Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
SUBSTITUTED IMIDAZOPYRIDINES, THEIR PREPARATION AND THEIR USE AS PHARMACEUTICALS
Document Type and Number:
WIPO Patent Application WO/2017/066876
Kind Code:
A1
Abstract:
This application relates to substituted imidazopyridines, compositions comprising them and their uses in the treatment of diseases and conditions in which inhibition of a bromodomain is indicated. For example, the application relates to substituted imidazopyridines and to their use as bromodomain inhibitors. The present application is also related to the treatment or prevention of proliferative disorders, auto-immune disorders, inflammatory disorders, dermal disorders, and neoplasms, including tumors and/or cancers.

Inventors:
POURASHRAF MEHRNAZ (CA)
BEAULIEU MARC-ANDRÉ (CA)
CLARIDGE STEPHEN (CA)
BAYRAKDARIAN MALKEN (CA)
JOHNSTONE SHAWN (CA)
ALBERT JEFFREY S (CA)
GRIFFIN ANDREW (CA)
Application Number:
PCT/CA2016/051215
Publication Date:
April 27, 2017
Filing Date:
October 20, 2016
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
NEOMED INST (CA)
International Classes:
C07D471/04; A61K31/444; A61K31/4545; A61K31/553; A61P29/00; A61P35/00; A61P37/06
Foreign References:
CA2874953A12013-12-19
CA2875016A12013-12-19
CA2932030A12015-06-18
Other References:
YAMANAKA, M. ET AL.: "Imidazo[1, 2-a]pyridines.I Synthesis and Inotropic Activity of New 5-Imidazo[1,2-a]pyridiny-2(1H)-pyridinone Derivatives''.", CHEM. PHARM. BULL., vol. 39, no. 6, 1991, pages 1556 - 1567, XP055376396
YAMANAKA, M. ET AL.: "Imidazo[1,2-a]pyridines. III. Synthesis and Bradycardic Activity of New 5-Imidazo[1,2-a]pyridin-6-ylpyridine Derivatives''.", CHEM. PHARM. BULL., vol. 40, no. 6, 1992, pages 1486 - 1493, XP055376399
Attorney, Agent or Firm:
ROBIC LLP (CA)
Download PDF:
Claims:
A compound of Formula I:

Formula I

wherein,

R1 is a substituted or unsubstituted group selected from d-C6alkyl, C2-C6alkenyl, C2- C6alkynyl, C3-C10cycloalkyl, C3-C10heterocycloalkyl, d-docycloalkyld-dalkyl-, C3- doheterocycloalkyld-Cealkyl-, d-Ci0aryld-dalkyl-, Cs-doheteroaryld-dalkyl-, C(0)Ra, ORa, NHRa, N(Ra)2, C(0)NH2, C(0)NHRa, C(0)N(Ra)2, NHC(0)Ra, N(Ra)C(0)Ra, NHC(0)NHRa, N(Ra)C(0)NHRa, NHC(0)N(Ra)2, N(Ra)C(0)N(Ra)2, NHS02Ra, N(Ra)S02Ra, NHS02NHRa, N(Ra)S02NHRa, NHS02N(Ra)2, N(Ra)S02N(Ra)2, S02Ra, S02NH2, S02NHRa, and S02N(Ra)2;

Ra is, independently in each occurrence, a substituted or unsubstituted group selected from Ci-C6alkyl, C2-C6alkenyl, C2-C6alkynyl, C3-Ci0cycloalkyl, C3-Ci0heterocycloalkyl, C3- Ci0cycloalkylCi-C6alkyl-, C3-Ci0heterocycloalkylCi-C6alkyl-, C6-Ci0aryl, C5-Ci0heteroaryl, C6- Ci0arylCi-C6alkyl-, and C5-CioheteroarylCi-C6alkyl-;

R2 is selected from H, NH2, CN or a substituted or unsubstituted group selected from d- C6alkyl, C2-C6alkenyl, C2-C6alkynyl, C3-Ciocycloalkyl, C3-Cioheterocycloalkyl, C6-Ci0aryl, C5- Cioheteroaryl, C(0)Ra, ORa, SRa, NHRa, N(Ra)2, C(0)NH2, C(0)NHRa, C(0)N(Ra)2, NHC(0)Ra, S02Ra, S02NHRa, S02N(Ra)2, NHS02Ra, N(Ra)S02Ra, NHS02NHRa, N(Ra)S02NHRa, NHS02N(Ra)2, and N(Ra)S02N(Ra)2;

R3 and R6 are each independently H, halogen, NH2, CN or a substituted or unsubstituted group selected from d-C6alkyl, C(0)Ra, ORa, NHRa, N(Ra)2, C(0)NH2, C(0)NHRa, C(0)N(Ra)2, NHC(0)Ra; and

one of R4 and R5 is H, halogen, NH2, CN or a substituted or unsubstituted group selected from C C6alkyl, C(0)Ra, NH2, NHRa, N(Ra)2, C(0)NH2, C(0)NHRa, C(0)N(Ra)2, and NHC(0)Ra; and the other of R4 and R5 is a group of Formula II:

Formula II

wherein,

R7 and R10 are each independently H, halogen (such as F, CI), CN, or a substituted or unsubstituted group selected from d-Cealkyl or C3-C6cycloalkyl group, OC Cealkyl, OC3- Cecycloalkyl, SC C6alkyl, SC3-C6cycloalkyl, NHC C6alkyl, NHC3-C6cycloalkyl, N(C C6alkyl)2, NHC(0)C C6alkyl and NHC(0)C3-C6cycloalkyl;

R8 is halogen (such as F, CI), CN, or a substituted or unsubstituted group selected from C C6alkyl or C3-C6cycloalkyl group, OC C6alkyl, OC3-C6cycloalkyl, SC C6alkyl, SC3- Cecycloalkyl, NHC C6alkyl, NHC3-C6cycloalkyl, N(C C6alkyl)2, NHC(0)C C6alkyl and NHC(0)C3-C6cycloalkyl;

R9 is a substituted or unsubstituted C C3alkyl or C3-C5cycloalkyl group;

X1, X2, and X3 are each selected from a nitrogen or carbon atom, wherein when X1, X2, or X3 is a nitrogen atom, then the R7, R8, or R10 attached thereto is absent, provided that at least two of X1 , X2, and X3 are C;

or a pharmaceutically acceptable salt, solvate, ester or prodrug thereof.

2. The compound of claim 1 , or a pharmaceutically acceptable salt, solvate, ester or prodrug thereof, wherein R4 is a group of Formula II.

3. The compound of claim 2, wherein said R5 is a substituted or unsubstituted C C3 alkyl. 4. The compound of claim 2, wherein said R5 is a hydrogen atom.

5. The compound of claim 1 , or a pharmaceutically acceptable salt, solvate, ester or prodrug thereof, wherein R5 is a group of Formula II.

6. The compound of claim 5, wherein said R4 is a substituted or unsubstituted C C3 alkyl.

7. The compound of claim 5, wherein said R4 is a hydrogen atom. 8. The compound of any one of claims 1 to 7, wherein X1, X2 and X3 are all carbon atoms.

9. The compound of any one of claims 1 to 7, wherein X1 is a nitrogen atom and R10 is absent, and X2 and X3 are carbon atoms.

10. The compound of any one of claims 1 to 9, wherein R9 is an unsubstituted Ci-C3alkyl or C3-C5cycloalkyl group or a fluorinated CrC3alkyl group or C3-C5cycloalkyl group. 1 1. The compound of claim 10, wherein R9 is selected from methyl, ethyl, n-propyl, isopropyl and cyclopropyl.

12. The compound of claim 8, wherein said R7 and R10 are each hydrogen atoms and R8 is selected from CI, CN, and a substituted or unsubstituted Ci-C3alkyl, C3cycloalkyl, NHCi-C3alkyl, or NH(CrC3alkyl)2 group. 13. The compound of any one of claims 1 to 12, wherein said R3 is H or a substituted or unsubstituted CrC6alkyl group.

14. The compound of claim 13, wherein said R3 is H.

15. The compound of any one of claims 1 to 14, wherein said R6 is H or a substituted or unsubstituted CrC6alkyl group. 16. The compound of claim 15, wherein said R6 is H.

17. The compound of claim 1 , wher a compound of Formula III:

Formula III

wherein R1, R2, R4 and R5 are as defined in claim 1 , or a pharmaceutically acceptable salt, solvate, ester or prodrug thereof.

18. The compound of claim 17, wherein said compound is a compound of Formula lll(a):

Formula lll(a)

wherein,

R1, R2, R7, R8, R9, and R10 are as defined in claim 1 , or a pharmaceutically acceptable salt, solvate, ester or prodrug thereof.

The compound of claim 17, wherein said compound is a compound of Formula lll(b):

Formula lll(b)

wherein,

R1, R2, R7, R8, R9, and R10 are as defined in claim 1 , or a pharmaceutically acceptable salt, solvate, ester or prodrug thereof.

20. The compound of claim 18 or 19, wherein R9 is an unsubstituted d-C^alky! or C3- C5cycloalkyl group.

21. The compound of claim 18 or 19, wherein R9 is selected from methyl, trifluoromethyl, ethyl, n-propyl, isopropyl and cyclopropyl.

22. The compound of any one of claims 18 to 21 , wherein said R7 and R10 are each hydrogen atoms and R8 is selected from CI, CN, and a substituted or unsubstituted Ci-C3alkyl, C3cycloalkyl, NHC C3alkyl, or NH(C C3alkyl)2 group.

23. The compound of claim 18 or 19, wherein R8 and R9 are each independently a methyl, ethyl, isopropyl, fluoromethyl, difluoromethyl, trifluoromethyl, cyclopropyl, or difluorocyclopropyl group.

24. The compound of any one of claims 1 to 23, wherein R2 is hydrogen or a substituted or unsubstituted group selected from d-C6alkyl, C3-C10cycloalkyl, or C3-C10heterocycloalkyl group.

25. The compound of claim 24, wherein R2 is a substituted or unsubstituted Ci-C3alkyl, C3- C6cycloalkyl, or C3-C6heterocycloalkyl group. 26. The compound of claim 24, wherein R2 is a substituted or unsubstituted group selected from methyl, ethyl, propyl, isopropyl, butyl, isobutyl, tert-butyl, cyclopropyl, cyclobutyl, cyclopentyl, tetrahydrofuranyl, tetrahydropyranyl, dioxolanyl, piperidinyl, and pyrrolidinyl.

27. The compound of claim 25, wherein R2 is a substituted or unsubstituted methyl or ethyl group. 28. The compound of claim 24, wherein R2 is hydrogen.

29. The compound of any one of claims 1 to 28, wherein R1 is a branched or linear C C6alkyl group, unsubstituted or substituted with one or more group(s) selected from halogen, OH, NH2, CN, or a substituted or unsubstituted group selected from C3-Ci0cycloalkyl, C3- Cioheterocycloalkyl, C6-Ci0aryl, C5-Ci0heteroaryl, C(0)Ra, ORa, NHRa, N(Ra)2, C(0)NH2, C(0)NHRa, C(0)N(Ra)2, NHC(0)Ra, N(Ra)C(0)Ra, NHC(0)NHRa, N(Ra)C(0)NHRa, NHC(0)N(Ra)2, N(Ra)C(0)N(Ra)2, NHS02Ra, N(Ra)S02Ra, NHS02NHRa, N(Ra)S02NHRa, NHS02N(Ra)2, N(Ra)S02N(Ra)2, S02Ra, S02NH2, S02NHRa, and S02N(Ra)2, wherein Ra is as defined in claim 1.

30. The compound of claim 29, wherein R1 is a branched or linear d-C3alkyl group, unsubstituted or substituted with one or more group selected from halogen, OH, NH2, CN, or a substituted or unsubstituted group selected from C3-C10cycloalkyl, C3-C10heterocycloalkyl, C6- C10aryl, C5-C10heteroaryl, C(0)Ra, ORa, NHRa, N(Ra)2, C(0)NH2, C(0)NHRa, C(0)N(Ra)2, NHC(0)Ra, N(Ra)C(0)Ra, NHC(0)NHRa, N(Ra)C(0)NHRa, NHC(0)N(Ra)2, N(Ra)C(0)N(Ra)2, NHS02Ra, N(Ra)S02Ra, NHS02NHRa, N(Ra)S02NHRa, NHS02N(Ra)2, N(Ra)S02N(Ra)2, S02Ra, S02NH2, S02NHRa, and S02N(Ra)2, wherein Ra is as defined in claim 1.

31. The compound of claim 30, wherein R1 is a Cialkyl substituted with one or more substituted or unsubstituted group selected from C3-Ci0cycloalkyl, C3-Ci0heterocycloalkyl, C6- Ci0aryl, C5-Ci0heteroaryl, ORa, and NHRa, wherein Ra is as defined in claim 1.

32. The compound of any one of claims 1 to 28, wherein R1 is selected from C(0)Ra, ORa, NHRa, N(Ra)2, C(0)NHRa, C(0)N(Ra)2, S02Ra, S02NHRa, and S02N(Ra)2, wherein Ra is as defined in claim 1.

33. The compound of any one of claims 1 to 28, wherein R1 is selected from C(0)C3- doheterocycloalkyl, ORa, NHRa, N(Ra)2, C(0)NHRa, SO2(C3-C10heterocycloalkyl), and

S02NHRa, wherein Ra is as defined in claim 1.

34. The compound of any one of claims 1 to 33, wherein Ra is, independently in each occurrence selected from a substituted or unsubstituted CrC6alkyl, C3-Ci0cycloalkyl, C3- Cioheterocycloalkyl, C6-Ci0aryl, or C5-Ci0heteroaryl. 35. The compound of claim 1 , wherein the compound is selected from Compounds 1 to 56 as herein defined, or a pharmaceutically acceptable salt, solvate, or prodrug thereof.

36. A pharmaceutical composition, comprising a compound of any one of claims 1 to 35, together with a pharmaceutically acceptable carrier, diluent or excipient.

37. Use of a compound according to any one of claims 1 to 35 for the treatment of a disease or condition for which a bromodomain inhibitor is indicated.

38. The use according to claim 37, wherein the disease or condition is an auto-immune disorder, an inflammatory disorder, a dermal disorder, or cancer.

39. The use according to claim 37, wherein the disease or condition is an auto-immune disorder. 40. The use according to claim 37, wherein the disease or condition is an inflammatory disorder.

41. The use according to claim 40, wherein the inflammatory disorder is rheumatoid arthritis, irritable bowel syndrome, or psoriasis.

42. The use according to claim 37, wherein the disease or condition is cancer. 43. The use according to claim 42, wherein the cancer is brain cancer, pancreatic cancer, breast cancer, lung cancer, or prostate cancer.

44. The use according to claim 42, wherein the cancer is brain cancer.

45. The use according to claim 44, wherein the brain cancer is glioblastoma multiforme.

46. Use of a compound according to any one of claims 1 to 35 in the manufacture of a medicament for the treatment of a disease or condition for which a bromodomain inhibitor is indicated.

47. The use according to claim 46, wherein the disease or condition is an auto-immune disorder, an inflammatory disorder, a dermal disorder or cancer.

48. The use according to claim 46, wherein the disease or condition is an auto-immune disorder. 49. The use according to claim 46, wherein the disease or condition is an inflammatory disorder.

50. The use according to claim 49, wherein the inflammatory disorder is rheumatoid arthritis, irritable bowel syndrome, or psoriasis.

51. The use according to claim 46, wherein the disease or condition is cancer. 52. The use according to claim 51 , wherein the cancer is brain cancer, pancreatic cancer, breast cancer, lung cancer, or prostate cancer.

53. The use according to claim 51 , wherein the cancer is brain cancer.

54. The use according to claim 53, wherein the brain cancer is glioblastoma multiforme.

55. The use according to any one of claims 46 to 54, wherein said medicament is an oral medicament.

56. Use of a compound according to any one of claims 1 to 35 for the treatment of a disease or condition selected from auto-immune disorders, inflammatory disorders, dermal disorders, and neoplasms.

57. The use according to claim 56, wherein the disease or condition is an auto-immune disorder.

58. The use according to claim 56, wherein the disease or condition is an inflammatory disorder.

59. The use according to claim 58, wherein the inflammatory disorder is rheumatoid arthritis, irritable bowel syndrome, or psoriasis. 60. The use according to claim 56, wherein the disease or condition is a neoplasm.

61. The use according to claim 60, wherein the neoplasm is brain cancer, pancreatic cancer, breast cancer, lung cancer, or prostate cancer.

62. The use according to claim 60, wherein the neoplasm is brain cancer.

63. The use according to claim 62, wherein the brain cancer is glioblastoma multiforme. 64. A method for treating a disease or condition for which a bromodomain inhibitor is indicated, which comprises administering to a subject in need thereof, a therapeutically effective amount of a compound according to any one of claims 1 to 35.

65. The method according to claim 64, wherein the disease or condition is an auto-immune disorder, an inflammatory disorder, a dermal disorder or cancer. 66. The method according to claim 64, wherein the disease or condition is an auto-immune disorder.

67. The method according to claim 64, wherein the disease or condition is an inflammatory disorder.

68. The method according to claim 67, wherein the inflammatory disorder is rheumatoid arthritis, irritable bowel syndrome, or psoriasis.

69. The method according to claim 64, wherein the disease or condition is cancer.

70. The method according to claim 69, wherein the cancer is brain cancer, pancreatic cancer, breast cancer, lung cancer, or prostate cancer.

71. The method according to claim 69, wherein the cancer is brain cancer. 72. The method according to claim 71 , wherein the brain cancer is glioblastoma multiforme.

73. The method according to any one of claims 64 to 72, wherein said method comprises orally administering a therapeutically effective amount of the compound.

74. A method for inhibiting a bromodomain which comprises contacting the bromodomain with a compound of according to any one of claims 1 to 35. 75. A method for the treatment of a disease or condition selected from auto-immune disorders, inflammatory disorders, dermal disorders, and neoplasms, which comprises administering to a subject in need thereof, a therapeutically effective amount of a compound according to any one of claims 1 to 35.

76. The method of claim 75, wherein the disease or condition is an auto-immune disorder. 77. The method of claim 75, wherein the disease or condition is an inflammatory disorder.

78. The method of claim 77, wherein the inflammatory disorder is rheumatoid arthritis, irritable bowel syndrome, or psoriasis.

79. The method of claim 75, wherein the disease or condition is a neoplasm.

80. The method of claim 79, wherein the neoplasm is brain cancer, pancreatic cancer, breast cancer, lung cancer, or prostate cancer.

81. The method of claim 80, wherein the neoplasm is brain cancer.

82. The method of claim 81 , wherein the brain cancer is glioblastoma multiforme.

Description:
SUBSTITUTED IMIDAZOPYRIDINES, THEIR PREPARATION AND

THEIR USE AS PHARMACEUTICALS

TECHNICAL FIELD

The technical field generally compounds, compositions and their uses in the treatment of diseases and conditions in which inhibition of bromodomains is indicated. For example, the application relates to imidazopyridines, to pharmaceutical compositions comprising the same, and to their use as bromodomain inhibitors. The present application is also related to the treatment or prevention of proliferative disorders, auto-immune disorders, inflammatory disorders, dermal disorders, and neoplasms, including tumors and/or cancers. BACKGROUND

Bromodomains are found in a variety of mammalian DNA-binding proteins. The bromodomain, which is the conserved structural module in chromatin-associated proteins and histone acetyltransferases, is known to recognize acetyl-lysine residues on proteins. Bromodomain inhibitors are believed to be useful in the treatment of a variety of diseases or conditions, such as cancer as well as chronic autoimmune and inflammatory conditions. There is therefore a need for compounds that could inhibit bromodomains.

