Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
INHIBITORS OF FIBROBLAST ACTIVATION PROTEIN
Document Type and Number:
WIPO Patent Application WO/2019/118932
Kind Code:
A1
Abstract:
Compounds and compositions for modulating fibroblast activation protein (FAP) are described. The compounds and compositions may find use as therapeutic agents for the treatment of diseases, including hyperproliferative diseases.

Inventors:
ALFARO JENNIFER (CL)
BELMAR SEBASTIAN (CL)
BERNALES SEBASTIAN (US)
PUJALA BRAHMAM (US)
PANPATIL DAYANAND (US)
BHATT BHAWANA (US)
Application Number:
PCT/US2018/065859
Publication Date:
June 20, 2019
Filing Date:
December 14, 2018
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
PRAXIS BIOTECH LLC (US)
International Classes:
A61K31/40; A61K31/4025; C07D207/10
Domestic Patent References:
WO2013107820A12013-07-25
WO2019083990A22019-05-02
Foreign References:
US20100081701A12010-04-01
US20120045509A12012-02-23
US20060276435A12006-12-07
US20100291020A12010-11-18
Other References:
SANCHEZ-GARRIDO, MA ET AL.: "Fibroblast Activation Protein (FAP) as a Novel Metabolic Target", MOLECULAR METABOLISM, vol. 5, no. 10, 16 July 2016 (2016-07-16), pages 1015 - 1024, XP055617682, DOI: 10.1016/j.molmet.2016.07.003
RYABTSOVA, O ET AL.: "Acylated Gly-(2-cyano)pyrrolidines as inhibitors of fibroblast activation protein (FAP) and the issue of FAP/prolyl oligopeptidase (PREP)-selectivity", BIOORGANIC & MEDICINAL CHEMISTRY LETTERS, vol. 22, 2012, pages 3412 - 3417, XP028479271, DOI: 10.1016/j.bmcl.2012.03.107
TSU, H ET AL.: "2-[3-[2-[(2S)-2-Cyano-1-pyrrolidinyl]-2-oxoethylamino]-3-methyl-1- oxobutyl]-1,2,3,4-tetrahydroisoquinoline: A Potent, Selective, and Orally Bioavailable Dipeptide-Derived Inhibitor of Dipeptidyl Peptidase IV", JOURNAL OF MEDICINAL CHEMISTRY, vol. 49, no. 1, 2006, pages 373 - 380, XP008100456, DOI: 10.1021/jm0507781
See also references of EP 3723753A4
Attorney, Agent or Firm:
REANEY, Shannon (US)
Download PDF:
Claims:
What is claimed is:

1. A compound of formula (I):

or a pharmaceutically acceptable salt thereof, wherein;

R is hydrogen, Ci-Cg alkyl, Cg-Cg cycloalkyl, 3- to 12-membered heterocyclyl, 5- to 10- membered heteroaryl, or Ce-Cn aryl, wherein the Ci-Cr, alkyl, Cg-Cg cydoalkyl, 3- to 12- membered heterocyclyl, 5- to lO-membered heteroaryl, and C6-C 4 aryl of R are independently optionally substituted by Rd; m is 0, l, 2, 3, or 4; n is 0, 1 , 2, 3, or 4, wherein m + n is 1 , 2, 3, or 4;

X is -P OK -0-, -CH(OH)-, -S-. -S( OK or -Si O h-.

L is wherein

* represents the point of attachment to the Y-X- moiety,

** represents the point of attachment to the remainder of the molecule,

Ra is hydrogen, C.-Ce alkyl, Cg-Cg cycloalkyl, 3- to 12-membered heterocyclyl, 5- to lO-membered heteroaryl, or C6-Ci4 aryl, wherein the Ci-C6 alkyl, Cg-Cg cycloalkyl, 3- to 12-membered heterocyclyl, 5- to 10-membered heteroaryl, and C6-C 4 aryl of Ra are independently optionally substituted by Rc. R1 and R2, independently of each other and independently at each occurrence, are hydrogen, CrC2 alkyl, C3-C8 cycloalkyl, 3- to 12-membered heterocyclyl, 5- to 10-membered heteroaryl, or Ce-Ci4 aryl, wherein the C3-C8 cycloalkyi, 3- to 12- membered heterocyclyl, 5- to 10-membered heteroaryl, and C6-Ci4 aryl of R1 and R2 are independently optionally substituted by R1, or R1 and R2 are taken together with the carbon atom or atoms to which they are attached to form a 3- to 8-membered cycloalkylene optionally substituted by Rf, q is l, 2, or 3,

RJ and R4, independently of each other and independently at each occurrence, are hydrogen, C -Cg cycloalkyi, 3- to 12-membered heterocyclyl, 5- to 10-membered heteroaryl, or Ce-Ci4 aryl, wherein the C3-Cg cycloalkyi, 3- to 12-membered heterocyclyl, 5- to 10-membered heteroaryl, and C6-CM aryl of RJ and R4 are independently optionally substituted by Rg, or R3 and R4 are taken together with the carbon atom to which they are attached to form a 3- to 8-membered cycloalkylene optionally substituted by R8, and p is 0, 1, or 2; , wherein

* represents the point of attachment to the Y-X- moiety,

** represents the point of attachment to the remainder of the molecule,

R5 and R6, independently of each other and independently at each occurrence, are

H, C -Ce alkyl, C3-Cg cycloalkyl, 3- to 12-membered heterocyclyl, 5- to 10- membered heteroaryl, or Ce-Ci4 aryl, wherein the Ci-Ce alkyl, C3-Cg cycloalkyi,

3- to 12-membered heterocyclyl, 5- to 10-membered heteroaryl, and C6-Cl4 aryl of R5 and R6 are independently optionally substituted by R1-', Rb and Rc are independently H, Ci-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, Cg-Cg cycloalkyl, 3- to 12-membered heterocyclyl, 5- to 10-membered heteroaryl, C6- C14 aryl, or -C(=0)0R1 wherein the C-.-Ce alkyl, Cg-Cg cycloalkyl, 3- to 12- membered heterocyclyl, 5- to lO-membered heteroaryl, and C6-Ci4 aryl of Rb and Rc are independently optionally substituted by R1, and r is 1, 2, or 3; or , wherein

* represents the point of attachment to the Y-X- moiety,

** represents the point of attachment to the remainder of the molecule,

R ' and R8, independently of each other and independently at each occurrence, are hydrogen, Cg-Cg cycloalkyl, 3- to 12-membered heterocyclyl, 5- to lO-membered heteroaryl, or Ce-Cu aryl, wherein the Cg-Cg cycloalkyl, 3- to 12-membered heterocyclyl, 5- to 10-membered heteroaryl, and Ce-Cn awl of R' and R8 are independently optionally substituted by RJ, or R' and R8 are taken together with the carbon atom to which they are attached to form a 3- to 8-membered cyc!oalky!ene optionally substituted by RJ,

R9 and R10, independently of each other and independently at each occurrence, are H, C -Cg alkyl, Cg-Cg cycloalkyl, 3- to 12-membered heterocyclyl, 5- to 10- membered heteroaryl, or Ce-Cu aryl, wherein the Cg-Cg alkyl, Cg-Cg cycloalkyl, 3- to 12-membered heterocyclyl, 5- to 10-membered heteroaryl, and Cg-Cig aryl of R9 and R10 are independently optionally substituted by Rk, s is 1, 2, or 3, t is 1, 2, or 3, wherein s + t is 2, 3, or 4, u is 0 or 1 , and v is 0 or 1 ;

Y is C6-C9 axyl optionally substituted by R11, 6- to 10-membered heteroaiyl optionally substituted by R12, or 3- to 12-membered heterocyclyl optionally substituted by R! i, wherein when Y is phenyl or naphthyl, the phenyl or naphthyl of Y is substituted by at least one R! !, and wherein when L is *-NH~CH2~** and Y is optionally substituted quinolinyl, the optionally subs tituted quinolinyl of Y is connected to the parent s tructure at the 2-, 3-, 5-, 6-, 7-, or 8- position, wherein

R11, R'2, and R1’, independently of each other and independently at each occurrence, are Ci-Cg alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C8 cycloalkyl, C4-C8 cycloalkenyl, 3- to 12-membered heterocyclyl, 5- to lO-membered heteroaryl, Ce-Cl4 aryl, halogen, cyano, oxo, -OR14, -NRI5R16, -SRi4, -NO?., -C=NH(OR14), -C(0)R14, -0C(0)R14, -C(Q)ORi4, -C(0)NR15R16, -NR14C(0)R1s, -NR14C(0)0R15, -NR14C(0)NR15R16, -S(Q)R14, -S(0)2R14, -NR14S(0)R15, -NR14S(0)2Ri5, -S(0)NRlsR16, ~S(0)2NR15R16,

or -P(0)(0R! ')(0R!6), wherein each Rn R12, and Ri 3 is independently optionally substituted by RL; each R14 is independently hydrogen, CrC6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3- C8 cycloalkyl, C6-C14 aryl, 5- to 10-membered heteroaiyl, or 3- to 12-membered heterocyclyl, wherein the Ci-Cf, alkyl, C2-Ce alkenyl, C2-C6 alkynyl, C3-C8 cycloalkyl, Ce-Cu aryl, 5- to 10- membered heteroaryl, and 3- to 12-membered heterocyclyl of R14 are independently optionally substituted by halogen, -OH, oxo, cyano, or C -C6 alkyl optionally substituted by halogen, -OH, or oxo;

R15 and R16, independently of each other and independently at each occurrence, are hydrogen, Cj-Ce alkyl, CVCV, alkenyl, C2-Ce alkynyl, C3-C8 cycloalkyl, C6-C]4 aryl, 5- to 10-membered heteroaiyl, or 3- to 12-membered heterocyclyl, wherein the C1-C5 alkyl, CrCe alkenyl, C2-C6 alkynyl, C3-C8 cycloalkyl, C6-Cl4 atyl, 5- to 1 Q-membered heteroard, and 3- to 12-membered heterocyclyl of R” and R16 are independently optionally substituted by halogen, -OH, oxo, cyano, or C -C6 alkyl, optionally substituted by halogen, -OH, or oxo, or Ri 5 and R16 are taken together with the atom to which they are attached to form a 3- to 6-membered heterocyclyl optionally substituted by halogen, oxo, cyano, or Ci-Cc, alkyl optionally substituted by halogen, -OH, or oxo;

Ra, Re, Rf, Rg, Rn, R1, R, and Rk, independently of each other and independently at each occurrence, are halogen, Ci-C6 alkyl, C2-C6 alkenyl, C2~Ce, alkynyl, Cj-Cg cycloalkyl, C6-C14 aryl, 5- to 10-membered heteroaryl, 3- to 12-membered heterocyclyl, -OR14, -NR15R16, cyano, or nitro; and each R1 IS independently halogen, CrC6 alkyl, C2-Ce alkenyl, C2-C6 alkynyl, CYCg cycloalkyl, C6-C 4 aryl, 5- to 10-membered heteroaryl, 3- to 12-membered heterocyclyl, -OR14, ~C(0)R14, - NRlsR16, cyano, oxo, or nitro.

2. The compound of claim 1, or a salt thereof, wherein X is -C(=0)-.

3. The compound of claim 1, or a salt thereof, wherein X is -0-.

4. The compound of claim 1, or a salt thereof, wherein X is -CH(OH)-.

5. The compound of any one of claims 1 to 4, or a salt thereof, wherein L is -NH-CR1R2-.

6. The compound of claim 5, or a salt thereof, wherein L is -NH-CH2-.

7. The compound of claim 5, or a salt thereof wherein L is -NH-CH(CH3)-.

8. The compound of claim 5, or a salt thereof wherein L is -NH-CRfR2-, wherein R1 and R2 are taken together with the carbon atom to which they are attached to form a 3- to 8-membered cycloalkylene.

9. Hie compound of claim 8, or a salt thereof, wherein R1 and R2 are taken together with the carbon atom to which they are attached to form a cyclopropylene.

10. The compound of any one of claims 1 to 4, or a salt thereof, wherein L is

-CR5R6-CH(NRbRc)-.

11. The compound of claim 10, or a salt thereof, wherein L is -CR5R6-CH(NRbRc)-, wherein R6, Rb, and Rc are H, and R5 is H or Ci-Ce alkyl.

12. The compound of any one of claims 1 to 4, or a salt thereof, wherein L is , where * represents the point of attachment to the Y-X- moiety, ** represents the point of attachment to the remainder of the molecule.

13. The compound of claim 12, or a salt thereof, wherein L is , wherein * represents the point of attachment to the Y-X- moiety', and ** represents the point of attachment to the remainder of the molecule.

14. The compound of any one of claims 1 to 13, or a salt thereof, wherein Y is 6- to 10- membered heteroaryl optionally substituted by R1"·, or 3- to 12-membered heterocyclyl optionally substituted by R!Y

15. The compound of claim 1, or a salt thereof, wherein Y is Ce-C6 aryl optionally substituted by R11, wherein when Y is phenyl or naphthyl, the phenyl or naphthyl of Y is substituted by at least one R“.

16. The compound of claim 15, wherein Y is phenyl substituted by 1 to 5 R11, which are independently selected from halogen, trihalomethyl, cyano, and -C(=0)NH2.

17. The compound of claim 15, wherein Y is unsubstituted 2,3-dihydro- li -inden-2-yl.

18. The compound of claim 14, or a salt thereof, wherein Y is 6- to 10-membered heteroaryl optionally substituted by R12, wherein when L is *-NH-CH2-** and Y is optionally substituted quinolinyl, the optionally substituted quinolinyl of Y is connected to the parent structure at the 2-, 3-, 5-, 6-, 7-, or 8-position.

19. The compound of claim 18, or a salt thereof, wherein either:

(a) L is *-CH2-CH(NH2)-** or *-CH(CH3)-CH(NH2)-** and Y is quinolin-4-yl optionally substituted by Rl or

(b) L is *-NH-CH2-**, Y is quinolin-6-yl optionally substituted by R12, which R32 is independently selected from -OH and phenyl.

20. The compound of claim 18, or a salt thereof, wherein Y is pyridin-4-yl substituted by R 2 in the 3-posiiion

21. The compound of claim 20, or a salt thereof, wherein Rl is pyridmyl optionally substituted by C-.-Cb alkyl.

22. The compound of claim 20, or a salt thereof, wherein Rlz is indolyl optionally substituted by Ci-Ce alkyl.

23. The compound of claim 20, or a salt thereof, wherein Riz is phenyl optionally substituted by Ci-Ce alkyl, halogen, or CrC6 alkoxy.

24. The compound of claim 18, or a salt thereof, wherein Y is pyrimidm-4-yi optionally substituted by R'2 and optionally fused to (',,··(' i i aryl or C5-C10 cycloalkyl, wherein C6-Ci4 aryl and C5-C10 cycloalkyl are optionally substituted by Rl2

25. The compound of claim 24, or a salt thereof, wherein Y is pyrimidin-4-yl fused to Ce-Cn aryl, wherein Ce-Cu and is optionally substituted by R12.

26. The compound of claim 24, or a salt thereof, wherein Y is optionally substituted pyridin-3- yi, imsubstituted quinazohn-4-yl or unsubstituted 6,7-dihydro-5//-cyclopentaj d]pyrimidin-4-yl.

27. The compound of claim 18, or a salt thereof, wherein Y is 2//-pyran-2-Qn-5-yl optionally substituted by R and optionally fused to C6-C14 aryl, which C6-C14 aryl is optionally substituted by R 2 .

28. The compound of claim 27, or a salt thereof, wherein Y is l -isochromen- 1 -on-4-yl optionally substituted by halogen.

29. The compound of claim 18, or a salt thereof, wherein Y is pyridm-2(l//)-on-5-yl optionally substituted by Rl2 and optionally fused to Ce-Cw aryl or 5- to ! O-memhered heterocyclyl, which C¾-C i4 axyl or 5- to 10-membered heterocyclyl, independently of each other and independently at each occurrence, are optionally substituted by R12.

30. The compound of claim 29, or a salt thereof, wherem Y is pyridin-2(l//)-on-5-yl optionally substituted by C -Cg alkyl or C6-Cl4 aryl.

31. The compound of claim 29, or a salt thereof, wherein Y is unsubstituted 7,8,9,10- tetrahy dropyrido[ 1 ,2-a] azepin-4(6//)-on-l -y!.

32. The compound of claim 29, or a salt thereof, wherein Y is isoquinolin-l(2//)-on-4-yl optionally substituted by halogen, Ci-Ce alkyl, CA-C aryl, or C3-C8 cycloalkyl.

33. The compound of claim 14, or a salt thereof, wherein Y is 3- to 12-membered heterocycly! optionally substituted by Rl .

34 The compound of claim 33, or a salt thereof, wherein Y is unsubstituted isoindolin-2-yl.

35 The compound of claim 33, or a salt thereof, wherein Y is piperidin-2-on-5-yl optionally substituted by C -Ce alkyl or C6-Ci4 aryl.

36 The compound of any one of claims 1 to 35, or a salt thereof, wherein m = n = 1.

37. The compound of any one of claims 1 to 36, or a salt thereof, wherein R is hydrogen.

38. The compound of claim 1, or a salt thereof, wherein the -X-L- moiety is selected from the

, and O ; wherein * represents the point of attachment to the

Y moiety, and ** represents the point of attachment to the remainder of the molecule.

39. A pharmaceutical composition comprising a compound of any one of claims 1-38, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier.

40. A method of treating a disease or disorder mediated by fibroblast activation protein (FAP) in an individual in need thereof comprising administering to the individual a therapeutically effective amount of a compound of any one of claims 1 -38, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition of claim 39.

41. A method of treating a disease or disorder characterized by proliferation, tissue remodeling, chronic inflammation, obesity, glucose intolerance, or insulin insensitivity m an individual in need thereof, comprising administering to the individual a therapeutically effective amount of a compound of any one of claims 1-38, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition of claim 39.

42. The method of claims 40 or 41, wherein the disease or disorder is breast cancer, colorectal cancer, ovarian cancer, prostate cancer, pancreatic cancer, kidney cancer, lung cancer, melanoma, fibrosarcoma, bone sarcoma, connective tissue sarcoma, renal cell carcinoma, giant cell carcinoma, squamous cell carcinoma, leukemia, skin cancer, soft tissue cancer, liver cancer, gastrointestinal carcinoma, or adenocarcinoma.

43. 'The method of claim 42, wherein the disease or disorder is metastatic kidney cancer, chronic lymphocytary leukemia, pancreatic adenocarcinoma, or non-small cell lung cancer.

44. The method of claims 40 or 41 , wherein the disease or disorder is fibrotic disease, wound healing, keloid formation, osteoarthritis, rheumatoid arthritis and related disorders involving cartilage degradation, atherosclerotic disease, Crohn’s disease, or Type II diabetes.

45. A method of reducing tumor growth, tumor proliferation, or tumorigenieity in an individual in need thereof, comprising administering to the individual a compound of any one of claims 1-38, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition of claim 39.

46. A method of inhibiting FAP in an individual comprising administering to the individual a compound of any one of claims 1 -38, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition of claim 39.

47. A method of inhibiting FAP in a cell comprising administering or delivering to the cell a compound of any one of claims 1-38, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition of claim 39, or a metabolite of the foregoing.

48. The method of claim 47, wherein the cell is a fibroblast.

49. Tire method of claims 47 or 48, wherein the cell is a cancer associated fibroblast (CAF) or a reactive stromal fibroblast.

50. A method of inhibiting FAP in a tumor comprising administering or delivering to the tumor a compound of any one of claims 1 -38, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition of claim 39, or a metabolite of the foregoing.

51. A method of inhibiting FAP in plasma comprising administering or delivering to the plasma a compound of any one of claims 1-38, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition of claim 39, or a metabolite of the foregoing.

52. The method of any one of claims 46-51, wherein inhibiting FAP comprises inhibiting an endopeptidase activity of FAP.

53. The method of any one of claims 46-51 , wherein inhibiting F AP comprises inhibiting an exopepiidase activity of FAP.

54. A method of enhancing an immune response in an individual comprising administering (a) an immune checkpoint inhibitor and (b) a compound of any one of claims 1-38, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition of claim 39.

55. A method of increasing the level of FGF21 expression in an individual comprising administering to the individual a compound of any one of claims 1 -38, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition of claim 39.

56. The method of claim 55, further comprising administering an inducer of FGF21 expression

57 The method of claim 56, wherein the inducer of FGF21 expression is PPARa agonist.

58. The method of claim 57, wherein the PPARa agonist is fibrate or fenofibrate.

59. The composition of claim 39 for use as a human or veterinary medicament.

60. Use of a compound of any one of claims 1-38, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition of claim 39, in the manufacture of a medicament for the prevention and/or treatment of a disorder or disease mediated by FAP.

Description:
INHIBITORS OF FIBROBLAST ACTIVATION PROTEIN

CROSS-REFERENCE TO RELATED APPLICATIONS [QQQ1] This application claims priority benefit of United States Provisional Patent Application No. 62/599,630, filed December 15, 2017, the disclosure of which is incorporated herein by reference in its entirety.

FIELD OF THE INVENTION

[0002] The present disclosure relates generally to therapeutic agents that may be useful in modulating fibroblast activation protein.

BACKGROUND

[0003] Fibroblast activation protein (FAP), also referred to as FAPa, Seprase or

<x2-antiplasmin converting enzyme, is a Ape II integral membrane serine protease that belongs to the prolyl oligopeptidase family S9, winch also includes DPPII, DPPiV, DPP8, DPP9, and PREP enzymes. This family is characterized for having an exo-dipeptidyl peptidase (DPP) activity. FAP is the only member that also has an endopeptidase activity (Aertgeerts, K., et al J Biol Chem, 2005. 280(20): p. 19441-4). FAP has a high degree of homology with DPPIV. It is mainly found as a cell surface homodimer but it has also been reported to form heterodimers with DPPIV in vivo (O'Brien, P., et al. Biochim Biophys Acta, 2008. 1784(9): p. 1130-45). Purported physiological substrates of FAP endopeptidase activity include a2-antiplasmin, type I collagen, gelatin, and Fibroblast growth factor 21 (FGF21) (Lee, K.N., et al., Biochemistry,

2009. 48(23): p. 5149-58), and for the exopeptidase activity include Neuropeptide Y, B-type natriuretic peptide, substance P and peptide YY (Brokopp, C.E., et al., Eur Heart J, 2011.

32(21): p. 2713-22: Coppage, A.L., et al., PLoS One, 2016. 11(3): p. e0151269; Dunshee, D.R., et al., J Biol Chem, 2016. 291(1 1): p. 5986-96; Lee, K.N., et al., J Thromb Haemost, 2011. 9(5): p. 987-96).

[0004] FAP has been implicated in diseases involving proliferation, tissue remodeling, chronic inflammation and/or fibrosis, including but not limited to fibrotic disease, 'ound healing, keloid formation, osteoarthritis, rheumatoid arthritis and related disorders involving cartilage degradation, atherosclerotic disease, and Crohn's disease.

[0005] FAP expression is related to poor prognosis in several types of cancer including gastric cancer, pancreatic adenocarcinoma and hepatocellular carcinoma, (Wen, X., et al, Oncol Res, 2016; Cohen, S.J., et ah, Pancreas, 2008. 37(2); p 154-8; Ju, M.J., et ah, Am J Clin Pathol,

2009. 131(4): p. 498-510) and in colon cancer, increased FAP expression has been associated with a more aggressive disease (Henry, L.R., et al., Clin Cancer Res, 2007. 13(6): p. 1736-41). Purportedly, FAPa on CAFs has critical roles in regulating antitumor immune response by inducing tumor-promoting inflammation (Chen, L., et al., Biochem Biophys Res Common.

2017; Wen, X., et al, Oncol Res, 2016; Hugo, W., et al.. Cell, 2016. 165(1): p. 35-44).

[0006] Yal-boroPro (Talabostat, PT-100) is the only FAP inhibitor that reached clinical stages. This compound was originally developed as a DPPIV inhibitor and subsequently evaluated as a FAP inhibitor regardless of its lack of selectivity (Cunningham, C.C., Expert Opin Investig Drugs, 2007. 16(9): p. 1459-65). This agent was tested in Phase II in a variety of cancers in combination with standard cytotoxic chemotherapy, however endpoints for efficacy were not met (Eager, R.M., et ah, BMC Cancer, 2009. 9: p. 263; Narra, K., et ah, Cancer Biol Ther, 2007 6(11): p. 1691-9; Eager, R.M., et ah, Clin Oncol R Coll Radiol, 2009. 21(6): p. 464-72). Two Phase III trials were early terminated, apparently because of both safety and efficacy concerns (Jansen, K., et ah, 1 Med Chem, 2014 57(7): p. 3053-74). Since Val-boroPro rapidly loses protease inhibitory activity due to cyclization upon standing in pH 7.8, effective concentrations were difficult to achieve in patients given the clinical toxicities seen with this agent at higher doses (Narra, K., et ah. Cancer Biol Ther, 2007. 6(1 1): p. 1691-9).

[0007] There is scope to improve FAP inhibitor selectivity and the properties of the inhibitors to improve safety and efficacy in vivo.

BRIEF SUMMARY

[0008] Provided herein are compounds, salts thereof, pharmaceutical compositions of the foregoing and methods of making and using the same. In one aspect is provided a compound of formula (I):

or a pharmaceutically acceptable salt thereof, wherein: R is hydrogen, Ci-Ce alkyl, C 3 -Cg cycloalkyl, 3- to 12-membered heterocyclyl, 5- to 10- membered heteroaryl, or Ce-Cu aryl, wherein the Cj-Ce alkyl, Cj-Cs cycloalkyl, 3- to 12- membered heterocyclyl, 5- to lO-membered heteroaryl, and Ce-Cn aryl of R are independently optionally substituted by R d ;

m is 0, 1, 2, 3, or 4;

n is 0, l, 2, 3, or 4,

wherein m + n is 1, 2, 3, or 4;

X is -C{ OK -0-, -CH(OH)-, -S-, -S{ OK or -Si Ob¬

L i

* represents the point of attachment to the Y-X- moiety',

** represents the point of attachment to the remainder of the molecule,

R a is hydrogen, Ci-C 6 alkyl, Cs-Cs cycloalkyl, 3- to 12-membered heterocyclyl,

5- to lO-membered heteroaryl, or C 6 -Ci4 aryl, wherein the Ci-Cg alkyl, C 3 -Cg cycloalkyl, 3- to 12-membered heterocyclyl, 5- to 10-membered heteroaryl, and Ce-Cu and of R a are independently optionally substituted by R e ,

R 1 and R , independently of each other and independently at each occurrence, are hydrogen, Ci-C 2 alkyl, C 3 -Cg cycloalkyl, 3- to 12-membered heterocyclyl, 5- to 10- membered heteroaryl, or Ce-Cn aryl, wherein the C 3 -Cg cycloalky-1, 3- to 12-membered heterocyclyl, 5- to 10-membered heteroaryl, and (VC H ary-1 of R 1 and R 2 are independently optionally substituted by R 1 , or R 1 and R 2 are taken together with the carbon atom or atoms to which they are attached to form a 3- to 8-membered

£

cycloaikyiene optionally substituted by R ,

q is 1 , 2, or 3,

R 3 and R 4 , independently of each other and independently at each occurrence, are hydrogen, Cg-Cg cycloalkyl, 3- to 12-membered heterocyclyl, 5- to 10-membered heteroaryl, or Cs-Cu ary-1, wherein the C: < -Cg cycloalkyl, 3- to 12-membered

heterocyclyl, 5- to 10-membered heteroaryl, and Ce-Cn ary-1 of R J and R 4 are independently optionally substituted by R g , or R 3 and R 4 are taken together with the carbon atom to winch they are attached to form a 3- to 8-membered cycloaikyiene optionally substituted by R 8 , and p is 0, 1 , or 2; , wherein

* represents the point of attachment to the Y-X- moiety,

** represents the point of attachment to the remainder of the molecule,

R s and R 6 , independently of each other and independently at each occurrence, are H, C r C 6 alkyl, C 3 -Cg cycloalky], 3- to 1 2-membered heterocyclyl, 5- to 10-membered heteroaryl, or C 6 -Ci4 aryl, wherein the Ci-Cg alkyl, C^-Cg cycloalkyl, 3- to 12-membered heterocyclyl, 5- to 10-membered heteroaryl, and Cg-Cu aryl of R J and R 6 are

independently optionally substituted by R h ,

R b and R c are independently H, CrCe alkyl, CVCg alkenyl, C 2 -C6 alkynyl, C3-C8 cycloalkyl, 3- to 12-membered heterocyclyl, 5- to 10-membered heteroaryl, C6-C14 and, or -C(=0)0R 17 , wherein the Ci-Cg alkyl, C3-C8 cycloalkyl, 3- to 12-membered heterocyclyl, 5- to lO-membered heteroaryl, and Cg-Cn a d of R b and R c are

independently optionally substituted by R 1 , and

r is 1, 2, or 3; or , wherein

* represents the point of attachment to the Y-X- moiety,

** represents the point of attachment to the remainder of the molecule,

R 7 and R 8 , independently of each other and independently at each occurrence, are hydrogen, C Cg cycloalkyl, 3- to 12-membered heterocyclyl, 5- to 10-membered heteroaryl, or Ce-Cu aryl, wherein the C 3 -Cg cycloalkyl, 3- to 12-membered

heterocyclyl, 5- to 10-membered heteroaryl, and Ce-Cty; aryl of R' and R 8 are

independently optionally substituted by R J , or R' and R 8 are taken together with the carbon atom to which they are attached to form a 3- to 8-membered cycloalkylene optionally substituted by R,

R 9 and R 10 , independently of each other and independently at each occurrence, are H, CrCe alkyl, Cs-Cg cycloalkyl, 3- to 12-membered heterocyclyl, 5- to 10- membered heteroaryl, or ( C M aryl, wherein the C -Cg alkyl, C 3 -C 8 cycloalkyl, 3- to 1 2- membered heterocyclyl, 5- to 10-membered heteroaryl, and C 6 -C l4 aryl of R 9 and R 10 are independently optionally substituted by R k ,

s is 1, 2, or 3,

t is 1, 2, or 3,

wherein s + t is 2, 3, or 4,

u is 0 or 1, and

v is 0 or 1;

Y is C 6 -C 9 aryl optionally substituted by R 11 , 6- to 10-membered heteroaryl optionally substituted by R l , or 3- to 12-membered heterocyclyl optionally substituted by R 13 , wherein when Y is phenyl or naphthyl, the phenyl or naphthyl of Y is substituted by at least one R 11 , and wherein when L is *-NH-CH 2 -** and Y is optionally substituted quinolinyl, the optionally substituted quinolinyl of Y is connected to the parent structure at the 2-, 3-, 5-, 6-, 7-, or 8-position, wherein

R n R l2 , and R l , independently of each other and independently at each occurrence, are Ci-Ce alkyl, C 2 -C 6 alkenyl, C 2 -Cg alkynyl, C 3 -C 8 cycloalkyl, C 4 -C 8 cycloalkenyl, 3- to 12- membered heterocyclyl, 5- to 10-membered heteroaryl, C 6 -C M aryl, halogen, cyano, oxo, -OR 14 , -NR 15 R 16 , -SR 34 , -NO 2 , -C=NH(OR 14 ), -C(0)R 14 , -0C(0)R 14 , -C(0)0R 14 , -C(0)NR 15 R 16 , -NR 14 C(0)R 15 , -NR 14 C(0)0R 1s , -NR 14 C(0)NR 15 R 16 , -S(0)R 14 , -S(0) 2 R 14 , -NR I4 S(0)R 15 , -NR 14 S(0) 2 R !5 , -S(0)NR 15 R 16 , -S(0) 2 NR I5 R 16 , or -P(0)(0R !5 )(0R 16 ), wherein each R n R 12 , and R 13 is independently optionally substituted by R L ;

each R 14 is independently hydrogen, Ci-Ce alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 - C 8 cycloalkyl, C 6 -C 14 aryl, 5- to 10-membered heteroaryl, or 3- to 12-membered heterocyclyl, wherein the Ci-Cg alkyl, C 2 -Cg alkenyl, C 2 -C 6 alkynyl, C -C 8 cycloalkyl, Ce-Cu aryl, 5- to 10- membered heteroaryl, and 3- to 12-membered heterocyclyl of R 14 are independently optionally substituted by halogen, -OH, oxo, cyano, or C -Cg alkyl optionally substituted by halogen, -OH, or oxo;

R 35 and R 16 , independently of each other and independently at each occurrence, are hydrogen, Ci-Ce alkyl, C 2 -C 6 alkenyl, C 2 -Cg alkynyl, C 3 -C 8 cycloalkyl, Cg-Cu aryl, 5- to 10- membered heteroaryl, or 3- to 12-membered heterocyclyl, wherein the Ci-Cg alkyl, C 2 -Cg alkenyl, C 2 -Cg alkynyl, C 3 -C 8 cycloalkyl, Cg-C 4 aryl, 5- to 10-membered heteroaryl, and 3- to 12-membered heterocyclyl of R 31 and R 36 are independently optionally substituted by halogen, - OH, oxo, cyano, or Ci-Cg alkyl, optionally substituted by halogen, -OH, or oxo, or R i 5 and R 16 are taken together with the atom to which they are attached to form a 3- to 6-membered heterocyclyl optionally substituted by halogen, oxo, cyano, or Ci-Cc, alkyl optionally substituted by halogen, -OH, or oxo; and

R d , R ® , R f , R 8 , R h , R , R J , and R k , independently of each other and independently at each occurrence, are halogen, C -Ce alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, Cj-C» cycloalkyl, C 6 -C 14 a ryl, 5- to lO-membered heteroaryl, 3- to 12-membered heterocyclyl, -OR 14 , -NR l5 R 16 , cyano, or nitro; and

each R L is independently halogen, Ci-Ce alkyl, alkenyl, C 2 -C 6 alkynyl, C 3 -C 8 cycloalkyl, C ¾ -C i 4 axyl, 5- to 10-membered heteroay , 3- to 12-membered heterocyclyl, -OR 14 , ~C(0)R 14 , - NR 15 R 16 , cyano, oxo, or nitro.

[0009] In one aspect, provided is a compound of formula (I) or a pharmaceutically acceptable salt thereof, wherein the compound has any one or more of the following features:

and

R z are taken together with the carbon atom to which they are attached to form a 3- to 8-membered eycloaikylene such as a cyclopropylene;

(b) -CR s R°-CH(NR b R c )- , including but not limited to, when R u , R b , and R are each H, and R 3 is H or Ci-C 6 alkyl; or iCR 7 R 8 ) ¾ — N > - (CR 9 R 10 ) t — ** , . * ,

(c) N / , wherem * represents the point of attachment to the Y-X- moiety, ** represents the point of attachment to the remainder of the molecule, such as is:

(a) C 6 -C 9 aryl optionally substituted by R 11 , such as unsubstituted 2,3- dihydro-li -inden-2-yl or a phenyl or naphthyl substituted by at least one R 11 , including but not limited to when each R 11 is independently selected from halogen, tribal ome thy 1, cyano, and - C(=0)NH 2 ; (b) 6- to 10-membered heteroaryl optionally substituted by R , such as a pyridinyl or pyrimidinyl substituted by at least one if’: or

(c) 3- to 12-membered heterocyclyl optionally substituted by R 13 , such as a piperidinyl substituted by at least one R 13 .

[0010] In another aspect is provided a compound of formula (I), or a salt thereof, wherein the

X-L- moiety is selected from the group consisting of O 0 CH 3

nts the point of attachment to the Y moiety, and ** represents the point of attachment to the remainder of the molecule.

[0011] Also provided is a pharmaceutical composition comprising a compound of any formula herein, including formula (I), or a pharmaceutically acceptable salt thereof, and a

pharmaceutically acceptable carrier.

[0012] Also provided is a method of treating a disease or disorder mediated by fibroblast activation protein (FAP) in an individual in need thereof comprising administering to the individual a therapeutically effective amount of a compound as detailed herein, including but not limited to a compound of formula (I), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising such compound or salt. Such disease or disorder in one aspect is characterized by proliferation, tissue remodeling, chronic inflammation, obesity, glucose intolerance, or insulin insensitivity. In one aspect, the disease or disorder is breast cancer, colorectal cancer, ovarian cancer, prostate cancer, pancreatic cancer, kidney cancer, lung cancer, melanoma, fibrosarcoma, bone sarcoma, connective tissue sarcoma, renal cell carcinoma, giant cell carcinoma, squamous cell carcinoma, leukemia, skin cancer, soft tissue cancer, liver cancer, gastrointestinal carcinoma, or adenocarcinoma. In a particular aspect, the disease or disorder is metastatic kidney cancer, chronic lymphocytary leukemia, pancreatic adenocarcinoma, or non-small cell lung cancer. In a further aspect, the disease or disorder is a fibrotic disease, wound healing, keloid formation, osteoarthritis, rheumatoid arthritis and related disorders involving cartilage degradation, atherosclerotic disease, Crohn's disease, or Type II diabetes. In another particular aspect is provided a method of reducing tumor growth, tumor proliferation, or tumoiigenicity in an individual in need thereof, comprising administering to the individual a compound as detailed herein, such as a compound of formula (I), or a

pharmaceutically acceptable salt thereof, or a pharmaceutical composition of the foregoing.

BRIEF DESCRIPTION OF THE DRAWINGS

[0013] FIG. 1 A shows PRXS-AMC degradation over time by rhFAP. FIG. IB shows Z-Gly- Pro-AMC degradation over time by rhFAP.

[0014] FIG. 2A shows PRXS-AMC degradation over time by rhPREP. FIG. 2B shows Z-Gly- Pro-AMC degradation over time by rhPREP.

[0015] FIG. 3A shows PRXS-AMC degradation over time by rhDPPIV. FIG. 3B shows PRXS-AMC degradation over time by rhDPP9.

[0016] FIG. 4 shows FAPoc activity in plasma of mice dosed with compound 13 after various time points post- administration.

[0017] FIG. 5 shows DPPIV activity in plasma of mice dosed with compound 13 after various time points post-administration.

[0018] FIG. 6A shows FAP activity in plasma of mice dosed with an exemplary compound after various time points post-administration. FIG. 6B show's DPPIV activity' in plasma of mice dosed with an exemplary' compound after various time points post- administration. FIG. 6C show's a comparison of FAP acti vity over time and the compound exposure as measured m in plasma of mice dosed with an exemplary compound.

[0019] FIG. 7 show's the volume of a MC38 colon cancer cell line-derived xenograft tumor in C57/BL6 mice after oral administration (on day 7) of test compound 24 (50 mg/kg, twice daily) or a vehicle control. FIG. 8 show's body weight gam of the C47/BL6 mice with the MC38 colon cancer cell line-derived xenograft tumor after administration (day 7) of test compound 24 (50 mg/kg, twice daily) or a vehicle control. FIG. 9 show's indi vidual recordings of tumor volume when mice were treated with test compound 24 or a control vehicle FIG. 10 shows mean and standard error of mean (SEM) of th e total mass of th e tumors when mice were treated with test compound 24 or a control vehicle.

[0020] FIG. 11 A show's the concentration of an exemplary compound in the plasma and tumor of mice bearing a B16-F 10 tumor, which were administered vehicle or compound 13 (an exemplary compound). FIG. 1 IB show's the FAP activity in the plasma and tumor of mice bearing a B16-F10 tumor, w'hich were administered vehicle or compound 13 (an exemplary' compound).

[QQ21] FIG. 12 A shows the concentration of an exemplary' compound in the plasma and tumor of mice bearing a MC38 tumor, which were administered vehicle or compound 13 (an exemplary compound). FIG. 12B shows the FAP activity in die plasma and tumor of mice bearing a MC38 tumor, which were administered vehicle or compound 13 (an exemplary compound).

[QQ22] FIG. 13A shows an image of an anti-FGF2l Western Blot demonstrating degradation of rhFGF21 by rhFAP. FIG. 13B show's the densitometry analysis of the Western Blot shown in FIG. 13A. FIG. 13C shows an image of an anti-FGF2l Western Blot demonstrating compound 13 (an exemplary' compound)-mediated inhibition of the degradation of rhFGF21 by rhFAP.

FIG. 13D shows the densitometry' analysis of the Western Blot shown in FIG. I3C.

[0023] FIG. 14A show's an image of an anti-FGF21 Western Blot demonstrating compound 13 (an exemplary compound)-rnediated inhibition of the degradation of rhFGF21 by rhFAP in a dose-dependent manor. FIG. 14B shows the densitometry analysis of the Western Blot shown in FIG. 14A and the ICso determined from the resulting curve.

[0024] FIG. 15 shows a summary of experimental protocol for orally administering vehicle or compound 13 (an exemplary' compound) to rats and subsequently administering rhFGF21 intravenously and also show's resulting anti-FGF21 Western Blots, FAP activity, and plasma exposure levels over time.

[0025] FIG. 16 show's FAP activity and FGF21 levels over time in hamsters after intravenous administration of compound 13 (an exemplary' compound).

[QQ26] Described herein are compounds according to formula (I):

and pharmaceutically acceptable salts thereof. The compounds can be useful for inhibiting fibroblast activation protein (FAPoc). In certain embodiments, the compound is used to treat a disease or a disorder mediated by FAPoc in an individual. Such diseases or disorders can include or be characterized by proliferation, tissue remodeling, chronic inflammation, obesity, glucose intolerance, and/or insulin insensitivity. In some embodiments, the compound is used to treat cancer.

Definitions

[0027] For use herein, unless clearly indicated otherwise, use of the terms“a”,“an” and the like refers to one or more.

[0028] Reference to“about” a value or parameter herein includes (and describes)

embodiments that are directed to that value or parameter per se. For example, description referring to“about X” includes description of“X”.

[QQ29] “Alkyl” as used herein refers to and includes, unless otherwise stated, a saturated linear (i.e., unbranched) or branched univalent hydrocarbon chain or combination thereof, having the number of carbon atoms designated (i.e., C -Cio means one to ten carbon atoms). Particular alkyl groups are those having 1 to 20 carbon atoms (a“C1-C20 alkyl”), having 1 to 10 carbon atoms (a “Ci-Cio alkyl”), having 6 to 10 carbon atoms (a“C 6 -Cio alkyl”), having 1 to 6 carbon atoms (a “Ci-Ce alkyl”), having 2 to 6 carbon atoms (a“C2-C6 alkyl”), or having 1 to 4 carbon atoms (a “C 1 -C 4 alkyl”). Examples of alkyl groups include, but are not limited to, groups such as methyl, ethyl, n-propyl, isopropyl, n-butyl, t-butyl, isobutyl, sec-butyl, n-pentyl, n-hexyl, n-heptyl, n- octy!, n-nonyi, n-decyl, and the like. [QQ3Q] “Alkylene” as used herein refers to the same residues as alkyl, but having bivalency. Particular alkylene groups are those having 1 to 20 carbon atoms (a“C1-C20 alkylene”), having 1 to 10 carbon atoms (a“Ci-C -o alkylene”), having 6 to 10 carbon atoms (a“Ce-C o alkylene”), having 1 to 6 carbon atoms (a“Ci-C 6 alkylene”), 1 to 5 carbon atoms (a“C 1 -C 5 alkylene”), 1 to 4 carbon atoms (a“C1-C4 alkylene”) or 1 to 3 carbon atoms (a“C1-C3 alkylene”). Examples of alkylene include, but are not limited to, groups such as methylene (-CH -), ethylene (-CH 2 CH 2 -), propylene (-CH 2 CH 2 CH 2 -), isopropylene (-CH 2 CH(CH 3 )-), butylene (-CH 2 (CH 2 ) 2 CH 2 -), isobutylene (-CH 2 CH(CH 3 )CH 2 -), pentylene (-CH 2 (CH 2 ) 3 CH 2 -), hexylene (-CH 2 (CH 2 ) 4 CH 2 -), heptylene ί-ί 1 1 ί( 1 I h( I I . ·-). octyiene (-CH 2 (CH 2 ) 6 CH 2 -), and the like.

[0031] “Alkenyl” as used herein refers to and includes, unless otherwise stated, an unsaturated linear (i.e., unbranched) or branched univalent hydrocarbon chain or combination thereof, having at least one site of olefmic unsaturation (i.e., having at least one moiety of the formula C=C) and havmg the number of carbon atoms designated (i.e., C 2 -C]o means two to ten carbon atoms). An alkenyl group may have“as” or“trans” configurations, or alternatively have“E” or “Z” configurations. Particular alkenyl groups are those having 2 to 20 carbon atoms (a“C 2 -C 2o alkenyl”), having 6 to 10 carbon atoms (a“C 6 -Cio alkenyl”), having 2 to 8 carbon atoms (a“C 2 - Cg alkenyl”), having 2 to 6 carbon atoms (a“C 2 -C 6 alkenyl”), or having 2 to 4 carbon atoms (a “C 2 -C 4 alkenyl”). Examples of alkenyl group include, but are not limited to, groups such as ethenyl (or vinyl), prop-l-enyl, prop-2-enyl (or allyl), 2-methylprop-l-enyl, but-l-enyl, but-2- enyl, but-3-enyl, buta-l,3-dienyl, 2-methylbuta-l,3-dienyl, pent-l-enyl, pent-2-enyl, hex-l-enyl, hex-2-enyl, hex-3-enyl, and the like.

[0032] “Alkynyl” as used herein refers to and includes, unless otherwise stated, an unsaturated linear (i.e., unbranched) or branched univalent hydrocarbon chain or combination thereof, having at least one site of acetylenic unsaturation (i.e., having at least one moiety of the formula CºC) and having the number of carbon atoms designated (i.e., C 2 -C lo means two to ten carbon atoms). Particular alkynyl groups are those having 2 to 20 carbon atoms (a“C 2 -C 2 o alkynyl”), having 6 to 10 carbon atoms (a“C 6 -Cio alkynyl”), having 2 to 8 carbon atoms (a“C 2 -Cg alkynyl”), having 2 to 6 carbon atoms (a“C 2 -C 6 alkynyl”), or having 2 to 4 carbon atoms (a“C 2 - C4 alkynyl”). Examples of alkynyl group include, but are not limited to, groups such as ethynyl (or acetyl enyl), prop-l-ynyl, prop-2-ynyl (or propargyl), but-l-ynyl, but-2-ynyl, but-3-ynyl, and the like. [QQ33] “Cycloalkyl” as used herein refers to and includes, unless otherwise stated, saturated cyclic univalent hydrocarbon structures, having the number of carbon atoms designated (i.e., C 3 - Cio means three to ten carbon atoms). Cycloalkyl can consist of one ring, such as cyclohexyl, or multiple rings, such as adamantyl A cycloalkyl comprising more than one ring may be fused, spiro or bridged, or combinations thereof. Particular cycloalkyl groups are those having from 3 to 12 annular carbon atoms. A preferred cycloalkyl is a cyclic hydrocarbon having from 3 to 8 annular carbon atoms (a "Cg-Cg cycloaikyl"), having 3 to 6 carbon atoms (a“Cg-C 6 cycloaikyl”), or having from 3 to 4 annular carbon atoms (a "C3-C4 cycloaikyl"). Examples of cycloalkyl include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cyclohepty!, norhornyi, and the like.

[0034] “Cycloalkylene” as used herein refers to the same residues as cycloaikyl, but having bivalency. Cycloalkylene can consist of one ring or multiple rings which may be fused, spiro or bridged, or combinations thereof. Particular cycloalkylene groups are those having from 3 to 12 annular carbon atoms. A preferred cycloalkylene is a cyclic hydrocarbon having from 3 to 8 annular carbon atoms (a "Cg-Cg cycloalkylene"), having 3 to 6 carbon atoms (a (VC,, cycloalkylene”), or having from 3 to 4 annular carbon atoms (a "C3-C4 cycloalkylene").

Examples of cycloalkylene include, but are not limited to, cyclopropylene, cyclobutylene, cyclopentylene, cyclohexylene, cycloheptylene, norbornylene, and the like. A cycloalkylene may attach to the remaining structures via the same ring carbon atom or different ring carbon atoms. When a cycloalkylene attaches to the remaining structures via two different ring carbon atoms, the connecting bonds may be cis- or trans- to each other. For example, cyclopropylene may include 1,1 -cyclopropylene and 1,2-cyclopropylene (e.g., cis~l,2-cyclopropylene or trans- 1, 2-cyclopropylene), or a mixture thereof

[0035] “Cycloalkenyl” refers to and includes, unless otherwise stated, an unsaturated cyclic non-aromatic univalent hydrocarbon structure, having at least one site of olefmic unsaturation (i.e., having at least one moiety of the formula 0 : C) and having the number of carbon atoms designated (i.e., C3-C10 means three to ten carbon atoms). Cycloalkenyl can consist of one ring, such as cyclohexenyl, or multiple rings, such as norbomenyl. A preferred cycloalkenyl is an unsaturated cyclic hydrocarbon having from 3 to 8 annular carbon atoms (a ( W

cycloalkenyl”). Examples of cycloalkenyl groups include, but are not limited to, cyclopropenyl, cyc!obuteny!, cyclopenteny!, cyclohexenyl, norbomenyl, and the like. [QQ36] “Cycloalkenylene” as used herein refers to the same residues as cycloalkenyl, hut having hi valency

[0037] “An or“Af’ as used herein refers to an unsaturated aromatic carbocyclic group having a single ring (e.g., phenyl) or multiple condensed rings (e.g., naphthyl or anthryl) which condensed rings may or may not be aromatic. Particular aryl groups are those having from 6 to 14 annular carbon atoms (a“C 6 -Ci4 aryl”). An aryl group having more than one ring where at least one ring is non-aromatic may be connected to the parent structure at either an aromatic ring position or at a non-aromatic ring position. In one variation, an aryl group having more than one ring where at least one ring is non-aromatic is connected to the parent structure at an aromatic ring position.

[0038] “Arylene” as used herein refers to the same residues as aryl, hut having bivalency. Particular arylene groups are those having from 6 to 14 annular carbon atoms (a“C 6 -Ci4 arylene”)

[QQ39] ‘Heteroaryl” as used herein refers to an unsaturated aromatic cyclic group having from 1 to 14 annular carbon atoms and at least one annular heteroatom, including but not limited to heteroatoms such as nitrogen, oxygen and sulfur. A heteroaryl group may have a single ring (e.g., pyridy!, fury!) or multiple condensed rings (e.g. , indohziny!, henzothienyl) which condensed rings may or may not be aromatic. Particular heteroaryl groups are 5 to 14-membered rings having 1 to 12 annular carbon atoms and 1 to 6 annular heteroatoms independently selected from nitrogen, oxygen and sulfur, 5 to 10-membered rings having 1 to 8 annular carbon atoms and 1 to 4 annular heteroatoms independently selected from nitrogen, oxygen and sulfur, or 5, 6 or 7-membered rings having 1 to 5 annular carbon atoms and 1 to 4 annular heteroatoms independently selected from nitrogen, oxygen and sulfur. In one variation, particular heteroaryl groups are monocyclic aromatic 5-, 6- or 7-membered rings having from 1 to 6 annular carbon atoms and 1 to 4 annular heteroatoms independently selected from nitrogen, oxygen and sulfur. In another variation, particular heteroaryl groups are polycyclic aromatic rings having from 1 to 12 annular carbon atoms and 1 to 6 annular heteroatoms independently selected from nitrogen, oxygen and sulfur. A heteroaryl group having more than one ring where at least one ring is non- aromatic may be connected to the parent structure at either an aromatic ring position or at a non aromatic ring position. In one variation, a heteroaryl group having more than one ring where at least one ring is non-aromatic is connected to the parent structure at an aromatic ring position. A heteroaryl group may be connected to the parent structure at a ring carbon atom or a ring heteroatom.

[0040] Where applicable, a heteroaryl group may be depicted in a tautomeric form. Such compounds would be considered to he heteroaryl even if certain tautomeric forms are, for

example, heterocyclyl. For example, the heteroaryl group OH may be depicted in the

heterocyclic tautomeric form O . Regardless of which tautomer is shown, the group is considered to be heteroaryl.

[0041] “Heterocycle”,“heterocyclic”, or“heterocyclyl” as used herein refers to a saturated or an unsaturated non-aromatic cyclic group having a single ring or multiple condensed rings, and having from 1 to 14 annular carbon atoms and from 1 to 6 annular heteroatoms, such as nitrogen, sulfur or oxygen, and the like. A heterocycle comprising more than one ring may be fused, bridged or spiro, or any combination thereof, but excludes heteroaryl groups. The heterocyclyl group may he optionally substituted independently with one or more substituents described herein. Particular heterocyclyl groups are 3 to 14-membered rings having 1 to 13 annular carbon atoms and 1 to 6 annular heteroatoms independently selected from nitrogen, oxygen and sulfur,

3 to 12-membered rings having 1 to 11 annular carbon atoms and 1 to 6 annular heteroatoms independently selected from nitrogen, oxygen and sulfur, 3 to lO-membered rings having 1 to 9 annular carbon atoms and 1 to 4 annular heteroatoms independently selected from nitrogen, oxygen and sulfur, 3 to 8-membered rings having 1 to 7 annular carbon atoms and 1 to 4 annular heteroatoms independently selected from nitrogen, oxygen and sulfur, or 3 to 6-membered rings having 1 to 5 annular carbon atoms and 1 to 4 annular heteroatoms independently selected from nitrogen, oxygen and sulfur. In one variation, heterocyclyl includes monocyclic 3-, 4-, 5-, 6- or 7-membered rings having from 1 to 2, 1 to 3, 1 to 4, 1 to 5, or 1 to 6 annular carbon atoms and 1 to 2, 1 to 3, or 1 to 4 annular heteroatoms independently selected from nitrogen, oxygen and sulfur. In another variation, heterocyclyl includes polycyclic non-aromatic rings having from 1 lo 12 annular carbon atoms and 1 to 6 annular heteroatoms independently selected from nitrogen, oxygen and sulfur.

[0042] “Halo” or“halogen” refers to elements of the Group 17 series having atomic number 9 to 85. Preferred halo groups include the radicals of fluorine, chlorine, bromine and iodine.

Where a residue is substituted with more than one halogen, it may be referred to by using a prefix corresponding to the number of halogen moieties attached, e.g., dihaloary 1, dihaloalkyl, trihaloaryl etc. refer to aryl and alkyl substituted with two (“di”) or three (“tri”) halo groups, which may be but are not necessarily the same halogen; thus 4-chloro-3-fiuorophenyl is within the scope of dihaloaryl. An alkyl group in which each hydrogen is replaced with a halo group is referred to as a“perhaloalkyl.” A preferred perhaloalkyl group is trifluoromethyl (-CF 3 ).

Similarly,“perhaloalkoxy” refers to an a!koxy group in which a halogen takes the place of each H in the hydrocarbon makin up the alkyl moiety of the alkoxy group. An example of a perhaloalkoxy group is trifluoromethoxy (-OCF 3 ).

[QQ43] “Carbonyl” refers to the group C=0.

[0044] “Oxo” refers to the moiety =0.

[0045] “Optionally substituted” unless otherwise specified means that a group may be unsubstituted or substituted by one or more (e.g. , 1, 2, 3, 4 or 5) of the substituents listed for that group in which the substituents may be the same of different. In one embodiment, an optionally substituted group has one substituent. In another embodiment, an optionally substituted group has twO substituents. In another embodiment, an optionally substituted group has three substituents. In another embodiment, an optionally substituted group has four substituents. In some embodiments, an optionally substituted group has 1 to 2, 1 to 3, 1 to 4, 1 to 5, 2 to 3, 2 to 4, or 2 to 5 substituents. In one embodiment, an optionally substituted group is unsubstituted.

[QQ46] Unless clearly indicated otherwise,“an individual” as used herein intends a mammal, including but not limited to a primate, human, bovine, horse, feline, canine, or rodent. In one variation, the individual is a human.

[0047] As used herein,“treatment” or“treating” is an approach for obtaining beneficial or desired results including clinical results. Beneficial or desired results include, but are not limited to, one or more of the follow ing: decreasing one more symptoms resulting from the disease, diminishing the extent of the disease, stabilizing the disease (e.g., preventing or delaying the worsening of the disease), preventing or delaying the spread of the disease, delaying the occurrence or recurrence of the disease, delay or sl owing the progression of the disease, ameliorating the disease state, providing a remission (whether partial or total) of the disease, decreasing the dose of one or more other medications required to treat the disease, enhancing effect of another medication, delaying the progression of the disease, increasing the quality of life, and/or prolonging survival. The methods described herein contemplate any one or more of these aspects of treatment.

[QQ48] As used herein, the term '‘effective amount” intends such amount of a compound of the invention which should be effective in a given therapeutic form. As is understood in the art, an effective amount may be in one or more doses, i.e., a single dose or multiple doses may be required to achieve the desired treatment. An effective amount may be considered in the context of administering one or more therapeutic agents, and a single agent may be considered to be given in an effective amount if, in conjunction with one or more other agents, a desirable or beneficial result may be or is achieved. Suitable doses of any of the co-administered compounds may optionally be lowered due to the combined action (e.g., additive or synergistic effects) of the compounds.

[0049] A“therapeutically effective amount” refers to an amount of a compound or salt thereof sufficient to produce a desired therapeutic outcome.

[0050] As used herein,“unit dosage form” refers to physically discrete units, suitable as unit dosages, each unit containing a predetermined quantity' of active ingredient calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier. Unit dosage forms may contain a single or a combination therapy.

[0051] As used herein, by“pharmaceutically acceptable” or“pharmacologically acceptable” is meant a material that is not biologically or otherwise undesirable, e.g , the material may be incorporated into a pharmaceutical composition administered to a patient without causing any significant undesirable biological effects or interacting in a deleterious manner with any of the other components of the composition in which it is contained. Pharmaceutically acceptable carriers or excipients have preferably met the required standards of toxicological and manufacturing testing and/or are included on the Inactive Ingredient Guide prepared by the U.S. Food and Drug administration. [QQ52] “Pharmaceutically acceptable salts’' are those salts which retain at least some of the biological activity of the free (non-salt) compound and which can be administered as drugs or pharmaceuticals to an individual. Such salts, for example, include: (1) acid addition salts, formed with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like; or formed with organic acids such as acetic acid, oxalic acid, propionic acid, succinic acid, maleic acid, tartaric acid and the like; (2) salts formed when an acidic proton present in the parent compound either is replaced by a metal ion, e.g., an alkali metal ion, an alkaline earth ion, or an aluminum ion; or coordinates with an organic base.

Acceptable organic bases include ethano!amine, diethanolamine, triethanolamine and the like. Acceptable inorganic bases include aluminum hydroxide, calcium hydroxide, potassium hydroxide, sodium carbonate, sodium hydroxide, and the like. Pharmaceutically acceptable salts can be prepared in situ in the manufacturing process, or by separately reacting a purified compound of the invention in its free acid or base form with a suitable organic or inorganic base or acid, respectively, and isolating the salt thus formed during subsequent purification.

[QQ53] The term“excipient” as used herein means an inert or inactive substance that may be used in the production of a drug or pharmaceutical, such as a tablet containing a compound of the invention as an active ingredient. Various substances may be embraced by the term excipient, including without limitation any substance used as a binder, dismtegrant, coating, compression/encapsulation aid, cream or lotion, lubricant, solutions for parenteral

administration, materials for chewable tablets, sweetener or flavoring, suspending/gelling agent, or wet granulation agent. Binders include, e.g., carbomers, povidone, xanthan gum, etc.; coatings include, e.g, cellulose acetate phthalaie, ethylcellulose, gellan gum, maltodextrin, enteric coatings, etc.; compression/encapsulation aids include, e.g. , calcium carbonate, dextrose, fructose dc (dc =“directly compressible”), honey dc, lactose (anhydrate or monohydrate;

optionally in combination with aspartame, cellulose, or microcrystalline cellulose), starch dc, sucrose, etc.; disintegrants include, e.g., croscarmellose sodium, gellan gum, sodium starch glycolate, etc.; creams or lotions include, e.g., maltodextrin, carrageenans, etc.; lubricants include, e.g., magnesium stearate, stearic acid, sodium stearyl fumarate, etc.; materials for chewable tablets include, e.g., dextrose, fructose dc, lactose (monohydrate, optionally in combination with aspartame or cellulose), etc.; suspending/gelling agents include, e.g. , carrageenan, sodium starch glycolate, xanthan gum, etc.; sweeteners include, e.g., aspartame, dextrose, fructose dc, sorbitol, sucrose dc, etc.; and wet granulation agents include, e.g., calcium carbonate, maltodextrin, microcrystalline cellulose, etc.

[0054] It is understood that aspects and embodiments described herein as‘comprising” include“consisting of” and“consisting essentially of’ embodiments.

[0055] When a composition is described as“consisting essentially of’ the listed components, the composition contains the components expressly listed, and may contain other components which do not substantially affect the disease or condition being treated such as trace impurities. However, the composition either does not contain any other components which do substantially affect the disease or condition being treated other than those components expressly listed; or, if the composition does contain extra components other than those listed which substantially affect the disease or condition being treated, the composition does not contain a sufficient concentration or amount of those extra components to substantially affect the disease or condition being treated. When a method is described as“consisting essentially of the listed steps, the method contains the steps listed, and may contain other steps that do not substantially affect the disease or condition being treated, but the method does not contain any other steps which substantially affect the disease or condition being treated other than those steps expressly listed.

[0056] When a moiety' is indicated as substituted by‘at least one” substituent, this also encompasses the disclosure of exactly one substituent.

