, the mixture was concentrated. The residue was purified by reverse phase HPLC (MeCN/H2O (0.05% TFA), 5-95%) to afford 72-2.
Step 2: To a solution of 72-2 (2.2 g, 9.28 mmol) in MeOH (2 mL) was added NaBH4 (0.7 g, 18.55 mmol) at 0° C. and the reaction mixture was stirred at room temperature for 2 h. The reaction mixture was diluted with DCM and saturated NH4Cl solution. The organic layer was separated and concentrated in vacuo. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=10/1) to afford 72-3.
Step 3: To a solution of 72-3 (1.12 g, 5.35 mmol) in DCM (15 mL) was added Dess-Martin periodinane (3.0 g, 6.96 mmol) at 0° C. and the reaction mixture was stirred at room temperature for 30 minutes. The reaction mixture was concentrated in vacuo. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=10/1) to afford 72-4.
Step 4: To a solution of 72-4 (460 mg, 2.21 mmol) in THF (10 mL) was added 4-fluorophenylmagnesium bromide (0.32 mL, 0.32 mmol) at −78° C., then the reaction mixture was stirred at room temperature for 30 minutes. The reaction mixture was diluted with ethyl acetate and saturated NH4Cl solution. The organic layer was separated and washed with saturated NaCl solution, then organic layer was concentrated in vacuo. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=4/1) to afford 72-5.
Step 5: To a solution of 72-5 (580 mg, 1.91 mmol) in DCM (12 mL) were added DMF (0.01 mL, 0.13 mmol) and SOCl2 (378 mg, 3.82 mmol) at 0° C., then the reaction mixture was stirred at room temperature for 30 minutes. The reaction mixture was concentrated in vacuo to afford 72-6 which was used for the next step directly without further purification.
Step 6: Compound 72 and 73 was prepared from compound 72-6 following the procedure for the synthesis of compound 28-15 in example 11. The product was obtained by reverse phase HPLC (MeCN/water (0.05% TFA), 0-80%). 72: 2.23 TFA salts, LCMS (EST, m/z): [M+H]+=598.4; 1H NMR (400 MHz, Methanol-d4, ppm): δ 8.98 (s, 1H), 8.22 (dd, J=8.0, 2.0 Hz, 1H), 8.14-8.01 (m, 2H), 7.76-7.60 (m, 3H), 7.25 (t, J=8.4 Hz, 2H), 6.43-5.91 (m, 2H), 5.42-5.32 (m, 1H), 4.10-3.99 (m, 1H), 3.60 (s, 3H), 3.20-3.02 (m, 2H), 1.91-1.59 (m, 5H), 1.39-1.28 (m, 1H), 0.85 (t, J=7.2 Hz, 3H). 19F NMR (376 MHz, Methanol-d4, ppm): δ −44.15 (3F), −77.27 (6.7F, TFA), −111.5 (1F). 73: 1.48 TFA salts, LCMS (ESI, m/z): [M+H]+=598.4; H NMR (400 MHz, Methanol-d4, ppm): δ 8.93 (s, 1H), 8.21 (dd, J=8.0, 2.0 Hz, 1H), 8.10 (d, J=8.8 Hz, 1H), 8.03 (d, J=8.8 Hz, 1H), 7.81-7.67 (m, 3H), 7.18 (t, J=8.4 Hz, 2H), 6.34-5.96 (m, 1H), 5.55-5.23 (m, 2H), 4.10-3.85 (m, 1H), 3.59 (s, 3H), 3.28-3.15 (m, 1H), 3.10-2.90 (m, 1H), 1.65-1.28 (m, 6H), 0.78-0.64 (m, 3H). 19F NMR (376 MHz, Methanol-d4, ppm): δ −4.24 (3F), −77.38 (4.45F, TFA), -111.5 (1F).
Step 1: To a mixture of 64-3 (80 mg, 0.14 mmol) in THF (10 mL) was added CDI (44.2 mg, 0.27 mmol). After stirred for 16 h at room temperature, NaBH4 (5.2 mg, 0.14 mmol) was added to the mixture. Then the mixture was stirred at room temperature for 20 minutes. The mixture was quenched by addition of water, the aqueous layer was extracted with ethyl acetate. The organic layer was combined and washed with brine, dried over anhydrous Na2SO4. The solvent was concentrated. The residue was purified by column chromatography on silica gel (DCM/MeOH=10/1) to afford 77. LCMS (ESI, m/z): [M+H]+=573.3; 1H NMR (400 MHz, DMSO-d6, ppm): δ 8.30-8.18 (m, 1H), 8.01-7.91 (m, 1H), 7.90-7.78 (m, 2H), 7.65-7.06 (m, 2H), 6.12-5.57 (m, 0.5H), 5.62-5.53 (m, 0.5H), 5.11-4.71 (m, 2H), 3.81-3.64 (m, 5H), 3.06-2.72 (m, 3H), 2.61-2.53 (m, 1H), 2.37-2.11 (m, 1H), 2.09-1.47 (m, 2H), 1.2-1.41 (m, 2H), 1.07-0.84 (m, 3H), 0.79-0.56 (m, 3H). 19F NMR (376 MHz, DMSO-d6, ppm): δ 88.41 (1F), 64.39 (4F).
Step 2: To a mixture of 77 (50 mg, 0.087 mmol) in THF (2 mL) was added NaH (7.0 mg, 0.17 mmol) and iodomethane (37.0 mg, 0.26 mmol). Then the mixture was stirred at 0° C. for 3 h. The reaction mixture was quenched by water, the aqueous layer was extracted with ethyl acetate. The organic layer was combined and washed with brine, dried over anhydrous Na2SO4. The solvent was concentrated. The residue was purified by reverse phase HPLC (MeCN/water from 1/0 to 2/3) to afford 78. LCMS (ESI, m/z): [M+H]+=587.3; 1H NMR (400 MHz, DMSO-d6, ppm): δ 8.31-8.17 (m, 1H), 8.03-7.94 (m, 1H), 7.96-7.83 (m, 2H), 7.70-7.58 (m, 2H), 6.09-5.78 (m, 0.5H), 5.53-5.26 (m, 0.5H), 5.09-4.75 (m, 1H), 3.98-3.80 (m, 1H), 3.69-3.48 (m, 3H), 3.43 (s, 3H), 3.20 (d, J=5.2 Hz, 3H), 3.14-2.81 (m, 2H), 2.37-2.16 (m, 1H), 2.13-1.70 (m, 2H), 1.56-1.31 (m, 2H), 1.01-0.83 (m, 3H), 0.77-0.55 (m 3H). 19F NMR (376 MHz, DMSO-d6, ppm): δ 88.66 (1F), 64.36 (4F).
Step 1: To a solution of 34-8 (2.39 g, 10.62 mmol) in MeCN (50 mL) was added DIPEA (14.04 mL, 84.97 mmol) and 28-11 (2.57 g, 10.62 mmol) at room temperature. Then the mixture was stirred at room temperature for 1 h. The mixture was concentrated under reduced pressure. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 10/1) to afford 84-1.
Step 2: To a solution of 84-1 (3.66 g, 8.49 mmol) in THF (30 mL) was added N2H4·H2O (5 mL), the mixture was stirred at 50° C. for 1 h. The mixture was concentrated under reduced pressure. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=10/0 to 0/1) to afford 84-2.
Step 3: 84-2 (3.6 g, 8.44 mmol) was dissolved in trimethoxymethane (30 mL), the mixture was stirred at 100° C. under N2 for 16 h. The mixture concentrated under reduced pressure to afford product 84-3.
Step 4: To a solution of 84-3 (3.65 g, 8.36 mmol) in DCM (30 mL) was added TFA (10 mL) at room temperature and stirred for 5 h. The mixture was concentrated under reduced pressure, the residue was dissolved in DCM and washed with saturated NaHCO3. The combined organic layer was dried over anhydrous Na2SO4, filtered and concentrated to afford 84-4.
Step 5: To a solution of 4-(trifluoromethyl)benzoic acid (100 mg, 0.53 mmol) in DMF (5 mL) was added DIPEA (0.13 mL, 0.79 mmol) and HATU (240.0 mg, 0.63 mmol), the mixture was stirred at 0° C. for 10 minutes, then 84-4 (176.9 mg, 0.53 mmol) was added to the mixture. The resulting mixture was stirred at room temperature for 16 h. The resulting mixture was purified by reverse phase HPLC (MeCN/water from 0/1 to 1/1) to afford 84. LCMS (ESI, m/z): [M+H]+=509.2. 1H NMR (400 MHz, DMSO-d6, ppm) δ 9.55 (s, 1H), 9.02-8.88 (m, 1H), 8.67-8.50 (m, 1H), 7.93-7.81 (m, 2H), 7.74-7.59 (m, 2H), 5.87-4.23 (m, 3H), 3.78-3.42 (m, 2H), 3.38-3.12 (m, 1H), 2.03-1.54 (m, 4H), 1.11-0.88 (m, 3H), 0.77-0.50 (m, 3H). 19F NMR (377 MHz, DMSO-d6, ppm) 6-61.21 (3F).
Step 1: To a solution of 91-1 (15 g, 99.25 mmol) in MeCN (100 mL) was added CH3NH2 (200 mL, 3.56 mmol, 40% in H2O). Then the reaction mixture was stirred at 80° C. for 16 h. The reaction mixture was quenched with H2O. The mixture was extracted with ethyl acetate. The combined organic layers were washed with H2O. The organic layer was dried over Na2SO4, filtered and concentrated to afford 91-2.
Step 2: To a solution of 91-2 (8.2 g, 50.56 mmol) in MeCN (150 mL) was added NCS (7.4 g, 55.61 mmol), then the reaction mixture was stirred at 60° C. for 70 minutes. The mixture was quenched by addition of water. The aqueous layer was extracted with ethyl acetate. The organic layer was combined, washed with brine, dried over anhydrous Na2SO4, filtered and concentrated. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 5/1) to afford 91-3.
Step 3: Compound 91-4 and 91-5 was prepared from compound 91-3 following the procedure for the synthesis of compound 5-8 in example 5, the product was obtained by reverse phase HPLC (MeCN/water, 55%-65%).
Step 4:91-4 (1.2 g) was purified by SFC (column: REGIS (S,S)WHELK-01, 250×25 mm I.D., 10 μm, A for CO2 and B for Ethanol) to afford 91-6 (200 mg) and 91-7 (250 mg) respectively. 91-6: SFC analysis: 100.00% ee; retention time: 5.932 min; column: ChiralPak AD, 250×30 mm I.D., 10 μm, A for CO2 and B for Ethanol, 40%; pressure: 100 bar; flow rate: 70 mL/min. 91-7: SFC analysis: 98.76% cc; retention time: 7.760 min; column: ChiralPak AD, 250×30 mm I.D., 10 μm, A for CO2 and B for Ethanol, 40%; pressure: 100 bar; flow rate: 70 mL/min.
Step 5: To a solution of 91-6 (180 mg, 0.39 mmol) in DCM (1 mL) under N2 at −78° C. was added dropwise BBr3 (3.86 mL, 3.86 mmol). After the addition was completed, the mixture was stirred at room temperature for 16 h. Ice water was added to the mixture to quench the reaction and the precipitation was collected by filtration, washed with H2O and dried to afford 91-8.
Step 6: The mixture of 91-8 (160 mg, 0.35 mmol), 3-bromotetrahydrofuran (67.84 mg, 0.45 mmol) and Cs2CO3 (266.2 mg, 0.82 mmol) in DMF (5 mL) was stirred at 100° C. for 6 h. Then the mixture was washed with water and extracted with ethyl acetate. The organic layer was dried over Na2SO4, filtered and concentrated to afford 91-9.
Step 7: 91-9 (110 mg) was purified by SFC (column: ChiralPak AD, 250×30 mm I.D., 10 μm, A for CO2 and B for Ethanol) to afford 91 (26 mg) and 92 (15 mg) respectively. 91: SFC analysis: 100.00% ee; retention time: 2.519 min; column: ChiralPak AD, 250×30 mm I.D., 10 μm, A for CO2 and B for Ethanol, 35%; pressure: 100 bar; flow rate: 80 mL/min. LCMS (ESI, m/z): [M+H]+=523.4; 1H NMR (400 MHz, DMSO-d(, ppm): δ 7.77 (s, 1H), 7.57 (d, J=8.4 Hz, 1H), 7.52 (s, 1H), 7.19 (d, J=8.4 Hz, 1H), 6.91 (s, 1H), 5.45-5.40 (m, 1H), 4.18-4.12 (m, 1H), 3.99-3.93 (m, 1H), 3.89-3.85 (m, 3H), 3.82-3.71 (m, 2H), 3.48 (s, 3H), 3.45-3.40 (m, 2H), 2.38-2.32 (m, 2H), 2.30-2.26 (m, 2H), 2.21-2.17 (m, 1H), 2.15-1.97 (m, 3H), 1.58-1.52 (m, 1H), 1.45-1.33 (m, 1H), 0.71 (t, J=7.6 Hz, 3H). 92: SFC analysis: 100.00% ee; retention time: 2.012 min; column: ChiralPak AD, 250×30 mm I.D., 10 μm, A for CO2 and B for Ethanol, 35%; pressure: 100 bar; flow rate: 80 mL/min. LCMS (ESI, m/z): [M+H]+=523.4; 1H NMR (400 MHz, DMSO-d6, ppm): δ 7.77 (s, 1H), 7.61-7.48 (m, 2H), 7.19 (d, J=8.0 Hz, 1H), 6.91 (s, 1H), 5.92-5.30 (m, 1H), 4.20-4.15 (m, 1H), 3.98-3.92 (m, 1H), 3.92-3.84 (m, 3H), 3.80-3.70 (m, 2H), 3.55-3.45 (m, 5H), 2.38-2.32 (m, 2H), 2.30-2.26 (m, 2H), 2.21-2.15 (m, 1H), 2.15-1.98 (m, 3H), 1.60-1.52 (m, 1H), 1.43-1.34 (m, 1H), 0.71 (t, J=7.0 Hz, 3H).
Step 1: To a solution of 84-4 (179.8 mg, 0.53 mmol) in DCM (2 mL) was added dropwise a solution of 1-isocyanato-4-(trifluoromethyl)benzene (100 mg, 0.53 mmol) in DCM (2 mL). The resulting mixture was stirred at room temperature for 16 h. The resulting mixture was concentrated, the residue was purified by column chromatography on silica gel (DCM/MeOH=10/1) to afford 95. LCMS (ESI, m/z): [M+H]+=524.2. 1H NMR (400 MHz, DMSO-d6, ppm) 8 9.55 (s, 1H), 8.98-8.91 (m, 2H), 8.63-8.55 (m, 1H), 7.75-7.66 (m, 2H), 7.64-7.57 (m, 2H), 5.51-4.83 (m, 2H), 4.41 (m, 1H), 4.20-4.07 (m, 1H), 3.58 (m, 1H), 3.33-3.13 (m, 1H), 1.86 1.63 (m, 4H), 0.97-0.85 (m, 6H). 19F NMR (377 MHz, DMSO-d6, ppm) 6-59.99 (s, 3F).
Step 1: Compound 98-1 was prepared from compound 28-12 following the procedure for the synthesis of compound 28-14 in example 11.
Step 2: To a 0° C. solution of 98-2 (4 g, 17.08 mmol) in DCM (100 mL) were added triethylamine (5.18 g, 51.24 mmol), DMAP (357.2 mg, 1.71 mmol) and 4-methylbenzenesulfonyl chloride (4.88 g, 25.62 mmol). Then the reaction mixture was stirred at room temperature for 4 h. The reaction was quenched with H2O, extracted with DCM. The organic layer was dried over anhydrous Na2SO4, filtered and concentrated. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 5/1) to afford 98-3.
Step 3: To a solution of 98-3 (3.8 g, 9.78 mmol) in THF (100 mL) were added TBAF (1M in THF, 14.7 mL, 14.7 mmol) and TMSCN (1.16 g, 11.74 mmol). The reaction mixture was stirred at 60° C. for 1 h. Then the reaction was quenched with H2O, extracted with ethyl acetate. The organic layer was dried over anhydrous Na2SO4, filtered and concentrated. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 4/1) to afford 98-4.
Step 4: To a solution of 98-4 (1 g, 4.11 mmol) in acetone (20 mL) was added K2CO3 (1.14 g, 8.22 mmol) and H2O2 (4 mL). Then the mixture was stirred at 40° C. for 48 h. Then the reaction was quenched with H2O, extracted with ethyl acetate. The organic layer was dried over anhydrous Na2SO4, filtered and concentrated. The residue was purified by column chromatography on silica gel (DCM/MeOH=1/0 to 10/1) to afford 98-5.
Step 5: To a solution of 98-5 (670 mg, 2.56 mmol) in CCl4 (10 mL) were added AIBN (42 mg, 0.26 mmol) and NBS (683.44 mg, 3.84 mmol). Then the reaction mixture was stirred at 90° C. for 16 h. Then the reaction was quenched with H2O, extracted with ethyl acetate. The organic layer was dried over anhydrous Na2SO4, filtered and concentrated. The residue was purified by column chromatography on silica gel (DCM/MeOH=1/0 to 10/1) to afford 98-6.
Step 6: To a solution of 98-6 (200 mg, 0.59 mmol) in MeCN (4 mL) were added 98-1 (265.5 mg, 0.59 mmol), DIPEA (380 mg, 2.94 mmol) and NaI (105.8 mg, 0.71 mmol). Then the reaction was stirred at 90° C. for 2 h. The solvent was removed under vacuum, and the residue was purified by reverse phase HPLC (MeCN/H2O, 5-80%) to afford 98-7.
Step 7: To a solution of 98-7 (110 mg, 0.19 mmol) in THF (4 mL) was added Lawesson's reagent (74.83 mg, 0.19 mmol). Then the reaction mixture was stirred at 50° C. for 30 h. The solvent was removed under vacuum, and the residue was purified by reverse phase HPLC (MeCN/H2O, 5-80%) to afford 98-8.
Step 8: To a solution of 98-8 (30 mg, 0.049 mmol) in EtOH (1 mL) was added 2-bromo-1-cyclopropylethan-1-one (16 mg, 0.098 mmol). Then the reaction mixture was stirred at 80° C. for 2 h. The solvent was removed under vacuum. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 2/3) to afford 98-9.
Step 9: To a mixture of 98-9 (20 mg, 0.03 mmol) in DMF (1 mL) was added Zn(CN)2 (10.6 mg, 0.09 mmol) and Pd(PPh3)4 (3.5 mg, 0.003 mmol). Then the mixture was stirred at 130° C. for 2 h under microwave. The reaction mixture was purified by reverse phase HPLC (MeCN/H2O (0.05% TFA), 5-80%) to afford 98 (5 mg) and 99 (7 mg). 98: LCMS (ESI, m/z): [M+H]+=666.4; 1H NMR (400 MHz, DMSO-d6, ppm): δ 8.23 (d, J=8.8 Hz, 1H), 8.01-7.88 (m, 3H), 7.79-7.70 (m, 2H), 7.25 (s, 1H), 5.96-5.82 (m, 0.5H), 5.42-5.38 (m, 0.5H), 5.22 (s, 1H), 4.98-4.83 (m, 1H), 3.62-3.58 (m, 0.5H), 3.43 (s, 3H), 3.38-3.32 (m, 0.5H), 2.80-2.71 (m, 1H), 2.23-2.18 (m, 1H), 2.05-1.88 (m, 3H), 1.49-1.38 (m, 3H), 0.86-0.77 (m, 6H), 0.70-0.64 (m, 1H), 0.62-0.45 (m, 3H). 19F NMR (376 MHz, DMSO-d6, ppm): δ 87.47 (1F), 64.17 (4F). 99: LCMS (ESI, m/z): [M+H]+=666.4; 1H NMR (400 MHz, DMSO-d(, ppm): δ 8.28-8.19 (m, 1H), 8.02-7.88 (m, 3H), 7.82-7.73 (m, 2H), 7.30 (s, 1H), 5.95-5.82 (m, 0.5H), 5.43-5.38 (m, 0.5H), 5.27 (s, 1H), 5.01-4.85 (m, 1H), 3.62-3.58 (m, 0.5H), 3.49-3.42 (m, 3.5H), 2.20-2.11 (m, 1H), 2.08-1.84 (m, 4H), 1.73-1.37 (m, 3H), 0.90-0.82 (m, 2H), 0.79-0.61 (m, 8H). 19F NMR (376 MHz, DMSO-dd, ppm): δ 87.33 (1F), 64.18 (4F).
Step 1: To a mixture of 102-1 (400 mg, 2.48 mmol), tert-butyl 2-ethyl-4-oxopiperidine-1-carboxylate (564.3 mg, 2.48 mmol) in THF (10 mL) was added dropwise TiCl4 (235.4 mg, 1.24 mmol) at room temperature. The mixture was stirred for another 30 minutes, then sodium triacetoxyborohydride (1052.3 mg, 4.97 mmol) was added and the mixture was stirred at room temperature for 12 h. The mixture was concentrated. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 4/1) to afford 102-2.
Step 2: To a mixture of 102-2 (646 mg, 1.74 mmol), acetaldehyde (382.1 mg, 8.68 mmol) in THF (10 mL) was added dropwise TiCl4 (164.5 mg, 0.87 mmol) at room temperature. The mixture was stirred for another 30 minutes, then and sodium triacetoxyborohydride (735.2 mg, 3.47 mmol) was added and the mixture was stirred at room temperature for 12 h. The reaction mixture concentrated. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=10/1 to 3/1) to afford 102-3.