SUM MARY

According to one aspect, the present application relates to compounds of Formula I, and pharmaceutically acceptable salts, solvates, esters or prodrugs thereof:

Formula I

wherein,

R 1 is a substituted or unsubstituted group selected from d-C 6 alkyl, C 2 -C 6 alkenyl, C 2 - C 6 alkynyl, C 3 -C 10 cycloalkyl, C 3 -C 10 heterocycloalkyl, Cs-Ciocycloalkyld-Cealkyl-, C 3 - Ci 0 heterocycloalkylCi-C 6 alkyl-, Ce-CioarylCi-Cealkyl-, Cs-doheteroaryld-Cealkyl-, C(0)R a , OR a , NHR a , N(R a ) 2 , C(0)NH 2 , C(0)NHR a , C(0)N(R a ) 2 , NHC(0)R a , N(R a )C(0)R a , NHC(0)NHR a , N(R a )C(0)NHR a , NHC(0)N(R a ) 2 , N(R a )C(0)N(R a ) 2 , NHS0 2 R a , N(R a )S0 2 R a , NHS0 2 NHR a , N(R a )S0 2 NHR a , NHS0 2 N(R a ) 2 , N(R a )S0 2 N(R a ) 2 , S0 2 R a , S0 2 NH 2 , S0 2 NHR a , and S0 2 N(R a ) 2 ;

R a is, independently in each occurrence, a substituted or unsubstituted group selected from CrC 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -Ci 0 cycloalkyl, C 3 -Ci 0 heterocycloalkyl, C 3 - Ci 0 cycloalkylCi-C 6 alkyl-, C 3 -CioheterocycloalkylCi-C 6 alkyl-, C 6 -Ci 0 aryl, C 5 -Ci 0 heteroaryl, C 6 - Ci 0 arylCi-C 6 alkyl-, and C 5 -CioheteroarylCi-C 6 alkyl-;

R 2 is selected from H, NH 2 , CN or a substituted or unsubstituted group selected from C C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -Ci 0 cycloalkyl, C 3 -Ci 0 heterocycloalkyl, C 6 -Ci 0 aryl, C 5 - Cioheteroaryl, C(0)R a , OR a , SR a , NHR a , N(R a ) 2 , C(0)NH 2 , C(0)NHR a , C(0)N(R a ) 2 , NHC(0)R a , S0 2 R a , S0 2 NHR a , S0 2 N(R a ) 2 , NHS0 2 R a , N(R a )S0 2 R a , NHS0 2 NHR a , N(R a )S0 2 NHR a , NHS0 2 N(R a ) 2 , and N(R a )S0 2 N(R a ) 2 ;

R 3 and R 6 are each independently H, halogen, NH 2 , CN or a substituted or unsubstituted group selected from C C 6 alkyl, C(0)R a , OR a , NHR a , N(R a ) 2 , C(0)NH 2 , C(0)NHR a , C(0)N(R a ) 2 , NHC(0)R a ; and

one of R 4 and R 5 is H, halogen, NH 2 , CN or a substituted or unsubstituted group selected from C C 6 alkyl, C(0)R a , NH 2 , NHR a , N(R a ) 2 , C(0)NH 2 , C(0)NHR a , C(0)N(R a ) 2 , and NHC(0)R a ; and the other of R 4 and R 5 is a group of Formula II:

Formula II

wherein,

R 7 and R 10 are each independently H, halogen (such as F, CI), CN, or a substituted or unsubstituted group selected from CrC 6 alkyl or C 3 -C 6 cycloalkyl group, OCrC 6 alkyl, OC 3 - Cecycloalkyl, SC C 6 alkyl, SC 3 -C 6 cycloalkyl, NHC C 6 alkyl, NHC 3 -C 6 cycloalkyl, N(C C 6 alkyl) 2 , NHC(0)C C 6 alkyl and NHC(0)C 3 -C 6 cycloalkyl; R is halogen (such as F, CI), CN, or a substituted or unsubstituted group selected from C C 6 alkyl or C 3 -C 6 cycloalkyl group, OC C 6 alkyl, OC 3 -C 6 cycloalkyl, SC C 6 alkyl, SC 3 - Cecycloalkyl, NHC C 6 alkyl, NHC 3 -C 6 cycloalkyl, N(C C 6 alkyl)2, NHC(0)C C 6 alkyl and NHC(0)C 3 -C 6 cycloalkyl;

R 9 is a substituted or unsubstituted C C 3 alkyl or C 3 -C 5 cycloalkyl group; and

X 1 , X 2 , and X 3 are each selected from a nitrogen or carbon atom, wherein when X 1 , X 2 , or X 3 is a nitrogen atom, then the R 7 , R 8 , or R 10 attached thereto is absent, provided that at least two of X 1 , X 2 , and X 3 are C.

In one embodiment, the compound is of Formula I, wherein R 4 is a group of Formula I I, preferably wherein said R 5 is hydrogen or a substituted or unsubstituted C C 3 alkyl. According to another embodiment, the compound is of Formula I, wherein R 5 is a group of Formula II, preferably wherein said R 4 is hydrogen or a substituted or unsubstituted C C 3 alkyl.

In one embodiment, in Formula II, groups X 1 , X 2 and X 3 are all carbon atoms. In another embodiment, X 1 is a nitrogen atom and R 10 is absent, and X 2 and X 3 are carbon atoms. For instance, the group of Formula II may be defined as a group of Formula ll(a):

Formula ll(a)

wherein R 7 , R 8 , R 9 and R 10 are as herein defined.

In one embodiment, R 9 is an unsubstituted Ci-C 3 alkyl or C 3 -C 5 cycloalkyl group or a fluorinated Ci-C 3 alkyl group or C 3 -C 5 cycloalkyl group, for instance trifluoromethyl, a methyl, ethyl, n-propyl, isopropyl or cyclopropyl group. In another embodiment, R 7 and R 10 are each hydrogen atoms and R 8 is selected from CI, CN, and a substituted or unsubstituted Ci-C 3 alkyl, C 3 cycloalkyl, NHC C 3 alkyl, or NH(C C 3 alkyl) 2 group.

Another embodiment of the application relates to compounds of Formula I (a), and pharmaceutically acceptable salts, solvates, esters or prodrugs thereof:

Formula 1(a)

wherein,

R 1 , R 2 , R 3 , R 5 , R 6 , R 7 , R 8 , R 9 , and R 10 are as herein defined.

According to another embodiment, the application also relates to compounds of Formula l(b), and pharmaceutically acceptable salts, solvates, esters or prodrugs thereof:

Formula l(b)

wherein,

R 1 , R 2 , R 3 , R 4 , R 6 , R 7 , R 8 , R 9 , and R 10 are as herein defined.

In one embodiment, the application relates to compounds as herein defined wherein R 3 is H or a substituted or unsubstituted d-C 6 alkyl group, for example, R 3 is H. In another embodiment, the application relates to compounds as herein defined wherein R 6 is H or a substituted or unsubstituted CrCealkyl group, for instance, R 6 is H.

In another embodiment, the present application relates to compounds of Formula III, and pharmaceutically acceptable salts, solvates, esters or prodrugs thereof:

Formula III

wherein R 1 , R 2 , R 4 and R 5 are as herein defined.

Another embodiment of the application relates to compounds of Formula lll(a), and pharmaceutically acceptable salts, solvates, esters or prodrugs thereof:

Formula lll(a)

wherein R 1 , R 2 , R 7 , R 8 , R 9 , and R 10 are as herein defined.

According to another embodiment, the application also relates to compounds of Formula lll(b), and pharmaceutically acceptable salts, solvates, esters or prodrugs thereof:

Formula lll(b)

wherein R 1 , R 2 , R 7 , R 8 , R 9 , and R 10 are as herein defined.

According to one embodiment, the application relates to compounds of Formula lll(a) or lll(b) as herein defined, wherein R 9 is an unsubstituted d-C 3 alkyl or C 3 -C 5 cycloalkyl group. In another embodiment, the application relates to compounds of Formula lll(a) or Formula lll(b) as herein defined, wherein R 9 is selected from methyl, trifluoromethyl, ethyl, n-propyl, isopropyl and cyclopropyl. In another embodiment, R 8 and R 9 are each independently a methyl, ethyl, isopropyl, fluoromethyl, difluoromethyl, trifluoromethyl, cyclopropyl, or difluorocyclopropyl group

According to another embodiment, the application relates to compounds of Formula 111(a) or 111(b) as herein defined, wherein said R 7 and R 10 are each hydrogen atoms and R 8 is selected from CI, CN, and a substituted or unsubstituted Ci-C 3 alkyl, C 3 cycloalkyl, NHCrC 3 alkyl, or NH(Ci-C 3 alkyl) 2 group.

In yet another embodiment, the present application relates to a compound of Formula I, l(a), l(b), III, lll(a) or lll(b) as herein defined, wherein R 2 is hydrogen or a substituted or unsubstituted group selected from CrC 6 alkyl, C 3 -Ci 0 cycloalkyl, or C 3 -Ci 0 heterocycloalkyl group. For instance, R 2 is a substituted or unsubstituted Ci-C 3 alkyl, C 3 -C 6 cycloalkyl, or C 3 -C 6 heterocycloalkyl group, or R 2 is a substituted or unsubstituted group selected from methyl, ethyl, propyl, isopropyl, butyl, isobutyl, tert-butyl, cyclopropyl, cyclobutyl, cyclopentyl, tetrahydrofuranyl, tetrahydropyranyl, dioxolanyl, piperidinyl, and pyrrolidinyl, preferably, R 2 is hydrogen or a substituted or unsubstituted methyl or ethyl group.

In a further embodiment, the present application relates to a compound of Formula I, l(a), l(b), III, lll(a) or lll(b) as herein defined, wherein R 1 is a branched or linear d-C^alky! group, unsubstituted or substituted with one or more group(s) selected from halogen, OH, NH 2 , CN, or a substituted or unsubstituted group selected from C 3 -C 10 cycloalkyl, C 3 -C 10 heterocycloalkyl, C 6 - C 10 aryl, C 5 -C 10 heteroaryl, C(0)R a , OR a , NHR a , N(R a ) 2 , C(0)NH 2 , C(0)NHR a , C(0)N(R a ) 2 , NHC(0)R a , N(R a )C(0)R a , NHC(0)NHR a , N(R a )C(0)NHR a , NHC(0)N(R a ) 2 , N(R a )C(0)N(R a ) 2 , NHS0 2 R a , N(R a )S0 2 R a , NHS0 2 NHR a , N(R a )S0 2 NHR a , NHS0 2 N(R a ) 2 , N(R a )S0 2 N(R a ) 2 , S0 2 R a , S0 2 NH 2 , S0 2 NHR a , S0 2 N(R a ) 2 , , wherein R a is as herein defined. In another embodiment, R 1 is a branched or linear C C 3 alkyl group, unsubstituted or substituted with one or more group(s) selected from halogen, OH, NH 2 , CN, or a substituted or unsubstituted group selected from C 3 - Ciocycloalkyl, C 3 -Ci 0 heterocycloalkyl, C 6 -Ci 0 aryl, C 5 -Ci 0 heteroaryl, C(0)R a , OR a , NHR a , N(R a ) 2 , C(0)NH 2 , C(0)NHR a , C(0)N(R a ) 2 , NHC(0)R a , N(R a )C(0)R a , NHC(0)NHR a , N(R a )C(0)NHR a , NHC(0)N(R a ) 2 , N(R a )C(0)N(R a ) 2 , NHS0 2 R a , N(R a )S0 2 R a , NHS0 2 NHR a , N(R a )S0 2 NHR a , NHS0 2 N(R a ) 2 , N(R a )S0 2 N(R a ) 2 , S0 2 R a , S0 2 NH 2 , S0 2 NHR a , S0 2 N(R a ) 2 , wherein R a is as herein defined. In a further embodiment, R 1 is a Ci-C 2 alkyl substituted with one or more group(s) selected from halogen or a substituted or unsubstituted group selected from C 3 -Ci 0 cycloalkyl, C 3 -Cioheterocycloalkyl, C 6 -Ci 0 aryl, C 5 -Ci 0 heteroaryl, C(0)R a , OR a , NHR a , N(R a ) 2 , C(0)NH 2 , C(0)NHR a , C(0)N(R a ) 2 , NHC(0)R a , N(R a )C(0)R a , NHS0 2 R a , N(R a )S0 2 R a , NHS0 2 NHR a , N(R a )S0 2 NHR a , NHS0 2 N(R a ) 2 , N(R a )S0 2 N(R a ) 2 , S0 2 R a , S0 2 NH 2 , S0 2 NHR a , S0 2 N(R a ) 2 , wherein R a is as herein defined. In yet another embodiment, R 1 is a dalkyl substituted with one or more substituted or unsubstituted group selected from C 3 -C 10 cycloalkyl, C 3 - C 10 heterocycloalkyl, C 6 -C 10 aryl, C 5 -C 10 heteroaryl, OR a , and NHR a , wherein R a is as herein defined. In a further embodiment, R 1 is selected from C(0)R a , OR a , NHR a , N(R a ) 2 , C(0)NHR a , C(0)N(R a ) 2 , S0 2 R a , S0 2 NHR a , and S0 2 N(R a ) 2 , wherein R a is as herein defined. In yet another embodiment, R 1 is selected from C(O)C 3 -Ci 0 heterocycloalkyl, OR a , NHR a , N(R a ) 2 , C(0)NHR a , S0 2 (C 3 -Cioheterocycloalkyl), and S0 2 NHR a , wherein R a is as herein defined.

In one embodiment, R a is, independently in each occurrence, selected from a substituted or unsubstituted CrC 6 alkyl, C 3 -Ci 0 cycloalkyl, C 3 -Ci 0 heterocycloalkyl, C 6 -Ci 0 aryl, or C 5 - Cioheteroaryl.

In a further embodiment, this application relates to a compound selected from Compounds 1 to 56 as herein defined, or a pharmaceutically acceptable salt, solvate, or prodrug thereof, for instance, any of these compounds or their isomers, or a pharmaceutically acceptable salt, solvate, or prodrug thereof, may be taken individually or as sub-groups.

Another aspect related to pharmaceutical composition, comprising a compound as defined in the present application, together with a pharmaceutically acceptable carrier, diluent or excipient.

A further aspect relates to the use of a compound as defined in the present application, or such a compound for use, in the treatment or prevention of a disease or condition for which a bromodomain inhibitor is indicated. Similarly, this aspect relates to the use of a compound of the present application in the manufacture of a medicament for the treatment or prevention of a disease or condition for which a bromodomain inhibitor is indicated. This aspect also further relates to a method for treating a disease or condition for which a bromodomain inhibitor is indicated, which comprises administering to a subject in need thereof, a therapeutically effective amount of a compound as herein defined. In one embodiment, the disease or condition for which a bromodomain inhibitor is indicated is an auto-immune disorder, an inflammatory disorder (such as rheumatoid arthritis, irritable bowel syndrome, or psoriasis), a dermal disorder, or cancer (for instance, brain cancer, pancreatic cancer, breast cancer, lung cancer, or prostate cancer). For instance the disease or condition is brain cancer, such as glioblastoma multiforme.

According to a further aspect, the application relates to the use of a compound as herein defined, or such a compound for use, in the treatment of a disease or condition selected from auto-immune disorders, inflammatory disorders, dermal disorders, and neoplasms. This aspect also relates to a method for the treatment or prevention of a disease or condition selected from auto-immune disorders, inflammatory disorders, dermal disorders, and neoplasms, which comprises administering to a subject in need thereof, a therapeutically effective amount of a compound as herein defined. For instance, the inflammatory disorder is rheumatoid arthritis, irritable bowel syndrome, or psoriasis. For example, the disease or condition is a neoplasm which is brain cancer (e.g. glioblastoma multiforme), pancreatic cancer, breast cancer, lung cancer, or prostate cancer.

Additional objects and features of the present compounds, compositions, methods and uses will become more apparent upon reading of the following non-restrictive description of exemplary embodiments, which should not be interpreted as limiting the scope of the invention.

DESCRIPTION

All technical and scientific terms used herein have the same meaning as commonly understood by one ordinary skilled in the art to which the present technology pertains. For convenience, the meaning of certain terms and phrases used herein are provided below.

To the extent the definitions of terms in the publications, patents, and patent applications incorporated herein by reference are contrary to the definitions set forth in this specification, the definitions in this specification control. The section headings used herein are for organizational purposes only, and are not to be construed as limiting the subject matter disclosed.

/ ' . Definitions

The terminology used herein is for the purpose of describing particular embodiments only and is not intended to be limiting. It should be noted that, the singular forms "a", "an", and "the" include plural forms as well, unless the content clearly dictates otherwise. Thus, for example, reference to a composition containing "a compound" also contemplates a mixture of two or more compounds. It should also be noted that the term "or" is generally employed in its sense including "and/or" unless the content clearly dictates otherwise. Furthermore, to the extent that the terms "including", "includes", "having", "has", "with", or variants thereof are used in either the detailed description and/or the claims, such terms are intended to be inclusive in a manner similar to the term "comprising".

The term "about" or "approximately" means within an acceptable error range for the particular value as determined by one of ordinary skill in the art, which will depend in part on how the value is measured or determined, i.e., the limitations of the measurement system. For example, "about" can mean within 1 or more than 1 standard deviation, per the practice in the art. Alternatively, "about" can mean a range of up to 20%, preferably up to 10%, more preferably up to 5%, and more preferably still up to 1 % of a given value. Alternatively, particularly with respect to biological systems or processes, the term can mean within an order of magnitude, preferably within 5-fold, and more preferably within 2-fold, of a value. Where particular values are described in the application and claims, unless otherwise stated the term "about" meaning within an acceptable error range for the particular value should be assumed.

As used herein, the terms "compounds herein described", "compounds of the present application" and equivalent expressions refer to compounds described in the present application, e.g., those encompassed by structural Formulae such as Formula I, l(a), l(b), III, lll(a), and lll(b), optionally with reference to any of the applicable embodiments, and also includes exemplary compounds, for example, Compounds 1-56, as well as their pharmaceutically acceptable salts, solvates, esters, and prodrugs when applicable. When a zwitterionic form is possible, the compound may be drawn as its neutral form for practical purposes, but the compound is understood to also include its zwitterionic form. Embodiments herein may also exclude one or more of the compounds. Compounds may be identified either by their chemical structure or their chemical name. In a case where the chemical structure and chemical name would conflict, the chemical structure will prevail.

Unless otherwise stated, structures depicted herein are also meant to include all isomeric (e.g., enantiomeric, diastereomeric, and geometric (or conformational)) forms of the structure; for example, the R and S configurations for each asymmetric center, Z and E double bond isomers, and Z and E conformational isomers. Therefore, single stereochemical isomers as well as enantiomeric, diastereomeric, and geometric (or conformational) mixtures of the present compounds are within the scope of the present description. Unless otherwise stated, all tautomeric forms of the compounds are within the scope of the present description. Additionally, unless otherwise stated, structures depicted herein are also meant to include compounds that differ only in the presence of one or more isotopically enriched atoms. For example, compounds having the present structures including the replacement of hydrogen by deuterium or tritium, or the replacement of a carbon by a 13 C- or 1 C-enriched carbon are within the scope of the present description. Such compounds are useful, for example, as analytical tools, as probes in biological assays, or as therapeutic agents in accordance with the present description.

Where a particular enantiomer is preferred, it may, in some embodiments be provided substantially free of the corresponding enantiomer, and may also be referred to as "optically enriched." "Optically-enriched," as used herein, means that the compound is made up of a significantly greater proportion of one enantiomer. In certain embodiments the compound is made up of at least about 90% by weight of a preferred enantiomer. In other embodiments the compound is made up of at least about 95%, 98%, or 99% by weight of a preferred enantiomer. Preferred enantiomers may be isolated from racemic mixtures by any method known to those skilled in the art, including chiral high pressure liquid chromatography (HPLC) and the formation and crystallization of chiral salts or prepared by asymmetric syntheses. See, for example, Jacques et al., Enantiomers, Racemates and Resolutions (Wiley Interscience, New York, 1981); Wilen, et al., Tetrahedron 33:2725 (1977); Eliel, E.L. Stereochemistry of Carbon Compounds (McGraw-Hill, NY, 1962); Wilen, S.H. Tables of Resolving Agents and Optical Resolutions, p. 268 (E.L. Eliel, Ed. , Univ. of Notre Dame Press, Notre Dame, IN 1972).

Definitions of specific functional groups and chemical terms are described in more detail below. For purposes of the present description, the chemical elements are identified in accordance with the Periodic Table of the Elements, CAS version, Handbook of Chemistry and Physics, 75 th , Ed., inside cover, and specific functional groups are generally defined as described therein. Additionally, general principles of organic chemistry, as well as specific functional moieties and reactivity, are described in Organic Chemistry, Thomas Sorrell, University Science Books, Sausalito, 1999; Smith and March March's Advanced Organic Chemistry, 5 th , Edition, John Wiley & Sons, Inc., New York, 2001 ; Larock, Comprehensive Organic Transformations, VCH Publishers, Inc., New York, 1989; Carruthers, Some Modern Methods of Organic Synthesis, 3 rd Edition, Cambridge University Press, Cambridge, 1987.

The number of carbon atoms in a hydrocarbyl substituent can be indicated by the prefix "C x -C y ," where x is the minimum and y is the maximum number of carbon atoms in the substituent. However, when the prefix "C x -C y " is associated with a group incorporating one or more heteroatom(s) by definition (e.g. heterocycloalkyl, heteroaryl, etc), then x and y define respectively the minimum and maximum number of atoms in the cycle, including carbons as well as heteroatom (s).

The prefix "halo" indicates that the substituent to which the prefix is attached is substituted with one or more independently selected halogen radicals. More specifically, the terms "halo" and "halogen" as used herein refer to an atom selected from fluorine (fluoro, -F), chlorine (chloro, - CI), bromine (bromo, -Br), and iodine (iodo, -I). For example, "haloalkyl" means an alkyl substituent wherein at least one hydrogen radical is replaced with a halogen radical.