Compounds

[0057] in one aspect, provided is a compound of formula (I):

or a pharmaceutically acceptable salt thereof, wherein:

R is hydrogen, C -Ce alkyl, CrCa cycloalkyl, 3- to 12-membered heterocyclyl, 5- to 10- membered heteroaryl, or Ce-Cu and, wherein the C1-C0 alkyl, Cs-Cg cycloalkyl, 3- to 12- membered heterocyclyl, 5- to 10-membered heteroaryl, and Cg-Cty aryl of R are independently optionally substituted by R d ; m is 0, 1, 2, 3, or 4; n is 0, 1, 2, 3, or 4, wherein m + n is 1, 2. 3. or 4:

X is -C(=0)-, -0-, -CH(OH)-, -S-, -S(=0)-, or -S(=0) 2 -;

L is . wherein

* represents the point of attachment to the Y-X- moiety,

** represents the point of attachment to the remainder of the molecule,

R a is hydrogen, C -Cg alkyl, Cg-Cg cycloalkyl, 3- to 12-membered heterocyclyl,

5- to lO-membered heteroaryl, or C 6 -C 14 and, wherein the C -C 6 alkyl, C 3 -Cg cycloalkyl, 3- to 12-membered heterocyclyl, 5- to 10-membered heteroaryl, and C 6 -C 14 aryl of R a are independently optionally substituted by R c ,

R 1 and R 2 , independently of each other and independently at each occurrence, are hydrogen, C]-C 2 alkyl, C 3 -Cg cycloalkyl, 3- to 12-membered heterocyclyl, 5- to 10-membered heteroaryl, or Cg-Cn aryl, wherein the C 3 -C 8 cycloalkyl, 3- to 12- membered heterocyclyl, 5- to 10-membered heteroaryl, and Cg~Ci 4 aryl of R 1 and R 2 are independently optionally substituted by R , or R 1 and R 2 are taken together with the carbon atom or atoms to which they are attached to form a 3- to 8-membered cycloalkylene optionally substituted by R f , q is 1, 2, or 3, R 3 and R 4 , independently of each other and independently at each occurrence, are hydrogen, C -Cg cycloalkyl, 3- to 12-membered heterocyclyl, 5- to 10-membered heteroaryl, or Ce-Cn aryl, wherein the C 3 -C 8 cycloalkyl, 3- to 12-membered heterocyclyl, 5- to 10-membered heteroaryl, and C 6 -C l4 aryl of R J and R 4 are independently optionally substituted by R g , or R 3 and R 4 are taken together with the carbon atom to which they are attached to form a 3- to 8-membered cydoalkylene optionally substituted by R 8 , and p is 0, 1 , or 2; wherein

* represents the point of attachment to the Y-X- moiety,

** represents the point of attachment to the remainder of the molecule,

R 1 and R 6 , independently of each other and independently at each occurrence, are H, Ci-Cty alkyl, C 3 -C 8 cycloalkyl, 3- to 12-membered heterocyclyl, 5- to 10- mernbered heteroaryl, or C 6 -C l4 aryl, wherein the (ty-Ce alkyl, C 3 -Cs cycloalkyl, 3- to 12-membered heterocyclyl, 5- to 10-membered heteroaryl, and Ce-Cw and of R 5 and R 6 are independently optionally substituted by R n ,

R b and R c are independently H, C-.-Ce alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -Cg cycloalkyl, 3- to 12-membered heterocyclyl, 5- to lO-me bered heteroaryl, C 6 - C 14 aryl, or -C(=0)OR 17 , wherein the Ci-Ce alkyl, C 3 -Cg cycloalkyl, 3- to 12- membered heterocyclyl, 5- to 10-membered heteroaryl, and Ce-Cu and of R b and R c are independently optionally substituted by R 1 , and r is 1, 2, or 3; or

* represents the point of attachment to the Y-X- moiety,

** represents the point of attachment to the remainder of the molecule,

R 7 and R 8 , independently of each other and independently at each occurrence, are hydrogen, C- r Cs cycloalkyl, 3- to 12-membered heterocyclyl, 5- to 10-membered heteroaiyl, or C 6 -Ci 4 aryl, wherein the Ch-Cg cycloalkyl, 3- to 12-membered heterocyclyl, 5- to 10-membered heteroaryl, and C 6 -C l4 aryl of R' and R 8 are independently optionally substituted by R J , or R ' and R 8 are taken together with the carbon atom to which they are attached to form a 3- to 8-membered cycloalkylene optionally substituted by R 1 ,

R 9 and R 3u , independently of each other and independently at each occurrence, are H, Ci-C 6 alkyl, Ci-C* cycloalkyl, 3- to 12-membered heterocyclyl, 5- to 10- membered heteroaiyl, or Ce-C^ aryl, wherein the Ci-CV, alkyl, C 3 -C 8 cycloalkyl, 3- to 12-membered heterocyclyl, 5- to 10-membered heteroaiyl, and C 6 -Ci 4 aryl of R 9 and R 10 are independently optionally substituted by R k , s is 1 , 2, or 3, t is 1, 2, or 3, wherein s + t is 2, 3, or 4, u is 0 or 1, and v is 0 or 1 ;

Y is C 6 -C 9 aryl optionally substituted by R 33 , 6- to 10-membered heteroaiyl optionally substituted by R’ 2 , or 3- to 12-membered heterocyclyl optionally substituted by R 13 , wherein when Y is phenyl or naphthyl, the phenyl or naphthyl of Y is substituted by at least one R 11 , and wherein when L is *-NH-CH 2 -** and Y is optionally substituted quinolinyl, the optionally substituted quinolinyl of Y is connected to the parent structure at the 2-, 3-, 5-, 6~, 7-, or 8~ position, wherein R n R l2 , and R l , independently of each other and independently at each occurrence, are C -Ce alkyl, C 2 -Ce alkenyl, C 2 -C6 alkynyl, C3-C8 cycloalkyl, C 4 -Cs cycloalkenyl, 3- to 12-membered heterocyclyl, 5- to 10-membered heteroaryl, C & -C l4 aryl, halogen, cyano, oxo, -OR 14 , ~NR 15 R 16 , -SR 14 , -NO2, -C=NH(OR 14 ), ~C(0)R 14 , -0C(0)R 14 , -C(0)0R 14 , -C(0)NR 1S R 16 , -NR 14 C(0)R 15 , -NR 14 C(0)0R 1s , -NR 14 C(0)NR 1S R 16 , ~S(0)R 14 ,

-S(0) ? R 14 , -NR 14 S(0)R 1s , -NR 14 S(0) 2 R , s , -S(0)NR 1S R 16 , -S(0) 2 NR 15 R 16 ,

or -P(0)(0R 15 )(0R 16 ), wherein each R n R 12 , and R 13 is independently optionally substituted by R L ; each R 14 is independently hydrogen, C 1 -C6 alkyl, C 2 -C 6 alkenyl, C2-C6 alkynyl, C3- Cg cycloalkyl, C 6 -C ]4 aryl, 5- to 10-membered heteroaryl, or 3- to 12-membered heterocyclyl, wherein the Ci-Ce alkyl, C 2 -Ce alkenyl, C 2 -C 6 alkynyl, C3-C8 cycloalkyl, Ce-Ci 4 aryl, 5- to 10- membered heteroaryl, and 3- to 12-membered heterocyclyl of R 4 are independently optionally substituted by halogen, -OH, oxo, cyano, or Ci-C 6 alkyl optionally substituted by halogen, -OH, or oxo;

R 15 and R lb , independently of each other and independently at each occurrence, are hydrogen, Ci-Ce alkyl, C 2 -C 6 alkenyl, C 2 -Ce alkynyl, Cs-Cg cycloalkyl, Ce-Ci 4 aryl, 5- to 10-membered heteroaryl, or 3- to 12-membered heterocyclyl, wherein the Ci-C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -Cs cycloalkyl, C 6 -C l4 aryl, 5- to 10-membered heteroaryl, and 3- to 12-membered heterocyclyl of R : " and R 16 are independently optionally substituted by halogen, -OH, oxo, cyano, or C -Ce alkyl, optionally substituted by halogen, -OH, or oxo, or R 15 and R 16 are taken together with the atom to which they are attached to form a 3- to 6-membered heterocyclyl optionally substituted by halogen, oxo, cyano, or Cj-Ce alkyl optionally substituted by halogen, -OH, or oxo:

R d , R e , R 1 , R 8 , R , R 1 , R j , and R k , independently of each other and independently at each occurrence, are halogen, C -C6 alkyl, C 2~ Ce, alkenyl, C 2 -C 6 alkynyl, Cg-Cg cycloalkyl, C 6 -C l4 aryl, 5- to 10-membered heteroaryl, 3- to 12-membered heterocyclyl, -OR 14 , ~NR 15 R 16 , cyano, or nitro; and each R L is independently halogen, Ci-Ce alkyl, alkenyl, C 2 -C 6 alkynyl, C^-Cs cycloalkyl, C ¾ -C i4 axyl, 5- to 10-membered heteroaryl, 3- to 12-membered heteroeycly!, -OR 14 , ~C(0)R 14 , -

NR 15 R 16 , cyano, oxo, or nitro.

[0058] In the descriptions herein, it is understood that every description, variation, embodiment or aspect of a moiety may be combined with every description, variation, embodiment or aspect of other moieties the same as if each and even combination of descriptions is specifically and individually listed. For example, every description, variation, embodiment or aspect provided herein with respect to R of formula (I) may be combined with every' description, variation, embodiment or aspect of Y, X, L, m, and/or n the same as if each and every' combination were specifically and individually listed. It is also understood that all descriptions, variations, embodiments or aspects of formula (I), where applicable, apply equally to other formulae detailed herein, and are equally described, the same as if each and every description, variation, embodiment or aspect were separately and individually listed for all formulae. For example, all descriptions, variations, embodiments or aspects of formula (I), where applicable, apply equally to any of formulae la, lb, II, Ila, lib, III, Ilia, Illb, IV, IV a, IVb, V, Va, Vb, VI, Via, \ lb.. VII, Vila, VHb, VIII, Villa and VUIb detailed herein, and are equally described, the same as if each and every description, variation, embodiment or aspect were separately and individually listed for all formulae.

[0059] In some embodiments, the compound of formula (I) is of the formula (la):

or a salt thereof, wherein Y, X, L, R, m, and n are as defined for formula (I).

[0060] In some embodiments, the compound of formula (I) is of the formula (lb):

or a salt thereof, wherein Y, X, L, R, m, and n are as defined for formula (I).

[0061] In some embodiments of th e compoun d of formula (I), where m is 1 and n is 1, the compound is of the formula (II):

or a salt thereof, wherein Y, X, L, and R are as defined for formula (I)

[0062] In some embodiments, the compound of formula (11) is of the formula (Ila):

or a salt thereof, wherein Y, X, L, and R are as defined for formula (I).

[QQ63] In some embodiments, the compound of formula (11) is of the formula (11b):

or a salt thereof, wherein Y, X, L, and R are as defined for formula (I).

[0064] In some embodiments of the compound of formula (II), where L is -NH-CH 2 -, the compound is of the formula (III):

or a salt thereof, wherein Y, X, and R are as defined for formula (I). [0065] In some embodiments, the compound of formula (III) is of the formula (Ilia):

or a salt thereof, wherein Y, X, and R are as defined for formula (I).

[0066] In some embodiments, the compound of formula (III) is of the formula (IHb):

or a salt thereof, wherein Y, X, and R are as defined for formula (I).

[QQ67] In some embodiments, the compound of formula (III) is of the formula (III-l):

or a salt thereof, wherein Y and X are as defined for formula (1).

[0068] In some embodiments of the compound of formula (II), where L is -NH-CH(CH 3 )-, the compound is of the formula (IV):

or a salt thereof, wherein Y, X, and R are as defined for formula (I). In one aspect of a compound of formula (IV), the carbon bearing the methyl group of L is in the S configuration. In one aspect of a compound of formula (IV), the carbon bearing the methyl group of L is in the R configuration.

[0069] In some embodiments, the compound of formula (IV) is of the formula (IVa):

or a salt thereof, wherein Y, X, and R are as defined for formula (I). In one aspect of a compound of formula (IV a), the carbon bearing the methyl group of L is in the S configuration in one aspect of a compound of formula (IV a), the carbon bearing the methyl group of L is m the R configuration.

[0070] In some embodiments, the compound of formula (IV) is of the formula (IVb):

or a salt thereof, wherein Y, X, and R are as defined for formula (I). In one aspect of a compound of

formula (IVb), the carbon bearing the methyl group of L is in the S configuration. In one aspect of a compound of formula (IVb), the carbon bearing the methyl group of L is in the R configuration.

[0071] In some embodiments of the compound of formula (II), the compound is of the formula (V):

or a salt thereof, wherein Y . X, and R are as defined for formula (I).

[0072] In some embodiments, the compound of formula (V) is of the formula (Va):

or a salt thereof, wherein Y, X, and R are as defined for formula (I). [0073] In some embodiments, the compound of formula (V) is of the formula (Vb):

or a salt thereof, wherein Y, X, and R are as defined for formula (I).

[0074] In some embodiments of the compound of formula (II), the compound is of the formula

(VI):

or a salt thereof, wherein Y, X, and R are as defined for formula (I).

[0075] In some embodiments, the compound of formula (VI) is of the formula (Via):

or a salt thereof, wherein Y, X, and R are as defined for formula (I).

[0076] In some embodiments, the compound of formula (VI) is of the formula (VIb):

or a salt thereof, wherein Y, X, and R are as defined for formula (I).

[0077] In some embodiments of the compound of formula (ΪΪ), where L is -CH 2 -CH(NH 2 )-, the compound is of the formula (VII):

or a salt thereof, wherein Y, X, and R are as defined for formula (I). In one aspect of a compound of formula (VII), the carbon hearing the ~NH 2 group of L is in the S configuration. In one aspect of a compound of formula (VII), the carbon bearing the -NH 2 group of L is in the R configuration.

[0078] In some embodiments, the compound of formula (VII) is of the formula (Vila):

or a salt thereof, wherein Y, X, and R are as defined for formula (I). In one aspect of a compound of formula (Vila), the carbon bearing the -NH 2 group of L is m the S configuration.

In one aspect of a compound of formula (Vila), the carbon hearing the ~NH 2 group of L is in the

R configuration.

[0079] In some embodiments, the compound of formula (VII) is of the formula (Vllb):

or a salt thereof, wherein Y, X, and R are as defined for formula (I). In one aspect of a compound of formula (Vllb), the carbon bearing the -NH 2 group of L is in the S configuration. In one aspect of a compound of formula (Vllb), the carbon bearing the -NH 2 group of L is in the R configuration.

[0080] In some embodiments of the compound of formula (II), where L is -CH(CH ¾ )~ CH(NH 2 )-, the compound is of the formula (VIII):

or a salt thereof, wherein Y, X, and R are as defined for formula (I). In one aspect of a

compound of formula (VIII), the carbon bearing the -NH 2 group of L is in the S configuration. In one aspect of a compound of formula (VIII), the carbon bearing the -NH 2 group of L is in the R configuration. In one aspect of a compound of formula (VIII), the carbon bearing the methyl group of L is in the S configuration. In one aspect of a compound of formula (VIII), the carbon bearing the methyl group of L is in the R configuration.

[0081] In some embodiments, the compound of formula (VIII) is of the formula (Villa):

or a salt thereof, wherein Y, X, and R are as defined for formula (I). In one aspect of a compound of formula (Villa), the carbon bearing the -NH group of L is in the S configuration. In one aspect of a compound of formula (Villa), the carbon bearing the -NH ? group of L is in the R configuration. In one aspect of a compound of formula (Villa), the carbon bearing the methyl group of L is in the S configuration. In one aspect of a compound of formula (Villa), the carbon bearing the methyl group of L is in the R configuration.

[QQ82] In some embodiments, the compound of formula (VIII) is of the formula (VUIb):

[0083] or a salt thereof, wherein Y, X, and R are as defined for formula (I). In one aspect of a compound of formula (VUIb), the carbon bearing the -NH group of L is in the S configuration. In one aspect of a compo und of formula (VUIb), the carbon bearing the -NH group of L is in the R configuration. In one aspect of a compound of formula (VUIb), the carbon bearing the methyl group of L is in the S configuration. In one aspect of a compound of formula (VUIb), the carbon bearing the methyl group of L is in the R configuration. In some embodiments of the compound of formula (II), the compound is of the formula (IX):

or a salt thereof, wherein Y, X, and R are as defined for formula (I) in one aspect of a compound of formula (IX), the 1,3-cyclobutylene is the cis isomer. In one aspect of a compound of formula (IX), the 1, 3-cyclobutylene is the (ran isomer.

[QQ84] In some embodiments, the compound of formula (IX) is of the formula (IXa):

or a salt thereof, wherein Y, X, and R are as defined for formula (I). In one aspect of a compound of formula (IXa), the 1 ,3-cyclobutylene is the cis isomer. In one aspect of a compound of formula (IXa), the 1,3-cyclobutylene is the (ran isomer.

[0085] In some embodiments, the compound of formula (IX) is of the formula (IXb):

or a salt thereof, wherein Y, X, and R are as defined for formula (I). In one aspect of a compound of formula (IXb), the 1,3-cyclobutylene is the cis isomer. In one aspect of a compound of formula (IXb), the 1 ,3-cyclobutylene is the tran isomer.

[0086] In some embodiments of the compound of formula (II), where L is -NH-CH 2 -, the compound is of the formula (X):

or a salt thereof, wherein Y, X, and R are as defined for formula (I).

[0087] In some embodiments, the compound of formula (X) is of the formula (Xa):

or a salt thereof, wherein Y, X, and R are as defined for formula (I).

[0088] in some embodiments, the compound of formula (X) is of the formula (Xb):

or a salt thereof, wherein Y, X, and R are as defined for formula (I).

[0089] In one variation a compound of formula (I), or a salt thereof, is provided wherein X is -C(=0)-, -O- or -CH(OH)-. In another variation a compound of formula (I), or a salt thereof, is provided wherein X is -S-, -S(=0)-, or -S(=0) 2 -. In some embodiments of the compound of formul a (I), or a salt thereof, X is -C(=0)-. In other embodiments of the compound of formula (I), or a salt thereof, X is -0-. All variations of X apply equally to any applicable formulae herein, such as formulae la, lb, II, Ila, lib, III, Ilia, Illb, IV, IVa, IVb, V, Va, Vb, VI, Via, VIb, VII, Vila, Vllb, VIII, Villa and Vlllb.

[0090] In some embodiments of the compound of formula (I), or a sal t thereof, L is . In a particular such embodiment, R a is H, and R 1 , R 2 , R 3 and R 4 , if

present, are each H. In one embodiment, L is X is - =0. In

another embodiment, R a

[0091] In some embodiments, L is CR 3 R p N CR 1 R 2 q ** . In one particular variation, R 1 and R 2 are attached to the same carbon atom. In another particular variation, R 1 and R are attached to different carbon atoms.

[0092] In some embodiments, L is -N(R 3 )-CR 3 R 2 - (Z. <?. , p is 0). In one particular variation, L is -NH-CR 3 R -. In another particular variation, L is -NH-CH 2 -. In another particular variation, L is -NH-CH(CH3)-. In another particular variation, L is -NH-CR^ 2 -, wherein R 1 and R 2 are taken together with the carbon atom or atoms to which they are attached to form a 3- to 8-membered eycloalkylene (e.g., cyclopropylene).

[0093] In some embodiments, L is -N(R a )-(CR 3 R 2 )3- (z. e., p is 0). In one particular variation, L is -NH-iCR^ 2 ) -. In another particular variation, L is -NH~(CH 2 )3~. In another particular variation, L is -NH-(CR ! R Z )3-, wherein R 3 and R 2 from two non-adjacent carbons are taken together with the carbon atoms to which they are attached and interstitial carbon to form a 3- to 8-membered eycloalkylene (e.g., 1,3-cyclobutylene).

[QQ94] In oilier embodiments of the compound of formula (I), or a salt thereof, L is

NR°R C . in one such embodiment, X is -C=0. In another such embodiment, X is - 0-. In a further such embodiment, X is -CH(OH)-. In yet another such embodiment, X is -S-. In still another such embodiment, X is -S(=0)-. In still another such embodiment, X is -S( : = : 0) 2 In one aspect of such embodiments, r is 1. In another aspect of such embodiments, r is 2. In still another aspect of such embodiments, r is 3. In any embodiment provided where L is e variation, R b and R are both H. In any embodiment provided where L another variation, R and " are both H, r is 1, R is H and R " is a Ci-Ce alkyl such as methyl.

[0095] In some of these embodiments, L is -CR R " -i ! h ' NR b R }- (i.e., r is i). In one particular variation, L is -CH(R 3 )-CH(NH 2 )-, including but not limited to aspects wherein R’ is hydrogen or CrCe alkyl. In one particular variation, L is -CH 2 -CH(NH 2 )-. In another particular variation, L is -( HίP ί O-P ίίNH In some embodiments of the compound of formula (I), or a salt thereof, L is . In one such embodiment, X is -C=0. In another such embodiment, X is -0-. In a further such embodiment, X is -S-. In still another such embodiment, X is -S(=0)-. In still another such embodiment, X is -S(=0) 2 . In a particular variation, L is

,

[0097] In one variation, L is In another particular variation, L is

In one variation, L is and X is selected from the group consisting of -C=0, -0-,

-S-,— S(=Q)- and -S(=0) 2.

[0098] In one variation, provided herein is a compound of formula (I), or a salt thereof, wherein the -X-L- moiety is selected from the group consisting of:

wherein * represents the point of attachment to the Y moiety and ** represents the point of attachment to the remainder of the molecule.

[0099] In another variation is provided a compound of formula (I), or a salt thereof, wherein the -X-L- moiety is selected from the group consisting of:

wherein * represents the point of attachment to the Y moiety and ** represents the point of attachment to the remainder of the molecule.

[0100] In one aspect, provided is a compound of formula (I) or a pharmaceutically acceptable salt thereof, wherein the compound has any one or more of the following features: (i) X is -( ' ! Ok -O- or -CH(OH)-;

(ii) L is: wherein R ! and R , independently of each other and independently at each occurrence, are hydrogen or Ci-C 2 alkyl, or R f and R 2 groups attached to the same carbon atom are taken together with the carbon atom to which they are attached to form a 3- to 5-membered cydoalkylene, or R f and R 2 groups attached to two different carbon atoms are taken together with the carbon atoms to which they are attached to form a 3- to 5-membered cydoalkylene (examples of such -NH-

(b) -CR 5 R 6 -CH(NH 2 )-, wherein R 5 and R 6 , independently of each other and independently at each occurrence, are hydrogen or C 1 -C 2 alky].

(examples of such -CR R -CH(NH 2 )- moieties mlucde NH, and

(c) wherein * represents the point of attachment to the Y-X- moiety and ** represents the point of attachment to the remainder of the molecule; and

(lii) Y is:

(a) C 6 -C 9 and optionally substituted by R ! ! , such as unsubstituted 2,3- dihydro-l / -inden-2-yl or a phenyl or naphthyl substituted by at least one R 11 , including but not limited to when each R 11 is independently selected from halogen, trihalomethyl, cyano, and - C(=0)NH 2 ;

(b) 6- to 10-membered heteroaryl optionally substituted by I1 1 , such as a pyridinyi, pyrimidmyl, pyridin-2(l/f)-onyl, quinoiin-6-yi, optionally substituted by R lz , wherein R 2 is phenyl or -OH; or

(c) 3- to 12-membered heterocycly! optionally substituted by R l , such as

2i/-pyran-2-only, isoindolinyl, piperidin-2-only and piperidmyl substituted by at least one R 1J .

In one aspect of this variation, (i), (ii)(a), and (iii)(a) apply. In another variation, (i), (ii)(a), and (iii)(b) apply apply. In another variation, (i), (ii)(a), and (iii)(c) apply. In another variation, (i), (ii)(b), and (iii)(a) apply. In another variation, (i), (ii)(b), and (iii)(b) apply. In another variation, (i), (ii)(b), and (iii)(c) apply. In another variation, (i), (ii)(c), and (iii)(a) apply. In another variation, (i), (ii)(c), and (iii)(b) apply. In another variation, (i), (ii)(c), and (iii)(c) apply.

[0101] All variations of L, or combinations of X and L, apply equally to any applicable formulae herein, such as formulae la, lb, P, II a. lib. Ill, Ilia, IHb, IV, IV a, IVb, V, Va, Vb, VI, Via, VIb, VII, Vila, VHb, VIII, Villa and VUIb.

[0102] In some embodiments, Y is C 6 -C 9 aryl optionally substituted by R. 3 ', 6- to 10- membered heteroaryl optionally substituted by R 12 , or 3- to 12-rnemhered heterocyclyl optionally substituted by R 13 , wherein when Y is phenyl or naphthyl, the phenyl or naphthyl of Y is substituted by at least one R 11 , and wherein when L is *-NH-CH 2 -** and Y is optionally substituted quinolinyi, the optionally substituted quinolinyi of Y is connected to the parent structure at the 2-, 3-, 5-, 6-, 7-, or 8-position,

[0103] In some embodiments, Y is Ce-C 6 aryl optionally substituted by R 1 '. In one aspect, Y is phenyl optionally substituted by R 11 . In one variation, Y' is substituted with 1 to 3 R 11 moieties which may be the same or different. In some embodiments, Y is phenyl substituted by 1 to 5 R f l independently selected from halogen, trihalomethyl, cyano, and -C(=0)NH 2 . In another particular embodiment, Y is a monosubstituted phenyl, such as phenyl substituted at the C4 position by R 11 . In a particular variation, ' is phenyl substituted in the C4 position by cyano or - C(=0)NH 2 . In another particular embodiment, Y is a disubstituted phenyl, such as phenyl substituted at the C2 and C4 positions or the C3 and C4 positions by two R 11 groups, which may be the same or different. In one aspect, the phenyl of Y is substituted with or two R‘‘ groups selected from the group consisting of cyano, -C(=0)NH 2 , halogen and trihalomethyl. In another embodiment, Y is optionally substituted 2,3-dihydro- l//-inden~2-yl

[0104] In some embodiments, Y is 6- to I G-membered heteroaryl optionally substituted by R 12 , wherein when L is *-NH-CH 2 -** and Y is optionally substituted quinolinyl, the optionally substituted quinolinyl of Y is connected to the parent structure at the 2-, 3-, 5-, 6-, 7-, or 8- position. In another embodiment, Y is quinoiin-6-y! optionally substituted by R i2 . In another embodiment, Y is quinolin-4-yl optionally substituted by R 12 and L is *-CH 2 -CH(NH 2 )-** or *- CH(CH 3 )-CH(NH 2 )-**. In another embodiment, Y is pyridm-4-yl optionally substituted by R’ 2 . In another embodiment, Y is pyrimidin-4-yl optionally substituted by R 12 and optionally fused to C 6 -Ci4 aryl or C5-C10 cycloalkyl, which C 6~ Ci4 aryl or C5-C10 cycloalkyl are optionally substituted by R 2 .

[0105] In some embodiments, Y is quinolin-6-yl optionally substituted by R ! , wherein R 12 is - OH or phenyl.

[0106] In some embodiments, Y is pyridin-3-yl substituted by R 12 . In one variation, Y is pyridin-3-yl substituted by R 12 , wherein R l2 is independently selected from optionally substituted C^-C H aryl or -OR 14 . In one variation, Y is pyridin-3-yl substituted in the C2 position by R 12 , wherein R l is optionally substituted C 6 -C l4 aryl. In one variation, Y is pyridin-3-yl substituted in the C6 position by R 12 , wherein R l2 is -OR 14 . In one variation, Y is pyridin-3-yl substituted in the C2 by optionally substituted Ce-Cw aryl and substituted in the C6 position by - OR 14 .

[0107] In some embodiments, Y is pyridin-4-yl substituted by R l in the C3 position. In one variation, Y is pyridin-4-yl substituted in the C3 position by R 32 , wherein R 12 is independently selected from optionally substituted Ci-Ce alkyl, optionally substituted C 2 -C6 alkenyl, optionally substituted C3-C8 cycloalkyl, optionally substituted C4-C8 cycloalkenyi, OR 14 , NR l5 R 16 , optionally substituted pyridinyl, optionally substituted quinolinyl, optionally substituted indolyl, optionally substituted indazolyl, optionally substitute pyridin-2(l//)-onyl, optionally substituted phenyl, and optionally substituted 3- to 12~membered heterocyclyl. [0108] In some embodiments, Y is pyridin-4-y] substituted in the C3 position by R lz , wherein R i2 is pyridinyl optionally substituted by C -Ce alkyl. In another embodiment, Y is pyridin-4-yl substituted in the C3 position by R 12 , wherein R' 2 is indolyl optionally substituted by C -Ce alkyl. In another embodiment, Y is pyridin-4-yl substituted in the C3 position by R 1 ", wherein R 12 is phenyl optionally substituted by Cj-Ce alkyl, halogen, or Cj-Ce alkoxy. In another embodiment, Y is pyridin-4-yl substituted in the C3 position by R l , wherein R 12 is cyclopropyl optionally substituted by C1-C0 alkyl or -OR 14 . In another embodiment, Y is pyridin-4-yl substituted in the C3 position by R 12 , wherein R 2 is -OR' 4 , and R 14 is optionally substituted Cj- Ce alkyl or optionally substituted phenyl.

[0109] In one variation, when Y is a 6-membered heteroaryl (e.g., pyridin-4-yl) substituted by Ci-C 2 alkyl, wherein the C1-C2 alkyl is substituted by R L , R 1 is selected from the group consisting of halogen, Cj-Ce alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C3-C8 cycloalkyl, Ce.-Cn aryl, 5- to lO-membered heteroaryl, 3- to 12-membered heterocyclyl, -OR 14 , -C(0)R l4 , cyano, oxo, and nitro.

[0110] In one variation, when Y is a 6-membered heteroaryl (e.g., pyridin-4-yl) substituted by C3-C2 alkyl, wherein the C3-C2 alkyl is substituted by R L , R L is selected from the group consisting of halogen and Ce-C^ aryl.

[0111] In some embodiments, Y is pyiimidin-4-yl optionally substituted by R and optionally fused to Ce-Cu aryl or C5-C10 cycloalkyl, which C 6 -C] 4 aryl or CS-C J O cycloalkyd are optionally substituted by R i2 . In a particular embodiment, Y is pyrimidin-4-yl fused to Ce-Cu aryl, wherein Ce-C aryl is optionally substituted by R 12 . In a further embodiment, Y is unsubstituted quinazolin-4-yl. In another particular embodiment, Y is pyrimidin-4-yI fused to C5-C10 cycloalkyl, wherein C5-C10 cycloalkyl is optionally substituted by R 12 . In a further embodiment, Y is 6,7-dihydro-5//-cyclopenta[d]pyrimidin-4-yl.

[0112] In some embodiments, Y is 2/7-pyran-2-onyl optionally substituted by R 12 and optionally fused to Ce-C^ aryl, which C6-C14 aryl is optionally substituted by R 12 . In a further embodiment, Y is 2/f-pyran-2-on-5-yl fused to CVCj 4 aiyl, wherein 2/f-pyran-2-onyl or Ce-C^ aryl are optionally substituted by R l2 . In yet another embodiment, Y is l/ -isochromen-l-on-4-yl optionally substituted by halogen. In some embodiments, Y is B’ or a tautomer thereof. Thus, it is understood that in

some embodiments, Y is A , wherein z is 0, 1,

2, 3,4, or 5; indicates tautomerism between A’ and B’; and R 12 and R 3 ’ are identical for any pair of tautomers. In some embodiments, Y is D or a tautomer thereof. Tims, it is

understood that in some embodiments, Y is 0 , wherein z is

0, 1, 2, 3,4, or 5; indicates tautomerism between C’ and D’; and R 32 and R 33 are identical for any pair of tautomers.

[0114] In one embodiment, Y is selected from the group consisting of

[0115] In some embodiments, Y is pytidin-2(li/)-onyl optionally substituted by R l and optionally fused to C 6 -C l4 aryl or 5- to lO-membered heterocyc!yl, which C 6 -Ci4 aryl or 5- to 10- membered heterocyelyl, independently of each other and independently at each occurrence, are optionally substituted by R 12 . In a further embodiment, Y is pyridin-2(l//)-on-5-yl optionally substituted by Ci-C 6 alkyl (e.g., methyl) or C 6 -Ci4 aryl (e.g phenyl, also referred to herein as “Ph’)). In yet another embodiment, Y is pyridin-2(li )-on-5-yl optionally substituted by R 12 and optionally fused to C 6 -C l 4 aryl, which C6-C34 aryl is optionally substituted by R 12 . In a further embodiment, Y is isoquinolin-l (2//)-on-4-yl optionally substituted by R 12 , such as halogen (e.g., flouro), Ci-Ce alkyl (e.g , methyl), Ce-Cu aryl (e.g., phenyl), or C -C cycloalkyl (e.g, cyclopropyl). In yet another embodiment, Y is pyridin-2(l//)-on-5-yl optionally substituted by R 12 and optionally fused to 5- to 10-membered heterocyclyl, which 5- to lO-membered heterocyclyl, is optionally substituted by R 12 . In a further embodiment, Y is unsubstituted 7,8,9, 10-tetrahy dropyndo] 1 ,2-a] azepm-4(6i/)-on- 1 -yl.

[0116] In some embodiments, Y is 3- to 12-membered heterocyclyl optionally substituted by R l . In a particular embodiment, Y is unsubstituted isoindolin-2-yl. In another embodiment, Y is piperidin-2-on-5-yl optionally substituted by R 13 , such as C -C alkyl (e.g., ethyl) and Ce-Cn aryl (e.g., phenyl).

[0117] In some of these embodiments, Y is selected from the group consisting of:

these embodiments, Y is selected from the group consisting of:

All variations of Y apply equally to any applicable formulae herein, such as formulae la, lb, 11, 11a, lib, III, 111a, Illb, IV, IVa, IVb, V, Va, Vb, VI, Via, VIb, VII, Vila, Vllb, VIII, Villa and Vlllb.

[0120] Also provided are salts of compounds referred to herein, such as

pharmaceutically acceptable salts. The invention also includes any or all of the stereochemical forms, including any enantiomeric or diastereomeric forms, and any tautomers or other forms of the compounds described.

[0121] Some of the compounds described herein exist in equilibrium with a tautomeric form. For example, amide A is a tautomeric form of B and imidic acid B is a tautomeric form of A. Similarly, amide C is a tautomeric form of D and imidic acid D is a tautomeric form of C. Amide A exists in equilibrium with a tautomeric form of imidic acid B, and amide C exists in equilibrium with a tautomeric form of imidic acid D Regardless of which tautomeric form is depicted, the compounds are understood by one of ordinary skill in the art to comprise both the amide and the imidic acid tautomers.

[0122] A compound as detailed herein may in one aspect be in a purified form and compositions comprising a compound in purified forms are detailed herein. Compositions comprising a compound as detailed herein or a salt thereof are provided, such as compositions of substantially pure compounds. In some embodiments, a composition containing a compound as detailed herein or a salt thereof is in substantially pure form. Unless otherwise stated, “substantially pure” intends a composition that contains no more than 35% impurity, wherein the impurity denotes a compound other than the compound comprising the majority of the composition or a salt thereof. In some embodiments, a composition of substantially pure compound or a salt thereof is provided wherein the composition contains no more than 25%, 20%, 15%, 10%, or 5% impurity. In some embodiments, a composition of substantially pure compound or a salt thereof is pro vided wherein the composition contains or no more than 3%, 2%, 1 % or 0 5% impurity

[0123] Representative compounds are listed in Table 1.

Additional representative compounds are listed in Table l-A.

[0125] Certain compounds depicted in Table 1 or Table l-A exist as tautomers.

Regardless of which tautomer is shown, all tautomeric forms are intended. [0126] In some embodiments, provided herein is a compound described in Table 1, or a tautomer thereof, or a salt of any of the foregoing, and uses thereof. In some embodiments, provided herein is a compound described in Table 1 or a pharmaceutically acceptable salt thereof. In some embodiments, provided herein is a compound described in Table l-A, or a tautomer thereof, or a salt of any of the foregoing, and uses thereof. In some embodiments, provided herein is a compound described in Table l-A or a pharmaceutically acceptable salt thereof. In some embodiments, provided herein is a compound described in Table 1 or l-A, or a tautomer thereof, or a salt of any of the foregoing, and uses thereof. In some embodiments, provided herein is a compound described in Table 1 or l-A or a pharmaceutically acceptable salt thereof.

[0127] In some embodiments, provided herein is a compound selected from Compound

Nos. 1 -129, or a tautomer thereof, or a salt of any of the foregoing, and uses thereof. In some embodiments, provided herein is a compound selected from Compound Nos. 1-129, or a pharmaceutically acceptable salt thereof.

[0128] In some embodiments, provided is a compound selected from Compound Nos. 1-

129, or a stereoisomer th ereof (in cluding a mixture of two or more stereoisomers th ereof), or a salt thereof In some embodiments, the compound is a salt of a compound selected from Compound Nos. 1-129, or a stereoisomer thereof

[0129] In one variation, the compound detailed herein is selected from the group consisting of:

N-(2-(2-eyano-4,4-difluoropyrrolidin-l-yl)-2-oxoethyl)-l-oxo -l,2-dihydroisoquinoiine-4- carboxamide;

N-(2-(2-cy ano-4,4-difluoropyrrolidin-l-yl)-2-oxoethyl)-l-oxo-2-pheny 1-1,2- dihydroisoquinoline-4-caxboxamide;

N-(2-(2-cyano-4,4-difluoropynOlidin-l-yl)-2-oxoethyl)-2-c yclopropyl-l -oxo-l ,2- dihydroisoquinoline-4-carboxamide;

N-(2-(2-cyano-4,4-difluoropynOlidin-l-yl)-2-oxoethyl)quin oline-6-carboxamide;

N-(2-(2-cyano-4,4-difluoropynOlidin-l-yl)-2-oxoethyl)-6,7 -dihydro-5H· cyclopenta[d]pyriniidine-4-carboxamide;

N-(2-(2-cyano-4,4-difluoropynOlidin-l-yl)-2-oxoethyl)-6-o xo-l ,6-dihydtOpyridine-3- carboxamide:

N-(2-(2-cyano-4,4-difluoropyrrolidin-l-yl)-2-oxoethyl)-2, 3-dihydro-lH-indene-2-carboxamide;

N-(2-(2-cyano-4,4-difluoropynOlidin-l-yl)-2-oxoethyl)isoi ndoline-2-carboxamide;

N -(2-(2-cy ano-4,4-difluoropy rrolidin- 1 -y l)-2-oxoethy l)-2-methy 1- 1 -oxo- 1,2- dihydroisoquinoline-4-carboxamide;

N-(2-(2-cyano-4,4-difluoropyrrolidin-l -yl)-2-oxoethyl)-2-hydroxyquinoline-6-carboxamide;

N-(2-(2-cyano-4,4-dilluoropyrrolidin-l -yl)-2-oxoethyl)terephthal amide;

4-cyano-N-(2-(2-cyano-4,4-diiluoropyrrolidin-l -yl)-2-oxoethyl)-3-(trif!uoromethyl)benzamide;

N-(2-(2-cyano-4,4-diiluoropyrrolidin-l -yl)-2-oxoethyl)-3-pheny]isonicotinamide;

N-(2-(2-cyano-4,4-diiluoropyrrolidin-l -yl)-2-oxoethyl)-6-oxo-5-phenyl-l,6-dihydropyridine-3- carboxamide;

4-cyano-N-(2-(2-cyano-4,4-difluoropynOlidin-l-yl)-2-oxoe hyl)-3-fluorobenzainide;

N-(2~(2-cy ano-4,4-difluoropyrrolidin- 1 -yl)-2-oxoethyl)- 1 -methyl-6-oxo- 1,6-dihy dropyridine-3- carboxamide;

4-cyano-N-(2-(2-cyano-4,4-difluoropyrrolidin-l-yl)-2-oxoe thyl)benzamide;

4-cyano-N-(2-(2-cyano-4,4-difluoropyrrolidin-l-yl)-2-oxoe thyl)-2-(trifluorometh}d)benzamide;

N-(2-(2-cyano-4,4-dif[uoropyrrolidin-l-yl)-2-oxoethyl)-4- phenylquinoline-6-carboxairade;

N-(2-(2-cyano-4,4-difluoropynOlidin-l-yl)-2-oxoethyl)-l-e thyl-6-oxo-2-phenylpiperidine-3- carboxamide;

N-(2-(2-cyano-4,4-difluoropyrrolidin-l-yl)-2-oxoethyl)-4- oxo-4,6,7,8,9,l 0-

^ 7 hexahydropyridoj 1 ,2-a]azepine-l -carboxamide;

N-(2-(2-cy ano-4,4-difluoropy rrolidin- 1 -yl)-2-oxoethyl)- 1 -oxo- 1 H-i sochromene-4- carboxamide:

N-(l-(2-cyano-4,4-difluoropyrrolidine-l-carbonyl)cyclopro pyl)-l-oxo-l,2- dihydroisoquinoline-4-carboxamide;

N-(2-(2-cyano-4,4-difluoropyriOiidm-l -yl)-2-oxoethyi)-3-cycIopropylisomcotmamide;

N-(l -(2-cy ano-4,4-difluoropyrrolidin- 1 -y 1)- 1 -oxopropan-2-yl)- 1 -oxo- 1 ,2-dihy droisoquinoiine-

4-carboxamide:

N-(2-(2-cyano-4,4-difluoropyrrolidin-l -yl)-2-oxoethyl)-6-fluoro-] -oxo-l,2- dihydroisoquinoline-4-carboxamide;

N-(2-(2-cyano-4,4-difluoropyrrolidin-l-yl)-2-oxoethyl)-7- fluoro-l-oxo-l,2- dihydroisoquinoline-4-carboxamide;

N-(2-(2-cyano-4,4-difluoropynOlidin-l-yl)-2-oxoethyl)-6-f luoro-l-oxo-lH-isochromene-4- carboxamide;

N-(2-(2-cy ano-4,4-difluoropyrrolidin- 1 -yl)-2-oxoethy l)-6~oxo-4-pheny! - 1 ,6-dihy dropyridine-3- carboxamide;

N-(2-(2-cyano-4,4-difluoropyrrolidin-l-yl)-2-oxoetbyl)-6- oxo-2-phenyl-l,6-dihydropyridine-3- carboxamide;

N-(2-(2-cyano-4,4-difluoropy rrolidin- l -yl)-2-oxoethyl)-l-methyl-6-oxo-2-phenyJ-l, 6- dihydropyridine-3-carboxamide;

4,4-difluoro-l-(l-(l-oxo-l ,2-dihydroisoquinoline-4-carbonyl)azetidine-3-carbonyl)pyrro lidine-

2-earbonitriIe:

N-(2-(2-cyano-4,4-difluoropyrrolidin-l-yl)-2-oxoethyl)-3- (dimethylamino)isonicotmamide;

N-(2-(2-cyano-4,4-difluoropyrroiidin-l-yl)-2-oxoethyi)-3- (diethylamino)isonicotinamide; N-(2-(2-cyano-4,4-difluoropynOlidin-l-yl)-2-oxoethyl)-3-(4-f luorophenoxy)isonicotinamide;

N-(2-(2-cyano-4,4-difluoropynOlidin-l-yl)-2-oxoethyl)-3-m ethoxyisonicotinamide;

N-(2-(2-cyano-4,4-dilluoropyrrolidin-l -yl)-2-oxoethyl)-3-(trifluoromethyl)isonicotinamide;

N-(2-(2-cyano-4,4-difluoropyrrolidin-l -yl)-2-oxoethyl)-3-isopropylisonicotmamide;

N-(2-(2-cyano-4,4-difluoropyrrolidin-l-yl)-2-oxoethyl)-3- yinylisonicotinamide;

N-(2-(2-cyano-4,4-difluoropyrrolidin-l -yi)-2-oxoethyl)-3-(pyrrolidin~] -yl)isonicoiinamide;

N-[2-[2-cy ano-4,4-difluoro-pyrrolidin- 1 -yl] -2-oxo-ethyl] -3-(l -piperidy l)pyridine-4- carboxamide;

N-(2-(2-cyano-4,4-difluoropyrrolidin-l-yl)-2-oxoethyl)-3- (cyclohex-l-en-l-yl)isonicotinamide;

3-(benzo d] [ l,3]dioxol-5-yl)-N-(2-(2-cyano-4,4-difluoropyrrolidin-l-yl)- 2- oxoeihy 1 )i sonicoti nami d e;

N-(2-(2-cyano-4,4-difluoropynOlidin-l-yl)-2-oxoethyl)-3-( 2,3-dihydrobenzo[b] [l,4]dioxin-6- yl)isonicotinamide;

N-(2-(2-cyano-4,4-difluoropyrrolidin-l-yl)-2-oxoethyl)-3- (2,6-difluorophenyl)isonicotinamide;

N-(2-(2-cyano-4,4-difluoropynOlidin-l-yl)-2-oxoethyl)-3-( 3,5-difluoiOphenyl)isonicotinamide;

N-(2-(2-cyaiio-4,4-difluoropyiTolidm-l~yl)-2-oxoethyl)~2 , -methyl-| 3,4'-bipyridine]-4- carboxamide;

N-(2-(2-cyano-4,4-difluoropyrrolidin- 1 -y l)-2-oxoethy l)-3-(l -methy 1- lH-indol-5- yl)isonicotinamide;

3-benz>4-N-(2-(2-cyano-4,4-difluoropyrrolidin-l-yl)-2- oxoeihyl)isonicotinamide;

N-(2-(2-cyano-4,4-difluoropyrrolidin-l-yl)-2-oxoethyl)-3- (quinolin-4-yl)isonicotinamide;

N-(2-(2-cyano-4,4-difluoropynOlidin-l -yl)-2-oxoethyl)-3-(4-fluorophenyl)isonicotinainide; N-(2-(2-cyano-4,4-difluoropynOlidin-l-yl)-2-oxoethyl)-3-(2,4 -dif]uorophenyl)isonicotinamide;

3-(5-chloro-2-fluoiOphenyl)-N-(2-(2-cyano-4,4-difluoropyn Olidin-l-yl)-2- oxoethyl)isonicotinamide;

N-(2-(2-cyano-4,4-difluoropynOlidin-l-yl)-2-oxoethyl)-3-( 2-methoxyphenyl)isonicotinamide;

N-(2-(2-cyano-4,4-difluoropyrrolidin-l-yl)-2-oxoethyl)-3- (3-methoxyphenyl)isonico†inamide;

N-(2-(2-cyano-4,4-difluoropyrrolidin-l-yl)-2-oxoethyl)-3- (4-methoxyphenyl)isonico†inamide;

N-(2-(2-cyano-4,4-difluoropynOlidin-l-yl)-2-oxoethyl)-[2, 3'-bipyridine]-4'-carboxamide;

N-(2-(2-cyano-4,4-difluoropynOlidin-l-yl)-2-oxoethyl)- 3,3'-bipyridine]-4-carboxamide;

N-(2-(2-cyano-4,4-difluoropynOlidin-l-yl)-2-oxoethyl)- 3,4'-bipyridine]-4-carboxamide;

3-(2-(tert-butyl)phenyl)-N-(2-(2-cyano-4,4-difluoropyrrol idin-l-yl)-2- oxoethy l)i soni cotinamide;

N-(2-(2-cyano-4,4-difluoropyrrolidin-l -yl)-2-oxoethyl)-3-(2-isopropylphenyl)isonicotinamide;

N-(2-(2-<yano-4,4-difluoropyrrolidin-l-yl)-2-oxoethyl) -3-(2-ethylphenyl)isonicotinamide;

N-(2-(2-cyano-4,4-difluoropyrrolidin-l-yl)-2-oxoethyl)-3- (o-tolyl)isonicotinamide;

N-(2-(2-cyano-4,4-difluoropyrrolidin-l-yl)-2-oxoethyl)-3- (lH-indol-4-yl)isonicotinamide;

N-(2-(2-cy ano-4,4-difluoropyrrolidin- 1 -yl)-2~oxoethyl)-3-(i -methyl- lH-indol-4- yl)isonicotinamide;

N-(2-(2-cyano-4,4-difluoropyrrolidin-l-yl)-2-oxoethyl)qui nazoline-4-carboxamide; l-(2-amino-4-hydroxy-4-(l -oxo-1, 2-dihydroisoquinolin-4-yl)butanoyl)-4,4-difluoropyrrolidine

2-carbonitrile;

4,4-difluoro-l -(0-(l -oxo-l,2-dihydroisoquinolin-4-yl)sery])pynOlidine-2-carbonit rile; l -(2-amino-4-oxo-4-(quinolin-4-yl)butanoyl)-4,4-dilluoropyrro lidine-2-carbonitrile; l -(2-amino-4-hydroxy-4-(quinolin-4-yl)butanoyl)-4,4-difluorop yrrolidine-2-carbonitrile;

4.4-difluoro-l-(0-(quinolin-4-yl)seiyl)pyrrolidine-2-carb onitrile; l-(2-amino-4~oxo-4-(l-oxo-l,2-dihydroisoquinolin-4-yl)butano yl)-4,4-difluoropyrrolidme-2- carbonitrile;

1 -(2-amino-4-hy droxy-3-methyl-4-( 1 -oxo- 1 ,2-dihy droisoquinolin-4-yl)butanoyl)-4, 4- difluoropyrrolidine-2-carbonitrile;

4.4-diiluoro-l-(0-(l-oxo-l,2-dihydroisoquinolin-4-yl)thre onyl)pynO]idine-2-carbonitrile; l-(2-amino-3-methyl-4-oxo-4-(quinolin-4-yl)butanoyl)-4,4-dif luoropyrrolidine-2-caxbonitrile; l-(2-armno-4-hydroxy-3-raethyl-4-(quinolin-4-yl)butanoyr)-4, 4-difluoropyrrolidine-2- carbonitrile;

4.4-difluoro-l-(0-(quinolin-4-yl)threonyl)pyrrolidine-2-c arbonitrile; l-(2-amino-3-methyl-4-oxo-4-(l-oxo-l ,2-dihydroisoquinolin-4-yl)butanoyl)-4,4- difluoropyrrolidine-2-carbonitrile;

N-(2-(2-cyano-4,4-difluoropyrrolidin-l-yl)-2-oxoethyl)-4- oxo-3,4,4a,8a-tetrahydrophthalazine-

1 -carboxamide;

N-[2-[2-cyano-4,4-difluoro-pynOlidin-l-yl]-2-oxo-ethyl]-3 -methyl-4-oxo-phthalazine-l - carboxamide;

N~(4-(2-cy ano-4,4~difluoropyrrolidin- 1 -yl)-4-oxobuty 1)- 1 -oxo- 1 ,2-dihy droisoquinoline-4- carboxamide;

N-(2-(2-cyano-4,4-difluoropyrrolidin-l -yl)-2-oxoethyl)-lH-indazole-5-carboxaniide;

N-(2-(2-cyano-4,4-difluoropyrrolidin-l -yl)-2-oxoethyl)furo[2,3-clpyridine-2-carboxamide; N-(2-(2-cyano-4,4-difluoropyrrolidin-l-yl)-2-oxoethyl)pyrazo lo[l,5-a]pytidine-5-carboxamide;

N-(2-(2-cyano-4,4-difluoropynOlidin-l-yl)-2-oxoethyl)-6-m ethoxy-2-pheny]nicotinaTnide;

N-(2-(2-cyano-4,4-difluoropyrrolidin-l-yl)-2-oxoethyl)-3- (l -methyl-lH-indazol-4- yl)isonicotinamide;

N-[2-[2-cyano-4,4-difluoro-pynOlidin-l-yll-2-oxo-ethyl]-3 -morpholino-pyridine-4- carboxamide;

N-(2-(2-cyano-4,4-difluoropyrrolidin-l-yl)-2-oxoethyl)-3- (l- meihy]cyclopropyl)isonicotinamide;

N-(2-(2-cyano-4,4-difluoropyrrolidin-l-yl)-2-oxoethyl)-3- (2- hydroxyeycIopropyl)isomcotinamide;

N-(2-(2-cyano-4,4-difluoropyrrolidin-l-yl)-2-oxoethyl)-3- (piperidin-4-yl)isonicotinamide;

N-(2-(2-cyano-4,4-difluoropyrrolidin-l-yl)-2-oxoethyl)-3- (tetrahydro-2H-pyran-4- yJ)isonicotinamide;

N-(2-(2-cyano-4,4-difluoropynOlidin-l-yl)-2-oxoethyl)-6,7 -dihydro-5H-cyclopenta[b]pyridine-

4-carboxamide;

N-(2-(2-cyano-4,4-difluoropyrrolidin-l-yl)-2-oxoethyl)-5, 6,7,8-tetrahydroquinoline-4- carboxamide;

N-(2-(2-cyano-4,4-difluoropyrrolidin-l-yl)-2-oxoethyl)pyr rolo[l,2-a]pyrimidine-4- carboxamide;

N-(2-(2-cyano-4,4-diiluoropyrrolidin-l -yl)-2-oxoethyl)-3-(trifluoromethoxy)isonicotinaniide;

N-(2-(2-cyano-4,4-diiluoropyrrolidin-l -yl)-2-oxoethyl)-3-phenoxyisonicotinaniide;

N-(2-(2-cyano-4,4-diiluoro-2-methy]pyrrolidin-l -y])-2-oxoethyl)-l -oxo-l,2- dihydroisoquinoline-4-carboxamide;

N-(2-(2-cyano-4,4-difluoropyrrolidin-]-yl)-2-oxoethyl)-6' -oxo- ,6'-dihydro-[3,3'-bipyridine]- 4-carboxamide;

N-(2-(2-cyano-4,4-difluoropynOlidin-l-yl)-2-oxoethyl)-5-c ydopropyl-6-oxo-5,6-dihydtO-l,5- naphthyridine-4-carboxamide;

N-(3-(2-cy ano-4,4-difluoropyrrolidine- 1 -carbonyl)cy clobutyl)- 1 -oxo- 1 ,2-dihy droisoquinoline- 4-carboxamide;

N-(2-(2-cyano-4,4-difluoropyrrolidin-l-yJ)-2-oxoethyl)-3- (phenylamino)isonicotinamide;

N-(2-(2-cyano-4,4-difluoropyrrolidin-l-yl)-2-oxoetbyl)-3- (cyclohex-2-en-l-yl)isonicotinamide;

N-(2-(2-cyano-4,4-dilluoropynOlidin-l -yl)-2-oxoethyl)-3-(indo]in-5-yl)isonicotinami de;

N-(2-(2-cy ano-4,4-difluoropyrrolidin-l-yl)-2-oxoethyl)-r, 2', 3', 6' -tetrahydro-[3,4' -bipyridine]- 4-carboxamide;

N-(2-(2-cyano-4,4-difluoropyrrolidin-l-yl)-2-oxoethyl)-3- (5-methylfuran-2- yl)isonicotinamide;

6'-amino-N-(2-(2-cyano-4,4-difluoropynOlidin-l-yl)-2-oxoe thyl)-[3,3 , -bipyridine]-4- carhoxamide;

3-(6-chloronaphthalen-2-yl)-N-(2-(2-cyano-4,4-difluoropyr rolidin-l -yl)-2- oxoethyljisonicotinamide;

N-(2-(2-cyano-4,4-difluoropyrrolidin-]-yl)-2-oxoethyi)-4- (4-fluorophenyi)-6-oxo-l,6- dihydropyridine-3-carboxamide;

N-(2-(2-cyano-4,4-difluoropyrrolidin-l-yl)-2-oxoethyl)-3- (3,5-dimethylisoxazol-4- y l)isonicotinami de;

3-(4-chloro-3-fluorophenyl)-N-(2-(2-cyano-4,4-difliioropy nOlidin-l-yl)-2- oxoethyl)isonicotinamide;

3-(3-(tert-butyl)phenyl)-N-(2-(2-cyano-4,4-difluoropyrrol idm-l-yl)-2- oxoethyl)isonicotinamide; N-(2-(2~cyano~4,4-difluoropyrrolidin-l -yl)-2~oxoeihyl)-3-ethoxyisonicotinamide;

N-(2-(2-cyano-4,4-difluoropyrrolidin-l -yl)-2-oxoethyl)-3-((4- fluorophenyl)amino)isonicotinamide;

N-(2-(2-cyano-4,4-difluoropyrrolidin-l-yl)-2-oxoethyl)-3- (phenylthio)isonicotinamide;

N-(2-(2-cyano-4,4-difluoropyrrolidin-l-yl)-2-oxoethyl)-6- oxo-4-(quinolin-4-yl)-l,6- dihydropyridine-3-carboxamide;

N-(2-(2-cyano-4,4-difluoropynOlidin-l-yl)-2-oxoethyl)-3-( pyridin-3-yJamino)isonicotinamide;

N-(2-(2-cyano-4,4-difluoropyrrolidin-l-yl)-2-oxoethyl)-3- (piperidin-4- ylamino)isonicotinamide;

N-(2-(2-cyano-4,4-difluoropyrrolidin-l-yl)-2-oxoethyl)-3- (quinolin-4- ylamino)isonicotinamide;

3-(4-aminopiperidin- 1 -yl)-N-(2-(2-cyano-4,4-difluoropyrrolidin- 1 -yl)-2- oxoethyl)isonicotinamide;

4-benzyl-N-(2-(2-cyano-4,4-difiuoropyrrolidin-l-yl)-2-oxo ethyl)-6-oxo-l ,6-dihydropyridine- 3-carboxamide;

N-(2-(2-cyano-4,4-difluoropyrrolidm-l-yl)-2-oxoethyl)-3-( l-phenylvmyl)isonicotinarnide;

N-(2-(2-cyano-4,4-difluoropyrrolidm-l-yl)-2-oxoethyl)-3-( l-phenylethyl)isonicotmamide;

N-(2-(2-cyano-4,4-difluoropyrrolidin-]-yl)-2-oxoethyl)-3- styrylisonicotinaimde;

N-(2-(2-cyano-4,4-difluoropyrrolidm-l-yl)-2-oxoethyl)-3-p henethylisonicotmaimde;

N-(l-(2-cyano-4,4-difluoropyrrolidin-l-yl)-l-oxopropan-2- yl)-3-phenylisonicotinamide;

N-(2-(2-cy ano-4,4-difluoropyrrolidin- 1 -yl)-2-oxoethyl)-3-(2-phenylprop- 1 -en- 1 - yl)isonicotinamide; N-(2-(2-cyano-4,4-difluoropyrrolidin-l -yl)-2-oxoethyl)-3-(2-phenylallyl)isonicotinamide, and

N-(2-(2~c ano~4,4-difluoropyrrolidin~l -y])-2-oxoethyl)-3-(2-pheny]propyl)isonicotinamide

[0130] The compounds depicted herein may be present as salts even if salts are not depicted and it is understood that the present disclosure embraces all salts and solvates of the compounds depicted here, as well as the non-salt and non-solvate form of the compound, as is well understood by the skilled artisan. In some embodiments, the salts of the compounds provided herein are pharmaceutically acceptable salts. Where one or more tertiary amine moiety is present in the compound, the N-oxides are also provided and described.

[0131] Where tautomeric forms may be present for any of the compounds described herein, each and every tautomeric form is intended even though only one or some of the tautomeric forms may be explicitly depicted. The tautomeric forms specifically depicted may or may not be the predominant forms m solution or when used according to the methods described herein.

[0132] The present disclosure also includes any or all of the stereochemical forms, including any enantiomeric or diastereomeric forms of the compounds described, such as the compounds of Table 1. The structure or name is intended to embrace all possible stereoisomers of a compound depicted. All forms of the compounds are also embraced by the invention, such as cr stalline or non-crystalline forms of the compounds. Compositions comprising a compound of the invention are also intended, such as a composition of substantially pure compound, including a specific stereochemical form thereof, or a composition comprising mixtures of compounds of the invention in any ratio, including two or more stereochemical forms, such as in a racemic or non-racemic mixture.

[0133] The invention also intends isotopically-iabeled and/or isotopically -enriched forms of compounds described herein. The compounds herein may contain unnatural proportions of atomic isotopes at one or more of the atoms that constitute such compounds. In some embodiments, the compound is isotopically-iabeled, such as an isotopically-iabeled compound of the formula (1) or variations thereof described herein, where a fraction of one or more atoms are replaced by an isotope of the same element. Exemplary isotopes that can be incorporated into compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen.

phosphorus, sulfur, chlorine, such as ¾ Ή. U C, i 3 C Certain isotope labeled compounds (e.g. 3 H and 4 C) is useful in compound or substrate tissue distribution studies. Incorporation of heavier isotopes such as deuterium (Ή) can afford certain therapeutic advantages resulting from greater metabolic stability, for example, increased in vivo half-life, or reduced dosage requirements and, hence may be preferred in some instances.

[0134] Isotopically-labeled compounds of the present invention can generally be prepared by standard methods and techniques known to those skilled in the art or by procedures similar to those described in the accompanying Examples substituting appropriate isotopically- labeled reagents in place of the corresponding non-labeled reagent.

[0135] Articles of manufacture comprising a compound described herein, or a salt or solvate thereof, m a suitable container are provided. The container may be a vial, jar, ampoule, preloaded syringe, i.v. bag, and the like.

[0136] Preferably, the compounds detailed herein are orally bioavailable. However, the compounds may also be formulated for parenteral (e.g., intravenous) administration.

[0137] One or several compounds described herein can be used in the preparation of a medicament by combining the compound or compounds as an active ingredient with a pharmacologically acceptable carrier, which are known in the art. Depending on the therapeutic form of the medication, the earner may be in various forms. In one vari ation, the manufacture of a medicament is for use in any of the methods disclosed herein, e.g., for the treatment of cancer.