Step 3: To a solution of 102-3 (345 mg, 0.86 mmol) in DCM (5 mL) was added ethyl acetate/HCl (0.43 mL) and the mixture was stirred at room temperature for 2 h. The reaction mixture was concentrated to dryness to afford 102-4 which was used for the next step directly without further purification.
Step 4: Compound 102 was prepared from compound 102-4 following the procedure for the synthesis of compound 84-3 in example 25. LCMS (ESI, m/z): [M+H]+=495.3; 1H NMR (400 MHz, DMSO-d6, ppm): δ9.52 (s, 1H), 8.95-8.93 (d, J=8.8 Hz, 1H), 8.58-8.56 (d, J=8.8 Hz, 1H), 7.39-7.37 (d, J=8.8 Hz, 2H), 6.83-6.81 (d, J=8.8 Hz, 2H), 4.75-4.69 (m, 2H), 4.05-4.03 (m, 1H), 3.72-3.71 (m, 1H), 3.45-3.34 (m, 2H), 2.34-2.33 (m, 1H), 2.14-1.75 (m, 5H), 1.17-0.98 (m, 6H). 19F NMR (376 MHz, DMSO-d6, ppm): δ −58.90 (3F).
Step 1: To a mixture of 103-1 (9.50 g, 26.79 mmol) and ethyl (2E)-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)prop-2-enoate (7.87 g, 34.83 mmol) in dioxane (95 mL) and H2O (9.5 mL) were added Pd(dppf)Cl2 (2.0 g, 2.68 mmol) and Na2CO3 (5.7 g, 53.59 mmol) at room temperature. Then the mixture was heated at 100 for 12 h. The mixture was filtered and the filtrate was concentrated under vacuo. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 1/4) to afford 103-2.
Step 2: To a mixture of 103-2 (12 g, 25.71 mmol) and CoCl2·6H2O (0.6 g, 2.57 mmol) in MeOH (360 mL) and THF (180 mL) was added NaBH4 (5.8 g, 154.23 mmol) in portions at 0. Then the mixture was stirred at room temperature for 1 h. H2O was added to the reaction mixture. This mixture was extracted with ethyl acetate. The combined organic layers were dried over Na2SO4, filtered and the filtrate was concentrated under vacuo. The residue was purified by column chromatography on silica gel (DCM) to afford 103-3.
Step 3: 103-3 (7.6 g, 23.12 mmol) in HCl/ethyl acetate (100 mL) was stirred at room temperature for 1 h. The mixture was concentrated under vacuo to afford 103-4.
Step 4: Compound 103 was prepared from compound 103-4 following the procedure for the synthesis of compound 59 in example 20. LCMS (ESI, m/z): [M+H]+=511.2; 1H NMR (400 MHz, DMSO-d6, ppm): δ 8.07 (s, 1H), 7.82-7.77 (m, 2H), 7.49-7.47 (m, 2H), 4.92-4.17 (m, 4H), 3.91-3.83 (m, 2H), 3.35 (s, 2H), 2.96-2.89 (m, 2H), 2.02-1.90 (m, 6H). 19F NMR (376 MHz, DMSO-d6, ppm): δ −56.75 (3F).
Step 1: The mixture of 104-1 (5.0 g, 44.59 mmol), 1-(pyridin-3-yl)propan-1-one (7.23 g, 53.51 mmol), AgNO3 (2.27 g, 13.38 mmol), H2SO4 (9.75 g, 89.18 mmol) and ammonium persulfate (10.17 g, 44.59 mmol) in H2O (70 mL) and DCM (70 mL) was flushed with N2 for 0.5 min, then the mixture was stirred at 25° C. for 18 h. The mixture quenched by addition of saturated NaHCO3 solution, the aqueous layer was extracted with DCM. The organic extracts were combined, washed with brine, dried over anhydrous Na2SO4. The solvent was concentrated and the residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=3/1) to afford 104-2.
Step 2: To a solution of 104-2 (560 mg, 2.78 mmol) in MeOH (10 mL) was added NaBH4 (12.3 mg, 3.34 mmol) at 0° C., the reaction mixture was stirred at room temperature for 2 h. The mixture was quenched by addition of H2O, and the aqueous layer was extracted with ethyl acetate. The organic extracts were combined, washed with brine, dried over anhydrous Na2SO4 and concentrated to afford 104-3.
Step 3: To a solution of 104-3 (580 mg, 2.85 mmol) in DCM (15 mL) were added DMF (21.0 mg, 0.29 mmol) and SOCl2 (509 mg, 4.28 mmol) at 0° C., the reaction mixture was stirred at room temperature for 3 hr. The solvent was removed under reduced pressure to afford 104-4 which was used for next step directly without further purification.
Step 4: The mixture of 28-9 (100 mg, 0.43 mmol), 104-4 (239 mg, 1.08 mmol), DIPEA (0.29 mL, 1.72 mmol) and NaI (129 mg, 0.86 mmol) in MeCN (2 mL) was stirred at 85° C. for 18 h. The mixture was filtered and concentrated, the residue was purified by reverse phase HPLC (0.05% NH4HCO3 in water/MeCN, 5-95%) to afford 104-5.
Step 5: To a solution of 104-5 (80 mg, 0.19 mmol) in 1,2-dichloroethane (1 mL) was added 1-chloroethyl chloromethanoate (274.5 mg, 1.92 mmol), the reaction was stirred at 110° C. for 96 h. The mixture was concentrated in vacuo, the residue was diluted with THF and H2O. Then the mixture was stirred at 70° C. for 3 h. The solvent was removed under reduced pressure to afford 104-6 which was used for next step directly without further purification.
Step 6: Compound 104 was prepared from compound 104-6 following the procedure for the synthesis of compound 84-3 in example 25. LCMS (ESI, m/z): [M+H]+=522.4. 1H NMR (400 MHz, DMSO-d6, ppm) δ 9.49-9.46 (m, 1H), 8.92-8.87 (m, 1H), 8.56-8.51 (m, 1H), 8.42-8.37 (m, 1H), 7.73-7.67 (m, 1H), 7.30-7.23 (m, 1H), 5.94-5.83 (m, 0.5H), 5.45-5.23 (m, 0.5H), 5.21-4.74 (m, 1H), 3.64-3.47 (m, 1H), 3.46-3.40 (m, 0.5H), 3.30-3.07 (m, 0.5H), 2.92-2.80 (m, 1H), 2.56-2.55 (m, 1H), 2.30-2.23 (m, 1H), 2.14-2.08 (m, 6H), 2.00-1.82 (m, 3H), 1.70-1.35 (m, 4H), 1.03-0.94 (m, 3H), 0.72-0.58 (m, 6H).
Step 1: To a stirred solution of 107-1 (500 g, 3.27 mol) in dry DCM (5000 mL) was added benzaldehyde (352 g, 3.32 mol), K2CO3 (494.9 g, 3.58 mol) and Na2SO4 (462 g, 3.25 mol). The reaction mixture was stirred at room temperature for 6 h. Then sodium triacetoxyborohydride (1030 g, 4.86 mol) was added in portion wise over 30 minutes to the mixture at ice-bath. The mixture was stirred at room temperature for 16 h. The solid was removed by filtration and the solvent was removed under reduced pressure. The residue was partitioned between ethyl acetate and HCl (1N). The mixture was extracted with ethyl acetate. The pH of the aqueous layer was adjusted to 8.0 with NaHCO3(aq) and the milky aqueous layer was extracted immediately with ethyl acetate. The combined organic layer was washed with brine, dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to afford 107-2.
Step 2: To a solution of 107-2 in DMF (3500 mL) were added DIPEA (885 mL, 5.08 mol) and HATU (966 g, 2.54 mol), the reaction mixture was stirred at 0° C. for 10 minutes, then methyl (2R)-2-(benzylamino)butanoate (351 g, 1.69 mol) was added to the reaction mixture. The result mixture was stirred at room temperature for 16 h. The reaction mixture was diluted with ethyl acetate and washed with water. The organic layer was dried over anhydrous Na2SO4, filtered and evaporated under reduced pressure to afford 107-3.
Step 3: Compound 107-4 was prepared from compound 107-3 following the procedure for the synthesis of compound 67-14 in example 22.
Step 4: To a stirred solution of 107-4 (4.8 g, 11.31 mmol) in isopropanol (150 mL) was added Pd/C (10%, 0.96 g, 9.02 mmol) at room temperature, the resulting mixture was stirred at 80 under H2 balloon for 15 h. The reaction mixture was filtered through a pad of celite and the filtrate was concentrated to afford 107-5.
Step 5: A mixture of 107-5 (1 g, 3.89 mmol), 34-2 (1.73 g, 7.78 mmol), NaI (1.2 g, 7.78 mmol), DIPEA (2.03 mL 11.66 mmol) and K2CO3 (6.1 g, 44.2 mmol) in CH3CN (15 mL) was stirred at 85 for 15 h. Another portion of DIPEA (2.03 mL, 11.66 mmol), NaI (1.2 g, 7.78 mmol) and 34-2 (1.73 g, 7.78 mmol) was added into the reaction mixture at 20° C. The resulting mixture was stirred at 85
under N2 for 15 h. The reaction mixture was concentrated. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 2/3) to afford 107-6.
Step 6: To a stirred solution of 107-6 (4.57 g, 10.09 mmol) in MeOH (20 mL) was added dropwise HCl in ethyl acetate (4M, 9.1 mL) at room temperature, the resulting mixture was stirred at 2 for 15 h. The reaction mixture was concentrated under reduced pressure to afford 107-7.
Step 7: Compound 107-8 was prepared from compound 107-7 following the procedure for the synthesis of compound 67-20 in example 22.
Step 8: To a stirred solution of 107-8 (350 mg, 0.63 mmol) in DMF (10 mL) was added CsF (285.5 mg, 1.88 mmol) at 15° C., the resulting mixture was stirred at 110 for 15 h. The reaction mixture was diluted with H2O and extracted with ethyl acetate. The organic layer was combined, washed with H2O and brine, dried over anhydrous Na2SO4 and concentrated. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 0/1) to afford 107 (60 mg) and 108 (crude, 110 mg), the crude 108 was purification again by reverse phase HPLC (MeCN/water (0.5% FA): 30/70) to afford pure 108 (44 mg) as 0.33 FA salt. 107: LCMS (ESI, m/z): [M+H]+=512.4. 1H NMR (400 MHz, Methanol-d4, ppm) δ 8.04 (d, J=8.7 Hz, 1H), 7.89 (d, J=8.7 Hz, 1H), 7.69-7.62 (m, 2H), 7.61-7.54 (m, 2H), 4.36-4.25 (m, 1H), 4.09-4.03 (m, 1H), 4.02-3.92 (m, 1H), 3.87-3.78 (m, 1H), 3.79-3.70 (m, 4H), 3.10-2.97 (m, 2H), 2.96-2.85 (m, 1H), 2.50-2.40 (m, 1H), 2.20-2.05 (m, 1H), 1.98-1.85 (m, 1H), 1.80-1.65 (m, 2H), 1.22 (t, J=7.4 Hz, 3H), 0.72 (t, J=7.3 Hz, 3H). 19F NMR (376 MHz, DMSO-d6, ppm): δ −63.83 (3F). 108: LCMS (ESI, m/z): [M+H]+=512.4. 1H NMR (400 MHz, Methanol-d4, ppm) δ 8.01 (s, 0.33 H), 8.00-7.98 (m, 1H), 7.86-7.83 (m, 1H), 7.65-7.63 (m, 2H), 7.48-7.46 (m, 2H), 4.72-4.60 (m, 1H), 4.40-4.32 (m, 1H), 4.15-4.00 (m, 2H), 3.68 (s, 3H), 3.45-3.38 (m, 1H), 3.30-3.26 (m, 1H), 3.23-3.16 (m, 1H), 2.38-2.37 (m, 1H), 2.26-2.20 (m, 1H), 2.10-1.92 (m, 2H), 1.90-1.70 (m, 2H), 1.03 (t, J=7.4 Hz, 3H), 0.87 (t, J=7.3 Hz, 3H); 19F NMR (376 MHz, DMSO-d6, ppm): δ −63.91 (3F).
Step 1: A solution of 34-9 (845 mg, 1.63 mmol), tert-butyl carbamate (287.2 mg, 2.45 mmol), Cs2CO3 (1.06 g, 3.27 mmol) and XantPhos Pd G2 (17.2 mg, 0.019 mmol) in dioxane (15 mL) was stirred at 100 under N2 for 6 h. The mixture was concentrated. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 3/1) to afford 109-1.
Step 2: To a solution of 109-1 (660 mg, 1.1 mmol) in DCM (8 mL) was added HCl/ethyl acetate (6 mL). The mixture was stirred at 30 for 12 h. The mixture was concentrated to afford 109-2.
Step 3: A solution of 109-2 (611 mg, 1.23 mmol), 2-chloroacetaldehyde (5 mL, 0.49 mmol) and AcOH (0.070 mL, 1.23 mmol) in EtOH (12 mL) was stirred at 80 for 16 h. The mixture was concentrated. The residue was purified by reverse phase HPLC (MeCN/water (0.5% FA): 5%-80%) to afford 109-3.
Step 4: 109-3 (80 mg) was purified by prep-SFC (column: ChiralPak AD, 250×30 mm I.D., 5 μm, Ethanol/CO2=40/60) to afford 109 (42 mg) and 110 (17 mg). 109: SFC analysis: 100% ee; retention time: 1.393 min; column: Chiralpak AD-3 150×4.6 mm I.D., 3 μm, 40% of Ethanol (0.05% of DEA) in CO2; pressure: 100 bar; flow rate: 2.5 mL/min; LCMS (ESI, m/z): [M+H]+=522.3; 1H NMR (400 MHz, DMSO-d6, ppm) δ 8.84-8.82 (d, J=8.8 Hz, 1H), 8.47-8.45 (d, J=8.8 Hz, 1H), 8.184-8.181 (d, J=1.2 Hz, 1H), 7.75-7.73 (d, J=8.0 Hz, 2H1), 7.59-7.57 (d, J=8.0 Hz, 2H), 7.328-7.325 (d, J=1.2 Hz, 1H), 5.45-4.80 (m, 2H), 3.73-3.54 (m, 1H), 3.35-3.26 (m, 1H), 3.01-2.91 (m, 1H), 2.88-2.80 (m, 1H), 2.36-2.31 (m, 1H), 2.12-1.88 (m, 3H), 1.67-1.39 (m, 3H), 1.03-0.86 (m, 3H), 0.71-0.52 (m, 6H). 19F NMR (377 MHz, DMSO-d6, ppm) 6-60.66 (3F). 110: SFC analysis: 100% ee; retention time: 3.046 min; column: Chiralpak AD-3 150×4.6 mm I.D., 3 μm, 40% of Ethanol (0.05% of DEA) in CO2; pressure: 100 bar; flow rate: 2.5 mL/min; LCMS (ESI, m/z): [M+H]+=522.4; 1H NMR (400 MHz, DMSO-d6, ppm) δ 8.86-8.84 (d, J=8.8 Hz, 1H), 8.49-8.47 (d, J=8.8 Hz, 1H), 8.26-8.14 (m, 1H), 7.73-7.71 (d, J=8.0 Hz, 2H), 7.60-7.58 (d, J=8.0 Hz, 2H), 7.42-7.26 (m, 1H), 5.75-4.55 (m, 2H), 3.61-3.41 (m, 2H), 3.22-3.02 (m, 1H), 2.66-2.53 (m, 1H), 2.27-2.12 (m, 1H), 2.03-1.73 (m, 3H), 1.69-1.36 (m, 3H), 1.06-0.90 (m, 3H), 0.71-0.49 (m, 6H). 19F NMR (377 MHz, DMSO-d6, ppm) 6-60.67 (3F).
Step 1: To a solution of 28-11 (3.0 g, 12.38 mmol) in DCM (30 mL) was added DIPEA (5.12 mL, 30.95 mmol) and benzyl carbonochloridate (3.49 mL, 24.76 mmol), then the reaction mixture was stirred at 20° C. for 18 h. The reaction was concentrated in vacuo. The residue was diluted with ethyl acetate and water. The mixture was extracted with ethyl acetate. The combined organic layer was washed with NaCl aqueous and dried by Na2SO4, the solvent was concentrated and the residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 3/1) to afford 113-1.
Step 2: To a solution of 113-1 (4.5 g, 11.98 mmol) in DCM (30 mL) was added TFA (6 mL, 11.98 mmol) and the reaction was stirred at 20° C. for 18 h. The reaction mixture was concentrated under vacuo, the residue was dilute with DCM and neutralized by NaHCO3 aqueous. The mixture was extracted with DCM. The combined organic layer was concentrated to afford 113-2.
Step 3: To a solution of 113-2 (500 mg, 1.81 mmol) in MeCN (20 mL) was added DIPEA (0.6 mL, 3.62 mmol), NaI (543 mg, 3.62 mmol) and 44-6 (900 mg, 3.62 mmol), and the reaction mixture was stirred at 80° C. for 24 h. The reaction was concentrated in vacuo. The residue was diluted with ethyl acetate and water. The mixture was extracted with ethyl acetate. The combined organic layer was washed with NaCl aqueous and dried by Na2SO4, filtered and the solvent was concentrated. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 1/1) to afford 113-3.
Step 4: To a solution of 113-4 (280 mg, 0.57 mmol) in MeOH (10 mL) was added Pd/C 10% (6.1 mg, 0.057 mmol) under N2. The then the reaction was stirred at 25° C. for 18 h at H2 atmosphere. The mixture was filtered and the filtrate was concentrated to afford 113-4.
Step 5: Compound 113-5 was prepared from compound 113-4 following the procedure for the synthesis of compound 84-3 in example 25.
Step 6: 113-5 (73 mg) was purified by prep-SFC (column: ChiralPak IG, 250×30 mm I.D., 10 μm, MeOH(0.1% NH3H2O)/CO2=40/60) to afford 113 (34.8 mg) and 114 (7.68 mg). 113: SFC analysis: 95% ee; retention time: 5.52 min; column: ChiralPak IG, 100×4.6 mm I.D., 3 μm, MeOH (0.05% DEA) in CO2, 5% to 40%; pressure: 100 bar; flow rate: 2.5 mL/min. LCMS (ESI, m/z): [M+H]+=549.4. 1H NMR (400 MHz, DMSO-d6, ppm): δ 9.50 (s, 1H), 8.93-8.91 (d, J=8.4 Hz, 1H), 8.56-8.54 (d, J=8.4 Hz, 1H), 7.53-7.40 (m, 3H), 6.18-4.42 (m, 2H), 3.86-3.77 (m, 1H), 3.69-3.40 (m, 2H), 3.04 (m, 1H), 2.83-2.71 (m, 1H), 2.62-2.53 (m, 1H), 2.15-1.90 (m, 5H), 1.87-1.71 (m, 2H), 1.65-1.48 (m, 3H), 1.39-1.27 (m, 1H), 1.06-0.97 (m, 3H), 0.68-0.51 (m, 3H). 19F NMR (376 MHz, DMSO-d6, ppm) 6-60.77 (3F). 114: SFC analysis: 100% ee; retention time: 5.82 min; column: ChiralPak IG, 100×4.6 mm I.D., 3 μm, MeOH (0.05% DEA) in CO2, 5% to 40%; pressure: 100 bar; flow rate: 2.5 mL/min. LCMS (ESI, m/z): [M+H]+=549.4. 1H NMR (400 MHz, methanol-d4, ppm): δ 9.33 (s, 1H), 8.80-8.74 (m, 1H), 8.35-8.25 (m, 1H), 7.43-7.35 (m, 3H), 6.00-5.65 (m, 1H), 5.15-4.89 (m, 1H), 3.82-3.77 (m, 1H), 3.73-3.63 (m, 1H), 3.06-3.00 (m, 1H), 2.78-2.67 (m, 2H), 2.36-2.03 (m, 6H), 1.78-1.70 (m, 1H), 1.67-1.50 (m, 3H), 1.44-1.29 (m, 2H), 1.16-1.08 (m, 3H), 0.68-0.51 (m, 3H). 19F NMR (376 MHz, methanol-d4, ppm): δ −63.91 (3F).
Step 1: To a solution of 28-9 (3 g, 12.91 mmol) and methyl 2-bromobutanoate (7.01 g, 38.73 mmol) in MeCN (50 mL) were added DIPEA (6.40 mL, 38.73 mmol) and NaI (1.9 g, 12.91 mmol) at room temperature. Then the mixture was heated at 80 for 12 h. The mixture was concentrated under vacuo, the residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 10/1) to afford 119-1.
Step 2: To a solution of 119-1 (3.4 g, 10.23 mmol) in propan-2-ol (80 mL) was added (Boc)2O (3.05 mL, 13.29 mmol) and Pd/C (10%, 0.5 g, 4.67 mmol) at room temperature. Then the mixture was stirred at 30 under H2 balloon for 12 h. The mixture was filtered and the filtrate was concentrated under vacuo. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 6/1) to afford 119-2.