The term "heteroatom" means one or more of oxygen, sulfur, nitrogen, phosphorus, or silicon (including, any oxidized form of nitrogen, sulfur, phosphorus, or silicon; the quaternized form of any basic nitrogen or; a substitutable nitrogen of a heterocyclic ring, for example N (as in 3,4- dihydro-2H-pyrrolyl), NH (as in pyrrolidinyl) or NR+ (as in N-substituted pyrrolidinyl).

As used herein a "direct bond" or "covalent bond" refers to a single, double or triple bond. In certain embodiments, a "direct bond" or "covalent bond" refers to a single bond.

Abbreviations may also be used throughout the application, unless otherwise noted, such abbreviations are intended to have the meaning generally understood by the field. Examples of such abbreviations include Me (methyl), Et (ethyl), Pr (propyl), i-Pr (isopropyl), Bu (butyl), t-Bu (tert-butyl), i-Bu (iso-butyl), s-Bu (sec-butyl), c-Bu (cyclobutyl), Ph (phenyl), Bn (benzyl), Bz (benzoyl), CBz or Cbz or Z (carbobenzyloxy), Boc or BOC (tert-butoxycarbonyl), and Su or Sue (succinimide). For greater certainty, examples of abbreviations used in the present application are listed in a table in the Examples section.

The chemical structures herein are drawn according to the conventional standards known in the art. Thus, where an atom, such as a carbon atom, as drawn appears to have an unsatisfied valency, then that valency is assumed to be satisfied by a hydrogen atom even though that hydrogen atom is not necessarily explicitly drawn. Hydrogen atoms should be inferred to be part of the compound.

The term "aliphatic" or "aliphatic group", as used herein, denotes a hydrocarbon moiety that may be straight-chain (i.e. , unbranched), branched, or cyclic (including fused, bridging, and spiro-fused polycyclic) and may be completely saturated or may contain one or more units of unsaturation, but which is not aromatic. Unless otherwise specified, aliphatic groups contain 1-6 carbon atoms. In some embodiments, aliphatic groups contain 1-4 carbon atoms, and in yet other embodiments aliphatic groups contain 1-3 carbon atoms. Aliphatic groups include, but are not limited to, alkyl, alkenyl, alkynyl, carbocycle. Suitable aliphatic groups include, but are not limited to, linear or branched, alkyl, alkenyl, and alkynyl groups, and hybrids thereof such as (cycloalkyl)alkyl, (cycloalkenyl)alkyl or (cycloalkyl)alkenyl.

The term "alkyl" as used herein, refers to a saturated, straight- or branched-chain hydrocarbon radical typically containing from 1 to 20 carbon atoms. For example, "Ci-C 8 alkyl" contains from one to eight carbon atoms. Examples of alkyl radicals include, but are not limited to, methyl, ethyl, propyl, isopropyl, w-butyl, tert-butyl, neopentyl, n-hexyl, heptyl, octyl radicals and the like. The term "alkenyl" as used herein, denotes a straight- or branched-chain hydrocarbon radical containing one or more double bonds and typically from 2 to 20 carbon atoms. For example, "C 2 -C 8 alkenyl" contains from two to eight carbon atoms. Alkenyl groups include, but are not limited to, for example, ethenyl, propenyl, butenyl, l-methyl-2-buten-l-yl, heptenyl, octenyl and the like.

The term "alkynyl" as used herein, denotes a straight- or branched-chain hydrocarbon radical containing one or more triple bonds and typically from 2 to 20 carbon atoms. For example, "C 2 - C 8 alkynyl" contains from two to eight carbon atoms. Representative alkynyl groups include, but are not limited to, for example, ethynyl,1-propynyl, 1-butynyl, heptynyl, octynyl and the like.

The terms "cycloalkyi", "alicyclic", "carbocyclic" and equivalent expressions refer to a group comprising a saturated or partially unsaturated (non aromatic) carbocyclic ring in a monocyclic or polycyclic ring system, including spiro (sharing one atom), fused (sharing at least one bond) or bridged (sharing two or more bonds) carbocyclic ring systems, having from three to fifteen ring members. Examples of cycloalkyi groups include, without limitation, cyclopropyl, cyclobutyl, cyclopentyl, cyclopenten-1-yl, cyclopenten-2-yl, cyclopenten-3-yl, cyclohexyl, cyclohexen-1-yl, cyclohexen-2-yl, cyclohexen-3-yl, cycloheptyl, bicyclo[4,3,0]nonanyl, norbornyl, and the like. The term cycloalkyi includes both unsubstituted cycloalkyi groups and substituted cycloalkyi groups. The term "C 3 -C n cycloalkyl" refers to a cycloalkyi group having from 3 to the indicated "n" number of carbon atoms in the ring structure. Unless the number of carbons is otherwise specified, "lower cycloalkyi" groups as herein used, have at least 3 and equal or less than 8 carbon atoms in their ring structure.

As used herein, the terms "heterocycle", "heterocycloalkyl", "heterocyclyl", "heterocyclic radical", and "heterocyclic ring" are used interchangeably and refer to a chemically stable 3- to 7- membered monocyclic or 7-10-membered bicyclic heterocyclic moiety that is either saturated or partially unsaturated, and having, in addition to carbon atoms, one or more, preferably one to four, heteroatoms, as defined above. When used in reference to a ring atom of a heterocycle, the term "nitrogen" includes a substituted nitrogen. As an example, in a saturated or partially unsaturated ring having 1-3 heteroatoms selected from oxygen, sulfur or nitrogen, the nitrogen may be N (as in 3,4-dihydro-2H-pyrrolyl), NH (as in pyrrolidinyl), or +NR (as in N-substituted pyrrolidinyl). A heterocyclic ring can be attached to its pendant group at any heteroatom or carbon atom that results in a chemically stable structure and any of the ring atoms can be optionally substituted. Examples of heterocycloalkyl groups include, but are not limited to, 1 ,3- dioxolanyl, pyrrolidinyl, pyrrolidonyl, pyrazolinyl, pyrazolidinyl, imidazolinyl, imidazolidinyl, piperidinyl, piperazinyl, oxazolidinyl, isoxazolidinyl, morpholinyl, thiazolidinyl, isothiazolidinyl, tetrahydrofuranyl, tetrahydropyranyl, tetrahydrothiopyranyl, tetrahydrodithienyl, tetrahydrothienyl, thiomorpholino, thioxanyl, azetidinyl, oxetanyl, thietanyl, homopiperidinyl, oxepanyl, thiepanyl, oxazepinyl, diazepinyl, thiazepinyl, 1 ,2,3,6-tetrahydropyridinyl, 2-pyrrolinyl, 3-pyrrolinyl, 2H-pyranyl, 4H-pyranyl, dioxanyl, dithianyl, dithiolanyl, dihydropyranyl, dihydrothienyl, dihydrofuranyl, 3-azabicyclo[3, 1 ,0]hexanyl, 3-azabicyclo[4, 1 ,0]heptanyl, quinolizinyl, quinuclidinyl, tetrahydroquinolinyl, tetrahydroisoquinolinyl, decahydroquinolinyl, and the like. Heterocyclic groups also include groups in which a heterocyclic ring is fused to one or more aryl, heteroaryl, or cycloaliphatic rings, such as indolinyl, 3H-indolyl, chromanyl, chromenyl, phenanthridinyl, 2-azabicyclo[2.2.1]heptanyl, octahydroindolyl, or tetrahydroquinolinyl, where the radical or point of attachment is on the heterocyclyl ring. A heterocyclyl group may be mono- or bicyclic. The term "heterocyclylalkyl" refers to an alkyl group substituted by a heterocyclyl, wherein the alkyl and heterocyclyl portions independently are optionally substituted.

As used herein, the term "partially unsaturated" refers to a ring moiety that includes at least one double or triple bond between ring atoms but is not aromatic. The term "partially unsaturated" is intended to encompass rings having multiple sites of unsaturation, but is not intended to include aryl or heteroaryl moieties, as herein defined.

The term "aryl" used alone or as part of a larger moiety as in "aralkyl", "aralkoxy", or "aryloxyalkyl", refers to a monocyclic moiety or to a bicyclic or tricyclic fused ring system having a total of six to 15 ring members, wherein at least one ring in the system is aromatic and wherein each ring in the system contains three to seven ring members. The term "aryl" may be used interchangeably with the term "aryl ring". In certain embodiments of the present description, "aryl" refers to an aromatic ring system which includes, but not limited to, phenyl, biphenyl, naphthyl, azulenyl, anthracyl and the like, which may bear one or more substituents. The term "aralkyl" or "arylalkyl" refers to an alkyl residue attached to an aryl ring. Examples of aralkyl include, but are not limited to, benzyl, phenethyl, 1-phenylethyl, and the like. Also included within the scope of the term "aryl", as it is used herein, is a group in which an aromatic ring is fused to one or more non-aromatic rings, such as indanyl, indenyl, phthalimidyl, naphthimidyl, fluorenyl, phenanthridinyl, or tetrahydronaphthyl, and the like.

The terms "heteroaryl" and "heteroar-", used alone or as part of a larger moiety, e.g., "heteroaralkyl", or "heteroaralkoxy", refer to groups having 5 to 18 ring atoms, preferably 5, 6, or 9 ring atoms; having 6, 10, or 14 π electrons shared in a cyclic array; and having, in addition to carbon atoms, from one to five heteroatoms. The term "heteroatom" includes but is not limited to nitrogen, oxygen, or sulfur, and includes any oxidized form of nitrogen or sulfur, and any quaternized form of a basic nitrogen. A heteroaryl may be a single ring, or two or more fused rings. Heteroaryl groups include, without limitation, thienyl, furanyl (furyl), thienyl, pyrrolyl, imidazolyl, pyrazolyl, triazolyl, tetrazolyl, oxazolyl, isoxazolyl, oxadiazolyl, thiazolyl, isothiazolyl, thiadiazolyl, pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, triazinyl, furopyridinyl, indolizinyl, purinyl, naphthyridinyl, and pteridinyl. The terms "heteroaryl" and "heteroar-", as used herein, also include groups in which a heteroaromatic ring is fused to one or more aryl, cycloaliphatic, or heterocyclyl rings, where the radical or point of attachment is on the heteroaromatic ring. Nonlimiting examples include indolyl, 3H-indolyl, isoindolyl, benzothienyl (benzothiophenyl), benzofuranyl, dibenzofuranyl, indazolyl, benzimidazolyl, benzoxazolyl, benzothiazolyl, quinolyl (quinolinyl), isoquinolyl (isoquinolinyl), quinolonyl, isoquinolonyl, cinnolinyl, phthalazinyl, quinazolinyl, quinoxalinyl, 4H-quinolizinyl, carbazolyl, acridinyl, phenanthridinyl, phenazinyl, phenothiazinyl, phenoxazinyl, tetrahydroquinolinyl, tetrahydroisoquinolinyl, and pyrido[2,3-b]-l,4- oxazin-3(4H)-one. A heteroaryl group may be mono- or bicyclic. Heteroaryl groups include rings that are optionally substituted. The term "heteroaralkyl" refers to an alkyl group substituted by a heteroaryl, wherein the alkyl and heteroaryl portions independently are optionally substituted. Examples include, but are not limited to, pyridinylmethyl, pyrimidinylethyl and the like.

The term "bivalent hydrocarbon" refers to a bivalent saturated or unsaturated hydrocarbon group. Such bivalent hydrocarbon groups include alkylene, alkenylene, and alkynylene groups.

The term "alkylene" refers to a divalent group derived from a straight or branched saturated hydrocarbyl chain typically containing from 1 to 20 carbon atoms, more typically from 1 to 8 carbon atoms. Examples of an "alkylene" include a polymethylene group, i.e., -(CH 2 )n-, wherein n is a positive integer, preferably from 1 to 6, from 1 to 4, from 1 to 3, from 1 to 2, or from 2 to 3; or -CH 2 -, -CH2CH2-, -CH2CH2CH2-, -CH2CH2CH2CH2-, and -CH 2 CH(CH 3 )CH 2 -. A substituted alkylene chain is a polymethylene group in which one or more methylene hydrogen atoms are replaced with a substituent. Suitable substituents include those described below for a substituted aliphatic group.

The term "alkenylene" refers to a divalent unsaturated hydrocarbyl group which may be linear or branched and which has at least one carbon-carbon double bond. An alkenylene group typically contains 2 to 20 carbon atoms, more typically from 2 to 8 carbon atoms. Non-limiting examples of alkenylene groups include -C(H)=C(H)-, -C(H)=C(H)-CH 2 -, -C(H)=C(H)-CH 2 -CH 2 -, -CH 2 - C(H)=C(H)-CH 2 -, -C(H)=C(H)-CH(CH 3 )-, and -CH 2 -C(H)=C(H)-CH(CH 2 CH 3 )-.

The term "alkynylene" refers to a divalent unsaturated hydrocarbon group which may be linear or branched and which has at least one carbon-carbon triple bond. Examples of alkynylene groups include, without limitation, -C≡C-, -C≡C-CH 2 -, -C≡C-CH 2 -CH 2 -, -CH 2 -C≡C-CH 2 -, -C≡C- CH(CH 3 )-, and -CH 2 -C≡C-CH(CH 2 CH 3 )-.

As described herein, compounds of the present description may contain "optionally substituted" moieties. In general, the term "substituted", whether preceded by the term "optionally" or not, means that one or more hydrogens of the designated moiety are replaced with a suitable substituent. Unless otherwise indicated, an "optionally substituted" group may have a suitable substituent at each substitutable position of the group, and when more than one position in any given structure may be substituted with more than one substituent selected from a specified group, the substituent may be either the same or different at each position. Combinations of substituents envisioned under the present description are preferably those that result in the formation of chemically stable or chemically feasible compounds. The term "chemically stable", as used herein, refers to compounds that are not substantially altered when subjected to conditions to allow for their production, detection, and, in certain embodiments, their recovery, purification, and use for one or more of the purposes disclosed herein.

The terms "optionally substituted", "optionally substituted alkyl," "optionally substituted alkenyl," "optionally substituted alkynyl", "optionally substituted carbocyclic," "optionally substituted aryl", " optionally substituted heteroaryl," "optionally substituted heterocyclic," and any other optionally substituted group as used herein, refer to groups that are substituted or unsubstituted by independent replacement of one, two, or three or more of the hydrogen atoms thereon with substituents including, but not limited to F, CI, Br, I, OH, C0 2 H, alkoxy, oxo, thiooxo, N0 2 , CN, CF 3 , NH 2 , protected amino, NHalkyl, NHalkenyl, NHalkynyl, NHcycloalkyl, NHaryl, NHheteroaryl, NHheterocyclic, dialkylamino, diarylamino, diheteroarylamino, O-alkyl, O-alkenyl, O-alkynyl, O-cycloalkyl, O-aryl, O-heteroaryl, O-haloalkyl, O- heterocyclic, C(0)alkyl, C(0)alkenyl, C(0)alkynyl, C(0)cycloalkyl, C(0)aryl, C(O) heteroaryl, C(0)heterocycloalkyl, C0 2 alkyl, C0 2 alkenyl, C0 2 alkynyl, C0 2 cycloalkyl, C0 2 aryl, C0 2 heteroaryl, C0 2 heterocycloalkyl, OC(0)alkyl, OC(0)alkenyl, OC(0)alkynyl, OC(0)cycloalkyl, OC(0)aryl, OC(O) heteroaryl, OC(0)heterocycloalkyl, C(0)NH 2 , C(0)NHalkyl, C(0)NHalkenyl, C(0)NHalkynyl, C(0)NHcycloalkyl, C(0)NHaryl, C(O) NHheteroaryl, C(0)NHheterocycloalkyl, OC0 2 alkyl, OC0 2 alkenyl, OC0 2 alkynyl, OC0 2 cycloalkyl, OC0 2 aryl, OC0 2 heteroaryl, OC0 2 heterocycloalkyl, OC(0)NH 2 , OC(0)NHalkyl, OC(O) NHalkenyl, OC(0)NHalkynyl, OC(0)NHcycloalkyl, OC(0)NHaryl, OC(O) NHheteroaryl, OC(0)NHheterocycloalkyl, NHC(0)alkyl, NHC(0)alkenyl, NHC(0)alkynyl, NHC(0)cycloalkyl, NHC(0)aryl, NHC(O) heteroaryl, NHC(0)heterocycloalkyl, NHC0 2 alkyl, NHC0 2 alkenyl, NHC0 2 alkynyl, NHC0 2 cycloalkyl, NHC0 2 aryl, NHC0 2 heteroaryl, NHC0 2 heterocycloalkyl, NHC(0)NH 2 , NHC(0)NHalkyl, NHC(0)NHalkenyl, NHC(0)NHalkenyl, NHC(0)NHcycloalkyl, NHC(0)NHaryl, NHC(0)NHheteroaryl, NHC(0)NHheterocycloalkyl, NHC(S)NH 2 , NHC(S)NHalkyl, NHC(S)NHalkenyl, NHC(S)NHalkynyl, NHC(S)NHcycloalkyl, NHC(S)NHaryl, NHC(S)NHheteroaryl, NHC(S)NHheterocycloalkyl, NHC(NH)NH 2 , NHC(NH)NHalkyl, NHC(NH)NHalkenyl, NHC(NH)NHalkenyl, NHC(NH)NHcycloalkyl, NHC(NH)NHaryl, NHC(NH)NHheteroaryl, NHC(NH)NHheterocycloalkyl, NHC(NH)alkyl, NHC(NH)alkenyl, NHC(NH)alkenyl, NHC(NH)cycloalkyl, NHC(NH)aryl, NHC(NH)heteroaryl, NHC(NH)heterocycloalkyl, C(NH)NHalkyl, C(NH)NHalkenyl, C(NH)NHalkynyl, C(NH)NHcycloalkyl, C(NH)NHaryl, C(NH)NHheteroaryl, C(NH)NHheterocycloalkyl, S(0)alkyl, S(0)alkenyl, S(0)alkynyl, S(0)cycloalkyl, S(0)aryl, S(0) 2 alkyl, S(0) 2 alkenyl, S(0) 2 alkynyl, S(0) 2 cycloalkyl, S(0) 2 aryl, S(0)heteroaryl, S(0)heterocycloalkyl, S0 2 NH 2 , S0 2 NHalkyl, S0 2 NHalkenyl, S0 2 NHalkynyl, S0 2 NHcycloalkyl, S0 2 NHaryl, S0 2 NHheteroaryl, S0 2 NHheterocycloalkyl, NHS0 2 alkyl, NHS0 2 alkenyl, NHS0 2 alkynyl, NHS0 2 cycloalkyl, NHS0 2 aryl, NHS0 2 heteroaryl, NHS0 2 heterocycloalkyl, CH 2 NH 2 , CH 2 S0 2 CH 3 , alkyl, alkenyl, alkynyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, heterocycloalkyl, cycloalkyl, carbocyclic, heterocyclic, polyalkoxyalkyl, polyalkoxy, methoxymethoxy, methoxyethoxy, SH, S-alkyl, S- alkenyl, S-alkynyl, S-cycloalkyl, S-aryl, S-heteroaryl, S-heterocycloalkyl, or methylthiomethyl.

In certain embodiments, suitable monovalent substituents on a substitutable carbon atom of an "optionally substituted" group are independently halogen; (CH 2 ) 0- 4R o ; (CH 2 ) 0 -4OR°; O(CH 2 ) 0- 4 C(0)OR°; (CH 2 ) 0-4 CH(OR°) 2 ; (CH 2 ) 0-4 SR°; (CH 2 ) 0-4 Ph, which may be substituted with R°; (CH 2 ) 0- 4 O(CH 2 ) 0 - 4 Ph which may be substituted with R°; -CH=CHPh, which may be substituted with R°; N0 2 ; CN; N 3 ; (CH 2 ) 0 - 4 N(R°) 2 ; (CH 2 ) 0-4 N(R o )C(O)R°; N(R°)C(S)R°; (CH 2 ) 0-4 N(R o )C(O)NR° 2 ; N(R°)C(S)NR° 2 ; (CH 2 ) 0-4 N(R o )C(O)OR°; N(R°)N(R°)C(0)R°; N(R°)N(R°)C(0)NR° 2 ; N(R°)N(R°)C(0)OR°; (CH 2 ) 0 - 4 C(O)R°; C(S)R°; (CH 2 ) 0 - 4 C(O)OR°; (CH 2 ) 0 - 4 C(O)SR°; (CH 2 ) 0- 4 C(0)OSiR° 3 ; (CH 2 )o -4 OC(0)R°; OC(0)(CH 2 ) 0 - 4 SR-, SC(S)SR°; (CH 2 ) 0 - 4 SC(O)R°; (CH 2 ) 0- 4 C(0)NR° 2 ; C(S)NR° 2 ; C(S)SR°; SC(S)SR°, (CH 2 ) 0 - 4 OC(O)NR° 2 ; C(0)N(OR°)R°; C(0)C(0)R°; C(0)CH 2 C(0)R°; C(NOR°)R°; (CH 2 ) 0 - 4 SSR°; (CH 2 ) 0 - 4 S(O) 2 R°; (CH 2 ) 0 - 4 (O) 2 OR°; (CH 2 ) 0- 4 OS(0) 2 R°; S(0) 2 NR° 2 ; (CH 2 ) 0 - 4 S(O)R°; N(R°)S(0) 2 NR° 2 ; N(R°)S(0) 2 R°; N(OR°)R°; C(NH)NR° 2 ; P(0) 2 R°; P(0)R° 2 ; OP(0)R° 2 ; OP(0)(OR°) 2 ; SiR° 3 ; (straight or branched Ci -4 alkylene)0-N(R°) 2 ; or (straight or branched Ci- 4 alkylene)C(0)0-N(R°) 2 , wherein each R° may be substituted as defined below and is independently hydrogen, Ci -6 aliphatic, CH 2 Ph, 0(CH 2 ) 0- iPh, or a 5-6- membered saturated, partially unsaturated, or aromatic ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or, notwithstanding the definition above, two independent occurrences of R°, taken together with their intervening atom(s), may form a 3 to 12 membered saturated, partially unsaturated, or aryl mono- or bicyclic ring having 0 to 4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, which may be substituted as defined below.