General synthetic methods

[0138] The compounds of the invention may be prepared by a number of processes as generally described below and more specifically in the Examples hereinafter (such as the schemes provided in the Examples below). In the following process descriptions, the symbols when used in the formulae depicted are to be understood to represent those groups described above in relation to the formulae herein.

[0139] Where it is desired to obtain a particular enantiomer of a compound, this may be accomplished from a corresponding mixture of enantiomers using any suitable conventional procedure for separating or resolving enantiomers. Thus, for example, diastereomeric deri vatives may be produced by reaction of a mixture of enantiomers, e.g., a racemate, and an appropriate chiral compound. The diastereomers may then be separated by any convenient means, for example by crystallization and the desired enantiomer recovered. In another resolution process, a racemate may be separated using chiral High Performance Liquid Chromatography. Alternatively, if desired a particular enantiomer may be obtained by using an appropriate chiral intermediate in one of the processes described.

[0140] Chromatography, recrystallization and other conventional separation procedures may also be used with intermediates or final products where it is desired to obtain a particular isomer of a compound or to otherwise purity a product of a reaction.

[0141] Solvates of a compound provided herein or a salt thereof are also contemplated.

Solvates contain either stoichiometric or non-stoichiometric amounts of a solvent, and are often formed during the process of crystallization. Hydrates are formed when the solvent is water, or alcoho!ates are formed when the solvent is alcohol.

[0142] Compounds of the formula (1-1) can be prepared according to Scheme 1, wherein R, R 1 , R 2 , Y, m, n and q are as detailed herein for formula (I), or any variation thereof detailed herein: Z and l ] are leaving groups; and PG 1 is an amine protecting group.

Scheme 1

[0143] Coupling of a compound of formula (1-1 a) with a compound of formula (I-lh) in the presence of a coupling agent (e.g., HATH, HOBt, or PyBOP) yields a compound of formula (I- lc). Deprotection of the amine of the compound of formula (I-lc) under acidic conditions (e.g., HC1 or TsOH) provides the compound of formula (I-l d) as a salt, which is coupled with a carboxylic acid in the presence of a couplin agent (e.g., HATU, HOBt, or PyBOP) to yield a compound of formula (1-1). [0144] An exemplary embodiment of the preparative method in Scheme 1 is shown in

Scheme la.

[0145] In some embodiments, Y is 6- to 10-membered heteroaryl optionally substituted by R 12 In a further embodiment, Y is pyridin-4-yl substituted by R 2 in the 3-position, wherein O

Y L OH i s represented by the compound of formula (P-1).

Scheme 2

[0146] It is understood that the schemes above may be modified to arrive at various compounds of the invention by selection of appropriate reagents and starting materials. For a general description of protecting groups and their use, see P.G.M. Wilts and T.W. Greene, Greene's Protective Groups in Organic Synthesis 4 th edition, Wiley-Interscience, New York, 2006.

Pharmaceutical Compositions and Formulations

[0147] Pharmaceutical compositions of any of the compounds detailed herein are embraced by this disclosure. Thus, the present disclosure includes pharmaceutical compositions comprising a compound as detailed herein or a salt thereof and a pharmaceutically acceptable carrier or excipient. In one aspect, the pharmaceutically acceptable salt is an acid addition salt, such as a salt formed with an inorganic or organic acid. Pharmaceutical compositions may take a form suitable for oral, buccal, parenteral, nasal, topical or rectal administration or a form suitable for administration by inhalation.

[0148] A compound as detailed herein may in one aspect be in a purified form and compositions comprising a compound in purified forms are detailed herein. Compositions comprising a compound as detailed herein or a salt thereof are provided, such as compositions of substantially pure compounds. In some embodiments, a composition containing a compound as detailed herein or a salt thereof is in substantially pure form.

[0149] In one variation, the compounds herein are synthetic compounds prepared for administration to an individual. In another variation, compositions are provided containing a compound in substantially pure form. In another variation, the present disclosure embraces pharmaceutical compositions comprising a compound detailed herein and a pharmaceutically acceptable carrier. In another variation, methods of administering a compound are provided.

The purified forms, pharmaceutical compositions and methods of administering the compounds are suitable for any compound or form thereof detailed herein.

[0150] A compound detailed herein or salt thereof may be formulated for any available delivery route, including an oral, mucosal (e.g., nasal, sublingual, vaginal, buccal or rectal), parenteral (e.g., intramuscular, subcutaneous or intravenous), topical or transdermal delivery form. A compound or salt thereof may be formulated with suitable carriers to provide delivery forms that include, but are not limited to, tablets, caplets, capsules (such as hard gelatin capsules or soft elastic gelatin capsules), cachets, troches, lozenges, gums, dispersions, suppositories, ointments, cataplasms (poultices), pastes, powders, dressings, creams, solutions, patches, aerosols (e.g., nasal spray or inhalers), gels, suspensions (e.g., aqueous or non-aqueous liquid suspensions, oil-in-water emulsions or water-in-oil liquid emulsions), solutions and elixirs.

[0151] One or several compounds described herein or a salt thereof can be used in the preparation of a formulation, such as a pharmaceutical formulation, by combining the compound or compounds, or a salt thereof, as an acti ve ingredient with a pharmaceutically acceptable carrier, such as those mentioned above. Depending on the therapeutic form of the system (e.g., transdermal patch vs. oral tablet), the carrier may be m various forms. In addition,

pharmaceutical formulations may contain preservatives, solubilizers, stabilizers, re-wetting agents, emulgators, sweeteners, dyes, adjusters, and salts for the adjustment of osmotic pressure, buffers, coating agents or antioxidants. Formulations comprising the compound may also contain other substances which have valuable therapeutic properties. Pharmaceutical formulations may be prepared by known pharmaceutical methods. Suitable formulations can be found, e.g., m Remington s Pharmaceutical Sciences, Mack Publishing Company, Philadelphia, PA, 20 th ed. (2000), which is incorporated herein by reference.

[0152] Compounds as described herein may he administered to individuals in a form of generally accepted oral compositions, such as tablets, coated tablets, and gel capsules in a hard or in soft shell, emulsions or suspensions. Examples of carriers, which may be used for the preparation of such compositions, are lactose, com starch or its derivatives, talc, stearate or its salts, etc. Acceptable carriers for gel capsules with soft shell are, for instance, plant oils, w ? ax, fats, semisolid and liquid poly-ols, and so on. In addition, pharmaceutical formulations may contain preservatives, solubilizers, stabilizers, re-wetting agents, emulgators, sweeteners, dyes, adjusters, and salts for the adj ustment of osmotic pressure, buffers, coating agents or antioxidants.

[0153] Compositions comprising a compound provided herein are also described. In one variation, the composition comprises a compound or salt thereof and a pharmaceutically acceptable carrier or excipient. In another variation, a composition of substantially pure compound is provided. In some embodiments, the composition is for use as a human or veterinary' medicament. In some embodiments, the composition is for use in a method described herein. In some embodiments, the composition is for use in the treatment of a disease or disorder described herein.

Methods of Use and Uses

[0154] Compounds and compositions detailed herein, such as a pharmaceutical composition comprising a compound of any formula provided herein or a salt thereof and a pharmaceutically acceptable carrier or excipient, may be used in methods of administration and treatment as provided herein. The compounds and compositions may also be used in in vitro methods, such as in vitro methods of administering a compound or composition to cells for screening purposes and/or for conducting quality control assays. [0155] Provided herein is a method of treating a disease or disorder in an individual in need thereof comprising administering a compound described herein or any embodiment, variation, or aspect thereof, or a pharmaceutically acceptable salt thereof. In some

embodiments, the compound, pharmaceutically acceptable salt thereof, or composition is administered to the individual according to a dosage and/or method of administration described herein.

[0156] The compounds or salts thereof described herein and compositions described herein are believed to he effective for treating a variety of diseases and disorders. In some embodiments, a compound or salt thereof described herein or a composition described herein may be used in a method of treating a disease or disorder mediated by fibroblast activation protein (FAP). In some embodiments, the disease or disorder is characterized by proliferation, tissue remodeling, fibrosis, chronic inflammation, excess alcohol consumption, or abnormal metabolism.

[0157] In some embodiments, a compound or salt thereof described herein or a composition described herein may be used in a method of treating a disease or disorder mediated by a physiological substrate of FAP peptidase activity. In some embodiments the FAP peptidase activity is endopeptidase activity. In some embodiments, the physiological substrate of FAP endopeptidase activity is a2-antiplasmin, type I collagen, gelatin, and Fibroblast growth factor 21 (FGF21). In some embodiments the FAP peptidase activity is exopeptidase activity. In some embodiments, the physiological substrate of FAP exopeptidase activity is Neuropeptide V . El- type natriuretic peptide, substance P and peptide YY. In some embodiments, a compound or salt thereof described herein or a composition described herein may be used in a method of treating a disease or disorder mediated by FGF21.

[0158] In some embodiments, a compound or salt thereof described herein or a composition described herein may be used m a method of treating a FGF2l-associated disorder, such as obesity, type I-and type II diabetes, pancreatitis, dyslipidemia, hyperlipidemia conditions, non-alcoholic fatty liver disease (NAFLD), non-alcoholic steatohepatitis (NASH), insulin resistance, hyperinsulinemia, glucose intolerance, hyperglycemia, metabolic syndrome, acute myocardial infarction, hypertension, cardiovascular· diseases, atherosclerosis, peripheral arterial disease, apoplexy, heart failure, coronary arter' heart disease, renal disease, diabetic complications, neuropathy, gastroparesis, disorder associated with a serious inactivation mutation in insulin receptor, and other metabolic disorders. In some embodiments, the FGF21- associated disorder is diabetes, obesity, dys!ipidemia, metabolic syndrome, non-alcoholic fatty liver disease, non-alcoholic steatohepatitis or cardiovascular diseases.

[0159] In some embodiments, a compound or salt thereof described herein or a composition described herein may be used in a method of treating a disease or disorder characterized by proliferation, tissue remodeling, fibrosis, chronic inflammation, excess alcohol consumption, or abnormal metabolism.

[0160] In some embodiments, a compound or salt thereof described herein or a composition described herein may be used in a method of treating cancer, such as breast cancer, colorectal cancer, ovarian cancer, prostate cancer, pancreatic cancer, kidney cancer, lung cancer, melanoma, fibrosarcoma, bone sarcoma, connective tissue sarcoma, renal cell carcinoma, giant cell carcinoma, squamous cell carcinoma, leukemia, skin cancer, soft tissue cancer, liver cancer, gastrointestinal carcinoma, or adenocarcinoma in some embodiments, the compound, salt, or composition may be used in a method of treating metastatic kidney cancer, chronic

lymphocytary leukemia, pancreatic adenocarcinoma, or non-small cell lung cancer.

[0161] In some embodiments, the administration of the compound, salt, or composition reduces tumor growth, tumor proliferation, or tumorigenicity in the individual . In some embodiments, the compound, salt, or composition may be used in a method of reducing tumor growth, tumor proliferation, or tumorigenicity in an individual in need thereof. In some embodiments, tumor growth is slowed or arrested. In some embodiments, tumor growth is reduced at least about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or more. In some embodiments, the tumor is reduced in size. In some embodiments, tumor metastasis is prevented or slowed. In some embodiments, the tumor growth, tumor proliferation, or tumorigenicity is compared to the tumor growth, tumor proliferation, or tumorigenicity in the individual prior to the administration of the compound, salt, or composition. In some embodiments, the tumor growth, tumor proliferation, or tumorigenicity is compared to the tumor growth, tumor proliferation, or tumorigenicity in a similar individual or group of individuals. Methods of measuring tumor growth, tumor proliferation, and tumorigenicity are known m the art, for example by repeated imaging of the indi vidual.

[0162] In some embodiments, a compound or salt thereof described herein or a composition described herein may be used m m a method of treating fibrotic disease. thrombosis, wound healing, keloid formation, osteoarthritis, rheumatoid arthritis and related disorders involving cartilage degradation, atherosclerotic disease, Crohn’s disease, hepatic cirrhosis, idiopathic pulmonary fibrosis, myocardial hypertrophy, diastolic dysfunction, obesity, glucose intolerance, insulin insensitivity, or diabetes mellitus. In some embodiments, the hepatic cirrhosis is viral hepatitis-induced, alcohol-induced, or biliary cirrhosis. In some embodiments, the diabetes mellitus is type 11 diabetes. In some embodiments, the disease or disorder is fibrotic liver degeneration.

[0163] In some embodiments, provided herein is a method of inhibiting FAP. The compounds or salts thereof described herein and compositions described herein are believed to be effective for inhibiting FAP.

[0164] In some embodiments, the method of inhibiting FAP comprises inhibiting FAP in a cell by administering or delivering to the cell a compound described herein, or a

pharmaceutically acceptable salt thereof, or a pharmaceutical composition described herein. In some embodiments, the cell is a fibroblast, such as a myofibroblast, a keloid fibroblast, a cancer associated fibroblast (CAF), or a reactive stromal fibroblast, among others ceils with FAP expression.

[0165] In some embodiments, the method of inhibiting FAP comprises inhibiting FAP in a tumor or in plasma by administering or delivering to the tumor or plasma a compound described herein, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition described herein.

[0166] In some embodiments, the inhibition of FAP comprises inhibiting an

endopeptidase and/or exopeptidase activity of FAP. In some embodiments, FAP is inhibited by at least about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 98% or more. Inhibition of FAP can be determined by methods known in the art

[0167] In some embodiments, the compound, salt thereof, or composition inhibits FAP with an IC50 of less than about 1 mM, such as less than about 750 nM, 600 nM, 500 nM, 300 nM, 200 nM, 100 nM, 80 nM, 60 nM, 40 nM, 25 nM, or less. In some embodiments, the compound, salt thereof, or composition inhibits FAP with an IC50 between about 7 nM and 1 mM, such between about 10 nM and 600 nM, 15 nM and 200 nM, or 20 nM and 180 nM. In some aspects, the half maximal inhibitory' concentration (!€%) is a measure of the effectiveness of a substance in inhibiting a specific biological or biochemical function. In some aspects, the ICso is a quantitative measure that indicates how much of an inhibitor is needed to inhibit a given biological process or component of a process such as an enzyme, cell, cell receptor or microorganism by half. Methods of determining IC50 in vitro and in vivo are known in the art.

[0168] In some embodiments, the compounds or salts thereof described herein and compositions described herein are administered in an amount wherein DPPTT, DPPIV, DPP8, DPP9, and/or PREP activity is not inhibited or is inhibited to a lesser extent. In some embodiments, inhibition of FAP is at least or at least about 2 fold greater than inhibition of DPPII, DPPIV, DPP8, DPP9, and/or PREP activity, for example at least or at least about 3 fold, 4 fold, 5 fold, 8 fold, 10 fold, 15 fold, 30 fold, 50 fold, 60 fold, 75 fold, or 100 fold greater.

[0169] Provided herein is a method of enhancing an immune response in an individual comprising administering to the individual a compound described herein, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition described herein. In some

embodiments, the individual has cancer in some embodiments, the enhanced immune response is directed to a tumor or cancerous cell. By way of example and not wishing to be bound by theory, FAP is believed to suppress immune responses, especially in the context of cancer, therefore inhibiting FAP may enhancing the immune response of an individual. Accordingly, provided herein are methods of treating cancer in an individual in need thereof comprising administering to the individual a compound described herein, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition described herein, wherein an immune response of the individual is increased.

[0170] Provided herein is a method of increasing the level of FGF21 expression in an individual comprising administering to the individual a compound described herein, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition described herein. Also provided herein is a method of increasing the level of FGF21 or an FGF21 analog in an individual comprising administering to the individual a compound described herein, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition described herein. In some embodiments, the method further comprises administering FGF21 or an FGF21 analog, such as a mutated FGF21, pegylated FGF21, PF-05231023, or LY2405319.

[0171] FGF21 is a peptidic endocrine hormone secreted primarily by the liver (Markan,

K.R. et al. Semin Cell Dev Biol, 2016, 53: 85-93). Upon entering circulation, FGF21 functions by signaling to specific tissues regulating carbohydrate and lipid metabolism (Kharitonenkov, A., et al., I Clin Invest, 2005, 115(6): 1627-35). FGF21 stimulates glucose uptake in adipocytes and is believed to protective against obesity and insulin insensitivity. Pharmacological administration of FGF21 to diabetic and obese animal models markedly ameliorates obesity, insulin resistance, dyslipidemia, fatty liver, and hyperglycemia in rodents (Markan, K.R. et al. Semin Cell Dev Biol, 2016, 53: 85-93). Small clinical trials have demonstrated that FGF21 analogs are efficacious in inducing weight loss and correcting hyperinsulinemia, dyslipidemia, and hypoadiponectinemia in obese individuals with type 2 diabetes (Gaich, G., et al., Cell Metab, 2013, 18(3): p 333-40; Dong, J.Q., et al., Br J Clin Pharmacol, 2015, 80(5): 1051-63.

[0172] By way of example and not wishing to be bound by theory , FAP is believed to be the enzyme responsible for cleavage and inactivation of FGF21; therefore inhibiting FAP may increase levels of FGF21 expression and may augment endogenous and/or exogenous FGF21 action. FGF2 ! interacts with FGFR1 through its N-terminus and with b-Klotho through its C- terminus. This C -terminal region of FGF21 is essential to activate the receptor complex to initiate signaling (Micanovic, R, et al., J Cell Physiol, 2009, 219(2): 227-34; Yie, J., et al.,

FEES Lett, 2009, 583(1): 19-24). Recently, FAPa has been identified as the protease responsible for the inactivation of circulating FGF21 through the C -terminal cleavage at Pro! 71 (Dunshee, D.R., et al., J Biol Chem, 2016, 291(11): 5986-96; Coppage, A.L., et al., PLoS One, 2016, 11(3): e0151269; Zhen, E.Y., et al, Biochem J, 2016, 473(5): 605-14). In rodents and primates, the half-life of exogenously administrated human FGF21 is short (~ 0.5-2 h) as result of FAP- mediated enzymatic degradation and susceptibility to renal clearance (Hager, T., et al., Anal Chem, 2013, 85(5): 2731-8; Xu, L, et al., Am J Physiol Endocrinol Metab, 2009, 297(5): El 105- 14; Kharitonenkov, A., et al. Endocrinology', 2007, 148(2): 774-81). Common half-life extension strategies have improved significantly the PK properties of these FGF21 analogs in vivo; however, proteolytic processing still persists in these analogs (Hecht, R., et al., PLoS One, 2012, 7(11): e49345; Mu, J., et al., Diabetes, 2012, 61(2): 505-12; Camacho, R.C., et al., Eur J Pharmacol, 2013, 715(1-3): 41 -5).

[0173] Accordingly, provided herein are methods of treating diabetes mellitus, insulin insensitivity, and/or obesity in an individual m need thereof comprising administering to the individual a compound described herein, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition described herein. In some embodiments, the method further comprises administering FGF21 or an FGF21 analog. In some embodiments, the FGF21 analog

* 7 ^ is pegylated FGF21, PF-05231023, or LY2405319. Also provided herein are methods of treating diabetes mellitus, insulin insensitivity, and/or obesity in an individual in need thereof comprising administering to the individual a compound described herein, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition described herein, wherein FGF21 expression is increased. In some embodiments, the diabetes mellitus is type II diabetes.

[0174] In some embodiments, the individual is a mammal. In some embodiments, the individual is a primate, bovine, ovine, porcine, equine, canine, feline, iapine, or rodent. In some embodiments, the individual is a human. In some embodiments, the individual has any of the diseases or disorders disclosed herein. In some embodiments, the individual is a risk for developing any of the diseases or disorders disclosed herein.

[0175] In some embodiments, the individual is human. In some embodiments, the human is at least about or is about any of 21, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, or 85 years old. In some embodiments, the human is a child. In some embodiments, the human is less than about or about any of 21, 18, 15, 12, 10, 8, 6, 5, 4, 3, 2, or 1 years old.

[0176] Also provided herein are uses of a compound described herein or a

pharmaceutically acceptable salt thereof, or a pharmaceutical composition described herein, in the manufacture of a medicament. In some embodiments, the manufacture of a medicament is for the treatment of a disorder or disease described herein. In some embodiments, the manufacture of a medicament is for the prevention and/or treatment of a disorder or disease mediated by FAP.

Combination Therapy

[0177] As provided herein, compounds or salts thereof described herein and

compositions described herein may be administered with an additional agent to treat any of the diseases and disorders disclosed herein.

[0178] In some embodiments, (a) a compound described herein, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition described herein and (b) an additional agent are sequentially administered, concurrently administered or simultaneously administered. In certain embodiments, (a) a compound described herein, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition described herein and (b) an additional agent are administered with a time separation of about 15 minutes or less, such as about any of 10, 5, or 1 minutes or less. In certain embodiments, (a) a compound described herein, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition described herein and (b) an additional agent are administered with a time separation of about 15 minutes or more, such as about any of 20, 30, 40, 50, 60, or more minutes. Either (a) a compound described herein, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition described herein and (b) an additional agent may be administered first in certain embodiments, (a) a compound described herein, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition described herein and (b) an additional agent are administered simultaneously.

[0179] In some embodiments, the additional agent targets an immune checkpoint protein.

In some embodiments, the additional agent is an antibody that targets an immune checkpoint protein. In some embodiments, the additional agent targets PD-1, PD-LI, PD-L2, CTLA4, TIMS, LAGS, CCR4, 0X40, OX40L, IDO, and A2AR. In some embodiments, the additional agent is an anti-PD-l antibody, an anti-PD-Ll antibody, or an anti-CTLA-4 antibody.

[0180] In some embodiments, the additional agent is an inducer of FGF21 expression, such as a PPARtx agonist. In some embodiment, the PPARa agonist is fibrate or fenofibrate. In some embodiments, the additional agent is FGF-21 or an FGF-21 analog. In some

embodiments, the FGF-21 analog is a mutated FGF21 and/or pegylated FGF21. In some embodiments, the FGF-21 analog is PF-05231023 or LY24G53I9.

[0181] In some embodiments, the additional agent is a KLB/FGFR complex agonist, a

DDPIV antagonist, a GLP-1 receptor agonist, or a glucagon receptor agonist.

[0182] Provided herein is a method of enhancing an immune response in an individual comprising administering to the individual (a) a compound described herein, or a

pharmaceutically acceptable salt thereof, or a pharmaceutical composition described herein and (b) an agent that targets an immune checkpoint protein. In some embodiments, the individual has cancer. In some embodiments, the enhanced immune response is directed to a tumor or cancerous cell.

[0183] Also provided herein are methods of treating cancer in an individual in need thereof comprising administering to the individual (a) a compound described herein, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition described herein and (b) an agent that targets an immune checkpoint protein, wherein an immune response of the individual is increased. [0184] Provided herein is a method of increasing the level of FGF21 expression in an individual comprising administering to the individual (a) a compound described herein, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition described herein and (b) an agent that induces FGF21 expression.

[0185] Also provided herein are methods of treating diabetes mellitus, insulin insensitivity, and/or obesity in an individual m need thereof comprising administering to the individual (a) a compound described herein, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition described herein and (b) an agent that induces FGF2 ! expression, wherein FGF21 expression is increased. In some embodiments, the diabetes mellitus is type II diabetes.

[0186] As provided herein, compounds or salts thereof described herein and

compositions described herein are administered as part of a treatment regimen that includes an exercise regimen, such as strength- training or cardiovascular exercise. In some embodiments, the compounds or salts thereof described herein and compositions described herein are administered with an additional agent and as part of a treatment regimen that includes an exercise regimen, such as strength-training or cardiovascular exercise. In some embodiments, the exercise regimen comprises exercising at least once per week, such as twice per week, 3x per week, 4x per week, 5x per week, 6x per week, or 7x per week. In some embodiments, the exercise regimen comprises exercising at least one day per week, such as two days per week, 3 days per week, 4 days per week, 5 days per week, 6 days per week, or 7 days per week. In some embodiments, the exercise regimen comprises exercising once per day, twice per day, or 3x per day. In some embodiments, the exercise regimen comprises exercising for at least 10 minutes per session, such as for at least 15 min, 20 min, 25 m , 30 min, 35 min, 40 min, 45 min, 50 min, 55 min, 1 hour, 1.25 hours, or 1.5 hours.

Dosing and Method of Administration

[0187] The dose of a compound administered to an indi vidual (such as a human) may vary with the particular compound or salt thereof, the method of administration, and the particular disease, such as type and stage of cancer, being treated. In some embodiments, the amount of the compound or salt thereof is a therapeutically effective amount. [0188] The effective amount of the compound may in one aspect be a dose of between about 0.01 and about 100 mg/kg. Effective amounts or doses of the compounds of the invention may be ascertained by routine methods, such as modeling, dose escalation, or clinical trials, taking into account routine factors, e.g., the mode or route of administration or drug delivery, the pharmacokinetics of the agent, the severity and course of the disease to be treated, the subject’s health status, condition, and weight. An exemplary dose is in the range of about from about 0.7 mg to 7 g daily, or about 7 mg to 350 mg daily, or about 350 mg to 1.75 g daily, or about 1.75 to 7 g daily.

[0189] Any of the methods provided herein may in one aspect comprise administering to an individual a pharmaceutical composition that contains an effective amount of a compound provided herein or a salt thereof and a pharmaceutically acceptable excipient.

[0190] A compound or composition of the invention may be administered to an individual in accordance with an effective dosing regimen for a desired period of time or duration, such as at least about one month, at least about 2 months, at least about 3 months, at least about 6 months, or at least about 12 months or longer, which in some variations may be for the duration of the individual’s life in one variation, the compound is administered on a daily or intermittent schedule. The compound can be administered to an individual continuously (for example, at least once daily) over a period of time. The dosing frequency can also be less than once daily, e.g., about a once weekly dosing. The dosing frequency can be more than once daily, e.g., twice or three times daily. The dosing frequency can also be intermittent, including a ’drug holiday’ {e.g., once daily dosing for 7 days followed by no doses for 7 days, repeated for any 14 day time period, such as about 2 months, about 4 months, about 6 months or more). Any of the dosing frequencies can employ any of the compounds described herein together with any of the dosages described herein.

Articles of Manufacture and Kits

[0191] The present disclosure further provides articles of manufacture comprising a compound described herein or a salt thereof, a composition described herein, or one or more unit dosages described herein in suitable packaging. In certain embodiments, the article of manufacture is for use in any of the methods described herein. Suitable packaging is known m the art and includes, for example, vials, vessels, ampules, bottles, jars, flexible packaging and the like. An article of manufacture may further be sterilized and/or sealed.

[0192] The present disclosure further provides kits for camring out the methods of the invention, which comprises one or more compounds described herein or a composition comprising a compound described herein. The kits may employ any of the compounds disclosed herein. In one variation, the kit employs a compound described herein or a salt thereof. The kits may be used for any one or more of the uses described herein, and, accordingly, may contain instructions for the treatment any disease or described herein, for example for the treatment of cancer.

[0193] Kits generally comprise suitable packaging. The kits may comprise one or more containers comprising any compound described herein. Each component (if there is more than one component) can be packaged in separate containers or some components can be combined in one container where cross-reactivity and shelf life permit.

[0194] The kits may be in unit dosage forms, bulk packages (e g., multi-dose packages) or sub-unit doses. For example, kits may be provided that contain sufficient dosages of a compound as disclosed herein and/or an additional pharmaceutically active compound useful for a disease detailed herein to provide effective treatment of an individual for an extended period, such as any of a week, 2 weeks, 3 weeks, 4 weeks, 6 weeks, 8 weeks, 3 months, 4 months, 5 months, 7 months, 8 months, 9 months, or more. Kits may also include multiple unit doses of the compounds and instructions for use and be packaged in quantities sufficient for storage and use in pharmacies (e.g., hospital pharmacies and compounding pharmacies).

[0195] The kits may optionally include a set of instructions, generally written instructions, although electronic storage media (e.g., magnetic diskette or optical disk) containing instructions are also acceptable, relating to the use of component(s) of the methods of the present invention. The instructions included with the kit generally include information as to the components and their administration to an individual.

[0196] The invention can be further understood by reference to the following examples, which are provided by way of illustration and are not meant to be limiting.

[0197] Ail references throughout, such as publications, patents, patent applications and published patent applications, are incorporated herein by reference in their entireties.

The following enumerated embodiments are representative of some aspects of the invention.

Embodiment 1. A compound of formula (I):

or a pharmaceutically acceptable salt thereof, wherein:

R is hydrogen, Ci-Ce alkyl, C 3 -C 8 cycloalkyl, 3- to 12-membered heterocydyl, 5- to 10- membered heteroaryl, or C 6 -Ci4 and, wherein the Ci-Cg alkyl, C 3 -C 8 cycloalkyl, 3- to 12- membered heterocydyl, 5- to 10-membered heteroaryl, and Ce-C^ aryl of R are independently optionally substituted by R d ; m is 0, 1, 2, 3, or 4; n is 0, 1, 2, 3, or 4, wherein m + n is 1, 2, 3, or 4;

X is -( ' { OK -0-, -CH(OH)-, -S-, -Si O}··. or -Si O}·-;

L is . wherein represents the point of attachment to the Y-X- moiety, represents the point of attachment to the remainder of the molecule,

R a is hydrogen, C -Ce alkyl, C 3 -C 8 cycloalkyl, 3- to 12-membered heterocydyl, 5- to 10-membered heteroaryl, or Ce-Cn aryl, wherein the Ci-Ce alkyl, C 3 -C 8 cycloalkyl, 3- to 12-membered heterocycly], 5- to 10-membered heteroaxyl, and Ce~Ci4 aryl of R a are independently optionally substituted by R c ,

R 1 and R , independently of each other and independently at each occurrence, are hydrogen, C3-C2 alkyl, C 3 -C 3 cycloalkyl, 3- to 12-membered heterocyclyl, 5- to l O-membered heteroaxyl, or C 6 -C l4 aryl, wherein the C3-C8 cycloalkyl, 3- to 12- membered heterocyclyl, 5- to 10-membered heteroaxyl, and Ce-Cu ary'l of R 1 and R 2 are independently optionally substituted by R f , or R 1 and R are taken together with the carbon atom to which they are attached to form a 3- to 8-membered cycloalkylene optionally substituted by R , q is l, 2, or 3,

R J and R 4 , independently of each other and independently at each occurrence, are hydrogen, C -Cg cycloalkyl, 3- to 12-membered heterocyclyl, 5- to 10-membered heteroaryd, or Ce-Cn aryl, wherein the t Cg cycioalkyl, 3- to 12-membered heterocyclyl, 5- to 10-membered heteroaryd, and C 6 -C M aryl of R J and R 4 are independently optionally substituted by R s , or R 3 and R 4 are taken together with the carbon atom to which they are attached to form a 3- to 8-membered cycloalkylene optionally substituted by R 8 , and p is 0, 1, or 2; wherein

* represents the point of attachment to the Y-X- moiety,

** represents the point of attachment to the remainder of the molecule,

R 5 and R 6 , independently of each other and independently at each occurrence, are H, C -Ce alkyl, C 3 -Cg cycloalkyl, 3- to 12-membered heterocyclyl, 5- to 10- membered heteroaryd, or C6-C34 ary l, wherein the Ci-Ce alkyl, Ch-Cg cycloalkyl, 3- to 12-membered heterocyclyl, 5- to lO-membered heteroaryl, and Ce-Cw aryl of R 5 and R 6 are independently optionally substituted by R h ,

R b and R £ are independently H, Ci-Ce alkyl, C 2 -C 6 alkenyl, CVCe alkynyl, Cg-Cg cycloalkyl, 3- to 12-membered heterocyclyl, 5- to lO-membered heteroaryl, C 6 - C l4 aryl, or -C( : =0)OR 1 ', wherein the C -C 6 alkyl, C 3 -Cg cycloalkyl, 3- to 12- membered heterocyclyl, 5- to lO-membered heteroaxyl, and Ce-Cu and of R b and R c are independently optionally substituted by R 1 , and r is 1, 2, or 3; wherein

* represents the point of attachment to the Y-X- moiety,

** represents the point of attachment to the remainder of the molecule,

R 7 and R 8 , independently of each other and independently at each occurrence, are hydrogen, Cg~Cg cycloalkyl, 3- to 12-membered heterocyclyl, 5- to 10-membered heteroaryl, or Ce-Cir aryl, wherein the Cg-Cg cycloalkyl, 3- to 12-membered heterocyclyl, 5- to 10-membered heteroaryl, and C 6 -C l4 aryl of R' and R 8 are independently optionally substituted by R J , or R' and R 8 are taken together with the carbon atom to which they are attached to form a 3- to 8-membered cydoalkylene optionally substituted by R’,

R 9 and R lu , independently of each other and independently at each occurrence, are H, Ci-Ce alkyl, Cg~Cg cycloalkyl, 3- to 12-membered heterocyclyl, 5- to 10- membered heteroaxyl, or Ce-C^ aryl, wherein the Ci-CV, alkyl, C 3 -Cg cycloalkyl, 3- to 12-membered heterocyclyl, 5- to 10-membered heteroaxyl, and C 6 -C l4 aryl of R 9 and R 10 are independently optionally substituted by R k , s is l, 2, or 3, t is 1, 2, or 3, wherein s + t is 2, 3, or 4, u is 0 or 1 , and v is 0 or 1;

Y is C6-C9 axyl optionally substituted by R 11 , 6- to 10-membered heteroaryl optionally substituted by R 12 , or 3- to 12-membered heterocyclyl optionally substituted by R ! i , wherein when Y is phenyl or naphthyl, the phenyl or naphthyl of Y is substituted by at least one R ! ! , and wherein when L is *-NH~CH 2~ ** and Y is optionally substituted quinolinyl, the optionally subs tituted quinolinyl of Y is connected to the parent s tructure at the 2-, 3-, 5-, 6-, 7-, or 8- position, wherein

R 1 R : and R 1 ’, independently of each other and independently at each occurrence, are Ci-Cg alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, Cs-Cg cycloalkyl, C 4 -C 8 cycloalkenyl, 3- to 12-membered heterocyclyl, 5- to lO-membered heteroaryl, Ce-C l4 aryl, halogen, cyano,

or -P(0)(0R ! ' )(0R !6 ), wherein each R n R 12 , and R i 3 is independently optionally substituted by R L ; each R 14 is independently hydrogen, C r C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C3- C 8 cycloalkyl, C6-C14 aryl, 5- to 10-membered heteroaiyl, or 3- to 12-membered heterocyclyl, wherein the Ci-Cg alkyl, C 2 -Ce alkenyl, C 2 -C 6 alkynyl, C3-C8 cycloalkyl, Ce-Cu aryl, 5- to 10- membered heteroaryl, and 3- to 12-membered heterocyclyl of R 14 are independently optionally substituted by halogen, -OH, oxo, cyano, or G.-Cg alkyl optionally substituted by halogen, -OH, or oxo;

R 15 and R 16 , independently of each other and independently at each occurrence, are hydrogen,

C -Ce alkyl, C?-C 6 alkenyl, C 2 -Cg alkynyl, C3-C8 cycloalkyl, Ce-Cw aryl, 5- to 10-membered heteroaiyl, or 3- to 12-membered heterocyclyl, wherein the Ch-Cg alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C -Cg cycloalkyl, C 6 -C l4 atyl, 5- to 1 Q-membered heteroard, and 3- to 12-membered heterocyclyl of R” and R 16 are independently optionally substituted by halogen, -OH, oxo, cyano, or C -Cg alkyl, optionally substituted by halogen, -OH, or oxo, or R i5 and R 16 are taken together with the atom to which they are attached to form a 3- to 6-membered heterocyclyl optionally substituted by halogen, oxo, cyano, or Ci-Cc, alkyl optionally substituted by halogen, -OH, or oxo;

R a , R e , R f , R g , R n , R 1 , R', and R k , independently of each other and independently at each occurrence, are halogen, Ci-C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C3-C8 eycloalkyl, C6-C14 and, 5- to lO-membered heteroaryl, 3- to 12-membered heterocyclyl, -OR 14 , -NR 15 R 16 , cyano, or nitro; and each R 1 IS independently halogen, Cj-Ce alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, CYCg eycloalkyl, C6-C 4 aryl, 5- to 10-rnembered heteroaryl, 3- lo [2-membered heterocyclyl, -OR 14 , ~C(0)R 14 , - NR ls R 16 , cyano, oxo, or nitro.

Embodiment 2. The compound of embodiment 1, or a salt thereof, wherein X is -C(=0)-.

Embodiment 3. The compound of embodiment 1, or a salt thereof, wherein X is -0-

Embodiment 4. The compound of embodiment 1, or a salt thereof, wherein X

is -Ci !iGl l K

Embodiment 5. The compound of any one of embodiments 1 to 4, or a salt thereof, wherein L is -NH-CR^-.

Embodiment 6 The compound of embodiment 5, or a salt thereof, wherein L is -NH-CH 2 -

Embodiment 7. The compound of embodiment 5, or a salt thereof, wherein L is -NH-

CH(CH 3 )-.

Embodiment 8. The compound of embodiment 5, or a salt thereof, wherein L is -NH- CR R -. wherein R 1 and R 2 are taken together with the carbon atom to which they are attached to form a 3- to 8-membered cycloalkylene.

Embodiment 9. The compound of embodiment 8, or a salt thereof, wherein R 1 and R 2 are taken together with the carbon atom to which they are attached to form a cyclopropylene. Embodiment 10. The compound of any one of embodiments 1 to 4, or a salt thereof, wherein L is

-CR 5 R 6 -CH(NR b R c )-.

Embodiment 11. The compound of embodiment 10, or a salt thereof, wherein L is -CR 5 R 6 - CH(NR b R ^ )-, wherein R u , R b , and R" are H, and R 5 is H or Ci-Ce alkyl.

Embodiment 12. The compound of any one of embodiments 1 to 4, or a salt thereof, wherein L is , wherein * represents the point of attachment to the Y-X- moiety, ** represents the point of attachment to the remainder of the molecule.

Embodiment 13. The compound of embodiment 12, or a salt thereof, wherein L is , wherein * represents the point of attachment to the Y-X- moiety, and * 3 represents the point of attachment to the remainder of the molecule.

Embodiment 14. The compound of any one of embodiments 1 to 13, or a salt thereof, wherein Y is C6-C9 a yl optionally substituted by R 11 , 6- to 10-membered heteroaryl optionally substituted by IV 2 , or 3- to 12-membered heterocyclyl optionally substituted by R 1 ’.

Embodiment 15. The compound of embodiment 14, or a salt thereof, wherein Y is Ce-Cg aryl optionally substituted by R 11 , wherein when Y is phenyl or naphthyl, the phenyl or naphthyl of Y is substituted by at least one R 11 .

Embodiment 16. The compound of embodiment 15, wherein Y is phenyl substituted by 1 to 5 R 11 , which are independently selected from halogen, trihalomethyl, cyano, and C(=0)NH .

Embodiment 17. The compound of embodiment 15, wherein Y' is unsubstituted 2,3- dihy d o- 1 H -inden-2-y 1.

Embodiment 18. The compound of embodiment 14, or a salt thereof, wherein Y^ is 6- to 10- membered heteroaryl optionally substituted by R , wherein when L is *-NH-CH 2 -** and Y is optionally substituted qumolinyl, the optionally substituted qumolinyl of Y is connected to the parent structure at the 2-, 3-, 5-, 6-, 7-, or 8-position.

Embodiment 19. The compound of embodiment 18, or a salt thereof, wherein either: (a.) L is *-CH 2 -CH(NH 2 )-** or *-CH(CH 3 )-CH(NH 2 )-** and Y is quinolin-4-yl optionally substituted by R l or

(b) L is *-NH-CH 2 -**, Y is quinolin-6-yl optionally substituted by R’ 2 , which R l is independently selected from -OH and phenyl.

Embodiment 20. The compound of embodiment 18, or a salt thereof, wherein Y is pyridm- 4-yl substituted by R lz in the 3-position.

Embodiment 21. The compound of embodiment 20, or a salt thereof, wherein R 12 is pyridiny! optionally substituted by Ci-Cr, alkyl.

Embodiment 22. The compound of embodiment 20, or a salt thereof, wherein R 12 is indolyl optionally substituted by Ci-CY alkyl.

Embodiment 23. The compound of embodiment 20, or a salt thereof, wherein R 12 is phenyl optionally substituted by Ci-Cg alkyl, halogen, or CrCg alkoxy.

Embodiment 24. The compound of embodiment 18, or a salt thereof, wherein Y is pyrimidin~4~yl optionally substituted by R 12 and optionally fused to C6-C l4 aryl or C5-C10 cycloalkyl, wherein Ce-Cu aryl and Cs-CThe compound of embodiment 24, or a salt thereof, wherein Y is pyrirnidin-4-yl fused to C 6 -Ci4 aryl, wherein C6-C 4 aryl is optionally substituted by R 2 .

Embodiment 26. The compound of embodiment 24, or a salt thereof, wherein Y is optionally substituted pyndin-3-yl, unsubstituted qumazolin-4-yl or unsubstituted 6,7-dihydro- 5//-cyclopenta[djpyrimidm-4-yl.

Embodiment 27. The compound of embodiment 18, or a salt thereof, wherein Y is 2/7- pyran-2-on-5~yl optionally substituted by R lz and optionally fused to Ce-Cu and, which Ce-Cw aryl is optionally substituted by R l .

Embodiment 28. The compound of embodiment 27, or a salt thereof, wherein Y is 17/- isochromen-l-on-4-yl optionally substituted by halogen. Embodiment 29. The compound of embodiment 18, or a salt thereof, wherein Y is pyridin- 2(li/)~on-5-yl optionally substituted by R 12 and optionally fused to Ce-C l4 aryl or 5- to 10- membered heterocyclyl, which C & -C l4 aryl or 5- to lO-membered heterocyclyl, independently of each other and independently at each occurrence, are optionally substituted by R l .

Embodiment 30. The compound of embodiment 29, or a salt thereof, wherein Y is pyridin- 2(l//)-on-5-yl optionally substituted by CrCe alkyl or Ce-C^ aryl.

Embodiment 31. The compound of embodiment 29, or a salt thereof, wherein Y is unsubstituted 7,8,9,l0-tetrahydropyrido[l,2-a]azepin-4(6E/)-on-l-yl.

Embodiment 32. The compound of embodiment 29, or a salt thereof, wherein Y is isoquin olin-l(2//)-on-4~yl optionally substituted by halogen, Ch-Cg alkyl, C 6 -Ci aryl, or C 3 -Cs cycloalkyl.

Embodiment 33. The compound of embodiment 14, or a salt thereof, wherein Y is 3- to 12- membered heterocyclyl optionally substituted by R 13 .

Embodiment 34. The compound of embodiment 33, or a salt thereof, wherein Y is unsubstituted isoindolin-2-yl.

Embodiment 35. The compound of embodiment 33, or a salt thereof, wherein Y is piperidin-2-on-5-yl optionally substituted by Ci-Cg alkyl or C 6 -Ci aryl.

Embodiment 36. The compound of any one of embodiments 1 to 35, or a salt thereof, wherein m = n = 1.

Embodiment 37. The compound of any one of embodiments 1 to 36, or a salt thereof, wherein R is hydrogen.

Embodiment 38. The compound of embodiment 1 , or a salt thereof, wherein the -X-L

moiety is selected from the group consisting of , wherein * represents the point of attachment to the Y moiety, and ** represents the point of attachment to the remainder of the molecule.

Embodiment 39. A pharmaceutical composition comprising a compound of any one of embodiments 1-38, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier.

Embodiment 40 A method of treating a disease or disorder mediated by fibroblast activation protein (FAP) in an individual in need thereof comprising administering to the individual a therapeutically effective amount of a compound of any one of embodiments 1-38, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition of embodiment 39.

Embodiment 41. A method of treating a disease or disorder characterized by proliferation, tissue remodeling, chronic inflammation, obesity, glucose intolerance, or insulin insensitivity in an individual in need thereof, comprising administering to the individual a therapeutically effective amount of a compound of any one of embodiments 1-38, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition of embodiment 39.

Embodiment 42, The method of embodiments 40 or 41 , wherein the disease or disorder is breast cancer, colorectal cancer, ovarian cancer, prostate cancer, pancreatic cancer, kidney- cancer, lung cancer, melanoma, fibrosarcoma, bone sarcoma, connective tissue sarcoma, renal cell carcinoma, giant cell carcinoma, squamous cell carcinoma, leukemia, skin cancer, soft tissue cancer, liver cancer, gastrointestinal carcinoma, or adenocarcinoma.

Embodiment 43. The method of embodiment 42, wherein the disease or disorder is metastatic kidney cancer, chronic lymphocytary leukemia, pancreatic adenocarcinoma, or non small cell lung cancer. Embodiment 44. The method of embodiments 40 or 41, wherein the disease or disorder is fibrotic disease, wound healing, keloid formation, osteoarthritis, rheumatoid arthritis and related disorders involving cartilage degradation, atherosclerotic disease, Crohn’s disease, or Type II diabetes.

Embodiment 45. A method of reducing tumor growth, tumor proliferation, or

tumorigenicity in an individual in need thereof, comprising administering to the individual a compound of any one of embodiments 1-38, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition of embodiment 39.

Embodiment 46. A method of inhibiting FAP in an individual comprising administering to the individual a compound of any one of embodiments 1-38, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition of embodiment 39.

Embodiment 47. A method of inhibiting FAP in a cell comprising administering or delivering to the cell a compound of any one of embodiments 1-38, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition of embodiment 39, or a metabolite of the foregoing.

Embodiment 48. The method of embodiment 47, wherein the ceil is a fibroblast.

Embodiment 49. The method of embodiments 47 or 48, wherein the ceil is a cancer associated fibroblast (CAF) or a reactive stromal fibroblast.

Embodiment 50. A method of inhibiting FAP in a tumor compri sing administering or delivering to the tumor a compound of any one of embodiments 1-38, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition of embodiment 39, or a metabolite of the foregoing.

Embodiment 51. A method of inln biting FAP m plasma comprising administering or delivering to the plasma a compound of any one of embodiments 1 -38, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition of embodiment 39, or a metabolite of the foregoing.

Embodiment 52. The method of any one of embodiments 46-51 , wherein inhibiting F AP comprises inhibiting an endopeptidase activity of FAP.

Embodiment 53. The method of any one of embodiments 46-51, wherein inhibiting FAP comprises inhibiting an exopeptidase activity of FAP.

Embodiment 54. A method of enhancing an immune response in an individual comprising administering (a) an immune checkpoint inhibitor and (b) a compound of any one of embodiments 1-38, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition of embodiment 39.

Embodiment 55. A method of increasing the level of FGF21 expression in an individual comprising administering to the individual a compound of any one of embodiments 1-38, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition of embodiment 39.

Embodiment 56. The method of embodiment 55, further comprising administering an inducer of FGF21 expression

Embodiment 57. The method of embodiment 56, wherein the inducer of FGF2 ! expression is PPARa agonist.

Embodiment 58. The method of embodiment 57, wherein the PPARa agonist is fibrate or fenofibrate.

Embodiment 59. The composition of embodiment 39 for use as a human or veterinary medicament.

Embodiment 60. Use of a compound of any one of embodiments 1-38, or a

pharmaceutically acceptable salt thereof, or a pharmaceutical composition of embodiment 39, in the manufacture of a medicament for the prevention and/or treatment of a disorder or disease mediated by FAP.

EXAMPLES

Synthetic Examples

[0199] The chemical reactions in the Synthetic Examples described can be readily adapted to prepare a number of oilier compounds of the invention, and alternative methods for preparing the compounds of this invention are deemed to be within the scope of this invention. For example, the synthesis of non-exemplified compounds according to the invention can be successfully performed by modifications apparent to those skilled in the art, e.g., by appropriately protecting interfering groups, by utilizing other suitable reagents known in the art other than those described, or by making routine modifications of reaction conditions.

Alternatively, other reactions disclosed herein or known in the art will be recognized as having applicability for preparing other compounds of the invention.

Example 1

Synthesis of ( S)-N-(2-(2-cyano-4 , 4-difluoropyrrolidin-l-yl)-2-oxoethyl)-l -oxo-1 2- dihydroisoquinoline-4-carboxamide

[Q2QQ] Compound la: To a stirred solution of 1 -(tert-buty 1) 2-methyl (S)-4- oxopyrroIidine-l,2-dicarboxylate (1.9 g, 7.36 mmol, 1.0 equiv.), in DCM (15 mL), was added DAST (2,6 ml, 19.85 mmol, 2.6 equiv) drop wise at 0 °C over a period of 30 min, the reaction mixture was stirred at RT for overnight. Progress of the reaction was monitored by NMR. Water (50 mL) was added to the reaction mixture, stirred for 5 min and extracted with DCM (50 mL c 3). Combined organic layer was washed with saturated sodium bicarbonate solution (100 mL), brine (100 mL), dried over anhydrous sodium sulphate and concentrated under reduced pressure to obtain 1 -(tert-buty 1) 2-methyl (5)-4,4-difluoropyrrolidine-l,2-dicarboxylate (1.25 g, 61 % Yield) as a red oil.

[0201] 1H NMR (400 MHz, DMSO-de) d 4.41 - 4.53 (m, 1H), 3.65 - 3.84 (m, 5H), 2.82 - 3.01 (m, 21 1). 1.41 (s, 11 1). 1.35 (s, 51 1). [Q2Q2] Compound lb: To a stirred solution of 1 -(tert-butyl) 2-methyl (5)-4,4- difluoropyrro!idine~l,2-dicarboxylate (200 mg, 0.75 mmol, 1.0 equiv.) in MeOH (1.0 mL) was added a solution of 7.0 M in ammonia in methanol (5.0 mL) drop wise at 0 °C. The mixture was allowed to stir at RT for 12 h. Progress of the reaction was monitored by NMR. After completion of reaction the solvent was evaporated under reduced pressure and the obtained crude material was crystallized by hexane and pentane to afford tert-butyl S)-2-carbamoyl-4,4- difluoropyrrolidine-l-carboxylate (185 mg, 98 % Yield) as an off-white solid.

[0203] 'l l NMR (400 MHz, DY!SO-d,.) d 7 48 (d, J = 15.16 Hz, i l l ). 7 16 (br. s . H I). 4.24 (dd, J= 5.87, 8.80 Hz, 1H), 3 58 - 3.83 (m, 2H), 2.77 (dd, ,/= 8 31, 13.69 Hz, 1H), 2.20 - 2 38 (m, 11 1). 1.26 - 1.55 (m, 9H).

[0204] Compound lc: To a stirred solution of tert-butyl (S)-2-carbamoyl-4,4- difluoropyrrolidine-l-carboxylate (100 mg,0.4mmol, l .Oequiv) in DCM (5 0 mL), pyridine (0.36 ml, 0.48 mmol, 1.2 equiv) was added dropwise at 0 °C. The reaction mixture was allowed to stir for 15 mm at 0 °C. Trifluoroacetic anhydride (54.72 mg, 0.48 mmol, 1.2 equiv) was added drop wise at 0 °C. The reaction mixture was allowed stir for 1 h at RT. The reaction progress was monitored by NMR and TLC, after completion of reacti on, quenched by water (20 mL), and extracted with ethyl acetate (50 mL c 3). Combined organic layer was washed with saturated NaHCO, solution (10 mL) and brine (20 mL), dried over anhydrous sodium sulphate and concentrated under reduced pressure. The crude product obtained was purified by combi- flash chromatography (0-30 % ethyl acetate in hexane as an eluent) to obtain tert-butyl (5)-2-cyano- 4,4-difluoropyrrolidine-l-carboxylate (70 mg, 76 % Yield).

[0205] 'l l NMR (400 MHz, DMSO-de) 6 4.96 (d, J = 8.80 Hz, 11 1 ). 3.65 - 3.89 (m, 21 1 ). 2,82 - 3.03 (ra, 1H), 2 75 (br. s., 1H), 1.35 - 1.56 (m, 9H).

[0206] Compound Id: To a stirred solution of tert- butyl (5)-2-cyano-4,4-difluoropyrro!idine- 1-carboxylate (250 mg, 1.07 mmol, 1.0 equiv) in acetonitrile (10 mL ) was added 4.0 M HC1 in dioxan (0.5 mL) drop wise at 0 °C over a period of 10 min. The resultant reaction mixture was allowed to stir at RT for 16 h. Progress of the reaction was monitored by TLC and NMR. The reacti on mixture was concentrated under reduced pressure, the crude material obtained was washed with ethyl acetate and hexane (1 : 1 (20 mL)) to obtain S)-4,4-difluoropyrrolidine-2- carbomtnle hydrochloride (70 mg, 38 % Yield) as an off-white solid. [0207] ! H NMR (400 MHz, DMSO-de) d 4 56 - 4.67 (m, 1 H), 3.27 - 3.50 (ra, 2H), 2 54 - 2.75 (m, 2H).

[0208] Compound If: To a stirred solution of (tert-butoxy carbonyl) glycine (921.8 mg, 5.26 mmoI,l .5equiv.) and HATU(2667 mg, 7.025 mmol, 2.0 equiv) in DMF (10 mL) was added (S)- 4,4-difluoropyrrolidine-2-carbonitrile hydrochloride (590 mg, 3.51 mmol, 1.0 equiv) and stirred for 10 min. DIPEA (1.8 mL, 10.53 mmol, 3.0 equiv) was added and the reaction mixture was allowed to stir for 16 h at RT. Progress of the reaction monitored by NMR and TLC. The reaction mixture was diluted with cold water (50 mL) and extracted with ethyl acetate (50 ml. c 3). Combined organic layer was washed with water (25 mi x 4), dried over anhydrous sodium sulphate and concentrated under reduced pressure. The crude material obtained was purified by combi flash chromatography to obtain tert-butyl (5)-(2-(2-cyano-4,4-difluoropyrrolidin-l-yl)-2- oxoethyl)carbamate (300 mg, 30 % Yield).

[0209] 1H NMR (400 MHz, DMSO-de) d 7.08 (t, J= 5.92 Hz, 1H), 4.98 - 5.16 (m, J f t } 4.09 - 4.24 (m, i l l }. 3.93 - 4.09 (m, 1H), 3.77 (d, J= 6.14 Hz, 2H), 2.71 - 2.97 (m, 2H), 1.31 - 1.42 (m 9H).

[0210] Compound !g: To a stirred solution of tert-butyl (ri)~(2~(2~cyano~4,4~

difluoropyrrolidin-l-y!)~2-oxoethyl)carbamate (300 mg, 1.03 mmol, 1.0 equiv) in acetonitrile (10 mL) was added 4.0M HC1 in dioxan (2 mL) dropwise at 0 °C over a period of 10 min. The mixture was allowed to stir at RT for 16 h. Progress of the reaction was monitored by TLC and NMR. The solvent was evaporated under reduced pressure to obtain residue which was washed with 20 mL ethyl acetate and hexane (1: 1) to obtain (5)-4,4-difluoro-l-glycylpyrrolidine-2- carbomtrile hydrochloride (200 mg, 86 % Yield) as an off-white solid.

[0211] 1H NMR (400 MHz, DMSO-de) d 8.31 (br. s , 2H), 5.18 (d, ./= 7.89 Hz, !H), 4.21 (d, J = 1 1.40 Hz, 1H), 3 97 - 4.10 (m, 1H), 3.94 (br. s., 1H), 3.82 (d, J= 12.28 Hz, 1 1 1). 2.81 - 2.97 (m, 2H).

[0212] Compound 1: To a stirred solution of l-oxo-l,2-dihydroisoquino!ine-4-carboxylic acid

(315 rag, 1.6 mmol, 1.5 equiv) and HATH (843mg,2.22 mmol, 2.0 equiv) in DMF (5 mL) was added (S)-4,4-difluoro-l-glycylpyrrolidine-2-carbonitrile hydrochloride (250 mg,l . i i mmol, 1.0 equiv) and stirred for 10 min. DIPEA (429 mg, 3.33 mmol, 3.0equiv.) was added and the mixture was allowed to stir at RT for 16 h. The reaction progress was monitored by NMR and TLC. The reaction mixture was diluted with cold water (50 mL) and extracted with ethyl acetate (50 ml, c 3). Combined organic layer was washed with water (50 ml. x 3), dried over anhydrous sodium sulphate and concentrated under reduced pressure. The crude product obtained was purified by reversed phase HPLC to obtain (5)-N-(2-(2-cyano-4, 4-difluoropyrrolidin-l -yl)-2-oxoethyl)-l - oxo-1, 2-dihydroisoquinoline-4-carboxamide (38 mg, 6 % Yield) as an off-white solid.

[0213] LCMS 361 | VI · I i |

[0214] *H NMR (400 MHz, DMSO-de) d 11.62 (br s., 1H), 8.68 (1 J = 5.62 Hz, TH), 8.25 (t ./ = 8.31 Hz, 2H), 7.75 it. ./ 7.09 Hz, 1H), 7.47 - 7.62 (m, 2H), 5.03 - 5.18 (m, 1H), 4.22 - 4.36

(m, 1H), 3.99 - 4.20 (m, 3H), 2.77 - 2.98 (m, 2H).

Example 2

Synthesis of (S)~N~(2~(2~cyano~4, 4-difluoropyrrolidin-l -yl)~2~oxoethyl)-i-oxo~2~phenyl-l, 2- dihydroisoquinoline-4-carboxamide

Compound 2

[0215] Compound 2a: To a stirred solution of l -oxo-l,2-dihydroisoquinoline-4-carboxylic acid (500 mg, 2.6 mmol, 1 equiv) in DCM (10 rnL), was added pheny!boronic acid (480 mg, 3.9 mmol, 1.5 equiv), copper acetate (2360 mg, 13 mmol, 5 equiv), molecular sieves and Et 3 N (4 mL, 26 mmol, 10 equiv). The reaction mixture was allowed to stir for overnight at RT. Progress of the reaction was monitored by TLC and LCMS. After completion of the reaction, the reaction mixture was passed through the celite® bed and the filtrate was concentrated under reduced pressure to obtain crude, which was diluted with water (100 mL) and washed with ethyl acetate (100 mL). Aqueous layer was acidified with 3N HC1 (30 mL) to pH-3 and extracted with ethyl acetate (100 ml, c 2). Organic layer was dried over anhydrous sodium sulphate and concentrated under reduced pressure to obtain l-oxo-2-phenyl-l , 2-dihydroisoquinoline-4-carboxylic acid (650 mg, 93%) as a white solid

[0216] LCMS 266 j M 1 f |

[0217] Compound 2: To a stirred solution of l-oxo-2-phenyl-l, 2-dihydroisoquinoline-4- carboxylic acid (200 mg, 0.75 mmol, 1 equiv) in DMF (5 mL), was added DIPEA (0.5 mL, 3 mmol, 4 equiv) and HATH (802 mg, 2.1 mmol, 2.8 equiv) and the reaction mixture was allowed to stir for 30 mm under nitrogen Atmosphere. S)-l-(2-aminoacetyl)-4,4-difluoropyrrolidine-2- carbonitnle hydrochloride (204 mg, 0.905 mmol, 1.2 equiv) was added to above mixture and allowed to stir for overnight at RT. Progress of the reaction was monitored by TLC and LCMS. After completion of the reaction, the reaction mixture was diluted with water (30 mL) and extracted with ethyl acetate (100 mL. c 2). Organic layer was washed with water (100 mL), brine solution (100 mL), dried over anhydrous sodium sulphate and concentrated under reduced pressure to obtain crude compound, which was purified by reversed phase HPLC to obtain (S)- N-(2-(2-cyano-4,4-difluoiOpyrrolidin~l-yi)-2-oxoethyl)-l-oxo -2-phenyl-l,2

dihydroisoquinoiine-4-carboxamide (Free base) (25 mg, 7 %) as a white solid compound.

[0218] LCMS 437 3 I M l ! |

[0219] ! HNMR (400 MHz, DMSO-^6) d 8.78 (t, J= 5.9 Hz, 1H), 8.35 - 8.27 (m, 2H), 7.82 (q, J= 5.3, 3.2 Hz, 2H), 7.66 - 7.54 (m, 5H), 7.51 (p, J= 4.6 Hz, 1H), 5.15 - 5.07 (m, 1H), 4.29 (ddd, J= 16.2, 1 1.9, 4.5 Hz, 1 1 1). 4.14 (qd, J ------ 18.6, 17.9, 7.2 Hz, 21 1 ). 3.29 is , 1 1 1). 2.99 - 2.73

(m, 2H).

Example 3

Synthesis of (S) -N-(2-(2-cyano-4,4-difiuoropyrroiidin-l-yl)-2-oxoethy!)-2-cy ciopropyl-l-oxo- l,2-dihydroisoquinoline-4-carboxamide

[0220] Compound 3a: To a stirred solution of 1 -oxo- 1 ,2-dihy droisoquinoline-4-carboxy lie acid (1000 mg, 5.2 mmol, 1 equiv) in DCM (15 mL), was added cyclopropylboronic acid (683 mg, 7.9 mmol, 1.5 equiv), copper acetate (4732 mg, 26 mmol, 5 equiv), molecular sieves and Et3N (7.2 mL, 52 mmol, 10 equiv). The reaction mixture was allowed to stir for overnight at RT. Progress of the reaction was monitored by TLC and LCMS. After completion of the reaction, the reaction mixture was passes through the celite bed and the filtrate was concentrated under reduced pressure to obtain crude, which was diluted with water (100 ml.) and extracted with ethyl acetate (100 mL). Aqueous layer was acidified with 3N HC1 (30 mL) to pH-3 and extracted with ethyl acetate (100 mL c 2). Organic layer was dried over anhydrous sodium sulphate and concentrated under reduced pressure to obtain 2-cyclopropyl- 1 -oxo- 1, 2- dihydroisoquinoline-4-carboxylic acid (550 mg, 45%) as a light brown solid compound.