Step 3: To a solution of 119-2 (3.0 g, 8.76 mmol) in THF (30 mL) and H2O (20 mL) was added LiOH (0.6 g, 13.14 mmol) at room temperature. Then the mixture was stirred at 30 for 12 h. NaOH (1.1 g, 26.28 mmol) was added to the reaction mixture. The mixture was heated at 80
for 12 h. The solvent was removed under vacuo. The pH of the residue was adjusted to 3 with HCl(6N). The mixture was extracted with DCM. The combined organic layers were dried over Na2SO4, filtered and the filtrate was concentrated to afford 119-3.
Step 4: To a mixture of 119-3 (2.7 g, 8.22 mmol) and N-hydroxycyclopropanecarboximidamide (0.82 g, 8.22 mmol) in DMF (80 mL) was added BOP (10.9 g, 24.66 mmol) and triethylamine (3.43 mL, 24.66 mmol) at room temperature. Then the mixture was stirred at room temperature for 0.5 h and heated at 90 for 4 h. The mixture was concentrated under vacuo. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 1/1) to afford 119-4.
Step 5: To a solution of 119-4 (400 mg, 1.02 mmol) in DCM (4 mL) was added TFA (4 mL) at room temperature. Then the mixture was stirred at room temperature for 1 h. The mixture was concentrated under vacuo to afford 119-5 which was used for the next step directly without further purification.
Step 6: Compound 119-6 was prepared from compound 119-5 following the procedure for the synthesis of compound 84-3 in example 25.
Step 7: 119-6 (160 mg) was purified by SFC (column: ChiralCel OX, 250×30 mm I.D., 10 μm, methanol/CO2=30/70) to afford 119 (97.92 mg) and 120 (12.41 mg) respectively. 119: SFC analysis: 99.54% ee; retention time: 7.29 min; column: ChiralCel OX, 100×4.6 mm T.D., 3 μm, 40% of methanol (0.05% DEA) in CO2; pressure: 100 bar; flow rate: 2.5 mL/min. LCMS (ESI, m/z): [M+H]+=487.4. 1H NMR (400 MHz, DMSO-d6, ppm): δ 9.49 (s, 1H), 8.93-8.91 (d, J=8.2 Hz, 1H), 8.58-8.56 (d, J=8.4 Hz, 1H), 5.51-5.21 (m, 1H), 5.12-4.91 (m, 1H), 4.03 (m, 1H), 3.50-3.47 (m, 1H), 3.04-3.02 (m, 1H), 2.68 (m, 2H), 2.16 (s, 1H), 1.96-1.72 (m, 4H), 1.44-1.03 (m, 4H), 0.97-0.76 (m, 11H); 120: SFC analysis: 98.66% ee; retention time: 8.28 min; column: ChiralCel OX, 100×4.6 mm I.D., 3 μm, 40% of methanol (0.05% DEA) in CO2; pressure: 100 bar; flow rate: 2.5 mL/min. LCMS (ESI, m/z): [M+H]+=487.4. 1H NMR (400 MHz, DMSO-d6, ppm): δ 9.50 (s, 1H), 8.92-8.90 (d, J=8.7 Hz, 1H), 8.6-8.54 (d, J=8.6 Hz, 1H), 7.38-7.18 (m, 1H), 5.25-4.90 (m, 1H), 4.04-4.00 (m, 1H), 3.53-3.44 (m, 1H), 3.03-2.94 (m, 1H), 2.87-2.78 (m, 1H), 2.62-2.55 (m, 1H), 2.16-2.09 (m, 1H), 2.08-1.94 (m, 1H), 1.89-1.72 (m. 3H), 1.56-1.44 (m, 1H), 1.10-1.02 (m. 3H), 0.92-0.79 (m, 11H).
Step 1: A solution of 121-1 (4 g, 18.49 mmol), 34-2 (5.35 g, 24.04 mmol), NaI (0.6 g, 3.69 mmol) and DIPEA (7.64 mL, 46.24 mmol) in MeCN (30 mL) was stirred at 85 for 12 h. The mixture was concentrated. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 1/1) to afford 121-2.
Step 2: To a solution of 121-2 (2 g, 4.97 mmol) in DCM (12 mL) was added HCl/ethyl acetate (6 mL) at 15 . The mixture was stirred at 15
for 2 h. The mixture was concentrated to afford 121-3.
Step 3: Compound 121-4 was prepared from compound 121-3 following the procedure for the synthesis of compound 67-20 in example 22.
Step 4: To a solution of 121-4 (225 mg, 0.42 mmol) in DMF (30 mL) was added NaH (50.9 mg, 2.12 mmol) at 15 . The mixture was stirred at 110
for 16 h. water was added to quench the reaction. The mixture was extracted with ethyl acetate. The organic phase was concentrated. The residue was purified by column chromatography on silica gel (DCM/MeOH=1/0 to 10/1) to afford 121-5.
Step 5: To a solution of 121-5 (130 mg, 0.26 mmol) in THF (6 mL) was added triethyl amine (0.072 mL, 0.52 mmol) and TFAA (0.14 mL, 1.04 mmol) at 0 . The mixture was stirred at 15
for 48 h. The mixture was diluted with H2O, extracted with ethyl acetate. The organic phase was concentrated. The residue was purified by reverse phase HPLC (0.5% TFA in water/MeCN, 5-70%) to afford 121-6.
Step 6: 121-6 (70 mg) was purified by SFC (column: ChiralCel OD, 250×30 mm I.D., 5 μm, Ethanol/CO2=30/70) to afford 121 (18 mg) and 122 (5 mg) respectively. 121: SFC analysis: 100% ee; retention time: 2.69 min; column: ChiralCel OD, 150×4.6 mm I.D., 3 μm, 40% of ethanol (0.05% DEA) in CO2; pressure: 100 bar; flow rate: 2.5 mL/min. LCMS (ESI, m/z): [M+H]+=484.4. 1H NMR (400 MHz, DMSO-d6, ppm): δ 8.13-8.00 (m, 2H), 7.78-7.66 (m, 2H), 7.58-7.42 (m, 2H), 4.80-4.50 (m, 1H), 4.34-4.20 (m, 1H), 4.12-3.96 (m, 1H), 3.58 (s, 3H), 3.50-3.42 (m, 1H), 3.34-3.23 (m, 2H), 2.84-2.70 (m, 2H), 2.35-2.25 (m, 1H), 2.22-2.10 (m, 1H), 2.03-1.88 (m, 1H), 1.83-1.68 (m, 1H), 0.73 (t, J=7.0 Hz, 3H). 19F NMR (376 MHz, DMSO-d6, ppm) 6-60.73 (3F); 122: SFC analysis: 99.34% ee; retention time: 3.16 min; column: ChiralCel OD, 150×4.6 mm I.D., 3 μm, 40% of ethanol (0.05% DEA) in CO2; pressure: 100 bar; flow rate: 2.5 mL/min. LCMS (ESI, m/z): [M+H]+=484.4. 1H NMR (400 MHz, DMSO-d6, ppm): δ 8.11-7.97 (m, 2H), 7.78-7.66 (m, 2H), 7.56-7.42 (m, 2H), 4.65-4.35 (m, 1H), 4.32-4.20 (m, 1H), 4.16-4.02 (m, 1H), 3.58 (s, 3H), 3.51-3.40 (m, 1H), 3.33-3.25 (m, 2H), 2.92-2.76 (m, 1H), 2.70-2.57 (m, 1H), 2.34-2.20 (m, 2H), 2.02-1.89 (m, 1H), 1.80-1.66 (m, 1H), 0.74 (t, J=7.2 Hz, 3H). 19F NMR (376 MHz, DMSO-d6, ppm) δ −60.75 (3F).
Step 1: To a stirred solution of 67-14 (2 g, 5.98 mmol) in isopropanol (35 mL) was added Pd/C (10%, 0.5 g, 0.470 mmol) at 25, the resulting mixture was stirred at 50
under H2 balloon for 15 h. The reaction mixture was filtered and the filtrate was concentrated to afford 123-1.
Step 2: Compound 123-2 was prepared from compound 123-1 following the procedure for the synthesis of compound 67-20 in example 22.
Step 3: To a stirred solution of 123-2 (140 mg, 0.3 mmol) in DMF (15 mL) was added potassium tert-butoxide (99.7 mg, 0.89 mmol) at 15, the resulting mixture was stirred at 110
for 20 h. The reaction mixture was concentrated, the residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 0/1) to afford 123-3.
Step 4: Compound 123 and 124 was prepared from compound 123-3 following the procedure for the synthesis of compound 28-15 in example 11. The product was obtained by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 0/1). 123: LCMS (ESI, m/z): [M+H]+=544.4. 1H NMR (400 MHz, DMSO-d6, ppm): δ 8.11-8.06 (m, 2H), 7.71-7.69 (d, J=7.8 Hz, 2H), 7.53-7.51 (d, J=7.8 Hz, 2H), 5.32-5.25 (m, 1H), 4.16-4.14 (m, 1H), 3.83-3.79 (m, 1H), 3.61-3.52 (m, 4H), 3.35-3.31 (m, 1H), 3.09-2.99 (m, 2H), 2.38-2.33 (m, 2H), 2.01-1.86 (m, 2H), 1.79-1.72 (m, 2H), 1.04-1.00 (m, 3H), 0.59-0.52 (m, 3H). 19F NMR (376 MHz, DMSO-d6, ppm) 6-42.17 (3F). 124: LCMS (ESI, m/z): [M+H]+=544.4. 1H NMR (400 MHz, DMSO-d6, ppm): δ 8.05 (s, 2H), 7.72-7.70 (d, J=8.0 Hz, 2H), 7.50-7.48 (d, J=7.8 Hz, 2H), 4.72-4.68 (m, 1H), 4.38-4.35 (m, 1H), 4.04-3.99 (m, 1H), 3.75-3.71 (m, 1H), 3.59 (s, 3H), 3.42-3.39 (m, 1H), 3.11-3.08 (m, 1H), 2.92-2.89 (m, 1H), 2.67-2.59 (m, 1H), 2.36-2.34 (m, 1H), 2.00-1.94 (m, 1H), 1.68-1.66 (m, 3H), 0.75-0.61 (m, 6H). 19F NMR (376 MHz, DMSO-d6, ppm) 6-42.31 (3F).
Step 1: Compound 67 and 145 was prepared from compound 123-3 following the procedure for the synthesis of compound 28-15 in example 11. The product was obtained by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 0/1). 145: LCMS (ESI, m/z): [M+H]+=512.2; 1H NMR (400 MHz, CD3OD, ppm) δ 8.08-8.04 (m, 1H), 7.94-7.90 (m, 1H), 7.67-7.62 (m, 2H), 7.59-7.55 (m, 2H), 5.62-5.52 (m, 1H), 4.18-4.11 (m, 1H), 3.91-3.84 (m, 1H1), 3.71 (s, 3H1), 3.68-3.61 (m, 11H), 3.49-3.41 (m, 11H), 3.25-3.11 (m, 3H1), 2.45-2.41 (m, 1H), 2.05-1.92 (m, 1H), 1.91-1.81 (m, 1H), 1.68-1.53 (m, 2H), 1.11 (t, J=7.4 Hz, 3H), 0.65 (t, J=7.3 Hz, 3H). 19F NMR (376 MHz, CD3OD, ppm) 6-63.86 (3F).
Step 1: To a solution of 154-1 in acetone (200 mL) was added ethyl 4-bromobutanoate (10.63 g, 54.51 mmol), K2CO3 (9.4 g, 68.14 mmol), and NaI (1.0 g, 6.81 mmol), the reaction mixture was stirred at 70° C. for 18 h. The mixture was concentrated in vacuo. The residue was diluted with ethyl acetate and water. The organic layer was separated and washed with NaCl aqueous. The organic layer was dried by Na2SO4, filtered and concentrated, the residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 4/1) to afford 154-2.
Step 2: To a solution of 154-2 (13.7 g, 40.98 mmol) in DMF (300 mL) was added C2H5OH (2.5 mL, 42.82 mmol) and NaH (3.8 g, 94.26 mmol), the mixture was stirred at room temperature for 18 h. Then the reaction mixture was heated to 100° C. for 2 h. The reaction mixture was cooled to room temperature, quenched with HCl. The mixture was concentrated in vacuo. The residue was diluted with ethyl acetate and water. The organic layer was separated and washed with NaCl aqueous. The organic layer was dried by Na2SO4, filtered and concentrated, the residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 1/1) to afford 154-3 and 154-4.
Step 3: To a mixture of 154-3 (3.3 g, 10.92 mmol) and 154-4 (3.3 g, 11.45 mmol) in dioxane (100 mL) was added HCl (25 mL, 10.92 mmol) and the reaction was stirred at 100° C. for 18 h. The mixture was concentrated in vacuo. The residue was diluted with ethyl acetate and neutralized with NaHCO3 aqueous. The organic layer was separated and washed with NaCl aqueous. The organic layer was dried by Na2SO4, filtered and concentrated, the residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 3/2) to afford 154-5.
Step 4: To a solution of 154-5 (4.5 g, 19.55 mmol) in CH3OH (40 mL) was added NaBH4 (813.5 mg, 21.5 mmol) and the reaction was stirred at 0° C. for 18 h. The reaction was quenched with NH4Cl aqueous and concentrated in vacuo. The residue was diluted with ethyl acetate and water. The organic layer was separated and washed with NaCl aqueous. The organic layer was dried by Na2SO4, filtered and concentrated, the residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 3/2) to afford 154-6.
Step 5: Compound 154 was prepared from compound 154-6 following the procedure for the synthesis of compound 44-7 in example 17. The product was obtained by reverse phase HPLC (acetonitrile/H2O: 5%˜95%) to afford 154. LCMS (ESI, m/z): [M+H]+=541.3; 1H NMR (400 MHz, CDCl3, ppm) δ 7.88-7.77 (m, 1H), 7.62-7.54 (m, 1H), 7.43-7.33 (m, 1H), 7.30-7.26 (m, 2H), 6.23-5.74 (m, 1H), 5.63-5.32 (m, 1H), 5.32-5.03 (m, 1H), 4.45-3.93 (m, 2H), 3.88-3.67 (m, 2H), 3.60-3.53 (m, 3H), 3.40-2.91 (m, 1H), 2.86-2.76 (m, 1H), 2.70-2.31 (m, 2H), 2.24-2.13 (m, 2H), 2.02-1.87 (m, 2H), 1.45-1.37 (m, 2H), 1.08-0.91 (m, 3H), 0.83-0.47 (m, 3H). 19F NMR (376 MHz, CDCl3, ppm) 6-62.52 (3F).
Step 1: A solution of 192-1 (6 g, 25.84 mmol), NBS (5.1 g, 28.42 mmol) and AIBN (0.4 g, 2.584 mmol) in CCl4 (60 mL) was stirred at 80 under N2 for 15 h. The reaction mixture was concentrated under vacuo. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 3/1) to afford 192-2.
Step 2: Compound 192-4 was prepared from compound 192-3 following the procedure for the synthesis of compound 67-14 in example 22.
Step 3: To a stirred solution of 192-4 (3.6 g, 10.76 mmol) in isopropanol (50 mL) was added Pd/C (10%, 0.72 g) at 25 T, the resulting mixture was stirred at 50 under H2 balloon for 15 h. The reaction mixture was filtered and the filtrate was concentrated to afford 192-5.
Step 4: To a stirred solution of 192-2 (3.02 g, 9.72 mmol), 192-5 (2.5 g, 9.72 mmol) and NaI (0.15 g, 1.0 mmol) in CH3CN (50 mL) was added DIPEA (8.47 mL, 48.60 mmol) at 25, the resulting mixture was stirred at 95
for 15 h. The reaction mixture was concentrated. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 1/1) to afford 192-6.
Step 5: To a stirred solution of NaBH4 (132.4 mg, 3.50 mmol) in THF (3 mL) was added a cooled solution of 192-6 (500 mg, 1.17 mmol) in THF (6 mL) and Boron trifluoride diethyl etherate (2.9 mL, 1.17 mmol) dropwisely at 0-10 under N2. After addition, the reaction mixture was stirred at 90
for 4.5 h. A cooled solution of HCl was added to quench the reaction. The reaction mixture was diluted with H2O and extracted with ethyl acetate. The organic layer was combined, washed with brine, dried over anhydrous Na2SO4 and concentrated. The residue was purified by column chromatography on silica gel (DCM/methanol=1/0 to 5/1) to afford 192-7.
Step 6: Compound 192 was prepared from compound 192-7 following the procedure for the synthesis of compound 84-3 in example 25. The product was obtained by reverse phase HPLC (0-60% acetonitrile in water) to afford 192 (77 mg). LCMS (ESI, m/z): [M+H]+=509.2; 1H NMR (400 MHz, CD3OD, ppm) δ 9.36 (s, 1H), 8.82-8.76 (m, 1H), 8.32-8.28 (m, 1H), 7.72-7.59 (m, 4H), 5.47 (s, 1H), 4.85-4.61 (m, 1H), 4.01-3.87 (m, 1H), 3.82-3.73 (m, 1H), 3.60-3.48 (m, 2H), 3.448-3.40 (m, 1H), 3.13 (s, 1H), 2.73 (t, J=10.0 Hz, 2H), 2.34-2.02 (m, 3H), 0.88 (t, J=7.5 Hz, 3H). 19F NMR (376 MHz, CD3OD, ppm) δ −64.02 (3F).
Step 1: To a stirred solution of 123-1 (7.5 g, 30.7 mmol) in THF (50 mL) and H2O (50 mL) were added NaHCO3 (7.7 g, 92.09 mmol) and CbzOSu (8.4 g, 33.76 mmol). The reaction mixture was stirred at 25 for 3 h. The reaction mixture was diluted with ethyl acetate, washed
with brine and dried over anhydrous Na2SO4. Evaporation of the solvent under reduced pressure, the residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 3/1) to afford 198-1.
Step 2: NaIO4 (9.9 g, 46.3 mmol) was dissolved in water (50 mL) and CH3CN/dimethyl carbonate (40 ml, 1:1), then Ruthenium(III) chloride trihydrate (60.5 mg, 0.23 mmol) was added to the mixture, followed by a solution of 198-1 (8.76 g, 23.15 mmol) in CH3CN/dimethyl carbonate (60 ml, 1:1) at 0 over 1 minutes. The solution was stirred at 25
for 1 h. The reaction mixture was diluted with water and extracted with ethyl acetate. The combined organics were washed with brine and dried over Na2SO4. The mixture was filtered and concentrated. The residue was partitioned between ethyl acetate and NaHCO3 aqueous. The layers were separated and the aqueous layer was extracted with ethyl acetate. The aqueous layer was adjusted to pH=3 with 2M HCl and the aqueous layer was extracted immediately with ethyl acetate. The combined organic layers were washed with brine, dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to afford 198-2.
Step 3: To a stirred solution of 198-2 (6.4 g, 16.31 mmol) in CH3CN (120 mL) were added K2CO3 (3.4 g, 24.46 mmol) and CH3I (3.5 g, 24.46 mmol). The reaction mixture was stirred at 60° C. for 2 h. The reaction mixture was partitioned between ethyl acetate and H2O. The separated organic layer was washed with brine and dried over anhydrous Na2SO4, filtered and concentrated, the residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 5/11) to afford 198-3.
Step 4: To a stirred solution of 198-3 (5.76 g, 14.17 mmol) in propan-2-ol (50 mL) was added Pd/C (10%, 0.8 g). The reaction mixture was stirred at 25° C. under H2 for 18 h. The reaction mixture was filtered, washed with propan-2-ol, evaporated under reduced pressure to afford 198-4.
Step 5: The mixture of 67-7 (4.4 g, 13.81 mmol), 198-4 (3.76 g, 13.81 mmol) and DIEA (6.85 mL, 41.44 mmol) in CH3CN (50 mL) was stirred at 85° C. for 3 h. The solvent was removed under reduced pressure, the residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 3/1) to afford 198-5.
Step 6: To a stirred solution of 198-5 (6.9 g, 12.45 mmol) in AcOH (70 mL) at 25° C. was added Fe (2.4 g, 43.56 mmol). The resultant suspension was stirred at 80 for 1 h. The reaction mixture was concentrated under reduced pressure. The residue was suspended in CH2Cl2 and aqueous NaHCO3 was added until the solution was adjusted to pH=8. The mixture was filtered. The phases were separated and the aqueous was washed with CH2Cl2. The combined organic extracts were washed with brine, dried over anhydrous Na2SO4, filtered and concentrated, the residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 1/1) to afford 198-6.
Step 7: The mixture of 198-6 (300 mg, 0.61 mmol), K2CO3 (168.4 mg, 1.22 mmol), and CH3I (864.8 mg, 6.09 mmol) in CH3CN (5 mL) was stirred at 70° C. for 18 h. The solvent was removed under reduced pressure, the residue was purified by C18 reverse phase column (CH3CN/H2O (0.05% TFA), 5-95%) to afford 198-7.