Examples of monovalent substituents on R° (or the ring formed by taking two independent occurrences of R° together with their intervening atoms), are independently halogen, -(CH 2 ) 0- 2 R*, -(haloR*), -(CH 2 )o- 2 OH, -(CH 2 ) 0-2 OR*, -(CH 2 ) 0 - 2 CH(OR*) 2 , -O(haloR'), -CN, -N 3 , -(CH 2 ) 0- 2 C(0)R*, -(CH 2 )o- 2 C(0)OH, -(CH 2 )o- 2 C(0)OR*, -(CH 2 ) 0-2 SR*, -(CH 2 ) 0 - 2 SH, -(CH 2 ) 0-2 NH 2 , -(CH 2 ) 0- 2 NHR*, -(CH 2 ) o- 2 NR*2 , -N0 2 , -SiR* 3 , -OSiR* 3 , -C(0)SR* -(C^straight or branched alkylene)C(0)OR*, or -SSR*, wherein each R* is unsubstituted or where preceded by "halo" is substituted only with one or more halogens, and is independently selected from Ci -4 aliphatic, - CH 2 Ph, -O(CH 2 ) 0 -iPh, or a 5-6-membered saturated, partially unsaturated, or aryl ring having 0- 4 heteroatoms independently selected from nitrogen, oxygen, or sulfur. Suitable divalent substituents on a saturated carbon atom of R° include =0 and =S.

Examples of divalent substituents on a saturated carbon atom of an "optionally substituted" group include the following: =0, =S, =NNR* 2 , =NNHC(0)R*, =NNHC(0)OR*, =NNHS(0) 2 R*, =NR*, =NOR*, -O(C(R* 2 )) 2-30 -, or -S(C(R* 2 )) 2-3 S-, wherein each independent occurrence of R* is selected from hydrogen, C 1-6 aliphatic which may be substituted as defined below, or an unsubstituted 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur. Suitable divalent substituents that are bound to vicinal substitutable carbons of an "optionally substituted" group include: -O(CR 2 ) 2-30 -, wherein each independent occurrence of R is selected from hydrogen, C 1-6 aliphatic which may be substituted as defined below, or an unsubstituted 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.

Examplary substituents on the aliphatic group of R* include halogen, -R*, -(haloR*), -OH, -OR*, -O(haloR'), -CN, -C(0)OH, -C(0)OR*, -NH 2 , -NHR*, -NR* 2 , or -N0 2 , wherein each R* is unsubstituted or where preceded by "halo" is substituted only with one or more halogens, and is independently Ci -4 aliphatic, -CH 2 Ph, -O(CH 2 ) 0- iPh, or a 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.

The expression "pharmaceutically acceptable salt" refers to those salts of the compounds formed by the process of the present description which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals without undue toxicity, irritation, allergic response and the like, and are commensurate with a reasonable benefit/risk ratio. Pharmaceutically acceptable salts are well known in the art. For example, S. M. Berge, et al. describes pharmaceutically acceptable salts in detail in J. Pharmaceutical Sciences, 66: 1-19 (1977). The salts can be prepared in situ during the final isolation and purification of the compounds of the present description, or separately by reacting a free base function of the compound with a suitable organic or inorganic acid (acid addition salts) or by reacting an acidic function of the compound with a suitable organic or inorganic base (base-addition salts). Examples of pharmaceutically acceptable salts include, but are not limited to, nontoxic acid addition salts, or salts of an amino group formed with inorganic acids such as hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid and perchloric acid or with organic acids such as acetic acid, maleic acid, tartaric acid, citric acid, succinic acid or malonic acid or by using other methods used in the art such as ion exchange. Other pharmaceutically acceptable salts include, but are not limited to, adipate, alginate, ascorbate, aspartate, benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphorate, camphorsulfonate, citrate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, formate, fumarate, glucoheptonate, glycerophosphate, gluconate, hemisulfate, heptanoate, hexanoate, hydroiodide, 2-hydroxy-ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, malate, maleate, malonate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oleate, oxalate, palmitate, pamoate, pectinate, persulfate, 3- phenylpropionate, phosphate, picrate, pivalate, propionate, stearate, succinate, sulfate, tartrate, thiocyanate, p-toluenesulfonate, undecanoate, valerate salts, and the like. Representative base addition alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, or magnesium salts, and the like. Further pharmaceutically acceptable salts include, when appropriate, nontoxic ammonium, quaternary ammonium, and amine cations formed using counterions such as halide, hydroxide, carboxylate, sulfate, phosphate, nitrate, sulfonate and aryl sulfonate.

The term "solvate" refers to a physical association of one of the present compounds with one or more solvent molecules. This physical association includes hydrogen bonding. In certain instances, the solvate will be capable of isolation, for example when one or more solvent molecules are incorporated in the crystal lattice of a crystalline solid. "Solvate" encompasses both solution-phase and isolable solvates. Exemplary solvates include, without limitation, hydrates, hemihydrates, ethanolates, hemiethanolates, n-propanolates, iso-propanolates, 1- butanolates, 2-butanolate, and solvates of other physiologically acceptable solvents, such as the Clas 3 solvents described in the International Conference on Harmonization (ICH), Guide for Industry, Q3C Impurities: Residual Solvents (1997). The compounds as herein described also include each of their solvates and mixtures thereof.

As used herein, the term "pharmaceutically acceptable ester" refers to esters of the compounds formed by the process of the present description which hydrolyze in vivo and include those that break down readily in the human body to leave the parent compound or a salt thereof. Suitable ester groups include, for example, those derived from pharmaceutically acceptable aliphatic carboxylic acids, particularly alkanoic, alkenoic, cycloalkanoic and alkanedioic acids, in which each alkyl or alkenyl moiety advantageously has not more than 6 carbon atoms. Examples of particular esters include, but are not limited to, formates, acetates, propionates, butyrates, acrylates and ethylsuccinates.

The expression "pharmaceutically acceptable prodrugs" as used herein refers to those prodrugs of the compounds formed by the process of the present description which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals with undue toxicity, irritation, allergic response, and the like, commensurate with a reasonable benefit/risk ratio, and effective for their intended use. "Prodrug", as used herein means a compound which is convertible in vivo by metabolic means (e.g. by hydrolysis) to afford any compound delineated by the formulae of the instantdescription. Various forms of prodrugs are known in the art, for example, as discussed in Bundgaard, (ed.), Design of Prodrugs, Elsevier (1985); Widder, et al. (ed.), Methods in Enzvmology, vol. 4, Academic Press (1985); Krogsgaard-Larsen, et al., (ed). "Design and Application of Prodrugs, Textbook of Drug Design and Development", Chapter 5, 1 13-191 (1991); Bundgaard, et al., Journal of Drug Deliver Reviews, 8:1-38(1992); Bundgaard, J. of Pharmaceutical Sciences, 77:285 et seq. (1988); Higuchi and Stella (eds.) Prodrugs as Novel Drug Delivery Systems, American Chemical Society (1975); and Bernard Testa & Joachim Mayer, "Hydrolysis In Drug And Prodrug Metabolism: Chemistry, Biochemistry And Enzvmology", John Wiley and Sons, Ltd. (2002).

Combinations of substituents and variables envisioned by the present description are only those that result in the formation of stable compounds. The term "stable", as used herein, refers to compounds which possess stability sufficient to allow manufacture and which maintains the integrity of the compound for a sufficient period of time to be useful for the purposes detailed herein (e.g., therapeutic or prophylactic administration to a subject). / ' / ' . Compounds

The compounds of the present application may be prepared by conventional chemical synthesis, such as exemplified in Schemes 1 to 3 and in Examples 1 to 40. As can be appreciated by the skilled artisan, further methods of synthesizing the compounds of the formulae herein will be evident to those of ordinary skill in the art. Additionally, the various synthetic steps may be performed in an alternate sequence or order to give the desired compounds. In addition, the solvents, temperatures, reaction durations, etc. delineated herein are for purposes of illustration only and one of ordinary skill in the art will recognize that variation of the reaction conditions can produce the desired products of the present description. Synthetic chemistry transformations and protecting group methodologies (protection and deprotection) useful in synthesizing the compounds described herein are known in the art and include, for example, those such as described in R. Larock, Comprehensive Organic Transformations, VCH Publishers (1989); T.W. Greene and P.G.M. Wuts, Protective Groups in Organic Synthesis, 2d. Ed., John Wiley and Sons (1991); L. Fieser and M. Fieser, Fieser and Fieser's Reagents for Organic Synthesis, John Wiley and Sons (1994); and L. Paquette, ed., Encyclopedia of Reagents for Organic Synthesis, John Wiley and Sons (1995), and subsequent editions thereof. The synthesized compounds can be separated from a reaction mixture and further purified by standard methods such as column chromatography, high pressure liquid chromatography, or recrystallization.

The compounds of the present description may be modified by appending various functionalities via any synthetic means delineated herein to enhance selective biological properties. Such modifications are known in the art and include those which increase biological penetration into a given biological system (e.g., blood, lymphatic system, central nervous system), increase oral availability, increase solubility to allow administration by injection, alter metabolism and alter rate of excretion.

The recitation of a listing of chemical groups in any definition of a variable herein includes definitions of that variable as any single group or combination of listed groups. The recitation of an embodiment for a variable herein includes that embodiment as any single embodiment or in combination with any other embodiments or portions thereof. The recitation of an embodiment herein includes that embodiment as any single embodiment or in combination with any other embodiments or portions thereof. As such, the following embodiments are present alone or in combination if applicable: The present application relates to substituted imidazopyridine compounds of general Formula I, as well as to their pharmaceutically acceptable salts, solvates, esters or prodrugs thereof:

wherein,

substituted or unsubstituted group selected from Ci-C 6 alkyl, C 2 -C 6 alkenyl, C 2 -

C 6 alkynyl, C 3 -C 10 cycloalkyl, C 3 -C 10 heterocycloalkyl, Cs-doCycloalkyld-Cealkyl-, C 3 - Ci 0 heterocycloalkylCi-C 6 alkyl-, Ce-doaryld-Cealkyl-, Cs-doheteroaryld-dalkyl-, C(0)R a , OR a , NHR a , N(R a ) 2 , C(0)NH 2 , C(0)NHR a , C(0)N(R a ) 2 , NHC(0)R a , N(R a )C(0)R a , NHC(0)NHR a , N(R a )C(0)NHR a , NHC(0)N(R a ) 2 , N(R a )C(0)N(R a ) 2 , NHS0 2 R a , N(R a )S0 2 R a , NHS0 2 NHR a , N(R a )S0 2 NHR a , NHS0 2 N(R a ) 2 , N(R a )S0 2 N(R a ) 2 , S0 2 R a , S0 2 NH 2 , S0 2 NHR a , and S0 2 N(R a ) 2 ;

R a is, independently in each occurrence, a substituted or unsubstituted group selected from CrC 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -Ci 0 cycloalkyl, C 3 -Ci 0 heterocycloalkyl, C 3 - Ci 0 cycloalkylCi-C 6 alkyl-, C 3 -Ci 0 heterocycloalkylCi-C 6 alkyl-, C 6 -Ci 0 aryl, C 5 -Ci 0 heteroaryl, C 6 - Ci 0 arylCi-C 6 alkyl-, and C 5 -CioheteroarylCi-C 6 alkyl-;

R 2 is selected from H, NH 2 , CN or a substituted or unsubstituted group selected from d-

C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -Ci 0 cycloalkyl, C 3 -Ci 0 heterocycloalkyl, C 6 -Ci 0 aryl, C 5 - Cioheteroaryl, C(0)R a , OR a , SR a , NHR a , N(R a ) 2 , C(0)NH 2 , C(0)NHR a , C(0)N(R a ) 2 , NHC(0)R a , S0 2 R a , S0 2 NHR a , S0 2 N(R a ) 2 , NHS0 2 R a , N(R a )S0 2 R a , NHS0 2 NHR a , N(R a )S0 2 NHR a , NHS0 2 N(R a ) 2 , and N(R a )S0 2 N(R a ) 2 ;

R 3 and R 6 are each independently H, halogen, NH 2 , CN or a substituted or unsubstituted group selected from C C 6 alkyl, C(0)R a , OR a , NHR a , N(R a ) 2 , C(0)NH 2 , C(0)NHR a , C(0)N(R a ) 2 , and NHC(0)R a ; and

one of R 4 and R 5 is H, halogen, NH 2 , CN or a substituted or unsubstituted group selected from C C 6 alkyl, C(0)R a , NH 2 , NHR a , N(R a ) 2 , C(0)NH 2 , C(0)NHR a , C(0)N(R a ) 2 , and NHC(0)R a ; and the other of R 4 and R 5 is a group of Formula II:

Formula II

wherein,

R 7 and R 10 are each independently H, halogen (such as F, CI), CN, or a substituted or unsubstituted group selected from d-Cealkyl or C 3 -C 6 cycloalkyl group, OC Cealkyl, OC 3 - Cecycloalkyl, SC C 6 alkyl, SC 3 -C 6 cycloalkyl, NHC C 6 alkyl, NHC 3 -C 6 cycloalkyl, N(C C 6 alkyl)2, NHC(0)C C 6 alkyl and NHC(0)C 3 -C 6 cycloalkyl;

R 8 is halogen (such as F, CI), CN, or a substituted or unsubstituted group selected from C C 6 alkyl or C 3 -C 6 cycloalkyl group, OC C 6 alkyl, OC 3 -C 6 cycloalkyl, SC C 6 alkyl, SC 3 - Cecycloalkyl, NHC C 6 alkyl, NHC 3 -C 6 cycloalkyl, N(Ci-C 6 alkyl) 2 , NHC(0)C C 6 alkyl and NHC(0)C 3 -C 6 cycloalkyl;

R 9 is a substituted or unsubstituted Ci-C 3 alkyl or C 3 -C 5 cycloalkyl group; and

X 1 , X 2 , and X 3 are each a nitrogen or carbon atom, wherein when X 1 , X 2 , or X 3 is a nitrogen atom, then the R 7 , R 8 , or R 10 attached thereto is absent, provided that at least two of X 1 , X 2 , and X 3 are C.

In one embodiment, the compound is of Formula I, wherein R 4 is a group of Formula I I, preferably wherein said R 5 is hydrogen or a substituted or unsubstituted C C 3 alkyl. According to another embodiment, the compound is of Formula I, wherein R 5 is a group of Formula II, preferably wherein said R 4 is hydrogen or a substituted or unsubstituted C C 3 alkyl.

In one embodiment, in Formula II, X 1 is a nitrogen atom and R 10 is absent, and X 2 and X 3 are carbon atoms. In another embodiment, X 1 , X 2 and X 3 are all carbon atoms. For instance, the group of Formula II may be defined as a group of Formula l l(a):

Formula ll(a) wherein R 7 , R 8 , R 9 and R 10 are as herein defined.

In one embodiment, R 9 is an unsubstituted CrC 3 alkyl or C 3 -C 5 cycloalkyl group or a fluorinated CrC 3 alkyl group or C 3 -C 5 cycloalkyl group, for instance trifluoromethyl, a methyl, ethyl, n-propyl, isopropyl or cyclopropyl group. In another embodiment, R 7 and R 10 are each hydrogen atoms and R 8 is selected from CI, CN, and a substituted or unsubstituted Ci-C 3 alkyl, C 3 cycloalkyl, NHC C 3 alkyl, or NH(C C 3 alkyl) 2 group.

Another embodiment of the application relates to compounds of Formula I (a), and pharmaceutically acceptable salts, solvates, esters or prodrugs thereof:

Formula l(a)

wherein,

R 1 , R 2 , R 3 , R 5 , R 6 , R 7 , R 8 , R 9 , and R 10 are as herein defined.

According to another embodiment, the application also relates to compounds of Formula l(b), and pharmaceutically acceptable salts, solvates, esters or prodrugs thereof:

Formula l(b)

wherein,

R 1 , R 2 , R 3 , R 4 , R 6 , R 7 , R 8 , R 9 , and R 10 are as herein defined. In one embodiment, the application relates to compounds as herein defined wherein R 3 is H or a substituted or unsubstituted Ci-C 6 alkyl group, e.g. R 3 is H. In another embodiment, the application relates to compounds as herein defined wherein R 6 is H or a substituted or unsubstituted Ci-C 6 alkyl group, e.g. R 6 is H.

In another embodiment, the present application relates to compounds of Formula III, and pharmaceutically acceptable salts, solvates, esters or prodrugs thereof:

Formula III

wherein R 1 , R 2 , R 4 and R 5 are as herein defined.

Another embodiment of the application relates to compounds of Formula lll(a), and pharmaceutically acceptable salts, solvates, esters or prodrugs thereof:

Formula lll(a)

wherein R 1 , R 2 , R 7 , R 8 , R 9 , and R 10 are as herein defined;

or to compounds of Formula lll(b), and pharmaceutically acceptable salts, solvates, esters or prodrugs thereof:

Formula lll(b) wherein R 1 , R 2 , R 7 , R 8 , R 9 , and R 10 are as herein defined.

According to one embodiment, the application relates to compounds of Formula lll(a) or lll(b) as herein defined, wherein R 9 is an unsubstituted CrC 3 alkyl or C 3 -C 5 cycloalkyl group. In another embodiment, the application relates to compounds of Formula lll(a) or Formula lll(b) as herein defined, wherein R 9 is selected from methyl, trifluoromethyl, ethyl, n-propyl, isopropyl and cyclopropyl. In another embodiment, R 8 and R 9 are each independently a methyl, ethyl, isopropyl, fluoromethyl, difluoromethyl, trifluoromethyl, cyclopropyl, or difluorocyclopropyl group

According to another embodiment, the application relates to compounds of Formula lll(a) or lll(b) as herein defined, wherein said R 7 and R 10 are each hydrogen atoms and R 8 is selected from CI, CN, and a substituted or unsubstituted Ci-C 3 alkyl, C 3 cycloalkyl, NHCrC 3 alkyl, or NH(C C 3 alkyl) 2 group.

In one aspect, the present application relates to a compound of Formula I, l(a), l(b), III, ll l(a) or lll(b) as herein defined in combination with any of the described embodiments or subgroup thereof, wherein R 2 is hydrogen or a substituted or unsubstituted group selected from C C 6 alkyl, C 3 -C 10 cycloalkyl, or C 3 -C 10 heterocycloalkyl group. For instance, R 2 is a substituted or unsubstituted C C 3 alkyl, C 3 -C 6 cycloalkyl, or C 3 -C 6 heterocycloalkyl group, or R 2 is a substituted or unsubstituted group selected from methyl, ethyl, propyl, isopropyl, butyl, isobutyl, tert-butyl, cyclopropyl, cyclobutyl, cyclopentyl, tetrahydrofuranyl, tetrahydropyranyl, dioxolanyl, piperidinyl, and pyrrolidinyl, preferably, R 2 is hydrogen or a substituted or unsubstituted methyl or ethyl group.