[0221] LCMS 230 j M ! f j

[0222] Compound 3: To a stirred solution of 2-cyclopropyl- 1 -oxo- 1, 2-dihydroisoquinohne-4- carboxyhc acid (200 mg, 0.87 mmol, 1 equiv) in DMF (5 mL), wus added DIPEA (0.6 mL, 3.48 mmol, 4 equiv) and HATU (926 mg, 2 43 mmol, 2.8 equiv). The reaction mixture was allowed to stir for 30 min under nitrogen. (<S)-l-(2-aminoacetyl)-4,4-difluoropyrrolidine-2-carbonit rile hydrochloride (236 mg, 1.04 mmol, 1.2 equiv) wus added to above mixture and allowed to stir for overnight at RT. Progress of the reaction was monitored by TLC and LCMS. After completion of the reaction, the reaction mixture was diluted with water (30 mL) and extracted with ethyl acetate (100 mL c 2). Organic layer was washed with water (100 mL), brine solution (100 mL), dried over anhydrous sodium sulphate and concentrated under reduced pressure to obtain crude compound, which was purified by reversed phase HPLC to obtain (S)-N-(2-(2- cyano-4,4-difluoropyrrolidin-l-yl)-2-oxoethyl)-2-cyclopropy 1-1 -oxo-l, 2-dihydroisoquinoline-4- carboxamide (Free base) (60 mg, 17 %) as a white solid compound.

[0223] LCMS 437.3[M+H] +

[0224] 'i !NYIR (400 MHz, DMSO-d6) d 8.75 (t, J = 5.9 Hz, i l l ). 8.24 (dd, I = 19.7, 8.1 Hz, 2H ). 7.78 - 7 66{ . 2H), 7.55 (t, I = 7.5 Hz, 1H), 5.14 (d, J = 9.2 Hz, i 1 0.4.37 - 4.24 i ns. 0 0. 4 14 (qd, I = 17.0, 16.5, 7.9 Hz, 3H), 3.35(s , 1H), 2.88 (dd, j = 40.5, 13.1 Hz, 21 0.1 75 (s , OH), 1.05 (d, J = 7.3 Hz, 2H), 0.98 (s , 2H).

Example 4

Synthesis of (S)-N-(2-(2-cyano-4,4-difluoropyrrolidin-l-yl)-2-oxoethyl)qu inoline-6-carboxamide

[0225] To a stirred solution of quinoline-6-carboxylic acid (230 mg, 1.32 mmol, 1.0 equiv) and HATH (1003 mg, 2,64 mmol, 2.0 equiv) in DMF (15 ml) was added (S)-4,4-difluoro-l- glycylpyrrolidine-2-carbomtrile hydrochloride (250 mg, 1.32 mmol, 1.0 equiv.) and stirred the reaction mixture for 10 min. DIPEA (510 mg, 3.96 mmol, 3.0 equiv) was added and the reaction mixture was allowed to stir at RT for 16 h. The progress of the reaction was monitored by NMR and TLC. The reaction mixture was diluted with water (50 mL) and extracted with ethyl acetate (50 mL x 3). Combined organic layer was washed with water (25 mL c 4), dried over anhydrous sodium sulphate and concentrated under reduced pressure. The erode material obtained was purified by reversed phase HPLC to obtain (S)-N-(2-(2-cyano-4,4-difluoropynOlidin-l-yl)-2- oxoethyl)quinoline-6-carboxamide (175 mg, 38 % Yield) as an off-white solid.

[0226] LCMS 345

[0227] l H NMR (400 MHz, DMSO-de) d 11.62 (br s., 1H), 8.68 (t J = 5.62 Hz, 1H), 8.25 (t, ./ = 8.31 Hz, 2H), 7.75 (t. ./ 7.09 Hz, 1H), 7.47 - 7.62 (m, 2H), 5.03 - 5.18 (m, 1H), 4.22 - 4.36

(m, 1H), 3 99 - 4.20 (m, 3H), 2.77 - 2.98 (m, 2H).

Example 5 Synthesis of ( S)-N-(2-(2-cyano-4,4-difluoropyrroiidin-l-yl)-2-oxoethyi)-6 , 7-dihydro-5H- cyclopentafd/pyrimidine-4-carhoxamide

Compound 5

[Q228] To a stirred solution of (S)-4,4-difluoro-l-glycylpynOlidine~2~carbonitrile

hydrochloride (100 mg, 0.44 mmol, 1 equiv) in DMF (3 mL), was added quinoline-4-carboxy!ic acid (73 mg, 0.44 mmol, 1 equiv), Et 3 N (0.06 mL, 0.48 mmol, 1.1 equiv), HOBt (68 mg, 0.44 mmol, 1 equiv), DMAP (3 mg, 0.02 mmol, 0.05 equiv) and EDC.HCI (93 mg, 0.48 mmol, 1.1 equiv). The reaction mixture was allowed to stir the overnight at RT. Progress of the reaction was monitored by TLC and LCMS. After completion of the reaction, the reaction mixture was diluted with water (10 mL) and extracted with ethyl acetate (30 mL c 2). Organic layer was washed with water (50 mL), brine solution (50 mL). Organic layer was dried over anhydrous sodium sulphate and concentrated under reduced pressure to obtain crude compound, which was purified by normal phase combi flash to obtain (5)-N-(2-(2-cyano-4,4-difluoropyrrolidin~l-yi)-2- oxoethyl)-6,7-dihydro-5H-cyclopenta[d]pyrimidine-4-carboxami de (40 mg, 27 %) as a white solid compound.

[0229] LCMS 336 [ \ 1 f 11

[0230] 'l ! MR (400 MHz, DVlSi. /6) d 9.02 (s, M i). 5.09 - 5.01 (m, 1H), 4.33 - 3.92 (m, 4H), 3.24 (t, ./ 7.7 Hz, 2H), 2.97 (t, J ------ 7.8 Hz, 2H), 2.93 - 2.70 (m, 21 11 2.06 i n. J ------ 7.7 Hz,

2H).

Example 6

Synthesis of (S)-N-(2-(2-cyano-4, 4-difluoropyrroiidin-l-yl)-2-oxoethyl)-6-oxo-L 6- dihydropyridine-3-carboxamide

Compound

[0231] To a stirred solution of 6-oxo-l, 6-dihydropyridine-3-carboxylic acid (50 mg , 0.22 mmol, 1 equiv) in DMF (3 mL), was added Et ¾ N (0.03 mL, 0.24 mmol, 1.1 equiv), HOBt (34 mg, 0.22 mmol, 1 equiv), DMAP (1.3 mg, 0.01 mmol, 0.05 equiv) and EDC.HCI (47 mg, 0.24 mmol, 1.1 equiv). The reaction mixture was allowed to stir for overnight at RT. Progress of the reaction was monitored by TLC and LCMS. After completion of the reaction, the reaction mixture was diluted with water (10 mL) and extracted with ethyl acetate (30 mL x 2). Organic layer was washed with water (50 mL), brine solution (50 mL). Organic layer was dried over anhydrous sodium sulphate and concentrated under reduced pressure to obtain crude compound, which was purified by reversed phase HPLC to obtain (S)~N-(2-(2-cyano-4, 4- ditluoropyrrolidin~l~yl)-2-oxoethyl)~6-oxo-l, 6-dihydropyridine-3-carboxamide (Free base) (50 mg, 58 %) as a white solid compound.

[0232] LCMS 3 ! ! | M i 11

[0233] 'l IN.YIR (400 MHz, D.MSC /6) 6 ! 1.78 (s, 11 1). 8.60 (q, ,/ 7.8, 6.0 Hz, i l l ). 8 03 i d. J

= 2.7 Hz, 11 0.7.87 (dd, ,/= 9 6, 2.8 Hz, 1H), 6 37 (d, J= 9.6 Hz, 1H), 5.07 (dd, J= 9.3, 2.8Hz, 1H), 4.32 - 4.20 (m, 1H), 4.07 (qt, J= 16.8, 7.9 Hz, 3H), 2.97 - 2.71 (m, 2H).

Example 7

Synthesis of (S)-N-(2-(2-cyano-4, 4-difluoropyrrolidin-l-yl)-2-oxoethyl)-2, 3-dihydro- 1H- indene-2-carboxamide

Compound 7

[0234] To a stirred solution of 2, 3-dihydro-lH-indene-2-carboxylic acid (100 mg, 0.617 mmol, 1 equiv) in DMF (3 mL), was added DIPEA (0.4 mL, 2.46 mmol, 4 equiv) and HATU (657 mg, 1.72 mmol, 2.8 equiv) and the mixture was allowed to stir for 30 min. (5)-l-(2- aminoacetyl)-4,4-difiuoropyrrolidine-2-carbonitrile hydrochloride (167 mg, 0.74 mmol, 1.2 equiv) was added to above mixture and the resulting mixture was allowed to stir for overnight at RT. Progress of the reaction was monitored by LCMS. After completion of the reaction, the reaction mixture was diluted with water (30 mL) and extracted with ethyl acetate (50 mL x 2). Organic layer was washed with water (50 mi), brine solution (50 mL). Organic layer was dried over anhydrous sodium sulphate and concentrated under reduced pressure to obtain crude compound, which was purified by reversed phase HPLC to obtain (A)-N-(2-(2-cyano-4,4- difl uoropy rrolidin- 1 -yl)-2-oxoeth l)-2,3-dihy dro- 1 H-indene-2-carboxami de (Free base) ( 100 mg, 49 %) as a white solid compound.

[0236] ’l IN MR (400 MHz, DY1SC /6) 6 8.29 (t, J= 5.8 Hz, i l l }. 7.19 (dt, J= 7.3, 3.6 Hz, 2H ). 7.12 (dd, J= 5.5, 3.2 Hz, 21 0. 5 07 (dd, J = 9.3, 2.8 Hz, 11 1). 4.22 (ddd, ./= 15.8, 11.3, 4.5 Hz, 1H), 4.16 - 4 00 (m,lH), 3 97 (t, ./= 6.0 Hz, I I I ). 3.29 (dd, J= 17.8, 9.2 Hz, 2H), 3.08 (t, J = 6.3 Hz,4H), 2.96 - 2.71 (m, 2H).

Example 8

Synthesis of (S)-N-(2-(2-cyano-4 4-difluoropyrrolidin-l-yl)-2-oxoelhyl) isoindoline-2- carboxamide.

Compound 8

[Q237] To a stirred solution of (>S)-l-(2-aminoacetyl)-4, 4-difluoropyrrolidine-2-carbonitrile hydrochloride (50 mg, 0.22 mmol, 1 equiv) m DCM (3 raL), was added Et 3 N (0.2 ml , 1.1 mmol, 5 equiv). The reaction mixture was allowed to stir for 15 min under nitrogen. Isoindoiine (26 mg, 0.22 mmol, 1.0 equiv) was added to above mixture and cool the reaction mixture to 0 °C. Phosgene (20% in Toluene) (0.6 ml,) was added to above mixture drop wise. Raise the temperature to RT and allowed to stir for overnight at RT. Progress of the reaction was monitored by LCMS. After completion of reaction, the reaction mixture was diluted with water (30 niL) and extracted with DCM (50 mL x 2) Organic layer was washed with water (50 mL), brine solution (50 mL). Organic layer was dried over anhydrous sodium sulphate and concentrated under reduced pressure to obtain crude compound, which w as purified by reversed phase HPLC to obtain (S)-N-(2-(2-cyano-4, 4-difluoropyrroli din- 1 -y 1 )-2-oxoethy 1) isoindoline- 2-carboxamide (Free base) (20 mg, 27%) as a white solid compound.

[0238] LCMS 335.1 [M+H] +

[0239] 1HNMR (400 MHz, DVlSi.K/6) d 7.34 (dt, J = 7.1, 3.6 Hz, 2H), 7.31 - 7.25 (m, 2H), 6.70 (t, J= 5.8 Hz,IH), 5.07 (dd, J = 9.3, 2.9 Hz, 1 1 1). 4.62 (s, 4H), 4.24 (ddd, J= 16.1, 11.5, 4.6

Hz, 11 1 ). 4.15 - 4.01 (ns. i l l). 3.90 (t, ./ 5.3 Hz, 2! !). .97 - 2,71 (ra, 21 1 )

Example 9

Synthesis of (S)-N-(2-(2-cyano-4, 4-difluoropyrrohdin-l-yi)-2-oxoethyl)-2-methyl-l-oxo-l, 2- dihydroisoquinoline-4-carboxamide.

Compound 9

[0240] To a stirred solution of (,S)-4,4-difluoro-l-glycylpyrrolidine-2-carbonitrile

hydrochloride (100 mg, 0.44 mmol, 1 equiv) in DMF (3 mL), was added 2-methyl- l-oxo-l, 2- dihydroisoquinoiine-4-carboxylic acid (89 mg, 0.44 mmol, 1 equiv), Et < N (0.06 mL, 0.48 mmol, 1.1 equiv), HOBt (68 mg, 0.44 mmol, 1 equiv), DMAP (3 mg, 0.02 mmol, 0.05 equiv) and EDC.HCI (93 mg, 0.48 mmol, 1.1 equiv). The reaction mixture was allowed to stir for overnight at RT. Progress of the reaction was monitored by TEC and LCMS. After completion of the reaction, the reaction mixture was diluted with water (10 mL) and extracted with ethyl acetate (30 mL x 2). Organic layer was washed with water (50 mL), brine solution (50 mL). Organic layer was dried over anhydrous sodium sulphate and concentrated under reduced pressure to obtain crude compound, which was purified by normal phase combi flash to obtain (S)-N-(2-(2- cyano-4,4-difluoropynOlidin-l-yl)-2-oxoethyl)-2-methyl-l-oxo -l ,2-dihydroisoquinoline-4- carboxamide (Free base) (80 mg, 48 %) as a white solid compound.

[0241] LCMS 375 [M+H] +

[0242] *HNMR (400 MHz, DMSO-r 6) d 8.69 (t, J= 5.7 Hz, 1H), 8.26 (d, J= 8.1 Hz, 1H),

8.16 (d, J 8 2 1 Iz. I I I ). 7.83 (s, I I I ). 7.79 - 7.70 (m, I f ! ) 7.56 (t, ./ 7 6 Hz, 1H), 5.07 (dd, J = 9.5, 2.6 Hz, i l l ). 4.29 - 3.99(m, il l ). 3.54 (s, 3H), 2.97 - 2.70 (m, 21 1 ).

Example 10

Synthesis of (S)-N-(2-(2-cyano-4,4-difluoropyrrolidin-l-yl)-2-oxoethyl)-2 -hydroxyquinoline-6~ carboxamide

[0243] To a stirred solution of 2 -hydroxy quinoline-6-carboxylic acid (0.200 g, 1.05 mmol, 1.0 equiv) in DMF (5 mL), was added (,S)-4,4-difluoro-l -glycylpyrrolidme-2-carbomtrile hydrochloride (0.261 g, 1.16 mmol, 1.1 equiv), HOBt (0.156 g, 1.16 mmol, 1.1 equiv) and EDC.HCI (0.221 g, 1.16 mmol, 1.1 equiv). The mixture was allowed to stir at RT for 10 min. Triethyl amine (0.73 mL) was added and the mixture was allowed to stir at RT for overnight. Product formation was confirmed by LCMS and TLC The reaction mixture was diluted with water (50 mL) and extracted with ethyl acetate (50 mL x 2). Combined organic layer was washed with water (20 mL c 4), dried over anhydrous Na 2 SC>4 and concentrated. The crude material obtained was purified by reversed phase HPLC to obtain (5)-N-(2-(2-cyano-4,4- difluoropyrrolidin-l -y])-2-oxoethyl)-2-hydroxyquinoline-6-carboxamide (0.010 g, ~ 5 % Yield) as an off-white solid.

[0244] LCMS 361.2[M+H] +

[0245] *H NMR (400 MHz, DMSO-de) d 11.97 (br. s., 1 1 1). 8.80 (br. s., 11 1 )_ 8.23 (br. s„ i l l ). 7.89 - 8.04 (m, 21 1 ). 7 35 (d, ./ 8.33 Hz, 1H), 6.56 (d, J= 7.45 Hz, H I). 5.10 (d, J 10.96 Hz,

1H), 4.30 (br. s„ 21 1 ). 4.15 (d, J= 10.96 Hz, 21 1 ). 2 85 (br. s , 1H), 2.80 (br s., 11 1)

Example 11

Synthesis of ( S)-N-(2-(2-cyano-4,4-difluoropyrrolidin-l-yl)-2-oxoethyl)ter ephthalamide

Compound 11

[Q246] To a stirred solution of (<S)-4,4-difluoro-l-glycylpynOlidine-2-carbonitrile

hydrochloride (136 mg, 0.606 mmol, 1 equiv) in DMF (10 mL), was added 4-carbamoylbenzoic acid (100 mg, 0.606 mmol, 1 equiv), Et ¾ N (0.2 mL, 1.818 mmol, 3 equiv), HOBT (90 mg, 0.666 mmol, 1.1 equiv), EDC.HCL (128 mg, 0.666 mmol, 1.1 equiv). The reaction mixture was allowed to stir for overnight at RT. Progress of the reaction was monitored by TEC and LCMS. After completion of the reaction, the reaction mixture was diluted with water (50 mL) and extracted with ethyl acetate (50 mL x 2). Organic layer was washed with water (20 mL x 4), brine solution (50 mL). Organic layer was dried over anhydrous sodium sulphate and concentrated under reduced pressure to obtain crude compound, which was purified by normal phase combi flash to obtain (5)-N-(2-(2-cyano-4,4-difluoropyrrolidin-l-yl)-2- oxoethySjterephthalamide (Free base) (25 mg, 12 % Yield ) as an off white solid compound.

[0247] LCMS 337 [M+H] +

[0248] ! HNMR (400 MHz, DVISC /,.)□ 2.79 - 3.00 (m, 3 1 1 ) 4.04 - 4.22 (m, 3 1 1 } 4.24 - 4.41 (m, 1 H) 5.10 (d, J 7.02 Hz, 1 H) 7.51 (hr. s., 1 H) 7 83 - 8.02 (m, 3 H) 8.09 (hr. s„ 1 H) 8 92 (d, .7=5 70 Hz, 1 H).

Example 12

Synthesis of (S)-4-cyano-N-(2-(2-cyano-4A-difluoropyrrolidin-l-yl)-2-oxoe thyl)-3-

(trifluoromethyl)henzamide

[0249] Compound 12a. To a solution of 4-methyl-2-(trifluoromethyl)benzonitrile (0.790 g, 4.270 mmol, 1 equiv) in pyridine (8 ml.) and water (2 mL) was added KMn0 4 (0.675 g, 4.270 mmol, 1.0 equiv). The resultant reaction mixture was heated at 100 °C in microwave reactor for 1 h. Product formation was confirmed by LCMS (-ve mode mass). After completion of reaction, the mixture was filtered through celite bed and washed with ethyl acetate. Filtrate was acidified with 1 N HC! and extracted with ethyl acetate (50 mL c 2). Combined organic layer was dried over anhydrous Na 2 S0 4 and concentrated. The crude product obtained was purified by flash chromatography (0-50 % ethyl acetate in hexane as an eluent) to obtain 4-cyano-3- (triiluoromethyl)benzoic acid (0.200 g, 22 % Yield) as an off-white solid.

[0250] LCMS 214.2(M-1)

[0251] ! H NMR (400 MHz, DMSO-d6) d 8.66 (d, I = 4.38 Hz, 1H), 8.32 - 8.42 (m, 2H).

[0252] Compound 12. To a stirred solution of 4-cyano-3-(trifluoromethyl)benzoic acid (0.200 g, 0.930 mmol, 1.0 equiv) in DMF (5 mL), was added (S)-4,4-difluoro-l -glycylpyrrolidine-2- carbonitrile hydrochloride (0.210 g, 0.930 mmol, 1.0 equiv), HOBt (0.151 g, 1.116 mmol, 1.2 equiv) and EDC.HCI (0.212 g, 1.116 mmol, 1.2 equiv). The reaction mixture was allowed to stir at RT for 10 min. Tri ethyl amine (0.4 mL) was added and the mixture was allowed to stir at RT for overnight. Product formation was confirmed by LCMS and TLC After completion of reaction, the mixture was diluted with water (30 mL) and extracted with ethyl acetate (50 mL c 2). Combined organic layer was washed with water (20 mL c 4), dried over anhydrous Na 2 S0 4 and concentrated. The crude product obtained wus purified by flash chromatography (0-50 % ethyl acetate m hexane as an eluent) to obtain (5)-4-cyano-N~(2-(2-cyano-4,4~difiuoropyrrolidm- l-yl)-2-oxoethyl)-3-(trifluoromethyl)benzamide (0.060 g, 17 % Yield) as an off-white solid.

[0253] LCMS 387.1 [M+H] + [0254] ! H NMR (400 MHz, DMSO-d6) d 9 35 (br. s , IH), 8.41 (s, IH), 8.35 (s, 2H), 5.10 (d, I = 7 89 Hz, IH), 4.31 (L I = 11.62 Hz, IH), 4.05 - 4.22 (m, 2H), 2.77 - 3.00 (m, 2H), 2.73 (s,

IH).

Example 13

Synthesis of (S) N (2-(2-cyano-4,4 dijluoropyrroiidin-l-yl)-2-oxoeihyl)-3-phenylisonicotinaniid e

Compound 13

[0255] Compound 13a. To a solution of ethyl 3-bromoisonicotmate (1.00 g, 4.34 mmol, 1. equiv) in THF (20 rnL) was added phenylboronic acid (0.584 g, 4 78 mmol, 1.1 equiv), Na 2 C0 3 (0.922 g, 8.68 mmol, 2.0 equiv) followed by the addition of Pd(PPh3) 2 Cl 2 (0.153 g, 0.217 mmol. 0.05 equiv). The resulting reaction mixture was heated at 100 °C for overnight. Product formation was confirmed by LCMS. After the completion of reaction, the mixture was filtered through celite bed, washed with ethyl acetate (100 rnL). Filtrate was concentrated under reduced pressure. The crude product obtained was purified by flash chromatography (0-30 % ethyl acetate in hexane as an eluent) to obtain ethyl 3-phenylisonicotinate (0.300 g, 31 % Yield) as an off-white solid.

[0257] ! H NMR (400 MHz, DMSO-d6) d 8 68 - 8 76 (m, 2H), 7.67 (d, I = 4.82 Hz, 1H), 7.41 - 7.51 (m, 31 1). 7 37 (d, I = 6 14 Hz, 2H), 4.11 (q, J = 7.02 Hz, 2H), 0.98 (t, J = 7.24 Hz, 31 1 ).

[0258] Compound 13b. To a stirred solution of ethyl 3-phenylisonicotinate (0.250 g, 1.10 mmol, 1.0 equiv) in THF (5 mL) and water (5 mL), was added LiOH (0.053 g, 2.20 mmol, 2.0 equiv). The mixture was allowed to stir at RT for overnight. Product formation was confirmed by LCMS and ! H NMR Spectroscopy. The reaction mixture was diluted with water (15 mL) and washed with ethyl acetate (15 mL). Aqueous layer was separated and freeze dried on lyophilyzer to obtain 3-phenylisonicotinic acid (0.300 g, 99 % Yield) as an off-white solid.

[0259] LCMS 200. i I VI 1 11

[0260] ! H NMR (400 MHz, DVISO-d,.) d 8 30 - 8.47 (m, 2H), 7.58 (d, ./= 7.02 Hz, 2H), 7.26 - 7.45 (m, 3H), 7.13 (d, ./ 4.82 Hz, M l).

[0261] Compound 13. To a stirred solution of 3-phenylisomcotinic acid (0.200 g, 1.00 mmol, 1.0 equiv) in DMF (5 mL), was added (Y)-4,4-difluoro-l-glycylpyrrolidine-2-carbomtrile hydrochloride (0.224 g, 1 00 mmol, 1.0 equiv), HOBt (0.163 g, 1.206 mmol, 1.2 equiv) and EDC.HC1 (0.230 g, 1.206 mmol, 1.2 equiv). The mixture was allowed to stir at RT for 10 mm. Triethyl amine (0.7 mL) was added and the mixture was allowed to stir at RT for overnight. Product formation was confirmed by LCMS and TLC. After completion of reaction, the mixture was diluted with vrater (50 mL) and extracted with ethyl acetate (50 mL * 2). Combined organic layer was washed with water (20 mL c 4), dried over anhydrous Na 2 S0 4 and concentrated. The crude product obtained was purified by flash chromatography (0-50 % ethyl acetate in hexane as an eluent) to obtain (S)-N-(2-(2~cyano~4,4-difluoropyrrolidin~l-yl)-2~oxoethyl)-3 - phenylisonicotinamide (0.050 g, 14 % Yield) as an off-white solid.

[0262] LCMS 371 2| VI · 1 11

[0263] 1H NMR (400 MHz, DMSO-d6) d 8 96 (t, J - 5.48 Hz, ill), 8.62 - 8.70 (ra, 1H), 7 54 (d, J = 6 58 Hz, i l l ). 7.29 - 7.51 (m, 31 1 ). 5 10 (d, I = 7.02 Hz, 1 1 1). 4.21 (hr. s., 1H), 3.93 - 4.12 (m, 2H), 2.80 (d, J = 13.59 Hz, 2H).

Example 14

Synthesis of (S)-N-(2-(2-cyano-4, 4-difluoropyrrolidin-l-yl)-2-oxoethyl)-6-oxo-5-phenyl-l, 6- dihydropyridine-3-carboxamide

Compound 14

[Q264] To a stirred solution of (S)-4,4-difluoro-l-glycylpyrro]idine~2~carbonitrile

hydrochloride (50 mg, 0.22 mmol, 1 equiv) in DMF (2 mi), was added 6-oxo-5-phenyl-l,6- dihydropyridine-3-carboxylic acid (48 mg, 0.22 mmol, 1 equiv), EΪ3N (0.03 mL, 0.24 mmol, 1.1 equiv), HOBt (34 mg, 0.22 mmol, 1 equiv), DMAP (2 mg, 0.01 mmol, 0.05 equiv) and EDC.HCI (46 mg, 0.24mmol, 1.1 equiv). The reaction mixture was allowed to stir for overnight at RT. Progress of the reaction was monitored by TLC and LCMS. After completion of the reaction, the reaction mixture was diluted with water (10 mL) and extracted with ethyl acetate (30 mL x 2). Organic layer was washed with water (50 mL), brine solution (50 mL). Organic layer was dried over anhydrous sodium sulphate and concentrated under reduced pressure to obtain crude compound, which w¾s purified by normal phase combi flash to obtain (5)-N-(2-(2- cyano-4, 4-difluoropyrrolidin-l-yl)-2-oxoethyl)-6-oxo-5-phenyl-l, 6-dihydropyridine-3- carboxamide (Free base) (30 mg, 35%) as an off white solid compound.

[0265] LCMS 387 | \ I · I I |

[0266] ' l ! MR (400 MHz, DVISi.K/6) d 12.22 (s, 1H), 8.72 (t, J= 5.8 Hz, 11 I s. 8.11 (d, J = 2.7 Hz, 1H), 8.04(s, 1H), 7.74 i d. ./ 7.5 Hz, 2H), 7.42 (t, J= 7.5 Hz, 2H), 7.35 (t, ./ 7.2 Hz,

I f !) 5.09 Odd,/ 9.4, 2 8 Hz, l i t) 4.29 (ddd, J= 15.9, 1 1.6, 4.6 Hz, 1H), 4 18 - 3.93 (m, 31 1). 2.98- 2.70 (m, 2H).

Example 15

Synthesis of (S)-4-cyano-N-(2-(2-cyano-4, 4-difluoropyrrolidin-l-yl)-2-oxoethyl)-3- fluorohenzamide

Compound 15

[0267] To a stirred solution of (S)-4,4-difluoro-l-glycylpyrrolidine-2-carbonitrile

hydrochloride (100 mg, 0.44 mmol, 1 equiv) in DMF (3 mL), was added 4-cyano-3- fluorobenzoic acid (73 mg, 0.44 mmol, 1 equiv), Eΐ-,N (0.06 mL, 0.48 mmol, 1.1 equiv), HOBt (67 mg, 0.44 mmol, 1 equiv), DMAP (3 mg, 0.02 mmol, 0.05 equiv) and EDC.HCl (92 mg, 0.48 mmol, 1.1 equiv). The reaction mixture was allowed to stir for overnight at RT. Progress of the reaction was monitored by TLC and LCMS. After completion of the reaction, the reaction mixture was diluted with water (10 mL) and extracted with ethyl acetate (30 mL c 2). Organic layer was washed with water (50 mi), brine solution (50 mL). Organic layer was dried over anhydrous sodium sulphate and concentrated under reduced pressure to obtain crude compound, which was purified by normal phase combi flash to obtain (A)-4-cyano-N-(2-(2-cyano-4, 4- difluoropyrrolidin-1 -y])-2-oxoethyl)-3-fluorobenzamide (Free base) (45 mg, 30%) as a white solid compound.

[0268] LCMS 337 [M+H] +

[0269] 'HNMR (400 MHz, DMSi. /6) d 9.18 (q, J= 8.4, 5.8 Hz, 1H), 8.10 (t, J= 7.3 Hz,

I f !) 7.91 (dd, J 18.9. 9.1 FIz, 2H), 5.10 (dd. J= 9 3, 2.8 Hz, i l l }. 4 31 (ddt, J= 16 3, 1 1.9, 5.9 Hz, 1H), 4.25 - 4.00 (m,3H), 2.99 - 2.74 (m, 2H)

Example 16

Synthesis of (S)-N-(2-(2-cyano-4, 4-difluoropyrrolidin-l-yl)-2-oxoethyl)-l-methyl-6-oxo-l, 6- dihydropyridine-3-carboxamide

Compound 16

[0270] To a stirred solution of (S)~4,4-difluoro-l-glycylpyrrolidine~2~carbonitrile

hydrochloride (100 mg, 0.44 mmol, 1 equiv) in DMF (3 mL), was added l -methyl-6-oxo-l,6- dihydropyridine-3-carboxylic acid (68 mg, 0.44 mmol, 1 equiv), Et 3 N (0.06 mL, 0.48 mmol, 1.1 equiv), HOBt (67 mg, 0.44 mmol, 1 equiv), DMAP (3 mg, 0.02 mmol, 0.05 equiv) and

EDC.HCI (92 mg, 0.48 mmol, 1.1 equiv). The reaction mixture was allowed to stir for overnight at RT. Progress of the reaction was monitored by TLC and LCMS. After completion of the reaction, the reaction mixture was diluted with water (10 ml.) and extracted with ethyl acetate (30 mL x 2). Organic layer was washed with water (50 mL), brine solution (50 mL). Organic layer was dried over anhydrous sodium sulphate and concentrated under reduced pressure to obtain crude compound, which was purified by normal phase combi flash to obtain (5)~N~(2~(2~ cyano-4,4~difiuoropyrrohdin~l~yl)-2-oxoethyl)~l~methy!~6~oxo ~l,6~dihydropyridine~3~ carboxamide (Free base) (30 mg, 21%) as a white solid compound.

[0271] LCMS 325 [M+H] +

[0272] 1 HNMR (400 MHz, DMSO-t/6) 6 8.33-8.66 (d, J = 2.6 Hz, 21 H. 7.98 (dd, J = 9.4, 2.7 Hz, 11 1 ). 6.68(d, J= 9.4 Hz, 1 1 1). 5.14 (dd, ./ 8.4, 4.4 Hz, i l l ). 4.26 (dd, J 12.1, 7.1 1 1/.11 1).

4.22 (s, 2H), 4.20 - 4.06 (m, 11 1). 3.65 (s, 3H), 3.06 -2.82 (m, 2H).

Example 17

Synthesis of (S)-4-cyano-N-(2-(2-cyano-4,4-difluoropyrroiidm-l-yl)-2-oxoe thyi)benzamide

Compound 17

[0273] To a stirred solution of (S)-4,4-difluoro-l -glycylpyrrolidine-2-carbonitrile

hydrochloride (100 mg, 0.44 mmol, 1 equiv) in DMF (3 mi), was added 4-cyanobenzoic acid (68 mg, 0.44 mmol, 1 equiv), Et 3 N (0.06 mi, 0.48 mmol, 1.1 equiv), HOBt (67 mg, 0.44 mmol, 1 equiv), DMAP (3 mg, 0.02 mmol, 0.05 equiv) and EDC.HC1 (92 mg, 0.48 mmol, 1.1 equiv). The reaction mixture was allowed to stir for overnight at RT. Progress of the reaction was monitored by TLC and LCMS. After completion of the reaction, the reaction mixture was diluted with water (10 mi) and extracted with ethyl acetate (30 mi c 2). Organic layer was washed with water (50 mL), brine solution (50 mL). Organic layer was dried over anhydrous sodium sulphate and concentrated under reduced pressure to obtain crude compound, which was purified by normal phase combi flash to obtain (,S’)-4-cyano-N-(2-(2-eyano-4,4- diftuoropy rrolidin- 1 -yl)-2-oxoethy l)benzamide (Free base) (35 g, 25%) as a white solid compound.

[0274] LCMS 319 [M+H] +

[0275] ] HNMR (400 MHz, DMSO-^6) d 9.10 (q, J= 7 5, 5.8 Hz, 11 1). 8 01 (q, J= 8.0 Hz, 5H), 5.10 (dd, 7=9.4, 2.8 Hz, 1H), 4.30 (ddd, J = 16.1, 11.8, 4.8 Hz, 11 1). 4.14 (qt , ./= 17.2, 8.4 Hz, 31 1 i. 17 9, 14 3, 9.9 Hz, 2H).

Example 18

Synthesis of (S)-4-cyano-N-(2-(2-cyano-4,4-difluoropyrrolidin-l-yl)-2-oxo ethyl)-2-

(trifluoromethyl)benzamide

[0276] To a stirred solution of 3-phenylisonicotinic acid (0.100 g, 0 46 mmol, 1.0 equiv) in DMF (10 mL), was added (<S)-4,4-difluoro-l-glycylpyrrolidine-2-carbonitrile hydrochloride (0.104 g, 0.46 mmol, 1.0 equiv), HOBt (0.068 g, 0.50 mmol, 1.1 equiv) and EDC.HCi (0.96 g, 0.50 mmol, 1.1 equiv). The mixture was allowed to stir at RT for 10 min. Triethyl amine (0.19 mL) was added and the mixture was allowed to stir at RT for overnight. Product formation was confirmed by LCMS and TLC. The reaction mixture was diluted with water and extracted with ethyl acetate (50 mL. c 2). Combined organic extracts were washed with water (20 mL. c 4), dried over anhydrous Na 2 S0 4 and concentrated. The crude product obtained was purified by reversed phase HPLC to obtain (5)-4-cy ano-N-(2-(2-cy ano-4,4-difluoropyrrolidin- 1 -yl)-2~ oxoethyl)-2-(trifluoromethyJ)benzamide (0.030 g, 17 % Yield) as an off-white solid.

[0277] LCMS 387.2 j M ! ! | +

[0278] h-J NMR (400 MHz, DMSO-d6) d 9 05 (hr. s., 1H), 8.40 (s, 1H), 8.28 (d, I = 7.45 Hz, 1H), 7.78 (d, J = 7.89 Hz, 1H), 5.12 (d, J = 7.02 Hz, 1H), 4.27 (t, I = 15.13 Hz, 1 ! 1 ) 4.01 - 4.21 (m, 3H), 2.72 - 2.99 (m, 3H).

Example 19

Synthesis of ( ' S)-N-(2-(2-cyano-4,4-difluoropyrrolidin-l-yl)-2-oxoeth yl)-4-phenylquinoline-6 - carboxamide

[0279] Compound 19a. To a solution of quinoline-6-carboxylic acid (1.00 g, 5.7 mmol,

1 equiv) in MeOH (10 mL) was added SOCl 2 (2.06 mL, 17.30 mmol, 3.0 equiv) at 0 °C. The reaction mixture was heated at 50 °C for overnight. After completion of reaction (TLC) the mixture was basified with saturated NaHCO- and extracted with DCM (100 mL x 2). Combined organic layer was dried over anhydrous Na 2 S0 4 and concentrated under reduced pressure to obtain methyl quinoline-6-carboxylate (1.00 g, 98 % Yield) as a brown solid.

[0280] LCMS 188.2 [M+H]+

[0281] Compound 19b. To a solution of methyl quinoline-6-carboxylate (1.00 g, 5.3 mmol, 1 equiv) in DCM (30 mL) was added mCPBA (1.84 g, 10.6 mmol, 2equiv) and the mixture was allowed to stir at RT for overnight. After completion of reaction (TLC) the mixture was diluted with saturated NaHCCL (40 ml.) and extracted with DCM (100 mL x 2). The combined organic layer was dried over anhydrous Na 2 S0 4 and concentrated under reduced pressure to obtain a residue, which was crystallized in ethyl acetate to obtain 6-(rnethoxycarbonyi)quinoline 1-oxide (1.00 g, 92 % Yield)

[0282] LCMS 204.2 [M+H]+

[0283] Compound 19c. The 6-(methoxycarbonyl)quinoiine 1-oxide (0.900g, 4.4mmol lequiv) was taken in 50 ml. RB under nitrogen atmosphere, to this was added POCl 3 (5 mL) and then resulting mixture was sti rred for 2 h under nitrogen atmosphere. After completion of reaction (TLC) the mixture was concentrated under reduced pressure. The resulting residue was dissolved m DCM (100 mL) and washed with saturated NaHCCh (25 mL c 3). Organic layer was separated and dried over anhydrous Na 2 S0 4 and concentrated under reduced pressure to obtain crude material which was purified by flash chromatography (0-20 % Ethyl acetate in Hexane as an eluent) to obtain methyl 4-chloroquinoline-6-carboxylate (0.300 g, 30 % Yield).

[0284] LCMS 222.1 [M+HJ+

[0285] Compound 19d. To the solution of methyl 4-chloroquinohne-6-carhoxylate (300 mg, 1.30 mmol, 1.0 equiv) in THF (5 mL), was added phenylboronic acid (198 mg, 1.60 mmol, 1.2 equiv), Na 2 C0 3 (287 mg, 2.70 mmol, 2.0 equiv) and a catalytieal amount of Pd(PPh 3 ) 2 Cl 2 (47 mg, 0.069 mmol, 0.05 equiv). The resulting reaction mixture was heated at 100 °C for overnight. Product formation was confirmed by LCMS. After completion of reaction the reaction mixture was diluted with water (30 mL) and extracted with ethyl acetate (50 mL * 2). Combined organic layer was dried over anhydrous Na 2 S0 4 and concentrated under reduced pressure to obtain crude material which was purified by flash chromatography (0-20 % Ethyl acetate in Hexane as an eluent) to obtain methyl 4-phenylquino!ine-6-carboxyiate (0.300 g, 84 % Yield).

[0286] LCMS 264. 1 j M 1 11

[0287] 'l l NMR (400 MHz, DMSO-de) 6 9.08 (d, J = 4.38 Hz, i l l ). 8.53 (s, i l l ). 8.08 - 8.35 (m, 2H), 7.48 - 7.69 (m, 5H), 3.86 (s, 3H).

[Q288] Compound 19e. To a stirred solution of ethyl 3-phenylisonicotinate (0.360 g, 1.30 mmol, 1.0 equiv) in THF (5 mL) and water (5 mL), was added LiOH (0.098 g, 4.10 mmol, 3.0 equiv). The mixture was allowed to stir at RT for overnight. Product formation was confirmed by LCMS and T-I NMR The reaction mixture was concentrated to obtain 4-phenylquinoline-6- carboxyiic acid (0.420 g, 100 % Yield) as an off-white solid.

[0289] LCMS 249.9 [M+H]+ [0290] ! H NMR (400 MHz, DY!SO-d,.) d 8 88 id. J = 4.38 Hz, ! ! ! ). 8 37 is. H i ). 8 27 (d, J = 8.77 Hz, 1H), 7.95 (d, ./= 8.77 Hz, 1H), 7.49 - 7.65 (m, 4H), 7.39 (d, J= 4.38 Hz, 1H).

[0291] Compound 19. To a stirred solution of 4-phenylqumolme~0-earboxylie acid (0.200 g, 0.80 mmol, 1.0 equiv) in DMF (5 mL), was added (5)-4,4-difluoro-l-glycylpyrrolidine-2- carbonitrile hydrochloride (0 180 g, 0.80 mmol, 1.0 equiv), HOBt (0.118 g, 0 88 mmol, 1.1 equiv) and EDC.HC1 (0.168 g, 0.88 mmol, 1.2 equiv). The mixture was allowed to stir at RT for 10 min. Triethyl amine (0.4 mL) was added and the mixture was allowed to stir at RT for overnight. Product formation was confirmed by LCMS and TLC, After completion of reaction, the mixture was diluted with water (50 mL) and extracted with ethyl acetate (50 mL x 2).

Combined organic extracts were washed with water (20 mL c 4), dried over anhydrous Na 2 S0 4 arid concentrated. The crude product obtained wns purified reversed phase HPLC to obtain (S)- N-(2-(2-cyano-4,4-difluoropyrroli din-1 -yl)-2-oxoethyl)-4-phenylquinoline-6-carboxamide (0.030 g, 9 % Yield) as an off-white solid.

[0292] LCMS 421.2 [M+H]+

[0293] 'l l NMR (400 MHz, DMSO-de) 6 9.04 (d, J = 4.38 Hz, 2H), 8.43 (s, i l l ). 8.12 - 8.28 (m, 2H ). 7.61 (s, 31 0. 7.55 (d, ./ 4.38 Hz, 2H), 5.09 (d, J 8.33 Hz, 11 1). 4.29 (br s., 11 1). 4.04 - 4 23 (m, 2H), 2.90 (br s., 2H), 2.81 (d, ./= 17.54 Hz, 21 1 }

Example 20

Synthesis of (2R3R)-N-(2-((S)-2-cyano-4, 4-difluoropyrrolidin-l-yl)-2-oxoethyl)-l-ethyl-6-oxo-

2-phenylpiperidine-3-carboxamide

Compound 20

[0294] To a stirred solution of (2R,3R)-l-ethyi-6-Qxo-2-phenylpiperidine-3-carboxylic acid (0 200 g, 0.809 mmol, 1 0 equiv) in DMF (5 mL), was added (5 -4,4-difluoro- 1 - giyeylpyrrolidine-2-carbomtriie hydrochloride (0.200 g, 0.890 mmol, 1.0 equiv), HOBt (0.163 g, 1.21 mmol, l .Sequiv) and EDC.HC1 (0.230 g, 1.21 mmol, 1.5 equiv). The mixture was allowed to stir at RT for 10 min. Triethy] amine (0.4 mL) was added and the mixture was allowed to stir at RT for overnight. Product formation was confirmed by LCMS and TLC. The reaction mixture was diluted with water and extracted with ethyl acetate (50 mL X 2). Combined organic layer was washed with water (20 mL c 4), dried over anhydrous Na 2 S0 4 and concentrated. The crude product obtained was purified by reversed phase HPLC to obtain (2i?,3i?)-N-(2-((»S)-2-cyano- 4, 4~difluoiOpyrrolidin-l~yi)-2-oxoethyl)-l~ethyi-6-oxo-2-pheny lpiperidine-3 -carboxamide (0.040 g, 12 % Yield) as an off-white solid.

[0295] LCMS 419.3 j M I S j +

[0296] h-J NMR (400 MHz, DMSO-de) d 8 26 (d, J = 4.82 Hz, 1H), 7 17 - 7.43 (m, 4H), 5.07

(t, J= 9 65 Hz, 1H), 4.88 (d, J 5 26 Hz. 4. 17 (t, J = 12.06 Hz, i l l ). 3.95 - 4.11 (m, 2H)

3.77 - 3.90 (m, 11 1 ). 3.58 - 3.74 (m, 1H), 2.87 (dd, J = 5.26, 9.65 Hz, 1H), 2.72 - 2.83 (m, 2H), 2.27 - 2.46 (m, 3H), 1.84 (d, J 6 58 Hz, 2H), 0.92 (t, ./= 6.36 Hz, 31 1 ).

Example 21

Synthesis of (S)-N-(2-(2-cyano-4,4-difluoropyrrolidin-l-yl)-2-oxoethyl)-4 -oxo-4, 6, 7,8,9.10- hexahydropyrido [ 1 , 2-a Jazepine-l -carboxamidt

Compound 21

[0297] To a stirred solution of 4-oxo-4,6,7,8,9,10-hexahydropyrido[l,2-a]azepine-l- carboxylic acid (0.050 g, 0.241 mmol, 1.0 equiv) in DMF (2 mL), was added (5)-4,4-difluoro-l- glycylpyrrolidine-2-carbonitrile hydrochloride (0.060 g, 0.265 mmol, 1.1 equiv), DMAP (0.002 g, 0.0120 mmol, 0.05 equiv), HOBt (0.050 g, 0 362 mmol, 1 5equiv) and EDC.HCI (0.070 g, 0.362 mmol, 1.5 equiv). The mixture was allowed to stir at RT for 10 min, Et 3 N (0.1 mL) was added and the mixture was allowed to stir at RT for overnight. Product formation was confirmed by LCMS and TLC. The reaction mixture was diluted with water and extracted with ethyl acetate (25 mL c 2) Combined organic extracts were washed with water (10 mL x 4), dried over anhydrous Na 2 S0 4 and concentrated. The crude product obtained was purified by reversed phase HPLC to obtain (5)-N-(2-(2-c 'ano-4,4-difluoropyrrolidin-l-yl)-2-oxoethyl)-4-oxo-4,6,7,8, 9,l0- hexahydropyrido[! ,2-a]azepine-l-carboxamide (0.015 g, 17 % Yield) as an off-white solid.

[0298] LCMS 379.3 [M+H] +

[0299] l H NMR (400 MHz, DMSO-de) d 8.51 (d, J = 5 70 Hz, 1H), 7.37 (d, ./= 9 21 Hz, 1H), 6.30 (d. ./ 9.65 Hz, 11 1). 5.10 (d, J = 7.02 Hz, 1H), 4.34 (br. s., 2H), 4.17 - 4.31 (m, 2H), 3.96 - 4.15 (ra, 3H), 3 09 (br. s., 2H), 2.72 - 2.92 (rn, 2H), 1.69 (br. s„ 3H), 1.59 (br. s , 2H).

Example 22

Synthesis of (S)-N-(2-(2-cyano-4.4-difluoropyrrolidin-l-yl)-2-oxoethyl)-l -oxo-lH-isochromene-

4-carhoxamide

[0300] To a stirred solution of l-oxo-lH-isochromene-4-carboxyJic acid (0.200 g, 1.04 mmol, 1.0 equiv) in DMF (5 mL), was added (5')-4,4-difluoro-l-glycylpyrrolidine-2-carbonitrile hydrochloride (0.235 g, 1 04 mmol, 1.0 equiv), HOBt (0.168 g, 1.24 mmol, 1.2 equiv) and EDC.HCI (0.235 g, 1.24 mmol, 1.2 equiv). The mixture was allowed to stir at RT for 10 min. Triethyl amine (0.5 mL) was added and the mixture was allowed to stir at RT for overnight. Product formation was confirmed by LCMS and TLC The reaction mixture was diluted with water and extracted with ethyl acetate (50 mL c 2). Combined organic extracts were washed with water (20 mL c 4), dried over anhydrous Na 2 S0 4 and concentrated. The crude product obtained was purified by reversed phase HPLC to obtain (5)-N-(2-(2-cyano-4,4- difluoropyrrohdin~l~yl)-2-oxoethy!)-l -oxo-lH-isochromene-4-carboxamide (0.030 g, 15 % Yield) as an off-white solid.

[0301] LCMS 362.2 [M+H] f

[0302] 'l l NMR 400 MHz, DMSO-d6) 6 8 90 (hr. s . H I). 8.22 (d, I = 7.02 Hz, 1H), 8 05 (d, J 7.89 Hz, 1 1 1). 7.93 (s, 2H), 7 57 - 7 74 (m, 1H), 5.14 (d, j = 6.58 Hz, 1H), 4.01 - 4.22 (m, 2H), 2.83 (d, J = 17.98 Hz, 2H), 1.87 (s, 2H).

Example 23

dihydroisoquinoline-4-carhoxamide

[0303] Compound 23a. To a stirred solution of l-((tert-butoxycarbonyl)amino)cyclopropane- 1 -carboxylic acid (201 mg, 1.00 mmol, 1 equiv) in DMF (2 mL), was added HATU (760 mg, 2.00 mmol, 2.0 equiv) followed by the addition of (5)-4,4-difluoropyrrolidine-2-carbonitrile hydrochloride (305 mg, 1.00 mmol, 1.0 equiv). The reaction mixture was allowed to stir at RT for 10 min. DIPEA (0 86 mL, 5.00 mmol, 5.0 equiv) was added and the reaction mixture was allowed to stir for overnight at RT. Progress of th e reaction was monitored by NMR. After completion of the reaction, the reaction mixture was diluted with water (25 mL) and extracted with ethyl acetate (50 mL. c 2). Combined organic layer was washed with water (25 mL. c 4), dried over anhydrous sodium sulphate and concentrated under reduced pressure. Crude product obtained was enriched by flash chromatography (0-50 % Ethyl acetate in hexane as an eluent) to obtain tert-butyl (S)-(l-(2-cyano-4,4-difluoropyrrolidine-l-carbonyl)cycloprop yl)carbamate (300 mg, 95 % Yield) as an off-white solid.

[0304] i .C.YiS 315.2 [M+H] + [Q3Q5] Compound 23b To a stirred solution of tert-butyl (S)-(l -(2-cyano-4,4- diiluoropyrro!idine~l~carbonyl)cyc!opropyl)carbamate (468 mg, 1.48 mmol, 1 equiv) in MeCN (5 mL), was added pTsOH (383 mg, 2.22 mmol, 1.5 equiv). The reaction mixture was allowed to stir for overnight at RT. Progress of the reaction was monitored by NMR. After completion of the reaction, solvent was removed under reduced pressure to obtain C.V)~ ! -( ! - aminocyclopropane-l-carbon}T)-4,4-difluoropyrrolidine-2-carb onitnle 4- methylbenzenesulfonate (898 mg, qant.) as a white solid compound.

[0306] h-J NMR (400 MHz, DMSO-de) d 8 66 (br. s , IH), 7.42 - 7.54 (m, 2H), 7.09 - 7 20 (m, J= 7.89 Hz, 2H), 5 10 - 5.28 (m, 1H), 4.14 - 4.31 (m. 1 1 1). 3 87 - 4.11 (m, 11 0. 2.74 - 3.03 (m, 1H), 2.29 (s, 3H), 1.42 - 1.67 (m, 1H), 1.31 - 1.41 (m, 1H).

[0307] Compound 23. To a stirred solution of l-oxo-l,2-dihydroisoquinoline-4-carboxyhc acid (0.398 g, 2 10 mmol, 1.0 equiv) in DMF (5 mL), was added (5)-l-(l-aminocyclopropane-l - carbonyl)-4,4-difluoropyrrolidine-2-carbonitrile 4-methylbenzenesulfonate (0.816 g, 2.10 mmol,

I.0 equiv), HOBt (0.311 g, 2.31 mmol, 1.1 equiv) and EDC.HC1 (0.441 g, 2.31 mmol, 1.1 equiv). The mixture was allowed to stir at RT for 10 mm. Triethyl amine (0.87 mL) was added and the mixture was allowed to stir at RT for overnight. Product formation was confirmed by LCMS and TLC. The reaction mixture was diluted with water (30 mL) and extracted with ethyl acetate (50 mL * 2). Combined organic layer was washed with water (20 mL * 4), dried over anhydrous Na 2 S04 and concentrated. The crude product obtained was crystallized in MeOH to obtain (S)- N-(l-(2-cyano-4,4-difluoropyrrolidine-l-carbonyi)cyclopropyl )-l-oxo~l ,2-dihydroisoquinoline- 4-carboxamide (0.140 g, 17 % Yield) as an off-white solid.

[0308] LCMS 387.2

[0309] 'l l NMR (400 MHz, DMSO-d,.} 6 11.79 (br. s., 1H), 10.72 (br. s., 11 1 ). 8.24 (d, J 7.89 Hz, 1H), 7.76 (t, J = 7.67 Hz, IH), 7.54 (t, .7 = 7.45 Hz, IH), 5.40 (br. s., 1H), 4.22 (d, / =

I I.40 Hz, I I I ). 3.68 (d, J= 11.84 Hz, I I I ). 2.87 (br. s., IH), 2.78 id. ./ 14.03 Hz, IH), 1.65 (br. s , I I I). 1.40 (br s., IH), 1.15 - 1.23 (m, I H ). 1.12 (br. s , IH).

Example 24

Synthesis of (S)-N-(2~(2~cyano~4,4-difiuoropyrrolidin~l~yl)~2~oxoethyl)-3 - cyclopropylisonicotinamide

[0310] Compound 24a. To a stirred solution of ethyl 3-bromoisonicotinate (0.5 g, 2.32 mmol, 1.0 equiv) in toluene (30 L) was added cyclopropylboronic acid (0.398 g, 4.63 mmol, 2,0 equiv), K 2 C(¾ (0.958 g, 6.95 mmol, 3.0 equiv) and resulting reaction mixture purged with N 2 gas for 10 min, followed by the addition of Pd(PPh 3 )4 (0.133 g, 0.116 mmol, 0.05 equiv). The resulting reaction mixture was heated at 100 °C for overnight. Product formation was confirmed by LCMS. After the completion of reaction, the mixture was filtered through celite® bed, washed with ethyl acetate (100 mL). Filtrate w¾s concentrated under reduced pressure. The crude product obtained was purified by flash chromatography (0-15 % ethyl acetate in hexane as an eluent) to obtain methyl 3-cyclopropylisonicotinate (0 360 g, 87.8 % Yield) as a yellow liquid.

[0311] LCMS 178.2[M+H] +

[0312] l H NMR (400 MHz, DMSO-de) d 8.51 (d, ,/= 5 26 Hz, 1H), 8.37 (s, 1H), 7.56 (d, ./ = 4.82 Hz, 1 1 1 ). 3.89 (s, 3H), 2.35 id. ./= 14.03 Hz, 1H), 0.96 - 1.04 (m, 2H), 0.80 - 0 87 Cm. 2H).

[0313] Compound 24b. To a stirred solution of methyl 3-cyclopropylisonicotinate (0.420 g, 2.37 mmol, 1.0 equiv) in THF (10 mL) and water (5 mL), was added LiOH (0.170 g, 7.11 mmol, and 3 0 equiv). The mixture was allowed to stir at RT for overnight. Product formation was confirmed by LCMS and 1H NMR. The reaction mixture was concentrated under reduced pressure and diluted with water (15 mL) and washed with ethyl acetate (15 mL). Aqueous layer was separated and freeze dried on lyophilyzer to obtain 3-cyclopropylisonicotinic acid (0.380 g, 100 % Yield) as an off-white solid.

[0314] 1.( MS 164.1 | \ 1 · ! 11

[0315] ! H NMR (400 MHz, DMSO-de) d 8.54 (d, J = 4.82 Hz, 1H), 8.38 (s, 1H), 7.66 (d, J = 5.26 Hz, i l l ). 2.42 - 2.46 (m, i H ) 0.99 - 1 07 (m, 2H), 0.78 - 0.94 (m, 2H).

[0316] Compound 24. To a stirred solution of 3-cyclopropylisonicotinic acid (0.200 g, 1.22 mmol, 1.0 equiv) in DMF (5 mL), was added (S)-4,4-difluoro-l-glycylpyrroIidine-2-carbonitrile hydrochloride (0.33 g, 1.46 mmol, 1.2 equiv) and TBTU (0.587 g, 1.83 mmol, 1.5 equiv) and the mixture was continued to stir at RT for 10 min. Y-Metbyitnorphoime (0.4 mL) was added and the mixture was allowed to stir at RT for overnight. Product formation was confirmed by LCMS and TLC. After completion of reaction, the mixture was diluted with water (50 ml.) and extracted with ethyl acetate (50 mL x 2). Combined organic extracts were washed with water (20 mL x 4), dried over anhydrous Na 2 S0 4 and concentrated. The crude product obtained was purified by flash chromatography (0-50 % ethyl acetate in hexane as an eluent) followed by reverse phase purification to obtain (M-A-(2~(2~cyano~4,4-difluoropyrrolidin-l -yl)-2-oxoethyl)-3- cyc!opropylisonicotinamide (0.065 g, 16 % Yield) as an white solid.

[0317] LCMS 335.2[M+H] +

[0318] ! H NMR (400 MHz, DMSO-d 6 ) d 8.85 (br. s., 1H), 8.44 (d, J = 4.82 Hz, 1H), 8.24 (s, 1H), 7.26 (d, J= 4.82 Hz, 1H), 5.12 (d, ./= 8.33 Hz, 1H), 4.27 (d, J = 11.84 Hz, 1H), 4.05 - 4.17 (m, 2H), 2.90 (br. s., i l l). 2.81 (d, J= 17.10 Hz, 2H), 2.22 (d, J= 5.26 Hz, i l l). 0.97 id. ./ 8.33

Hz, 2H), 0.74 - 0.90 (m, 2H).

Example 25

Synthesis ofN-(l-((S)-2-cyano-4,4-difluoropyrrolidin-l-yl)-l-oxopropan -2-yl)-l-oxo-ll,2- dihydroisoquinoline-4-carboxamide

[0319] Compound 25a. To a stirred solution of (tert-butoxycarbonyl)alanme (140 mg, 0.73 mmol, 1 equiv) in DMF (3 mL), was added HATU (555 mg, 1.46 mmol, 2.0 equiv) followed by the addition of (S)-4,4-difluoropyrrolidine-2-carbonitrile hydrochloride (225 mg, 0.73 mmol, 1.0 equiv). The reaction mixture was allowed to stir at RT for 10 min. DIPEA (0.63 mL, 3.65 mmol, 5.0 equiv) was added and the reaction mixture was allowed to stir for overnight at RT. Progress of the reaction was monitored by 1 H NMR. After completion of the reaction, the reaction mixture was diluted with water (25 mL) and extracted with ethyl acetate (50 mL c 2). Combined organic layer was washed with water (25 mL c 4), dried over anhydrous sodium sulphate and concentrated under reduced pressure. Crude product obtained was enriched by flash

chromatography (0-50 % Ethyl acetate in hexane as an eluent) to obtain tert-butyl (l-((S)-2- cyano-4,4-difluoropyrrolidin-!-yl)~l~oxopropan~2~yl)carbamat e (280 mg, quant.) as an off-white solid.

[0320] LCMS 304.2.2 [M+H] '

[0321] Compound 25b. To a stirred solution of tert-butyl (l-((S)-2-cyano-4,4- chfluoropyrrolidin~l-yl)-l-oxopropan-2-yl)carbamate (500 mg, 1.65 mmol, 1 equiv) in ACN (5 mL), was added PTSA (425 mg, 2.40 mmol, 1.5 equiv). The reaction mixture was allowed to stir the mixture for overnight at RT. Progress of the reaction was monitored by NMR. After completion of the reaction, solvent was removed under reduced pressure to obtain (25)-l-alanyl- 4,4-difluoropyrroIidine-2-carbonitrile 4-methylbenzenesulfonate (630 mg, quant.) as a white solid compound.

[0322] h-J NMR (400 MHz, DMSO-de) d 822 (br. s, 2H), 7.44 - 7.52 (m, 2H), 706 -717 (m, ./ 7.89 Hz, 2H), 5.12 id../ 9.21 Hz, 111}.4.21 (br. s., 111).3.61 (d../ 4.38 Hz, III).3.07 -

326 (m, 111).2.77 - 2.96 (ra, ill).229 (s, 311).1.20 - 126 (m, 311).

[0323] Compound 25. To a stirred solution of i-oxo~l,2-dihydroisoquinoline-4-carboxy!ic acid (0.100 g, 0.53 mmol, 1.0 equiv) in DMF (5 mL), was added (25)-l-alanyl-4,4- difiuoropyrrolidine-2-carbonitnle 4-methylbenzenesulfonate (0.200 g, 0.53 mmol, 1.0 equiv), HOBt (0078 g, 058 mmol, 11 equiv) and EDC.HC1 (0.111 g, 0.58 mmol, 1.1 equiv). The mixture was allowed to stir at RT for 10 min. Triethyl amine (0.22 mL) was added and the mixture was allowed to stir at RT for overnight. Product formation was confirmed by LCMS and TLC. The reaction mixture was diluted with water and extracted with ethyl acetate (50 mL c 2). Combined organic extracts were washed with water (20 mL x 4), dried over anhydrous Na 2 S0 4 and concentrated. The crude product obtained was crystallized in pure MeOH to obtain N-(l- ((S)-2-cyano-4,4-difluoropyrrolidin-l-yl)-l-oxopropan-2-yl)- l-oxo-l,2-dihydroisoquinoline-4- carboxamide (0.120 g, 60 % Yield) as an off-white solid.

[0324] LCMS 375.3 [M+H] +

[0325] 'll NMR (400 MHz, DMSO-de) d 11.64 (br. s., Ml).8.77 (dd, J = 6.80, 18.20 Hz, ill). 8.22 (d, J= 789 Hz, 211) 773 (br. s„ ill).745 - 7.65 (m, 211).4.98 - 516 (m, ill).462 (d, J = 5.70 Hz, 1H), 4.31 (d, ./= 17.54 Hz, 2H), 2.92 (br s., 1H), 2.85 (br. s., ill).1.26 - 1.44 (m, 3H).