Step 8: Compound 198 and 199 was prepared from compound 198-7 following the procedure for the synthesis of compound 28 and 29 in example 11. The product was obtained by SFC (column: ChiralPak AD, 250><30 mm, 10 μm, Ethanol (0.1% NH3H2O)/Supercritical CO2=25/75) to afford 198 (9.5 mg) and 199 (11.5 mg). 198: LCMS (ESI, m/z): [M+H]+=539.4; SFC analysis: 100% ee; retention time: 1.341 min; column: ChiralPak AD, 150×4.6 mm, 3 μm, 40% EtOH (0.05% DEA) in CO2; pressure: 100 bar; flow rate: 2.5 mL/min. 1H NMR (400 MHz, DMSO-d6, ppm): δ 8.18-8.12 (m, 2H), 7.70 (d, J=8.1 Hz, 2H), 7.58 (d, J=8.1 Hz, 2H), 5.17-5.04 (m, 1H), 3.78-3.68 (m, 2H), 3.63 (s, 3H), 3.49-3.39 (m, 1H), 3.24 (s, 3H), 3.09-3.01 (m, 1H), 2.94-2.76 (m, 2H), 1.94-1.83 (m, 1H), 1.69-1.47 (m, 3H), 0.94 (t, J=7.3 Hz, 3H), 0.62 (t, J=7.2 Hz, 3H); 19F NMR (376 MHz, DMSO-d6, ppm): δ −60.69 (3F). 199: LCMS (ESI, m/z): [M+H]+=539.4; SFC analysis: 99.48% ee; retention time: 1.603 min; column: ChiralPak AD, 150×4.6 mm, 3 μm, 40% EtOH (0.05% DEA) in CO2; pressure: 100 bar; flow rate: 2.5 mL/min. 1H NMR (400 MHz, DMSO-d6, ppm): δ 8.18-8.06 (m, 2H), 7.72 (d, J=8.1 Hz, 2H), 7.55 (d, J=8.1 Hz, 2H), 4.03-3.93 (m, 1H), 3.90-3.83 (m, 1H), 3.75-3.65 (m, 2H), 3.63 (s, 3H), 3.42-3.35 (m, 1H), 3.33 (s, 3H), 2.70-2.62 (m, 1H), 2.61-2.53 (m, 1H), 2.10-2.00 (m, 1H), 1.77-1.61 (m, 2H), 1.51-1.43 (m, 1H), 0.85-0.75 (m, 6H); 19F NMR (376 MHz, DMSO-d6, ppm): δ −60.77 (3F) Example 42 Synthesis of Compounds 210 and 211
Step 1: To a stirred solution of 210-1 (10 g, 66.16 mmol) in AcOH (60 mL) and H2O (120 mL) was added iodine (16.8 g, 66.16 mmol) over a period of 10 minutes. The reaction mixture was stirred at room temperature for 48 h. The reaction mixture was filtered and the solid was collected and dried under vacuum to afford 210-2.
Step 2: To a stirred solution of 210-2 (12.1 g, 43.67 mmol) in dioxane (120 mL) was added triphosgene (7.1 g, 24.02 mmol) at 25, the resulting mixture was stirred at 100
for 15 h. The reaction mixture was concentrated under reduced pressure to afford 210-3 which was used for the next step directly.
Step 3: NaH (1.8 g, 45.89 mmol) was added to a mixture of diethyl malonate (27.28 mL, 180.56 mmol) and DMF (240 mL) with stirring at room temperature. A mixture of 210-3 (12 g, 37.62 mmol) and DMF (240 mL) was added dropwise to this solution, the mixture was stirred at 120 for 4 h. The mixture was filtered and the solid was added to H2O (1 L) and HCl (30%, 200 mL), the mixture was stirred and filtered. The solid was collected and dried to afford 210-4.
Step 4: A solution of ethyl 210-4 (12 g, 32.16 mmol) in TFA (95 mL) and HCl (95 mL) was stirred at 100 for 16 h. The reaction mixture was diluted with H2O and filtered. The cake was washed with a large amount of H2O and dried under reduced pressure to afford 210-5.
Step 5: To a stirred solution of 210-5 (3 g, 9.96 mmol) in AcOH (26 mL) was added HNO3 (1.6 mL, 2.46 mmol) at 25, the resulting mixture was stirred at 80
for 1.5 h. The reaction mixture was cooled to 25
and diluted with H2O, after stirred for 10 minutes, the resulting solid was filtered and dried to afford 210-6.
Step 6: To a stirred solution of 210-6 (2.7 g, 7.80 mmol) and Zn (CN)2 (2.7 g, 23.41 mmol) in DMF (40 mL) was added Pd(PPh3)4 (0.9 g, 0.78 mmol) at room temperature, the resulting mixture was stirred at 110 for 36 h under N2. The reaction mixture was concentrated under reduced pressure. The residue was purified by reverse phase column chromatography (0-20% CH3CN in H2O (0.5% FA)) to afford 210-7.
Step 7: To a stirred solution of 210-7 (700 mg, 2.86 mmol) in toluene (15 mL) were added DIPEA (2.98 mL, 17.13 mmol) and POCl3 (1.33 mL, 14.28 mmol) at room temperature, the resulting mixture was stirred at 110 under N2 for 1.5 h. The reaction mixture was concentrated under reduced pressure. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 0/1) to afford 210-8.
Step 8: To a stirred solution of 67-18 (680 mg, 1.76 mmol) in CH3CN (20 mL) were added DIPEA (3.07 mL, 17.63 mmol) and 210-8 (820.4 mg, 2.65 mmol) at room temperature, the resulting mixture was stirred at 85 under N2 for 5 h. The reaction mixture was concentrated under reduced pressure. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 1/2) to afford 210-9.
Step 9: To a stirred solution of 210-9 (320 mg, 0.57 mmol) in NMP (9 mL) was added t-BuOK (193.2 mg, 1.72 mmol) in one portion at room temperature, the resulting mixture was stirred at 120 for 18 h. The reaction mixture was diluted with H2O and extracted with ethyl acetate. The organic layer was combined, washed with brine, dried over anhydrous Na2SO4 and concentrated. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 2/3) by column chromatography (eluting with 0-60% EtOAc in petroleum ether) to afford 210 (8.56 mg) and 211 (22.04 mg). 210: LCMS (ESI, m/z): [M+H]+=511.2. 1H NMR (400 MHz, CDCl3, ppm) δ 7.97 (s, 1H), 7.62-7.52 (m, 3H), 7.45-7.39 (m, 2H), 7.36-7.30 (m, 1H), 4.20 (t, J=10.2 Hz, 1H), 3.90-3.81 (m, 1H), 3.80-3.73 (m, 1H), 3.67 (s, 3H), 3.34-3.28 (m, 1H), 3.09-2.98 (m, 2H), 2.99-2.89 (m, 1H), 2.81-2.72 (m, 1H), 2.51-2.42 (dd, 1H), 1.87-1.71 (m, 2H), 1.68-1.54 (m, 2H), 0.90 (t, J=7.4 Hz, 3H), 0.82 (t, J=7.3 Hz, 3H). 19F NMR (376 MHz, CDCl3, ppm) δ −62.39 (3F). 211: LCMS (ESI, m/z): [M+H]+=511.2. 1H NMR (400 MHz, CDCl3, ppm) δ 7.95-7.86 (m, 1H), 7.62-7.53 (m, 3H), 7.32-7.27 (m, 3H), 4.55 (t, J=10.5 Hz, 1H), 4.25-4.18 (m, 1H), 3.97 (t, J=7.5 Hz, 1H), 3.71-3.64 (m, 3H), 3.27-3.21 (m, 1H), 3.16-3.04 (m, 1H), 3.01-2.92 (m, 1H), 2.77-2.64 (m, 1H), 2.57-2.42 (m, 2H), 1.96-1.82 (m, 1H), 1.80-1.64 (m, 3H), 0.93-0.77 (m, 6H). 19F NMR (376 MHz, CDCl3, ppm) δ −62.46 (3F).
Step 1: To a solution of propionaldehyde (4 g, 68.87 mmol) in THF (60 mL) was added (R)-(2-methylprop-2-yl)(oxo)-λ4-sulfanamine (10.0 g, 82.65 mmol) and Ti(OEt)4 (47.1 g, 206.61 mmol) at room temperature. Then the mixture was heated at 60 for 2 h. The mixture was cooled to room temperature and quenched with H2O. The mixture was filtered and the filtrate was concentrated under vacuo. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 5/1) to afford 219-2.
Step 2: A mixture of 219-3 (6.6 g, 36.83 mmol), Boc2O (12.69 mL, 55.24 mmol) and triethylamine (15.36 mL, 110.48 mmol) in DCM (100 mL) was stirred at room temperature for 2 h. H2O was added to the mixture, then the mixture was extracted with DCM. The combined organic layers were dried over Na2SO4, filtered and the filtrate was concentrated. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 1/1) to afford 219-4.
Step 3: To a solution of 219-4 (4 g, 14.32 mmol) in THF (60 mL) was added LiHMDS (1M, 14.32 mL) at −78 After the mixture was stirred at −78
for 30 minutes, 219-2 (2.31 g, 14.32 mmol) was added. Then the mixture was stirred at −78
for 30 minutes. The mixture was quenched with saturated NH4Cl. The mixture was extracted with ethyl acetate. The combined organic layers were dried over Na2SO4, filtered and the filtrate was concentrated under vacuo. The reside was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 1/2) to afford 219-5.
Step 4: To a solution of 219-5 (6 g, 13.62 mmol) in THF (60 mL) was added LiAlH4 (0.8 g, 20.43 mmol) at 0. Then the mixture was stirred at 0
for 0.5 h. The mixture was quenched with H2O, NaOH (15%) and H2O. The mixture was filtered and the filtrate was concentrated. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 1/2) to afford 219-6.
Step 5: To a solution of 219-6 (2.26 g, 5.48 mmol) in dioxane (10 mL) was added HCl/dioxane (40 mL) at room temperature. Then the mixture was stirred at room temperature for 1 h. This mixture was concentrated under vacuo to afford 219-7.
Step 6: To a mixture of 219-7 (1141 mg, crude) and ethyl 2-ethoxy-2-oxoacetate (1.6 g, 10.95 mmol) in DCM (6 mL) and methanol (24 mL) was added CH3ONa (1183.6 mg, 21.91 mmol) at room temperature. Then the mixture was stirred at room temperature for 12 h and stirred at 60 for 2 h. The mixture was concentrated under vacuo, the residue was purified by column chromatography on silica gel (DCM/methanol=1/0 to 5/1) to afford 219-8.
Step 7: To a solution of 219-8 (1.0 g, 3.81 mmol) in THF (8 mL) was added borane tetrahydrofuran (22.87 mL, 22.87 mmol) at room temperature. Then the mixture was heated at 80 for 1 h. The mixture was quenched with methanol and HCl. This mixture was concentrated under vacuo to afford 219-9.
Step 8: To a solution of 219-9 (893 mg, crude) in THF (10 mL) and H2O (10 mL) were added NaHCO3 (3201.4 mg, 38.11 mmol) and (Boc)2O (1.05 mL, 4.57 mmol) at room temperature. Then the mixture was stirred at room temperature for 30 minutes. H2O was added to the mixture and the mixture was extracted with ethyl acetate. The combined organic layers were dried over Na2SO4, filtered and the filtrate was concentrated under vacuo. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 5/1) to afford 219-10.
Step 9: Compound 219-11 was prepared from compound 219-10 following the procedure for the synthesis of compound 192-7 in example 40.
Step 10: Compound 219 (26 mg) was prepared from compound 219-11 following the procedure for the synthesis of compound 5 in example 5. LCMS (ESI, m/z): [M+H]+=499.2; 1H NMR (400 MHz, DMSO-d6, ppm) δ 8.30-8.19 (m, 1H), 8.05-7.97 (m, 1H), 7.77-7.64 (m, 4H), 5.85-5.32 (m, 1H), 5.24-4.56 (m, 1H), 3.96-3.66 (m, 2H), 3.58-3.47 (m, 1H), 3.45 (s, 3H), 3.40-3.34 (m, 1H), 3.31-3.24 (m, 1H), 3.16-3.06 (m, 1H), 2.78-2.54 (m, 2H), 2.37-2.20 (m, 1H), 2.10-1.99 (m, 1H), 1.74-1.57 (m, 1H), 0.81 (t, J=7.4 Hz, 3H). 19F NMR (376 MHz, DMSO-d6, ppm): δ −60.88-60.90 (3F).
Step 1: The mixture of 67-7 (4 g, 12.56 mmol), 123-1 (3.07 g, 12.56 mmol) and DIPEA (4.9 g, 37.68 mmol) in MeCN (40 mL) was stirred at 85° C. for 3 h. The solvent was removed under reduced pressure, the residue was purified by column chromatography on silica gel (ethyl acetate/DCM=1/3) to afford 220-1.
Step 2: To a stirred solution of 220-1 (3.79 g, 7.2 mmol) in ethyl acetate (40 mL) was added 5% Pt/C (0.7 g, 3.6 mmol). The reaction mixture was stirred at 25° C. under H2 for 18 h. The reaction mixture was filtered, washed with ethyl acetate, evaporated under reduced pressure to afford 220-2.
Step 3: To a solution of 220-2 (3.57 g, 7.19 mmol) and triethylamine (4 mL, 28.77 mmol) in CH2Cl2 (30 mL) was added MsCl (1.6 g, 14.38 mmol) at 0° C. Then the reaction mixture was stirred at 25° C. for 1 h, the solvent was removed under reduced pressure to afford 220-3 which was used for next step directly.
Step 4: To a solution of 220-3 (4.13 g, crude) in DMF (30 mL) was added DIPEA (1.9 g, 14.38 mmol). Then the reaction was stirred at 120° C. for 5 h. The reaction was purified by C18 reverse phase column (MeCN/H2O(0.05% TFA), 5-95%) to afford 220-4.
Step 5: To a solution of 220-4 (1.0 g, 2.09 mmol) in anhydrous THF (10 mL) was added NaH (334.5 mg, 8.36 mmol) at 0° C., the mixture further stirred at 0° C. under N2 for 30 mins. Then CH3I (890.1 mg, 6.27 mmol) in THF (10 mL) was added dropwise and the mixture stirred at 25° C. for 1 h. The reaction was quenched with H2O at 0° C. The mixture was partitioned between ethyl acetate and H2O, the aqueous layer was extracted with ethyl acetate. The combined organic layers were washed with brine. The organic phase was separated, dried over Na2SO4, filtered and concentrated to afford 220-5.
Step 6: Compounds 220 and 221 were prepared from compound 220-5 following the procedure for the synthesis of compound 28-15 in example 11. The products, 220 (14 mg) and 221 (17 mg), were obtained by C18 reverse phase column (MeCN/H2O (0.05% TFA), 5-95%) and column chromatography on silica gel (ethyl acetate/DCM=1/1). 220: LCMS (ESI, m/z): [M+H]+=525.3. 1H NMR (400 MHz, methanol-d4, ppm): δ 8.01 (d, J=8.7 Hz, 1H), 7.87 (d, J=8.7 Hz, 1H), 7.65 (d, J=8.1 Hz, 2H), 7.60 (d, J=8.1 Hz, 2H), 5.33-5.24 (m, 1H), 3.68 (s, 3H), 3.67-3.61 (m, 1H), 3.21-3.10 (m, 2H), 3.07-2.98 (m, 1H), 2.86-2.77 (m, 4H), 2.75-2.67 (m, 1H), 2.42-2.36 (m, 2H), 2.13-1.98 (m, 2H), 1.72-1.60 (m, 2H), 1.29-1.23 (m, 3H), 0.67 (t, J=7.3 Hz, 3H); 19F NMR (376 MHz, methanol-d4, ppm): δ −63.83 (3F). 221: LCMS (ESI, m/z): [M+H]+=525.3. 1H NMR (400 MHz, methanol-d4, ppm): δ 7.97 (d, J=8.7 Hz, 1H), 7.81 (d, J=8.7 Hz, 1H), 7.66 (d, J=8.1 Hz, 2H), 7.57 (d, J=8.1 Hz, 2H), 4.84-4.79 (m, 1H), 3.82-3.74 (m, 1H), 3.68 (s, 3H), 3.29-3.24 (m, 1H), 3.09-2.96 (m, 3H), 2.94-2.86 (m, 4H), 2.66-2.57 (m, 1H), 2.52-2.45 (m, 1H), 2.13-1.93 (m, 2H), 1.72-1.63 (m, 2H), 0.92 (t, J=7.4 Hz, 3H), 0.71 (t, J=7.3 Hz, 3H); 19F NMR (376 MHz, methanol-d4, ppm): δ −63.86 (3F).
Step 1: A mixture of 67-5 (50 g, 196.08 mmol), aniline (50 g, 537.63 mmol), and trimethyl orthoformate (56.3 g, 530.53 mmol.) in ethyleneglycol (400 mL) was heated to 70° C. under stirring. The temperature was increased to 170° C., then kept at 170° C. for 2 h. After cooling the solid was collected with filtration and washed with EtOH, dried at 55˜60° C. to afford 227-1.
Step 2: To a solution of 227-1 (63 g, 175.88 mmol) in anhydrous DMF (400 mL) was slowly added POCl3 (42 g, 273.94 mmol) and the mixture was stirred at room temperature for 20 h. The mixture was poured into ice water (3.2 L), and the precipitate was collected with filtration and dried at 55-60° C. to afford 227-2.
Step 3: To a solution of 67-18 (150 mg, 0.45 mmol) in DCM (10 mL) were added 1H-imidazole (494.5 mg, 7.26 mmol), DMAP (5.5 mg, 0.045 mmol) and TBSCl (136.9 mg, 0.91 mmol). The reaction mixture was stirred at 25° C. for 4 h under N2. The reaction was partitioned between DCM and H2O. The separated organic layer was washed with brine and dried over anhydrous Na2SO4. Evaporation of the solvent under reduced pressure afford 227-3.
Step 4: The mixture of 227-2 (140 mg, 0.46 mmol), 227-3 (206.5 mg, crude), and Na2CO3 (98.4 mg, 0.93 mmol) in THF (4 mL) was stirred at 75° C. for 3 h. The solvent was removed under reduced pressure, the residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 1/1) to afford 227-4.
Step 5: To a solution of 227-4 (200 mg, 0.28 mmol) and TMSOTf (62.6 mg, 0.28 mmol) in DCM (25 mL) was added Et3SiH (65.5 mg, 0.56 mmol) at −78. The mixture was stirred at 25
for 3 h. The reaction was quenched by addition of H2O, and the aqueous layer was extracted with DCM. The organic extracts were combined, washed with brine, dried over anhydrous Na2SO4 and concentrated to afford 227-5.
Step 6: To a solution of 227-5 (250 mg, 0.43 mmol) in DMF (5 mL) were added Zn(CN)2 (152.0 mg, 1.29 mmol), Zn (8.5 mg, 0.13 mmol) and palladium(0)bis[tris(2-methylprop-2-yl)phosphane](22.0 mg, 0.043 mmol). The reaction mixture was stirred at 85° C. for 3 h under N2. The reaction was concentrated to give a residue. The residue was purified by C18 reverse phase column (MeCN/H2O (0.05% NH3·H2O), 5-95%) to afford 227 (53 mg). LCMS (ESI, m/z): [M+H]+=526.2. 1H NMR (400 MHz, DMSO-d6, ppm): δ 8.11-8.00 (m, 2H), 7.73 (d, J=8.1 Hz, 2H), 7.48 (d, J=8.1 Hz, 2H), 4.81-4.72 (m, 1H), 4.68-4.55 (m, 2H), 4.43-4.32 (m, 1H), 4.20-4.10 (m, 1H), 4.05-3.94 (m, 1H), 3.87-3.78 (m, 1H), 3.53 (s, 3H), 2.98-2.84 (m, 2H), 2.44-2.35 (m, 1H), 2.13-1.98 (m, 2H), 1.97-1.85 (m, 1H), 1.77-1.64 (m, 1H), 1.58-1.40 (m, 1H), 0.96 (t, J=7.2 Hz, 3H), 0.87 (t, J=7.2 Hz, 3H); 19F NMR (376 MHz, DMSO-d6, ppm): δ −60.79 (3F).
Step 1: To a solution of 235-1 (5 g, 24.49 mmol) in CCl4 (50 mL) was added NBS (4.8 g, 26.94 mmol) and AIBN (0.4 g, 2.45 mmol). The reaction mixture was stirred at room temperature for 2 h. The reaction was diluted with ethyl acetate and water. The organic layer was separated, washed with saturated NaCl and concentrated in vacuo. The residue was purified by C18 column and was eluted with (Acetonitrile in water (0.1% FA), 0-70%, keep in 50% 25 min) to afford 235-2.
Step 2: To a solution of 235-2 (500 mg, 1.77 mmol) in MeOH (10 mL) was added H2SO4 (0.1 mL). This reaction mixture stirred at 85° C. for 18 h. The reaction mixture was diluted with ethyl acetate and saturated NaHCO3. The organic layer was separated, washed with saturated NaCl and concentrated in vacuo. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 1/2) to afford 235-3.
Step 3: Compound 235-4 was prepared from compound 235-3 and 123-3 following the procedure for the synthesis of compound 28-15 in example 11.
Step 4: To a solution of 235-4 (300 mg, 0.55 mmol) in THF (3 mL) and water (0.5 mL) was added LiOH (232.5 mg, 5.54 mmol) The reaction mixture was stirred at 0° C. at N2 overnight. The crude product was purified by reverse HPLC (0.1% TFA in water/MeCN, 0-70%) to afford 235-5.
Step 5: 235-5 (37 mg) was purified by SFC (column: ChiralCel OD, 250×30 mm I.D., 10 μm, Methanol/Supercritical CO2=50/50) to afford 235-6 (15 mg) and 235-7 (8 mg). 235-6: SFC analysis: 100% ee; retention time: 6.129 min; column: ChiralCel OD, 100×4.6 mm I.D., 3 μm, 40% of Ethanol (0.05% DEA) in CO2; pressure: 100 bar; flow rate: 2.5 mL/min. 235-7: SFC analysis: 100% ee; retention time: 8.821 min; column: ChiralCel OD, 100×4.6 mm I.D., 3 μm, 40% of Ethanol (0.05% DEA) in CO2; pressure: 100 bar; flow rate: 2.5 mL/min.