According to a further aspect, the present application relates to a compound of Formula I, l(a), l(b), III, lll(a) or lll(b) in combination with any of the described embodiments or subgroups thereof, wherein R 1 is a branched or linear d-C^alky! group, unsubstituted or substituted with one or more group(s) selected from halogen, OH, NH 2 , CN, or a substituted or unsubstituted group selected from C 3 -Ci 0 cycloalkyl, C 3 -Ci 0 heterocycloalkyl, C 6 -Ci 0 aryl, C 5 -Ci 0 heteroaryl, C(0)R a , OR a , NHR a , N(R a ) 2 , C(0)NH 2 , C(0)NHR a , C(0)N(R a ) 2 , NHC(0)R a , N(R a )C(0)R a , NHC(0)NHR a , N(R a )C(0)NHR a , NHC(0)N(R a ) 2 , N(R a )C(0)N(R a ) 2 , NHS0 2 R a , N(R a )S0 2 R a , NHS0 2 NHR a , N(R a )S0 2 NHR a , NHS0 2 N(R a ) 2 , N(R a )S0 2 N(R a ) 2 , S0 2 R a , S0 2 NH 2 , S0 2 NHR a , S0 2 N(R a ) 2 , , wherein R a is as herein defined. In another embodiment, R 1 is a branched or linear Ci-C 3 alkyl group, unsubstituted or substituted with one or more group(s) selected from halogen, OH, NH 2 , CN, or a substituted or unsubstituted group selected from C 3 -Ci 0 cycloalkyl, C 3 - Cioheterocycloalkyl, C 6 -Ci 0 aryl, C 5 -Ci 0 heteroaryl, C(0)R a , OR a , NHR a , N(R a ) 2 , C(0)NH 2 , C(0)NHR a , C(0)N(R a ) 2 , NHC(0)R a , N(R a )C(0)R a , NHC(0)NHR a , N(R a )C(0)NHR a , NHC(0)N(R a ) 2 , N(R a )C(0)N(R a ) 2 , NHS0 2 R a , N(R a )S0 2 R a , NHS0 2 NHR a , N(R a )S0 2 NHR a , NHS0 2 N(R a ) 2 , N(R a )S0 2 N(R a ) 2 , S0 2 R a , S0 2 NH 2 , S0 2 NHR a , S0 2 N(R a ) 2 , wherein R a is as herein defined. In a further embodiment, R 1 is a C C 2 alkyl substituted with one or more group(s) selected from halogen or a substituted or unsubstituted group selected from C 3 -C 10 cycloalkyl, C 3 -C 10 heterocycloalkyl, C 6 -C 10 aryl, C 5 -C 10 heteroaryl, C(0)R a , OR a , NHR a , N(R a ) 2 , C(0)NH 2 , C(0)NHR a , C(0)N(R a ) 2 , NHC(0)R a , N(R a )C(0)R a , NHS0 2 R a , N(R a )S0 2 R a , NHS0 2 NHR a , N(R a )S0 2 NHR a , NHS0 2 N(R a ) 2 , N(R a )S0 2 N(R a ) 2 , S0 2 R a , S0 2 NH 2 , S0 2 NHR a , S0 2 N(R a ) 2 , wherein R a is as herein defined. In yet another embodiment, R 1 is a Cialkyl substituted with one or more substituted or unsubstituted group selected from C 3 -Ci 0 cycloalkyl, C 3 - Cioheterocycloalkyl, C 6 -Ci 0 aryl, C 5 -Ci 0 heteroaryl, OR a , and NHR a , wherein R a is as herein defined. In a further embodiment, R 1 is selected from C(0)R a , OR a , NHR a , N(R a ) 2 , C(0)NHR a , C(0)N(R a ) 2 , S0 2 R a , S0 2 NHR a , and S0 2 N(R a ) 2 , wherein R a is as herein defined. In yet another embodiment, R 1 is selected from C(O)C 3 -Ci 0 heterocycloalkyl, OR a , NHR a , N(R a ) 2 , C(0)NHR a , S0 2 (C 3 -Cioheterocycloalkyl), and S0 2 NHR a , wherein R a is as herein defined.

In an alternative definition, R 1 is represented as a R a group linked to the imidazopyridine bicyclic core via a linker, wherein said linker is selected from a linear or branched -C C 3 akyl-, -C C 3 akylO-, -OC C 3 akyl-, -C C 3 akylN(H)-, -NHC C 3 akyl-, -C C 3 akylN(R a )-, -NR a C C 3 akyl-, C(O), O, NH, -N(R a )-, -C(0)N(H)-, -C(0)N(R a )-, S0 2 , -S0 2 N(H)-, and -S0 2 N(R a )- group.

In one embodiment, R a is, independently in each occurrence, selected from a substituted or unsubstituted d-C^alky!, C 3 -C 10 cycloalkyl, C 3 -C 10 heterocycloalkyl, C 6 -C 10 aryl, or C 5 - C 10 heteroaryl. In one embodiment, R a is, when within the definition of R 1 , a C 3 -C 10 cycloalkyl, C 3 - C 10 heterocycloalkyl, C 6 -C 10 aryl, or C 5 -C 10 heteroaryl group.

Examples of compounds of the present application include, without limitation:

Compound 1 Compound 2

Compound 7 (RT: 9.61)

Compound 1 1 Compound 12

Compound 16

Compound 18

Compound 21 Compound 22

Compound 23

Compound 29

Compound 31 Compound 32

Compound 41 Compound 42 51215

31

Compound 51 Compound 52 (Isomer RT4.17)

Compound 55 Compound 56

or a pharmaceutically acceptable salt, solvate, ester or prodrug thereof.

In a further embodiment, this application relates to a compound selected from Compounds 1 to 56 as herein defined, or a pharmaceutically acceptable salt, solvate, or prodrug thereof, for instance, any of these compounds or their isomers, or a pharmaceutically acceptable salt, solvate, or prodrug thereof, may be taken individually or as sub-groups.

/ ' / ' / ' . Methods, Uses, Formulations and Administration

As used herein, the term "effective amount" means that amount of a drug or pharmaceutical agent that will elicit the biological or medical response of a tissue, system, animal or human that is being sought, for instance, by a researcher or clinician. Furthermore, the term "therapeutically effective amount" means any amount which, as compared to a corresponding subject who has not received such amount, results in treatment, healing, prevention, or amelioration of a disease, disorder, or side effect, or a decrease in the rate of advancement of a disease or disorder. The term also includes within its scope amounts effective to enhance normal physiological function.

As used herein, the terms "treatment," "treat," and "treating" refer to reversing, alleviating, delaying the onset of, or inhibiting the progress of a disease or disorder, or one or more symptoms thereof, as described herein. In some embodiments, treatment may be administered after one or more symptoms have developed. In other embodiments, treatment may be administered in the absence of symptoms. For example, treatment may be administered to a susceptible individual prior to the onset of symptoms (e.g., in light of a history of symptoms and/or in light of genetic or other susceptibility factors). Treatment may also be continued after symptoms have resolved, for example to prevent or delay their recurrence.

As used herein, the term "bromodomain inhibitor" denotes a compound which inhibits the binding of a bromodomain with its cognate acetylated proteins. In one embodiment the bromodomain inhibitor is a compound which inhibits the binding of a bromodomain to acetylated lysine residues. In a further embodiment the bromodomain inhibitor is a compound which inhibits the binding of a bromodomain to acetylated lysine residues on histones, particularly histones H3 and H4.

In a particular embodiment the bromodomain inhibitor is a compound that inhibits the binding of BET family bromodomains to acetylated lysine residues (hereafter referred to as a "BET family bromodomain inhibitor"). The BET family of bromodomain containing proteins comprises 4 proteins (BRD2, BRD3, BRD4 and BRD-t) which contain tandem bromodomains capable of binding to two acetylated lysine residues in close proximity, increasing the specificity of the interaction.

As used herein, the term "inhibitor" is defined as a compound that binds to and/or inhibits the target bromodomain-containing protein (such as a BET protein, e.g. , BRD2, BRD3, BRD4, and/or BRDT) with measurable affinity.

The terms "measurable affinity" and "measurably inhibit," as used herein, means a measurable change in activity of at least one bromodomain-containing protein between a sample comprising a provided compound, or composition thereof, and at least one histone methyltransferase, and an equivalent sample comprising at least one bromodomain-containing protein, in the absence of said compound, or composition thereof.

The term "patient or subject" as used herein refers to a mammal. A subject therefore refers to, for example, dogs, cats, horses, cows, pigs, guinea pigs, and the like. Preferably the subject is a human. When the subject is a human, the subject may be either a patient or a healthy human.

In some embodiments, the disease or condition can be an auto-immune disorder, an inflammatory disorder, a dermal disorder, or cancer. In some optional embodiments, the disease or condition can be an auto-immune disorder. In some other optional embodiments, the disease or condition can be an inflammatory disorder. In further optional embodiments, the inflammatory disorder can be rheumatoid arthritis, irritable bowel syndrome, or psoriasis.

In some other optional embodiments, the disease or condition can be cancer. In further optional embodiments, the cancer can be brain cancer, pancreatic cancer, breast cancer, lung cancer, or prostate cancer. In further optional embodiments, the cancer can be brain cancer. In further optional embodiments, the brain cancer is glioblastoma multiforme.

In some embodiments, the cancer is selected from the group consisting of: brain (gliomas), glioblastomas, leukemias, lymphomas, Bannayan-Zonana syndrome, Cowden disease, Lhermitte-Duclos disease, breast, inflammatory breast cancer, Wilm's tumor, Ewing's sarcoma, Rhabdomyosarcoma, ependymoma, medulloblastoma, colon, gastric, bladder, head and neck, kidney, lung, liver, melanoma, renal, ovarian, pancreatic, prostate, sarcoma, osteosarcoma, giant cell tumor of bone and thyroid.

In some embodiments, the disorder can be a proliferative disorder, inflammatory disease, sepsis, autoimmune disease, or viral infection. In some optional embodiments, the proliferative disorder can be cancer.

The term "proliferative disorder" refers to cells having the capacity for autonomous growth, i.e., an abnormal state of condition characterized by rapidly proliferating cell growth which generally forms a distinct mass that show partial or total lack of structural organization and functional coordination with normal tissue.

The terms "neoplasm", "neoplastic disorder", "neoplasia" "cancer," and "tumor" are meant to encompass hematopoietic neoplasms (e.g. lymphomas or leukemias) as well as solid neoplasms (e.g. sarcomas or carcinomas), including all types of pre-cancerous and cancerous growths, or oncogenic processes, metastatic tissues or malignantly transformed cells, tissues, or organs, irrespective of histopathologic type or stage of invasiveness. Hematopoietic neoplasms are malignant tumors affecting hematopoietic structures (structures pertaining to the formation of blood cells) and components of the immune system, including leukemias (related to leukocytes (white blood cells) and their precursors in the blood and bone marrow) arising from myeloid, lymphoid or erythroid lineages, and lymphomas (related to lymphocytes). Solid neoplasms include sarcomas, which are malignant neoplasms that originate from connective tissues such as muscle, cartilage, blood vessels, fibrous tissue, fat or bone. Solid neoplasms also include carcinomas, which are malignant neoplasms arising from epithelial structures, including external epithelia (e.g., skin and linings of the gastrointestinal tract, lungs, and cervix), and internal epithelia that line various glands (e.g., breast, pancreas, thyroid). Examples of neoplasms include leukemia, and hepatocellular cancers, sarcoma, vascular endothelial cancers, breast cancers, central nervous system cancers (e.g. astrocytoma, gliosarcoma, neuroblastoma, oligodendroglioma and glioblastoma), prostate cancers, lung and bronchus cancers, larynx cancers, esophagus cancers, colon cancers, colorectal cancers, gastro- intestinal cancers, melanomas, ovarian and endometrial cancer, renal and bladder cancer, liver cancer, endocrine cancer (e.g. thyroid), and pancreatic cancer.

In some aspects, examples of cancers treated using the compounds and methods described herein include, but are not limited to, acinic cell carcinoma, acoustic neuroma, acral lentiginous melanoma, acrospiroma, acute eosinophilic leukemia, acute erythroid leukemia, acute lymphoblastic leukemia, acute lymphocytic leukemia, acute megakaryoblastic leukemia, acute monocytic leukemia, acute myelogenous leukemia, acute myelognous leukemia, acute promyelocytic leukemia, adrenal cancer, adenocarcinoma, adenoid cystic carcinoma, adenoma, adenomatoid odontogenic tumor, adenosquamous carcinoma, adipose tissue neoplasm, adrenal cancer, adrenocortical carcinoma, adult T-cell leukemia/lymphoma, aggressive NK-cell leukemia, AIDS-related lymphoma, alveolar rhabdomyosarcoma, alveolar soft part sarcoma, ameloblastic fibroma, anaplastic large cell lymphoma, anaplastic thyroid cancer, angioimmunoblastic T-cell lymphoma, angiomyolipoma, angiosarcoma, astrocytoma, atypical teratoid rhabdoid tumor, Bannayan-Zonana syndrome, basal cell carcinoma, B-cell chronic lymphocytic leukemia, B-cell lymphoma, B-cell prolymphocytic leukemia, biliary tract cancer, bladder, bladder cancer, blastoma, bone cancer, brain (gliomas), brain cancer, breast, breast cancer, Brenner tumor, Brown tumor, Burkitt's lymphoma, carcinoma, carcinoma in situ, carcinosarcoma, cartilage tumor, cementoma, cervical cancer, chondroma, chordoma, choriocarcinoma, choroid plexus papilloma, chronic lymphocytic leukemia, clear-cell sarcoma of the kidney, colon, colorectal cancer, Cowden disease, craniopharyngioma, cutaneous T-cell lymphoma, Degos disease, desmoplastic small round cell tumor, diffuse large B-cell lymphoma, dysembryoplastic neuroepithelial tumor, dysgerminoma, embryonal carcinoma, endocrine gland neoplasm, endodermal sinus tumor, enteropathy-associated T-cell lymphoma, ependymoma, esophageal cancer, Ewing's sarcoma, fetus in fetu, fibroma, fibrosarcoma, follicular lymphoma, follicular thyroid cancer, gallbladder cancer, ganglioneuroma, gastric, gastric cancer, gastrointestinal cancer, germ cell tumor, gestational choriocarcinoma, giant cell fibroblastoma, giant cell tumor of bone and thyroid, giant cell tumor of the bone, glial tumor, glioblastoma multiforme, glioblastomas, glioma, gliomatosis cerebri, glucagonoma, gonadoblastoma, granulosa cell tumor, gynandroblastoma, hairy cell leukemia, head and neck, head and neck cancer, hemangioblastoma, hemangiopericytoma, hematological malignancy, hepatoblastoma, hepatosplenic T-cell lymphoma, Hodgkin's lymphoma, inflammatory breast cancer, intestinal cancer, invasive lobular carcinoma, kidney, kidney cancer, laryngeal cancer, lentigo maligna, lethal midline carcinoma, leukemia, leukemias, leydig cell tumor, Lhermitte-Duclos disease, liposarcoma, liver, liver cancer, lung, lung cancer, lymphangio sarcoma, lymphangioma, lymphoepithelioma, lymphoma, lymphomas, malignant fibrous histiocytoma, malignant peripheral nerve sheath tumor, malignant triton tumor, MALT lymphoma, mantle cell lymphoma, marginal zone B-cell lymphoma, mast cell leukemia, mediastinal germ cell tumor, medullary carcinoma of the breast, medullary thyroid cancer, medulloblastoma, melanoma, meningioma, merkel cell cancer, mesothelioma, metastatic urothelial carcinoma, mixed Mullerian tumor, mucinous tumor, multiple myeloma, muscle tissue neoplasm, mycosis fungoides, myeloid sarcoma, myxoid liposarcoma, myxoma, myxosarcoma, nasopharyngeal carcinoma, neurinoma, neuroblastoma, neurofibroma, neuroma, nodular melanoma, non-Hodgkin's lymphoma, non- small cell lung cancer, ocular cancer, oligoastrocytoma, oligodendroglioma, oncocytoma, optic nerve sheath meningioma, optic nerve tumor, oral cancer, osteosarcoma, ovarian, ovarian cancer, Pancoast tumor, pancreatic, pancreatic cancer, papillary thyroid cancer, paraganglioma, pharyngeal cancer, pinealoblastoma, pineocytoma, pituicytoma, pituitary adenoma, pituitary tumor, plasmacytoma, polyembryoma, precursor T-lymphoblastic lymphoma, primary central nervous system lymphoma, primary effusion lymphoma, primary peritoneal cancer, prostate, prostate cancer, pseudomyxoma peritonei, rectal cancer, renal, renal cell carcinoma, renal medullary carcinoma, retinoblastoma, rhabdomyoma, Rhabdomyosarcoma, Richter's transformation, sarcoma, Schwannomatosis, seminoma, Sertoli cell tumor, sex cord-gonadal stromal tumor, Sezary's disease, signet ring cell carcinoma, skin cancer, small blue round cell tumors, small cell carcinoma, small cell lung cancer, small intestine cancer, soft tissue sarcoma, somatostatinoma, soot wart, spinal tumor, splenic marginal zone lymphoma, squamous carcinoma, squamous cell carcinoma, stomach cancer, synovial sarcoma, T-cell lymphoma, testicular cancer, thecoma, throat cancer, thyroid cancer, transitional cell carcinoma, urachal cancer, urogenital cancer, urothelial carcinoma, uterine cancer, uveal melanoma, vaginal cancer, verrucous carcinoma, visual pathway glioma, vulvar cancer, Waldenstrom' s macroglobulinemia, Warthin's tumor, and Wilm's tumor.

In some embodiments, the cancer can be adenocarcinoma, adult T-cell leukemia/lymphoma, bladder cancer, blastoma, bone cancer, breast cancer, brain cancer, carcinoma, myeloid sarcoma, cervical cancer, colorectal cancer, esophageal cancer, gastrointestinal cancer, glioblastoma multiforme, glioma, gallbladder cancer, gastric cancer, head and neck cancer, Hodgkin's lymphoma, non-Hodgkin's lymphoma, intestinal cancer, kidney cancer, laryngeal cancer, leukemia, lung cancer, lymphoma, liver cancer, small cell lung cancer, non-small cell lung cancer, mesothelioma, multiple myeloma, ocular cancer, optic nerve tumor, oral cancer, ovarian cancer, pituitary tumor, primary central nervous system lymphoma, prostate cancer, pancreatic cancer, pharyngeal cancer, renal cell carcinoma, rectal cancer, sarcoma, skin cancer, spinal tumor, small intestine cancer, stomach cancer, T-cell lymphoma, testicular cancer, thyroid cancer, throat cancer, urogenital cancer, urothelial carcinoma, uterine cancer, vaginal cancer, or Wilms' tumor. In some other embodiments, the cancer can be acute myelognous leukemia or Burkitt's lymphoma.

In some embodiments, the autoimmune and inflammatory diseases or conditions can involve an inflammatory response to infections with bacteria, viruses, fungi, parasites or their toxins, as well as viruses. In some other embodiments, the autoimmune and inflammatory diseases or conditions can be selected from the group consisting of acute lung injury, acute pancreatitis, acute renal failure, ARDS (adult respiratory distress syndrome), burns, coronavirus, encephalitis, endotoxaemia, fulminant hepatitis, herpes simplex, herpes zoster, Herxheimer reactions, malaria and SIRS associated with viral infections such as influenza, meningitis, multi- organ dysfunction syndrome, myelitis, post-surgical syndromes, sarcoidosis, sepsis, sepsis syndrome, septic shock, systemic inflammatory response syndrome (SI RS), toxic shock syndrome.

In some embodiments, the present description provides a method of treating other conditions. Such other conditions include, but are not limited to, acne, acute inflammatory responses (such as acute respiratory distress syndrome and ischemia/reperfusion injury, glioblastoma, Graves' disease, HIV, HPV, inflammatory disease, keloids and related scarring, lung cancer, meningitis (bacterial and viral), multiple sclerosis, neoplasm, neuroblastoma, pancreatic cancer, scleroderma, skin cancer, toxic shock, viral infections, viral infections and diseases.

In some embodiments, the present description provides a method of treating a benign proliferative disorder. Such benign proliferative disorders include, but are not limited to, benign soft tissue tumors, bone tumors, brain and spinal tumors, eyelid and orbital tumors, granuloma, lipoma, meningioma, multiple endocrine neoplasia, nasal polyps, pituitary tumors, prolactinoma, pseudotumor cerebri, seborrheic keratoses, stomach polyps, thyroid nodules, cystic neoplasms of the pancreas, hemangiomas, vocal cord nodules, polyps, and cysts, Castleman disease, chronic pilonidal disease, dermatofibroma, pilar cyst, prolactinoma, pseudotumor cerebri, pyogenic granuloma, and juvenile polyposis syndrome.