Example 26

Synthesis of (S)-N-(2-(2-cyano-4, 4-difluoropyrrolidin-l-yl)-2-oxoethyl)-6-fluoro-l-oxo-l, 2- dihydroisoquinoline-4-carboxamide

O

Compound 26

[0326] To a stirred solution of 6-fluoro-l-oxo-l,2-dihydroisoquinoime-4Carboxylic acid (0.80 g, 0386 mmol, 1.0 equiv) in DMF (12 mL), was added (S)~4,4-difluoro~l~glycylpyrrolidine-2- carbonitri!e hydrochloride (0.173 g, 0.772 mmol, 2.0 equiv) and TBTU (0.136 g, 0.425 mmol,

1.1 equiv). The mixture was allowed to stir at RT for 10 min. A ' -methy Imorpholine (0.3 mL) was added and the mixture was allowed to stir at RT for overnight. Product formation was confirmed by LCMS and TLC. After completion of reaction, the mixture was diluted with water (15 mL) and extracted with ethyl acetate (15 mL x 2). Combined organic layer was washed with water (20 mL x 4), dried over anhydrous NaS04 and concentrated. The crude product obtained was purified by flash chromatography (0-5 % methanol in DCM as an eluent) to obtain (S)-N-(2-(2- cy ano-4,4~difiuoropyrroiidin- 1 -yl)-2-oxoethyi)~6-fiuoro- 1 -oxo~ 1 ,2-dihy droisoquinoline-4- carboxamide (0.015 g, 10.3 % Yield) as an off white solid.

[0327] LCMS 379.2 [M+H] +

[0328] 1HNMR (400 MHz, DMSO-de) 611.74 (br. s., 111).8.72 (br. s., III).8.29 (br. s., ill:·. 8.07 (d, J 1316 Hz, ill).7.66 (br. s., ill).741 id. J= 7.89 Hz, 1H), 513 (d, J= 8.33 Hz, 1H), 4.28 (d, ./= 1622 Hz, ill).4.05 - 4.21 (m, 311).282 (d, ./ 10.09 Hz, 2H).

Example 27

Synthesis ofN-[2-[(2S)-2-cyano-4,4-difluoro-pyrrolidin-l-ylj-2-oxo-eth yl]-3-methoxy-pyridine

4-carboxamide

Compound 38

[Q329] To a stirred solution 3-methoxypyridine-4-carboxylic acid (0.100 g, 0.65 mmol, 1.0 equiv) in DMF (3 mL), was added (2S)-l-(2-aminoacetyl)-4,4-difluoro-pyrrolidine-2- carbonitrile hydrochloride (0 146 g, 0.65 mmol, 1.0 equiv), HOBt (0.105 g, 0.78 mmol, 1.2equiv) and EDC.HCI (0.149 g, 0.78 mmol, 1.2 equiv). The mixture was allowed to stir at RT for 10 min. Triethyl amine (0.3 mL) was added and the mixture was allowed to stir at RT for overnight. Product formation was confirmed by LCMS and TLC. The reaction mixture was diluted with water and extracted with ethyl acetate (40 mL X 2) Combined organic layer was washed with water (15 mL X 4), dried over anhydrous Na 2 S0 4 and concentrated. The crude product was washed with hexane and crystallized in diethyl ether to obtain N-[2-[(2S)-2-cyano- 4,4-dif]uoro-pyrrolidin-I-yl]-2-oxo-ethyl]-3-methoxy-pyridin e-4-carboxamide (0.060 g, 28 % Yield) as an off-white solid.

[0330] LCMS 325.2 [M+H] +

[0331] ! H NMR (DMSO-d ft ,400MHz): d = 8.66 - 8.72 (m, 1 H), 8.59 (s, 1 H), 8.35 (d, 7=4.8

Hz, 1 H), 7.66 (d, J= 4.8 Hz, 1 H), 5.12 (dd, .7=9 0, 2.4 Hz, 1 H), 4.15 - 4.34 (m, 2 H), 4.11 (d, 7=11.8 Hz, 2 H), 3.99 (s, 3 H), 2.89 (d, ./ 3. 1 Hz, 1 H), 2.82 ppm (d, .7=8.8 Hz, 1 H).

Example 28

Synthesis of { ' S)-N-(2-(2-cyano-4,4-difluoropyrrolidin-l-yl)-2-oxoeth yl)-3

(trifluoromeihyl)isonicotinamide

Compound 38

[0332] To a stirred solution of 3-(trifluoromethyl)isonicotinic acid (0.100 g, 0.523 mmol, 1.0 equiv) in DMF (7 mL), was added (S)-4,4-difluoro-l-glycylpyrrolidine-2-carbonitrile 4- methylbenzenesulfonate (0.226 g, 0.627 mmol, 1.2 equiv) and TBTU (0.201 g, 0.627 mmol, 1.2 equiv). The mixture was allowed to stir at RT for 10 min. iV-Methylmorpholine (0.2 mL) was added and the mixture was allowed to stir at RT for overnight. Product formation was confirmed by LCMS and TLC After completion of reaction, the mixture was diluted with water (25 mL) and extracted with ethyl acetate (25 mL x 2). Combined organic layer vras washed with water (20 mL x 4), dried over anhydrous Na 2 S0 4 and concentrated. The crude product obtained was crystalized with 20% DCM in hexane to afford (S)-N-(2-(2-cyano-4,4-difluoropyrrolidin-l -yl)- 2-oxoethyl)-3-(trifluoromethyl)isonicotinamide (0.070 g, 37 % Yield) as an off white solid.

[0333] LCMS 363.2 [M+H] +

[0334] 1H NMR (400 MHz, DMSQ-d 6 ) d 9.09 (br. s., 1H), 9.04 (s, 1H), 8.98 (d, J = 4.82 Hz, 1H), 7.61 (d, ./= 4.82 Hz, 1H), 5.13 (d, J= 8.33 Hz, 1 1 1). 4 26 (d, J= 15.79 Hz, 1 1 1). 4 18 (br. s., 1H), 4.11 (d. ./ 11.84 Hz, 2H), 2.82 (d, /= 15.35 Hz, 1H), 2.73 (s, 1H).

Example 29

Synthesis of (S)-N~(2~(2~cyano~4,4~difluoropyrrolidin~l~yl)~2~oxoethyl)-3 -vinylisonicotin mide

[Q335] Compound 40a. To a solution of 3-bromoisonicotinic acid (0.300 g, 1.49 mmol, 1.0 equiv) in Dioxane : water (1 : 1) (12 ml.) was added compound 4,4,5,5-tetramethyl-2-vinyl-l ,3,2- dioxaborolane (0.34 g, 2.21 mmol, 2.0 equiv), K ·( () ; (0.31 g, 2.23 mmol, 1.5 equiv) and resulting reaction mixture purged with N 2 gas for 10 minute, followed by the addition of Pd(PPh 3 )4 (0.086 g, 0.074 mmol. 0.05 equiv). The resulting reaction mixture was heated at 100° C for overnight. Product formation was confirmed by LCMS. The reaction mixture was concentrated and diluted with water (15 mi.) and washed with ethyl acetate (10 ml, c 2).

Aqueous layer was separated and freeze dried to obtain 3-vinylisonicotinic acid (Quant. Yield) as a white solid.

[0336] LCMS 150.2 [ M i l l

[0337] 1H NMR (400 MHz, DY!SO-d,.) d 8 65 (s, i l l ). 8 29 (d, J= 4.82 Hz, 1H), 7 26 - 7.39 (m, i l l). 7.23 id. ./ 4.38 Hz. 1H), 5.74 Id. ./ 1 7.98 Hz. I I I). 5. 19 id. ./ 10.96 Hz, 1H).

[0338] Compound 40. To a stirred solution of compound 3-vinylisonicotinic acid (0.200 g, 1.342 mmol, 1.0 equiv) in DMF (15 mL), was added (S)-4,4-difluoro-l-glycylpyrrolidine-2- carbonitri!e hydrochloride (0.453 g, 2 013 mmol, 1.5 equiv) and TBTU (0.646 g, 2.013 mmol, 1.5 equiv). The mixture was allowed to stir at RT for 10 min. /v-Methyimorphoime (0.5 mL) was added and the mixture was allowed to stir at RT for overnight. Product formation was confirmed by LCMS and TLC. After completion of reaction, the mixture was diluted with water (50 mL) and extracted with ethyl acetate (50 mL c 2). Combined organic extracts wore washed with water (20 mL c 6), dried over anhydrous Na 2 S0 4 and concentrated. The crude product obtained w¾s purified by flash chromatography (5 % MeOH in DCM as an eluent) followed by reverse phase purification to obtain (S)-N-(2-(2-cyano-4,4-difluoropyrrolidin-l-yl)-2-oxoethyl)-3 - vinyhsonieotinamide (0.012 g, 3 % Yield) as a white solid.

[0339] LCMS 320.2 j VI · 1 1 f [0340] ! H NMR (400 MHz, DMSO-de) d 8 85 - 9 00 (m, 1H), 8.56 (d, J= 4 82 Hz, 1H), 7.35 (d, J= 4.82Hz, 11 1 ). 7.02 (dd, J = 11.40, 17.54 Hz, I I I ). 6.01 (d, ./= 17.54 Hz, I H i. 5.43 (d, ./ = 10.96 Hz, 1H), 5.14 (d. ./ 8.33 Hz, 1H), 4.28 (br. s., 1H), 3.99 - 4.21 (m, 3H), 2.91 (br. s., 1H),

2 82 Id. ./ 17.54 Hz, 1H), 2 67 (br. s , 11 1).

Example 30

Synthesis ofN-[2-[(2S)-2-cyano-4,4-difluoro-pyrrolidin-l-yl]-2-oxo-eth yl]-3-(l- piperidyl)pyridine-4 -carboxamide

Compound 42

[0341] To a stirred solution 3-(l-piperidyl)pytidine-4-carboxy]ic acid (0.100 g, 0.49 mmol, 1.0 equiv) in DMF (3 mL), was added (2S)-]-(2-aminoacetyl)-4,4-difluoro-pyrrolidine-2- carbonitrile hydrochloride (0.1 10 g, 0.49 mmol, 1.0 equiv), HOBt (0.078 g, 0.58 mmol, 1.2equiv) and EDC.HCI (0 1 10 g, 0.58 mmol, 1.2 equiv). The mixture was allowed to stir at RT for 10 min. Triethyl amine (0.21 mL) was added and the mixture was allowed to stir at RT for overnight. Product formation was confirmed by LCMS and TLC. The reaction mixture was diluted with water and extracted with ethyl acetate (40 mL X 2). Combined organic layer was washed with water (15 mL X 4), dried over anhydrous Na 2 S0 4 and concentrated. The crude product was purified by column chromatography (2% MeOH in DCM) to obtain N-[2~[(2S)-2~ cyano-4,4-difluoro-pyrrolidin-l-yl]-2-oxo-ethyl]-3-(l-piperi dyl)pyridine-4-carboxamide (0.050 g, 27.32 % Yield) as an off-white solid.

[0342] LCMS 378 3 I VM ! | +

[0343] 1H NMR (DMSO-d e, ,400MHz) d 9.78 (br. s„ 1 H), 8.58 (br s., 1 H), 8.40 (br. s , 1 H), 7.64 (d, J 4.8 Hz, 1 H), 5.14 (d, J 9 2 Hz, 1 H), 4 17 - 4 37 (m, 2 H), 4 01 - 4 17 (m, 1 H), 2 94 - 3 07 (m, 3 H), 2.90 (br. s., 1 H), 2.82 (d, .7=9 6 Hz, 1 H), 1.72 (br s., 3 H), 1.53 ppm (br. s., 2 H).

Example 31 Synthesis of (S)-N-(2-(2-cyano-4, 4-difluoropyrrolidin-l-yl)-2-oxoethyl)-3-(3, 5- difluorophenyi)isonicotinamide

Compound 47

[0344] Compound 47a. To a solution of ethyl 3-bromoisonicotinate (0.25 g, 1.16 mmol, 1.0 equiv) in toluene (15 mL) was added (3,5-difluorophenyl)boronic acid (0.366 g, 2 315 mmol, 2.0 equiv), K2CO3 (0.48 g, 3.472 mmol, 3.0 equiv) and resulting reaction mixture purged with N gas for 10 minute followed by the addition of Pd(PPh 3 )4 (0.69 g, 0.058 mmol. 0.05 equiv). The resulting reaction mixture was heated at 100 °C for overnight. Product formation was confirmed by LCMS After the completion of reaction, the mixture was filtered through celite bed, washed with ethyl acetate (100 mL). Filtrate was concentrated under reduced pressure. The crude product obtained was purified by flash chromatography (0-10% ethyl acetate in hexane as an eluent) to obtain methyl 3-(3,5-difluoropheny])isonicotinate (0.34 g, quant.) as an off white solid.

[0345] LCMS 250.2] VI P ]

[0346] ! H NMR (400 MHz, DY!SO-d,.) 6 8 80 id.. J= 4.82 Hz, 1H), 8.72 (s, 1H), 8.40 (s, 1H), 7 77 id. ./ 5.26 1 1/. 1H), 7.27 - 7.45 (m, 1H), 7.16 (d, J= 6.58 Hz, 1H), 3.71 (s, 3H).

[0347] Compound 47b. To a stirred solution of methyl 3-(3,5-difluorophenyl)isonicotinate (0.34 g, 1.364 mmol, 1.0 equiv) in THF (10 mL) and w¾ter (5 mL), was added LiOH (0.98 g, 4.08 mmol, 3 0 equiv). The mixture was allowed to stir at RT for overnight. Product formation was confirmed by LCMS and ! H NMR Spectroscopy The reaction mixture was concentrated and diluted with water (15 mL) and washed with ethyl acetate (15 mL). Aqueous layer w ? as separated and freeze dried on lyophilyzer to obtain 3-(3,5-difluorophenyl)isonicotinic acid (0.340 g, quant as an off-white solid).

[0348] LCMS 236.2 [M+H] +

[0349] ! H NMR (400 MHz, DVISO-d,.) d 8 45 (s, 1H), 8 41 (d, J= 4.82 Hz, 1H), 7.32 (d, J = 7 02 Hz, 2H), 7.05 - 7 22 (m, 2H).

[Q350] Compound 47. To a stirred solution of 3-(3,5-difluorophenyl)isonicotinic acid (0.200 g, 0.845 mmol, 1.0 equiv) in DMF (10 mL), was added (S)-4,4-difluoro-l-glycylpyrrolidine-2- carbonitrile hydrochloride (0.38 g, 1.46 mmol, 2.0 equiv) and TBTU (0.587 g, 1.83 mmol, 1.5 equiv). The mixture was allowed to stir at RT for 10 min. /V-metbyImorpboIine (0.4 mL) was added and the mixture was allowed to stir at RT for overnight. Product formation was confirmed by LCMS and TLC. After completion of reaction, the mixture was diluted with water (50 mL) and extracted with ethyl acetate (50 ml. c 2). Combined organic layer was washed with water (20 mL x 4), dried over anhydrous Na 2 S0 4 and concentrated. The crude product obtained was purified by flash chromatography (0-50 % ethyl acetate m hexane as an eluent) followed by reversed phase purification to obtain (S)-N-(2-(2-cyano-4,4-difluoropyrrolidin-l -yl)-2- oxoethyl)-3-(3,5-difiuoropheny!)isomcotinamide (0.020 g, 5.8 % Yield) as an white solid

[0351] LCMS 407.3 [M+H] +

[0352] ' l l NMR ( DV!SO-d.. ,400MHz) d 9.01 (br. s., 1 H), 8.65 - 8.74 (m, 1 H), 7.49 (d, ,/ 4 8 Hz, 1 H), 7 22 - 7 33 (m, 1 H), 5 08 (d, 7=7.0 Hz, 1 H), 4.15 - 4.33 (m, 1 H), 3.92 - 4.15 (m, 3 H), 2.89 (br. s., 1 H), 2.79 ppm (d, 7=13.2 Hz, 1 H).

Example 32

Synthesis of (S)-N-(2-(2-cyano-4, 4-difluoropyrrolidin-l-yl)-2-oxoethyl)-3-(4- fluorophenyljisonicohnamide

[0353] Compound 52a. To a solution of ethyl 3-bromoisonicotinate (0.2 g, 0.93 mmol, 1.0 equiv) in Toluene (15 ml.) was added (4-fluorophenyl)boronic acid (0.21 g, 1.85 mmol, 2.0 equiv), K 2 C(¾ (0.38 g, 2.78 mmol, 3.0 equiv). The resulting reaction mixture purged with N 2 gas for 10 minute, followed by the addition of Pd(PPh 3 )4 (0.054 g, 0.046 mmol. 0.05 equiv). The reaction mixture was heated at 100° C for overnight. Product formation was confirmed by LCMS. After the completion of reaction, the mixture was filtered through celite bed, washed with ethyl acetate (100 mL). Filtrate was concentrated under reduced pressure. The crude product obtained was purified by flash chromatography (0-20 % ethyl acetate in hexane as an eluent) to obtain methyl 3-(4-fluorophenyl)isonicotinate (0 185 g, 83.3%) as an off white solid

[0354] LCMS 232.2 1 VM ! |

[0355] ! H NMR (400 MHz, DMSO-d 6 ) d 8.74 (d, J= 5.26 Hz, 1H), 8.70 (s, 1H), 7.70 (d, J = 4.82 Hz, 11 1 }. 7.42 i dd. ./ 5.70, 8.33 Hz, 2H), 7.31 <i. ./ 8.77 Hz, 21 1 }. 3.68 (s, 3H).

[0356] Compound 52b. To a stirred solution of methyl 3-(4-fluorophenyl)isonicotinate (0.37 g, 1.601 mmol, 1.0 equiv) in THF (20 mL) and water (10 mL), was added LiOH (0.192 g, 8.01 mmol, 5.0 equiv). The mixture was allowed to stir at RT for overnight. Product formation was confirmed by LCMS and 3 H NMR Spectroscopy. The reaction mixture was concentrated and diluted with water (20 mL), washed with ethyl acetate (15 mL c 2) Aqueous layer was separated and freeze dried to obtain 3-(4-fluorophenyl)isonicotinic acid (Quant. Yield) as a white solid

[0357] LCMS 218.2 [M+H] +

[0358] 'l l NMR (400 MHz, DMSO-d 6 ) d 8.38 (s, I I I ). 8.36 (d, J= 4.82 Hz, ! ! ! ). 7.61 (dd, ,/ 5 92, 8.11 Hz, 21 1 ). 7 18 (t, J 8 77 Hz, 21 ! ). 7.13 (d, J 4.82 Hz, i l l }

[0359] Compound 52. To a stirred solution of 3-(4~fiuorophenyl)isonicotinic acid (0.200 g, 0.92 mmol, 1.0 equiv) in DMF (20 mL), was added (S)-4,4-difluoro-i-glyeylpyrrolidine-2- carbonitnle hydrochloride (0.228 g, 1.014 mmol, 1.1 equiv) and TBTU (0.325 g, 1.014 mmol, 1.5 equiv). The mixture was allowed to stir at RT for 10 min. A r ~Methylmorpholine (0.3 mL) was added and the mixture was allowed to stir at RT for overnight. Product formation was confirmed by LCMS and TLC. After completion of reaction, the mixture was diluted with water (50 mL) and extracted with ethyl acetate (50 mL. c 2). Combined organic extracts were washed with water (20 mL x 4), dried over anhydrous Na 2 S0 4 and concentrated. The crude product obtained was purified by flash chromatography (5% MeOH DCM as an eluent) followed by reverse phase purification to obtain (S)-N-(2-(2-cyano-4,4-difluoropyrrolidin-l-yl)-2-oxoethyl)-3 -(4- fluorophenyl)isonicotinamide (0.005 g, 2 % Yield) as a white solid.

[0360] LCMS 389 3 [M+H] +

[0361] 1H NMR (400 MHz, DMSO-de) d 8 97 (br. s , 1H), 8.65 (br s., 2H), 7.57 (br. s., 2H), 7.45 (br. s., 11 1). 7.23 (br. s., 21 ! }. 5.08 (br. s., i l l ). 4.20 (br. s., 11 1 ). 4.05 (br. s., 2H), 2.87 (br. s., i l l ). 2.81 (br. s , 21 1).

Example 33

Synthesis of (S)-3-(5-chloro-2-fluorophenyl)-N-(2-(2-cyano-4,4-difluoropy rrolidin-l-yl)-2- oxoethyl)isonicotinamide

[0362] Compound 54a. To a solution of ethyl 3-bromoisonicotinate (0.25 g, 1.16 mmol, 1.0 equiv) in Toluene (15 mL) was added (5-chloro-2-fluorophenyl)boronic acid (0.403 g, 2.315 mmol, 2.0 equiv), K 2 C0 3 (0.48 g, 3.472 mmol, 3.0 equiv) and resulting reaction mixture purged with N 2 gas for 10 minute, followed by the addition of Pd(PPh 3 ) 4 (0.69 g, 0.058 mmol. 0.05 equiv). The resulting reaction mixture was heated at 100° C for overnight. Product formation was confirmed by LCMS. After the completion of reaction, the mixture was filtered through celite bed, washed with ethyl acetate (100 mL). Filtrate was concentrated under reduced pressure. The crude product obtained was purified by flash chromatography (0-10 % ethyl acetate in hexane as an eluent) to obtain methyl 3-(5-chloro-2-iiuoropheny!)isomcotinate (0.400 g, quant as an off white solid).

[0363] LCMS 266.2[M+H] +

[0364] 'l l NMR (400 MHz, DMSO-d,.} 6 8.84 (d, ,/ 5.26 Hz, 11 1 )_ 8.73 (s, 11 1 )_ 7.83 id. ./

5.26 Hz, 1H), 7.49 - 7.68 (m, 2H), 7.29 - 7.41 (m, 2H), 3.72 (s, 3H).

[Q365] Compound 54b. To a stirred solution of compound methyl 3-(5-chloro-2- fluorophenyl)isonicotinate (0.4 g, 1.504 mmol, 1.0 equiv) in THF (20 mL) and water (10 rnL), was added LiOH (0.108 g, 4.51 mmol, 3.0 equiv). The mixture was allowed to stir at RT for overnight. Product formation was confirmed by LCMS and 'H NMR Spectroscopy. The reaction mixture was concentrated and diluted with water (15 mL) and washed with ethyl acetate (15 mL). Aqueous layer was separated and freeze dried on lyophilyzer to obtain 3~(5~chloro~2- fiuorophenyl)isonicotimc acid (0.400 quant as a white solid).

[0366] LCMS 252.2 [M+H] H

[0367] 1H NMR (400 MHz, DMSO-de) d 8.46 (d, J = 4.82 Hz, 1H), 8.34 (s, 1H), 7.32 - 7.52 (m, 3H), 7.24 (t, J= 9.21 Hz, i l l).

[0368] Compound 54. To a stirred solution of compound 3-(5-chloro-2- fluorophenyl)isonicotinic acid (0.200 g, 0.796 mmol, 1.0 equiv) in DMF (20 mL), was added (S)-4,4-difluoro-l-glycylpyrrolidine-2-carbonitrile hydrochloride (0.358 g, 1.59 mmol, 2.0 equiv) and TBTU (0.383 g, 1.19 mmol, 1.5 equiv). The mixture was allowed to stir at RT for 10 min. V-Methylmorpholine (0.3 mL) was added and the mixture was allowed to stir at RT for overnight. Product formation was confirmed by LCMS and TLC. After completion of reaction, the mixture was diluted with water (50 mL) and extracted with ethyl acetate (50 mL c 2).

Combined organic layer wes washed with water (20 mL c 4), dried over anhydrous Na 2 S0 4 and concentrated. The crude product obtained was purified by flash chromatography (0-50 % ethyl acetate in hexane as an eluent) followed by reverse phase purification to obtain (S)-3-(5-chloro- 2-fluorophenyl)-N-(2-(2-cy ano-4,4-difluoropyrrolidin- 1 -yl)-2-oxoethyl)isonicotinamide (0.055 g, 16.41 % Yield) as an white solid.

[0369] LCMS 423.2 I YM i |

[0370] 1H NMR 'l l NMR (400 MHz, DY!SO-d,.) d 8 92 (br. s , 11 1). 8.78 (d, ./ 4 82 Hz, 11 1). 8.64 (s, I I I). 7.61 (d, J= 4.82 Hz, I I I). 7.52 id. ./ 7.02 Hz, 21 1 }. 7.31 (s, i l l }. 5.07 (d. ./ 9.21

Hz, 11 1 ). 3.93 - 4.09 (m, 2H), 2.80 (br. s , 21 1).

Example 34 Synthesis qfN-(2-((S)-2-cyano-4,4-difluoropyrrolidin-I-yl)-2-oxoethyl) -4-oxo-3,4 4a,8a- tetrahydrophthalazine-1 -carboxamide

Compound 80

[0371] To a stirred solution of 4~oxo~3,4,4a,8a-tetrahydrophthalazine-l -carboxylic acid (0.200 g, 1.05 mmol, 1.0 equiv) in DMF (5 mL), was added (S)-4,4-difluoro-l-giycylpyrrolidine-2- carbonilrile 4-meihylbenzenesultbnate (0.380 g, 1.05 mmol, 1.0 equiv), HOBt (0.170 g, 1 26 mmol, 1.2 equiv) and EDC.HCI (0.240 g, 1 26 mmol, 1.2 equiv). The mixture was allowed to stir at RT for 10 min. Triethyl amine (0.3 mL) was added and the mixture was allowed to stir at RT for overnight. Product formation was confirmed by LCMS and TLC. The reaction mixture was diluted with water and extracted with ethyl acetate (50 mL X 2). Combined organic layer was washed with water (20 mL X 4), dried over anhydrous Na 2 S0 4 and concentrated. The crude product obtained was purified by reversed phase HPLC to obtain N-(2-((S)-2-cyano-4,4- difluoropyrrolidin-l -y])-2-oxoethyl)-4-oxo-3,4,4a,8a-teirahydrophthalazine-l-car boxamide (0.1 10 g, 29 % Yield) as an off-white solid.

[0372] LCMS 363 [M+H] +

[0373] 'l l NMR (400 MHz, DMSO-de) 513.04 (s, i l l). 8.84 (br. s., I I I). 8.61 (d, J = 7.45 Hz, i l l ). 8.30 (d, ./ 7.89 Hz, i l l ). 7.94 - 7.99 On. i l l ). 7.90 (d, ./ 7.02 Hz, I I I ). 5.14 (d, J ------

6.58 Hz, i l l). 4.30 (br s , I I I). 4.12 - 4.23 (m, 2H), 4 08 (d, ./ 1 1.84 Hz, 1H), 2.91 (br. s , i l l).

2.83 (d, ./ 17.98 Hz, 1H).

Example 33

Synthesis ofN-[ 2-/72S)-2-cycmo-4, 4-difluoro-pyrrolidin- I-yl J-2-oxo-eihyl ]-3-methyl-4-oxo- phthalazine-1 -carboxamide

Compound 81

[Q374] To a stirred solution 3-methyl-4-oxo-phthalazine-l -carboxylic acid (0.100 g, 0.49 mmol, 1.0 equiv) in DMF (3 mL), was added (2S)-l-(2-aminoacetyl)-4,4-difluoro-pyrrolidine-2- carbonitrile hydrochloride (0.1 10 g, 0.49 mmol, 1.0 equiv), HOBt (0.080 g, 0.59 mmol, l.2equiv) and EDC.HCI (0.1 13 g, 0.59 mmol, 1.2 equiv). The mixture was allowed to stir at RT for 10 min. Triethyl amine (0.21 mL) was added and the mixture was allowed to stir at RT for overnight. Product formation was confirmed by LCMS and TLC. The reaction mixture was diluted with water and extracted with ethyl acetate (30 mL X 2). Combined organic layer was washed with water (15 mL X 4), dried over anhydrous Na 2 S0 4 and concentrated. The crude product was washed with hexane and crystallized in diethyl ether to obtain (S)-N-(2-(2-cyano- 4,4-dif]uoropyrrolidin-l -yl)-2-oxoethyl)-3-methyl-4-oxo-3,4-dihydrophthalazine-l-car boxamide (0.040 g, 21 7 % Yield) as an off-white solid.

[0375] LCMS 376.3 [M+H] +

[0376] ! H NMR (DMSO-d ft ,400MHz): d = 8.93 (t, J= 5.7 Hz, 1 H), 8.66 (d, J=7.9 Hz, 1 H), 8.33 (d, ,7=7.9 Hz, 1 H), 7.86 - 8.03 (in, 2 H), 5.14 id. .7=7.0 Hz, 1 H), 4 31 (br. s., 1 H), 4.07 - 4.24 (m, 3 H), 2.92 (br. s., 1 H), 2.83 ppm (d, J=17.1 Hz, 1 H).

Example 36

Synthesis of (S)-N-(4-(2-cyano-4, 4-difluoropyrrolidin-l-yl)-4-oxobutyl)-l-oxo-l, 2- dihydroisoquinolim-4-carboxamide

[0377] Compound 82a. To a stirred solution of 4-((tert-butoxycarbonyl)amino)butanoic acid (200 mg, 0.98 mmol, 1 equiv) in DMF (4 mL), was added HATU (745 mg, 1.96 mmol, 2.0 equiv) followed by the addition of (S)-4,4-difluoropyrrolidine-2-carbonitrile hydrochloride(599 mg, 1.97 mmol, 2.0 equiv). The reaction mixture was allowed to stir at RT for 10 min. DIPEA (0.8 mL, 4.7 mmol, 5.0 equiv) was added and the reaction mixture was allowed to stir for overnight at RT. Progress of the reaction was monitored by LCMS. After completion of the reaction, the reaction mixture was diluted with writer (25 mL) and extracted with ethyl acetate (50 mL x 2). Combined organic layer was washed with water (25 mL x 4), dried over anhydrous sodium sulphate and concentrated under reduced pressure. Crude product obtained w¾s enriched by flash chromatography (0-50 % Ethyl acetate in hexane as an eluent) to obtain tert-butyl (S)- (4-(2-cyano-4,4~difluoropyrrolidin-l-yl)-4-oxobutyl)carbamat e (267 mg, 85 % Yield) as an off- white solid.

[0378] LCMS 318.3 [M+H] +

[0379] Compound 82b. To a stirred solution of tert-butyl (S)-(4-(2-cyano-4,4- difluoropyrrohdin-l -y])-4-oxobutyl)carhamaie _(267 mg, 0.84 mmol, 1 equiv) in ACN (3 mL), was added PTSA (217 mg, 1.26 mmol, 1.5 equiv). The reaction mixture was allowed to stir the mixture for overnight at RT. Progress of the reaction was monitored by NMR. After completion of the reaction, solvent was removed under reduced pressure to obtain (S)-l-(4-aminobutanoyl)- 4,4-dif]uoropyrrolidine-2-carbonitrile 4-methylbenzenesulfonate (425 mg, qant.) as a white solid compound.

[0380] ! H NMR (DMSO-di ,400MHz) d 7.68 (br. s., 2 H), 7.48 id. J=8.3 Hz, 2 Hi.. 7.12 (d,

J= 7.9 Hz, 2 H), 5.04 (d, .7=9 2 Hz, 1 H), 4.07 (d, ,7=15.8 Hz, 1 H), 3 91 - 4.02 (m, 1 H), 2 77 - 2.93 (m, 2 H), 2.38 - 2.44 (m, 1 H), 2.29 (s, 4 H), 1.99 - 2.12 (m, 2 H), 1.69 ·· 1.86 ppm (m, 3 H).

[0381] Compound 82. To a stirred solution of l-oxo-l,2-dihydroisoquinoline-4-earboxyhc acid (206 mg, 1.09 mmol, 1.0 equiv) in DMF (3 mL), was added (S)-l-(4-armnobutanoyl)-4,4- difluoropyrro!idine-2-carbonitri!e 4~methy!benzenesulfonate (425 mg, 1.09 mmol, 1.0 equiv), HOBt (160 mg, 1.19 mmol, 1.1 equiv) and EDC.HC1 (229 mg, 1.19 mmol, 1.1 equiv). The mixture was allowed to stir at RT for 10 min. Triethyl amine (0.4 mL) was added and the mixture was allowed to stir at RT for overnight. Product formation was confirmed by LCMS and TLC. The reaction mixture was diluted with water and extracted with ethyl acetate (50 mL X 2). Combined organic layer was washed with water (20 mL X 4), dried over anhydrous Na 2 S0 4 and concentrated. The crude product obtained was crystallized in pure MeOH to obtain (S)-N-(4-(2- cyano-4,4-difluoropyrrolidin-l-yl)-4-oxobutyl)-l-oxo-l,2-dih ydroisoquinoline-4-carboxamide (17 mg, 4 % Yield ) as an off-white solid.

[0382] LCMS 389.3 [M+H] +

[0383] ! H NMR (DMSO-de ,400MHz) d 11.57 (br. s, 1 H), 8.34 (br. s, 1 H), 8.22 (d, J=1.9 Hz, 2 H), 7.73 (t, J= 7.5 Hz, 1 H), 7.42 - 7.60 (m, 2 H), 5.05 (d, .7=8.3 Hz, 1 H), 3.90 - 4.20 (m, 4 H), 3.22 - 3.29 (m, 2 H), 2.86 (br. s., 1 H), 2,78 (d, ,7=12.3 Hz, 1 H), 2.20 - 2.40 (m, 2 H), 1.69 - 1 86 ppm (m, 2 H).

Example 37

Synthesis of ( ' S)-N-(2-(2-cyano-4,4-difluoropyrrolidin-l-yl)-2-oxoeth yl)-lH-indazole-5 - carboxamide

[0384] To a stirred solution of lH-indazole-5 -carboxylic acid (0.200 g, 1.2 mmol, 1.0 equiv) in DMF (10 mL), was added (S)~4,4~difluoro~l~glycylpyrrolidine-2-carbonitrile PTS A (0.444 g, 1.2 mmol, 1 0 equiv), HOBt (0.198 g, 1.46 mmol, 1.2equiv) and EDC.HCI (0.280 g, 1.46 mmol, 1.2 equiv). The mixture was allowed to stir at RT for 10 min. Triethyl amine (0.4 mL) was added and the mixture was allowed to stir at RT for overnight. Product formation was confirmed by LCMS and TLC. The reaction mixture was diluted with water and extracted with ethyl acetate (50 mL X 2). Combined organic layer was washed with water (20 mL X 4), dried over anhydrous Na 2 S0 4 and concentrated. The crude product obtained was purified by reverse phase HPLC to obtain (S)-N-(2-(2-cyano-4,4-difluoropy rrolidin- 1 -yl)-2-oxoethyl)- lH-indazole-5- carboxamide (0.020 g, 10 % Yield) a white solid.

[0385] LCMS 334 [M+H] +

[0386] ! H NMR (400 MHz, DMSO-d 6 ) d 13.29 (br. s., 1H), 8.79 (hr. s., 1H), 8.38 (s, 1H), 8.23 (s, i l l). 7.88 (d, ./ 8.77 Hz, 11 1). 7.60 (d, J= 8.77 Hz, I I I ). 5.10 (d, ./ 8.77 Hz, I I I). 4.31 (br. s., 1 1 1). 4 04 - 4 21 (m, 3H), 2.90-2.81 (m, 2H).

Example 38

Synthesis of ( S)-N-(2-(2-cyano-4,4-difluoropyrrolidin-l-yl)-2-oxoethyl)fur o[2,3-c]pyridine-2 carboxamide

Compound 84

[0387] To a stirred solution of†uro[2,3-c]pyridine-2-carboxytic acid (0.050 g, 0.30 mmol, 1.0 equiv) in DMF (10 mL), was added (S)-4,4-difluoro-l-glycylpyrrolidine-2-carbonitrile 4- methylbenzenesulfonate (0.110 g, 0.30 mmol, 1.0 equiv), HOBt (0.049 g, 0.36 mmol, 1.2equiv) and EDC.HCI (0.069 g, 0.36 mmol, 1.2 equiv). Hie mixture was allowed to stir at RT for 10 mm. Triethyl armne (0.1 mL) was added and the mixture was allowed to stir at RT for overnight. Product formation was confirmed by LCMS and TLC. The reaction mixture was diluted with water and extracted with ethyl acetate (50 mL c 2). Combined organic extracts were washed with water (20 mL. c 4), dried over anhydrous Na 2 S0 4 and concentrated. The crude product obtained was purified by reverse phase FIPLC to obtain (S)-N-(2-(2-cyano-4,4- difluoropyrrolidin-l-y!)~2-oxoethyl)furo[2,3-c]pyridine-2-ca rboxamide (0.020 g, 19 % Yield) as a white solid.

[0388] LCMS 335 [M+H] +

[0389] 'l l NMR (DMSO-d6 ,400MHz) 6 (br s , 1 H), 9 09 (s, 1 H), 8.49 (d, .1 5.3 Hz, 1 FT), 7 84 id.. 1=5.3 Hz, 1 H), 7.69 (s, 1 H), 5.1 1 (d, 1=9.6 Hz, 1 H), 4.25 - 4.39 (m, 1 H), 4.04 - 4.22 (m, 3 H), 2.72 - 3.00 ppm (m, 3 H).

Example 39

Synthesis of (S)-N-(2-(2-cyano-4, 4~difluoropyrrolidm-J-yl)~2-oxoe(hyl)pyrazolo[ 1, 5~a ]pyridine-

5 -carboxamide

Compound 85

[0390] To a stirred solution of pyrazoloj L5-a]pyridine-5-carboxylic acid (0.100 g, 0.61 mmol, 1.0 equiv) in DMF (2 mL), was added (S)-4,4-difluoro-l-glycylpyrro]idine~2~carbonitri]e 4- methylbenzenesulfonate (0.222 g, 0.61 mmol, 1.0 equiv), HOBt (0.93 g, 0.67 mmol, 1.1 equiv) and EDC.HCI (0.129 g, 0.67 mmol, 1.1 equiv). Hie mixture was allowed to stir at RT for 10 mm. Tri ethyl amine (0.2 mL) was added and the mixture was allowed to stir at RT for overnight. Product formation was confirmed by LCMS and TLC. The reaction mixture was diluted with water and extracted with ethyl acetate (50 mL c 2). Combined organic extracts were washed with water (20 mL. c 4), dried over anhydrous Na 2 S0 4 and concentrated. The crude product obtained was purified by reverse phase HPLC to obtain (S)-N-(2-(2-cyano-4,4- difluoropyrrolidin-l-yl)-2-oxoethyl)pyrazolo[l ,5~a]pyndine-5~carboxamide (0.030 g, 15 % Yield) as an off-white solid.

[0391] LCMS 334.2 j Y1 · 1 11 +

[0392] 'l l NMR (400MHz .DMSO-d,.) d 9.02 (hr. s., 1 1 1 }. 8.78 id. J 7.5 Hz, 1 H), 8.29 (br. s., 1 H), 8.11 (s, 1 H), 7.30 (d, .7= 7.0 Hz, 1 H), 6.85 (br s., 1 H), 5.11 (d, .7= 7 9 Hz, 1 H), 4 42 - 4.23 (m, 1 H), 4.23 - 4.05 (m, 3 H), 3.01 - 2.72 (m, 3 H).

Example 40

Synthesis of (S)~N~(2-(2-cyano-4,4~difluoropyrrolidin-l-yl)-2-oxoethyl)-6 -methoxy~2- phenylnicotinamide

Compound 86

[0393] Compound 86a. To a solution of 2,6-dichloronicotinic acid (2.0 g , 10.41 mmol, 1.0 equiv) in MeOH (50 mL) was added compound potassium tert-hutoxide (4.7 g, 41.66 mmol, 4 0 equiv), and resulting reaction mixture was heated at 70 °C for 3 h. Product formation was confirmed by LCMS and TLC. After the completion of reaction, the reaction mixture was concentrated and diluted with water (50 ml). Aqueous layer extracted with ethyl acetate (30 mL x 3). Combined organic extracts were washed with brine (50 mL), dried over anhydrous Na 2 S0 and concentrated. The crude product obtained was purified by flash chromatography (5 %

MeOH in DCM as an eluent) to obtain 2-chioro-6-methoxynicotinic acid (1.8 g, 96.4%) as white solid

[0394] LCMS 188 j M ! f j

[0395] ! H NMR ! H NMR (400 MHz, DMSO-de) d 13.33 (br s., 1H), 8.06 - 8 24 (m, 1H),

6.92 (d, J= 8.77 Hz, 1H), 3.91 (s, 3H).

[0396] Compound 86b. To a solution of 2-chloro-6-methoxynicotinic acid (0.200 g, 1.07 mmol, 1.0 equiv) in Dioxane (10 mL) was added phenylboronic acid (0.195 g, 1.604 mmol, 1.5 equiv), Na 2 C0 3 (0.23 g, 2.14 mmol, 2.0 equiv) and resulting reaction mixture purged with N 2 gas for 10 minute, followed by the addition of Pd(PPh 3 ) 4 (0.062 g, 0.054 mmol. 0.05 equiv). The resulting reaction mixture was heated at 100° C for overnight. Product formation was confirmed by LCMS and TLC. After the completion of reaction, the mixture was filtered through celite bed, washed with ethyl acetate (100 mL). Filtrate was concentrated under reduced pressure. Tire crude product obtained was purified by flash chromatography (0-2 % MeOH in DCM as an eluent) to obtain 6-methoxy~2-phenylnicotinic acid (0.07 g, 28.6%) as white solid.

[0397] LCMS 230 [M+H] +

[0398] 'l l NMR (400 MHz, DMSO-de) d 12.89 (br. s., 1H), 8.17 (d, J= 6.58 Hz, 3H), 7.67 (d, J 7.45 Hz, 1H), 7.38 - 7.60 (m, 31 ! }. 4.04 (s, 3H).

[0399] Compound 86. To a stirred solution of 6-methoxy-2-phenylnicotinic acid (0.050 g, 0.218 mmol, 1.0 equiv) in DMF (5 mL), was added (S)-4,4-difluoro-l -glycylpyrrolidine-2- carbonitrile hydrochloride (0.073 g, 0.327 mmol, 1.5 equiv) and TBTU (0.104 g, 0.327 mmol,

1.5 equiv). The mixture was allowed to stir at RT for 10 min. iV-Methylmorpholine (0.1 mL) was added and the mixture was allowed to stir at RT for overnight. Product formation was confirmed by LCMS and TLC. After completion of reaction, the mixture was diluted with water (15 mL) and extracted with ethyl acetate (10 mL c 2). Combined organic extracts were washed with water (10 mL c 4), dried over anhydrous Na 2 S0 4 and concentrated. The crude product obtained was purified by flash chromatography (5 % MeOH in DCM as an eluent) followed by reverse phase purification to obtain (S)-N-(2-(2-cyano-4,4-difluoropyrrolidin-l-yl)-2-oxoethyl)-6 - methoxy-2-phenylnicotinamide (0.015 g, 17.2 % Yield) as a white solid.

[0400] LCMS 401 2 I M l 11 +

[0401] 1H NMR (400 MHz, DMSO-de) d 8.76 (br. s., 1H), 8.48 (s, 2H), 7.79 (d, J = 8.33 Hz,

2H), 7.39 (br.s., 2H), 6.86 (d, J= 8.33 Hz, 1H), 5.10 (d, J= 7.45 Hz, 1H), 4.23 (br. s., 1H), 3.98 - 4.12 (m, 2H), 3.94 (s, 31 1). 2.90 (d, J= 8.33 Hz, 1 1 1 ). 2.70 - 2.86 (m, i l l ). 2.67 (br. s., 11 1 ).

Example 41

Synthesis of (S)-N-(2-(2-cyano-4, 4-diftuoropyrrolidin-l-yl)-2-oxoethyl)-3-( l -methyl- IH-indazol-

4-yl) isonicotinamide

[Q4Q2] Compound 87a. To a solution of ethyl 3-bromoisonicotinate (0.2 g, 0.93 mmol, 1.0 equiv) in Toluene (15 mi.) was added (l-methyl-lH-indazol-4-yl)boronic acid (0.325 g, 1.85 mmol, 2.0 equiv), K 2 C0 3 (0.38 g, 2.78 mmol, 3.0 equiv) and resulting reaction mixture purged with N 2 gas for 10 minute, followed by the addition of Pd(PPh 3 ) 4 (0.054 g, 0.046 mmol. 0.05 equiv). The resulting reaction mixture was heated at 100° C for overnight. Product formation was confirmed by LCMS. After the completion of reaction, the mixture was filtered through celite bed, washed with ethyl acetate (100 mL). Filtrate was concentrated under reduced pressure. The crude product obtained was purified by flash chromatography (0-30 % ethyl acetate in hexane as an eluent) to obtain methyl 3~(1 -methyl- iH-indazol-4-yi)isonicotinate (0.150 g, 60.7%) as brown liquid.

[0403] LCMS 268.2 ! \ 1 1 1 ]

[0404] 'l l NMR (400 MHz, DYISO-d,.) 6 8.72 - 8.87 (m, 21 1 K 7.79 (d, J = 5.26 Hz, i l l ). 7.64 - 7 76 (m, 2H), 7.49 (s, ill), 7.07 (d, ./ 7 02 Hz, 1H), 4.10 (s, 3H), 3.53 (s, 3H).

[0405] Compound 87b. To a stirred solution of methyl 3~(l-methyi~lH~indazoS-4- yl)isonicotinate (0.27 g, 1.011 mmol, 1.0 equiv) in THF (10 mL) and wnter (10 mL), was added LiOH (0.072 g, 3.03 mmol, 3.0 equiv). The mixture was allowed to stir at RT for overnight. Product formation was confirmed by LCMS and NMR Spectroscopy. ' The reaction mixture was concentrated and diluted with water (20 ml.) and washed with ethyl acetate (15 ml. x 2). Aqueous layer was separated and freeze dried to obtain 3-(l -methyl- lH-indazol-4- yl)isonicotinic acid (Quant. Yield) as a white solid.

[0406] LCMS 254.2 j M 1 11

[0407] 'l l NMR (400 MHz, DMSO-d 6 ) d 8.34 - 8.52 (ra, 2H), 7 90 (s, i l l ). 7 56 (d. J = 8.77 Hz, 11 1 ). 7 39 (s. 1 1 1 ). 7.16 - 7.31 (m, 2.1 1 ). 4.06 (s, 31 1 )

[Q4Q8] Compound 87. To a stirred solution of 3-(l-methyl-lH-indazol-4-yl)isonicotinic acid (0.100 g, 0.395 mmol, 1.0 equiv) in DMF (5 mL), was added (S)-4,4-difluoro-l- glycylpyrrolidine-2-carbonitrile hydrochloride (0.088 g, 0.395 mmol, 1.0 equiv), EDC.HC1 (0.075 g, 0.395 mmol, 1.0 equiv), HOBt (O.OoOg, 0.395 mmol, 1.0 equiv) and DMAP (0.003 g, 0.019 mmol, 0.05 equiv). The mixture was allowed to stir at RT for 10 min. TEA (0.5 mL) was added and the mixture was allowed to stir at RT for overnight. Product formation was confirmed by LCMS and TLC After completion of reaction, the mixture was diluted with water (50 mL) and extracted with ethyl acetate (50 mL c 2). Combined organic extracts were washed with water (20 mL x 4), dried over anhydrous Na 2 S0 4 and concentrated. The crude product obtained was purified by flash chromatography (5 % MeOH in DCM as an eluent) followed by reverse phase purification to obtain (S)-N-(2-(2-cyano~4,4-difluoropyrrolidin-l-yl)-2~oxoethyl)-3 -(l- methyl-iH-indazoi-4-yl)isonicotinamide (0.010 g, 6% Yield) as a white solid.

[0409] LCMS 425.3 [M+H] +

[0410] 1H NMR (400 MHz, D.YiSO-d,.} d 8.92 (br. s., 1H), 8.62 - 8 80 (m, 2H), 7.86 (s, 1H), 7 64 (d, J= 8.33 Hz, 1H), 7.56 (d, ./= 4.82 Hz, 1H), 7.35 - 7.45 (m, i l l ). 7.18 (d, J= 7.02 Hz, 1H), 5.05 (d, J = 6.58 Hz, 1H), 4.13 (br. s., 1H), 4.06 (s, 3H), 3.83 - 3.99 (m, 3H), 2.75 (d , J = 11.84 Hz, 2H), 2.65 (br. s., 1 1 1 ).

Example 42

Synthesis ofN-[2-[(2S)-2-cyano-4,4-difluoro-pyrrolidin-l-yl]-2-oxo-eth yl]-3-morpholino- pyridine-4-carboxamide

[0411] Compound 88a. In a 25 mL bottle, 3-fluoropyridine-4-carboxylic acid (0.500 g, 3.54 mmol, 1.0 equiv) and morpholine (0.616 g, 7.08 mmol, 2.0 equiv) were added and the reaction mixture w ? as heated at 120 °C for 1 h. Reaction progress was monitored by NMR and LCMS. After completion of reaction, reaction mixture was concentrated under reduced pressure and the crude compound was purified by normal phase flash chromatography (2-10% MeOH in DCM as an eluent) to obtain 3-morpholinopyridine-4-carboxylic acid (100 mg, 13 % Yield) as a yellow solid.

[0412] LCMS 209.3 j Y1 · 1 11 "

[0413] 1H NMR (400MHz ,DMSO-d 6 ) d 14 16 (br. s., 1 H), 8.55 (s, 1 H), 8.34 (d, ./= 4.8 Hz,

1 H), 7.53 (d, ./= 4 8 1 1/ 1 H), 3.82 - 3.63 (m, 4 H), 3.19 - 3.01 (m, 4 H).

[0414] Compound 88. To a stirred solution 3-morpholinopyridine-4-earboxylic acid (0.100 g, 0.48 mmol, 1.0 equiv) in DMF (3 mL), was added (2S)-l-(2-aminoacetyl)-4,4-difluoro- pyrrolidine-2-carbonitrile hydrochloride (0.108 g, 0.48 mmol, 1.0 equiv), HOB† (0.078 g, 0 58 mmol, !.2equiv) and EDC.HCI (0.111 g, 0.58 mmol, 1.2 equiv). The mixture was allowed to stir at RT for 10 min. Triethyl amine (0.21 mL) was added and the reaction mixture was allowed to stir at RT for overnight. Product formation was confirmed by LCMS. The reaction mixture w¾s diluted with water and extracted with ethyl acetate (40 mL c 2). Combined organic extracts were washed with water (15 mL c 4), dried over anhydrous Na 2 S0 4 and concentrated. The crude product obtained was purified by reverse phase HPLC to obtain (S)-N-(2-(2-cyano-4,4- difluoropyrrolidin-l-yl)-2-oxoethy])-3-morpholinoisonicotina mide (0.020 g, 11% Yield) as an off-white solid.

[0415] LCMS 380.5 [M+H] + [0416] ! H NMR (400MHZ, DMSO-d 6 ) d 9.48 (br. 8., i 1 1). 8.55 (s, 1 H), 8.40 (d, J= 4.8 Hz, 1 H), 7.57 (d, J= 4.8 Hz, 1 H), 4.37 - 4.09 (m, 4 H), 3.78 (br s., 4 H), 3.08 (br s, 4 H), 2.90 (br. s., 2 H), 2.08 (s, 1 H).

Example 43

Synthesis of (S)-3-benzyl-N-(2-(2-cyano-4,4-diflmropyrrolidin-l-yl)-2-oxo ethyl)isonicotinamide

[0417] Compound 50a. To a solution of 3-bromoisonicotinic acid (0 5 g, 2.475 mmol, 1.0 equiv) in dioxane (6 mL) and water (12 mL) was added 2-benzyl-4,4,5,5-tetramethyl- 1,3,2- dioxaborolane (1.1 g, 4.95 mmol, 2.0 equiv), K 2 C0 3 (1.1 g, 7.45 mmol, 3.0 equiv) and resulting reaction mixture was purged with N 2 gas for 10 min. Pd(PPh 3 ) 4 (0.143 g, 0.124 mmol. 0.05 equiv) was added and the resulting reaction mixture was heated at 100° C for overnight. Product formation was confirmed by LCMS. After the completion of reaction, the reaction mixture was concentrated and diluted with water (10 mL). Aqueous layer washed with EtOAc (10 mL x 2) and acidify with 6N HC1 (pH ~ 3.0), extracted with EtOAc (10 mL x 3). Combined organic extracts were washed with water (30 mL), dried over anhydrous Na 2 S0 4 and concentrated. The crude product obtained was purified by flash chromatography (0-10 % Methanol in DCM as an eluent) to obtain 3-benzyhsonicotinic acid (0.100 g, 19 %) as an off white solid.

[0418] LCMS 214

[0419] Compound 50. To a stirred solution of 3-benzylisonicotinic acid (0.100 g, 0.467 mmol, 1.0 equiv) in DMF (8 mL), was added (5)-4,4-difluoro-l-glycylpyrrolidine-2-carbonitrile hydrochloride (0.105 g, 0.467 mmol, 1.0 equiv) and TBTU (0.164 g, 0.514 mmol, 1.1 equiv). The mixture ^as allowed to stir at RT for 10 min. iV-methylmorpholine (0.4 mL) was added and the mixture was allowed to stir at RT for overnight. Product formation was confirmed by LCMS and TLC. After completion of reaction, the reaction mixture was diluted with water (20 mL) and extracted with ethyl acetate (20 mL c 2). Combined organic extracts were washed with water (20 mL. x 4), dried over anhydrous Na?SC>4 and concentrated. The crude product obtained was purified by flash chromatography (5 % MeOH in DCM as an eluent) followed by reversed phase purification to obtain (5)-3-benzyl~N-(2~(2-cyano-4,4-difluoropyrrolidin-l-yl)~2- oxoethyl)isonicotinamide (0.025 g, 14 % Yield) as an white solid.

[0420] LCMS 385.2 j M i i j

[0421] 'l l NMR (400MHz, DMSO-d 6 ) 6 8.94 (br. s., 1 1 1 }. 8.61 - 8.42 (m, 2 1 1 }. 7.34 id. ./

4.8 Hz, 1 1 1}. 7.30 - 7 21 (m, 3 1 1 } 7.18 (d, J= 7.0 Hz, 2 H), 5.13 id. ./= 7.0 Hz, 1 H), 4 27 (d, J = 15.3 Hz, 2 H), 4.23 - 4.01 (m, 4 1 1 }. 2.90 (br. s., 1 H), 2.82 (d. ./ 17.5 Hz, 1 H).

Example 44

Synthesis ofN-(2-((S)-2-cyano-4,4-diflmropyrrolidin-l-yl)-2-oxoethyl)- 3-(cyclohex-l-en-l- yljisonicolinamide

Compound 43

[Q422] Compound 43a. To a solution of ethyl 3-bromoisonicotinate (0 4 g, 1.85 mmol, 1.0 equiv) in toluene (20 mi.) was added 2-(cy clohex- 1 -en- 1 -yl)-4,4,5,5-tetramethyl- 1,3,2- dioxaborolane (0.58 g, 2.77 mmol, 1.5 equiv), K 2 C0 3 (0.51 g, 3.78 mmol, 2.0 equiv) and resulting reaction mixture purged with N 2 gas for 10 min. followed by the addition of Pd(PPh 3 ) 4 (0.065 g, 0.093 mmol. 0 05 equiv). The resulting reaction mixture was heated at 100 °C for overnight. Product formation was confirmed by LCMS. After the completion of reaction, the reaction mixture was filtered through celite® bed, washed with ethyl acetate (100 niL). Filtrate was concentrated under reduced pressure. The crude product obtained was purified by flash chromatography (0-10 % ethyl acetate m hexane as an eluent) to obtain methyl 3-(cyclohex-l- en-l-yl)isonicotinate (0.170 g, 42.4%) as colorless oil.

[0423] LCMS 2 ! 8.2| M i i |

[0424] 'l l NMR (4QQMHz .DMSO-d,.) 6 8.60 (d, ,/ 5.3 Hz, 1 H), 8.52 (s, 1 H), 7.56 (d, ,/

5 3 Hz, 1 H), 5.62 (br s., 1 H), 2 24 - 2.06 (m, 4 H), 1 77 - 1.54 (m, 4 H)

[0425] Compound 43b. To a stirred solution of methyl 3-(c clohex-1 -en- l-yl)isonicotinate

(0.17 g, 0.779 mmol, 1.0 equiv) in THF (5 mL) and water (5 mL), was added LiOH (0.037 g,

1.56 mmol, 2.0 equiv). The mixture was allowed to stir at RT for overnight. Product formation was confirmed by LCMS and *H NMR. The reaction mixture was concentrated and diluted with water (20 mL) and washed with ethyl acetate (15 ml. x 2). Aqueous layer was separated and iyophihzed to obtain 3-(cyclohex-l-en-l-yl)isonicotinic acid (Quant. Yield) as a white solid.

[0426] LCMS 204.2] VI · ί ! |

[0427] 'li NMR (400MHz, DYlSO-d,.) d 8.25 (d, .7= 4.8 Hz, 1 H i. 8 17 (s, 1 I I). 7.10 (d. ./

4 8 Hz, 1 H), 5.66 (br s., 1 1 1 ). 2.29 (br. s , 2 H), 2.12 - 2 05 Cm. 2 H), 1.72 - 1 53 Cm. 4 H)

[Q428] Compound 43. To a stirred solution of 3-(cyclohex-l-en-l-yl)isonicotinic acid (0.100 g, 0.493 mmol, 1.0 equiv) in DMF (5 mL), was added (S)-4,4-difluoro-l-glycyJpyrrolidine-2- carbonitrile hydrochloride (0.110 g, 0.493 mmol, 1.0 equiv), TBTU (0.174 g, 0.542 mmol, 1.1 equiv). The mixture was allowed to stir at RT for 10 min. L'-Methylmorpholine (0.4 ml.) was added and the mixture was allowed to stir at RT for overnight. Product formation was confirmed by LCMS and TLC. After completion of reaction, the reaction mixture was diluted with water (20 mL) and extracted with ethyl acetate (20 mi x 2). Combined organic extracts were washed with water (20 mL x 4), dried over anhydrous Na 2 S0 4 and concentrated. The crude product obtained was purified by flash chromatography (5 % MeOH in DCM as an eluent) followed by reverse phase purification to obtain N-(2-((S)-2-cyano-4,4-difluoropyrrolidin-l-yl)-2-oxoethyl)- 3-(cy clohex- 1 -en- 1 -yl)isonicotinamide (0.004 g, 2 4 % Yield) as an white solid.

[0429] LCMS 375.3[M+H] +

[0430] 'l l NMR (500 MHz, DMSO-d6) 6 8.69 (t, J = 5.8 Hz, i l l ). 8.53 (d, I = 4.9 Hz, 11 1 }. 8 46 (s, 11 1 ). 7 33 (d, J = 4.9 Hz, i l l }. 5.79 (d, I = 3 9 Hz, I I !}. 5.10 (dd, J = 9.3, 2.7 Hz, i l l}. 4 33 - 4.23 (m, i l l ). 4.15 -4.03 (m, 3H), 2.95 - 2.74 (m, 2H), 2.25 (s, 2H), 2.12 (d, I = 6.4 Hz, 2H), 1.70 - 1.61 (m, 2H), 1.58 (q, J = 6.0, 5.5 Hz, 2H).

Example 45

Synthesis of ( S)-N-(2-(2-cyano-4,4-difluoropyrrolidin-l-yl)-2-oxoethyl)-3- (l-melhyl-JH-indol-4 - yljisonicotinamide

[0431] Compound 66a. To a solution of ethyl 3-bromoisonicotinate (0.2 g, 0.93 mmol, 1.0 equiv) in mixture of toluene (16 ml,) & water (4 mL) was added 1 -methyl-4-(4, 4,5,5- tetramethyl-l,3,2-dioxaborolan-2-yl)-lH-indole (0.48 g, 1.85 mmol, 2.0 equiv), K 2 C0 3 (0.382 g, 2.78 mmol, 3.0 equiv) and resulting reaction mixture purged with N 2 gas for 10 min. followed by the addition of Pd(PPh 3 ) 2 Cl 2 (0.033 g, 0.046 mmol. 0.05 equiv). The resulting reaction mixture was heated at 100 °C for overnight. Product formation was confirmed by l .C YIS After the completion of reaction, the mixture was filtered through celite bed, washed with ethyl acetate (100 mL). Filtrate was concentrated under reduced pressure. The crude product obtained was purified by flash chromatography (0-20 % ethyl acetate in hexane as an eluent) to obtain methyl 3-(l-methyl-lH-indol-4-yl)isonicotinate (0.180 g, 73.4%) as a yellow oil.

[0432] LCMS 267.2 j M 1 1 j

[0433] 'l l NMR (4QQMHz, DMSO-de) 6 8.74 (s, 1 H), 8.67 id. J = 4.8 Hz, 1 H), 7.67 - 7.49 (m, 3 H), 7 40 (d, ./ 3.1 Hz, 1 H), 7. 12 (d, J= 8.8 Hz, 1 H), 6.49 (d, ./= 3.1 Hz, 1 H), 3 92 (s, 3 H), 3.63 (s, 3 H).

[Q434] Compound 66b. To a stirred solution of methyl- lH-indol-4-yl)isonicotinate (0.18 g, 0.67 mmol, 1.0 equiv) in THF (5 mL) and water (5 mL), was added LiOH.H 2 0 (0.085 g, 2.63 mmol, 3.0 equiv). The mixture was allowed to stir at RT for overnight. Product formation was confirmed by LCMS. The reaction mixture was concentrated and diluted with water (20 mL) and washed with ethyl acetate (15 mL x 2) Aqueous layer was separated and lyophilized to obtain 3- (l-methyi-lH-indol-4-yl)isomcotinic acid (Quant. Yield) as a white solid.