Step 6: To a solution of 235-6 (20 mg, 0.038 mmol) in THF (2 mL) was added CDI (15.4 mg, 0.095 mmol), the reaction mixture stirred at 60° C. for 18 h. NaBH4 (3.6 mg, 0.095 mmol) was added in and stirred at room temperature for 2 h. This reaction mixture was diluted with ethyl acetate and saturated NaCl. The organic layer was separated, washed with saturated NaCl and concentrated in vacuo. The residue was purified by reverse HPLC (MeCN in water (0.1% NH4HCO3), keep in 50%) to afford 235 (3 mg). LCMS (ESI, m/z): [M+H]+=514.2. 1H NMR (400 MHz, methanol-d4, ppm) δ 8.05 (d, J=8.8 Hz, 1H), 7.91 (d, J=8.8 Hz, 1H), 7.67-7.52 (m, 4H), 5.46 (d, J=15.2 Hz, 1H), 4.17 (d, J=11.0 Hz, 1H), 3.93-3.81 (m, 4H), 3.71 (s, 3H), 3.57-3.46 (m, 1H), 3.29-3.19 (m, 1H), 3.15-3.05 (m, 1H), 2.59-2.48 (m, 2H), 1.95-1.82 (m, 1H), 1.75-1.63 (m, 1H), 1.10 (t, J=7.4 Hz, 3H). 19F NMR (376 MHz, methanol-d4, ppm) 8-63.88 (3F).
Step 1: To a solution of 241-1 (10 g, 49.98 mmol) in DCM (100 mL) was added acetic anhydride (5.63 mL, 59.97 mmol) and triethylamine (13.89 mL, 99.96 mmol), then the reaction mixture was stirred at room temperature for 2 h. The reaction was quenched with H2O. The mixture was extracted with ethyl acetate. Combined the organic layer and washed with NaHCO3. The organic layer was dried over Na2SO4, filtered and concentrated. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 1/1) to afford 241-2.
Step 2: To a stirred solution of 241-2 (9.4 g, 38.82 mmol) in DCM (100 mL) was added oxalyl Chloride (3.94 mL, 46.59 mmol). The mixture was warmed up to room temperature and stirred for 1 h, then FeCl3 (12.6 g, 77.65 mmol) was added, and stirred at room temperature for 24 h. The reaction was quenched with 1M HCl. The mixture was extracted with DCM. The organic layers were combined and dried over Na2SO4, filtered and concentrated to afford 241-3 which was used for the next step directly.
Step 3: To a solution of 241-3 (10 g, crude) in MeOH (100 mL) was added H2SO4 (25 mL). The reaction mixture was stirred at 65° C. for 4 h. After cooling to room temperature, the reaction mixture was quenched with NH3·H2O. The mixture was extracted with DCM. The organic layers were dried over Na2SO4, filtered and concentrated to afford 241-4 which was used for the next step directly.
Step 4: To a solution of 241-4 (5.5 g, crude) in EtOH (20 mL) was in portions added NaBH4 (0.84 g, 22.10 mmol). After stirred at 0 for 2 h, HCl (15 mL, 5M) was added and the mixture was stirred at room temperature for 16 h. The solvent was removed and EtOH was added to the residue, followed by NH3H2O at 0° C. to quench the reaction. The mixture was extracted with DCM. The organic layers were dried over Na2SO4, filtered and concentrated to afford 241-5 which was used for the next step directly.
Step 5: To a stirred solution of 241-5 (5 g, crude) in DCM (10 mL) were added TEA (9.22 mL, 66.33 mmol) and (BOC)2O (4.32 mL, 18.8 mmol) at 0° C. the reaction mixture was stirred at room temperature for 16 h. The reaction was quenched with H2O, extracted with ethyl acetate. The organic layer was dried over Na2SO4, filtered and concentrated. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 4/1) to afford 241-6.
Step 6: To a solution of 241-6 (2 g, 6.13 mmol) in DMF (4 mL) and H2O (1 mL) were added K2CO3 (3.4 g, 24.52 mmol) and Pd(PPh3)2Cl2 (0.5 g, 0.61 mmol). Then the reaction mixture was stirred at 80° C. for 24 h. After cooling to room temperature, the reaction was quenched with H2O. The mixture was extracted with DCM. The organic layer was dried over Na2SO4, filtered and concentrated. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 2/1) to afford 241-7.
Step 7: To a solution of 241-7 (2.6 g, 6.64 mmol) in DCM (21 mL) was added TFA (7 mL). Then the reaction mixture was stirred at room temperature for 2 h. The mixture was concentrated to afford 241-8 which was used for the next step directly.
Step 8: Compound 241-9 was prepared from compound 241-8 following the procedure for the synthesis of compound 5 in example 5.
Step 9: 241-9 (100 mg) was purified by SFC (column: ChiralPak AD, 250×30 mm, 10 μm, Ethanol/Supercritical CO2=35/65) to afford 241 (25 mg) and 242 (28 mg) respectively. 241: SFC analysis: 100% ee; retention time: 1.637 min; column: Chiralpak AD 150×4.6 mm I.D., 3 μm, 40% EtOH (0.05% DEA) in CO2; pressure: 100 bar; flow rate: 2.5 mL/min. LCMS (ESI, m/z): [M+H]+=476.4. 1H NMR (400 MHz, methanol-d4, ppm): δ 8.13-8.03 (m, 1H), 8.01-7.90 (m, 1H), 7.80-7.70 (m, 2H), 7.59-7.49 (m, 2H), 7.42-7.20 (m, 2H), 7.15-7.05 (m, 1H), 6.78-6.65 (m, 0.5H), 6.21-6.11 (m, 0.5H), 5.61-5.50 (m, 0.5H), 4.86-4.75 (m, 0.5H), 3.82-3.71 (m, 0.5H), 3.58 (s, 3H), 3.51-3.40 (m, 0.5H), 3.22-3.02 (m, 1H), 2.78-2.55 (m, 1H), 1.91-1.60 (m, 3H); 19F NMR (376 MHz, methanol-d4, ppm): δ −60.87 (3F). 242: SFC analysis: 99.56% ee; retention time: 2.105 min; Column: Chiralpak AD 150×4.6 mm I.D., 3 μm, 40% EtOH (0.05% DEA) in CO2; pressure: 100 bar; flow rate: 2.5 mL/min. LCMS (ESI, m/z): [M+H]+=476.4. 1H NMR (400 MHz, methanol-d4, ppm): δ 8.13-8.03 (m, 1H), 8.01-7.90 (m, 1H), 7.79-7.69 (m, 2H), 7.53-7.42 (m, 2H), 7.45-7.20 (m, 2H), 7.15-7.05 (m, 1H1), 6.76-6.65 (m, 0.5H1), 6.21-6.09 (m, 0.5H), 5.60-5.50 (m, 0.5H), 4.86-4.76 (m, 0.5H), 3.82-3.75 (m, 0.5H), 3.58 (s, 3H), 3.50-3.39 (m, 0.5H), 3.22-3.02 (m, 1H), 2.78-2.54 (m, 1H), 1.93-1.60 (m, 3H); 19F NMR (376 MHz, methanol-d4, ppm): δ −60.87 (3F).
Step 1: Compound 252-1 was prepared from compound 67-3 following the procedure for the synthesis of compound 67-7 in example 22.
Step 2: Compound 252-2 was prepared from compound 252-1 following the procedure for the synthesis of compound 67 in example 22.
Step 3: To a stirred solution of 252-2 (5 mg, 0.008 mmol) in TFA (1.5 mL) was stirred at 70° C. under N2 for 18 h. The reaction mixture was concentrated. The residue was purified by C18 reverse phase column (MeCN/H2O(0.05% NH3·H2O), 5-95%) to afford 252 (3 mg). LCMS (ESI, m/z): [M+H]+=498.2. 1H NMR (400 MHz, methanol-d4, ppm): δ 7.91 (d, J=8.5 Hz, 1H), 7.75 (d, J=8.5 Hz, 1H), 7.66 (d, J=8.1 Hz, 2H), 7.56 (d, J=8.1 Hz, 2H), 5.30-5.20 (m, 1H), 4.48-4.35 (m, 1H), 4.12-4.03 (m, 1H), 3.81-3.72 (m, 1H), 3.68-3.57 (m, 1H), 3.19-3.12 (m, 1H), 3.10-3.03 (m, 1H), 2.75-2.65 (m, 1H), 2.55-2.45 (m, 1H), 2.11-2.02 (m, 1H), 1.79-1.60 (m, 3H), 0.81 (t, J=7.4 Hz, 3H), 0.70 (t, J=7.3 Hz, 3H); 19F NMR (376 MHz, methanol-d4, ppm): δ −63.87 (3F).
Step 1: To a solution of 261-1 (2 g, 9.38 mmol) in EtOH (20 mL) was added TMSCN (1.76 mL, 14.07 mmol) and (NH4)2CO3 (4.5 g, 46.89 mmol) at room temperature. Then the mixture was stirred at 60° C. for 6 h. This mixture was concentrated under vacuo. The residue was purified by column chromatography on silica gel (DCM/MeOH=1/0 to 6/1) to afford 261-2.
Step 2: To a solution of 261-2 (1.7 g, 6.0 mmol) in THF (30 mL) was added Boc2O (5.51 mL, 24.0 mmol), DMAP (0.1 g, 1.20 mmol) and triethylamine (2.50 mL, 18.0 mmol) at room temperature. Then the mixture was stirred at room temperature for 2h. This mixture was washed with H2O, extracted with ethyl acetate, dried and concentrated to afford 261-3 which was used for the next step directly.
Step 3: To a solution of 261-3 (2.8 g, crude) in THF (30 mL) and H2O (10 mL) was added KOH (9.7 g, 172.88 mmol). Then the mixture was stirred at room temperature for 3 h. The mixture was concentrated under vacuo. The residue was purified by reverse-HPLC (57% MeCN in H2O) to afford 261-4.
Step 4: A solution of LiAlH4 (161.6 mg, 4.26 mmol) in THF (15 mL) was stirred at 0° C. under N2 for 0.5 h. Then 261-4 (550 mg, 2.13 mmol) was added and stirred at 25° C. for 16 h. The mixture was diluted with THF, quenched with saturated Na2SO4, filtered and the filtrate was concentrated to afford 261-5 which was used for the next step directly.
Step 5: Compound 261 and 262 was prepared from compound 261-5 following the procedure for the synthesis of compound 36 in example 15. The product was purified by reverse-HPLC (80% ACN in H2O) and SFC (Column: ChiralPak IG, 250×30 mm I.D., 10 μm; 30% Ethanol in CO2, Flow rate: 150 mL/min, Back pressure: 100 bar) to afford 261 (8 mg) and 262 (8 mg). 261: SFC analysis: 100% ee; retention time: 2.666 min; column: ChiralPak IG, 100×4.6 mm I.D., 3 μm, 40% EtOH (0.05% DEA) in CO2, pressure: 100 bar; flow rate: 2.5 mL/min. LCMS (ESI, m/z): [M+H]+=499.2; 1H NMR (400 MHz, methanol-d4, ppm) δ 8.06 (d, J=8.8 Hz, 1H), 7.99 (d, J=8.7 Hz, 3H), 7.35 (d, J=8.2 Hz, 2H), 4.66-4.40 (m, 2H), 4.36-4.23 (m, 2H), 4.23-3.93 (m, 2H), 3.59 (s, 3H), 2.38-2.12 (m, 3H), 2.07-1.80 (m, 3H). 19F NMR (376 MHz, methanol-d4, ppm) δ −59.37 (3F). 262: SFC analysis: 98.98% ee; retention time: 3.100 min; column: ChiralPak IG, 100×4.6 mm I.D., 3 μm, 40% EtOH (0.05% DEA) in CO2, pressure: 100 bar; flow rate: 2.5 mL/min. LCMS (ESI, m/z): [M+H]+=499.2; 1H NMR (400 MHz, methanol-d4, ppm) δ 8.11-8.03 (m, 1H), 7.99 (d, J=8.7 Hz, 3H), 7.35 (d, J=8.2 Hz, 2H), 4.64-4.39 (m, 2H), 4.35-4.24 (m, 2H), 4.24-3.92 (m, 2H), 3.59 (s, 3H), 2.39-2.11 (m, 3H), 2.07-1.79 (m, 3H). 19F NMR (376 MHz, methanol-d4, ppm) δ −59.37 (3F).
Step 1: A solution of 264-1 (3 g, 17.09 mmol) and cyclopropanamine (7.5 mL, 108.23 mmol) in dioxane (9 mL) was stirred at 85 for 10 h in a sealed tube. The mixture was concentrated. The residue was dissolved in ethyl acetate and H2O. The mixture was adjusted pH to 5 by 2M HCl, the mixture was extracted with ethyl acetate. The organic phases were dried over Na2SO4, filtered and concentrated to afford 264-2.
Step 2: To a solution of 264-2 (3.62 g, crude) in EtOH (30 mL) was added SOCl2 (6.18 mL, 85.12 mmol) at 0. The mixture was stirred at 20
for 12 h. The mixture was concentrated. The residue was dissolved in ethyl acetate and H2O, the mixture was extracted with ethyl acetate. The organic phase was washed with H2O, dried over Na2SO4, filtered and concentrated. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 4/1) to afford 264-3.
Step 3: To a solution of 264-3 (1.38 g, 5.73 mmol) in DCE (10 mL) was added acetyl chloride (1.63 mL, 22.93 mmol) at 0. The mixture was stirred at 60
for 4 h. The mixture was quenched with H2O, extracted with ethyl acetate. The combined organic phases were washed with H2O and concentrated. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 1/20) to afford 264-4.
Step 4: Compound 264-5 was prepared from compound 264-4 following the procedure for the synthesis of compound 67-7 in example 22.
Step 5: Compound 264 (29 mg) was prepared from compound 264-5 following the procedure for the synthesis of compound 67 in example 22. LCMS: m/z 538.2 [M+H]f. 1H NMR (400 MHz, DMSO-d6, ppm) δ 8.28 (d, J=8.6 Hz, 1H), 8.03 (d, J=8.6 Hz, 1H), 7.72 (d, J=8.0 Hz, 2H), 7.56 (d, J=8.0 Hz, 2H), 4.74-4.62 (m, 1H), 4.42-4.34 (m, 1H), 4.00-3.90 (m, 1H), 3.81-3.73 (m, 1H), 3.41-3.36 (m, 1H), 3.06-2.87 (m, 3H), 2.63-2.54 (m, 1H), 2.40-2.32 (m, 1H), 2.04-1.91 (m, 1H), 1.76-1.66 (m, 1H), 1.65-1.51 (m, 2H), 1.32-1.17 (m, 2H), 0.85-0.74 (m, 4H), 0.72-0.60 (m, 4H). 19F NMR (376 MHz, DMSO-d6, ppm) δ −60.73 (3F).
Step 1: To a solution of 4-(trifluoromethyl)benzoic acid (1 g, 5.26 mmol) in DMF (6 mL) were added 67-15 (1.3 g, 5.55 mmol), DIPEA (1.38 mL, 8.32 mmol) and HATU (3.2 g, 8.32 mmol), the reaction was stirred at 0° C. for 3 h. The mixture was diluted with ethyl acetate and water. The organic layer was separated and washed with brine. The organic layer was dried by Na2SO4, filtered and concentrated. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 3/3) to afford 272-1.
Step 2: To a solution of 272-1 (1.2 g, 2.95 mmol) in DMF (5 mL) were added imidazole (0.3 g, 4.43 mmol) and TBSCl (0.5 g, 3.54 mmol), the mixture was stirred at 0° C. for 3 h. The mixture was diluted with ethyl acetate and water. The organic layer was separated and washed with brine. The organic layer was dried by Na2SO4, filtered and concentrated. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 4/1) to afford 272-2.
Step 3: To a solution of 272-2 (450 mg, 0.86 mmol) in THF (4.5 mL) were added IrCl(CO)(PPh3)3 (67.4 mg, 0.086 mmol) at room temperature under N2. Then the reaction mixture was cooled to −78° C., 1,1,3,3-Tetramethyldisiloxane(101.1 mg, 0.130 mmol) was added into the mixture, the mixture was stirred at −78° C. for 1.5 h, then bromo(prop-2-yl)magnesium (2.59 mL, 2.59 mmol) was added into the reaction, the mixture was stirred for another 18 h at room temperature under N2. The reaction was quenched with saturated NH4Cl aqueous and diluted with ethyl acetate and water. The organic layer was separated and washed with brine. The organic layer was dried by Na2SO4, filtered and concentrated. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 4/1) afford 272-3.
Step 4: To a solution of 272-3 (410 mg, 0.75 mmol) in EtOH (10 mL) was added Pd/C (10%, 4.0 mg) under N2. The suspension was degassed with H2 for 4 times, and then the mixture was stirred at 25° C. for 2 h under H2. The mixture was filtered and the filtrate was concentrated. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 1/1) to afford 272-4.
Step 5: Compound 272-5 was prepared from compound 272-4 following the procedure for the synthesis of compound 5 in example 5.
Step 6: To a solution of 272-5 (130 mg, 0.19 mmol) in DMF (5 mL) was added CsF (115.0 mg, 0.76 mmol) and the mixture was stirred at 110° C. for 36 h. The mixture was diluted with ethyl acetate and water. The organic layer was separated and washed with brine. The organic layer was dried by Na2SO4, filtered and concentrated. The residue was purified by column chromatography on silica gel (DCM/MeOH=1/0 to 10/1) to afford 272-6.
Step 7: 272-6 (100 mg) was purified by SFC(column: ChiralPak AD, 250>30 mm I.D., 10 μm, Ethanol/Supercritical CO2=20/80) to afford 272 (48.85 mg) and 273 (5.2 mg) respectively. 272: SFC analysis: 100% ee; retention time: 2.185 min; column: ChiralPak AD, 150×4.6 mm I.D., 3 μm, 5-40% of ethanol (0.05% DEA) in CO2; pressure: 100 bar; flow rate: 2.5 mL/min. LCMS (ESI, m/z): [M+H]+=526.2. 1H NMR (400 MHz, CDCl3, ppm) δ 7.75-7.65 (m, 2H), 7.61-7.51 (m, 2H), 7.48-7.40 (m, 2H1), 5.70-5.58 (m, 1H), 4.22-4.12 (m, 1H), 3.92-3.81 (m, 1H), 3.71 (s, 3H), 3.57-3.41 (m, 2H), 3.12-2.97 (m, 2H), 2.44-2.31 (m, 2H), 2.28-2.17 (m, 1H), 1.93-1.77 (m, 1H), 1.55-1.44 (m, 1H), 1.20-1.08 (m, 3H), 0.85-0.66 (m, 6H). 19F NMR (376 MHz, CDCl3, ppm) δ −62.26 (3F). 273: SFC analysis: 100% ee; retention time: 2.487 min; column: ChiralPak AD, 150×4.6 mm I.D., 3 μm, 5-40% of ethanol (0.05% DEA) in CO2; pressure: 100 bar; flow rate: 2.5 mL/min. LCMS (ESI, m/z): [M+H]+=526.2. 1H NMR (400 MHz, CDCl3, ppm) δ 7.71-7.64 (m, 2H), 7.63-7.54 (m, 2H), 7.40-7.31 (m, 2H), 4.42-4.34 (m, 1H), 4.34-4.25 (m, 1H), 4.17-4.08 (m, 1H), 3.74-3.70 (m, 3H), 3.68-3.63 (m, 1H), 3.57-3.49 (m, 1H), 3.45-3.36 (m, 1H), 2.86-2.75 (m, 2H), 2.54-2.45 (m, 1H), 2.28 (dt, J=12.8, 6.4 Hz, 1H), 1.71-1.64 (m, 2H), 0.92-0.84 (m, 6H), 0.82-0.76 (m, 3H). 19F NMR (376 MHz, CDCl3, ppm) δ −62.41 (3F).
Step 1: To a solution of 278-1 (2 g, 11.39 mmol) and DMF (0.088 mL, 1.14 mmol) in DCM (40 mL) was dropwise added oxalyl dichloride (1.45 mL, 17.09 mmol) at 0° C. and stirred for 12 h at room temperature. The reaction mixture was concentrated to dryness afford 278-2 which was used for next step directly.
Step 2: To a solution of 1H-1,2,4-triazol-5-amine (800 mg, 9.52 mmol) in DMF (30 mL) was added DIPEAe (7.86 mL, 47.57 mmol), then 278-2 (2214.9 mg, 11.42 mmol) in DMF (10 mL) was added to the mixture at 0° C. The resulting mixture was stirred at room temperature for 12 h. The reaction mixture used for the next step directly.
Step 3: K2CO3 (3.8 g, 27.32 mmol) was added to the reaction mixture from step 2, stirred at 100° C. under N2 for 16 h. The reaction was quenched with HCl (2 N) until the pH to 4. The solid was collected with filtration to afford 278-4.