The present description further relates to a method for treating infectious and noninfectious inflammatory events and autoimmune and other inflammatory diseases by administration of an effective amount of a provided compound to a mammal, in particular a human in need of such treatment. Examples of autoimmune and inflammatory diseases, disorders, and syndromes treated using the compounds and methods described herein include inflammatory pelvic disease, urethritis, skin sunburn, sinusitis, pneumonitis, encephalitis, meningitis, myocarditis, nephritis, osteomyelitis, myositis, hepatitis, gastritis, enteritis, dermatitis, gingivitis, appendicitis, pancreatitis, cholecystitis, agammaglobulinemia, psoriasis, allergy, Crohn's disease, irritable bowel syndrome, ulcerative colitis, Sjogren's disease, tissue graft rejection, hyperacute rejection of transplanted organs, asthma, allergic rhinitis, chronic obstructive pulmonary disease (COPD), autoimmune polyglandular disease (also known as autoimmune polyglandular syndrome), autoimmune alopecia, pernicious anemia, glomerulonephritis, dermatomyositis, multiple sclerosis, scleroderma, vasculitis, autoimmune hemolytic and thrombocytopenic states, Goodpasture's syndrome, atherosclerosis, Addison's disease, Parkinson's disease, Alzheimer's disease, Type I diabetes, septic shock, systemic lupus erythematosus (SLE), rheumatoid arthritis, psoriatic arthritis, juvenile arthritis, osteoarthritis, chronic idiopathic thrombocytopenic purpura, Waldenstrom macroglobulinemia, myasthenia gravis, Hashimoto's thyroiditis, atopic dermatitis, degenerative joint disease, vitiligo, autoimmune hypopituitarism, Guillain-Barre syndrome, Behcet's disease, scleracierma, mycosis fungoides, acute inflammatory responses (such as acute respiratory distress syndrome and ischemia/reperfusion injury), and Graves' disease. Other examples of infectious and noninfectious inflammatory events, autoimmune and other inflammatory diseases include, but are not limited to, Addison's disease, agammaglobulinemia, allergic rhinitis, allergy, Alzheimer's disease, appendicitis, asthma, atherosclerosis, atopic dermatitis, autoimmune alopecia, autoimmune hemolytic and thrombocytopenic states, autoimmune hypopituitarism, autoimmune polyglandular disease (also known as autoimmune polyglandular syndrome), Behcet's disease, cholecystitis, chronic idiopathic thrombocytopenic purpura, chronic obstructive pulmonary disease (COPD), Crohn's disease, degenerative joint disease, dermatitis, dermatomyositis, encephalitis, enteritis, gastritis, gingivitis, glomerulonephritis, Goodpasture's syndrome, Guillain-Barre syndrome, Hashimoto's thyroiditis, hepatitis, hyperacute rejection of transplanted organs, inflammatory pelvic disease, irritable bowel syndrome, juvenile arthritis, meningitis, multiple sclerosis, myasthenia gravis, mycosis fungoides, myocarditis, myositis, nephritis, osteoarthritis, osteomyelitis, pancreatitis, Parkinson's disease, pernicious anemia, pneumonitis, psoriasis, psoriatic arthritis, rheumatoid arthritis, scleracierma, scleroderma, septic shock, sinusitis, Sjogren's disease, skin sunburn, systemic lupus erythematosus (SLE), tissue graft rejection, Type I diabetes, ulcerative colitis, urethritis, vasculitis, vitiligo, and Waldenstrom macroglobulinemia.

In some embodiments, the present description provides a method of treating systemic inflammatory response syndromes such as LPS-induced endotoxic shock and/or bacteria- induced sepsis by administration of an effective amount of a provided compound to a mammal, in particular a human in need of such treatment.

The present description further relates to a method for treating viral infections and diseases by administration of an effective amount of a provided compound to a mammal, in particular a human in need of such treatment. Examples of viral infections and diseases treated using the compounds and methods described herein include episome-based DNA viruses including, but not limited to, human papillomavirus, Herpesvirus, Epstein-Barr virus, human immunodeficiency virus, hepatis B virus, and hepatitis C virus.

The present description further relates to a method of treating a subject, such as a human, suffering from one of the abovementioned conditions, illnesses, disorders or diseases. The method comprises administering a therapeutically effective amount of one or more provided compounds, which function by inhibiting a bromodomain and, in general, by modulating gene expression, to induce various cellular effects, in particular induction or repression of gene expression, arresting cell proliferation, inducing cell differentiation and/or inducing apoptosis, to a subject in need of such treatment.

The present description further provides a therapeutic method of modulating protein methylation, gene expression, cell proliferation, cell differentiation and/or apoptosis in vivo in diseases mentioned above, in particular cancer, inflammatory disease, and/or viral disease comprising administering to a subject in need of such therapy a pharmacologically active and therapeutically effective amount of one or more provided compounds.

The present description further provides a method of regulating endogenous or heterologous promoter activity by contacting a cell with a provided compound.

In certain embodiments, the present description provides a method of treating a disorder (as described herein) in a subject, comprising administering to the subject identified as in need thereof, a compound of the present description. The identification of those patients who are in need of treatment for the disorders described above is well within the ability and knowledge of one skilled in the art. Certain of the methods for identification of patients which are at risk of developing the above disorders which can be treated by the subject method are appreciated in the medical arts, such as family history, and the presence of risk factors associated with the development of that disease state in the subject patient. A clinician skilled in the art can readily identify such candidate patients, by the use of, for example, clinical tests, physical examination and medical/family history. A method of assessing the efficacy of a treatment in a subject includes determining the pre- treatment extent of a disorder by methods well known in the art (e.g., determining tumor size or screening for tumor markers where the cell proliferative disorder is cancer) and then administering a therapeutically effective amount of a compound of the present description, to the subject. After an appropriate period of time after the administration of the compound (e.g., 1 day, 1 week, 2 weeks, one month, six months), the extent of the disorder is determined again. The modulation (e.g., decrease) of the extent or invasiveness of the disorder indicates efficacy of the treatment. The extent or invasiveness of the disorder may be determined periodically throughout treatment. For example, the extent or invasiveness of the disorder may be checked every few hours, days or weeks to assess the further efficacy of the treatment. A decrease in extent or invasiveness of the disorder indicates that the treatment is efficacious. The method described may be used to screen or select patients that may benefit from treatment with a compound of the present description.

According to one aspect, there is provided a method for identifying compounds for use in treating autoimmune and inflammatory diseases or conditions which comprises the step of determining whether the compound inhibits the binding of a bromodomain with its cognate acetylated protein.

According to another embodiment, the description provides a method of inhibiting a bromodomain-containing protein (such as a BET protein, e.g., BRD2, BRD3, BRD4, and/or BRDT) using a composition comprising a compound of the present description or a pharmaceutically acceptable derivative thereof and a pharmaceutically acceptable carrier, adjuvant, or vehicle. The amount of a compound of the present description in a provided composition is such that is effective to measurably inhibit one or more bromodomain-containing proteins (such as a BET protein, e.g., BRD2, BRD3, BRD4, and/or BRDT), or a mutant thereof, in a biological sample or in a patient. In certain embodiments, the amount of compound in a provided composition is such that is effective to measurably inhibit one or more bromodomain- containing proteins (such as a BET protein, e.g., BRD2, BRD3, BRD4, and/or BRDT), or a mutant thereof, in a biological sample or in a patient. In certain embodiments, a provided composition is formulated for administration to a patient in need of such composition. In some embodiments, a provided composition is formulated for oral administration to a patient.

In some embodiments, the therapeutically effective amount of a compound as defined herein can be administered to a patient alone or admixed with a pharmaceutically acceptable carrier, adjuvant, or vehicle. The expression "pharmaceutically acceptable carrier, adjuvant, or vehicle" and equivalent expressions, refer to a non-toxic carrier, adjuvant, or vehicle that does not destroy the pharmacological activity of the compound with which it is formulated. Pharmaceutically acceptable carriers, adjuvants or vehicles that may be used in the compositions of this disclosure include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, serum proteins, such as human serum albumin, buffer substances such as phosphates, glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, cellulose-based substances, polyethylene glycol, sodium carboxymethylcellulose, polyacrylates, waxes, polyethylene-polyoxypropylene-block polymers, polyethylene glycol and wool fat.

A "pharmaceutically acceptable derivative" means any non-toxic salt, ester, salt of an ester, prodrug, salt of a prodrug, or other derivative of a compound of the present description that, upon administration to a recipient, is capable of providing, either directly or indirectly, a compound of the present description or an inhibitory active metabolite or residue thereof.

As used herein, the term "inhibitory active metabolite or residue thereof" means that a metabolite or residue thereof is also an inhibitor of one or more bromodomain-containing protein(s) (such as a BET protein, e.g., BRD2, BRD3, BRD4, and/or BRDT), or a mutant thereof.

Compositions described herein may be administered orally, parenterally, by inhalation spray, topically, rectally, nasally, buccally, vaginally or via an implanted reservoir. The term "parenteral" as used herein includes subcutaneous, intravenous, intramuscular, intraarticular, intrasynovial, intrasternal, intrathecal, intrahepatic, intralesional and intracranial injection or infusion techniques. Other modes of administration also include intradermal or transdermal administration.

Liquid dosage forms for oral administration include, but are not limited to, pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs. In addition to the active compounds, the liquid dosage forms may contain inert diluents commonly used in the art such as, for example, water or other solvents, solubilizing agents and emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1 ,3-butylene glycol, dimethylformamide, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor, and sesame oils), glycerol, tetrahydrofurfuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof. Besides inert diluents, the oral compositions can also include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents.

Injectable preparations, for example, sterile injectable aqueous or oleaginous suspensions may be formulated according to the known art using suitable dispersing or wetting agents and suspending agents. The sterile injectable preparation may also be a sterile injectable solution, suspension or emulsion in a nontoxic parenterally acceptable diluent or solvent, for example, as a solution in 1 ,3-butanediol. Among the acceptable vehicles and solvents that may be employed are water, Ringer's solution, U.S. P. and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose any bland fixed oil can be employed including synthetic mono- or diglycerides. In addition, fatty acids such as oleic acid are used in the preparation of injectables.

Injectable formulations can be sterilized, for example, by filtration through a bacterial -retaining filter, or by incorporating sterilizing agents in the form of sterile solid compositions which can be dissolved or dispersed in sterile water or other sterile injectable medium prior to use.

In order to prolong the effect of a provided compound, it is often desirable to slow the absorption of the compound from subcutaneous or intramuscular injection. This may be accomplished by the use of a liquid suspension of crystalline or amorphous material with poor water solubility. The rate of absorption of the compound then depends upon its rate of dissolution that, in turn, may depend upon crystal size and crystalline form. Alternatively, delayed absorption of a parenterally administered compound form is accomplished by dissolving or suspending the compound in an oil vehicle. Injectable depot forms are made by forming microencapsule matrices of the compound in biodegradable polymers such as polylactide-polyglycolide. Depending upon the ratio of compound to polymer and the nature of the particular polymer employed, the rate of compound release can be controlled.

Examples of other biodegradable polymers include poly(orthoesters) and poly(anhydrides). Depot injectable formulations are also prepared by entrapping the compound in liposomes or microemulsions that are compatible with body tissues.

Compositions for rectal or vaginal administration are preferably suppositories which can be prepared by mixing the compounds of the present description with suitable non-irritating excipients or carriers such as cocoa butter, polyethylene glycol or a suppository wax which are solid at ambient temperature but liquid at body temperature and therefore melt in the rectum or vaginal cavity and release the active compound.

Solid dosage forms for oral administration include capsules, tablets, pills, powders, and granules. In such solid dosage forms, the active compound is mixed with at least one inert, pharmaceutically acceptable excipient or carrier such as sodium citrate or dicalcium phosphate and/or a) fillers or extenders such as starches, lactose, sucrose, glucose, mannitol, and silicic acid, b) binders such as, for example, carboxymethylcellulose, alginates, gelatin, polyvinylpyrrolidinone (PVP), sucrose, and acacia, c) humectants such as glycerol, d) disintegrating agents such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate, e) solution retarding agents such as paraffin, f) absorption accelerators such as quaternary ammonium compounds, g) wetting agents such as, for example, cetyl alcohol and glycerol monostearate, h) absorbents such as kaolin and bentonite clay, and i) lubricants such as talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate, and mixtures thereof. In the case of capsules, tablets and pills, the dosage form may also comprise buffering agents.

Solid compositions of a similar type may also be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polyethylene glycols and the like. The solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings and other coatings well known in the pharmaceutical formulating art. They may optionally contain opacifying agents and can also be of a composition that they release the active ingredient(s) only, or preferentially, in a certain part of the intestinal tract, optionally, in a delayed manner. Examples of embedding compositions that can be used include polymeric substances and waxes. Solid compositions of a similar type may also be employed as fillers in soft and hard- filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polyethylene glycols and the like.

Provided compounds can also be in micro-encapsulated form with one or more excipients as noted above. The solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings, release controlling coatings and other coatings well known in the pharmaceutical formulating art. In such solid dosage forms the active compound may be admixed with at least one inert diluent such as sucrose, lactose or starch. Such dosage forms may also comprise, as is normal practice, additional substances other than inert diluents, e.g., tableting lubricants and other tableting aids such a magnesium stearate and microcrystalline cellulose. In the case of capsules, tablets and pills, the dosage forms may also comprise buffering agents. They may optionally contain opacifying agents and can also be of a composition that they release the active ingredient(s) only, or preferentially, in a certain part of the intestinal tract, optionally, in a delayed manner. Examples of embedding compositions that can be used include polymeric substances and waxes.

Dosage forms for topical or transdermal administration of a compound of the present description include ointments, pastes, creams, lotions, gels, powders, solutions, sprays, inhalants or patches. The active component is admixed under sterile conditions with a pharmaceutically acceptable carrier and any needed preservatives or buffers as may be required. Ophthalmic formulation, ear drops, and eye drops are also contemplated as being within the scope of the present description. Additionally, the description contemplates the use of transdermal patches, which have the added advantage of providing controlled delivery of a compound to the body. Such dosage forms can be made by dissolving or dispensing the compound in the proper medium. Absorption enhancers can also be used to increase the flux of the compound across the skin. The rate can be controlled by either providing a rate controlling membrane or by dispersing the compound in a polymer matrix or gel.

Pharmaceutically acceptable compositions provided herein may also be administered by nasal aerosol or inhalation. Such compositions are prepared according to techniques well-known in the art of pharmaceutical formulation and may be prepared as solutions in saline, employing benzyl alcohol or other suitable preservatives, absorption promotors to enhance bioavailability, fluorocarbons, and/or other conventional solubilizing or dispersing agents.

Pharmaceutically acceptable compositions provided herein may be formulated for oral administration. Such formulations may be administered with or without food. In some embodiments, pharmaceutically acceptable compositions of this disclosure are administered without food. In other embodiments, pharmaceutically acceptable compositions of this disclosure are administered with food.

The amount of provided compounds that may be combined with carrier materials to produce a composition in a single dosage form will vary depending upon the patient to be treated and the particular mode of administration. Provided compositions may be formulate such that a dosage of between 0.01 - 100 mg/kg body weight/day of the inhibitor can be administered to a patient receiving these compositions. It should also be understood that a specific dosage and treatment regimen for any particular patient will depend upon a variety of factors, including age, body weight, general health, sex, diet, time of administration, rate of excretion, drug combination, the judgment of the treating physician, and the severity of the particular disease being treated. The amount of a provided compound in the composition will also depend upon the particular compound in the composition.

Compounds or compositions described herein may be administered using any amount and any route of administration effective for treating or lessening the severity of the disorders or diseases as contemplated herein. The exact amount required will vary from subject to subject, depending on the species, age, and general condition of the subject, the severity of the infection, the particular agent, its mode of administration, and the like. Provided compounds are preferably formulated in unit dosage form for ease of administration and uniformity of dosage. The expression "unit dosage form" as used herein refers to a physically discrete unit of agent appropriate for the patient to be treated. It will be understood, however, that the total daily usage of the compounds and compositions of the present disclosure will be decided by the attending physician within the scope of sound medical judgment. The specific effective dose level for any particular patient or organism will depend upon a variety of factors including the disorder being treated and the severity of the disorder; the activity of the specific compound employed; the specific composition employed; the age, body weight, general health, sex and diet of the patient; the time of administration, route of administration, and rate of excretion of the specific compound employed; the duration of the treatment; drugs used in combination or coincidental with the specific compound employed, and like factors well known in the medical arts.

Pharmaceutically acceptable compositions of this disclosure can be administered to humans and other animals orally, rectally, parenterally, intracisternally, intravaginally, intraperitoneally, topically (as by powders, ointments, or drops), buccally, as an oral or nasal spray, or the like, depending on the severity of the infection being treated. In certain embodiments, provided compounds may be administered orally or parenterally at dosage levels of about 0.01 mg/kg to about 50 mg/kg and preferably from about 1 mg/kg to about 25 mg/kg, of subject body weight per day, one or more times a day, to obtain the desired therapeutic effect.

Compounds and compositions described herein are generally useful for the inhibition of activity of one or more proteins involved in epigenetic regulation. Thus, in some embodiments, the present description provides a method of inhibiting one or more proteins involved in epigenetic regulation, such as proteins containing acetyl-lysine recognition motifs, also known as bromodomains (e.g., BET proteins, such as BRD2, BRD3, BRD4, and/or BRDT), by administering a provided compound or composition.

Examples of proteins inhibited by the compounds and compositions described herein and against which the methods described herein are useful include bromodomain-containing proteins, such as BET proteins, such as BRD2, BRD3, BRD4, and/or BRDT, or an isoform or mutant thereof.

The activity of a provided compound, or composition thereof, as an inhibitor of a bromodomain- containing protein, such as a BET protein, such as BRD2, BRD3, BRD4, and/or BRDT, or an isoform or mutant thereof, may be assayed in vitro, in vivo, or in a cell line. In vitro assays include assays that determine inhibition of bromodomain-containing proteins, such as BET proteins, such as BRD2, BRD3, BRD4, and/or BRDT, or a mutant thereof. Alternatively, inhibitor binding may be determined by running a competition experiment where a provided compound is incubated with a bromodomain-containing protein, such as a BET protein, such as BRD2, BRD3, BRD4, and/or BRDT bound to known ligands, labeled or unlabeled. Detailed conditions for assaying a provided compound as an inhibitor of a bromodomain-containing protein, such as a BET protein, such as BRD2, BRD3, BRD4, and/or BRDT or a mutant thereof.

The present description provides for a method of treating a subject with a MYC-dependent cancer, comprising: identifying a subject in need of treatment; administering to the subject a BET inhibitor; determining at least one of MYC mRNA expression, MYC protein expression and tumor mass, and wherein following administration, there is a decrease in at least one of MYC mRNA expression, MYC protein expression and tumor mass, thereby treating the disease.

In one embodiment, the identification step comprises determining whether the subject has at least one of a MYC translocation, a genetic rearrangement of MYC, MYC amplification, MYC over-expression and at least one cellular function that facilitates cellular and/or tumor growth and is altered upon reduction of MYC mRNA or protein expression.

The present description also provides for a method of treating a subject with a MYC-dependent cancer, comprising: determining at least one of MYC mRNA expression, MYC protein expression and tumor mass; administering to the subject a BET inhibitor; and comparing at least one of MYC mRNA expression, MYC protein expression and tumor mass in the subject before and after administration of the BET inhibitor.

The present description also provides a method of treating a subject with a MYC-dependent cancer, comprising: administering to the subject a BET inhibitor that is identified as capable of decreasing at least one of MYC mRNA expression, MYC protein expression and tumor mass; and determining at least one of MYC mRNA expression, MYC protein expression and tumor mass; wherein following the administration, there is a decrease in at least one of MYC mRNA expression, MYC protein expression and tumor mass, thereby treating the disease.

The present description also provides for a method of treating a subject with a disease, comprising: administering a BET inhibitor that is identified as capable of decreasing at least one of MYC mRNA expression, MYC protein expression and tumor mass, wherein following the administration, there is a decrease in at least one of MYC mRNA expression, MYC protein expression and tumor mass, thereby treating the disease.

Acetylated histone recognition and bromodomain-containing proteins (such as BET proteins) have been implicated in proliferative disease. BRD4 knockout mice die shortly after implantation and are compromised in their ability to maintain an inner cell mass, and heterozygotes display pre- and postnatal growth defects associated with reduced proliferation rates. BRD4 regulates genes expressed during M/GI, including growth- associated genes, and remains bound to chromatin throughout the cell cycle (Dey, et al. (2009) Mol. Biol. Cell 20:4899-4909). BRD4 also physically associates with Mediator and P-TEFb (CDK9/cyclin Tl) to facilitate transcriptional elongation (Yang, et al. (2005) Oncogene 24: 1653-1662; Yang, et al. (2005) Mol. Cell 19:535- 545). CDK9 is a validated target in chronic lymphocytic leukemia (CLL), and is linked to c-MYC- dependent transcription (Phelps, et al. Blood 1 13:2637-2645; Rahl, et al. (2010) Cell 141 :432- 445).

BRD4 is translocated to the NUT protein in patients with lethal midline carcinoma, an aggressive form of human squamous carcinoma (French, et al. (2001) Am. J. Pathol. 159: 1987-1992; French, et al. (2003) Cancer Res. 63:304-307). In vitro analysis with RNAi supports a causal role for BRD4 in this recurrent t(15;19) chromosomal translocation. Pharmacologic inhibition of the BRD4 bromodomains results in growth arrest/differentiation of BRD4-NUT cell lines in vitro and in vivo (Filippakopoulos, et al. "Selective Inhibition of BET Bromodomains," Nature (published online September 24, 2010)).