[0435] LCMS 253.2 1 VM 11 f

[0436] Compound 66. To a stirred solution of 3-( l-methyl-lH-indol-4-yl)isonicotinic acid (0.050 g, 0.198 mmol, 1.0 equiv) in DMF (2 mL), was added (S)-4,4~difluoro~l- glyeylpyrrolidine-2-carbonitriIe hydrochloride (0.044 g, 0.198 mmol, 1.0 equiv), EDC1.HC1 (0.057 g, 0.297 mmol, 1.5 equiv), HOBt (0.04 g, 0.297 mmol, 1.5 equiv). The resulting reaction mixture was allowed to stir at RT for 10 min. Et 3 N (0.2 mL) was added and the mixture was allowed to stir at RT for overnight. Product formation was confirmed by LCMS and TLC. After completion of reaction, the mixture was diluted with water (10 mL) and extracted with ethyl acetate (10 mL c 2). Combined organic extracts were washed with water (10 mL c 4), dried over anhydrous Na 2 S0 4 and concentrated. The crude product obtained was purified by flash chromatography (5 % MeOH in DCM as an eluent) to obtain (S)-N-(2-(2-cyano-4,4- difluoropyrrolidin-l-yl)-2-oxoethyl)-3-(l-methyl-lH-indol-4- yl)isonicotinamide (0.013 g,

15.5 % Yield) as a while solid.

[0437] LCMS 424.3[M+H] +

[0438] ! H NMR (500 MHz, DMSO-d6) d 8.91 (t, J = 6.0 Hz, 1H), 8.69 (s, 1H), 8.61 (d, I =

4.9 Hz, i l l }. 8.19 (t, I = 5.8 Hz, i l l ). 7.74 (d, J = 1.8 Hz, IH), 7.49 - 7.40 (m, 2H), 7.40 - 7.29 (m, 2H), 6.47 (d, J = 3.1 Hz, i l l). 5.08 {tid'd. J = 15.6, 9.3, 2 8 Hz, 2H), 4.18 (t, J = 13.4 Hz, 2H),

4.10 - 3.89 (m, 6H), 3.81 (s, 11 1 ). 2.91 - 2.72 (m, il l ). 1.88 (s, 2H), 1 32 - 1.21 (m, 5H), 0.84 (q, 1 = 10.7, 8.5 Hz, IH).

Example 46

Synthesis of (S)-N-(2-(2-cyano-4,4-difluoropyrrolidin-l-yl)-2-oxoethyl)-3 -(indolin-5- yljisonicotinarnide

104c Compound 104

[0439] Compound 104a. To a solution of ethyl 3-bromoisonicotinate (0.2 g, 0.922 mmol, 1.0 equiv) in toluene (8 mL) and water (2 mL) was added tert-butyl 5-(4,4,5,5-tetramethyl-l,3,2- dioxaborolan-2-yl)indoline~l -carboxylate (0.381 g, 1.106 mmol, 1.2 equiv), K 2 CO 3 (0 38 g, 2.764 mmol, 3.0 equiv) and resulting reaction mixture purged with N 2 gas for 10 min, followed by the addition of Pd(PPh 3 ) 4 (0.033 g, 0.046 mmol. 0.05 equiv). The resulting reaction mixture was heated at 100 °C for overnight. Product formation was confirmed by LCMS. After the completion of reaction, the mixture was filtered through celite® bed, washed with ethyl acetate (100 mL). Filtrate was concentrated under reduced pressure. The crude product obtained was purified by flash chromatography (0-20 % ethyl acetate in hexane as an eluent) to obtain compound of tert-butyl 5-(4-(methoxycarbonyl)pyridin-3-yl)indoline-l -carboxylate (0.200 g, 61 34 %) as a yellow oil.

[0440] LCMS 355 4 [M+H] +

[0441] Compound 104b. To a stirred solution of compound tert-butyl 5-(4- (methoxycarbonyl)pyridin-3-yl)indoline-l-carboxylate (0.200 g, 0.565 mmol, 1.0 equiv) in THF (8 mL) and water (8 mL), was added LiOH (0.028 g, 1.29 mmol, 2.0 equiv). The mixture was allowed to stir at RT for overnight. Product formation was confirmed by LCMS. The reaction mixture was concentrated and diluted with water (10 mL) and washed with ethyl acetate (10 mL x 2) Aqueous layer was separated and freeze dried on lyophilyzer to obtain compound 3-(l- (tert-butoxycarbonyl)indolin~5~yl)isonicotinic acid (Quant. Yield) as a white solid.

[0443] Compound 104c. To a stirred solution of compound 3-il-(tert-butoxycarbonyi)indoiin- 5-yl)isonicotinic acid (0.050 g, 0.147 mmol, 1.0 equiv) in DMF (3 mL), was added (S)-4,4- difluoro-i-g!ycylpyrrolidine-2-carbonitrile hydrochloride (0.034 g, 0.147 mmol, 1.0 equiv), EDCI.HCT (0.042 g, 0.225 mmol, 1.5 equiv), HOST (0.03 g, 0.225 mmol, 1.5 equiv). The mixture was allowed to stir at RT for 30 min. Triethylamine (0.2 mL) was added and the mixture was allowed to stir at RT for overnight. Product formation was confirmed by LCMS. After completion of reaction, the mixture was diluted with water (0 lmL) and extracted with ethyl acetate (10 mL * 2). Combined organic extracts w'ere washed with w¾ter (10 mL c 4), dried over anhydrous Na 2 S0 4 and concentrated to obtained tert-buiyl (S)-5-(4-((2-(2-cyano-4,4- difluoropyrrolidin-l~yl)-2-oxoethyl)carbamoyl)pyridin-3-yl)i ndoline-i-carboxylate (0.035 g,

46 % Yield) as an yellow solid.

[0444] LCMS 512.4

[0445] Compound 104. To a stirred solution of tert-butyl (S)-5-(4-((2-(2-cyano-4,4- difluoropyrrolidin-l-yl)~2-oxoethyl)carbamoyl)pyridin-3-yl)i ndoline-l-carboxylate (0.07 g, 0.14 mmol, 1.0 equiv) in DCM (5 mL), w¾s added trifloroacetic acid (0.5 mL). The mixture was allowed to stir at RT overnight. Product formation was confirmed by LCMS. The reaction mixture concentrated and the crude was purified by reverse phase purification to obtain (S)-N- (2-(2-cyano-4,4-difluoropyrrolidin-] -yl)-2-oxoethyl)-3-(indolin-5-yl)isonicotinamide (0.022 g, 40 % Yield) as a white solid.

[0446] LCMS 512.31 M 1 1 ]

[0447] h-J NMR (400MHz ,DMSO-d 6 ) d 8.83 (br s., 1 H), 8.58 (s, 1 H), 8.52 (d, J 4.8 Hz,

1 H), 7.33 (d, ./ 4.8 Hz, 1 H), 7.26 (s, 1 H), 7.08 id../ = 6.6 Hz, 1 H), 6 50 id. J= 7.9 Hz, 1 H), 5.68 (br. s., 1 H), 5.09 (d. ./ 7.5 Hz, 1 H), 4.21 (br. s., 1 H), 4.06 (br. s., 1 H), 4.00 (br. s., 3 H),

3.45 (t, J = 8.6 Hz, 2 H), 2.96 (d, J= 9.2 Hz, 2 H), 2.80 (d, J = 17.5 Hz, 1 H).

Example 47

Synthesis of (S)-3-(6-chloronaphthalen-2-yl)-N-(2-(2-cyano-4, 4-difluoropyrrolidin-l-yl)-2- oxoethyljisonicotinamide

[0448] Compound 108a To a solution of methyl 3-bromoisonicotinate (0 150 g, 0.694 mmol, 1.0 equiv) in Dioxan (4 mL) was added 2-(6-chloronaphthalen-2-yl)-4,4,5,5-tetramethyl-l,3,2- dioxaborolane (0.300 g, 1.04 mmol, 1.5 equiv), K 2 C0 3 (0.192 g, 1.38 mmol, 2.0 equiv) and resulting reaction mixture purged with N 2 gas for 10 minute, followed by the addition of Pd(PPh 3 )Cl 2 (0.024 g, 0.0347 mmol. 0.05 equiv). The resulting reaction mixture was heated at 100° C for overnight. Product formation was confirmed by LCMS. After the completion of reaction, the mixture was filtered through celite bed, washed with ethyl acetate (100 mL).

Filtrate was concentrated under reduced pressure. The crude product obtained was purified by flash chromatography (0-20 % ethyl acetate m hexane as an eluent) to methyl 3-(6- chioronaphthalen-2-yl)isonicotinate (0.200 g, 97.43% yield) as an yellow' solid.

[0449] LCMS 298 1 i M l 11

[0450] Compound 108b. To a stirred solution of methyl 3-(6-chloronaphthalen-2- yi)isonicotmate (0.200 g, 0.673 mmol, 1.0 equiv) in THF (4 mL) and w¾ter (4 mL), w¾s added LiOH.H 2 0 (0.043 g, 1.011 mmol, 1.5 equiv). The mixture was allowed to stir at RT for overnight. Product formation was confirmed by LCMS and l H NMR Spectroscopy. The reaction mixture was concentrated and diluted with water (20 mL) and washed with ethyl acetate (10 mL x 2). Aqueous layer was separated and freeze dried on lyophilyzer to obtain 3-(6- chioronaphthalen-2-yi)isonicotinic acid (Quant. Yield) as an yellow' solid.

[0451] LCMS 284.2 j M i l l

[0452] Compound 108. To a stirred solution of 3-(6-chloronaphthaJen-2-yl)isonicotinic acid

(0.130 g, 0.459 mmol, 1.0 equiv) in DMF (5 mL), w¾s added (S)-4,4-difluoro-l- glycylpyrrolidine-2-carbonitrile hydrochloride (0.103 g, 0.459 mmol, 1.0 equiv), EDCLHC1

(0.133 g, 0 688 mmol, 1.5 equiv) & HOBt (0.094 g, 0.688 mmol, 1.5 equiv). The mixture was allowed to stir at RT for 10 min. TEA (0.3 mL) was added and the mixture was allowed to stir at

RT for overnight. Product formation was confirmed by LCMS and TLC. After completion of reaction, the mixture was diluted with water (20 mL) and extracted with ethyl acetate (20 mL c 2). Combined organic extracts w¾re washed with wilier (20 mL c 4), dried over anhydrous Na 2 S0 4 and concentrated. The crude product obtained was purified by flash chromatography (5 % MeOH m DCM as an eluent) to obtain (S)-3-(6-chloronaphthalen-2-yl)-N-(2-(2-cyano-4,4- difluoropyrrolidin-l -yl)-2-oxoethyl)isonicotinamide (0.100 g, 48 % Yield) as an off-white solid.

[0453] LCMS 455 2] M 1 ! ]

[0454] ‘H NMR (400MHz ,DMSO-d 6 ) d 9.12 (t, .7= 5.9 Hz, 1 H), 8.82 (s, 1 H), 8.72 (d, .7 = 4.8 Hz, 1 H), 8.23 (s, 1 H), 8.12 - 8.00 (m, 2 H), 7.95 (d, J= 8.3 Hz, 1 H), 7.75 (dd, J = 1.8, 8.3 Hz, 1 H), 7.61 - 7.45 (m, 2 H), 5.12 (dd. ./ 2.9, 9.0 Hz, 1 H), 4.27 - 3.97 (m, 3 H), 2.95 - 2.76

(m, 2 H).

Example 48

Synthesis of(S)-N-(2-(2-cyano-4,4-difluoropyrrolidin-l-yl)-2-oxoethyl) -6-hydroxy-2- phenylmcotinamide

o

Compound 30

[Q455] Compound 30a. To a solution of 2,6-dichloronicotinic acid (2.0 g, 10.41 mmol, 1.0 equiv) in MeOH (50 mL) was added compound potassium tert-butoxide (4.7 g, 41.66 mmol, 4.0 equiv), and resulting reaction mixture was heated at 70° C for 3 hour. Product formation was confirmed by LCMS and TLC. After the completion of reaction, the reaction mixture was concentrated up to maximum and diluted with water (50 ml). Aqueous layer extracted with ethyl acetate (30 mL c 3). Filtrate was concentrated under reduced pressure. Combined organic extracts wore washed w th brine (50 mL), dried over anhydrous Na 2 S0 4 and concentrated. The crude product obtained was purified by flash chromatography (5 % MeOH in DCM as an eluent) to obtain 2-chloro-6-methoxynicotinic acid (1.8 g, 96.4%) as a white solid

[0456] 1.( MS 188[M+H] +

[0457] 'l l NMR (400 MHz, DMSO-de) d 13.33 (br. s., ! I I ). 8.06 - 8.24 (m, 11 1 )_ 6.92 id. ./ 8 77 Hz, 1H), 3.91 (s, 3H).

[0458] Compound 30b. To a solution of 2-chloro-6-methoxymcotimc acid (0.200 g, 1.07 mmol, 1.0 equiv) in Dioxane (10 mL) was added compound (0.195 g, 1.604 mmol, 1.5 equiv), Na 2 C0 3 (0.23 g, 2.14 mmol, 2.0 equiv) and resulting reaction mixture purged with N 2 gas for 10 minute, followed by the addition of Pd(PPh 3 )4 (0.062 g, 0.054 mmol. 0.05 equiv). The resulting reaction mixture was heated at 100° C for overnight. Product formation was confirmed by LCMS and TLC. After the completion of reaction, the mixture was filtered through celite bed, washed with ethyl acetate (100 mL). Filtrate was concentrated under reduced pressure. The crude product obtained was purified by flash chromatography (0-2 % MeOH in DCM as an eluent) to obtain 6-methoxy-2-phenylnicotinie acid (0.07 g, 28.6% Yield) as a white solid.

[0459] LCMS 230[M+H] +

[0460] 'l l NMR (400 MHz, DMSO-de) d 12.89 (br. s., I I I ). 8.17 (d, J= 6.58 Hz, 31 1). 7.67 (d, J ----- 7.45 Hz, 1 H ). 7 38 - 7.60 (m, 3H), 4.04 (s, 31 1}

[0461] Compound 30c. To a stirred solution of 6-methoxy~2-pheny!nicotinie acid (0.050 g, 0.218 mmol, 1.0 equiv) in DMF (5 mL), w¾s added (S)-4,4-difluoro-l-glycylpyrrolidine-2- carbonitrile hydrochloride (0.073 g, 0.327 mmol, 1.5 equiv) and TBTU (0.104 g, 0.327 mmol, 1.5 equiv). The mixture was allowed to stir at RT for 10 min. Y-Methylmorpholine (0.1 mL) was added and the mixture was allowed to stir at RT for overnight. Product formation was confirmed by LCMS and TLC. After completion of reaction, the mixture was diluted with water (15 mL) and extracted with ethyl acetate (10 mL x 2). Combined organic extracts were washed with water (10 mL x 4), dried over anhydrous Na 2 SC>4 and concentrated. The crude product obtained was purified by flash chromatography (5 % MeOH in DCM as an eluent) followed by reverse phase purification to obtain (S)-N-(2-(2-cyano-4,4-difiuoropyrroiidin-l-yl)-2-oxoethyi)-6 - methoxy-2 phenylnicotinamide (0.015 g, 17.2 % Yield) as an white solid

[0462] LCMS 401.2[M+H] + [0463] ! H NMR (400 MHz, DMSO-de) d 8 76 (hr. s., 1H), 8.48 (s, 2H), 7.79 (d, ./= 8.33 Hz, 2H), 7.39 (br.s , 2.1 1 ). 6.86 (d, ,/= 8 33 Hz, 1H), 5.10 (d, ./= 7 45 Hz, 1H), 4.23 (hr. s„ 11 1 ). 3 98 - 4.12 (m, 21 1 }. 3.94 (s, 31 1 }. 2.90 (d , J ------ 8.33 Hz, i l l ). 2.70 - 2.86 (m, 1 1 1 ). 2.67 (br. s., i l l }.

[0464] Compound 30. To a stirred solution of (S)-N-(2-(2-cyano-4,4-difluoropyrrolidin-l-yl)- 2-oxoethyl)-6-methoxy-2 phenylnicotinamide (0.035 g , 0.087 mmol, 1.0 equiv) in DCM (5 mL), was added TMS1 (1 ml) dissolved in DCM (5 ml) drop wise. The mixture was allowed to stir at RT for 10 min and then stir at 50°C for 2h. Product formation was confirmed by LCMS and TLC. After completion of reaction, the mixture was concentrated up to dry, residue diluted with Ethyl acetate (15 mL) and washed with water (10 mi c 2), dried over anhydrous Na 2 S0 4 and concentrated. The crude product obtained was purified by reverse phase purification to obtain (S)-N-(2-(2-cyano-4,4-difluoropyrroJidin-l-yl)-2-oxoethyl)-6 -hydroxy-2- phenylnicotinamide (0.003 g, 8.9 % Yield) as an off white solid.

[0465] LCMS 387.21 VM 1 |

[0466] ! H NMR (400MHz ,DMSO-d 6 ) d 10.18 (s, 1 H), 8.37 (d, J= 7.9 Hz, 1 H), 7.82 (br. s., 2 H), 7.54 (br. s., 2 H), 6.81 (d, J ------ 7.0 Hz, 1 H), 5.12 (d, ./ 8.8 Hz, 1 H), 4.37 - 4.18 (m, 2 H),

4.18 - 3.95 (m, 1 H), 2.89 (s, 1 H), 2 81 (d, J = 8.8 Hz, 2 H).

Example 49

Synthesis of (S)-N-(2-(2-cyano-4,4-difluoropyrrolidin-l-yl)-2-oxoethyl)-3 -(o- tolyljisonicotinamide

[0467] Compound 64a. To a solution of ethyl 3-bromoisonicotinate (0.3 g, 1.38 mmol, 1.0 equiv) in Toluene (15 mL) and water (2 mL) was added o-tolylboronic acid (0.288 g, 1.66 mmol, 1.2 equiv), K 2 C0 3 (0.571 g, 4.14 mmol, 3.0 equiv) and resulting reaction mixture purged with N 2 gas for 10 minute, followed by the addition of Pd(PPh 3 ) 4 (0 08 g, 0.007 mmol. 0.05 equiv). The resulting reaction mixture was heated at 90° C for overnight. Product formation was confirmed by LCMS. After the completion of reaction, the mixture was filtered through celite bed, washed with ethyl acetate (100 ml ). Filtrate was concentrated under reduced pressure. The crude product obtained was purified by flash chromatography (0-20 % ethyl acetate in hexane as an eluent) to obtain methyl 3-(o-tolyl)isonicotinate (0.300 g, 95 %) as yellow liquid.

[0468] LCMS 22H 3] M 1 ! j

[0469] 1H NMR (400MHz ,DMSO-d 6 ) d 8 76 (d, J= 5.3 Hz, 1 H), 8.55 (s, 1 H), 7 77 (d, J = 4.8 Hz, 1 H), 7.39 - 7.14 (m, 3 H), 7.08 (d, J = 7.5 Hz, 1 H), 3.33 (s, 3 H), 2.02 (s, 3 H)

[0470] Compound 64b. To a stirred solution of methyl 3-(o-tolyl)isonicotinate (0.4 g, 1.762 mmol, 1.0 equiv) in THF (5 mL) and water (5 mL), was added LiOH.H 2 0 (0.148 g, 3.524 mmol, 2.0 equiv). The mixture was allowed to stir at RT for overnight. Product formation was confirmed by LCMS. The reaction mixture was concentrated and diluted with water (20 mL) and washed with ethyl acetate (10 mL c 2). Aqueous layer was separated and freeze dried on lyophilyzer to obtain 3-(o-to!yl)isonicotinic acid (Quant Yield) as a white solid

[0471] LCMS 2 1 4 2| M ! f j

[0472] Compound 64. To a stirred solution of 3-(o-tolyl)isomcotinic acid (0.100 g, 0.469 mmol, 1.0 equiv) in DMF (4 mL), was added (S)-4,4-difluoro-l-glycylpyrrolidine-2-carbonitrile hydrochloride (0 105 g, 0.469 mmol, 1.0 equiv), EDCI.HC1 (0.135 g, 0.704 mmol, 1.5 equiv) & HOBt (0 096 g, 0.704 mmol, 1.5 equiv). The mixture was allowed to stir at RT for 10 min. Tnethylamine (0.5 mL) was added and the mixture was allowed to stir at RT for overnight. Product formation was confirmed by LCMS and TLC. After completion of reaction, the mixture was diluted with water (20 mL) and extracted with ethyl acetate (20 mL c 2). Combined organic extracts were washed with water (20 mL c 4), dried over anhydrous Na 2 S0 4 and concentrated. The crude product obtained was purified by flash chromatography (3 % MeOH in DCM as an eluent) to obtain (S)-N-(2-(2-cyano-4,4-difluoropyrrolidin-l -yl)-2-oxoethyl)-3-(o- tolyl)isonicotinamide (0.1 g, 55 5 % Yield) as a white solid

Synthesis of ( ' R)-N-(2-(2-cyano-4,4-difluoropyrrolidin-l-yl)-2-oxoeth yl)-3 - cyclopropylisonicotinamide

[0475] Compound 37. To a stirred solution of 3-cyclopropylisonicotinic acid (0 100 g, 060 mmol, 1.0 eq) in DMF (05 mL), was added (R)-4,4-difluoro-l-glycylpyrrolidine-2- carbomtnle.HCl (0.138 g, 0.60 mmol, 1.0 eq), HOBt (0.098 g, 0.72 mmol, 1.2eq) and EDC.HC1 (0.138 g, 0.72 mmol, 1.2 eq). The mixture was allowed to stir at RT for 10 min. Triethyl amine (0.2 ml.) was added and the mixture was allowed to stir at RT for overnight. Product formation was confirmed by LCMS and TLC. The reaction mixture was diluted with water and extracted with ethyl acetate (50 mL c 2). Combined organic extracts were washed with w'ater (20 mL * 4), dried over anhydrous Na 2 S0 4 and concentrated. The crude product obtained was purified by reverse phase HPLC to obtain (R)-N-(2-(2-cyano-4,4-difiuoropyrrolidin~l-yl)-2-oxoethyl)-3 - cyclopropylisonicotinamide (0.005 g, 03 % Yield) a white solid.

[0476] LCMS 335 [M+H] +

[0477] 'l l NMR (4QQMHz ,DMSO-6 8.85 (br. s., 1 1 1}. 8.44 id. J = 4.4 Hz, 1 H i. 8.24 (s, 1 H), 7.26 id. J = 4.8 Hz, 1 1 1 ). 5 12 (d, I = 7.5 Hz, 1 H), 4.27 (d, J = 12.3 Hz, 2 H), 4.21 - 3.99 (m, 2 H), 2.98 - 2.86 (m, 1 H), 2.81 (d, J = 17.1 Hz, 1 H), 2.73 (s, 1 H), 2.21 (br. s., 2 H), 1.24 (br. s.,

2 H).

Example 51

Synthesis of(S)-N-(2-(2-cyano-4,4-difluoropyrrolidm-l-yl)-2-oxoethyl)- 6-oxo-4-phenyl-l, 6- dihydropyridine-3-carhoxamide

[0478] Compound 29a. To a stirred solution of methyl 4-chloro-6-oxo-l,6-dihydropyridine-3- carboxylate (0.200 g, 1.07 mmol, 1 equiv) in Dioxan (4 mL) and water (lmL), was added Na 2 C0 3 (0.227 g, 2.14 mmol, 2 equiv) and phenylboronic acid (0.196 g, 1.61 mmol, 1.5 equiv). Aerated the reaction mixture with nitrogen gas for 15 min. Pd(PPh3) 2 Cl 2 (0.078 g, 0.11 mmol,

0.1 equiv) was added to the reaction mixture. Reaction mixture was again aerated with nitrogen gas for 15 mm. The reaction mixture was allowed to stirrer for 16 h at 100°C. Progress of the reaction was monitored by LCMS. After completion of the reaction, the reaction mixture was diluted with water (15 mL) and extracted with ethyl acetate (25 mL X 2). Combined organic extracts were washed with brine (25 mL), dried over anhydrous Na2SG4 and concentrated under reduced pressure to obtain methyl 6-oxo-4-phenyl-l,6-dihydropyridine-3-carboxylate (0.170 g, 69 % Yield) as an off-white solid

[0479] LCMS 230.3 [M+H] f

[0480] Compound 29b. To a stirred solution of methyl 6-oxo-4-phenyl- 1 ,6-dihy dropyridine-3- earboxyiate (0.170 g, 0.74 mmol, 1 equiv) in EtOH and water (1: 1 )(6 mL), w¾s added NaOH (0.059 g, 1.48 mmol, 2 equiv). The mixture was allowed to stir at 50°C for 16 h. Progress of the reaction was monitored by LCMS. After completion of the reaction, the reaction mixture was diluted 1 M HC1 (10 mL) and extracted with ethyl acetate (20 mL X 3). Combined organic extracts were washed with brine (25 mL), dried over anhydrous Na2SQ4 and concentrated under reduced pressure. Crude product was purified by flash chromatography (0- 10 % MeOH in DCM as an eluent) to obtain 6-oxo-4-phenyl-l,6-dihydropyridine-3-carboxylic acid (0.090 g, 56 % Yield) as an off-white solid.

[0481] LCMS 216.2 ] \ 1 1 11

[0482] Compound 29. To a stirred solution of 6^xo-4-phenyl-L6--dihydrGpyridine--3- carboxylic acid (0.080 g, 0.37 mmol, 1 equiv) in DMF (3 mL), was added (S)-4,4-difluoro-l- glycylpyrrolidine-2-carbonitrile hydrochloride (0.083g, 0.37 mmol, 1.0 equiv), HOBt (0.059 g, 0.44 mmol, l .2equiv) and EDC.HC1 (0.085 g, 0.44 mmol, 1.2 equiv). The mixture was allowed to stir at RT for 10 min. Triethylamine (0.112 g, 1.11 mmol, 3.0 equiv) was added and the mixture was allowed to stir at RT for overnight. Product formation was confirmed by LCMS.

The reaction mixture was diluted with water and extracted with ethyl acetate (40 mL c 2).

Combined organic extracts were washed with water (15 mL x 4), dried over anhydrous Na2S04 and concentrated. The crude product obtained was purified by normal phase combi-flash chromatography to obtain ( S)-N-(2-(2-cy ano-4,4-difluoropyrroli din-1 -yl)-2-oxoethyl)-6-oxo-4- phenyl- i,6-dihydropyridine-3-carboxamide (0.045 g, 31 25 % Yield) as an off-white solid.

[0483] LCMS 387.3 [M+H] +

[0484] ! H NMR (400MHZ ,DMSO-d6) d 12.39 - 11.70 (m, 1H), 8.66 (t, J = 5.9 Hz, 1H), 7.64 (s, 1 1 1). 7.44 - 7.32 (m, 51 U. 6.29 is. 1H), 5.08 idd. ./ 9.3, 2,9 Hz, 11 1 ). 4.20 (ddd, J ------ 16.0,

11 6, 4.4 Hz, i l l ). 4 01 (dd, J = 19.7, 8.6 Hz, 31 1). 2.96 - 2,70 (ra, 21 1 ). 1.38 - 1.21 (m, 31 1 ). 1.16 (s, 1H).

Example 52

Synthesis of (S)-N-(2-(2-cyano-4, 4-difluoropyrrolidin-l-yl)-2-oxoethyl)-3- phenoxyisonicotinarnide

[0485] Compound 97a. To a stirred solution of methyl 3-bromoisonicotinate (1.0 g, 4.65 mmol, 1 equiv) in DMF (10 mL), was added phenol (0.437 g, 4.65 mmol, 1 equiv), Cul (1.762 g, 9.3 mmol, 2 equiv) and CS 2 CO 3 (3.03 g, 10.0 mmol, 2 equiv). Heated the reaction mixture at 110 °C for 18 h. Reaction progress was checked by LCMS. The reaction mixture was diluted with water (20 mL) and added few drops of dil. HC1 till pH was slightly acidic. Aqueous layer was extracted with ethyl acetate (50 mL c 2). Combined organic extracts were washed with brine (50 mL), dried over anhydrous Na 2 S0 4 , filtered and concentrated under reduced pressure to obtain 3-phenoxyisonieotimc acid (0.200 g, 20 % Yield) as an off-white solid. [0486] LCMS : 216.1 [M+H] +

[0487] Compound 97. To a stirred solution of 3-phenoxyisonicotinic acid (0.140 g, 0.65 mmol, 1 equiv) in DMF (3 mL), was added (S)-4,4-difluoro-l-glycylpyrrolidine-2-carbonitrile hydrochloride (0.146 g, 0.65 mmol, 1.0 equiv), HOBt (0.105 g, 0.78 mmol, l.2equiv) and EDC.HCI (0.149 g, 0.78 mmol, 1.2 equiv). The mixture was allowed to stir at RT for 10 min. Triethyl amine (0.197 g, 1.95 mmol, 3 equiv) was added and the mixture was allowed to stir at ambient temperature for 16 h. Product formation was confirmed by LCMS. Tire reaction mixture was diluted with water (l OmL) and extracted with ethyl acetate (40 mL c 2). Combined organic extracts were washed with water (25 mL x 5). Organic extract was dried over anhydrous Na S04, filtered and concentrated under reduced pressure to obtain erode product. The crude product obtained was purified by normal phase combi-flash chromatography to obtain (S)-N-(2- (2-cyano-4,4-difluoropyrrolidin-l-yl)-2-oxoethyl)-3-phenoxyi sonicotinamide (0.050 g, 19.9 % Yield) as an off-white solid.

[0488] LCMS 387.2 [M+H] +

[0489] 'l l NMR (400MHz ,DMSO-d6) 6 8.80 (t, J == 5.2 Hz, 11 1 ). 8.49 (d, J = 4.9 Hz, 1 H),

8.25 (s, 11 1). 7.67(d, ,/ 4.9 Hz, 11 1). 7.43 (t, J = 7.9 Hz, 21 1 ). 7.21 U. ./ 7.5 Hz, i l l ). 7 1 1 (d, J

= 8.0 Hz, 21 1 ). 5 10 (dd, ./ 9 3, 3.0 Hz, 1 1 1 ). 4 29 - 3.98 (m, 11 1 ). 2.93 - 2.73 (m, 2H), 1 23 (d, J = 3.6 Hz, 11 1).

Example 33

Synthesis of N-(2-( (2R)-2-cyono-4, 4-difluorocyclopentyl)-2-oxoethyl)-3- (phenylaminojisonicotinamide

[0490] Compound 102a. To a stirred solution of methyl 3-bromoisonicotinate (1 0 g, 4.65 mmol, 1 equiv) in toluene (6 mL), was added aniline (0.476 g, 5.12 mmol, 1.1 equiv),

Tris(dibenzyiideneaceione)dipal3adium(0) (0.431 g,0.47 mmol, 0.1 equiv), xantphos (0.539 g. 0.93 mmol, 0 2 equiv) and CS 2 CO 3 (2,28 g, 6.98 mmol, 1.5 equiv). The resulting reaction mixture was heated at 190 °C for 1 h in microwave. Reaction progress was checked by LCMS. The reaction mixture was diluted with w¾ter (15 mL) and extracted with ethyl acetate (30 mL c 2) Combined organic extracts were washed with brine (30 mL), dried organic extract over anhydrous Na 2 SC> 4 , filtered and concentrated under reduced pressure. Crude compound was purified by normal phase combi-flash chromatography to obtain methyl-3- (phenylamino)isonicotinate (0.300 g, 28 % Yield ) as yellow' semisohd.

[0491] LCMS: 229.2 [M+H]

[0492] Compound 102b. To a stirred solution of methyl-3-(phenylarnino)isonicotinate (0.300 g, 1.31 mmol, 1 equiv) in T ' H F and water (l: l)(4 mL), was added LiOH.H 2 0 (0.083 g, 1.97 mmol, 1.5 equiv). The mixture was allowed to stir at ambient temperature for 16 h. Product formation was confirmed by LCMS. The reaction mixture was diluted with water (20 mL), extracted with ethyl acetate and concentrated the aqueous layer under reduced pressure and lyophilized to obtain 3-(phenylamino)isonicotmic acid (0.260 g.) as an off-white solid

[0493] LCMS 215.2 [M+H] +

[0494] Compound 102. To a stirred solution of 3-(phenylamino)isonicotinic acid (0 260 g,

1.21 mmol, 1 equiv) in DMF (3 mL), was added (lR)-4,4-dif!uoro-2~gly c lc clopentane- 1 - carbonitrile hydrochloride (0.272 g, 1.21 mmol, 1.0 equiv), HOBt (0.196 g, 1.45 mmol, 1.2equiv) and EDC.HCJ (0.278 g,1.45 mmol, 1.2 equiv). and triethyl amine (0.366 g, 3.63 mmol, 3 equiv) was added and the mixture was allowed to stir at ambient temperature for 16 h. Product formation was confirmed by LCMS. The reaction mixture was diluted with water (15mL) and extracted with ethyl acetate (40 mL c 3). Combined organic extracts were washed with water (25 mL x 5), dried over anhydrous Na 2 S0 4 , filtered and concentrated under reduced pressure to obtain crude product which was purified by reverse phase HPLC N-(2-((2R)-2-cyano-4,4- difluorocyclopentyl)-2-oxoethyl)-3-(phenylamino)isonicotinam ide (0.004 g, 0.8 % Yield) .

[0495] LCMS 386.2 [M+H] +

[0496] 1 H NMR (400MHz ,DMSO-d6) 6 9 22 - 9.07 (m, 21 1). 8.64 (s, 1 1 1 ). 8.12 (d, ./ 5.0

Hz, i l l ). 7 58 (d, J= 5 0 Hz, l i t) 7.34 (t, J 7.7 Hz, 21 1). 7.22 (d, J= 7 8 Hz, 2H), 7.03 (t, ./

7.4 Hz, 1H), 5.13 (dd, J= 9.4, 2.9 Hz, 1H), 4.36 - 4.25 (m, 1H), 4.25 - 4.04 (m, 3H), 3.00 - 2.74 (m, 2H), 2.09 (s, 3H), 1.24 (s, 1H). Example 54

Synthesis of (S)-N-( 2-(2-cyano-4, 4-difiuoropyrrolidin-l-yl)-2-oxoethyl)-l 2 3 6-tetrahydro- [3.4’-bipyridine j -4-carboxamide 2, 2, 2-trifluoroacetate

Compound 105-TFA

[Q497] Compound 105a. To a solution of ethyl 3-bromoisonicotinate (0.2 g, 0.925 mmol, 1.0 equiv) in Toluene (8 ml.) and water (2 mL) was added tert-butyl 4-(4,4,5,5-tetramethyl-l ,3,2- dioxaborolan-2-yl)-3,6-dihydropyridine-l(2H)-carboxylate (0.429 g, 1.39 mmol, 1.5 equiv),

K‘CO; (0.255 g, 1.85 mmol, 2.0 equiv) and resulting reaction mixture purged with N 2 gas for 10 rmnute, followed by the addition of Pd(PPh 2 )Cl 2 (0.033 g, 0.046 mmol. 0.05 equiv). The resulting reaction mixture was heated at 90° C for overnight. Product formation was confirmed by LCMS. After the completion of reaction, the mixture was filtered through celite bed, washed with ethyl acetate (100 mL). Filtrate was concentrated under reduced pressure. The erode product obtained was purified by flash chromatography (0-20 % ethyl acetate in hexane as an eluent) to obtain T-(tert-butyl) 4-methyl 3',6'-dihydro-[3,4'-bipyridine]-r,4(2'H)-dicarboxylate (0.130 g, 44.4 % Yield) as an yellow oil.

[0498] LCMS 3 19.2j VI · 1 11

[0499] 'l l NMR (400MHz .DMSO-d,.} 6 8.65 (d, ./ 4.8 Hz, 1 H), 8.59 - 8.52 (m, 1 H), 7.63 (d, ./= 4.8 Hz, 1 H), 5 68 (br. s., 1 H), 3.96 (br. s., 2 H), 3.87 - 3.69 (m, 3 H), 3.52 (br. s„ 2 H), 2 27 (br. s, 2 H), 1.53 - 1.25 (m, 9 H), 1.07 (s, 3 H).

[0500] Compound 105b.To a stirred solution of T-(tert-buiyl) 4-methyl 3',6'-dihydro-[3,4'- bipyridine|-r,4(2'H)-dicarboxylate (0.13 g, 0.408 mmol, 1.0 equiv) in THF (5 mL) and water (5 mL), was added Li OH (0 020 g, 0.817 mmol, 2.0 equiv). The mixture was allowed to stir at RT for overnight. Product formation was confirmed by LCMS and‘H NMR Spectroscopy. The reaction mixture was concentrated and diluted with water (20 rnL) and washed with ethyl acetate (15 mL x 2). Aqueous layer was separated and freeze dried on lyophilyzer to obtain 1 '-(tert- butoxycarbony])-r,2',3',6'-tetrahydro-[3,4'-bipyridine]-4-ca rboxylic acid (Quantative Yield) as white solid.

[0501] LCMS 305.2[M+H] +

[0502] l H NMR (400MHz, DMSO-d 6 ) d 8.30 (d, J= 4.8 Hz, 2 H), 8.22 (s, 1 H), 7.17 (d, J = 4.8 Hz, 1 H), 3.92 (br. s., 2 H), 2.38 (br. s., 2 H), 2.06 (s, 1 H), 2.04 - 1.92 (m, 1 H), 1.43 (s, 9 H), 1.06 (s, 2 H)

[0503] Compound 105c. To a stirred solution of r-(tert-butoxycarbonyl)-r,2',3',6'- tetrahydro-[3,4'-bipyridine]-4-carboxylic acid (0.100 g, 0.32 mmol, 1.0 equiv) in DMF (5 mL), was added (S)-4,4-dif!uoro-l-glycylpyrrolidine-2-carbonitrile hydrochloride (0.074 g, 0.32 mmol, 1.0 equiv), EDCI.HC1 (0.092 g, 0.48 mmol, 1.5 equiv) & HOBt (0.065 g, 0.48 mmol, 1.5 equiv). The mixture was allowed to stir at RT for 10 mm. Triethylamine (0.5 mL) was added and the mixture was allowed to stir at RT for overnight. Product formation was confirmed by LCMS and TLC, After completion of reaction, the mixture was diluted with water (20 mL) and extracted with ethyl acetate (20 mL x 2). Combined organic extracts were washed with water (20 mL x 4), dried over anhydrous Na S04 and concentrated. The crude product obtained was purified by flash chromatography (5 % MeOH in DCM as an eluent) followed by reverse phase purification to obtain tert-butyl (S)-4-((2-(2-cyano-4,4-difluoropyrrolidin-l-yl)-2- oxoethyl)carbamoyl)-3 ! ,6'-dihydiO-[3,4 ! -bipyridine]-r(2'H)-carboxyiate (0.020 g, 13.15 % Yield) as a white solid.

[0504] LCMS 476.31 l ! |

[0505] 1H NMR (400MHz ,DMSO~d 6 ) d 8.83 (br. s„ 1 H), 8.57 (d, J= 4 8 Hz, 1 H),

8.52 (s, 1 H), 7.36 (d, ./ 5.3 Hz, 1 H), 5.83 (br. s„ 1 H), 5.09 (br. s., 1 H), 4.25 id. J = 13.6 Hz,

2 H), 4.11 (d, J= 5.3 Hz, 2 H), 3.94 (br. s., 2 H), 3.48 (br. s., 2 H), 2.81 (d , J ------ 17.5 Hz, 3 H),

2.67 (hr. s , 2 H), 2 35 (d, J= 14.5 Hz, 3 H), 1.43 (s, 9H).

[0506] Compound 103 TFA.To a stirred solution of N-(2-((S)-2-cyano-4,4-difluoropyrrolidin- l-yl)-2-oxoethyl)-3-(cyclohex-2-en-l-yl)isonicotinamide (0.020 g, 0.042 mmol ,1.0 equiv) in DCM (2 mL), was added TFA (0.02 mL, 0.126 mmol ,3.0 equiv) at room temperature and the mixture was allowed to stir at RT for overnight.. Product formation was confirmed by LCMS. After completion of reaction, the mixture concentrated up to dried to obtained crude. The crude product obtained was purified by reverse phase purification to obtain (S)-N-(2-(2-cyano-4,4- difluoropyrrolidin-l -y])-2-oxoethyl)-r,2',3',6'-teirahydro-[3,4'-bipyridine]-4-c arboxamide 2,2,2- trifluoroacetate (0.005 g, 31.72 % Yield) as off white solid.

[0507] LCMS 376.3I .V1 · I I f

[0508] l H NMR (400MHz ,DMSO-d 6 ) d 8.71 (br. s., 1 H), 8.54 id. ./ 4.4 Hz, 1 H), 8.47 (s, 1 H), 7.33 i d. ./ 4.4 Hz, 2 H), 5.85 (br. s., 1 H), 5.11 (d, J ------ 8.3 Hz, 1 H), 4.26 id. ./ 12.3 Hz, 1

H), 4.15 - 4 00 (m, 2 H), 3.31 (br. s., 4 H), 2.98 - 2.82 (m, 3 H), 2.79 (br. s., 1 H), 2.24 (br s., 2 H), 1.80 (br. s., 3 H), 1.75 (s, 2 H).

Example 55

Synthesis of (S)-N-(2-(2-cyano-4,4-difluoropyrrolidin-l-yl)-2-oxoethyl)-3 -(5-methylfiiran-2- yljisonicotinamide

[0509] Compound 106a. To a stirred solution methyl 3-bromoisonicotinate (0.300 g, 1.4 mmol, 1.0 equiv) in Dioxan (5 mi.) and water (5 mL), was added (5-methylfuran-2-yl)boronic acid (0.212 g, 1.68 mmol, 1.2 equiv), Na 2 C0 3 (0.297 g, 2.8 mmol, 2.0 eqmv) and aerated the reaction mixture with nitrogen for 15 min and added

Dichlorobis(triphenylphosphine)paJladium(II) (0.098 g, 0.14 mmol, 0.1 equiv). Aerated the reaction mixture again with nitrogen for 15 mm. The mixture was allowed to stir at 100°C for 2 h. Product formation was confirmed by LCMS. The reaction mixture was diluted with water (50 rnL) and washed with ethyl acetate (20 rnL c 2). Aqueous layer was separated and freeze dried over lyophiolizer to obtain 3-(5-methylfuran-2-yl)isonicotinic acid (0.250 g).

[0510] LCMS 204.2 [M+H] + [0511] Compound 106. To a stirred solution 3-(5-methylfuran-2-yl)isonicotinic acid (0.100 g, 0.5 mmol, 1 0 equiv) in DMF (3 mL), was added (S)-4,4-difluoro-l-glycylpyrrolidine-2- carbonitrile hydrochloride (0.112 g, 0.5 mmol, 1.0 equiv), HOBt (0.081 g, 0.6 mmol, 1.2equiv) and EDC.HC! (0.115 g, 0.6 mmol, 1.2 equiv). The mixture was allowed to stir at RT for 10 min. Triethyl amine (0.151 g, 1.5 mmol, 3.0 equiv) was added and the mixture was allowed to stir at RT for overnight. Product formation was confirmed by LCMS. The reaction mixture was diluted with water and extracted with ethyl acetate (40 mL c 2). Combined organic extracts were washed with water (25 mL c 6), dried over anhydrous Na 2 S0 4 and concentrated. The crude product obtained was purified by reversed phase HPLC to obtain (S)-N-(2-(2-cyano-4,4- diiluoropyrrolidin-l-yl)-2-oxoethyl)-3-(5-methylfuran-2-yi)i sonicotinamide (0.010 g, 5.04 % Yield) as an off- white solid.

[0512] LCMS 375.3 [M+H] +

[0513] 1H NMR (400 MHz, DMSO-de) d 8.97 (t, J= 5.8 Hz, 1H), 8.92 (s, 1H), 8.52 (d, J = 4.9 Hz, M l ). 7.32 (d, ./ 4.9 Hz, i l l). 6.90 id. ./ 3.4 Hz, 1H), 6.21 (d, J = 3.4 Hz, 1H), 5.13 (dd, J = 9.3, 2.8 Hz, 1 1 1). 4.29 (ddd, J= 16.0, 11.2, 4.1 Hz, i l l ). 4.16 (d , J ------ 5.8 Hz, 2H), 4.12 - 4.03

(m, i l l). 2 97 - 2.74 (m, 2H), 2.34 (s, 3H), 1.24 (s, 1H).

Example 56

Synthesis of (S)-6'-amino-N-(2-(2-cyano-4, 4-difluoropyrrolidin-l-yl)-2-oxoethyl)-[ 3, 3

bipyridine] -4-carboxamide formate

[0514] Compound 107a. To a stirred solution of 3-bromoisonicotinic acid (0.500 g, 2,34 mmol, 1 equiv) in Dioxan (5 mL) and water (5 ml,), was added Na 2 C0 3 (0.496 g, 4.68 mmol, 2 equiv) and 5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyridin-2-ami ne (0.618 g, 2.81 mmol, 1.2 equiv). Aerated the reaction mixture with nitrogen gas for 15 min.

Bis(tripheny!phosphine)pal!adium chloride (0.164 g, 0.234 mmol, 0.1 equiv) was added to the reaction mixture. Reaction mixture was again aerated with nitrogen gas for 15 min. The reaction mixture was allowed to heat at 120°C for 16 h. Progress of the reaction was monitored by LCMS. After completion of the reaction, the reaction mixture was diluted with water (35 niL) Aqueous phase was washed with ethyl acetate (20 mL c 2) and freeze dried on Lyoplulzer to obtain 6'-ammo-|3,3'-bipyridine]-4-carhoxyiic acid (0.600 g, Quant. Yield).

[0515] LCMS 216.2 I M l 11 +

[0516] Compound 107. To a stirred solution of 6'-amino-[3,3'-bipyridine]-4-carboxylic acid (0.200 g, 0.93 mmol, 1 equiv) in DMF (4 mL), was added (S)-4,4-difluoro-l-glycylpyrrolidine- 2-carbonitrile hydrochloride (0.208 g, 0.93 mmol, 1.0 equiv), HOBt (0.151 g, 1.12 mmol,

1 2equiv) and EDC.HC1 (0.214 g, 1.12 mmol, 1.2 equiv). The mixture was allowed to stir at RT for 10 min. Triethylamine (0.282 g, 2.79 mmol, 3.0 equiv) was added and the mixture was allowed to stir at RT for overnight. Product formation was confirmed by LCMS. The reaction mixture was diluted with water and extracted with ethyl acetate (40 mL c 2). Combined organic extracts were washed with water (15 mL x 4), dried over anhydrous Na2S04 and concentrated. The crude product was purified by reversed phase HPLC to obtain (S)-6'-amino-N-(2-(2-cyano- 4,4-difluoropyrrolidin-l-yi)-2-oxoethyl)-[3,3'-bipyridine]-4 -carboxamide formate (0.003 g,

0 8 % Yield).

[0517] LCMS 387.3 [M+H] +

[0518] ! H NMR (400MHZ ,DMSO-d6) d 8 88 (t, ./= 5 8 Hz, 1 1 1 ). 8.64 - 8 55 (m, 2H), 8.18 (s, 1H), 8.03 (s,lH), 7.55 id../ 8.6 Hz. 1H), 7.38 (d, J = 4.9 Hz, 1H), 6.46 (d, J = 8.6 Hz, 1H),

6.11 (s, 2H), 5.13 - 5.03 (m, 1H), 4 30 - 4.17 (m, 1H), 4.05 (dt, ./ 21.0, 8 8 Hz, 3H), 3.95 - 3.80 (m, 1 1 1). 3 26 (s, 3H), 2 94 2.72 (m, 2H), 2 13 (d, J= 16.6 Hz, i l l ). 1.23 is. i l l ). 1.12 id.. J = 16.9 Hz, M l).

Example 57

Synthesis of (S)-N-(2-(2-cyano-4,4-difluoropyrrolidin-l-yl)-2-oxoethyl)-3 -(quinolin-4- yljisonicotinamide

Compound 51

[0519] Compound 51a. To a solution of ethyl 3-bromoisonicotinate (0 3 g, 1.38 mmol, 1.0 equiv) in Toluene (15 mL) and water (2 mi.) was added quinolin-4-ylboronic acid (0.288 g, 1.66 mmol, 1.2 equiv), K3PO43H 2 0 (0.887 g, 4.14 mmol, 3.0 equiv) and resulting reaction mixture purged with N 2 gas for 10 minute, followed by the addition of Pd(OAc) (0.032 g, 0.139 mmol

0.1 equiv) and tricyclohexylphosphine (0.053 g, 0.139 mmol. 0.1 equiv).. The resulting reaction mixture was heated at 100° C for overnight. Product formation was confirmed by LCMS. After the completion of reaction, the mixture was filtered through celite bed, washed with ethyl acetate (100 rnL). Filtrate was concentrated under reduced pressure. The crude product obtained was purified by flash chromatography (100 % ethyl acetate as an eluent) to obtain methyl 3- (quinolin-4-yl)isonicotinate (0 230 g, 62.7 % Yield) as white solid.

[0520] LCMS 265 3[M+H] i

[0521] Compound 51b. To a stirred solution of methyl 3-(quinolin-4-yl)isonicotinate (0.21 g, 0.795 mmol, 1 0 equiv) in THF (7 mi.) and water (7 rnL), was added LiQH.FLQ (0.067 g, 1.59 mmol, 2.0 equiv). The mixture was allowed to stir at RT for overnight. Product formation was confirmed by LCMS. The reaction mixture was concentrated and diluted with water (20 mL) and washed with ethyl acetate (10 mL x 2). Aqueous layer was separated and freeze dried on lyophilyzer to obtain 3-(quinolin-4-yl)isonicotinic acid (Quant. Yield) as white solid.

[0522] LCMS .251 2| VI · 1 11

[0523] Compound 51. To a stirred solution of 3-(quinolin-4-yl)isonicotinic acid (0.12 g, 0.48 mmol, 1.0 equiv) in DMF (5 mL), was added (S)-4,4-diiluoro-l-glycylpyrrolidine-2-carbonitrile hydrochloride (0 108 g, 0.48 mmol, 1.0 equiv), EDCI.HC1 (0.138 g, 0.72 mmol, 1.5 equiv) & HOBt (0.097 g, 0.72 mmol, 1.5 equiv). The mixture was allowed to stir at RT for 10 min.

Triethylamine (1 mL) was added and the mixture was allowed to stir at RT for overnight.

Product formation was confirmed by LCMS and TLC. After completion of reaction, the mixture was diluted with water (20 mL) and extracted with ethyl acetate (20 mL x 2). Combined organic extracts were washed with water (20 mL c 4), dried over anhydrous Na 2 S0 4 and concentrated. The crude product obtained was purified by flash chromatography (5 % MeOH in DCM as an eluent) to obtain (S)-N-(2-(2-cy ano-4,4-difluoropy rrolidin- 1 -yl)-2-oxoethy l)-3-(quinolin-4- yl)isonicotinamide (0.040g, 19.8 % Yield) as an white solid.

[0524] LCMS 422 2| M · 1 11

[0525] ' l l NMR (400MHz ,DMSO-d 6 ) d 9.00 (br s., 1 H), 8.92 - 8.81 (m, 2 H), 8.65 (s, 1 H), 8.08 id. ./ 8.3 Hz, 1 H), 7.78 (d, J= 4.8 Hz, 1 H), 7.70 (d, J = 4.8 Hz, 1 H), 7.55 (br. s., 2 H), 7.47 id. ./ 4.4 Hz, 1 H), 5.03 (d, J= 8.3 Hz, 1 H), 4.1 1 (br. s., 1 H), 3.96 - 3.74 (m, 3 H), 2.82 (br. s., 1 H), 2.74 (d, ./ 9.2 Hz, 1 H)

Example 58

Synthesis of (S)-N-(2-(2-cyano-4, 4-difluoropyrrolidin-l-yl)-2-oxoethyl) -4- (4 -fluorophenyl) -6-

[0526] Compound 109a. To a solution of methyl 4-chloro-6-oxo-l ,6-dihydropyridine-3- carboxylate (1 g, 5.3191 mmol, 1.0 equiv) in Dioxan (30 mL): water (10 mL) was added compound 4-fluorophenyl)boronic acid (0.82 g, 5.8510 mmol, 1.1 equiv), Na 2 C0 3 (1.12 g, 10.63 mmol, 2.0 equiv) and resulting reaction mixture purged with N 2 gas for 10 minute, followed by the addition of Pd(PPh 3 ) 2 Cl2 (0.187 g, 0.2659 mmol. 0.05 equiv). The resulting reaction mixture was heated at 100° C for overnight. Product formation was confirmed by LCMS. After the completion of reaction, the mixture was filtered through celite bed, washed with ethyl acetate (100 mL). Filtrate was concentrated under reduced pressure. Diluted with water & extracted with EtOAc (3 x 50 mL), organic layer was dried over Na 2 S0 4 & concentrated under reduced pressure .The crude product obtained was purified by flash chromatography (0-50 % ethyl acetate in hexane as an eluent) to obtain compound of methyl 4-(4-fluorophenyl)-6-oxo-l,6- dihydropyridine-3-carboxylate . (1.2g, 81.34 %) as a white solid.

[0527] LCMS 247.8 [M+H] + [0528] ! H NMR (400 MHz, DMSO-de) d 12 22 (br. s., 1 H), 8 05 (s, 1 H), 7.65 - 7.58 (m, 1 H), 7.56 (d, J= 6.6 Hz, 1 H), 7.35 - 7.26 (m, 1 H), 7.22 - 7.14 (m, 1 H), 6.21 (s, 1 H), 3.61 - 3 52 (m, 3 H)

[0529] Compound 109b. To a stirred solution of compound methyl 4-(4-fluorophenyl)-6-oxo-

1.6-dihy dropyridine-3-carboxylate (1.2 g, 4.8582 mmol, 1 0 equiv) in THF (30 mL) and water (10 mL), was added LiOH (0.225 g, 9.7165 mmol, 2.0 equiv). The mixture was allowed to stir at 80°C for overnight. Product formation was confirmed by LCMS. The reaction mixture was concentrated and diluted with water (30 mL) and washed with ethyl acetate (10 mL x 2).

Aqueous layer was separated and freeze dried on lyophilyzer to obtain compound 4-(4~ fluorophenyl)-6-oxo-l,6-dihydropyridine-3-earboxylic acid (1 gm, 88%) as a white solid.

[0530] LCMS: 234.0[M+H] +

[0531] 'l l NMR (400 MHz, DMSO-d,.} 511.07 (s, i l l :· 7.63 (s, 1 H), 7.34 (dd, J ------ 5.7, 8.3 Hz,

2 H), 7.11 (t, ./ = 9.0 Hz, 2 H), 6 03 (s, 1 H), 3.53 (s, 1 H).

[0532] Compound 109. To a stirred solution of 4-(4~fiuoropheny!)-6-oxo-l,6-dihydropyridine- 3-carboxylic acid (0.4 gm, 1.7167 mmol, l.Oequiv), HATU(0.278 gm, 2.064 mmol, 1.2 equiv) in DMF (10 ml) after 10 min was added a (S)-4,4~difluoro-l -glycylpyrrolidine-2-carbonitrile hydrochloride (0 464 gm, 2.06 mmol, 1.2 equiv) and stirred the reaction mixture for10 min, followed by drop wise addition of Et 3 N (0.5ml, 3.4334 mmol, 3 equiv) allowed the reaction for 16h stirring at rt. reaction progress was monitored by LCMS and TLC, workup done by addition of chilled water(50 mL) to the reaction mass and extracted by ethyl acetate (3 c 50 ml) and collected all the organic layers, washed by water by three times and once by sodium bicarbonate and once by brine solution. Organic layer was dried over anhydrous Na 2 S04, concentrated on reduced pressure crude was purified by reverse phase chromatography to afford the desired product (S)-N-(2-(2-cyano-4,4-difluoropyrrolidin-l-yl)-2-oxoethyl)-4 -(4-fluorophenyl)-6-oxo-

1.6-dihydropyridine-3-carboxamide (100 mg, 20 %).

[0533] LCMS: 405.2[M+H] +

[0534] 'l l NMR (400 MHz, DMSO-d,.} 511.93 (br. s., 1 H), 8.69 (br. s., 1 H), 7.64 (s, 1H),

7 44 (br. s , 2 H), 7 17 (t, J= 8 8 Hz, 2 H), 6.30 (s, 1 H), 5.08 (br. s., 1 H), 4.20 (d, J= 11 0 Hz, 2 H), 3.99 - 3.89 (m, 2 H), 2.80 (d, J = 13.6 Hz, 2 H).

Example 59 Synthesis of (S)~N~(2-(2-cyano-4,4~difluoropyrrolidin-l-yl)-2-oxoethyl)-3 ~(3,5-dimethylisoxazol~

4-yl)isonicotinamide

ompoun

[0535] Compound 1 10a. To a solution of ethyl 3-bromoisonicotinate (0 1 g, 0.463 mmol, 1.0 equiv) in Toluene (5 mL) was added 3,5-dimethyl-4-(4,4,5,5-tetramethyi-l,3,2-dioxaborolan-2- yi)isoxaz.oie (0.126 g, 0.926 mmol, 2 equiv), Is. ' CO ; (0.192 g,l.39 mmol, 3.0 equiv) and resulting reaction mixture purged with N 2 gas for 10 minute, followed by the addition of Pd(PPh 3 )4 (0.027 g, 0.023 mmol. 0.05 equiv). The resulting reaction mixture was heated at l00°C for overnight. Product formation was confirmed by LCMS. After the completion of reaction, the mixture was filtered through celite bed, washed with ethyl acetate (100 mL).

Filtrate was concentrated under reduced pressure. The crude product obtained was purified by flash chromatography (0-30 % ethyl acetate in hexane as an eluent) to obtain methyl 3-(3,5- dimethylisoxazol-4-yl)isonicotinate (0.105 g, 97.2% Yield) as an yellow liquid.

[0536] LCMS 233.2[M+H] +

[0537] h-J NMR (400MHz, DMSO-de) d 8.81 id, J = 4.8 Hz, 1 H), 8.65 (s, 1 H), 7 85 (d, J = 5.3 Hz, 1 H), 3.32 (s, 3 H), 2.23 (s, 3 H), 2.09 - 1.90 (m, 3 H).

[0538] Compound 110b. To a stirred solution of 3-(3,5-dimethylisoxazol-4-yl)isonicotinate (0.120 g, 0.518 mmol, 1.0 equiv) in THF (5 mL) and water (5 mL), was added Li0H.2H 2 0 (0.043 g, 1.034 mmol, 2,0 equiv). The mixture was allowed to stir at RT for overnight Product formation was confirmed by LCMS. The reaction mixture was concentrated and diluted with water (20 mL) and washed with ethyl acetate (10 mL x 2). Aqueous layer was separated and freeze dried on lyophilyzer to obtain 3-(3,5-dimethylisoxazol-4-yl)isonicotinic acid (Quant. Yield) as a white solid.

[0539] LCMS 2 19 21 VM i f [Q54Q] Compound 1 10. To a stirred solution of 3-(3,5-dimethylisoxazol-4-yl)isonicotinic acid (0.1 g, 0.46 mmol, 1.0 equiv) in DMF (4 mL), was added (S)-4,4-difluoro-i-glycylpyrrolidine~2~ carbonitrile hydrochloride (0.103 g, 0.46 mmol, 1.0 equiv), EDCI.HC1 (0.132 g, 0.69 mmol, 1.5 equiv) & HOBt (0.093 g, 0.69 mmol, 1.5 equiv). The mixture was allowed to stir at RT for 10 min. Triethylamine (0.5 mL) was added and the mixture was allowed to stir at RT for overnight. Product formation was confirmed by LCMS and TLC. After completion of reaction, the mixture was diluted with water (20 mL) and extracted with ethyl acetate (50 mL c 3). Combined organic extracts were washed with water (50 mL c 5), dried over anhydrous Na 2 S0 4 and concentrated. The crude product obtained was purified by flash chromatography (5 % MeOH in DCM as an eluent to obtain (S)-N-(2-(2-cyano-4,4-difluoropyrrolidin-l-yl)-2-oxoethyl)-3 -(3,5- dimethylisoxazol-4-y])isonicotinamide (0 Q55g, 30 7 % Yield) as an white solid.

[0541] LCMS 390.2[M+H] +

[0542] 1H NMR (400MHz .DVISO-d,.) d 8 85 (d, J= 5.7 Hz, 1 H), 8.75 (d, ./= 4 8 Hz, 1 H), 8.56 (s, 1 H), 7.56 (d, J = 4.8 Hz, 1 H), 5.10 - 4.98 (m, 1 H), 4.27 - 4.18 (m, 1 H), 4.11 - 3.95 (m, 2 H), 2.95 - 2.74 (m, 3 H), 2.25 (s, 3 H), 2.06 (s, 3 H).