Step 4: Compound 272-5 was prepared from compound 272-4 following the procedure for the synthesis of compound 5 in example 5. The crude product was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 1/4) to afford 278 (3 mg) and 279 (2 mg). 278: LCMS (ESI, m/z): [M+H]+=523.4. 1H NMR (400 MHz, DMSO-d6, ppm)8 8.76-8.67 (m, 1H), 8.53-8.43 (m, 1H), 8.32 (s, 1H), 7.79-7.71 (m, 2H), 7.64-7.55 (m, 2H), 5.60-4.71 (m, 2H), 3.67 (s, 1H), 2.99-2.92 (m, 1H), 2.90-2.83 (m, 1H), 2.31-1.88 (m, 4H), 1.69-1.59 (m, 1H), 1.56-1.37 (m, 3H), 1.05-0.95 (m, 3H), 0.75-0.59 (m, 6H). 19F NMR (376 MHz, DMSO-d6, ppm) δ −60.67 (3F). 279: LCMS (ESI, m/z): [M+H]+=523.4. 1H NMR (400 MHz, DMSO-d6, ppm) 68.78-8.64 (m, 1H), 8.52-8.42 (m, 1H), 8.33 (s, 1H), 7.78-7.69 (m, 2H), 7.65-7.56 (m, 2H), 6.21-4.53 (m, 2H), 3.53 (s, 1H), 2.62-2.58 (m, 1H), 2.26 (s, 1H), 2.07-1.94 (m, 2H), 1.85 (s, 2H), 1.66-1.54 (m, 2H), 1.46 (s, 2H), 1.08-0.99 (m, 3H), 0.67-0.57 (m, 6H). 19F NMR (376 MHz, DMSO-d6, ppm) δ −60.67 (3F) Example 53 Synthesis of Compounds 282 and 283
Step 1: To a solution of 235-5 (153 mg, 0.29 mmol) in THF (5 mL) was added CDI (70.5 mg, 0.44 mmol) and the reaction was stirred at room temperature overnight. NaBH4 (11.0 mg, 0.29 mmol) was added to this mixture and stirred at room temperature for 1 h. The reaction mixture was diluted with ethyl acetate and water. The organic layer was separated, washed with brine and concentrated in vacuo. The residue was by reverse HPLC (MeCN in water (0.1% FA), 0-70%) to afford 282-1.
Step 2: To a solution of 282-1 (72 mg, 0.14 mmol) in THF (3 mL) was added NaH (6.2 mg, 0.15 mmol) at 0° C., the mixture was stirred at 0° C. for 2 h, then CH3I (29.9 mg, 0.21 mmol) was added to the mixture. The reaction mixture was stirred at 30° C. for 2 h. The mixture was diluted with ethyl acetate and saturated NaCl aqueous. The organic layer was separated, washed with brine, and concentrated in vacuo. The residue was purified by column chromatography on silica gel (DCM/ethyl acetate=1/0 to 1/3) to afford 282 (10 mg) and 283 (18 mg). 282: LCMS (ESI, m/z): [M+H]+=528.2. 1H NMR (400 MHz, methanol-d4, ppm) δ 8.05 (d, J=8.8 Hz, 1H), 7.91 (d, J=8.8 Hz, 1H), 7.63 (s, 4H), 5.42-5.31 (m, 1H), 4.19-4.09 (m, 1H), 4.09-3.99 (m, 1H), 3.95-3.85 (m, 1H), 3.76-3.66 (m, 4H), 3.65-3.55 (m, 1H), 3.55-3.45 (m, 2H), 3.28 (s, 3H), 3.09-2.99 (m, 1H), 2.62-2.47 (m, 2H), 1.92-1.80 (m, 1H), 1.75-1.63 (m, 1H), 1.07 (t, J=7.4 Hz, 3H). 19F NMR (376 MHz, methanol-d4, ppm): δ −63.89 (3F). 283: LCMS (ESI, m/z): [M+H]+=528.2. 1H NMR (400 MHz, methanol-d4, ppm) δ 8.04 (d, J=8.8 Hz, 1H), 7.88 (d, J=8.8 Hz, 1H), 7.68-7.57 (m, 4H), 5.27-5.17 (m, 1H), 4.41-4.31 (m, 1H), 4.11-4.00 (m, 2H), 3.78-3.73 (m, 1H), 3.71 (s, 3H), 3.66-3.57 (m, 2H), 3.29 (s, 3H), 3.14-3.00 (m, 2H), 2.83-2.72 (m, 1H), 2.54-2.44 (m, 1H), 1.87-1.72 (m, 1H), 1.69-1.56 (m, 1H), 0.85 (t, J=7.4 Hz, 3H). 19F NMR (376 MHz, methanol-d4, ppm): δ −63.91 (3F).
Step 1: To a solution of 220-1 (10 g, 19.0 mmol) in DMF (100 mL) was added Zn(CN)2 (6.69 g, 57 mmol), Zn (372.6 mg, 5.7 mmol) and palladium(0) bis[tris(2-methylprop-2-yl)phosphane](970.9 mg, 1.90 mmol). The reaction mixture was stirred at 85° C. for 3 h under N2. The reaction mixture was diluted with ethyl acetate, washed with brine and dried over anhydrous Na2SO4. Evaporation of the solvent under reduced pressure, the residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 1/3) to afford 300-1.
Step 2: To a stirred solution of 300-1 (5.5 g, 11.64 mmol) in EtOH (60 mL) and H2O (15 mL) at room temperature was added Fe (3.2 g, 58.20 mmol) and NH4Cl (6.2 g, 116.40 mmol). The reaction mixture was stirred at 80 for 1 h. The hot reaction mixture was filtered and the filter cake was washed with EtOH. The filtrate was concentrated to dryness. The residue was purified by C18 reverse phase HPLC (MeCN/H2O(0.05% TFA), 5-95%, 50 min) to afford 300-2.
Step 3: Compound 300-3 was prepared from compound 300-2 following the procedure for the synthesis of compound 220-4 in example 44.
Step 4: Compound 300-4 was prepared from compound 300-3 following the procedure for the synthesis of compound 28-15 in example 11.
Step 5: 300-4 (167 mg) was purified by SFC (column: ChiralCel OJ, 250×30 mm I.D., 10 μm, A for CO2 and B for Ethanol (0.10% NH3H2O) to afford 300 (25 mg) and 301 (15 mg) respectively. 300: SFC analysis: 100.0% ee; retention time: 3.542 min; column: ChiralCel OJ, 150×4.6 mm I.D., 3 μm, A for CO2 and B for Ethanol (0.05% DEA), 5-40%; pressure: 100 bar; flow rate: 2.5 mL/min. LCMS (ESI, m/z): [M+H]+=511.4; 1H NMR (400 MHz, DMSO-d6, ppm) δ 7.96 (d, J=8.8 Hz, 1H), 7.85 (d, J=8.8 Hz, 1H), 7.71 (d, J=8.0 Hz, 2H), 7.58 (d, J=8.0 Hz, 2H), 6.09-5.99 (m, 1H), 4.49-4.39 (m, 1H), 3.74-3.64 (m, 1H), 3.64 (s, 3H), 3.28-3.18 (m, 1H), 3.04-2.90 (m, 4H), 2.57-2.47 (m, 1H), 2.41-2.31 (m, 1H), 1.95-1.82 (m, 1H), 1.78-1.46 (m, 3H), 1.01 (t, J=7.2 Hz, 3H), 0.66 (t, J=7.2 Hz, 3H). 19F NMR (376 MHz, DMSO-d6, ppm): δ −60.65 (3F). 301: SFC analysis: 98.18% ee; retention time: 3.740 min; column: ChiralCel OJ, 150×4.6 mm I.D., 3 μm, A for CO2 and B for Ethanol (0.05% DEA), 5-40%; pressure: 100 bar; flow rate: 2.5 mL/min. LCMS (ESI, m/z): [M+H]+=511.4; 1H NMR (400 MHz, DMSO-d6, ppm) δ 7.92 (d, J=8.8 Hz, 1H), 7.79 (d, J=8.8 Hz, 1H), 7.73 (d, J=8.0 Hz, 2H), 7.52 (d, J=8.0 Hz, 2H), 6.48-6.38 (m, 1H), 4.01-3.92 (m, 1H), 3.65 (s, 3H), 3.54-3.40 (m, 2H), 3.35-3.25 (m, 2H), 3.01-2.91 (m, 2H), 2.59-2.49 (m, 1H), 2.36-2.26 (m, 1H), 2.10-1.95 (m, 1H), 1.80-1.56 (m, 3H), 0.89-0.71 (m, 6H). 19F NMR (376 MHz, DMSO-d6, ppm): δ −60.76 (3F1).
Step 1: To a solution of 192-4 (3.6 g, 10.76 mmol) in DMF (40 mL) was added 1H-imidazole (1.8 g, 26.91 mmol) and TBSCl (2.4 g, 16.15 mmol), and the mixture was stirred at 0° C. for 3 h. The mixture was diluted with ethyl acetate and water. The organic layer was separated, washed with brine and dried by Na2SO4, filtered and concentrated. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 3/1) to afford 302-1.
Step 2: To a solution of 302-1 (4.06 g, 9.05 mmol) in propan-2-ol (50 mL) was added Pd/C (10%, 1.0 g) at room temperature. Then the mixture was stirred at room temperature under H2 for 12 h. The mixture was filtered and the filtrate was concentrated under vacuo to afford 302-2.
Step 3: A mixture of 302-2 (3.12 g, 8.70 mmol), 192-2 (4.06 g, 13.05 mmol), DIPEA (4.31 mL, 26.10 mmol) and NaI (2.6 g, 17.40 mmol) in MeCN (70 mL) was heated at 80 for 16 h. The mixture was concentrated under vacuo and the residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 1/1) to afford 302-3.
Step 4: To a solution of 302-3 (4.5 g, 7.64 mmol) in THF (50 mL) was added LiAlH4 (0.4 g, 11.46 mmol) at 0. Then the mixture was stirred at 0
for 0.5 h. The reaction was quenched with H2O. The mixture was filtered and the filtrate was concentrated. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 4/1) to afford 302-4 and 302-5.
Step 5: To a solution of 302-4 (1.77 g, 3.24 mmol) in THF (40 mL) was added TBAF (9.71 mL, 9.71 mmol) and the reaction mixture was stirred at 30° C. for 1 h. The mixture was diluted with ethyl acetate and water. The organic layer was separated, washed with brine and dried by Na2SO4, filtered and concentrated. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 3/1) to afford 302-6.
Step 6: To a solution of 302-6 (300 mg, 0.69 mmol) in THF (5 mL) was added NaH (83.2 mg, 2.08 mmol). The mixture was stirred at room temperature for 30 minutes, then the mixture was cooled to 0. A solution of 4-methylbenzenesulfonyl chloride (132.2 mg, 0.69 mmol) in THF (0.5 mL) was slowly added to the mixture. The reaction mixture was stirred at room temperature for 12 h. The resulting mixture was quenched by saturated NH4Cl aqueous. The mixture was concentrated, then ethyl acetate and water were added to the residue. The organic layer was separated and washed with brine, dried by Na2SO4. The organic solvent was removed in vacuo, the residue was purified by reverse HPLC (MeCN/H2O(0.05% TFA), 5-95%) to afford 302-7.
Step 7: Compound 302 (21 mg) was prepared from compound 302-7 following the procedure for the synthesis of compound 30 in example 12. LCMS (EST, m/z): [M+H]+ =499.2; 1H NMR (400 MHz, DMSO-d(, ppm) δ 8.26-8.15 (m, 1H), 8.00-7.90 (m, 1H), 7.85-7.76 (m, 2H), 7.73-7.64 (m, 2H), 5.51-5.26 (m, 1H), 5.04-4.59 (m, 1H), 4.18-3.87 (m, 3H), 3.85 (s, 1H), 3.47-3.41 (m, 1H), 3.40 (s, 3H), 2.89-2.60 (m, 3H), 2.20-1.83 (m, 3H), 1.01-0.79 (m, 3H). 19F NMR (376 MHz, DMSO-d6, ppm) δ −60.92 (3F).
Step 1: To a stirred solution of 325-1 (10 g, 41.11 mmol) in DMF (10 mL) was added Cs2CO3 (6.7 g, 20.55 mmol) and iodomethane (7.0 g, 49.33 mmol) at 0° C. The reaction mixture was stirred at room temperature for 4 h. The solvent was removed under vacuum, the residue was purified by reverse HPLC (MeCN/0.05% FA in water: 5%˜60%) to afford 325-2.
Step 2: To a stirred solution of 325-2 (0.3 g, 1.17 mmol) in MeOH (3 mL) was added NaBH4 (88.22 mg, 2.33 mmol) in portions at 0° C. The mixture was stirred at room temperature for 1 h. The reaction was quenched with NH4Cl solution. The mixture was extracted with EtOAc and washed with H2O. The organic layer was dried over Na2SO4, filtered and concentrated. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 1/1) to afford 325-3.
Step 3: Compound 325-4 was prepared from compound 325-3 following the procedure for the synthesis of compound 2-2 in example 2.
Step 4: To a stirred solution of 325-4 (900 mg, 2.23 mmol) in THF (9 mL) was added dropwise LiAlH4 (4.46 mL, 4.46 mmol) at −78° C. The mixture was stirred at −78° C. for 1 h under N2. The reaction was quenched with H2O. The mixture was extracted with EtOAc and washed with H2O. The organic layer was dried over Na2SO4, filtered and concentrated. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 2/1) to afford 325-5.
Step 5: Compound 325 was prepared from compound 325-5 following the procedure for the synthesis of compound 107 in example 32. LCMS (ESI, m/z): [M+H]+=457.2; 1H NMR (400 MHz, DMSO-d6, ppm): δ 8.15-8.05 (m, 2H), 7.66-7.64 (m, 2H), 7.22-7.20 (m, 2H), 5.20-5.09 (m, 1H), 4.87-4.75 (m, 1H), 4.35-4.24 (m, 1H), 4.11-4.01 (m, 1H), 3.66-3.54 (m, 4H), 3.14-3.02 (m, 1H), 2.30-2.20 (m, 1H), 2.19-2.09 (m, 1H), 1.70-1.50 (m, 2H); 19F NMR (376 MHz, DMSO-d6, ppm): δ −59.78 (3F).
Step 1: To a solution of 330-1 (4 g, 14.04 mmol) in toluene (30 mL) was added n-BuLi (5.89 mL, 14.74 mmol) at −78 under N2, the mixture was stirred at −78
for 0.5 h. propanal
(1.11 mL, 15.45 mmol) was added to the mixture and stirred at −78 C for 0.5 h. Then the mixture was warmed to 20
for 1 h. The reaction was quenched with H2O and extracted with EtOAc. The combined organic layer was washed with H2O and concentrated. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 4/1) to afford 330-2.
Step 2: To a solution of 330-2 (983 mg, 4.53 mmol) in DCM (20 mL) were added PPh3 (1.78 g, 6.79 mmol) and CBr4 (2.25 g, 6.79 mmol) at 0 . The mixture was stirred at 20
for 5 h. The mixture was concentrated. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 6/1) to afford 330-3.
Step 3: Compound 330-4 was prepared from compound 123-4 following the procedure for the synthesis of compound 28-15 in example 11.
Step 4: A solution of 330-4 (90 mg, 0.17 mmol), methyl methyl 2,2-difluoro-2-(fluorosulfonyl)acetate (164.85 mg, 0.86 mmol) and CuI (6.5 mg, 0.034 mmol) in DMF (3 mL) was stirred at 100 for 16 h in a sealed tube. The reaction mixture was concentrated in vacuo. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 0/1) to afford 330 and 331. 330: LCMS (ESI, m/z): [M+H]+=514.2. 1H NMR (400 MHz, CDCl3, ppm): δ 8.93 (s, 2H), 7.66-7.59 (m, 2H), 5.05-4.95 (m, 1H), 4.30-4.12 (m, 1H), 4.08-3.90 (m, 1H), 3.92-3.80 (m, 1H), 3.64 (s, 3H), 3.55-3.10 (m, 3H), 2.85-2.70 (m, 1H), 2.60-2.50 (m, 1H), 2.10-1.90 (m, 2H), 1.85-1.65 (m, 1H), 1.50-1.35 (m, 1H), 1.10-0.95 (m, 3H), 0.70-0.62 (m, 3H). 19F NMR (376 MHz, CDCl3, ppm): δ −62.30 (3F). 331: LCMS (ESI, m/z): [M+H]+=514.2. 1H NMR (400 MHz, CDCl3, ppm): δ 8.91 (s, 2H), 7.63-7.58 (m, 2H), 4.39-4.18 (m, 2H), 4.20-4.16 (m, 1H), 3.86-3.75 (m, 1H), 3.66 (s, 3H), 3.45-3.35 (m, 1H), 3.33-3.12 (m, 1H), 3.14-3.02 (m, 1H), 2.99-2.85 (m, 1H), 2.59-2.52 (m, 1H), 2.00-1.72 (m, 2H), 1.72-1.66 (m, 2H), 0.90-0.70 (m, 6H). 19F NMR (376 MHz, CDCl3, ppm): δ −62.34 (3F).
Step 1: To a solution of ethyl 3-chloro-3-oxopropanoate (7.37 g, 48.96 mmol) and 1-5 (10 g, 40.80 mmol) in DCM (100 mL) was added DIPEA (20.23 mL, 122.41 mmol) at 0° C. The mixture was stirred at room temperature for 16 h. Then the solvent was removed and the residue was dissolved in EtOH (100 mL), followed by the addition of sodium ethoxide (11.1 g, 163.21 mmol). The resulting mixture was stirred at room temperature for 4 h. The reaction was quenched by the addition of water. The reaction mixture was acidified to pH=4 by the addition of HCl (1N). The resulting mixture was filtered, the filter cake was dried to afford 332-1.
Step 2: To a solution of 332-1 (5 g, 15.97 mmol) in DMF (50 mL) was added Sodium hydride (1.6 g, 39.92 mmol) at 0° C. The mixture was stirred at room temperature for 1 h. CH3I (2.3 g, 15.97 mmol) was added to the mixture and the resulting mixture was stirred at room temperature for 16 h. The reaction was quenched by the addition of water. The reaction mixture was acidified to pH=3 by the addition of HCl (1N). The mixture was extracted with EtOAc and the organic layers were combined, dried over Na2SO4, filtered and concentrated under vacuum. The residue was purified by reverse HPLC (0-100% of MeCN in water) to afford 332-2.
Step 3: To an ice-cold solution of 1H-imidazole (1.2 g, 17.94 mmol) and Ph3P (4.7 g, 17.94 mmol) in DCM (30 mL) was added I2 (4.6 g, 17.94 mmol). The mixture was stirred at room temperature for 15 minutes, then 67-14 (3 g, 8.97 mmol) was added to the mixture. The resulting mixture was stirred at room temperature for 2 h. The reaction was quenched with water and extracted with DCM, the combined organic layer was dried and concentrated. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 1/1) to afford 332-3.
Step 4: To a solution of 332-3 (2.8 g, 6.30 mmol) and TMSCN (0.8 g, 7.56 mmol) in THF (50 mL) was added TBAF (7.56 mL, 7.56 mmol). The resulting mixture was stirred at room temperature for 16 h. The reaction was quenched with water and extracted with DCM. The combined organic layer was dried and concentrated, the residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 1/1) to afford 332-4.
Step 5: A mixture of 332-4 (1.7 g, 4.95 mmol) in HCl (4M in MeOH, 10.92 mL) was stirred at 70° C. for 16 h. The solvent was removed under vacuum to afford 332-5.
Step 6: Compound 332-6 was prepared from compound 332-5 following the procedure for the synthesis of compound 67-18 in example 22.
Step 7: To a solution of N-Phenyl-bis(trifluoromethanesulfonimide) (1.9 g, 5.37 mmol) and 332-2 (1.58 g, 4.83 mmol) in MeCN (10 mL) was added K2CO3 (3.7 g, 26.85 mmol). The mixture was stirred at room temperature for 4 h, then 332-6 (1 g, 2.69 mmol) was added to the mixture. The resulting mixture was stirred at room temperature for another 16 h. The reaction mixture was diluted with water and extracted with EtOAc. The combined organic layer was dried and concentrated, the residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 1/1) to afford 332-7.
Step 8: To a solution of Sodium ethoxide (249.6 mg, 3.67 mmol) in EtOH (10 mL) was added 332-7 (500 mg, 0.73 mmol). The resulting mixture was stirred at 50° C. for 4 h. The reaction was quenched with water and extracted with EtOAc. The combined organic layer was dried and concentrated to afford 332-8 which was used for the next step directly without further purification.
Step 9: To a solution of 332-8 (500 mg, crude) in MeOH (20 mL) was added HCl (6 N, 20 mL). The resulting mixture was stirred at 80° C. for 16 h. The reaction was quenched by the addition of water. The reaction mixture was adjusted the pH to 10 by the addition of NaOH (4 N). The mixture was extracted with EtOAc, the organic layers were combined, dried and concentrated to afford 332-9 which was used for the next step directly without further purification.
Step 10: To an ice-cold solution of 332-9 (350 mg, crude) in MeOH (20 mL) was added NaBH4 (27.5 mg, 0.73 mmol). The resulting mixture was stirred at 0° C. for 1h. The mixture was diluted with water and extracted with EtOAc. The combined organic layer was dried and concentrated, the residue was purified by preparative TLC (MeOH/DCM=20:1) to afford 332-10.