Bromodomain-containing proteins (such as BET proteins) have also been implicated in inflammatory diseases. BET proteins {e.g., BRD2, BRD3, BRD4, and BRDT) regulate assembly of histone acetylation-dependent chromatin complexes that control inflammatory gene expression (Hargreaves, et al. (2009) Cell 138: 129-145; LeRoy, et al. (2008) Mol. Cell 30:51-60; Jang, et al. (2005) Mol. Cell 19:523-534; Yang, et al. (2005) Mol. Cell 19:535-545). Key inflammatory genes (secondary response genes) are down-regulated upon bromodomain inhibition of the BET subfamily, and non-responsive genes (primary response genes) are poised for transcription. BET bromodomain inhibition protects against LPS-induced endotoxic shock and bacteria-induced sepsis in vivo (Nicodeme, et al. "Suppression of Inflammation by a Synthetic Histone Mimic," Nature (published online November 10, 2010)).

Bromodomain-containing proteins (such as BET proteins) also play a role in viral disease. For example, BRD4 is implicated in human papilloma virus (HPV). In the primary phase of HPV infection of basal epithelia, the viral genome is maintained in an extra-chromosomal episome. In some strains of HPV, BRD4 binding to the HPV E2 protein functions to tether the viral genome to chromosomes. E2 is critical for both the repression of E6/E7 and te activation of HPV viral genes. Disruption of BRD4 or the BRD4-E2 interaction blocks E2-dependent gene activation. BRD4 also functions to tether other classes of viral genomes to host chromatin (e.g., Herpesvirus, Epstein-Barr virus).

In certain embodiments, a provided compound inhibits one or more of BRD2, BRD3, BRD4, BRDT, and/or another member of the bromodomain-containing proteins, or a mutant thereof. In some embodiments, a provided compound inhibits two or more of BRD2, BRD3, BRD4, BRDT, and/or another member of the bromodomain-containing proteins, or a mutant thereof. Provided compounds are inhibitors of one of more of the bromodomain-containing proteins, such as BRD2, BRD3, BRD4, and/or BRDT and are therefore useful for treating one or more disorders associated with activity of one or more of the bromodomain-containing proteins, such as BRD2, BRD3, BRD4, and/or BRDT. Thus, in certain embodiments, the present description provides a method for treating an bromodomain-containing protein-mediated disorder, such as a BET- mediated, a BRD2-mediated, a BRD3-mediated, a BRD4-mediated disorder, and/or a BRDT- mediated disorder comprising the step of inhibiting a bromodomain-containing protein, such as a BET protein, such as BRD2, BRD3, BRD4, and/or BRDT, or a mutant thereof, by administering to a patient in need thereof a provided compound, or a pharmaceutically acceptable composition thereof.

As used herein, the terms "bromodomain-containing protein-mediated", "BET-mediated", "BRD2-mediated", "BRD3-mediated", "BRD4-mediated", and/or "BRDT-mediated" disorders or conditions means any disease or other deleterious condition in which one or more of the bromodomain-containing proteins, such as BET proteins, such as BRD2, BRD3, BRD4 and/or BRDT, or a mutant thereof, are known to play a role.

In some embodiments, the BET family bromodomain can be BRD2, BRD3 or BRD4. Accordingly, another embodiment of the present description relates to treating or lessening the severity of one or more diseases in which one or more of the bromodomain-containing proteins, such as BET proteins, such as BRD2, BRD3, BRD4, and/or BRDT, or a mutant thereof, are known to play a role.

Diseases and conditions treatable according to the methods of the present description include, but are not limited to, cancer and other proliferative disorders, inflammatory diseases, sepsis, autoimmune disease, and viral infection. Thus one aspect is a method of treating a subject having a disease, disorder, or symptom thereof the method including administration of a compound or composition herein to the subject. In one embodiment, a human patient is treated with a compound of the present description and a pharmaceutically acceptable carrier, adjuvant, or vehicle, wherein said compound is present in an amount to measurably inhibit bromodomain- containing protein activity (such as BET protein, e.g., BRD2, BRD3, BRD4, and/or BRDT) in the patient.

The present description further relates to the use of provided compounds for the production of pharmaceutical compositions which are employed for the treatment and/or prophylaxis and/or amelioration of the diseases, disorders, illnesses and/or conditions as mentioned herein.

The present description further relates to the use of provided compounds for the production of pharmaceutical compositions which are employed for the treatment and/or prophylaxis of diseases and/or disorders responsive or sensitive to the inhibition of bromodomain-containing proteins, particularly those diseases mentioned above, such as e.g. cancer, inflammatory disease, viral disease.

According to some embodiments, the present description relates to a method of inhibiting bromodomain-containing proteins in a biological sample comprising the step of contacting said biological sample with a provided compound, or a composition thereof.

According to some embodiments, the present description relates to a method of inhibiting a bromodomain-containing protein, such as a BET protein, such as BRD2, BRD3, BRD4 and/or BRDT, or a mutant thereof, activity in a biological sample comprising the step of contacting said biological sample with a provided compound, or a composition thereof.

The term "biological sample", as used herein, includes, without limitation, cell cultures or extracts thereof, biopsied material obtained from a mammal or extracts thereof, and blood, saliva, urine, feces, semen, tears, or other body fluids or extracts thereof. Inhibition of activity of a protein, e.g., a bromodomain-containing protein such as a BET protein (e.g. BRD2, BRD3, BRD4 and/or BRDT), or a mutant thereof, in a biological sample is useful for a variety of purposes that are known to one of skill in the art. Examples of such purposes include, but are not limited to, blood transfusion, organ-transplantation, biological specimen storage, and biological assays.

According to another embodiment, the present description relates to a method of inhibiting activity of one or more bromodomain-containing protein, such as a BET protein, such as BRD2, BRD3, BRD4, and/or BRDT, or a mutant thereof, in a patient comprising the step of administering to said patient a provided compound, or a composition comprising said compound. In certain embodiments, the present description provides a method for treating a disorder mediated by one or more bromodomain-containing proteins, such as a BET protein, such as BRD2, BRD3, BRD4, and/or BRDT, or a mutant thereof, in a patient in need thereof, comprising the step of administering to said patient a provided compound or pharmaceutically acceptable composition thereof. Such disorders are described in detail herein.

Depending upon the particular condition, or disease, to be treated, additional therapeutic agents that are normally administered to treat that condition may also be present in the compositions of this disclosure or administered separately as a part of a dosage regimen. As used herein, additional therapeutic agents that are normally administered to treat a particular disease, or condition, are known as "appropriate for the disease, or condition, being treated."

In some embodiments, the composition of a compound or compounds described herein can be in combination with an additional therapeutic agent.

In some embodiments, the additional therapeutic agent is an epigenetic drug. As used herein, the term "epigenetic drug" refers to a therapeutic agent that targets an epigenetic regulator. Examples of epigenetic regulators include the histone lysine methyltransferases, histone arginine methyl transferases, histone demethylases, histone deacetylases, histone acetylases, and DNA methyltransferases. Histone deacetylase inhibitors include, but are not limited to, vorinostat.

Other therapies, chemotherapeutic agents, or other anti-proliferative agents may be combined with a provided compound to treat proliferative diseases and cancer. Examples of therapies or anticancer agents that may be used in combination with compounds herein described include surgery, radiotherapy (e.g., gamma-radiation, neutron beam radiotherapy, electron beam radiotherapy, proton therapy, brachytherapy, and systemic radioactive isotopes), endocrine therapy, a biologic response modifier (e.g., an interferon, an interleukin, tumor necrosis factor (TNF), hyperthermia and cryotherapy, an agent to attenuate any adverse effects (e.g., an antiemetic), and any other approved chemotherapeutic drug.

A provided compound may also be used to advantage in combination with one or more antiproliferative compounds. Such antiproliferative compounds include an aromatase inhibitor; an anti-estrogen; an anti-androgen; a gonadorelin agonist; a topoisomerase I inhibitor; a topoisomerase II inhibitor; a microtubule active agent; an alkylating agent; a retinoid, a carotenoid, or a tocopherol; a cyclooxygenase inhibitor; an MMP inhibitor; an mTOR inhibitor; an antimetabolite; a platin compound; a methionine aminopeptidase inhibitor; a bisphosphonate; an antiproliferative antibody; a heparanase inhibitor; an inhibitor of Ras oncogenic isoforms; a telomerase inhibitor; a proteasome inhibitor; a compound used in the treatment of hematologic malignancies; a Flt-3 inhibitor; an Hsp90 inhibitor; a kinesin spindle protein inhibitor; a MEK inhibitor; an antitumor antibiotic; a nitrosourea; a compound targeting/decreasing protein or lipid kinase activity, a compound targeting/decreasing protein or lipid phosphatase activity, or any further anti-angiogenic compound.

Exemplary aromatase inhibitors include steroids, such as atamestane, exemestane and formestane, and non-steroids, such as aminoglutethimide, rogletimide, pyridoglutethimide, trilostane, testolactone, ketoconazole, vorozole, fadrozole, anastrozole and letrozole.

Exemplary anti-estrogens include tamoxifen, fulvestrant, raloxifene and raloxifene hydrochloride. Anti-androgens include, but are not limited to, bicalutamide. Gonadorelin agonists include, but are not limited to, abarelix, goserelin and goserelin acetate.

Exemplary topoisomerase I inhibitors include topotecan, gimatecan, irinotecan, camptothecin and its analogues, 9-nitrocamptothecin and the macromolecular camptothecin conjugate PNU- 166148. Topoisomerase II inhibitors include, but are not limited to, the anthracyclines such as doxorubicin, daunorubicin, epirubicin, idarubicin and nemorubicin, the anthraquinones mitoxantrone and losoxantrone, and the podophillotoxins etoposide and teniposide.

Exemplary microtubule active agents include microtubule stabilizing, microtubule destabilizing compounds and microtubulin polymerization inhibitors including, but not limited to taxanes, such as paclitaxel and docetaxel; vinca alkaloids, such as vinblastine or vinblastine sulfate, vincristine or vincristine sulfate, and vinorelbine; discodermolides; colchicine and epothilones and derivatives thereof. Exemplary alkylating agents include cyclophosphamide, ifosfamide, melphalan or nitrosoureas such as carmustine and lomustine.

Exemplary cyclooxygenase inhibitors include Cox-2 inhibitors, 5-alkyl substituted 2- arylaminophenylacetic acid and derivatives, such as celecoxib, rofecoxib, etoricoxib, valdecoxib or a 5-alkyl-2-arylaminophenylacetic acid, such as lumiracoxib.

Exemplary matrix metalloproteinase inhibitors ("MMP inhibitors") include collagen peptidomimetic and non-peptidomimetic inhibitors, tetracycline derivatives, batimastat, marimastat, prinomastat, metastat, BMS-279251 , BAY 12-9566, TAA21 1 , MMI270B, and AAJ996.

Exemplary mTOR inhibitors include compounds that inhibit the mammalian target of rapamycin (mTOR) and possess antiproliferative activity such as sirolimus, everolimus, CCI-779, and ABT578.

Exemplary antimetabolites include 5-fluorouracil (5-FU), capecitabine, gemcitabine, DNA demethylating compounds, such as 5-azacytidine and decitabine, methotrexate and edatrexate, and folic acid antagonists such as pemetrexed.

Exemplary platin-containing compounds include carboplatin, cisplatin, nedaplatin, and oxaliplatin.

Exemplary methionine aminopeptidase inhibitors include bengamide or a derivative thereof and PPI-2458.

Exemplary bisphosphonates include etidronic acid, clodronic acid, tiludronic acid, pamidronic acid, alendronic acid, ibandronic acid, risedronic acid and zoledronic acid.

Exemplary antiproliferative antibodies include trastuzumab, trastuzumab-DMI, cetuximab, bevacizumab, rituximab, PR064553, and 2C4. The term "antibody" is meant to include intact monoclonal antibodies, polyclonal antibodies, multispecific antibodies formed from at least two intact antibodies, and antibody fragments, so long as they exhibit the desired biological activity.

Exemplary heparanase inhibitors include compounds that target, decrease or inhibit heparin sulfate degradation, such as PI- 88 and OGT2115.

The term "an inhibitor of Ras oncogenic isoforms," such as H-Ras, K-Ras, or N-Ras, as used herein refers to a compound which targets, decreases, or inhibits the oncogenic activity of Ras; for example, a farnesyl transferase inhibitor such as L-744832, DK8G557, tipifarnib, and lonafarnib. Exemplary telomerase inhibitors include compounds that target, decrease or inhibit the activity of telomerase, such as compounds which inhibit the telomerase receptor, such as telomestatin.

Exemplary proteasome inhibitors include compounds that target, decrease or inhibit the activity of the proteasome including, but not limited to, bortezomib.

The phrase "compounds used in the treatment of hematologic malignancies" as used herein includes FMS-like tyrosine kinase inhibitors, which are compounds targeting, decreasing or inhibiting the activity of FMS-like tyrosine kinase receptors (Flt-3R); interferon, Ι-β-D- arabinofuransylcytosine (ara-c) and busulfan; and ALK inhibitors, which are compounds which target, decrease or inhibit anaplastic lymphoma kinase.

Exemplary Flt-3 inhibitors include PKC412, midostaurin, a staurosporine derivative, SU11248 and MLN518.

Exemplary HSP90 inhibitors include compounds targeting, decreasing or inhibiting the intrinsic ATPase activity of HSP90; degrading, targeting, decreasing or inhibiting the HSP90 client proteins via the ubiquitin proteosome pathway. Compounds targeting, decreasing or inhibiting the intrinsic ATPase activity of HSP90 are especially compounds, proteins or antibodies which inhibit the ATPase activity of HSP90, such as 17-allylamino, 17-demethoxygeldanamycin (17AAG), a geldanamycin derivative; other geldanamycin related compounds; radicicol and HDAC inhibitors.

The phrase "a compound targeting/decreasing a protein or lipid kinase activity; or a protein or lipid phosphatase activity; or any further anti-angiogenic compound" as used herein includes a protein tyrosine kinase and/or serine and/or threonine kinase inhibitor or lipid kinase inhibitor, such as a) a compound targeting, decreasing or inhibiting the activity of the platelet-derived growth factor-receptors (PDGFR), such as a compound which targets, decreases, or inhibits the activity of PDGFR, such as an N-phenyl-2-pyrimidine-amine derivatives, such as imatinib, SU101 , SU6668 and GFB-1 11 ; b) a compound targeting, decreasing or inhibiting the activity of the fibroblast growth factor-receptors (FGFR); c) a compound targeting, decreasing or inhibiting the activity of the insulin-like growth factor receptor I (IGF-IR), such as a compound which targets, decreases, or inhibits the activity of IGF-IR; d) a compound targeting, decreasing or inhibiting the activity of the Trk receptor tyrosine kinase family, or ephrin B4 inhibitors; e) a compound targeting, decreasing or inhibiting the activity of the Axl receptor tyrosine kinase family; f) a compound targeting, decreasing or inhibiting the activity of the Ret receptor tyrosine kinase; g) a compound targeting, decreasing or inhibiting the activity of the Kit/SCFR receptor tyrosine kinase, such as imatinib; h) a compound targeting, decreasing or inhibiting the activity of the c-Kit receptor tyrosine kinases, such as imatinib; i) a compound targeting, decreasing or inhibiting the activity of members of the c-Abl family, their gene-fusion products (e.g. Bcr-Abl kinase) and mutants, such as an N-phenyl-2-pyrimidine-amine derivative, such as imatinib or nilotinib; PD180970; AG957; NSC 680410; PD173955; or dasatinib; j) a compound targeting, decreasing or inhibiting the activity of members of the protein kinase C (PKC) and Raf family of serine/threonine kinases, members of the MEK, SRC, JAK, FAK, PDK1 , PKB/Akt, and Ras/MAPK family members, and/or members of the cyclin-dependent kinase family (CDK), such as a staurosporine derivative disclosed in US 5,093,330, such as midostaurin; examples of further compounds include UCN-01 , safingol, BAY 43-9006, bryostatin 1 , perifosine; ilmofosine; RO 318220 and RO 320432; GO 6976; ISIS 3521 ; LY333531/LY379196; a isochinoline compound; a farnesyl transferase inhibitor; PD184352 or QAN697, or AT7519; k) a compound targeting, decreasing or inhibiting the activity of a protein-tyrosine kinase, such as imatinib mesylate or a tyrphostin such as Tyrphostin A23/RG-50810; AG 99; Tyrphostin AG 213; Tyrphostin AG 1748; Tyrphostin AG 490; Tyrphostin B44; Tyrphostin B44 (+) enantiomer; Tyrphostin AG 555; AG 494; Tyrphostin AG 556, AG957 and adaphostin (4-{ [(2,5- dihydroxyphenyl)methyl] amino} -benzoic acid adamantyl ester; NSC 680410, adaphostin); 1) a compound targeting, decreasing or inhibiting the activity of the epidermal growth factor family of receptor tyrosine kinases (EGFR, ErbB2, ErbB3, ErbB4 as homo- or heterodimers) and their mutants, such as CP 358774, ZD 1839, ZM 105180; trastuzumab, cetuximab, gefitinib, erlotinib, OSI-774, CI-1033, EKB-569, GW-2016, antibodies El. I, E2.4, E2.5, E6.2, E6.4, E2.I I, E6.3 and E7.6.3, and 7H-pyrrolo-[2,3-d]pyrimidine derivatives; and m) a compound targeting, decreasing or inhibiting the activity of the c-Met receptor.

Exemplary compounds that target, decrease or inhibit the activity of a protein or lipid phosphatase include inhibitors of phosphatase 1 , phosphatase 2A, or CDC25, such as okadaic acid or a derivative thereof.

Further anti-angiogenic compounds include compounds having another mechanism for their activity unrelated to protein or lipid kinase inhibition, e.g. thalidomide and TNP-470.

Additional exemplary chemotherapeutic compounds, one or more of which may be used in combination with provided compounds, include: daunorubicin, adriamycin, Ara-C, VP- 16, teniposide, mitoxantrone, idarubicin, carboplatinum, PKC412, 6-mercaptopurine (6-MP), fludarabine phosphate, octreotide, SOM230, FTY720, 6-thioguanine, cladribine, 6- mercaptopurine, pentostatin, hydroxyurea, 2-hydroxy-IH-isoindole-l,3-dione derivatives,!- (4- chloroanilino)-4-(4-pyridylmethyl)phthalazine or a pharmaceutically acceptable salt thereof, l-(4- chloroanilino)-4-(4-pyridylmethyl)phthalazine succinate, angiostatin, endostatin, anthranilic acid amides, ZD4190, ZD6474, SU5416, SU6668, bevacizumab, rhuMAb, rhuFab, macugen; FLT-4 inhibitors, FLT-3 inhibitors, VEGFR-2 IgGI antibody, RPI 4610, bevacizumab, porfimer sodium, anecortave, triamcinolone, hydrocortisone, 1 1a-epihydrocotisol, cortexolone, 17a- hydroxyprogesterone, corticosterone, desoxycorticosterone, testosterone, estrone, dexamethasone, fluocinolone, a plant alkaloid, a hormonal compound and/or antagonist, a biological response modifier, such as a lymphokine or interferon, an antisense oligonucleotide or oligonucleotide derivative, shRNA or siRNA, or a miscellaneous compound or compound with other or unknown mechanism of action.

For a more comprehensive discussion of updated cancer therapies see: The Merck Manual, 17 th Ed. 1999. See also the National Cancer Institute (CNI) website (www.nci.nih.gov) and the Food and Drug Administration (FDA) website for a list of the FDA approved oncology drugs.

Other examples of additional therapeutic agents, one or more of which a provided compound may also be combined with include: a treatment for Alzheimer's Disease such as donepezil and rivastigmine; a treatment for Parkinson's Disease such as L-DOPA/carbidopa, entacapone, ropinirole, pramipexole, bromocriptine, pergolide, trihexyphenidyl, and amantadine; an agent for treating multiple sclerosis (MS) such as beta interferon {e.g. , Avonex® and Rebif®), glatiramer acetate, and mitoxantrone; a treatment for asthma such as albuterol and montelukast; an agent for treating schizophrenia such as zyprexa, risperdal, seroquel, and haloperidol; an antiinflammatory agent such as a corticosteroid, a TNF blocker, IL-1 RA, azathioprine, cyclophosphamide, and sulfasalazine; an immunomodulatory agent, including immunosuppressive agents, such as cyclosporin, tacrolimus, rapamycin, mycophenolate mofetil, an interferon, a corticosteroid, cyclophosphamide, azathioprine, and sulfasalazine; a neurotrophic factor such as an acetylcholinesterase inhibitor, an MAO inhibitor, an interferon, an anti-convulsant, an ion channel blocker, riluzole, or an anti-Parkinson's agent; an agent for treating cardiovascular disease such as a beta-blocker, an ACE inhibitor, a diuretic, a nitrate, a calcium channel blocker, or a statin; an agent for treating liver disease such as a corticosteroid, cholestyramine, an interferon, and an anti- viral agent; an agent for treating blood disorders such as a corticosteroid, an anti-leukemic agent, or a growth factor; or an agent for treating immunodeficiency disorders such as gamma globulin.