Example 60

Synthesis of (S)-3-(4-chloro-3-fluorophenyl)-N-(2-(2-cyano-4, 4-difluoropyrrolidin-J-yl)-2 oxoethyljisonicotinamide

Compound 111

[0543] Compound 1 1 la. To a solution of ethyl 3-bromoisonicotinate (1) (0.2 g, 0.926 mmol,

1.0 equiv) in Toluene (10 mL) was added 3,5-dimethyl-4~(4,4,5,5~tetramethyl~l,3,2- dioxaborolan-2-yl)isoxazole (2) (0.242 g, 1.39 mmol, 1.5 equiv), K2CO3 (0.383 g, 2.79 mmol,

3.0 equiv) and resulting reaction mixture purged with N 2 gas for 10 minute, followed by die addition of Pd(PPh 3 )4 (0.053 g, 0.046 mmol. 0.05 equiv). The resulting reaction mixture was heated at 100°C for overnight. Product formation was confirmed by LCMS. After the completion of reaction, the mixture was filtered through celite bed, washed with ethyl acetate (100 mL). Filtrate was concentrated under reduced pressure. The crude product obtained was purified by flash chromatography (0-20 % ethyl acetate in hexane as an eluent) to obtain methyl 3-(3,5-dimethylisoxazol-4-yl)isonicotinate (3) (0 250 g, 98% Yield ) as an off-white solid

[0544] LCMS 266 11 M · 1 11

[0545] Compound 11 lb. To a stirred solution of methyl 3~(4-ehloro-3- fluorophenyl)isomcotinic acid (0.25 g, 0.94 mmol, 1.0 equiv) in THF (8 mL) and water (8 niL), was added LiOH.H 2 0 (0 080 g, 1.89 mmol, 2 0 equiv). The mixture was allowed to stir at RT for overnight. Product formation was confirmed by LCMS. The reaction mixture was

concentrated and diluted with water (20 mL) and washed with ethyl acetate (10 mL x 2).

Aqueous layer was separated and freeze dried on lyophi!yzer to obtain 3-(4-chloro-3- fluorophenyl)isonicotinic acid (Quant. Yield) as a white solid.

[0546] LCMS 252 1 [M+H] +

[0547] ! H NMR (400MHZ, DMSO-de) 68 47 - 8.35 (m, 2 H), 7 67 - 7.51 (m, 2 H), 7 42 (d, J = 8.3 Hz, 1 H), 7.19 (d, J = 4.8 Hz, 1 H).

[0548] Compound 111. To a stirred solution of 3-(4-chioro-3-fiuoropheny!)isonicotimc acid (0.12 g, 0.48 mmol, 1.0 equiv) in DMF (5 mL), was added (S)-4,4-diiluoro-l-glycylpyrrolidine~ 2-carbonitrile hydrochloride (0.108 g, 0.48 mmol, 1.0 equiv), EDCLHCl (0.138 g, 0.717 mmol,

I.5 equiv) & HOBt (0.097 g, 0.717 mmol, 1.5 equiv). The mixture was allowed to stir at RT for 10 min. Triethylamine (1 mL) was added and the mixture was allowed to stir at RT for overnight. Product formation was confirmed by LCMS and TLC. After completion of reaction, the mixture was diluted with water (20 mL) and extracted with ethyl acetate (50 mL * 3).

Combined organic extracts ware washed with water (50 mL c 5), dried over anhydrous Na 2 S0 4 and concentrated. The crude product obtained was purified by flash chromatography (5 % MeOH in DCM as an eluent) to obtain (S)-3-(4-chloro-3-fluorophenyl)-N-(2-(2-cyano-4,4- difluoropyrrolidin-l-yl)-2-oxoethyl)isonicotinamide (0.120g, 59.1 % Yield) as an white solid.

[0549] LCMS 423.51 Al i i ]

[0550] i NMR (400MHz ,DMSO-d 6 ) d 9 04 (t, ./ = 5.9 Hz, 1 H), 8.76 - 8.63 (m, 2 H), 7.65 - 7 54 (m, 2 H), 7 49 (d, J= 4.8 Hz, 1 H), 7.41 (d, J= 7 0 Hz, 1 H), 5.09 (br. s , 1 H), 4 22 (d, J =

I I.8 Hz, 1 H), 4.12 - 3.96 (m, 2 H), 3.92 id. ./ 5.3 Hz, 1 H), 2.97 - 2.76 (m, 2 H). Example 61

Synthesis of (S)~3~( 3~( tert-butyl)phenyl)-N~( 2~(2~cyano~4, 4-difluoropyrrolidin-l -yl)~2- oxoethyl)isonicotinamide

[0551] Compound 1 12a. To a solution of l-bromo-3-(tert-butyl)benzene (0.2 g, 0.938 mmol, 1.0 equiv) in DMSO (10 mL) was added 4,4,4',4',5,5,5',5'-octamethyl-2,2'-bi(l,3,2- dioxabofolane) (0.356 g, 1.41 mmol, 1.5 equiv), KOAc (0.184 g, 1.876 mmol, 2.0 equiv) and resulting reaction mixture purged with N 2 gas for 10 minute, followed by the addition of Pd(PPh 3 ) 4 (0.055 g, 0.047 mmol. 0.05 equiv). The resulting reaction mixture was heated at l00°C for overnight. Product formation was confirmed by LCMS. After the completion of reaction, the mixture was filtered through celite bed, washed with ethyl acetate (100 mL). Filtrate was concentrated under reduced pressure. The crude product obtained was purified by flash chromatography (hexane as an eluent) to obtain 2-(3-(tert-butyl)phenyl)-4, 4,5,5- tetramethyl-l,3,2-dioxaborolane (0.230 g, 94.26% Yield) as an off-white solid.

[0552] LCMS 26 i .3I M i i |

[0553] h-J NMR (400MHz, DMSO-de) 57.67 (s, 1 H), 7 57 - 7 45 (m, 2 H), 7 32 (t, J 7 5 Hz, 1 H), 1 44 (d, J= 6.6 Hz, 1 H), 1.39 - 1.23 (m, 9 H), 1.23 - 1.04 (m, 12 H)

[0554] Compound 112b. To a solution of 2-(3-(tert-butyl)phenyl)-4,4,5,5-tetramethyl-l,3,2- dioxaborolane (0.216 g, 0.833 mmol, 1.2 equiv) in Toluene (15 mL) w as added ethyl 3- bromoisonicotinate (0.150 g, 0.649 mmol, 1.0 equiv), K 2 C0 3 (0.287 g, 2 08 mmol, 3.0 equiv) and resulting reaction mixture purged with N gas for 10 minute, followed by the addition of Pd(PPh 3 ) 4 (0.040 g, 0.035 mmol. 0.05 equiv). The resulting reaction mixture w¾s heated at 100°C for overnight. Product formation wus confirmed by LCMS. After the completion of reaction, the mixture was filtered through celite bed, washed with ethyl acetate (100 mL). Filtrate w¾s concentrated under reduced pressure. The crude product obtained was purified by flash chromatography (0-20 % ethyl acetate in hexane as an eluent) to obtain methyl 3-(3-(tert- butyl)phenyl) isomcotinate (0.150 g, 80.6 %) as yellow semi solid.

OSSSj LCMS 270.5[M+H] +

[0556] 'l l NMR (400MHz, DMSO-d,.} d 8.75 - 8.66 (m, 2 H), 7.74 (d, J= 4.8 Hz, 1 H), 7.66 (d, J 4.8 Hz, 1 H), 7 51 - 7.39 (m, 2 H), 7 24 (d, J = 7.5 Hz, 1 H), 3.66 (s, 3 H), 1.30 (s, 9 H).

[0557] Compound 112c. To a stirred solution of methyl 3-(3-(tert-butyl)phenyl)isonicotinate (0.150 g, 0.555 mmol, 1.0 equiv) in THF (4 mL) and water (4 mL), was added LiOH.H?0 (0.047 g, 1.11 mmol, 2.0 equiv). The mixture was allowed to stir at RT for overnight. Product formation was confirmed by LCMS The reaction mixture was concentrated and diluted with water (20 mL) and washed with ethyl acetate (10 ml, x 2). Aqueous layer was separated and freeze dried on lyophilyzer to obtain 3-(3-(tert-butyl)phenyl)isonicotinic acid (Quant. Yield) as a white solid.

[0558] LCMS 256.3[M+H]'

[0559] Compound 112. To a stirred solution of 3-(3-(tert-butyl)phenyl)isonicotinic acid (0.1 g, 0.392 mmol, 1.0 equiv) in DMF (5 mL), was added (S)-4,4-difluoro-l-glycylpyrrolidine-2- carbonitnle hydrochloride (0.89 g, 0.392 mmol, 1.0 equiv), EDCI.HC1 (0.113 g, 0.59 mmol, 1.5 equiv) & HOBt (0.080 g, 0.59 mmol, 1.5 equiv). The mixture was allowed to stir at RT for 10 mm. Triethyiamine (0.5 mL) was added and the mixture was allowed to stir at RT for overnight. Product formation was confirmed by LCMS and TLC. After completion of reaction, the mixture was diluted with water (20 mL) and extracted with ethyl acetate (50 mL x 3). Combined organic extracts were washed with water (50 mL c 5), dried over anhydrous Na 2 S0 4 and concentrated. The crude product obtained was purified by flash chromatography (5 % MeOH in DCM as an eluent) followed by prep purification to obtain (S)-3-(3-(tert-butyJ)phenyJ)-N-(2-(2-cyano-4,4- difluoropyrrolidin-l-yl)-2-oxoethyl)isonicotinamide (0.028g, 16 86 % Yield) as an white solid

[0560] LCMS 427.3[M+H] +

[0561] 'H NMR (400MHz .DMSO-d, ,) d 8.92 (br. s., 1 H), 8.76 - 8.56 (m, 2 H), 7.50 (s, 1 H), 7.48 - 7.40 (m, 2 H), 7.40 - 7.29 (m, 2 H), 5.14 - 5.04 (m, 1 H), 4.21 (br. s., 1 H), 4.03 i d. ./ 2.6 Hz, 2 H), 2.87 (br s., 1 H), 2.79 (d, J 14.5 Hz, 1 H), 2.67 (br. s., 1 H).

Example 62 Synthesis of (S)-N-(2-(2-cyano-4,4-difluoropyrrolidin-l-yI)-2-oxoethyl)-3 -ethoxyisonicotinamide

Compound 113

[0562] Compound 113. To a stirred solution of 3-ethoxyisomcotinic acid (0.05 g, 0.299 mmol, 1.0 equiv) in DMF (3 mL), was added (S)-4,4-dif!uoro-l-glycylpyrrolidine-2-carbonitrile hydrochloride (0.067 g, 0.299 mmol, 1.0 equiv), EDCT.HC1 (0.086 g, 0.488 mmol, 1.5 equiv) & HOBt (0.06 g, 0.488 mmol, 1.5 equiv). The mixture was allowed to stir at RT for 10 min.

Triethylamine (0.5 mL) was added and the mixture was allowed to stir at RT for overnight. Product formation was confirmed by LCMS and TLC. After completion of reaction, the mixture was diluted with water (20 mL) and extracted with ethyl acetate (50 mL c 3). Combined organic extracts were washed with water (50 mL x 5), dried over anhydrous Na 2 S0 4 and concentrated. The crude product obtained was purified by flash chromatography (5 % MeOH in DCM as an eluent to obtain (S)-N-(2-(2-cyano-4,4-difluoropynOlidin-l-yl)-2-oxoethyl)-3- ethoxyisonicotinamide (0.07 g, 30.7 % Yield) as an off-white solid.

[0563] LCMS 339.2| V1 · 1 1 |

[0564] 'l l NMR (400MHz .DMSO-d,.} 6 8.71 (br. s., 1 H), 8.59 (s, 1 1 1 }. 8.34 i d. ./ 4.8 Hz, 1

H), 7.70 (d, J ----- 4.8 Hz, 1 H), 5.15 (dd, J 2.9, 9 0 Hz, 1 H), 4.36 (q, J 7.0 Hz, 2 H), 4 30 - 4.12 (m, 3 H), 4.12 - 3.98 (m, 1 H), 2.90 - 2.68 (m, 2 H), 1.46 (t, J= 6.8 Hz, 3 H).

Example 63

Synthesis of (S)-N-(2-(2-cyano-4,4-dijluoropyrrolidin-l-yl)-2-oxoethyl)-3 -((4- fluorophenyijaminojisonicotinarnide 2, 2, 2-trifluoroacetate

[0565] Compound 114a. To a stirred solution of methyl 3-bromoisonicotinate (0.200 g, 0.93 mmol, 1 equiv) in toluene (3 mL), was added 4-fluoroaniline (0.103 g, 0.93 mmol, 1.0 equiv), Tris(dibenzyiideneacetone)dipaiiadium(0) (0.043 g, 0.046 mmol, 0.05 equiv), xantphos (0.053 g, 0.093 mmol, 0.1 equiv) and CS 2 CO 3 (0.456 g, 1.4 mmol, 1.5 equiv). The resulting reaction mixture was heated at 190 °C for 1 h in microwave. Reaction progress was checked by LCMS. The reaction mixture was diluted with w¾ter (15 mL) and extracted with ethyl acetate (30 mL c 2) Combined organic extracts were washed with brine (30 mL), dried organic extract over anhydrous Na 2 SC> 4 , filtered and concentrated under reduced pressure. Crude compound was purified by normal phase combi-flash chromatography to obtain methyl 3-((4- iluorophenyl)amino)isonicotinate (0.120 g, 53 % Yield ) as an off-white solid.

[0566] LCMS: 246.8

[0567] Compound 1 14b. To a stirred solution of methyl 3-((4- fluorophenyl)annno)isonicotinate (0.350 g, 1.4 mmol, 1 equiv) in THF and water (1:1 )(6 mL), w¾s added LiOHTLO (0.089 g, 2.1 mmol, 1.5 equiv). The mixture was allowed to stir at ambient temperature for 16 h. Product formation was confirmed by LCMS. The reaction mixture was diluted with water (20 mL), extracted with ethyl acetate and concentrated the aqueous layer under reduced pressure and lyophilized to obtain 3-((4-fluorophenyl)amino)isonicotinic acid (0.350 g. Quant Yield) as an off-white solid

[0568] LCMS 233.2 I M l 11 +

[0569] Compound 114. To a stirred solution of 3-((4-fluoropheny!)amino)isonicotinic acid (0.350 g, 1.5 mmol, 1 equiv) in DMF (4 mL), was added (lR)-4,4-difluoro-2- glycylcyclopentane-l-carbonitrile hydrochloride (0.337 g, 1.5 mmol, 1.0 equiv), HOBt (0.243 g, 1.8 mmol, 1 2equiv), EDC.HCI (0.343 g,l 8 mmol, 1.2 equiv) and triethyl amine (0 455 g, 4.5 mmol, 3 equiv) was added and the mixture was allowed to stir at ambient temperature for 16 h. Product formation was confirmed by LCMS. The reaction mixture was diluted with water (15mL) and extracted with ethyl acetate (40 mL x 3). Combined organic extracts were washed with water (25 mL c 5), dried over anhydrous Na 2 S0 4 , filtered and concentrated under reduced pressure to obtain crude product which was purified by reverse phase HPLC to obtain (S)-N-(2- (2-cyano-4,4-difluoropyrrolidin-l-yl)-2-oxoethyl)-3-((4-fluo rophenyl)amino)isonicotinamide as a ΐ! ; A salt (0.003 g, 0.5 % Yield).

[0570] LCMS 404.2 [M+H] +

[0571] 'l l NMR (400MHz ,DMSO-d6) 6 9.22 (t, J= 6.0 Hz, 1H), 9.12 (s, 1H), 8.58 (s, 1H), 8.50 (q, J= 15.7 Hz, 1H), 8.15 (s, 1H), 7.68 - 7.63 (m, 1H), 7.32 - 7.14 (m, 4H), 5.12 idd. J 9.3, 2.9 Hz, i l l). 4.30 (ddd, ./= 15.9, 1 1 5, 4.6 Hz, i l l ). 4 20 - 4.06 (m, 31 1 ). 2.92 - 2.73 (m, 2H), 2.33 - 2.25 (m, i l l). 1.56 - 1.49 (m, 1H), 1.23 (s, 1H), 0.87 (d, J= 12.4 Hz, 1H).

Example 64

Synthesis of (S)-N-(2-(2-cyano-4, 4-difluoropyrrolidin-l-yl)-2-oxoethyl)-3-(4- fluorophenoxyjisonicoiinamide

[0572] Compound 35a. To a stirred solution of 3-bromopyridine-4-carboxylic acid (1.0 g, 5.0 mmol, 1 equiv) in DMF (10 mi), was added 4-fluorophenol (0.560 g, 5.0 mmol, 1 equiv), Cul (1.90 g, 10.0 mmol, 2 equiv) and Cs CCL (3.260 g, 10.0 mmol, 2 equiv). Heated the reaction mixture at 110 °C for 18 h. Reaction progress was checked by LCMS. The reaction mixture was diluted with water (20 mL) and added few drops of dil. HC1 till pH was slightly acidic. The aqueous layer was extracted with ethyl acetate (50 mL c 2). Combined organic extracts were washed with brine (50 mL), dried over anhydrous Na 2 S04, filtered and concentrated under reduced pressure to obtain 3-(4-fluorophenoxy)pyridine-4-carboxylic acid (0.200 g, 18 % Yield )

[0573] LCMS: 234 2 [M+H] + [Q574] Compound 35. To a stirred solution of 3-(4-fluorophenoxy)pyridine-4-carboxylic acid (0.200 g, 0.86 mmol, 1 equiv) in DMF (3 mL), was added (S)-4,4-difluoro~ 1 ~gly cylpyrrolidine- 2-carbonitrile hydrochloride (0.194 g, 0.86 mmol, 1.0 equiv), HOBt (0.140 g, 1.03 mmol, 1.2equiv) and EDC.HC1 (0.197 g, 1 03 mmol, 1.2 equiv). The mixture was allowed to stir at RT for 10 min. Triethyl amine (0 260 g, 2.58 mmol, 3 equiv) was added and the mixture was allowed to stir at ambient temperature for 16 h. Product formation was confirmed by LCMS. The reaction mixture was diluted with water (lOmL) and extracted with ethyl acetate (40 mL c 2). Combined organic extracts were washed with water (25 mL c 5). Organic layer was dried over anhydrous Na^SCL, filtered and concentrated under reduced pressure to obtain crude product.

The crude product obtained was purified by reversed phase HPLC to obtain (S)-N-(2-(2-cyano- 4,4-difluoropyrrolidin-l-yl)-2-oxoethyl)-3-(4-iluorophenoxy) isonicotinamide (0.040 g, 1 1.5 % Yield) as an off-white solid.

[0575] LCMS 405.2 [M+H] +

[0576] 1H NMR (400MHz ,DMSO-d6) d 8.80 (t, J= 5.6 Hz, 1H), 8.48 (d, J= 4.8 Hz, 1H), 8.24 (s, I I I). 7.65! d. J 5.0 Hz, 11 1 ). 7.26 (t, J = 8.7 Hz, 2H), 7.18 (dd, J 9.0, 4.5 Hz, 2H), 5.10 (dd, J 8 9, 3.0 Hz, 1H),4.30 - 3.98 (m, 4H), 2 93 - 2.72 (m, 2.1 1 ).

Example 65

Synthesis of (S)-N-(2-(2-cyano-4,4-difluoropyrrolidin-l-yl)-2-oxoethyl)-3 - (phenylthio)isomcotinamide

[0577] Compound 115a. To a solution of methyl 3-bromoisonicotinate (0.2 g, 0.93 mmol, 1.0 equiv) in Dioxane (10 mL) was added benzenethiol (0.204 g, 1.85 mmol, 2.0 equiv), KOAc (0.184 g, 1.876 mmol, 2.0 equiv) and resulting reaction mixture purged with N 2 gas for 10 minute, followed by the addition of Pd 2 (dba)3 (0.042 g, 0.046 mmol. 0.05 equiv, ), Xanthphose (0.026 g, 0.046 mmol, 0.05 equiv) and resulting reaction mixture). The resulting reaction mixture was heated at 90°C for overnight. Product formation was confirmed by LCMS. After the completion of reaction, the mixture was filtered through celite bed, washed with ethyl acetate (100 mL). Filtrate was concentrated under reduced pressure. The crude product obtained was purified by flash chromatography (20% ethyl acetate hexane as an eluent) to obtain methyl 3- (phenylthio)isonicotinate (0.100 g, 44.4 % Yield) as yellow oil.

[0578] LCMS 246.1 [M+H] +

[0579] l H NMR (400MHz ,DMSO-d 6 ) d 8.92 (br. s., 1 H), 8.76 - 8.56 (m, 2 H), 7.50 (s, 1 H), 7.48 - 7.40 (m, 2 H), 7.40 - 7.29 (m, 2 H), 5.14 - 5.04 (m, 1 H), 4.21 (br. s., 1 H), 4.03 (d, J= 2.6 Hz, 2 H), 2 87 (br. s., 1 H), 2.79 (d, J 14 5 Hz, 1 H), 2.67 (br s., 1 H).

[0580] Compound 115b. To a stirred solution of obtain methyl 3-(phenylthio)isonicotinate (0.150 g, 0.614 mmol, 1.0 equiv) in THF (5 mL) and water (5 mL), was added LiOH.H 2 0 (0.052 g, 1.23 mmol, 2.0 equiv). The mixture was allowed to stir at RT for overnight. Product formation was confirmed by LCMS. The reaction mixture was concentrated and diluted with water (20 mL) and washed with ethyl acetate (10 mL x 2). Aqueous layer was separated and freeze dried on lyophilyzer to obtain 3-(phenylthio)isonicotinate (Quant. Yield) as white solid.

[0581] LCMS 232.31 M i l |

[0582] Compound 1 15. To a stirred solution of 3-(phenylthio)isonicotinate (0.15 g, 0.65 mmol, 1.0 equiv) in DMF (5 mL), was added (S)-4,4-difluoro-l-glycylpyrrolidine-2-carbonitrile hydrochloride (0.146 g, 0.65 mmol, 1.0 equiv), EDCI.HC1 (0.190 g, 0.97 mmol, 1.5 equiv) & HOBt (0.132 g, 0.97 mmol, 1.5 equiv). The mixture was allowed to stir at RT for 10 min

Tri ethyl amine (0.3 mL) was added and the mixture was allowed to stir at RT for overnight. Product formation was confirmed by LCMS and TLC. After completion of reaction, the mixture was diluted with water (20 mL) and extracted with ethyl acetate (50 mL c 3). Combined organic extracts were washed with water (50 mL c 5), dried over anhydrous Na 2 S0 4 and concentrated. The crude product obtained was purified by flash chromatography (5 % MeOH in DCM as an eluent) followed by prep purification to obtain (S)-N-(2-(2-cyano-4,4-difluoropyrrolidin-l -yl)-2- oxoethyl)-3-(phenylthio)isonicotinamide (O.OlOg, 3.8 % Yield) as a white solid.

[0583] LCMS 403.2| YM 1 | [0584] ! H NMR (400MHZ, DMSO-de) d 9.03 (br. s , 1 H), 8.52 (d, J= 4.8 Hz, 1 H), 8.18 (s, 1 H), 7.56 - 7.32 (m, 6 H), 5.13 (d, J= 7.5 Hz, 1 H), 4.29 (br s., 1 H), 4.24 - 4.05 (m, 2 H), 2 82 (d , J = 14.9 Hz, 3 H).

Example 66

Synthesis of (S)-N-(2-(2-cyano-4,4-diflmropyrrolidin-l-yl)-2-oxoethyl)-6- oxo-4-(quinolin-4-yl)- l, 6-dihydropyridine-3-carhoxamide

[Q585] Compound 116a. To a solution of methyl 4-chloro-6-oxo-l,6-dihydropyridine-3- carboxylate (0.2 g, 1.07 mmol, 1.0 equiv) in Dioxane (10 mL) was added quinolin-4-ylboronic acid (0.22 g, 1.26 mmol, 2,0 equiv), K3PO4.3H2O (0 455 g, 2.14 mmol, 2 0 equiv) and resulting reaction mixture purged with N 2 gas for 10 minute, followed by the addition of Pd 2 (dba) 3 (0.024 g, 0.11 mmol. 0.1 equiv, ), tncyclohexylphoshpine (0.030 g, 0.11 mmol, 0.1 equiv). The resulting reaction mixture was heated at 140°C for overnight. Product formation was confirmed by LCMS. After the completion of reaction, the mixture was filtered through celite bed, washed with 10% methanol in DCM (100 mL). Filtrate was concentrated under reduced pressure. The crude product obtained was purified by flash chromatography (5 methanol in DCM as an eluent) to obtain methyl 6-oxo-4-(quinolin-4-yl)-l,6-dihydropyridine-3-carboxylate (3) (0.058 g,

19.4 %) as off white solid.

[0587] ! H NMR (400MHz ,DMSO-d 6 ) d 12.47 (br. s., 1 H), 8.91 (d, ./ 4.4 Hz, 1 H),

8 23 (s, 1 H), 8.11 - 7.96 (m, 1 H), 7.84 - 7.70 (m, 1 H), 7.64 - 7.43 (m, 2 H), 7.35 (d, J= 4 4 Hz, 1 H), 6.30 (s, 1 H), 3.35 (s, 3 H).

[0588] Compound 116b. To a stirred solution of obtain methyl 6-oxo-4-(quinoiin-4-yl)-l,6- dihydropyridine-3-carboxylate (0.115 g, 0.412 mmol, 1.0 equiv) in THF (4 mL) and water (4 mL), was added LiOH.H 2 0 (0.026 g, 0.616 mmol, 1.5 equiv). The mixture was allowed to stir at RT for overnight. Product formation was confirmed by LCMS. The reaction mixture was concentrated and diluted with water (20 mL) and washed with ethyl acetate (10 mL x 2).

Aqueous layer was separated and freeze dried on lyophilyzer to obtain 6-oxo-4-(qmnoIm-4-yl)- l,6-dihydropyridine-3-carboxylate (Quant. Yield) as white solid.

[0589] LCMS 267.2[M+H] +

[0590] ' l l NMR (400MHz, DMSO-de) d 11.54 (d, J = 1 7 1 Hz, 1 H), 8 80 (d, J= 4.4 Hz, 1 H), 8.05 - 7.86 (m, 2 H), 7.67 (t, J= 7.2 Hz, 1 H), 7.58 (d, J = 8.3 Hz, 1 H), 7.21 (d, J 4.4 Hz,

1 H), 5.98 (s, 1 H)

[0591] Compound 1 16. To a stirred solution of 6-oxo-4-(quinolin-4-yl)-l,6-dihydropyridine-3- carboxylate (0.1 g, 0.38 mmol, 1.0 equiv) in DMF (5 mL), was added (S)-4,4-difluoro-l- glycylpyrrolidme-2-carbomtrile hydrochloride (0.085 g, 0.38 mmol, 1.0 equiv), HATU (0.286 g, 0.754 mmol, 2.0 equi v). The mixture was allowed to stir at RT for 10 min. DIPEA (0.3 mL) was added and the mixture was allowed to stir at RT for overnight. Product formation was confirmed by LCMS and TLC. After completion of reaction, the mixture was diluted with water (20 mL) and extracted with ethyl acetate (50 mL c 3). Combined organic extracts were washed with water (50 ml, c 5), dried over anhydrous Na 2 S0 4 and concentrated. The crude product obtained was purified prep purification to obtain (S)-N-(2-(2-c ano-4,4-difluoropyrrolidin- 1 -yl)-2- oxoethyl)-6-oxo-4-(quinolin-4-yl)-l,6-dihydropyridine-3-carb oxamide (0.045g, 27.12 %Yield) as an white solid.

[0592] LCMS 438.2| VI 1 1 ]

[0593] ' l l NMR (400MHz .DVISO-d,.) d 12.24 (br. s, 1 H), 8.86 (hr. s, 1 H), 8.59 (br. s, 1 H), 8.03 (d, J = 8.3 Hz, 1 H), 7.94 (br. s, 1 H), 7.74 (br. s, 1 H), 7.67 (d, J = 7.9 Hz, 1 H), 7.60 - 7.45 (m, 1 H), 7.33 (d, J= 4.4 Hz, 1 H), 6.29 (br. s, 1 H), 4.98 (br. s, 1 H), 4.05 (d, ./ 14.9 Hz,

1 H), 3.89 - 3.73 (m, 2 H), 2 83 - 2.60 (m, 3 H).

Example 67

Synthesis of (S)-N-(2-(2-cyano-4,4-difluoropyrrolidin-l-yl)-2-oxoethyl)-3 -(pyridin-3- ylaminopsonicotinamide

Compound 117a. To a solution of methyl 3-bromoisonicotinate (0.5 g, 2.315 mmol, 1.0 equiv) in Dioxane (10 mL) was added pyridin-3-amine (0.218 g, 2.315 mmol, 1.0 equiv), Cs CCL (1.5 g, 4.63 mmol, 2.0 equiv) and resulting reaction mixture purged with N 2 gas for 10 minute, followed by the addition of Pd?(dba):¾ (0.106 g, 0.116 mmol. 0.05 equiv, ), Xanthphose (0. 134 g, 0.232 mmol, 0.1 equiv) and resulting reaction mixture). The resulting reaction mixture was heated at 120°C for overnight. Product formation was confirmed by LCMS. After the completion of reaction, the mixture was filtered through celite bed, washed with ethyl acetate (100 mL). Filtrate was concentrated under reduced pressure. The crude product obtained was purified by flash chromatography (3% methanol in DCM as an eluent) to obtain methyl 3- (pyridin-3-ylamino)isonicotinate (0.170 g, 32.07 %) as dark brown liquid.

LCMS 230. i I VI 1 11

!H NMR (400MHZ, DMSO-de) d 8.88 (s, 1 H), 8 57 - 8.44 (m, 2 H), 8 29 id. J= 3.5 Hz, 1 H), 8.13 (d, J= 5.3 Hz, 1 H), 7.79 - 7.62 (m, 2 H), 7.39 - 7.29 (m, 1 H), 3.88 (s, 3 H).

[0597] Compound 117b. To a stirred solution of obtain methyl 3-(pyridin-3- ylaminojisonicotinate (0.170 g, 0.742 mmol, 1.0 equiv) in THF (4 niL) and water (4 mL), was added LIOH.H 2 0 (0.048 g, 1.114 mmol, 1.5 equiv). The mixture was allowed to stir at RT for overnight. Product formation was confirmed by LCMS. The reaction mixture was concentrated and diluted with water (20 mL) and washed with ethyl acetate (10 mL x 2). Aqueous layer was separated and freeze dried on lyophilyzer to obtain 3-(pyridin-3-ylamino)isonicotinate

(Quantative Yield) as white solid.

[0598] LCMS 216.1

[0599] 'l l NMR (400MHz .DYISO-d,.) 6 11.84 (s, 1 H), 8.55 (s, 1 H), 8.39 (d, J= 2.6 Hz, 1 H), 8.10 (d, J--- 3.9 Hz, 1 H), 7 96 (d, J= 4.8 Hz, 1 H), 7.70 (d, J= 4 8 Hz, ! H), 7.65 - 7.55 (rn, 1 H), 7.28 (dd, ./= 4 8, 8.3 Hz, 1 H). [Q6QQ] Compound 1 17. To a stirred solution of 3-(pyridin-3-ylamino)isonicotinate (0.10 g, 0.465 mmol, 1.0 equiv) m DMF (5 mL), was added (S)-4,4-difluoro- 1 -glycylpyrrolidine-2- carbonitrile hydrochloride (0.104 g, 0.465 mmol, 1.0 equiv), HATH (0.353 g, 0.93 mmol, 2.0 equiv). The mixture was allowed to stir at RT for 10 min. DIPEA (0.35 rnL) was added and the mixture was allowed to stir at RT for overnight. Product formation was confirmed by LCMS and TEC. After completion of reaction, the mixture was diluted with water (20 mL) and extracted with ethyl acetate (50 mL c 3). Combined organic extracts were washed with water (50 mL c 5), dried over anhydrous Na 2 S0 4 and concentrated. The crude product obtained was purified by flash chromatography (10 % MeOH in DCM as an eluent) followed by prep purification to obtain (S)-N-(2-(2-cyano-4,4-difluoropyrrolidin-l-yl)-2-oxoethyl)-3 -(pyridin-3- ylaminojisonicotinamide (0.072g, 40 % Yield) as an off white solid.

[0601] LCMS 387.21 VI 1 11

[0602] ' l l NMR (400MHz .DVISO-d,.) d 9.17 (br. s . 1 H), 9.09 (s, 1 H), 8.62 (s, 1 H), 8.45 (br. s., 1 H), 8.19 (br. s., 2 H), 7.64 (d, J= 7.5 Hz, 1 H), 7.58 (d, J= 4.8 Hz, 1 H), 7.38 - 7.22 (m, 1 H), 5.12 (d, J ------ 7.0 Hz, 1 H), 4.31 i d. ./ 1 1.4 Hz, 1 H), 4.18 - 3.98 (m, 2 H), 2.90 (br. s., 1 H),

2.82 (d, .7= 15 3 Hz, 2 H).

Example 68

Synthesis of (S)-N-(2-(2-cyano-4,4-difluoropyrrolidin-l-yl)-2-oxoethyl)-3 -(piperidin-4 - ylaminojisonicotinamide

[Q6Q3] Compound 118a. To a stirred solution of methyl 3-bromoisonicotinate (0.500 grn, 2.3148 mmol, 1 equiv) & tert-butyl 4-aminopiperi dine- l-carboxy late (0.463gm, 2.3l48mmol, 1 equiv) and CS 2 CO 3 (l.Sgrn, 4.6296 mmol, 2equiv)in dioxane (15 mL). The resulting mixture was purged with nitrogen for 10 min followed by addition of Pd 2 (dba)3 (0.106gm, 0.1 157 mmol, 0.05 equiv) and xantphos (0 134gm, 0.2314 mmol, 0.1 equiv), again purged with nitrogen for 10 min. The reaction mixture wns heated at 120 °C for overnight. The progress of reaction was monitored by LCMS. The reaction mixture was diluted with water (30 mL), extracted with EtOAc (2x50 mL). The combined organic layers were washed with water (30 mL), with brine (30 mL), dried over Na S0 4 , concentrated to afford the crude which w r as purified by flash chromatography to obtain methyl 3-((l-(tert-butoxycarbonyl)piperidin-4-yl)amino)isonicotinat e (400 mg, 51.49%) as a white solid.

[0604] LCMS: 336.31. VI H i

[0605] 'l l NMR: (400MHz, DMSO-d 6 ) d 8.41 (s, 1 H), 7.60 - 7.51 (m, 2 H), 7.27 (d, ./ 7.9

Hz, 1 H), 3.00 (m., 4 H), 2.04 - 1.88 (s, 3 H), 1.44 - 1.35 (s, 9 H), 1.31 - 1.12 (m, 4 H).

[0606] Compound 11 8b. To a stirred solution of compound methyl 3-(( l-(tert-butoxy carbonyl) piperidin-4-yl) amino)isonicotinate (0.400 gm, 1.1904 mmol, 1 equiv) in fi ll (8 ml.) and water (4 mL), was added LiOH (0.058 gm, 2.3809 mmol, 2 equiv). The mixture wns allowed to stir at 80°C for overnight. Product formation was confirmed by LCMS. The reaction mixture was concentrated and diluted with wnter (10 mL) and washed with ethyl acetate (10 mL c 2).

Aqueous layer was separated and freeze dried on lyophilyzer to obtain compound 3-((l-(tert- butoxy carbonyl) piperidin-4-yl) amino)isonicotinic acid (330 mg, 78% Yield) as a white solid.

[0607] LCMS: 322

[0608] 1H NMR: (400MHz, DMSO-de) d 11 .70(s, 1H), 8.41 (s, 1 H), 7.60 - 7.51 (m, 2 H),

7.27 (d, 7.9 Hz, 1 H), 3.00 (m, 4 H), 1.44 - 1.35 (s, 9 H), 1.31 - 1.12 (m, 4 H).

[0609] Compound 11 8c. To a stirred solution of 3-((l -(tert-butoxycarbonyl)piperidin-4- yl)amino)isonicotinic acid (0.200 gin, 0.6211 mmol, 1 equiv), HATU(0354 gm, 0.9316 mmol,

1.5 equiv) in DMF (10 ml) after 10 min was added a (S)-4,4-difluoro-l-glycylpyrrolidine-2- carbonitnle hydrochloride (0.168 gm, 0.7453 mmol, 1.2 equiv) and stirred the reaction mixture forl O min, followed by drop wise addition of DIPEA (0 2ml, 0.9316 mmol, 1.5 equiv) allowed the reaction for 16h stirring at rt reaction progress was monitored by LCMS and TLC, workup done by addition of water (30 ml) and extracted by ethyl acetate (3 c 50 ml.). Combined organic layer was washed with water (4 x 20 mL), dried over anhydrous Na 2 S0 4 , concentrated under reduced pressure to obtain ten-butyl (S)-4-((4-((2-(2-cyano-4,4-difluoropyrrolidin-l-yl)-2- oxoethyl)carbamoyl)pyridin-3-yl)amino)piperidine-l -carboxylate (220 mg, 71.89 % Yield) as a brown solid

[Q61Q] LCMS: 493.3[M+H] +

[0611] ! H NMR: (400 VII Iz ,DMSO-d 6 ) d 8.95 (hr. s„ 1 H), 8.28 (s, 1 1 1 ). 7 86 (d, J= 5.3 Hz,

1 H), 7.51 - 7.38 (m, 2 H i. 5.76 (s, 1 H), 4.12 - 4.02 On. 4 H), 3 81 (d, ./ 13.2 Hz, 2 H),

3.50( m, 1H), 2.05 - 1.93 (m, 4 H), 1.40 (s, 9 H), 1.28 - 1.10 (m, 4 H).

[0612] Compound 118. To a stirred solution of tert-butyl (S)-4-((4-((2-(2-cyano-4,4- difluoropyrrolidin-l-yl)-2-oxoethyl)carbamoyl)pyridin-3-yl)a mino)piperidine-l -carboxylate (0.190 gm, 0.3861 mmol, 1 equiv) in DCM (5 mL), was added trifloroacetic acid (1.5 mL). The mixture was allowed to stir at RT overnight. Product formation was confirmed by LCMS. The reaction mixture concentrated and the crude was purified by reversed phase HPLC to obtain (S)- N-(2-(2-cyano-4,4-difluoropyrroli din-1 -yl)-2-oxoethyl)-3-(piperidin-4-ylamino)isonicotinamide ( 60 mg, 39.73% Yield) as a white solid.

[0613] LCMS: 393.3[M+H] +

[0614] ' l l NMR: (400MHz ,DMSG-d 6 ) d = 8.95 (br. s, 1 H), 8.28 (s, 1 H), 7.86 (d, J = 5.3 Hz, 1 H), 7.51 - 7.38 (m, 2 H i. 5.76 (s, 1 H), 4.12 - 4.02 (m, 4 H), 3 81 (d, ./ 13.2 Hz, 2 H),

3.50( m, I I ! }. 2.05 - 1.93 (m, 4 H), 1.28 - 1.10 (m, 4 H).

Example 69

Synthesis of (S)-N-(2-(2-cyano-4,4-difluoropyrrolidin-l-yl)-2-oxoethyl)-3 -(quinolin-4- ylaminojisonicofinamide

[0615] Compound 1 19a. To a solution of methyl 3-bromoisonicotinate (0 5 g, 2.315 mmol, 1.0 equiv) in Dioxane (10 mL) was added pyridin-3-amine (0.334 g,2.315 mmol, 1.0 equiv), CS 2 CO 3 (1.5 g, 4.63 mmol, 2.0 equiv) and resulting reaction mixture purged with N gas for 10 minute, followed by the addition of Pd 2 (dba) 3 (0.106 g, 0.116 mmol. 0 05 equiv, ), Xanthphose (0.134 g, 0.232 mmol, 0.1 equiv) and resulting reaction mixture). The resulting reaction mixture was heated at 120°C for overnight. Product formation was confirmed by LCMS. After the completion of reaction, the mixture diluted with water (30 mL) washed with ethyl acetate (50 mL). Aqueous layer was separated and freeze dried on lyophilyzer to obtain 3-(quinolin-4- ylamino)isonicotinic acid (Quant. Yield) as a white solid.

[0616] LCMS 266. 1 [ VI ! 1 ]

[0617] 'l l NMR (400MHz .DYISO-d,.) 6 13.66 (s, 1 1 11 8.93 is. 1 IT), 8.58 (d, J= 5.3 Hz, 1 H), 8.28 - 8.18 (m, 1 H), 8.14 (d, ./= 4.8 Hz, 1 H), 7.91 (d, J= 8.8 Hz, 1 H), 7.79 (d, J= 4 8 Hz,

1 H), 7.72 (t, ./ = 7.0 Hz, 1 H), 7.58 (t, ./= 7 2 Hz, 1 H), 7.45 - 7.33 (m, 1 H), 6.76 (br s., 1 H).

[0618] Compound 119. To a stirred solution of 3-(quinolin-4-ylamino)isonicotinic acid (0.10 g, 0.377 mmol, 1.0 equiv) in DMF (5 mL), was added (S)-4,4-difluoro-l-glycylpyrrolidine-2- carbonitrile hydrochloride (0.085 g, 0.377 mmol, 1.0 equiv), HATU (0.286 g, 0.754 mmol, 2.0 equiv). The mixture was allowed to stir at RT for 10 min. DIPEA (0.2 mL) was added and the mixture was allowed to stir at RT for overnight. Product formation was confirmed by LCMS and TLC. After completion of reaction, the mixture was diluted with water (20 mL) and extracted with ethyl acetate (50 mL x 3). Combined organic extracts were washed with water (50 ml x 5), dried over anhydrous Na 2 S0 4 and concentrated. The crude product obtained was purified prep purification to obtain (S)-N-(2-(2-cyano-4,4-difluoropyrrolidin-l-yJ)-2-oxoethyl)-3 -(quinolin-4- ylamino)isonicotinamide (0.004 g, 2,4 % Yield) as an off white solid.

[0619] LCMS 437.2[M+H] +

[0620] 'H NMR (400 MHz, DYISO-d, .) 6 8 76 (br. s., 1H), 8.48 (s, 2H), 7.79 (d, .7 = 8.33 Hz,

21 i ). 7.39 (br.s., 2H), 6.86 (d, J = 8.33 Hz, I I I ). 5.10 (d, J ------ 7.45 Hz, 11 1 ). 4.23 (hr. s., ! ! ! ). 3.98

- 4.12 (m, 21 !}. 3.94 (s, 31 1 }. 2.90 (d, J --- 8.33 Hz, i l l ). 2 70 - 2.86 (m, 11 1}. 2,67 (br. s., I l l }

Example 70

Synthesis of (S)-3-(4-aminopiperidin-l-yl)-N-(2-(2-cyano-4,4-difluoropyrr olidin-l-yl)-2- oxoethyljisonicotinamide 2.2, 2-trifluoroacetate

[0621] Compound 120a. To a stirred solution of methyl 3-bromoisonicotinate (0.200 gm,

0.925 mmol, 1.0 equiv) & tert-butyl piperidin-4-ylcarbamate (0.185gm, 0.925 mmol, 1.0 equiv) and CS .( () : (0.601 gm, 1.85 mmol, 2.0 equiv) in Dioxan (5 mL). The resulting mixture was purged with nitrogen for 10 min followed by addition of Pd 2 (dba) 3 (0.042 gm, 0.046 mmol, 0.05 equiv) and xantphos (0.053 gm, 0.092 mmol, 0.1 equiv), again purged with nitrogen for 10 mm. The reaction mixture was heated at 120 °C for overnight. The progress of reaction was monitored by LCMS. The reaction mixture was diluted with water (30 mL), exlraeted with EtOAc (2 x 50 mL). The combined organic layers were washed with water (30 mL), with brine (30 mL), dried over Na 2 S0 4 , concentrated to afford the crude which was purified by column chromatography to obtain methyl 3-(4-((tert-butoxycarbony])amino)piperidin-l-yl)isonicotinat e (0.120 g, 38 % Yield) as an yellow semi solid.

[0622] LCMS: 336.5[M+H] +'

[0623] Compound 120b. To a stirred solution of compound methyl 3-(4-((tert- butoxycarbonyl)amino)piperidin-l-yl)isonicotinate (0.100 gin, 0.298 mmol, 1.0 equiv) m THF (3 mL) and water (2 mL), was added LiOH (0.014 gm, 0.597 mmol, 2.0 equiv). The mixture was allowed to stir at RT for overnight. Product formation was confirmed by LCMS. The reaction mixture wns concentrated and diluted with wnter (10 mL) and washed with ethyl acetate (10 L x 2). Aqueous layer was separated and freeze dried on lyophilyzer to obtain 3-(4-((tert~ butoxycarbonyl)amino)piperidin-l-yl)isonicotinic acid, lithium salt ( 0.120 g, Quant. Yield) as a white solid.

[0624] LCMS: 322.21 VI H i +

[0625] Compound 120c. To a stirred solution of 3-(4-((tert-butoxycarbonyl)amino)piperidin-l- yl)isonicotinic acid, lithium salt (0.070 gm, 0.213 mmol, 1.0 equiv), HATH (0.162 gm, 0.426 mmol, 2.0 equiv) in DMF (3 ml) after 10 min was added a (S)-4,4-difluoro-l -glycylpyrrolidine- 2-carbonitrile hydrochloride (0.048 gm, 0.213 mmol, 1.0 equiv) and stirred the reaction mixture for 10 min, followed by drop wise addition of DIPEA (0.082 g, 0.640 mmol, 3.0 equiv) allowed the reaction for 16 h stirring at RT. Reaction progress was monitored by LCMS and TLC, workup done by addition of water (30 ml) and extracted with ethyl acetate (2 c 50 ml).

Combined organic layer was washed with water (4 c 20 mL), dried over anhydrous Na 2 S0 4 , concentrated under reduced pressure to obtain (S)-(l-(4-((2-(2-cyano-4,4-difluoropyrrolidin-l- yl)-2-oxoethyl)carbamoyl)pyridin-3-yl)piperidin-4-yl)carbama te (0.1 10 g, ) as a crude product which was directly used for next step.

[0626] LCMS: 493.3[M+H] +

[0627] Compound 120-TFA. To a stirred solution of (S)-(l-(4-((2-(2-cyano-4,4- difluoropyrrolidin-l -y])-2-oxoethyl)carbamoyl)pyridin-3-yl)piperidin-4-yl)carbam ate (0.1 10 gm, 0.223 mmol, 1.0 equiv) in DCM (3 mL), was added trifloroacetic acid (1.0 mL). The mixture was allowed to stir at RT overnight. Product formation was confirmed by LCMS. The reaction mixture concentrated and the crude was purified by reverse phase HPLC to obtain (S)-3-(4- aminopiperidin~l -yl)-N-(2-(2-cyano-4,4-difluoropyrro!idin-l~yl)-2-oxoethyl)i sonicotinamide 2,2,2-trifluoroacetate ( 0.015 g ) as an off-white solid.

[0628] LCMS: 393.3[M+H] +

[0629] 'l l NMR (4QQMHz ,DMSO-d 6 ) 59.25 (d, ./ 5.7 Hz, 1 H), 8.50 (hr. s., 1 H), 8.34 id. J

= 4.8 Hz, 1 H), 7 93 (br. s , 2 H), 7.49 (d, J= 4.8 Hz, 1 H), 7.11 (s, 1 H), 5 10 (d, J = 6.6 Hz, 1 H), 4.22 - 4.11 (m, 2 H), 3.51 - 3.35 (m, 3 H), 3.19 (br. s., 1 H), 3 04 - 2.75 (m, 4 H), 1.97 (d. J = 11.8 Hz, 2 H), 1.71 id../ 11.4 Hz, 2 H), 1.55 (br. s., 1 H), 0.94 - 0.77 (m, 2 H)

Example 71

Synthesis of (S)-4-benzyl-N-(2-(2-cyano-4, 4-difluoropyrrolidin-l-yl)-2-oxoeihyl)-6-oxo-l, 6- dihydropyridine-3-carboxamide

[0630] Compound 121a. To a stirring solution of methyl 4-chloro-6-oxo-l,6-dihydropyridine- 3-carboxyiate (0.250 g, 1.34 mmol, 1.0 equiv) m Dioxan (4 ml) and water(4 nil) were added Na 2 C(>3 (0.284 g, 2.68 mmol, 2.0 equiv), 2-benzy 4-4,4,5,5-tetramethyl-l,3,2-dioxaboiOlane (0.584 g, 2.68 mmol, 2.0 equiv) and Pd(dppi)Ci2.DCM complex (0.058 g, 0.07 mmol, 0.05 equiv). The reaction mixture was allowed to heat at 150°C for 70 min in microwave. Product formation was confirmed by TLC and LCMS. The reaction mixture was diluted with water (15 mL) and extracted with ethyl acetate (20 mL X 3). Combined organic layer was washed with brine (30 mL). Organic layer was dried over anhydrous sodium sulphate, filtered and concentrated under reduced pressure. Purified the compound by normal phase combi-flash chromatography to obtain methyl 4-benzyl-0-oxo-l,0-dihydropyridme-3-carboxylate (0.030 g).

[0631] LCMS 244.0

[0632] Compound 121b. To a stirred solution of methyl 4-benzyl~6~oxo-l ,6~dihydropyridine~ 3-carboxylate (0.300 g, 1 23 mmol, 1 equiv) in THF and water (1 : i)(6 ml), was added LIOH.H 2 0 (0.104 g, 2.47 mmol, 2.0 equiv). The mixture vras allowed to stir at 60°C for 16 h. Product formation was confirmed by LCMS. The reaction mixture was diluted with water (25mL), extracted with ethyl acetate (20 mL) and concentrated the aqueous layer under reduced pressure and lyophilized to obtain 4-benzyl-6-oxo-l,6-dihydropyridine~3-carboxylic acid (0 260 o)

[0633] LCMS 230.0 j M ! ! | + [Q634] Compound 121. To a stirring solution of 4-benzyl-6-oxo-l ,6-dihydropyridine-3- carboxylic acid (0.500 g, 2.18 mmol, 1.0 equiv) in DMF (17 ml) were added (S)-4,4-difluoro-l - glycylpyrrolidine-2-carbonitrile hydrochloride (0.590 g, 2.62 mmol, 1.2 equiv), EDC.HC1 (0.503 g, 2 62 mmol, 1.2 equiv), HOBT (0.354 g, 2.62 mmol, 1.2 equiv), DMAP (0.001 g) and stirred the reaction mixture at RT for 10 min followed by addition of TEA (0.661 g, 6.54 mmol, 3.0 equiv). The reaction mixture was allowed to stir at RT for 16 h. Product formation was confirmed by LCMS. The reaction mixture was diluted with water (35 mL) and extracted adi ethyl acetate (60 mL X 3). Combined organic layer was washed with water (40 mL X 6) and brine (60 mL). Organic layer was dried over anhydrous sodium sulphate, filtered and concentrated under reduced pressure. Purified the compound by reverse phase HPLC to obtain pure (S)-4-benzyl-N-(2-(2-cyano-4,4-difluoropyrrolidin-I -yl)-2-oxoethyl)-6-oxo-l,6- dihydropyridine-3-carboxamide (0.070 g 8.5%) as an off-white solid.

[0635] LCMS 401.1 [M+H] +

[0636] 1H NMR (400 MHz, DMSO-de) d 11.74 (s, 1H), 8.55 (t, J = 5.9 Hz, 1H), 7.58 (s, 1H), 7.33 - 7.15 (m, 5H), 6.05 (s, 1H), 5.09 (dd, J = 9.1, 2.8 Hz, 1 1 1 ). 4.26 (dd, J= 14.5, 9.9 Hz, i l l :·. 4.16 - 3.95 (m, 5H), 2 98 - 2.72 (m, 2H).

Example 72

Synthesis of ( S)-N-(2-(2-cyano-4,4-difluoropyrrolidin-l-yl)-2-oxoethyl)-3- (l - phenylvinyljisonicotinamide

[0637] Compound 122a. To a solution of methyl 3-bromoisonicotinate (1.0 g, 4.6 mmol, 1.0 equiv) in toluene (10 mL) was added 4,4,5,5-tetramethyl-2-(l-phenylvinyl)-l,3,2-dioxaborolane (1.06 g, 4.6 mmol, 1.0 equiv), K3O4P (1.96 g, 9.2 mmol, 2.0 equiv) followed by the addition of Palladium(li) acetate (0.104 g, 0.46 mmol. 0.1 equiv), and Tricyclohexylphosphine (0.130 g. 0.46 mmol. 0.1 equiv) The resulting reaction mixture was heated at 100° C for overnight.

Product formation was confirmed by LCMS. After completion of reaction, the mixture was diluted with water (200 mL) and extracted with ethyl acetate (150 mL c 2). Combined organic extracts were washed with water (20 mL c 2), dried over anhydrous Na 2 S0 4 and concentrated. The crude product obtained was purified by flash chromatography (0-20 % ethyl acetate in hexane as an eluent) to obtain methyl 3-(l-phenylvinyl)isonicotinate (0.560 g, 50 % Yield) as brown semi solid.

[0638] LCMS 240[M+H] h

[0639] Compound 122b. To a stirred solution of methyl 3-(l-phenylviny])isonicotinate (0.100 g, 0.41 mmol, 1.0 equiv) m THF (5 mL) and water (5 mL), w-as added LiOH (0.020 g, 0.82 mmol, 2.0 equiv). The mixture was allowed to stir at RT for overnight. Product formation was confirmed by LCMS and 1H NMR Spectroscopy. The reaction mixture was diluted with water (15 mL) and washed with ethyl acetate (15 mL). Aqueous layer was separated and freeze dried on lyophilyzer to obtain 3-(l-phenylvinyl)isonicotinic acid (0.140 g, Qaunt. Yield ) as a brown solid.

[0640] LCMS 225.91 YL I ! |

[0641] Compound 122. To a stirred solution of 3-(!-phenylvinyl)isonicotinic acid (0.140 g, 0.44 mmol, 1.0 equiv) in DMF (5 mL), wns added (S)-4,4-difluoro-l-glycylpyrrolidine-2- carbonitrile hydrochloride (0.100 g, 0.44 mmol, 1.0 equiv), HOBt (0.072 g, 0.53 mmol, 1.2 equiv) and EDC.HC1 (0.102 g, 0.53 mmol, 1 2 equiv). The mixture was allowed to stir at RT for 10 min. Triethyl amine (0.2 mL) was added and the mixture was allowed to stir at RT for overnight. Product formation w¾s confirmed by LCMS. After completion of reaction, the mixture was diluted with water (50 mL) and extracted w th ethyl acetate (50 ml. c 2). Combined organic extracts were washed with water (20 mL c 4), dried over anhydrous Na 2 S0 4 and concentrated. The crude product was purified by reversed phase HPLC to obtain (S)-N-(2-(2- cyano-4,4-difluoropyrrolidin-l-yi)-2-oxoethyl)-3-(l-phemTvin yl)isonicotinamide (0.010 g, 07 % Yield) as a white solid.

FI), 4. 16 (br. s„ 1 ! ! }. 3.97 (d, J= 9.2 Hz, 1 H i. 3.86 (br. s , 2 ! ! ). 2 78 (d, J = 10.5 Hz, 2 ! ! }. 2.67 (br. s. , 1 H).

Example 73

Synthesis ofN-(2-((S)-2-cyano-4,4-difluoropyrrolidin-l-yl)-2-oxoethyl) -3-(l- phenylethyl)isonicotinamide

[0644] Compound 123 a. To a stirred solution of methyl 3-(l -phenylvinyl)isonicotinate (0.200 g, 0.83 mmol, 1.0 equiv) inTHF: Methanol (10 : 04 mL) was added TEA (0.425 g, 4.16 mmol,

5.0 equiv) under nitrogen and Palladium on Carbon[Pd/C](0.045 g, 0.41 mmol, 0.5 equiv) was added. Purge the reaction mixture with H 2 gas for lh. Product formation was confirmed by LCMS. After the completion of reaction, reaction mixture was filtered through Celite bed & Filtrate was concentrated under reduced pressure to obtain methyl 3-(l-phenylethyi)isonicotinate (0.180 g, 89 % Yield) as a brown semisolid.

[0645] LCMS 241.28 1 M l l j

[0646] Compound 123b. To a stirred solution of methyl 3-(1 -phenylethyllisonicotinate (0.180 g, 0.74 mmol, 1.0 equiv) in THF (15 mL) and water (10 mL), was added LiOH (0.036 g, 1.48 mmol, 2.0 equiv). The mixture was allowed to stir at RT for overnight. Product formation was confirmed by LCMS and ! H NMR Spectroscopy. The reaction mixture was diluted with water (15 mL) and washed with ethyl acetate (15 mL) Aqueous layer was separated and freeze dried on !yophiiyzer to obtain 3-(! -phenylethy!)isonicotinic acid (0 210 g, Quant Yield ) as a brown solid.

LCMS 227.9] M · 1 1 [

[0648] Compound 123. To a stirred solution of 3-(l-phenylethyl)isonicotinic acid (0.100 g, 0.43 mmol, 1.0 equiv) in DMF (5 rnL), was added DIPEA (0.3 ml , 1.29 mmol, 3.0 equiv) and HATU (0.327 g, 0.86 mmol, 2.0 equiv). Allow to stir the mixture for 30 min. under nitrogen Atm (S)-4,4-difluoro-l -glycy]pyrrolidine-2-carbonitrile hydrochloride (0.099 g, 0.43 mmol, 1.0 equiv) was added to above mixture and allowed to stir for overnight at RT. Progress of the reaction was monitored by TLC and LCMS. After completion of the reaction, the reaction mixture was diluted with water (30 mL) and extracted with ethyl acetate (100 mL c 2). Organic layer was washed with water (100 mL), brine solution (100 mL), dried over anhydrous sodium sulphate and concentrated under reduced pressure to obtain crude compound, which was purified by reversed phase HPLC to obtain N-(2-((S)-2-cyano-4,4-difluoropyrrolidin-l-yl)-2-oxoethyl)- 3-(l -phenylethyl)isonicotinamide (Free base) (0.005 g, 03% Yield) as a white solid

[0649] LCMS 399[M+H] +

[0650] ! H NMR (400MHZ .D SO-d, ) d 8.97 (d, J = 8 8 Hz, 1 H), 8.56 - 8.38 (m, 2 H), 7.46 - 7.33 (m, 2 H), 7.29 (t, ./ 7.2 Hz, 3 H), 7.18 (t, J = 7.0 Hz, 1 H), 5.14 (d, ./ 9.6 Hz, 1 H), 4.69 -

4 59 (m, 1 H), 4.29 (br. s . 1 H), 4.22 - 3.99 (m, 3 H), 2.82 id. ./ 18.4 Hz, 2 H), 1.63 (t, .7 = 7.0

Hz, 3 H).

Example 74

Synthesis ofN~(2~((S)-2-cyano-4,4~difluoropyrrolidm-l-yl)-2-oxoethyl)- 3-((R)-l- phenylethyl)isonicotinamide and N-(2-((S)-2-cyano-4,4-difluoropyrroiidin-]-yl)-2-oxoethyl) 3

((S)-l-phenylethyl)isonicotinamide

second eiuting isomer [0651] Chiral resolution method: The enantiomers were separated by chiral SFC (Chiralpak- IC, 250X20 mm, 5m). Isocratic Program with analytical grade liquid carbon dioxide with HPLC grade 0.2% DBA in Hexane and Isopropyl alcohol.

[0652] LCMS 399.3 j M 1 11

[0653] 'l l NMR (4QQMHz ,DMSO-d 6 ) 6 8 97 (d, J = 8.8 Hz, 1 H), 8.56 - 8.38 (m, 2 ! !}. 7.46 - 7 33 (m, 2 H), 7.29 (t, J 7.2 Hz, 3 H), 7.18 (t, J= 7.0 Hz, 1 H), 5.14 (d, J 9.6 Hz, 1 H), 4.69 - 4.59 (m, 1 H), 4.29 (br. s., 1 H), 4.22 - 3.99 (m, 3 H), 2.82 (d, J= 18.4 Hz, 2 H), 1.63 (t, J= 7.0 Hz, 3 FI).

Example 75

Synthesis of (S,E)-N-(2-(2-cyano-4, 4-difluoropyrrolidin-l-yl)-2-oxoethyl)-3- styrylisonicotinamide

124a 124b Compound 124

[0654] Compound 124a. To a solution of ethyl 3-bromoisomcotinate (0.2 g, 0.925 mmol, 1.0 equiv) in dioxane (10 mL) and water (1 mL) was added (E)-4,4,5,5-tetramethyl-2-styiyl-I ,3,2- dioxaborolane (0.319 g, 1.39 mmol, 1.5 equiv), K CO ; (0.26 g, 1.852 mmol, 2.0 equiv) and resulting reaction mixture purged with N 2 gas for 10 minute, followed by the addition of

Pd(PPh3)Cl 2 (0.033 g, 0.046 mmol. 0 05 equiv). The resulting reaction mixture was heated at 120° C for overnight. Product formation was confirmed by LCMS. After the completion of reaction, the mixture was filtered through celite bed, washed with ethyl acetate (100 mL).

Filtrate was concentrated under reduced pressure. The crude product obtained was purified by flash chromatography (0-30 % ethyl acetate in hexane as an eluent) to obatined methyl (E)~3~ styrylisonicotinate (0.150 g, 67.87 % yield) as a pale yellow solid.

[0655] LCMS 240.1 [M+H] + [0656] ! H NMR (DMSO-de, 400MHz) d 9.12 (s, 1 H), 8 63 (d. 7 5.4 Hz, 1 H). 7 65 - 111 (m, 2 H), 7.61 (d, J= 7.3 Hz, 2 H), 7.38 - 7 49 (m, 2 H), 7.26 - 7 38 (m, 2 H), 3.83 - 3.96 ppm (m, 3 H).

[0657] Compound 124b. To a stirred solution of methyl (E)-3-styrylisonicotinate (0.1 g, 0.418 mmol, 1.0 equiv) in THF (2 mL) and water (2 mL), was added LiOH.H 2 0 (0.021 g, 0.0.502 mmol, 1.2 equiv). The mixture was allowed to stir at RT for overnight. Product formation was confirmed by LCMS and ! H NMR Spectroscopy. The reaction mixture was concentrated and diluted with water (10 mL) and washed with ethyl acetate (10 mL x 2) Aqueous layer was separated and freeze dried on lyophilyzer to obtain (E)-3~styrylisonicotinic acid (Quant. Yield) as off white solid.