Step 11: To solution of 332-10 (100 mg, 0.17 mmol) in triethylsilane (5 mL) and 1,2-dichloroethane (5 mL) was added TFA (196.8 mg, 1.73 mmol) at room temperature. The mixture was stirred at room temperature for 2 h. The mixture was diluted with DCM, then the solvent was removed under vacuum to afford 332-11 which was used for the next step directly without further purification.
Step 12: A mixture of 332-11 (100 mg, crude), Bis(tri-tert-butylphosphine)palladium (18.1 mg, 0.035 mmol), zinc (34.8 mg, 0.53 mmol) and Zn(CN)2 (125.0 mg, 1.07 mmol) in DMF (0.5 mL) was stirred at 100° C. for 2 h. The mixture was diluted with water and extracted with EtOAc. The combined organic layer was dried and concentrated. The residue was purified by preparative TLC (100% of ethyl acetate) and reverse HPLC (MeCN/H2O (0.05% NH4HCO3), 0-100%) to afford 332 and 333. 332: LCMS (ESI, m/z): [M+H]+=510.2; 1H NMR (400 MHz, CDCl3, ppm): δ 7.72-7.63 (m, 2H), 7.59-7.57 (m, 2H), 7.44-7.42 (m, 2H), 5.09-5.05 (m, 1H), 3.67-3.63 (m, 4H), 3.37-3.25 (m, 1H), 3.01-2.98 (m, 2H), 2.88-2.83 (m, 1H), 2.69-2.59 (m, 2H), 2.45-2.43 (m, 1H), 2.01-2.94 (m, 2H), 1.79-1.73 (m, 2H), 1.56-1.50 (m, 2H), 0.85-0.81 (m, 3H), 0.68-0.64 (m, 3H). 19F NMR (376 MHz, CDCl3, ppm): δ −62.30 (3F). 333: LCMS (ESI, m/z): [M+H]+=510.2; 1H NMR (400 MHz, CD3OD, ppm): δ 8.06-8.04 (m, 1H), 7.96-7.94 (m, 1H), 7.64-7.57 (m, 4H), 5.49-5.47 (m, 1H), 3.66-3.58 (m, 4H), 3.21-3.12 (m, 3H), 2.72-2.67 (m, 1H), 2.55-2.46 (m, 3H), 2.05-1.95 (m, 2H), 1.78-1.75 (m, 1H), 1.64-1.54 (m, 3H), 1.20-1.15 (m, 3H), 0.66-0.63 (m, 3H). 19F NMR (376 MHz, CD3OD, ppm): δ −63.84 (3F).
Step 1: To a mixture of 252 (140 mg, 0.28 mmol) in toluene (5 mL) was added DIPEA (145.5 mg, 1.13 mmol) and POCl3 (172.6 mg, 1.13 mmol). Then the mixture was stirred at 1 10° C. for 3 h. The mixture was concentrated. The residue was purified by reverse HPLC (MeCN/H2O (0.05% NH3·H2O), 5-95%) to afford 334-1.
Step 2: To a stirred solution of 334-1 (80 mg, 0.16 mmol) in THF (5 mL) was added hydrazine hydrate (0.009 mL, 0.16 mmol). The reaction mixture was stirred at 50° C. for 1.5 h. The mixture was diluted with EtOAc and washed with water, brine and dried over anhydrous Na2SO4, filtered and concentrated to afford 334-2 which was used for the next step directly without further purification.
Step 3: To a solution of 334-2 (70 mg, 0.14 mmol) in trimethoxymethane (5 mL) was stirred at 100° C. for 18 h. The mixture was concentrated in vacuo, the residue was purified by reverse HPLC (MeCN/H2O(0.05% TFA), 5-80%) to afford 334 as 1.5 TFA salt. LCMS (ESI, m/z): [M+H]+=522.2. 1H NMR (400 MHz, DMSO-d6, ppm) δ 9.80 (s, 1H), 8.90-8.88 (m, 1H), 8.13-8.11 (m, 1H), 8.10-7.88 (m, 2H), 7.81-7.71 (m, 2H), 6.05-5.15 (m, 1H), 4.80-4.55 (m, 2H), 4.40-4.20 (m, 1H), 4.15-3.75 (m, 2H), 3.65-3.32 (m, 2H), 3.23-3.03 (m, 1H), 2.67-2.44 (m, 1H), 2.35-2.15 (m, 1H), 2.15-1.79 (m, 2H), 1.15-0.85 (m, 3H), 0.78 (m, 3H). 19F NMR (376 MHz, DMSO-d6, ppm): δ −64.39 (3F), −77.23 (4.5F).
Step 1: To a solution of 335-1 (10 g, 83.26 mmol) in DCM (40 mL) was added NBS (17.8 g, 99.91 mmol) at 0. The mixture was stirred at 20
for 4.5 h. The mixture was diluted with H2O and extracted with DCM. The combined organic phases were washed with H2O, dried over Na2SO4, filtered and concentrated to afford 335-2 which was used for the next step directly without further purification.
Step 2: A solution of 335-2 (15.87 g, 47.85 mmol), (bromomethyl)benzene (6.82 mL, 57.42 mmol) and K2CO3 (9.9 g, 71.77 mmol) in CH3CN (80 mL) was stirred at 75 for 2 h. The mixture was concentrated. The residue was diluted with H2O and extracted with EtOAc. The combined organic phases were washed with H2O and concentrated. The solid was filtered and the cake was washed with petroleum ether, dried under vacuum to afford 335-3.
Step 3: To a solution of 335-3 (4.72 g, 16.33 mmol) in THF (30 mL) was added Methylmagnesium bromide (16.33 mL, 48.98 mmol) at 0. The mixture was stirred at 20
for 3 h. Then the mixture was added to H2O (60 mL) and stirred at 0
for 0.5 h. The mixture was diluted with H2O and extracted with EtOAc. The combined organic phases were washed with H2O and concentrated. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 3/1) to afford 335-4.
Step 4: 335-4 (3.75 g, 12.25 mmol) in TFA (10 mL) was stirred at 80 for 2 h. The mixture was concentrated and the residue was diluted with NaHCO3 solution, extracted with EtOAc. The combined organic phases were was washed with H2O and concentrated. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 4/1) to afford 335-5.
Step 5: To a solution of 335-5 (2.61 g, 12.08 mmol) in DMF (20 mL) was added NaH (2.4 g, 60.41 mmol) at 0. The mixture was stirred at 0
for 0.5 h. Then ethyl ethoxymethanoate (2.20 mL, 18.12 mmol) was added and the mixture was stirred at 80
for 0.5 h. The reaction was quenched with H2O, the mixture was adjusted the pH to 4 by HCl (2N) and extracted with EtOAc. The combined organic phases were washed with H2O and concentrated. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 4/1) to afford 335-6.
Step 6: A solution of 335-6 (1.56 g, 5.42 mmol) and p-Toluenesulfonic acid (0.7 g, 4.33 mmol) in toluene (12 mL) was stirred at 90C for 10 h. The mixture was concentrated. The residue was purified by column chromatography on silica gel (DCM/MeOH=1/0 to 10/1) to afford 335-7.
Step 7: To a solution of 335-7 (400 mg, 1.65 mmol) in toluene (10 mL) was added DIPEA (1.37 mL, 8.26 mmol) and POCl3 (0.46 mL, 4.96 mmol) at 20. The mixture was stirred at 90
for 2 h. The mixture was concentrated. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 1/4) to afford 335-8.
Step 8: Compound 335-9 was prepared from compound 335-8 following the procedure for the synthesis of compound 67-20 in example 22.
Step 9: To a solution of 335-9 (127 mg, 0.25 mmol) in DCM (10 mL) was added Br2 (0.25 mL, 0.25 mmol) at 0. The mixture was stirred at 0
for 40 minutes. Then triethylamine (0.053 mL, 0.38 mmol) was added and the mixture was stirred at 20
for 0.5 h. The mixture was concentrated. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 1/1) to afford 335-10.
Step 10: A solution of 335-10 (77 mg, 0.13 mmol) and Cs2CO3 (86.6 mg, 0.27 mmol) in DMF (6 mL) was stirred at 90 for 1.5 h. The mixture was diluted with H2O and extracted with EtOAc. The organic phases were concentrated. The residue was purified by reverse HPLC (MeCN/water (0.05% FA): 5%-60%) to afford 335 as 0.5 FA salt. LCMS (ESI, m/z): [M+H]+=499.2; 1H NMR (400 MHz, DMSO-do, ppm): δ 8.46 (s, 0.5H), 8.37 (d, J=8.72 Hz, 1H), 8.20 (d, J=8.72 Hz, 1H), 7.78-7.68 (m, 2H), 7.62-7.53 (m, 2H), 5.20-5.10 (m, 1H), 4.63-4.52 (m, 1H), 4.46-4.34 (m, 1H), 3.80-3.72 (m, 11H), 3.70-3.60 (m, 1H), 3.19-3.13 (m, 1H), 3.11-3.04 (m, 1H), 2.65-2.55 (m, 1H), 2.49-2.44 (m, 1H), 2.06-1.90 (m, 1H), 1.71-1.46 (m, 3H), 0.84-0.71 (m, 3H), 0.67-0.57 (m, 3H). 19F NMR (376 MHz, DMSO-d6, ppm): δ −60.72 (3F).
Step 1: To a solution of 272-2 (1.1 g, 2.11 mmol) in MeOH (15 mL) was added Pd/C (10%, 22.5 mg) under N2. The suspension was degassed under vacuum and purged with H2 for 4 times, then the mixture was stirred at 25° C. for 3 h under H2. The mixture was filtered and concentrated, the residue was purified by column chromatography on silica gel (DCM/MeOH=1/0 to 5/1) to afford 346-1.
Step 2: To a solution of 346-1 (900 mg, 2.09 mmol) in DCM (15 mL) was added triethylamine (0.29 mL, 2.09 mmol), (Boc)2O (0.53 mL, 2.3 mmol) and DMAP (12.8 mg, 0.11 mmol). The reaction mixture was stirred at room temperature for 18 h. The mixture was concentrated in vacuo. The residue was purified by column chromatography on silica gel (petroleum ether/ethyl acetate=1/0 to 5/1) to afford 346-2.
Step 3: Compound 346-3 was prepared from compound 346-2 following the procedure for the synthesis of compound 272-3 in example 51.
Step 4: To a solution of 346-3 (580 mg, 1.04 mmol) in DCM (8 mL) was added TFA (2 mL, 1.04 mmol), the reaction mixture was stirred at room temperature for 3 h. The mixture was concentrated in vacuo. The residue was diluted with EtOAc and NaHCO3 solution. The organic layer was separated and washed with brine. The organic layer was separated and dried over Na2SO4, filtered and concentrated under vacuum to afford 346-4 which was used for the next step directly without further purification.
Step 5: Compound 346 and 347 was prepared from compound 346-4 following the procedure for the synthesis of compound 272-6 in example 51. The pure product 346 and 347 was obtained by reverse HPLC (MeCN/water: 5%-95%). 346: LCMS (ESI, m/z): [M+H]+=524.2; 1H NMR (400 MHz, CDCl3, ppm): δ 7.72-7.63 (m, 2H), 7.58-7.56 (m, 2H), 7.47-7.45 (m, 2H), 5.31-5.24 (m, 1H), 4.15-4.09 (m, 1H), 3.94-3.87 (m, 1H), 3.68 (s, 3H), 3.47-3.40 (m, 2H), 3.27-3.21 (m, 1H), 2.96-2.94 (m, 1H), 2.47-2.46 (m, 2H), 1.85-1.74 (m, 1H), 1.53-1.48 (m, 1H), 1.12-1.05 (m, 3H), 1.00-0.90 (m, 1H), 0.83-0.73 (m, 1H), 0.52-0.43 (m, 1H), 0.41-0.32 (m, 1H), 0.02-−0.02 (m, 1H). 19F NMR (376 MHz, CDCl3, ppm): δ −62.27 (3F). 347: LCMS (ESI, m/z): [M+H]+=524.2; 1H NMR (400 MHz, CDCl3, ppm): δ 7.66-7.60 (m, 2H), 7.56-7.54 (m, 2H), 7.42-7.40 (m, 2H), 5.23-5.17 (m, 1H), 4.38-4.32 (m, 1H), 4.07-4.00 (m, 1H), 3.67 (s, 3H), 3.65-3.57 (m, 1H), 3.42-3.35 (m, 1H), 2.97-2.91 (m, 1H), 2.77-2.71 (m, 1H), 2.64-2.55 (m, 1H), 2.50-2.43 (m, 1H), 1.69-1.59 (m, 1H), 1.45-1.35 (m, 1H), 0.97-0.88 (m, 1H), 0.82-0.75 (m, 4H), 0.43-0.28 (m, 2H), 0.02-−0.02 (m, 1H). 19F NMR (376 MHz, CDCl3, ppm) δ −62.31 (3F).
Compounds of the present disclosure can be synthesized by those having ordinary skill in the art in view of the present disclosure. Representative further compounds synthesized by following similar procedures/methods described herein in the Examples section. The structures and representative analytical data are shown in Table 1 below.
1H-NMR
1H NMR (400 MHz, DMSO-d6, ppm) δ 8.29-8.27 (m, 1H), 8.17-8.15 (m, 1H), 7.63-7.55 (m, 2H), 7.23-7.16 (m, 1H), 4.15-4.05 (m, 2H), 3.73-3.62 (m, 2H), 3.53 (s, 3H), 2.55-2.52 (m, 1H), 2.50-2.47 (m, 1H), 2.42 (s, 3H), 2.07-1.98 (m, 2H), 1.50 (s, 3H).
1H NMR (400 MHz, DMSO-d6, ppm) δ 8.20 (d, J = 8.8 Hz, 1H), 8.11 (d, J = 8.8 Hz, 1H), 7.70 (s, 1H), 7.59-7.53 (m, 3H), 7.20-7.17 (dd, J = 8.0, 1.2 Hz, 1H), 3.82-3.79 (m, 2H), 3.57 (s, 3H), 3.38-3.25 (m, 3H), 2.42 (s, 3H), 2.22-2.19 (m, 2H), 2.15-2.06 (m, 2H).
1H NMR (400 MHz, DMSO-d6, ppm) δ 8.26- 8.24 (m, 1H), 7.99-7.96 (m, 1H), 7.18-7.15 (m, 2H), 6.51-6.49 (m, 2H), 4.44 (s, 1H), 4.30 (t, J = 6.8 Hz, 1H), 3.90-3.73 (m, 6H), 3.45 (s, 3H), 2.32 (t, J = 6.8 Hz, 1H), 2.17 (t, J = 6.8 Hz, 1H). 19F NMR (376 MHz, DMSO-d6, ppm): δ −57.32 (3F).
1H NMR (400 MHz, DMSO-d6, ppm) δ 8.20- 8.19 (m, 1H), 8.09-8.10 (m, 1H), 7.64-7.45 (m, 4H), 7.18-7.20 (m, 1H), 3.63-3.66 (m, 2H), 3.56 (s, 3H), 3.19-3.24 (m, 2H), 2 .40-2.49 (m, 5H), 1.95-2.00 (m, 2H), 1.45 (s, 3H).
1H NMR (400 MHz, CDCl3-d, ppm) δ 7.89- 7.87 (m, 1H), 7.76-7.74 (m, 1H), 7.66 (s, 1H), 7.49 (s, 1H), 7.33-7.30 (m, 4H), 4.47- 4.44 (m, 2H), 3.72-3.62 (m, 5H), 3.07-3.01 (m, 1H), 2.28-2.25 (m, 5H), 2.10-2.00 (m, 2H).
1H NMR (400 MHz, CDCl3-d, ppm) δ 8.14 (s,1H), 7.83-7.80 (m, 1H), 7.72-7.70 (m, 1H), 7.63 (s, 1H), 7.44 (s, 1H), 7.30-7.27 (m, 4H), 6.15 (s, 1H), 4.03-4.00 (m, 2H), 3.64 (s, 3H), 3.44-3.38 (m, 2H), 3.04-2.99 (m, 1H), 2.24 (s, 3H), 2.14-2.12 (m, 2H), 2.03-2.00 (m, 2H).
1H-NMR (400 MHz, CDCl3-d, ppm) δ 7.80-7.77 (m, 1H), 7.56-7.50 (m, 1H), 7.40 (s, 1H), 7.33- 7.26 (m, 1H), 7.07-7.05 (m, 1H), 5.60-5.50 (m, 1H), 5.10-4.90(m,1H), 3.90-3.70 (m, 1H), 3.52 (s, 3H), 3.50-3.25 (m, 2H) 2.39 (s, 3H), 2.35- 2.25 (m, 2H), 2.20-2.05 (m, 2H).
1H NMR (400 MHz, DMSO-d6, ppm) δ 9.52-9.43 (m, 1H), 8.28-8.23 (m, 1H), 7.80-7.75 (m, 1H), 7.62-7.43 (m, 2H), 7.26-7.09 (m, 1H), 3.79-3.71 (m, 2H), 3.63-3.57 (m, 5H), 3.14-3.04 (m, 1H), 2.45-2.38 (m, 3H), 2.29-2.23 (m, 2H), 2.13-2.05 (m, 1H), 1.95-1.84 (m, 1H)
1H NMR (400 MHz, DMSO-d6, ppm): δ 8.30-8.24 (m, 1H), 8.06-8.00 (m, 1H), 7.86-7.72 (m, 2H), 7.55-7.40 (m, 2H), 4.80-3.85 (m, 4H), 3.47 (s, 3H), 3.37-3.34 (m, 2H), 2.05-1.89 (m, 4H). 19F NMR (376 MHz, DMSO-d6, ppm): δ −56.74 (3F).
1H NMR (400 MHz, DMSO-d6, ppm): δ 8.32- 8.20 (m, 1H), 8.07-7.92 (m, 3H), 7.53-7.40 (m, 2H), 4.65-4.28 (m, 4H), 3.96-3.78 (m, 2H), 3.50- 3.39 (m, 3H), 2.33-2.20 (m, 1H), 2.19-2.09 (m, 1H). 19F NMR (376 MHz, DMSO-d6, ppm): δ −56.68 (3F).
1H NMR (400 MHz, DMSO-d6, ppm): δ 8.06- 7.96 (m, 2H), 7.95-7.86 (m, 1H), 7.84-7.74 (m, 1H), 7.47 (d, J = 8.2 Hz, 2H), 4.65-4.21 (m, 4H), 3.95-3.73 (m, 2H), 3.49-3.39 (m, 3H), 2.30-2.07 (m, 2H). 19F NMR (376 MHz, DMSO-d6, ppm): δ −56.68 (3F).
1H NMR (400 MHz, DMSO-d6, ppm) δ 9.70 (s, 1H), 8.97-8.95 (m, 1H), 8.52-8.50 (m, 1H), 7.12- 7.10 (m, 1H), 6.98-6.92 (m, 2H), 4.08-4.05 (m, 2H), 2.89-2.86 (m, 2H), 2.03-2.00 (m, 2H), 1.62- 1.59 (m, 1H), 0.94-0.92 (m, 2H), 0.80-0.78 (m, 2H).
1H NMR (400 MHz, DMSO-d6, ppm) δ 8.20 (d, J = 8.9 Hz, 1H), 7.97-7.93 (m, 2H), 7.90 (s, 1H), 7.77 (d, J = 7.9 Hz, 1H), 7.69-7.64 (m, 1H), 4.70-3.90 (m, 4H), 3.42 (s, 3H), 3.40 (s, 2H), 2.01-1.89 (m, 4H). 19F NMR (376 MHz, DMSO-d6, ppm): δ −61.43 (3F).
1H NMR (400 MHz, DMSO-d6, ppm) δ 8.22 (d, J = 8.9 Hz, 1H), 7.97 (d, J = 9.0 Hz, 1H), 7.84 (d, J = 7.8 Hz, 1H), 7.78-7.74 (m, 1H), 7.69-7.64 (m, 2H), 4.65-3.95 (m, 4H), 3.43 (s, 3H), 3.28 (s, 2H), 2.02-1.94 (m, 4H). 19F NMR (376 MHz, DMSO-d6, ppm): δ −57.42 (3F).
1H NMR (400 MHz, DMSO-d6, ppm): δ 8.31- 8.23 (m, 1H), 8.07-7.99 (m, 1H), 7.93-7.79 (m, 4H), 4.95-3.75 (m, 4H), 3.46 (s, 3H), 3.39 (s, 2H), 2.05-1.89 (m, 4H). 19F NMR (376 MHz, DMSO-d6, ppm): δ −61.23 (3F).
1H NMR (400 MHz, DMSO-d6, ppm): δ 9.57 (s, 1H), 9.00-8.91 (m, 1H), 8.63-8.54 (m, 1H), 7.94-7.80 (m, 4H), 4.45-4.26 (m, 2H), 4.25- 4.04 (m, 2H), 3.41 (s, 2H), 2.12-1.94 (m, 4H). 19F NMR (376 MHz, DMSO-d6, ppm): δ −61.22 (3F).
1H NMR (400 MHz, DMSO-d6, ppm): δ 9.56 (s, 1H), 8.98-8.92 (m, 1H), 8.61-8.55 (m, 1H), 7.88-7.75 (m, 4H), 4.45-4.26 (m, 2H), 4.25-4.08 (m, 2H), 3.39 (s, 2H), 2.11- 1.92 (m, 4H). 19F NMR (376 MHz, DMSO-d6, ppm): δ −41.86 (3F).
1H NMR (400 MHz, Methanol-d4, ppm) δ 8.09-8.04 (m, 1H), 8.02-7.95 (m, 1H), 7.93- 7.84 (m, 2H), 7.80-7.76 (m, 2H), 6.33-5.78 (m, 1H), 5.33 (s, 1H), 5.28-5.03 (m, 1H), 3.78-3.64 (m, 1H), 3.57 (s, 3H), 3.10-2.79 (m, 1H), 2.71-2.52 (m, 1H), 2.44-2.30 (m, 1H), 2.20-2.06 (m, 1H), 1.73-1.52 (m, 3H), 1.15-1.05 (m, 2H), 1.05-0.96 (m, 2H), 0.95-0.87 (m, 3H), 0.86-0.78 (m, 2H). 19F NMR (376 MHz, Methanol-d4): δ 82.48 (1F), 61.33 (4F).
1H NMR (400 MHz, DMSO-d6, ppm) δ 8.29- 8.17 (m, 1H), 8.04-7.94 (m, 1H), 7.84-7.70 (m, 4H), 6.03-5.27 (m, 2H), 5.03-4.75 (m, 1H), 3.68- 3.48 (m, 1H), 3.44 (s, 3H), 3.29-2.80 (m, 1H), 2.71-2.54 (m, 1H), 2.42-2.31 (m, 1H), 2.20-1.69 (m, 3H), 1.65-1.28 (m 2H), 1.14-1.02 (m, 2H), 0.96-0.54 (m, 8H). 19F NMR (376 MHz, DMSO-d6, ppm) δ −41.97 (3F).
1H NMR (400 MHz, DMSO-d6, ppm) δ 9.54 (s, 1H), 8.99-8.89 (m, 1H), 8.63-8.54 (m, 1H), 7.74- 7.65 (m, 2H), 7.57-7.47 (m, 2H), 5.87-4.35 (m, 3H), 4.20 (s, 0.5H), 4.11-4.01 (m, 0.5H), 3.98- 3.78 (m, 2H), 3.57-3.34 (m, 1.5H), 3.08-2.95 (m, 0.5H), 1.83-1.50 (m, 4H), 0.98-0.73 (m, 6H). 19F NMR (376 MHz, DMSO-d6, ppm) δ −60.78 (3F).
1H NMR (400 MHz, CDCl3, ppm) δ 7.79-7.67 (m, 2H), 7.20-7.12 (m, 2H), 6.85 (d, J = 7.4 Hz, 1H), 5.43 (dd, J = 13.0, 2.3 Hz, 1H), 4.45 (dd, J = 11.1, 3.0 Hz, 1H), 4.38-4.21 (m, 2H), 4.17- 4.08 (m, 1H), 3.72 (s, 3H), 3.70-3.64 (m, 1H), 3.44-3.27 (m, 2H), 2.39 (s, 3H), 1.89-1.78 (m, 2H), 1.07 (t, J = 7.4 Hz, 3H).
1H NMR (400 MHz, DMSO-d6, ppm) δ 8.28-8.20 (m, 1H), 8.03-7.98 (m, 1H), 7.78-7.72 (m, 2H), 7.68-7.61 (m, 2H), 5.58-5.26 (m, 1H), 5.04-4.63 (m, 1H), 3.99-3.69 (m, 2H), 3.49-3.35 (m, 6H), 2.91 (t, J = 12.2 Hz, 1H), 2.62-2.51 (m, 2H), 2.24-2.10 (m, 1H), 2.09-1.81 (m, 2H), 0.72 (t, J = 7.2 Hz, 3H). 19F NMR (376 MHz, DMSO-d6, ppm) δ −60.88 (3F).
1H NMR (400 MHz, methanol-d4, ppm) δ 8.21 (d, J = 9.0 Hz, 1H), 7.81 (d, J = 8.9 Hz, 1H), 7.64 (d, J = 8.1 Hz, 2H), 7.54 (d, J = 8.1 Hz, 2H), 5.29 (s, 1H), 5.00 (dd, J = 12.2, 3.2 Hz, 1H), 4.59 (s, 1H), 4.38 (dd, J = 10.8, 2.9 Hz, 1H), 4.06-3.98 (m, 1H), 3.79-3.71 (m, 1H), 3.58-3.47 (m, 1H), 3.11-3.00 (m, 2H), 2.70- 2.60 (m, 1H), 2.46 (d, J = 7.5 Hz, 1H), 2.11- 1.98 (m, 1H), 1.85-1.72 (m, 1H), 1.70-1.64 (m, 1H), 1.60 (t, J = 7.4 Hz, 6H), 0.82 (t, J = 7.4 Hz, 3H), 0.68 (t, J = 7.3 Hz, 3H). 19F NMR (376 MHz, methanol-d4, ppm) δ −63.83 (3F).
1H NMR (400 MHz, CDCl3, ppm): δ 7.75- 7.65 (m, 2H), 7.62-7.52 (m, 2H). 7.47-7.36 (m, 2H), 4.77-4.67 (m, 1H), 4.38-4.28 (m, 3H), 4.17-4.07 (m, 1H), 3.74-3.64 (m, 1H), 3.52- 3.42 (m, 1H), 3.26-3.16 (m, 1H), 2.94-2.80 (m, 1H), 2.74-2.64 (m, 1H), 2.56-2.46 (m, 1H), 2.05-1.85 (m, 1H), 1.77-1.50 (m, 3H), 1.36- 1.29 (m, 3H), 0.86 (t, J = 8.0 Hz, 3H), 0.71 (t, J = 8.0 Hz, 3H). 19F NMR (376 MHz, CDCl3, ppm): δ −62.38 (3F).
1H NMR (400 MHz, methanol-d4, ppm) δ 8.04 (d, J = 8.8 Hz, 1H), 7.88 (d, J = 8.8 Hz, 1H), 7.67-7.55 (m, 4H), 5.24-5.14 (m, 1H), 4.41- 4.33 (m, 1H), 4.13-4.03 (m, 1H), 3.98-3.88 (m, 1H), 3.92-3.79 (m, 2H), 3.71 (s, 3H), 3.66-3.58 (m, 1H), 3.18-3.03 (m, 2H), 2.83-2.73 (m, 1H), 2.55-2.47 (m, 1H), 1.87-1.74 (m, 1H), 1.68-1.56 (m, 1H), 0.83 (t, J = 7.4 Hz, 3H). 19F NMR (376 MHz, methanol-d4, ppm): δ −63.91 (3F).
1H NMR (400 MHz, methanol-d4, ppm) δ 8.03 (d, J = 8.8 Hz, 1H), 7.87 (d, J = 8.7 Hz, 1H), 7.67 (d, J = 8.1 Hz, 2H), 7.57 (d, J = 8.1 Hz, 2H), 5.13 (d, J = 11.2 Hz, 1H), 4.39 (dd, J = 10.9, 2.9 Hz, 1H), 4.10-4.02 (m, 1H), 3.85-3.75 (m, 1H), 3.62-3.52 (m, 1H), 3.16-3.04 (m, 2H), 2.77-2.65 (m, 1H), 2.57-2.45 (m, 1H), 2.14-2.03 (m, 1H), 1.84-1.58 (m, 3H), 0.81 (t, J = 7.4 Hz, 3H), 0.70 (t, J = 7.3 Hz, 3H). 19F NMR (376 MHz, methanol-d4, ppm): δ −63.89 (3F).
1H NMR (400 MHz, CDCl3, ppm) δ 7.63 (dd, J = 12.8, 7.2 Hz, 4H), 7.45 (d, J = 7.9 Hz, 2H), 4.97- 4.77 (m, 2H), 4.37 (dd, J = 10.7, 2.4 Hz, 1H), 4.16-4.05 (m, 1H), 4.03-3.83 (m, 1H), 3.73 (dd, J = 8.7, 4.3 Hz, 1H), 3.53 (s, 1H), 3.10 (d, J = 11.0 Hz, 1H), 2.96 (d, J = 9.1 Hz, 1H), 2.83-2.73 (m, 1H), 2.70-2.47 (m, 5H), 2.08-1.73 (m, 5H), 0.91 (t, J = 7.3 Hz, 3H), 0.72 (t, J = 7.2 Hz, 3H). 19F NMR (376 MHz, CDCl3, ppm) δ −62.36 (3F).
1H NMR (400 MHz, DMSO-d6, ppm) δ 8.08 (s, 2H), 7.71 (d, J = 8.1 Hz, 2H), 7.60 (d, J = 8.0 Hz, 2H), 4.70 (dd, J = 12.2, 3.9 Hz, 1H), 4.27 (dd, J = 10.8, 2.7 Hz, 1H), 4.00 (dd, J = 10.7, 8.0 Hz, 1H), 3.85 (d, J = 14.3 Hz, 1H), 3.73 (d, J = 14.2 Hz, 1H), 3.61 (s, 3H), 3.44- 3.36 (m, 2H), 2.72-2.54 (m, 3H), 1.74-1.57 (m, 2H), 0.95 (t, J = 7.3 Hz, 3H). 19F NMR (376 MHz, DMSO-d6, ppm) δ −60.72 (3F).
1H NMR (400 MHz, DMSO-d6, ppm) δ 8.07 (s, 2H), 7.61 (dd, J = 4.9, 3.3 Hz, 2H), 7.37 (dd, J = 8.3, 1.7 Hz, 1H), 4.71 (dd, J = 12.1, 3.9 Hz, 1H), 4.27 (dd, J = 10.8, 2.7 Hz, 1H), 3.98 (dd, J = 10.7, 8.0 Hz, 1H), 3.74 (d, J = 14.2 Hz, 1H), 3.64 (d, J = 14.2 Hz, 1H), 3.60 (s, 3H), 3.42-3.37 (m, 1H), 3.32-3.28 (m, 1H), 2.70-2.54 (m, 3H), 1.73-1.53 (m, 2H), 0.94 (t, J = 7.3 Hz, 3H).
1H NMR (400 MHz, DMSO-d6, ppm) δ 8.24 (d, J = 8.8 Hz, 1H), 8.06 (d, J = 8.8 Hz, 1H), 7.72 (d, J = 7.6 Hz, 2H), 7.56 (d, J = 7.4 Hz, 2H), 6.31 (t, J = 54.8 Hz, 1H), 4.93-4.65 (m, 3H), 4.38 (d, J = 9.7 Hz, 1H), 4.13-3.96 (m, 1H), 3.82-3.72 (m, 1H), 3.12 (d, J = 11.2 Hz, 1H), 2.93 (d, J = 11.0 Hz, 1H), 2.68-2.60 (m, 1H), 2.42-2.30 (m, 1H), 2.03-1.93 (m, 1H), 1.74-1.50 (m, 3H), 1.23 (s, 1H), 0.73 (t, J = 7.0 Hz, 3H), 0.64 (t, J = 7.1 Hz, 3H). 19F NMR (400 MHz, DMSO-d6, ppm) δ −60.73 (3F), −120.83 (2F).
The DGKa and DGKz kinase assays were performed using Promega ADP-Glo kit (Promega, Cat #V9102). One microliter test compound was added to a 384-well plate. Reactions were performed in a 5 uL volume by adding 2 uL mixture of DGK enzyme and ATP in assay buffer (50 mM MOPS pH 7.5, 100 mM NaCl, 10 mM MgCl2, 1 mM CaCl2, 0.0025% BSA, and 1 mM DTT). The final concentration of DGKa, DGKz and ATP were 10 nM, 5 nM and 15 uM, respectively. The enzyme solution was incubated with test compound at 25 for 20 min. For the preparation of the substrate of micelles, 1 volume of a 16.1 mM solution of 1,2-dioleoyl-sn-glycerol in chloroform was slowly evaporated using a nitrogen stream. Subsequently, 22.55 volumes of a 510 mM solution of octyl-b-D-glucopyranoside in 50 mM MOPS buffer (pH 7.5) were added, and the mixture was sonicated in an ultrasonic bath for 20s. Then 35 volumes of 50 mM MOPS buffer (pH7.5) were added to yield a solution of 0.28 mM 1,2 dioleoyl-sn-glycerol and 200 mM octyl-b-D-glucopyranoside, which was aliquoted, flash-frozen in liquid nitrogen, and stored at −80° C. until use. The reaction was initiated by the addition of 2 uL of substrate solution (7 uM 1.2-dioleoyl-sn-glycerol, 5 mM octyl-b-D-glucopyranoside) in the 5 uL assay volume. The resulting mixture was incubated at 25° C. for 40 minutes. Next, 5 uL of ADP-Glo-reagent was added to the plate and incubated for 40 minutes. Then 10 uL of kinase detection reagent was added and incubated for 30 minutes. Luminescence was recorded using an Tecan SPARK microplate reader. 1% DMSO vehicle was used as control and no enzyme well was used as blank well. The percent inhibition was calculated with the formula: % inhibition=100-100*(RLUcmpd−RLUblank)/(RLUcontrol−RLUblank). Inhibition at 50% activity (IC50) was calculated with the equation of Y=Bottom+(Top−Bottom)/(1+10{circumflex over ( )}((Log IC50−X)*Hill Slope)).
PBMC (frozen) obtained from health donor were thawed in RPMI-1640 medium with 10% FBS and incubated for 4 h at 37° C. In a 96-well plate, −200,000 PBMC cells in 160 uL were added to each assay well. Ten microliter compound solution (10 uM to 0.6 nM, 8 points with 4-fold dilution) were added to cells in each well and incubated for 60 minutes at 37° C. Then 30 uL anti-CD3/CD28 antibody (Catalog #10991, Stemcell) were added to cells and incubated overnight at 37° C. After the incubation, 16 uL supernatants were transferred into a new 384-well white assay plate for cytokine analysis. IL-2 level was quantified using Human IL-2 kit (Catalog #62HIL02PEG, Cisbio) as described in the manufacturer manual. Activation at 50% activity (EC50) was calculated with the equation:
X is log of compound concentration, Y is the IL-2 concentration, increasing as X increases.
Jurkat cells (ATCC) were cultured in RPMI-1640 medium supplemented with 10% FBS. In a 96-well plate, −250,000 cells in 130 uL were added to each assay well. Ten microliter compound solution (5 uM to 2.3 nM, 8 points with 3-fold dilution) were added to cells in each well and incubated for 60 minutes at 37° C. Then 10 uL anti-CD3/CD28 antibody (Catalog #10991, Stemcell) were added and incubated overnight at 37° C. After the incubation, 16 uL supernatants were transferred into a new 384-well white assay plate for cytokine analysis. IL-2 was quantified using Human IL-2 kit (Catalog #62HIL02PEG, Cisbio) as described in the manufacturer manual.
Activation at 50% activity (EC50) was calculated with the equation:
X is log of compound concentration, Y is the IL-2 concentration, increasing as X increases.
This study aimed to assess the metabolic stability of a compound in human liver microsomes using a microsomal clearance assay.
A mixture containing 100 mM potassium phosphate, pH 7.4, 0.5 mg/mL liver microsomes, 2 mM NADPH, and 1 μM compound were prepared and added to 96-well plate. The plates were then incubated at 37 for different time points (0, 5, 15, 30, 45 minutes) and the reaction was stopped with acetonitrile solution containing an internal standard. The samples were then analyzed by LC/MS/MS to determine how much of the compound remained at each time point. The elimination rate constant and half-life were calculated from the data, and the in vitro intrinsic clearance (CLint) was determined as follows:
The in vitro intrinsic clearance, CLint, was calculated from the t1/2 as follows: CL′int=(0.693/t1/2)×(1/(microsomal protein concentration (0.5 mg/mL))) X Physiological Scaling Factor.
The whole blood from male C57BL6 mice (6-8 weeks old, Beijing Vital River Laboratory Animal Technology Co., Ltd.) was collected, pooled and dispensed at 200 uL per well in a 96-well plate. The blood was treated with compounds for one hour at 37° C. in a humidified 5% CO2 incubator. The blood was then stimulated with anti-mouse CD3 and anti-mouse CD28 antibody (BioLegend Cat #100340, #102116) at a final concentration of 0.6 ug/mL for 20 hours at 37° C. in a humidified 5% CO2 incubator. The blood was spun down, IFN-gamma in the supernatants was measured using ELISA kit (Excellbio Cat #EM002-96, #EM007-96) according the manufacture's instruction.
Murine colon adenocarcinoma cells MC-38 were obtained from American Type Culture Collection (ATCC) and cultured in RPMI 1640 media containing 10% fetal bovine serum (FBS) at 37° C. in a humidified 5% CO2 incubator. Female C57BL/6 mice (20 2 g, 6-8 weeks old) were purchased from SPF Biotechnology Co., Ltd. (Beijing, China). MC-38 cells were collected from flask and suspended in PBS. Suspension of 1×106 cells in 0.1 mL culture media without FBS was injected subcutaneously into animal armpits. Once the tumor reached 100-150 mm3, the mice were randomized into different groups and treatment was initiated. Mice were treated orally once per day (PO, QD) with compound 67 (0.6 mg/kg) or vehicle control. Anti-PD-1 antibody (BioXcell, Cat #BE0273) was diluted with PBS for dosing at 2 mg/kg and was administrated via intraperitoneal injection, twice per week. Tumor volume and body weight of mice were recorded twice per week.
All animals were housed in a room maintained at 23±3° C., 65%-75% humidity, with a controlled 12 h light-dark cycle for 5-7 days before experiments. Water and commercial laboratory complete food for mice were provided in the cage.
Tumor volume was calculated by measuring two perpendicular diameters using the following formula: (L×W2)/2, in which L and W refer to the length and width tumor diameter, respectively. Tumor growth inhibition (TGI) was calculated according to the following equation: TGI (%)=(1−(TVTreatment/Dn−TVTreatment/D0)/(TVControl/Dn−TVControl/D0))×100%, where the Dn is the final tumor volume and DO is starting tumor volume prior to treatment. Combination with anti-PD-1 shows significantly better tumor inhibition activity than either of the single agents (compound 67 or anti-PD-1) (see
Murine colorectal adenocarcinoma cells CT26 (purchased from ATCC) were cultured in RPMI 1640 medium with 10% FBS. For tumor implantation, 6-8 weeks old BALB/c female mice (Beijing Vital River Laboratory Animal Technology Co., Ltd.) were given a subcutaneous injection of CT26 cell (1×10{circumflex over ( )}7 cells/mL) in 0.1 mL suspension into the right flank. Once tumors grew to a volume of 50-100 mm3, mice were randomized into different groups and treatment was initiated. Mice were treated orally once per day (PO, QD) with DGK inhibitors (3 mg/kg) or vehicle control. Anti-PD-1 antibody (BioXcell, Cat #BE0273), anti-CTLA4 antibody (BioXcell, Cat #BE0164) were diluted with PBS for dosing at 10 mg/kg or 5 mg/kg, respectively. Antibody was administrated via intraperitoneal injection, twice per week. Tumor volume and body weight of mice were recorded twice per week.
All animals were housed in a room maintained at 23:3° C., 65%-75% humidity, with a controlled 12 h light-dark cycle for 5-7 days before experiments. Water and commercial laboratory complete food for mice were provided in the cage.
Exemplary compounds including compound 250 have been tested in this model and were found to inhibit tumor growth.
The Summary and Abstract sections may set forth one or more but not all exemplary embodiments of the present invention as contemplated by the inventor(s), and thus, are not intended to limit the present invention and the appended claims in any way.
The present invention has been described above with the aid of functional building blocks illustrating the implementation of specified functions and relationships thereof. The boundaries of these functional building blocks have been arbitrarily defined herein for the convenience of the description. Alternate boundaries can be defined so long as the specified functions and relationships thereof are appropriately performed.
With respect to aspects of the invention described as a genus, all individual species are individually considered separate aspects of the invention. If aspects of the invention are described as “comprising” a feature, embodiments also are contemplated “consisting of” or “consisting essentially of” the feature.
The foregoing description of the specific embodiments will so fully reveal the general nature of the invention that others can, by applying knowledge within the ordinary skill of the art, readily modify and/or adapt for various applications such specific embodiments, without undue experimentation, without departing from the general concept of the present invention. Therefore, such adaptations and modifications are intended to be within the meaning and range of equivalents of the disclosed embodiments, based on the teaching and guidance presented herein. It is to be understood that the phraseology or terminology herein is for the purpose of description and not of limitation, such that the terminology or phraseology of the present specification is to be interpreted by the ordinarily skilled artisan in light of the teachings and guidance.
The breadth and scope of the present invention should not be limited by any of the above-described exemplary embodiments.
All of the various aspects, embodiments, and options described herein can be combined in any and all variations.
All publications, patents, and patent applications mentioned in this specification are herein incorporated by reference to the same extent as if each individual publication, patent, or patent application was specifically and individually indicated to be incorporated by reference. To the extent that any meaning or definition of a term in this document conflicts with any meaning or definition of the same term in a document incorporated by reference, the meaning or definition assigned to that term in this document shall govern.
Number | Date | Country | Kind |
---|---|---|---|
PCT/CN2022/084377 | Mar 2022 | WO | international |
PCT/CN2022/118935 | Sep 2022 | WO | international |
This application claims priority to International Application Nos. PCT/CN2022/084377, filed on Mar. 31, 2022 and PCT/CN2022/118935, filed on Sep. 15, 2022, the contents of each of which are incorporated herein by reference in their entireties.
Filing Document | Filing Date | Country | Kind |
---|---|---|---|
PCT/CN2023/085320 | 3/31/2023 | WO |