[0658] LCMS 226.2| M i i |

[0659] 'l l NMR (DMSO-d 6 , 4! Hi VI I lz) 6 8.83 (s, 1 H), 8.30 (d, .7=4.9 Hz, 1 H), 7.86 (d, 7 1 7. 1 Hz, 1 H), 7.52 (d, J=7.3 Hz, 2 H), 7 38 (t, .7=7.6 Hz, 2 H), 7.20 - 7.29 (m, 2 H), 7.18 pp (s, 1 H).

[0660] Compound 124. To a stirred solution of (E)-3-styrylisonicotinic acid (0.06 g, 0.266 mmol, 1.0 equiv) in DMF (2 mL), was added (S)-4,4-difluoro-l-glycylpyrrolidine-2-carbonitrile hydrochloride (0.06 g, 0.266 mmol, 1.0 equiv), EDCI.HC1 (0.077 g, 0.39 mmol, 1.5 equiv) & HOBt (0.054 g, 0.39 mmol, 1.5 equiv). Hie mixture was allowed to stir at RT for 10 min.

Tnethylamine (0.2 mL) ivas added and the mixture was allowed to stir at RT for overnight. Product formation was confirmed by LCMS and TLC. After completion of reaction, the mixture was diluted with water (20 mL) and extracted with ethyl acetate (20 mL c 2). Combined organic extracts were washed with water (20 mL >< 4), dried over anhydrous Na 2 S0 4 and concentrated. The crude product obtained was purified by flash chromatography (5 % Methanol in DCM as an eluent) to obatine (S,E)-N~(2-(2-ey ano-4,4~difluoropyrrolidin- 1 -yl)-2-oxoethyl)-3- styrylisonicotinamide (0.046 g, 43.8 % Yield) as an off wliite solid.

[0661] LCMS 397.2| Vl ! ! j

[0662] 'l l NMR { DViSO-d,. ,400MHz) 6 9.14 (s, 1 H), 8.99 (t, 7=5.9 Hz, 1 H), 8.53 (d, 7=4.4 Hz, 1 H), 7.69 (d, 7=7 3 Hz, 2 H), 7 63 (s, 1 H), 7.47 (d, 7=16 1 FIz, 1 H), 7.33 - 7.44 (m, 3 H), 7.24 - 7.33 (m, 1 H), 5.10 - 5.24 (m, 1 H), 4.32 (hr. s., 1 H), 4.05 - 4.23 (m, 3 H), 2.94 (br. s., 1 H), 2.80 - 2.91 ppm (m, 1 H). Example 76

Synthesis of ( S)-N-(2-(2-cyano-4,4-difluoropyrrolidin-l-yl)-2-oxoethyl)-3 - phenethylisonicotinamide

[Q663] Compound 125a. To a solution of ethyl 3-bromoisonicotinate (0.2 g, 0.925 mmol, 1.0 equiv) in dioxane (10 mL) and water (1 mL) was added (E)-4,4,5,5-tetramethyl-2-styryl-l,3,2- dioxaborolane (0.319 g, 1.39 mmol, 1.5 equiv), K2CO 3 (0.26 g, 1.852 mmol, 2.0 equiv) and resulting reaction mixture purged with N 2 gas for 10 minute, followed by the addition of Pd(PPli 3 )Cl2 (0.033 g, 0.046 mmol. 0.05 equiv). The resulting reaction mixture was heated at 120° C for overnight. Product formation was confirmed by LCMS. After the completion of reaction, the mixture was filtered through celite bed, washed with ethyl acetate (100 mL).

Filtrate was concentrated under reduced pressure. The crude product obtained was purified by flash chromatography (0-30 % ethyl acetate in hexane as an eluent) to methyl (E)-3- styrylisomcotinate (0.150 g, 67 87 % yield) as pale yellow solid

[0664] LCMS 240.1 [M+H] +

[0665] ! H NMR (DMSO-de, 400MHz) 5 9.12 (s, 1 H), 8 63 (d, =5.4 Hz, 1 H), 7.65 - 7.77 (m, 2 H), 7.61 (d, J 7.3 Hz, 2 H), 7.38 - 7.49 (m, 2 H), 7.26 - 7.38 (m, 2 H), 3.83 - 3.96 ppm (m,

3 H).

[0666] Compound 125b. To a solution of methyl (E)-3-styrylisonicotinate (0.13 g, 0 544 mmol, 1.0 equiv) in methanol (9 mL) was purged with N 2 gas for 10 minute, followed by the addition of Pd/C (0.065 g). Then resulting reaction mixture was purged with TI 2 gas for 6 h. Product formation was confirmed by LCMS and NMR. After the completion of reaction, the mixture was filtered through celite bed, washed with methanol (30 mL). Filtrate was concentrated under reduced pressure to obtame methyl 3-phenethy!isonicotinate (0.1 g, 76.33 % yield) as a transparent oil.

[0667] LCMS 242.2| M 1 11

[0668] ' l l NMR ( DV!SO-d... 400MHz) d 8 57 (s, 2 1 1 ). 7.27 (d, ,7=6.8 Hz, 1 H), 7.19 (d, ./ 6.8 Hz, 2 H), 3.88 (s, 3 H), 3.15 (d, 7=8.8 Hz, 2 H), 2.83 ppm (br. s., 2 H).

[0669] Compound 125c. To a stirred solution of methyl 3-phenethylisonicotinate (0.1 g, 0.415 mmol, 1.0 equiv) in THF (4 mL) and water (4 L), was added LiOH.lLO (0.021 g, 0.5 mmol, and 1.2 equiv). The mixture was allowed to stir at RT for overnight. Product formation was confirmed by LCMS and H NMR Spectroscopy. The reaction mixture was concentrated and diluted with water (10 mL) and washed with ethyl acetate (10 mL x 2). Aqueous layer was separated and freeze dried on lyophilyzer to obtain 3-phenethylisonicotinic acid (Quant. Yield) as off white solid.

[0670] LCMS 228.2|M+H]

[0671] -de, 400MHz) d 8.22 (d, ,7=4.9 Hz, 1 H), 7.18 - 7.33 (m, 5 H), 7.08 - 7.18 (m, 1 H), 2,96 - 3.11 (m, 2 H), 2,74 - 2,87 pprn (rn, 2 H).

[0672] Compound 125. To a stirred solution of (E)-3~styrylisonicotinic acid (0.05 g, 0.22 mmol, 1.0 equiv) in DMF (3 mL), was added (S)-4,4-difluoro-l-glycylpyrrolidine-2-carbonitrile hydrochloride (0.05 g, 0.22 mmol, 1.0 equiv), EDCLHCl (0.063 g, 0.33 mmol, 1.5 equiv) & HOBt (0.046 g, 0.33 mmol, 1.5 equiv). The mixture was allowed to stir at RT for 10 min Triethylamine (0.2 mL) was added and the mixture was allowed to stir at RT for overnight. Product formation was confirmed by LCMS and TLC. After completion of reaction, the mixture was diluted with water (20 mL) and extracted with ethyl acetate (20 mL c 2) Combined organic extracts were wnshed with w¾ter (20 mL c 4), dried over anhydrous Na 2 S0 4 and concentrated. Hie crude product obtained w¾s purified by flash chromatography (5 % Methanol in DCM as an eluent) to obtain (S)-N-(2-(2-cyano-4,4-difluoropyrrolidin-l-yl)-2-oxoethyl)-3 - phenethylisonicotinamide (0.050 g, 57.12 % Yield) as a white solid.

[0673] LCMS 399.2[M+H] + [0674] ! H NMR (DMSO-de ,400MHz) d 8.92 (br. s„ 1 H), 8.40 - 8.53 (m, 2 H), 7.34 (d, .7=4.4 Hz, 1 H), 7.21 - 7 29 (m, 4 H), 7.17 (d, .7=6.4 Hz, 1 H), 5.12 (d, .7=7 3 Hz, 1 H), 4.31 (br s, 1 H), 4.02 - 4.22 (m, 3 H), 2.96 - 3.08 (m, 2 H), 2.77 - 2.95 ppm (m, 4 H).

Example 77

Synthesis of N-( l-( (S)-2-cyano-4, 4-difluoropyrrolidin-l-yl)-l-oxopropan-2-yl)-3- vhenylisonicotinamide

[0675] Compound 126a. To a stirred solution of alanine (2,0 g, 22.2 mmol, 1.0 equiv.) in ACN (20 mL) was added Di-tert-butyl dicarbonate (5.35 g, 24.4 mmol, 1.1 equiv) and stirred for 10 min. 0.5M NaOH solution (20 mL) was added and the reaction mixture was allowed to stir for overnight at RT. reaction progress was monitored by NMR . The reaction was acidify with diluted HCI (PH=2 5) and extracted with ethyl acetate (100 mL), and dried over anhydrous sodium sulphate and concentrated under reduced pressure to obtain (tert- butoxycarbonyl)alanine. (2.100 g, a white solid).

[0676] Compound 126b. To a stirred solution of (tert-butoxycarbonyl)alanine (0.623 g, 3 2 mmol, 1.1 equiv.) and HATU(2.20 g, 5.8 mmol, 2 0 equiv.) in DMF (05 mi was added (S)-4,4- difluoropyrrolidine-2-carbonitrile hydrochloride (0.500 g, 2.9 mmol, 1.0 equiv) and stirred forlO mm. DIPEA (1.5 mL, 8.7 mmol, 3.0 equiv.) was added and the reaction mixture was allowed to stir for overnight at RT. reaction progress was monitored by NMR and TLC The reaction was diluted with cold water (200 mi.) and extracted with ethyl acetate (200 mL c 2). Combined organic extracts were washed with water (200 mL * 3), dried over anhydrous sodium sulphate and concentrated under reduced pressure to obtain tert-butyl (1 -((S)-2-cyano-4,4- difluoropyrrohdin-l-y!)-l-oxopropan-2-yl)carbamate (0.700 g, as a brownish solid).

[0677] Compound 126c. To a stirred solution of tert-butyl (l-((S)-2-cyano-4,4- diiluoropyrrolidin-l-yl)-l-oxopropan-2-yl)carbamate (0.700 g, 2,31 mmol, l.Qequiv) in acetonitrile (10 ml. ) was added dropwise at 0° C. 4.0 M HC1 in Dioxan (5.0ml) over a period of 10 min. the mixture was allowed to stir at RT for overnight. The reaction progress was monitored by NMR. The solvent was evaporated under reduced pressure to obtain residue which was washed with 20 mL ethyl acetate and hexane (1 : 1) to obtain (2S)-l -alanyl-4,4- difluoropyrrolidine-2-carbonitrile hydrochloride (0.900 g ) as an yellow semi solid

[0678] Compound 126. To a stirred solution of 3-phenyhsonicotinic acid (0.753 g, 3.7 mmol, 1.0 equiv.) in DMF (5 mL) w¾s added (2S)-l-alanyl-4,4-difluoropyrrolidine-2-carbonitrile hydrochloride (0.900 g, 3.7 mmol, 1.0 equiv.) HOBT (0.594 g, 4 4 mmol, 1.2 equiv.) EDC.HC1 (0.848 g, 4.4 mmol, 1.2 equiv.) and stirred forlO min. TEA (1.0 mL, 7.4 mmol, 2.0 equiv.) w as added and the mixture w¾s allowed to stir at RT for 16h. The reaction progress w¾s monitored by NMR and TLC. The reaction mixture was diluted with cold water (50 mL) and extracted with ethyl acetate (150 x 2 mL). Combined organic extracts were washed with water by (50 mL x 4), dried over anhydrous sodium sulphate and concentrated under reduced pressure. The crude product obtained w¾s purified by reversed phase chromatography to obtain N-(l-((S)-2-cyano- 4,4-difiuoropyrrolidin-l-yl)-l-oxopropan-2-yl)-3-phenylisoni cotinamide (35 mg, 03 % Yield) as an off-white solid

[0679] LCMS 385 [M+H] +

[0680] l H NMR (400 MHz, DMSG -d 6 ) d 9.06 (d, J=7.45 Hz, 1 H) 8.66 (br. s., 2 H) 7.32 - 7.52 (m, 5 H) 5.05 (d, J= 7.02 Hz, 1 H) 4.56 (br s., 1 H) 4.07 (d, J 9.2 ! Hz, 1 H) 4.01 (br. s., 1 H) 2.82 (br. s., 2 H) 1.09 - 1.20 (m, 3 H)

Biological Examples

Example B1

Inhibition ofFAPa by test compounds was assessed by in vitro enzymatic activity assays

[0681] FAPa enzymatic exopeptidase (dipeptidase) activity assay. To assay baseline FAPa enzymatic exopeptidase activity, 40 ng of recombinant human FAPa (rhFAPa, R&S system, #3715-SE) or 40 ng of recombinant mouse FAPa (rrnFAPa, R&S system, #8647-SE) w¾s incubated with 100 mM of Z-Gly-Pro-AMC peptide (BACHEM, #L-I 145) in a FAPa assay buffer (50 mM Tris pH 7.4, 100 mM NaCl, 0.1 mg/ml bovine serum albumin) for 1 h at 37 °C protected from light in 96-well black plates (Nunc, #237108). To assay FAPa enzymatic exopeptidase activity inhibition by test compounds, all test compounds were pre-incubated with the enzyme for 15 min at 37 °C before starting the reaction by substrate addition in 96-well black plates (Nunc, #237108). 7-Amino-4-Methylcoumarin (AMC) release was detected by measuring fluorescence at Ex/Em 380/460 n using a Multifunction Microplate Reader (Synergy 4, Biotek). All measurements were carried out in duplicate. Val-boroPro, a non- specific prolyl peptidase inhibitor, was used as a positive control. Percent inhibition of rmFAPa or rhFAPa enzymatic exopeptidase activity at 1 mM was determined for certain compounds, as shown in Table 2. For the calculations, the average measurements from reactions containing only vehicle and substrate, without enzyme, were used as a blank and were subtracted from the rest of the measurements. Percent inhibition was calculated using the average measurements from reactions containing vehicle, enzyme, and substrate as the maximum of enzymatic activity. Additionally, IC 50 for the rmFAPa or rhFAPa enzymatic exopeptidase activity of certain compounds are also shown in Table 2. Measurements were performed as a single point.

[0682] FAPa enzymatic endopeptidase (eollagenase) activity' assay. To assay baseline FAPa enzymatic exopeptidase activity, 50 ng of recombinant human FAPa (rhFAPa) (R&S system, #3715-SE) diluted in FAPa assay buffer (50 mM Tris pH 7.4, 100 mM NaCl, 0.1 mg/ml bovine serum albumin) was incubated w ith 5 pg of substrate DQ collagen solution (Molecular Probes #0-12060) with for 5h at 37 °C and protected from light m 384-well optiplates (Perkin Elmer, #384-F). To assay FAPa enzymatic endopeptidase activity inhibition by test compounds, all test compounds were pre-incubated with the enzyme for 30 min at 37 °C before starting the reaction by substrate addition in 384- well OptiPlates (Perkin Elmer, #384-F). Collagen hydrolysis w'as determined by measuring fluorescence at Ex/Em 495/515 nm using a multifunction Microplate Reader (Synergy 4, Biotek). All measurements were performed as a single point. Val-boroPro, a non-specific prolyl peptidase inhibitor, was used as a positive control. IC50 for the rhFAPa enzymatic endopeptidase activity (as determined by the eollagenase assay) of certain compounds are also shown in Table 2

Table 2: Exopeptidase or Endopeptidase inhibition of rmFAPa or rhFAPa by Test

Compounds

Ref, Comp.: Compound 60 as described in Jansen, K., et aL, J Med Chem, 2014. 57(7): p. 3053-74; for % of inhibition: +++ refers to >50% inhibition at 1 mM test compound; ++ refers to 25% < % inhibition < 50% at 1 mM test compound; + refers to <25% inhibition at 1 mM; for iC ¾ a ++÷ refers to lCso < 1 mM; ++ refers to 1 mM < ICso < 10 mM; + refers to ICso >10mM; - represents compound not tested; rmFAPa: recombinant mouse fibroblast activation protein alpha; rhFAPa: recombinant human fibroblast activation protein alpha;

endo: endopeptidase; exo: exopeptidase; inh: inhibition.

Example B2

Selectivity of the inhibition ofFAPa by test compounds was assessed compared to other prolyl oligopeptidase family S9 members: DPPIV, PREP, and DPP9

DPP IV enzymatic activity assay

[0683] To assay baseline dipeptidyi peptidase-4 (DPPIV) activity, 40 ng of recombinant human DPPIV (rhDPPIV) (R&S system, #1180-SE) or 40 ng of recombinant mouse DPPIV (rmDPPIV) (R&S system, #954-SE) was incubated with 400 mM of H-Gly-Pro-pNA substrate (BACHEM, #L~1880) in a DPPIV assay buffer (25 mM Tris, pH 8.3) for 30 min at 37 °C protected from the light in 96-well black plates (Nunc, #237108). To assay DPPIV inhibition by test compounds, test compounds were pre-incubated with the enzyme for 15 mm at 37 °C before starting the reaction by substrate addition in 96-well black plates (Nunc, #237108). Para- nitroani!ine (pNA) release was detected by measuring absorbance at 405 nm using a

Multifunction Microplate Reader (Synergy 4, Biotek). All measurements were carried out in triplicate. V al-boroPro, a non-specific prolyl peptidase inhibitor, was used as a positive control.

PREP enzymatic activity assay

[0684] To assay baseline prolyl endopeptidase (PREP) activity , 20 ng of recombinant human PREP (rhPREP) (R&S system, #4308-SE) or 20 ng of recombinant mouse PREP (rmPREP) (R&S system, #6339-SE) was incubated with 100 mM of Z-Gly-Pro-AMC peptide (BACHEM, #L-1145) in a PREP assay buffer (25 rnM Tris, 250 rnM NaCl, 10 rnM DTT, pH 7.5) for 30 min at 37 °C protected from light in 96-well black plates (Nunc, #237108). To assay PREP activity inhibition by test compounds, test compounds were pre-incubated with the enzyme for 15 min at 37 °C before starting the reaction by substrate addition in 96-well black plates (Nunc, #237108). 7-Amino-4-Methylcoumarin (AMC) release was detected by measuring fluorescence at Ex/Em 380/460nm using a Multifunction Microplate Reader (Synergy 4, Biotek). All measurements were carried out in triplicate. Val-boroPro, a non-specific prolyl peptidase inhibitor, was used as a positive control.

DPP9 enzymatic activity assay

[0685] To assay baseline dipeptidyl peptidase 9 (DPP9) activity, 40 ng of recombinant human DPP9 (rhDPP9) (R&S system, #5419-SE) was incubated with 100 mM of H-Gly-Pro-AMC peptide (BACHEM, #L-1215) in a DDP9 assay buffer (50 mM HEPES, pH 8) for 30 min at 37 °C in 96-well black plates (Nunc, #237108). To assay rhDPP9 activity inhibition by test compounds, test compounds were pre-mcubated with the enzyme for 15 min at 37 °C before starting the reaction by substrate addition m 96-well black plates (Nunc, #237108). 7-Amino-4- Methylcoumarin (AMC) release was detected by measuring fluorescence at Ex/Em 380/460 nm using a Multifunction Microplate Reader (Synergy 4, Biotek). All measurements were carried out in triplicate. Val-boroPro, a non-specific prolyl peptidase inhibitor, was used as a positive control.

[Q686] To determine if new FAPa inhibitors were selective or if they also inhibited other prolyl peptidases, the IC 50 for rmDPPIV, rhDPPIV, rmPREP, and/or DPP9 of certain test compounds, a reference compound (compound 60 as described in Jansen, K., et al., J Med Chem, 2014. 57(7): p. 3053-74), and Val-boroPro were determined, as shown in Table 3.

Table 3: Selectivity of FAPa Inhibition by Test Compounds

Validation of selective PRXS-AMC substrate for FAPa activity measurements

[Q687] FAPa activity can be measured by a general fluorescence intensity assay for dipeptidyl-pepiidases using a peptide substrate attached to a chemically quenched dye, such as Ala-Pro-7-amino~4~trifluoromethyl~cournarin (AFC) or a substrate containing the consensus Gy -Pro dipeptide such as Z-Gly-Pro-AMC (Lew, M.T., et a! , Hepatology, 1999, 29(6): 1768- 78; Santos, A.M., et al., J Clm Invest, 2009, 119(12): 3613-25; Park, J.E., et ai., I Biol Chem, 1999, 274(51): 36505-12; Niedermeyer, I., et al. Mol Cell Biol, 2000, 20(3): 1089-94; Narra,

K., et al, Cancer Biol Ther, 2007, 6(11): 1691-9; Lee, K.N., et al., I Thromb Haemost, 2011 , 9(5): 987-96; Li, J., et al., Bioconjug Chem, 2012, 23(8): 1704-11). These substrates are likely targeted also by other circulating proline-specific endopeptidases such as PREP that could be present in the reaction. By contrast, a proprietary substrate reagent, named PRXS-AMC, can specifically monitor FAPa activity.

[0688] To validate the high selectivity of this proprietary substrate, enzymatic activity' assays for FAP, DPP I V. PREP and DPP9 were earned out using Z-Gly-Pro-AMC or PRXS-AMC as described in Examples B1 and B2 [Q689] To assay FAPa, DPPIV, DPP9 and PREP enzymatic activities, human recombinant enzymes were used at 5, 2.5, 2.5 and 5 nM final concentrations, respectively. Z-G!y-Pro-AMC or PRXS-AMC were used at 25, 50, 100 and 200 mM final concentrations. Reactions were carried out for 60 min at 37 °C and were protected from light. AMC release was detected by measuring fluorescence at Ex/Em 380/460 nm using a Multifunction Microplate Reader in kinetic mode. Measurements were performed as a single point. Resulting fluorescence over time for PRXS-AMC and Z-gly-pro-AMC in the presence of rhFAPa is shown in FIG. 1A and FIG. IB, respectively; resulting fluorescence over time for PRXS-AMC and Z-gly-pro-AMC in the presence of rhPREP is shown in FIG. 2A and FIG. 2B, respectively; and resulting fluorescence over time for PRXS-AMC m the presence of rhDPPIV or rhDPP9 is shown in FIG. 3A and FIG. 3B, respectively.

[0690] PRXS-AMC is processed to a lesser extent than Z-Gly-Pro-AMC by the closely related prolyl oligopeptidase PREP at similar concentrations (see FIGs. 2A-2B). PRXS-AMC is not processed by DPPIV or DPP9 (FIGs. 3A-3B). In addition, PRXS-AMC showed an improved solubility' in aqueous buffers.

[0691] Approximately 500 pL of whole blood from one C57BL/6 mouse was harvested into BD Microtainer ® ' tubes (K2) EDTA (#365974, Beeson Dickinson and Co.) via terminal cardiac puncture. The blood sample was immediately centrifuged at approximately 9000 g at 4°C for 5 minutes. Plasma was separated and stored at -80 ° C in aliquots of 300 pL. To assay baseline FAPa enzymatic exopeptidase activity, 5 mΐ. of thawed, plasma was diluted (1:5) with cFAP buffer (100 mM Tris-HCI, 400 mM Nad, 50 mM salicylic acid, 1 mM EDTA, pH 7.5) and mixed with 35 pL of the same buffer before being pre-incubated with different concentrations of 10 pL of test compounds or DMSO vehicle for 15 minutes at 37 ° C m 96-well black plates (Nunc, #237108). After pre-incubation, 50 pL of 200 mM dipeptide substrate Z-Gly-Pro-AMC (Bachem, #L-1145) or PRXS-AMC were added to the mixture. The assay was performed for 1 hour at 37 ° C protected from light. 7-Amino-4-Methylcoumarin (AMC) release was detected measuring fluorescence at an excitation wavelength of 380 nrn and an emission wavelength of 460 nm using a Multifunction Microplate Reader (Synergy 4, Biotek). All measurements were carried out at least as a single point. Results are shown in Table 4.

DPPIV enzymatic activity in mouse plasma

[0692] Approximately 500 pL of whole blood from one C57BL/6 mouse was harvested into BD Microtainer ® tubes (K2) EDTA (#365974, Becton Dickinson and Co.) via terminal cardiac puncture. The blood sample was immediately centrifuged at approximately 9000 g at 4 ° C for 5 minutes. Plasma was separated and stored at -80 ° C in aliquots of 300 pL. To assay baseline DPPIV enzymatic exopeptidase activity, 5 pL of thawed, mouse plasma was diluted (1 :5) m buffer (100 mM Tris-HCl, 400 mM NaCl, 50 mM salicylic acid, 1 mM EDTA, pH 7.5) and mixed with 35 pL of the same buffer before being pre-incubated with different concentrations of 10 pL of test compounds or DMSO vehicle for 15 minutes at 37 ° C m 96-well black plates (Nunc, #237108). After pre-incubation, 50 pL of 200 mM dipeptide substrate H-Gly-Pro-AMC (Bachem, #L-1225) was added to the mixture. The assay was performed for 1 hour at 37 ° C. 7- Amino-4-Methylcoumarin (AMC) release was detected measuring fluorescence at an excitation wavelength of 360 nm and an emission wavelength of 460 nm using a Multifunction Microplate Reader (Synergy 4, Biotek). All measurements were carried out at least in duplicate. Results are shown in Table 4.

Table 4: Inhibition and Specificity of Test Compounds in Biological Samples

FAPa enzymatic activity in human plasma

[0693] Human plasma is diluted 1/10 m PBS. To assay baseline FAPa enzymatic

exopeptidase activity, the diluted plasma is incubated with 100 mM Z-Gly-Pro-AMC peptide (BACHEM, #L-l 145) for 1 h at 37 °C in 96-well black plates (Nunc, #237108). Test compounds are pre-incubated with the diluted plasma for 15 min at 37 °C before starting the reaction by substrate addition in 96-well black plates (Nunc, #237108). 7-Amino-4-Methylcoumarin (AMC) release is detected measuring fluorescence at Ex/Em 380/460 nm using a Multifunction Microplate Reader (Synergy 4, Biotek). All measurements are carried out in triplicate.

Example B6

Ex vivo inhibition of circulating FAPa activity from plasma of different species Human plasma.

[0694] Human blood was obtained from healthy young volunteers. Blood samples were collected in tubes coated with EDTA-K2 by venipuncture method, mixed gently, then kept on ice and centrifuged at 2,500 xg for 15 minutes at 4° C. After plasma separation, samples were stored at -80 ° C in aliquots of 300 pL.

[0695] To determinate the inhibitory potency of exemplary' test compounds over circulating FAPa activity from human plasma, 20 mE of thawed plasma were mixed with 20 pL of cFAP buffer (100 mM Tris-HCl, 400 raM NaCl, 50 raM salicylic acid, 1 mM EDTA, pH 7.5) and 10 pL different concentrations of exemplar} test compounds or vehicle (DMSO).

[0696] Exemplar} compounds were allowed to interact with the enzyme for 15 minutes at 37 ° C. After pre-incubation, 50 mί of 200 mM PRXS-AMC substrate were added to the all mixtures. All reactions were carried out for 1 h at 37 C protected from light. AMC release was detected measuring fluorescence at an excitation/emission wavelength of 380/460 nm using a

Multifunction Microplate Reader. All measurements were carried out as single point.

Results of IC 50 of exemplary test compounds over circulating FAPa from human are shown in Table 5.

Hamster plasma

[0697] Male Golden Syrian hamsters were provided by National Laboratory Animal Center (NLAC) in Taiw'an. The animals w'ere maintained in a hygienic environment under controlled temperature (20 - 24 °C) and humidity (50% - 80%) with 12 hours light/dark cycles. Free access to standard lab diet [MFG (Oriental Yeast Co., Ltd. Japan)] and autoclaved tap water were granted. All aspects of this work including housing, experimentation and disposal of animals were performed in general accordance with the“Guide for the Care and Use of Laboratory Animals: Eighth Edition’ (National Academies Press, Washington, D.C., 2011). In addition, the a mal care and use protocol was reviewed and approved by the LACUC at Pharmacology' Discovery Services Taiwan, Ltd.

Immediately after the sacrifice of hamsters, blood samples were collected via terminal cardiac puncture in tubes coated with EDTA-K2, mixed gently, then kept on ice and centrifuged at 2,500 xg for 15 minutes at 4°C. After plasma separation, samples were stored at -80 °C in aliquots of 300 pL.

[0698] To assay exemplary compounds in hamster plasma, a similar protocol as described for human plasma was performed diluting thawed plasma 1:2 in cFAP buffer. In a 96-well black plate, 5 mΐ of diluted hamster plasma were mixed with 35 mΐ of the same buffer and 10 mΐ of exemplary test compounds at different concentrations or vehicle (DMSO).

[0699] Exemplary test compounds were allowed to interact with the enzyme for 15 minutes at 37 ° C. After pre-incubation, 50 mΐ of 200 mM PRXS-AMC substrate were added to the all mixtures. All reactions were carried out for 1 h at 37 ° C protected from light. AMC release was detected measuring fluorescence at an excitation/emission wavelength of 380/460 nm using a Multifunction Microplate Reader. All measurements were carried out as single point.

[0700] Results of IC50 of exemplary test compounds over circulating FAPa from hamster plasma are shown in Table 5.

Table 5 - Inhibition ex- vivo by exemplary compounds of circulating FAPa activity from human and hamster plasma.

For 1C so- ÷ + + refers to ICso < 1 mM; + + refers to 1 ,uM < ICso <: 10 mM; + refers to IC o >10mM.

Example B7

Intravenous and oral bioavailability

[0701] The pharmacokinetic properties of exemplar}' test compounds were assayed after administration of an intravenous (IV) 2 mg/kg or oral (PO) 10 mg/kg single dose in mice.

Exemplary test compounds were formulated at 0.4 and 1 mg/ l in a vehicle containing Poly- Ethylene Glycol 200 (PEG200; Cat No. # P3015, Sigma Aldrich) and distilled water (dH 2 0) (50/50, v/v) as dosing solutions for intravenous and oral administration, respectively.

[0702] C57BL/6J and Balb/e mice, approximately 8-10 weeks old, were obtained from the vivarium Fundacion Ciencia & Vida Chile (Santiago, Chile) and maintained in a temperature- controlled room with 12/12 hr light/dark schedule with food and water ad libitum. Animals were acclimated for a minimum period of 4 days upon arrival at the testing facility.

[Q7Q3] On the day of study, mice were weighed and identified by marking the tail with numbers using a non-toxic permanent marker for designation into the experimental groups (n=3 per group). Each mouse in the IV dosing groups received a systemic bolus of 2 mg/kg dosing solution via the caudal vein. Each mouse of PO dosing groups received an intragastric bolus of 10 mg/kg via feeding tubes 2QG (Cat. No.: FTP-2038; Instech Salomon Inc.).

[0704] Blood samples 'ere harvested by terminal cardiac puncture at 5, 10, 15, 30, 60, 120, 240, 360 and 480 min after dosing. Non-dosed mice were used to collect samples of zero time points. Whole blood was collected into microtamer tubes with (K2) EDTA (Cat. No. #365974, Becton Dickinson & Co.). Blood samples were centrifuged immediately at 9,000 g at 4°C for 5 min and the plasma was separated. Plasma samples were placed into individually labeled cryovia!s (Cat. No. #366656, Thermo Fisher Scientific, Inc.) and stored in a -80°C freezer until LC/MS/MS bioanalysis.

[0705] The plasma samples w¾re analyzed by QTRAP 4500 triple quadrupole mass spectrometer (Applied Biosystems SCIEX) in positive or negative ion mode depending on the tested compound and interfaced with an ekspert ultraLC 100-XL UPLC System (eksigent) to determine the concentration of the exemplary test compound. Calibration standards (0.001 to 10 mM) and QCs (0.02, 0.2 and 2 mM) were prepared from naive mouse plasma in parallel with mouse pl asma study samples (60 mΐ) by precipitation with three volumes of ice cold internal standard solution (acetonitrile containing 20mM of theophylline). The precipitated samples were centrifuged at 6,100 g for 30 min at 4 °C. Following centrifugation, an aliquot of each supernatant was transferred into a clean sample vial and diluted with two volumes of aqueous mobile phase (0.2% formic acid in water). Samples were injected onto a reverse phase analytical column (YMC Triart Cl 8; 2.0 x 50 mm; 1.9 pm; YMC CO) and eluted with a gradient of 0.1 or 0.2% formic acid m Acetonitrile. Test compound and internal standard w'ere monitored by a multiple reaction monitoring (MRM) experiment using Analyst software (v 1.6.2, Applied Biosystems SCIEX). Quantitation was conducted using MultiQuant software (v2.1 , Applied Biosystems SCIEX) and the resulting calibration curve was fitted with a linear or quadratic regression and l/x weighting. The lower limit of quantitation were between 0.003 - 0.01 mM.

[0706] IV and PC) PK parameters were calculated from the concentration-time data using Phoenix WinNonlin software (v6.4, Certara, Princeton, NJ) by noncornpartmental analysis.

Area under the concentration-time curve (AU Clast) vras estimated using a log-linear trapezoidal method, from the dosing time to the last measurable concentration. Results for AUC of exemplary compounds in mouse plasma are shown in Table 6.

Table 6 - AUC and bioavai!ability of exemplary compounds after oral administration in mice.

Ref. Comp. : Compound 60 as described in Jansen, K., et al., J Med Chem, 2014. 5 /(7): p. 3053-

Example B8

In vivo pharmacokinetics and pharmacodynamics of test compound. 13

[Q7Q7] Solutions of test compound 13 were prepared at 1 mg/niL in a vehicle containing 50% polyethylene glycol 200 (PEG200, #P3015-lKG: Sigma-Aldrich, Inc.) in distilled water for oral administration. [Q7Q8] Female C57BL/6 mice (approximately 9-10 weeks old; 20-21 grams) obtained from a vivarium (Fundacion Ciencia & Vida, Santiago, Chile) were weighed and divided into cohorts described in Table 7.

[0709] 100 mΐ, of whole blood was sampled from the tail vein of each mouse 24 hours prior to dosing. On the day of dosing, mice of all cohorts orally received a single dose (10 mg/kg) of test compound 13 using feeding needles (#FTP-2Q38/050312, Instech Laboratories, Inc,).

[0710] Depending on each cohort, approximately 500 pL of whole blood from each mouse was collected into BD Microtainer ® tubes (K2) EDTA (#365974, Becton Dickinson and Co.) via terminal cardiac puncture at the harvesting time point (Table 5). The blood sample w ? as immediately centrifuged at approximately 9000 g at 4 C for 5 minutes. Plasma was separated and placed into individual!} ' labeled cryotube vials (#366656; Thermo Fisher Scientific, Inc.) and stored at -80 ° C prior to being assayed for enzymatic activity or LC/MS/MS analysis.

[0711] All plasma samples were thawed and diluted with assay buffer (1 part plasma for 5 parts buffer). The assay buffer contained 100 mM Tris-HCl, 400 mM NaCl, 50 mM salicylic acid, 1 rnM EDTA, pH 7.5. All plasmas were assayed for FAP and DPPIV activity. To assay the plasma for FAPa or DPPIV enzymatic activity, 5 mΐ, of each diluted plasma sample was loaded into a well of a 96-well black plate (Nunc, #237108), which contained 45 mT of additional assay buffer. The plates were warmed at 37 ° C for 15 minutes before the assay began. To start the assay, 50 pL of 200 mM dipeptide substrate Z-Gly-Pro-AMC (Bachem, #L-1145) (FAPa enzymatic activity assay) or 50 pL of 200 mM dipeptide substrate H-Gly-Pro-AMC (Bachem, #L-1225) (DPPIV enzymatic activity assay) was added to each well. 7-Amino-4- Methylcoumann (AMC) release was detected measuring fluorescence at an excitation wavelength of 360 nm and an emission wavelength of 460 nm using a Multifunction Microplate Reader (Synergy 4, Biotek). All measurements were carried out at least in duplicate.

[0712] For each animal orally dosed with compound 13, the activities of FAP and DPPIV found m the plasma sample collected post-dosing were normalized as percentage to the respective acti vity found in pre-dosing plasma samples. Percentage of FAP activities found in plasma of mice orally dosed with compound 13 are summarized in FIG. 4. Percentages of DPPI activity are summarized in FIG. 5.

Example B9

Oral pharmacokinetics (PK) and pharmacodynamics (PD) studies in mice

[0713] To determine the PK/PD of exemplary test compounds, groups of mice were orally dosed with compound 13 (an exemplar ' compound) and then plasma samples collected at different time point were subjected to bioanalysis for concentrations of compound and FAP activity.

Female c57BL/6 mice, approximately 8-10 weeks old (19-21 gr), obtained from the Vivarium Fundacion Ciencia & Vida were acclimated for a minimum period of 4 days upon arrival at the testing facility. On the day of dosing, animals were weighed and identified by marking the tail with numbers using a non-toxic permanent marker for designation into the treatment groups described in Table 8. Table 8 - Experimental groups for PK/PD study.

[0714] The compound 13 (an exemplar} compound) was formulated at 5 mg/ml in a vehicle containing Poly-Ethylene Glycol 200 (PEG200; Cat. No. # P3015, Sigma Aldrich) and distilled water (dH 2 0) (50/50, v/v) as dosing solution for oral administration.

[0715] For oral administration, mice from groups 1 up to 6 received a single oral dose at 10 mL/Kg of vehicle PEG200/dH 2 0 (50/50, v/v). Mice from groups 7-12 received a single oral dose of compound 13 (an exemplary compound) formulated freshly at 5 mg/mL in vehicle.

[0716] Whole blood was collected via cardiac puncture at 1, 4, 8, 12, 16 and 24 h after administration of vehicle or compound 13 (an exemplary compound). Mice of group 13 were not dosed and were used to collect samples of the zero time point. Blood samples were immediately centrifuged at approximately 9000 g at 4 ° C for 5 minutes. Plasma was separated and placed into individually labeled cryotube vials and stored at -80 ° C prior to being assayed for enzymatic activity or bioanalvsis. [0717] All plasma samples were thawed and diluted 1 :5 with assay buffer (100 mM Tris-HCl, 400 mM NaC!, 50 mM salicylic acid, 1 mM EOT A, pH 7.5). All plasma was assayed for FAP and DPPIV activity. To assay the plasma for FAPoc or DPP1V enzymatic activity, 5 u.L of each diluted plasma sample was loaded into a well of a 96-well black plate, which contained 45 mT of additional assay buffer. The plates were warmed at 37 ° C for 15 minutes before the assay began. To start the reactions, 50 mT of 400 mM dipeptide substrate Z-Gly-Pro-AMC (FAPa enzymatic activity assay) or 50 L of 200 mM dipeptide substrate H-Gly-Pro-AMC (DPPIV enzymatic activity assay) diluted in assay buffer were added to each well. AMC release was detected measuring fluorescence at an excitation wavelength of 360 nm and an emission wavelength of 460 nm using a Multifunction Microplate Reader. All measurements were carried out at least in duplicate.

[0718] All fluorescence measurements were corrected subtracting the average fluorescence from blank reactions containing only substrate and assay buffer. To calculate the remaining FAP and DPPIV in plasma from mice dosed with vehicle or compound 13 (an exemplary compound), all fluorescence measurements were normalized against the fluorescence from samples of non-dose group assumed as one hundred percent of activity for each assay.

Percentage of remaining FAP activity found in plasma of mice orally dosed with vehicle or compound 13 (an exemplary compound) are shown in FIG. 6A. Percentage of DPPIV activity are summarized in FIG. 6B. A comparison of the FAPa activity and the PK/PD of the compound over time is shown in FIG. 6C

Example BIO

Tumor Inhibition In Vivo

[0719] Female C57/BL6 mice (approximately 8-9 weeks old; 20-21 gr) obtained from the vivarium of Fundaeion Ciencia & Vida (Santiago, Chile) were maintained in a temperature- controlled room with 12/12 hour light/dark schedule with food and water ad libitum. The mice were acclimated for a minimum period of 4 days upon arrival at the testing facility.

[0720] MC38 mouse colon cancer cell line was maintained as monolayer culture m

DMEM-F12 (Cat. No.: SH30023.01, Hyclone) supplemented with 10% fetal bovine serum (Cat No.: 16000, Gibco) and penicillin/streptomycin (Cat. No.: 15140122, Gibco) at 37°C in an atmosphere with 5% C0 2 . The cells were routinely subcultured every 3 days to maintain growth at exponential phase. The tumor cells growing in exponential growth phase were harvested using IX PBS with 0.05% trypsin-EDTA (Cat. No.: 15400054, Gibco), followed by centrifugation at 330 g x 3 min in a centrifuge at room temperature. The supernatant was subsequently removed by aspiration. Cell pellet was resuspended in approximately !Ox volume of cell culture medium and counted. Cell viability was determined to be >95% by trypan blue staining.

[0721] At the day of inoculation, female C57/B16 mice (n=15 total) were weighed and identified by marking the tail with numbers using a non-toxic permanent marker. Mice were inoculated subcutaneously in the right lower flank (near the dorsal thigh region) with a single volume of 0.1 mL cell suspension containing approximately 2 x 10 6 MC38 cells in IX PBS. Tumors were measured three times per week with digital calipers and tumors volumes, expressed in mm 3 , were calculated with the following formula:

Tumor volume (mm 3 ) = (a x !> 2 )/2

where“b” is the smallest diameter and“a” is the largest perpendicular diameter, [0722] Seven days after inoculation, mean tumor volume was about 100 mm’. The mice were weighted and randomized into two experimental groups (n = 5-6 mice), and receive the following treatments dosed orally twice daily until end: 1) Vehicle (PEG200 50% in water); or 2) 50 mg/kg test compound 24 m PEG200 50%/water. Body weight of the mice and tumor volume was recorded two or three times per week for a total of 22 days. At day 22, mice were sacrificed and all tumor mass were weighed.

[0723] Tumor volume is shown as mean with standard error of mean (SEM) in FIG. 7. Mouse body weight gam is shown as mean with standard error of mean (SEM) in FIG. 8. Individual recording of tumor volume is shown m FIG. 9. Tumor mass weight is shown as mean with standard error of mean (SEM) in FIG. 10. As shown by the figures, mice treated with test compound 24 had a small er volume of tumor than treatment with the vehicle alone. No statistical significance was observed in these studies.

Example Bll

FAPa enzymatic activity in murine tumors [0724] Mouse tumor proteins are extracted in a lysis buffer (Tris HC1 50mM pH 7 6, EDTA 1 mM, Glycerol 10%, protease/phosphatase inhibitors cocktail) for 20 min using an ultra Turrax (IKA, #3737000). To assay baseline FAPa enzymatic exopeptidase activity in the tumor extract, 10 pg of tumor extract sample is incubated with ! OOpM Z-Gly-Pro-AMC peptide (BACHEM, #L~1145) in PBS lx for 1 h at 37°C in 96-well black plates (Nunc, #237108). Inhibitors are preincubated with the tumor extract for 15 mm at 37 °C before starting the reaction by substrate addition in 96-well black plates (Nunc, #237108). 7-Amino-4-Methylcoumarin (AMC) release is detected measuring fluorescence at Ex/Em 380/460nm using a Multifunction Microplate Reader (Synergy 4, Biotek). All measurements are carried out in triplicate.

Example B12

PK/PD studies in tumors of murine cancer models

B16-F10 murine melanoma model

[Q725] Male C57B1/6 mice were engrafted intradermal with lxlO b B16-F10 murine melanoma cells suspended m 100 pL of sterile PBS IX (Day 0). On day 2 post-engraftment, groups of animals (n=3) were dosed with vehicle PEG200/dH 2 0 (50/50 v/v) or compound 13 (an exemplar compound) at 50mg/Kg orally (PO) twice a day (BID) until day 15.

[0726] At terminal day, all mice were sacrificed and immediately tumors and plasma samples were collected and stored at -80 ° C prior to being assayed for FAPcx enzymatic activity or bioanalysis.

[0727] Proteins of murine cancer tumors were extracted m lysis buffer (Tris HC1 50mM pH 7.6, EDTA 1 mM, Glycerol 10%, protease/phosphatase inhibitors cocktail) using an Ultra Turrax homogenizer (ΪKA, #3737000). Homogenates were clarified by centrifugation 21 ,000 g for 20 min at 4 °C. Supernatants were collected and proteins were quantified using a BCA Protein assay kit (Cat. No. #23225, Thermo Scientific). All protein samples were adjusted at 2 pg/pL with lysis buffer and then aliquoted for storing at -80 °C.

[0728] For assessment of FAPa activity in tumor homogenates, 10 pg of each tumoral extract were diluted with cFAP buffer assay in a 96-well black plate at final volume of 50 pL and then mixed with 100 pM PRXS-AMC peptide. Reactions were carried out for 1 h at 37 °C. AMC release tvas detected measuring fluorescence at Ex/Em 380/460n using a Multifunction Microplate Reader and expressed as relative fluorescence units (RFU). All measurements are carried out as single point.

[0729] Concentrations of compound 13 (an exemplar} compound) are shown in FIG. 11A. FAPcx activities in plasma and tumor from animals bearing B16-F10 murine melanoma tumors are shown in FIG. 1 IB.

MC38 murine melanoma model

[0730] : Female C57B1/6 mice were engrafted subcutaneously with 2xl0 6 MC38 mouse colon cancer cells suspended m 100 mΐί of sterile PBS IX (Day 0). On day 12 post-engraftment, mice were randomized based on tumor volume into two experimental groups (n = 6) with a mean tumor volume -160 mrnf Groups of animals received vehicle PEG200/dH 2 0 (50/50 v/v) or compound 13 (an exemplar} compound) 50 mg/kg PO bid until day 24.

[0731] At terminal day, all mice were sacrificed and immediately tumors and plasma samples w¾re collected and stored at -80 ° C prior to being assayed for FAPcx enzymatic activity or bioanalysis as described for the B 16-F10 murine melanoma model.

[0732] Concentrations of compound 13 (an exemplary compound) are shown in FIG. 12A. FAPa activities in plasma and tumor from animals bearing MC38 murine melanoma tumors are shown in FIG. 12B.

Example B13

In vitro FAP -mediated proteolytic cleavage ofFGF21 assays.

[Q733] Pharmacological administration of FGF21 to diabetic and obese animal models markedly ameliorates obesity , insulin resistance, dyslipidemia, fatty liver, and hyperglycemia in rodents (Markan, .R. et. al., Semm Cell Dev Biol, 2016, 53: 85-93), and FGF21 analogs have been efficacious in inducing weight loss and correcting hypermsulinemia, dyslipidemia, and hypoadiponectinemia in obese people with type 2 diabetes (Gaich, G., et al.. Cell Metab, 2013, 18(3): 333-40; Dong, J.Q., et al., Br J Clm Pharmacol, 2015, 80(5): 1051-63). However, m rodents and primates, the half-life of exogenously administrated human FGF21 is short (~ 0.5-2 h) as result of FAP -mediated enzymatic degradation and susceptibility to renal clearance (Hager, T., et al.. Anal Chem, 2013. 85(5): 2731-8; Xu, J., et al, Am I Physiol Endocrinol Metab, 2009, 297(5): El 105-14; Kharitonenkov, A., et al., Endocrinology, 2007, 148(2): 774-81) Common half-life extension strategies have improved significantly the PK properties of these FGF21 analogs in vivo; however, proteolytic processing still persists in these analogs (Hecht, R., et al., PLoS One, 2012, 7(11): e49345; Mu, I, et al., Diabetes, 2012, 61(2): 505-12; Camacho, R.C., et al., Eur J Pharmacol, 2013. 715(1-3): 41-5). To determine if the FAP inhibitor compounds described herein can inhibit the FGF-21 cleavage and can offer an oral therapy to augment endo- and/or exo- genous FGF21 action, FAP-mediated digestion of FGF21 in there presence and absence of exemplary FAP inhibitor compounds were compared.

FAP-mediated digestion ofFGF21 in vitro

[0734] Recombinant human FGF21 (rhFGF2l; Cat. No. #2539-FG-025/CF, R&D systems) was incubated overnight (16 h) at 37 °C with recombinant human FAP (rhFAP; Cat. No. #3715- SE-OiO, R&D systems) in digestion buffer (50 rnM Tris pH 7 4, 100 mM Nad, 0.1 mg/ml bovine serum albumin). Reactions were carried out at final concentrations of 1000 ng/ ' mL hFGF2l and 400, 800 or 1200 ng/mL rhFAP m a volume of 100 pL. For SDS-PAGE analysis, after incubation each sample received immediately 4x Laemmli protein sample buffer (Cat. No. #161 -0747, Bio-Rad) supplemented with 0.1 ml b-mercaptoethanol/lO ml aliquot and then boiled at 95 °C during 10 min. An aliquot of 15 m]_ of each sample were then loaded onto a reducing 20% Tris-Tricine SDS-PAGE gel. For immunoblot analysis, proteins were separated on SDS-PAGE gels and transferred to PVDF membranes (Cat. No. #1620177, Bio-Rad). For immunodetection, anti-FGF2l (Cat. No. #RDl 81108100, BioVendor) and anti-rabbit HRP conjugated (Cat. No. #611-1322, Rockland) 'ere used as primary and secondary antibodies, respectively. Proteins were detected using the ECL Western Blotting Substrate (Cat. No.

#32106, Thermo Fisher Scientific) and visualized using a ChemiDoc™ Imaging System

(Biorad) and Image lab software v5.2.1 build 11.

[0735] Immunodetection of intact and rhFAP -cleaved forms of rhFGF2l is shown in FIG. 13A and densitometr' analysis is shown in FIG. 13B. As shown by the immunobl oiling image and densitometry analysis, cleaved form of rhFGF21 increases in an rhFAP concentration- dependent fashion.

Inhibition of FAP -mediated digestion ofFGF2I in vitro. [Q736] To compare the efficacy for inhibiting the rhFAP-mediated proteolytic processing of rhFGF21, exemplary compounds and the commercial non-selective DDP inhibitor Val-boroPro were tested at low concentrations in the FGF21 cleavage assays in vitro.

Test compounds and Val-boroPro were prepared from powder as 10 mM stock solutions in DMSO and stored in presence of N2 neutral atmosphere at - 80°C. Stock solutions were pre dicted in DMSO to get diluted aliquots at 1 and 0.1 mM. These aliquots were 10-fold diluted again in digestion buffer and then 10 pL of these dilutions were added to 50 pL of rhFAP. The enzyme-inhibitor mixture was allowed to interact for 30 min at 37 °C and then 40pL of rhFGF2i were added to start the reaction. Reactions were carried out at final concentrations of 1000 ng /' mL hFGF21 and 1200 ng/mL rhFAP for 16 h at 37 °C. After incubation, samples were boiled immediately in Laemmli protein sample buffer and then proteins were resolved in a reducing 20% Tris-Trieine SDS-PAGE gel. The immunodetection of intact and rhFAP-cleaved forms of rhFGF2l was carried out as described above.

[0737] Comparison of efficacy between compound 13 (an exemplary compound) and Val- boroPro at 100 and 1000 nM for inhibiting the proteolytic processing of rhFGF2l by rhFAP in vitro is showed in the FIG. 13C and desitometry analysis is shown in FIG. 13D. As shown by the immunoblotting image and densitometry analysis, the intact form of rliFGF2I is preserved in presence of compound 13 (an exemplary compound) even at a low concentration.

[0738] Additionally, the TC50 for FAP-mediated proteolytic cleavage of FGF21, was determined in the using a serial dilution of compound 13 (an exemplary' compound). As shown m FIG. 14A, the exemplary' compound inhibited rliFAP-mediate proteolytic processing of FGF21 in a dose-dependent manor in vitro. As shown by the densitometry analysis in FIG. 14B, the exemplary' compound showed an IC50 in the nanomolar range in FAP-mediated proteolytic cleavage of FGF2l assays.

Example B14

rhFGF21 administration in conjunction with FAP inhibition in vivo.

[Q739] To determine if exemplary compounds of the invention can contribute to extend the half-life of hFGF2l in vivo, rats were orally dosed with vehicle or compound 13 (an exemplary compound) followed by sub-therapeutic dose of rhFGF21. [Q74Q] Male Sprague Dawley rats, approximately 7-8 weeks old (~250g), were obtained from the vivarium of Universidad Catolica de Chile (Santiago, Chile) Animals were acclimated for a minimum period of 4 days upon arrival at the testing facility. At the day of study, two rats were anesthetized using sevoflurane and subjected to surgical implantation of catheters in carotid artery and jugular vein. Catheter plugs were fixed to the external ends of the catheters and kept patent using an anti-coagulant heparinized saline solution containing 25 U/mL Heparin (Fresenius Kabi, Laboratorio Sanderson) in 0.9% NaCl Apirofiex (Fresenius Kabi, Laboratory Sanderson). After surgical recovery', rats were weighed and identified by marking the tail with numbers using a non-toxic permanen t marker for designation of treatments described in Table 9.

Table 9 - Experimental treatments of cannulated rats.

[0741] Compound 13 (an exemplary' compound) was formulated at 5 mg/ml in vehicle PEG200/dH 2 0 (50/50, v/v) for oral administration and rliFGF2i was formulated in 0.9% NaCl Apirofiex at 100 pg/mL.

Whole blood samples were collected via the implanted carotid artery catheter from both animals (time point = -15 min) and immediately after the first dose was administered (a single oral dose of 50 mg/Kg vehicle or compound 13 via feeding tubes (15 gauge) to Rat#l and #2, respectively). After 15 min, a second whole blood sample was collected (time point = 0 min) and immediately after the second dose was administered (a single intravenous dose of rhFGF2i 90 pg/Kg via the implanted jugular vein catheter). Whole blood samples were then collected at time points: 5, 15, 30, 60, 120, 240 and 480 min post- second dosing. Blood samples were immediately centrifuged at approximately 9000 g at 4 ° C for 5 minutes. Plasma was separated and placed into individually labeled cryotube vials and stored at -80 ° C prior to be assayed for immunodetection of rhFGF21 by Western blots technique, FAPa activity assays using PRXS- AMC or bioanalysis as described above in previous examples. [0742] For immunodetection, thawed plasma samples were centrifuged 2,000 g for 15 min at 4 °C and then supernatants were transferred into a clean Eppendorf tube. 1.5 pL of each plasma sample were mixed with 11.25 u.L of 4x Laemmli protein sample buffer and 32,25 pL of dH 2 0. Proteins were boiled and then 15pL of each sample were resolved in a reducing 20% Tris- Tricine SDS-PAGE gel and then immunob!otted for detection of FGF21 as described above.

[0743] Results are summarized in FIG. 15. The administration of compound 13 at an oral single dose of 50 mg/kg potently suppressed the plasma FAP activity. The pre-treatment with the exemplary compound increased the amount of rhFGF21 detected through time in the plasma.

Example B15

Oral PK/PD studies in hamsters.

[0744] hFAP hydrolyzes peptides that have both a Glycine (Gly) at P2 and a Proline (Pro) at PI . The Gly -Pro FAP consensus residues at position 170-171 in human FGF21 is conserved in most mammalian species with available FGF21 sequences (including predicted sequences). However, the FGF21 expressed in rats and mice possess Glu-Pro instead of the conserved Gly- Pro at the putative FAP cleavage site. It has been reported that hFAP does not process FGF21 containing the murine sequence (Dunshee, D.R., et al., J Biol Chem, 2016, 291(11): 5986-96). Interestingly, the corresponding peptide from Syrian hamster FGF21 with Leu at the RG position exhibited significant hydrolysis by hFAP (Dunshee, D.R., et al, J Biol Chem, 2016, 291(11): 5986-96).

[Q745] To assess the effect of FAP inhibition by an exemplary compound over the cleavage of endogenously produced FGF21, in vivo PK/PD studies were conducted in male Golden Syrian hamsters following single intravenous arid oral administration of an exemplary compound.

[0746] Male Golden Syrian hamsters were provided by National Laboratoxy Animal Center (NLAC) in Taiwan. The animals were maintained m a hygienic environment under controlled temperature (20 - 24 °C) and humidity (50% - 80%) with 12 hours light/dark cycles. Free access to standard lab diet [MFC (Oriental Yeast Co., Ltd. Japan)] and autoclaved tap water were granted. All aspects of this work including housing, experimentation and disposal of animals were performed in general accordance with the“Guide for the Care and Use of Laboratory' Animals: Eighth Edition” (National Academies Press, Washington, D.C., 2011) under the supervision of veterinarians. In addition, the animal care and use protocol was reviewed and approved by the IACUC at Pharmacology Discovery Services Taiwan, Ltd.

[0747] Compound 13 (an exemplary compound) was formulated in dimethyl sulfoxide (DMSO)/ Solutol® HS15 (BASF, Germany) / phosphate buffered saline (PBS, Sigma, USA) (5/5/90, v/v/v) at 0.2 mg/mL for IV injection and polyethylene glycol (PEG) 200 (Sigma, USA)/ water for injection (WFI; Tai-Yu, Taiwan) (50/50, v/v) at 1 mg/mL for PO administration. The dosing volumes were 5 mL/kg for IV and 10 mL/kg for PO.

[0748] On the day of study, hamsters were anesthetized and subjected to surgical implantation of catheters in the jugular vein. Catheter plugs were fixed to the external ends of the catheters and kept patent using an anti-coagulant heparinized saline solution. After surgical recovery', hamsters were weighed and identified by marking the tail with numbers using a non-toxic permanent marker for designation for IV or PO groups (n=3) as Table 10 details.

Table 10 - Experimental groups for PK/PD study in hamsters.

[0749] Hamsters from Group 1 received an intravenous bolus via j ugular vein of 1 mg/kg of compound 13 (an exemplary compound). On the other hand, hamsters from Group 2 received an intragastric bolus of 10 mg/kg of compound 13 (an exemplary compound) using feeding needles.

[0750] Plasma samples were collected at 5, 10, 15, 30, 60, 120, 240, 360, 480 and 1440 minutes post-dose in both IV and PO groups. Hamsters from group 3 were not dosed and were used to collect samples of zero time point and baseline levels of TAP activity and endogenous FGF21.

[0751] Blood aliquots (100 - 150 pL) were collected from jugular vein catheterized hamsters in tubes coated with EDTA-K2, mixed gently, then kept on ice and centrifuged at 2,500 xg for 15 minutes at 4°C, within 1 hour of collection. Plasma samples were placed into individually labeled cryotube vials and stored at -80 ° C prior to be assayed for FAPa activity assays (Z-Gly- Pro-AMC substrate) and bioanalysis as described above in previous examples.

[0752] Levels of endogenous hamster FGF21 in the plasma samples were quantified by using an ELISA kit (Cat. No. # EZRMFGF21-26K, Miilipore) according to the manufacturer’s instructions. The detection limit was 44.85 pg/rnl. Plasma samples of non-dosed animals were analyzed as a pool of plasma. All samples were assayed as single point. The results are shown in FIG. 16.

[0753] The AUC] asl of compound 13 (an exemplary compound) for intravenous and oral administration equaled 255 and 350 hr.ng/mL, respectively. Compared with baseline levels, FAP activity diminished quickly after the administration of compound 13 (an exemplary compound) and although the baseline levels of FGF21 differed between individuals, the levels of total FGF21 increased in the most of hamsters. Importantly, the time course of this increment correlated closely with the inhibition of activity of endogenous FAP in plasma. Therefore, inhibition of FAP in vivo by the administration of an exemplary compound resulted in an increase of endogenous serum FGF21, suggesting protection from cleavage by FAP.

Example B16

Efficacy on Diet-Induced Obesity (DIO) model in hamsters

[0754] Groups of 10 male golden Syrian hamsters weighing 90 ± 10 g are fed a high fat diet (HFD) (g/lOOg: com oil, 5; coconut oil, 5; cholesterol, 0.2; standard chow, 89.8) throughout the experiment. Seven days after the beginning of the diet, vehicle, ezetimibe (positive control) or an exemplary test compound are administered by oral gavage daily (PO, QD) for 28 consecutive days (Day 1 ). An extra non -treated group of animals (Group 5) are fed normal diet (control). Body weights (BW) are measured and recorded twice a week.

[0755] After fasting overnight, blood is obtained from the retro-orbital sinus of each animal 5 m before daily dosing on Days 1 , 8, 15, 22 and 29 after dosing for 0, 7, 14, 21 and 28 days.

[0756] The serum obtained from each hamster is assayed for fasted glucose, total cholesterol (Total), low density lipoprotein (LDL), high density lipoprotein (1 11)1.) and triglyceride (TG). Post-treatment values are calculated as a percentage of pre-treatment values. The percent change of treated relati ve to the vehicle control group is also determined. [Q757] At terminal Day 29, all animals are sacrificed and immediately subjected to cardiac puncture for collecting the maxi um volume of blood samples A half of blood sample is processed for serum and is used to assay for terminal adiponectin and insulin by ELISA methods. The other half of blood sample is processed for plasma. Plasma samples are placed into individually labeled cryotube vials and stored at -80 ° C prior to FAPa activity assays, quantitation of FGF21 levels by ELISA, and bioanalysis of the exemplary compound as described above in previous examples.

[0758] Additionally, tissues such as brain, liver, interscapular BAT, epididymal WAT, pancreas and gastrocnemius muscle are harvested for molecular and biochemical assessments of FGF21 target genes and proteins. Portions of the liver are also collected as non-fixed frozen samples and stained with Oil Red-0 staining, or formalin-fixed and paraffin-embedded samples for H&E staining and histopathological analyses.

[0759] All references throughout, such as publications, patents, patent applications and published patent applications, are incorporated herein by reference m their entireties.