The above-mentioned compounds, one or more of which can be used in combination with a provided compound, can be prepared and administered as described in the art. Provided compounds can be administered alone or in combination with one or more other therapeutic compounds, possible combination therapy taking the form of fixed combinations or the administration of a provided compound and one or more other therapeutic compounds being staggered or given independently of one another, or the combined administration of fixed combinations and one or more other therapeutic compounds. Provided compounds can besides or in addition be administered especially for tumor therapy in combination with chemotherapy, radiotherapy, immunotherapy, phototherapy, surgical intervention, or a combination of these. Long-term therapy is equally possible as is adjuvant therapy in the context of other treatment strategies, as described above. Other possible treatments are therapy to maintain the patient's status after tumor regression, or even chemopreventive therapy, for example in patients at risk.

Such additional agents may be administered separately from a composition containing a provided compound, as part of a multiple dosage regimen. Alternatively, those agents may be part of a single dosage form, mixed together with a provided compound in a single composition. If administered as part of a multiple dosage regimen, the two active agents may be submitted simultaneously, sequentially or within a period of time from one another normally within five hours from one another.

Upon improvement of a subject's condition, a maintenance dose of a compound, composition or combination of the present description may be administered, if necessary. Subsequently, the dosage or frequency of administration, or both, may be reduced, as a function of the symptoms, to a level at which the improved condition is retained when the symptoms have been alleviated to the desired level, treatment should cease. The subject may, however, require intermittent treatment on a long-term basis upon any recurrence of disease symptoms.

It will be understood, however, that the total daily usage of the compounds and compositions of the present description will be decided by the attending physician within the scope of sound medical judgment. The specific inhibitory dose for any particular patient will depend upon a variety of factors including the disorder being treated and the severity of the disorder; the activity of the specific compound employed; the specific composition employed; the age, body weight, general health, sex and diet of the patient; the time of administration, route of administration, and rate of excretion of the specific compound employed; the duration of the treatment; drugs used in combination or coincidental with the specific compound employed; and like factors well known in the medical arts.

The total daily inhibitory dose of the compounds of the present description administered to a subject in single or in divided doses can be in amounts, for example, from 0.01 to 50 mg/kg body weight or more usually from 0.1 to 25 mg/kg body weight. Single dose compositions may contain such amounts or submultiples thereof to make up the daily dose. In one embodiment, treatment regimens according to the present description comprise administration to a patient in need of such treatment from about 10 mg to about 1000 mg of the compound(s) of the present description per day in single or multiple doses.

As used herein, the term "combination," "combined," and related terms refers to the simultaneous or sequential administration of therapeutic agents in accordance with the present description. For example, a provided compound may be administered with another therapeutic agent simultaneously or sequentially in separate unit dosage forms or together in a single unit dosage form. Accordingly, an embodiment of the present description provides a single unit dosage form comprising a provided compound, an additional therapeutic agent, and a pharmaceutically acceptable carrier, adjuvant, or vehicle for use in the methods of the present description.

The amount of both, a provided compound and additional therapeutic agent (in those compositions which comprise an additional therapeutic agent as described above) that may be combined with the carrier materials to produce a single dosage form will vary depending upon the host treated and the particular mode of administration. Preferably, compositions should be formulated such that a dosage of between 0.01 - 100 mg/kg body weight/day of a provided compound can be administered.

In those compositions which comprise an additional therapeutic agent, that additional therapeutic agent and the provided compound may act synergistically. Therefore, the amount of additional therapeutic agent in such compositions will be less than that required in a monotherapy utilizing only that therapeutic agent. In such compositions a dosage of between 0.01 - 1 ,000 g/kg body weight/day of the additional therapeutic agent can be administered.

The amount of additional therapeutic agent present in the compositions of this disclosure will be no more than the amount that would normally be administered in a composition comprising that therapeutic agent as the only active agent. Preferably the amount of additional therapeutic agent in the presently disclosed compositions will range from about 50% to 100% of the amount normally present in a composition comprising that agent as the only therapeutically active agent. Provided compounds, or pharmaceutical compositions thereof, may also be incorporated into compositions for coating an implantable medical device, such as prostheses, artificial valves, vascular grafts, stents and catheters. Vascular stents, for example, have been used to overcome restenosis (re-narrowing of the vessel wall after injury). However, patients using stents or other implantable devices risk clot formation or platelet activation. These unwanted effects may be prevented or mitigated by pre-coating the device with a pharmaceutically acceptable composition comprising a provided compound. Implantable devices coated with a compound of the present description are another embodiment of the present description.

In another aspect, the present description provides a method of method of synthesizing a compound of any of the formulae herein. Another embodiment is a method of making a compound of any of the formulae herein using any one, or combination of, reactions delineated herein. The method can include the use of one or more intermediates or chemical reagents delineated herein.

The recitation of a listing of chemical groups in any definition of a variable herein includes definitions of that variable as any single group or combination of listed groups. The recitation of an embodiment for a variable herein includes that embodiment as any single embodiment or in combination with any other embodiments or portions thereof. The recitation of an embodiment herein includes that embodiment as any single embodiment or in combination with any other embodiments or portions thereof.

EXAMPLES

The Examples set forth herein below provide syntheses and experimental results obtained for certain exemplary compounds. Unless otherwise indicated, all numbers expressing quantities of ingredients, reaction conditions, concentrations, properties, stabilities, and so forth used in the specification and claims are to be understood as being modified in all instances by the term "about." At the very least, each numerical parameter should at least be construed in light of the number of reported significant digits and by applying ordinary rounding techniques. Accordingly, unless indicated to the contrary, the numerical parameters set forth in the present specification and attached claims are approximations that may vary depending upon the properties sought to be obtained. Notwithstanding that the numerical ranges and parameters setting forth the broad scope of the embodiments are approximations, the numerical values set forth in the specific examples are reported as precisely as possible. Any numerical value, however, inherently contain certain errors resulting from variations in experiments, testing measurements, statistical analyses and such.

The following is to be construed as merely illustrative, and not limitations of the preceding disclosure in any way whatsoever. Those skilled in the art will promptly recognize appropriate variations from the procedures both as to reactants and as to reaction conditions and techniques. In some cases, starting materials or intermediates may be commercially available. Commercial material may be generally available from known sources, for example, Sigma- Aldrich, Bachem, Lancaster, Alfa Aesar, etc. Chemical Synthesis of Exemplary Compounds

General:

All temperatures are in degrees Celsius (°C) and are uncorrected. Reagent grade chemicals and anhydrous solvent were purchased from commercial sources and unless otherwise mentioned, were used without further purification The names of the products were determined using the naming software included in the Contour Software AB electronic lab notebook iLabber™ version 4.1 1.3075.18678. Flash chromatography was performed on Teledyne Isco instruments using pre-packaged disposable Si0 2 stationary phase columns with eluent flow rate range of 5 to 300 mL/min, UV detection (254 and 280 nm). Unless otherwise indicated, HPLC purifications were performed on a Gilson™ HPLC with a Phenomenex Gemini™ column, C18, 150:30 mm, 5 micron, eluting at 40 mL/min with mixtures of MeOH and water containing 0.1 % (NH 4 ) 2 C0 3 (high pH), or mixtures of MeCN and water containing 0.1 % formic acid (low pH). Chiral isomer separations were performed, for instance, on a Minigram Semi- Preparative SFC™ from Mettler-Toledo. Analytical HPLC chromatograms were performed using an Agilent 1 100 series instrument. The mass spectra were recorded with a Waters Micromass ZQ™ detector at 120 °C. The mass spectrometer was equipped with an electrospray ion source (ESI) operated in a positive ion mode and was set to scan between m/z 150-750 with a scan time of 0.3 s. Products and intermediates were analyzed by HPLC/MS on a X-Bridge™ C 18 , (3.5 μΜ, 2.10 x 30 mm) using a high pH buffer gradient of 5% to 95% of MeOH in H 2 0 (0.03% (NH 4 ) 2 C0 3 / 0.375% NH 4 OH) over 4.5 min at 1 mL/min for a 6 min run. The 1 H NMR spectra were recorded on a Bruker UltraShield™ 500 MHz/54 mm instrument (BZH 43/500/70B, D221/54-3209) or on a Bruker Ultra Shield Avance 400 MHz/5 mm Probe (BBFO). The chemical shifts are reported in part-per-million from a tetramethylsilane standard.

As used herein, the following abbreviations may have the following meanings:

Abbreviation Term

AcOH Acetic acid

BF 3 « Et 2 0 Boron trifluoride diethyl etherate

CDCI 3 Deuterated chloroform

CHCI 3 Chloroform

m-CPBA 3-chloroperoxybenzoic acid CS 2 C0 3 Cesium carbonate

d Day(s)

DCE 1 ,2-dichloroethane

DCM Dichloromethane

DEA Diethylamine

DIPEA N,N-diisopropylethylamine

DME 1 ,2-dimethoxyethane

DMF Ν,Ν-dimethyl formamide

DMSO Dimethylsulfoxide

Et 2 0 Diethyl ether

EtOAc Ethyl acetate

Et 3 SiH Triethylsilane

h Hour(s)

HATU (dimethylamino)-/V,/\/-dimethyl(3/- -[1 ,2,3]tnazolo[4,5- jb]pyridin-3-yloxy)methaniminium hexafluorophosphate

HCI Hydrochloric acid

HNO3 Nitric Acid

HPLC High-performance liquid chromatography

H2SO4 Sulfuric Acid

I PA Isopropanol

K2CO3 Potassium carbonate

KOH Potassium hydroxide

LaCI 3 « 2LiCI Lanthanum(lll) chloride bis(lithium chloride) complex

LC-MS Liquid chromatography mass spectrum

ϋΟΗ·Η 2 0 Lithium hydroxide monohydrate

min Minute(s)

MeCN Acetonitrile

MeOD Deuterated methanol

MeOH Methanol

MgS0 4 Magnesium sulfate

MS (ESI) Electrospray ionization mass spectrometry

N 2 Nitrogen

NaBH 4 Sodium borohydride

Na 2 C0 3 Sodium carbonate

NaHC0 3 Sodium bicarbonate

Na(OAc) 3 BH Sodium triacetoxyborohydride

NaOH Sodium hydroxide

Na 2 S0 4 Sodium sulfate

NH 4 CI Ammonium chloride

N H4OH Ammonium hydroxide

NMR Nuclear magnetic resonance

Pd(PPh 3 ) 4 Tetrakis(triphenylphosphine)palladium(0)

POCI 3 Phosphorus(V) oxychloride

rt Room temperature

SFC Supercritical fluid chromatography

SOCI 2 Thionyl chloride

TEA Triethylamine

THF Tetrahydrofuran

TFA Trifluoroacetic acid Scheme 1. Preparation of a imidazo[1 ,2-a]pyridine core:

B

wherein R 5 is as herein defined, CH 2 R, CH(OH)R, CH(OMe)R and CH 2 XR each represents embodiment of R 1 as herein defined, wherein X is O or NH. Scheme 2. Preparation of the imidazo[1,2-a]pyridine carbonyl core:

wherein R x is hydrogen or R a , R y is R a , or R x and R y are taken together to form a heterocycloalkyi or heteroaryl, wherein R a is the same or different in each occurrence and is as defined herein.

Scheme 3. Preparation of the imidazo[1,2-a]pyridine sulfonyl core:

wherein R x is hydrogen or R a , R y is R a , or R x and R y are taken together to form a heterocycloalkyi or heteroaryl, wherein R a is the same or different in each occurrence and is as defined herein.

Intermediate 1: 6-bromo-3-(chloromethyl)imidazo[ 1 ,2-a]pyridine

Step 1: Preparation of (6-bromoimidazo[1 ,2-a]pyridin-3-yl) methanol

To solid NaBH 4 (8 mg, 0.227 mmol) was added water (0.1 mL) and then 6-bromoimidazo[ .2- a]pyridine-3-carbaldehyde (100 mg, 0.444 mmol), MeOH (0.7 mL) and DCM (0.7 mL) were added and the reaction mixture was stirred at rt for 2 h. The mixture was concentrated under reduced pressure and the material was triturated with ether and then concentrated under reduced pressure to provide the title compound (74 mg, 73%). 1 H NMR (500 MHz, DMSO) δ 8.65 (d, J = 1.1 Hz, 1 H), 7.57 (d, J = 9.1 Hz, 1 H), 7.53 (s, 1 H), 7.38 (dd, J = 9.5, 1.9 Hz, 1 H), 5.29 (bs, 1 H), 4.80 (s, 2H). MS (ESI) [M+H] + 227.0/229.0.

Step 2: Preparation of 6-bromo-3-(chloromethyl)imidazo[1 ,2-a]pyridine

To (6-bromoimidazo[1 ,2-a]pyridin-3-yl)methanol (0.1 g, 0.44 mmol) was added SOCI 2 (1 mL) and the mixture was heated to 60 °C for 2 h. The solution was cooled and then concentrated under reduced pressure to afford Intermediate 1 (140 mg), which was used in the next step without any further purification. 1 H NMR (500 MHz, DMSO) δ 9.33 - 9.15 (m, 1 H), 8.32 (s, 1 H), 8.1 1 (dd, J = 9.5, 1.7 Hz, 1 H), 7.98 (dd, J = 9.5, 0.8 Hz, 1 H), 5.39 (s, 2H). MS (ESI) [M+H] + 241.1 , 243.1.

Intermediate 2: 6-( 1, 5-dimethyl-6-oxo- 1, 6-dihydropyridin-3-yl)imidazo[ 1 ,2-a]pyridine-3- carboxylic acid

Step 1: Preparation of ethyl 6-(1,5-dimethyl-6-oxo-1,6-dihydropyridin-3-yl)imidazo[1,2- a ]pyridine-3-carboxylate

A stirred solution of ethyl 6-bromoimidazo[1 ,2-a]pyridine-3-carboxylate (1 g, 3.71 mmol) and 1 ,3-dimethyl-5-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)pyridin-2(1 H)-one (prepared as in US20130053362, 1.1 1 g, 4.46 mmol) in 1 ,4-dioxane (20 mL) was purged with N 2 for 10 min followed by addition of a solution of Na 2 C0 3 (1.2 g, 11.15 mmol) in water (4.2 mL) and was again purged with N 2 for 10 min. After 10 min, Pd(PPh 3 ) 4 (0.22 g, 0.19 mmol) was added and the reaction mixture was heated at 100 °C for 3 h. The solvent was evaporated under vacuum, water (100 mL) was added and the aqueous layer was extracted with EtOAc (3 x 50 mL). The combined organic layers were washed with brine (100 mL), dried over Na 2 S0 4 , filtered and evaporated under reduced pressure. The material was purified by flash chromatography on silica gel using a mixture of 2% methanol in DCM as eluent to afford the title compound (1 g, 86%) as a solid. 1 H NMR (400 MHz, DMSO) δ ppm 9.31 (s, 1 H), 8.29 (s, 1 H), 8.09 (d, J = 2.4 Hz, 1 H), 7.88 (d, J = 9.2 Hz, 1 H), 7.80 (dd, J = 2 and 1.6 Hz, 1 H), 7.70 (d, J = 1.2 Hz, 1 H), 4.37 (q, J = 7.2 Hz, 2H), 3.54 (s, 3H), 2.10 (s, 3H), 1.36 (t, 3H). MS (ESI) [M+H] + 312.3.

Step 2: Preparation of 6-(1,5-dimethyl-6-oxo-1,6-dihydropyridin-3-yl)imidazo[1,2-a] pyridin carboxylic acid

To a stirred solution of ethyl 6-(1 ,5-dimethyl-6-oxo-1 ,6-dihydropyridin-3-yl)imidazo[1 ,2- a]pyridine-3-carboxylate (1 g, 3.21 mmol) in THF (20 mL) and water (20 mL) was added LiOH « H 2 0 (0.15 g, 3.53 mmol) at rt and the reaction mixture was stirred for 3 h. The mixture was acidified with diluted HCI (pH= 2) and the resulting solid was filtered, washed with water (50 mL) and dried under vacuum to afford Intermediate 2 (0.8 g, 88%) as a solid. 1 H NMR (400 MHz, DMSO) δ ppm 13.39 (bs, 1 H), 9.34 (s, 1 H), 8.27 (s, 1 H), 8.06 (d, J = 2.4 Hz, 1 H), 7.85 (d, J= 9.2 Hz, 1 H), 7.74 (dd, J = 2.0 and 1.6 Hz, 1 H), 7.68 (d, J = 1.2 Hz, 1 H), 3.54 (s, 3H), 2.11 (s, 3H). MS (ESI) [M+H] + 284.3. Intermediate 3: 6-bromoimidazo[1,2-a]pyridine-3-sulfonyl chloride

Step 1: Preparation of 6-bromoimidazo[1 ,2-a]pyridine-3-sulfonic acid

To a stirred solution of 6-bromoimidazo[1 ,2-a]pyridine (1 g, 5.08 mmol) in chloroform (15 mL), a solution of chlorosulfonic acid (1 mL, 15.23 mmol) in chloroform (10 mL) was added and the reaction mixture was refluxed for 16 h. The mixture was concentrated under vacuum, the material was treated with a mixture of diethyl ether and ethanol (2: 1 , 30 mL), and the resulting solid was filtered to afford the title compound (1.2 g, 85%). 1 H NMR (400 MHz, DMSO) δ ppm 8.92 (t, J = 1 Hz, 1 H), 8.28 (s, 1 H), 8.10 (dd, J = 1.6 and 9.6 Hz, 1 H), 7.37 (dd, J t J 2 = 0.4 Hz, 1 H). MS (ESI) [M+H] + 279.5.

Step 2: Preparation of 6-bromoimidazo[1 ,2-a]pyridine-3-sulfonyl chloride

To 6-bromoimidazo[1 ,2-a]pyridine-3-sulfonic acid (1.2 g, 4.33 mmol) was added POCI 3 (12 mL, 128.53 mmol) and the reaction mixture was heated at reflux temperature for 16 h. The solution was evaporated under reduced pressure, the residue was diluted with water (50 mL) and the aqueous layer was extracted with DCM (3 x 50 mL). The organic layers were combined, dried over Na 2 S0 4 , filtered and evaporated under reduced pressure to afford Intermediate 3 (0.8 g, 62%) as a solid, which was used in the next step without purification. MS (ESI) [M+H] + 297.4. Intermediate 4: 5-(3-aminoimidazo[ 1,2-a]pyridin-6-yl)- 1 ,3-dimethyl-pyridin-2-one

Step 1: Preparation of 6-bromo-3-nitro-imidazo[1 ,2-a]pyridine

To a solution of 6-bromoimidazo[1 ,2-a]pyridine (100 mg, 0.508 mmol) in concentrated H 2 S0 4 (0.4 ml_) was added at rt concentrated HN0 3 (0.12 ml_, 2.69 mmol) and the reaction mixture was stirred for 15 min. The mixture was quenched by the addition of ice and the resulting solid (123 mg, 99%) was collected by filtration and used directly in the next step without further purification. 1 H NMR (500 MHz, DMSO) δ 9.40 (t, J = 1.4 Hz, 1 H), 8.78 (s, 1 H), 8.00 - 7.85 (m, 2H). MS (ESI) [M+H] + 242.1/244.1.

Step 2: Preparation of 1 ,3-dimethyl-5-(3-nitroimidazo[ 1,2-a]pyridin-6-yl)pyridin-2-one

To a suspension of 6-bromo-3-nitro-imidazo[1 ,2-a]pyridine (535 mg, 2.21 mmol) in DME (5 ml_) was added 1 ,3-dimethyl-5-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)pyridin-2-one (prepared using the procedure described in US20130053362, 661 mg, 2.65 mmol), Cs 2 C0 3 (1.8 g, 5.53 mmol), Pd(PPh 3 ) (256 mg, 0.22 mmol) and water (1 ml_). The reaction mixture was degassed with N 2 and then heated in a sealed tube at 90 °C for 18 h. The mixture was cooled to rt and the resulting solid was collected by filtration, washed with water and dried under vacuum to afford the title compound (472 mg, 75%), which was used directly in the next step without further purification. 1 H NMR (500 MHz, DMSO) δ 9.39 (s, 1 H), 8.78 (s, 1 H), 8.17 (d, J = 2.5 Hz, 1 H), 8.06 - 7.98 (m, 2H), 7.78 (d, J = 1.4 Hz, 1 H), 3.55 (s, 3H), 2.1 1 (s, 3H). MS (ESI) [M+H] + 285.2.

Step 3: Preparation of 5-(3-aminoimidazo[ 1 ,2-a]pyridin-6-yl)-1 ,3-dimethyl-pyridin-2-one

To a suspension of 1 ,3-dimethyl-5-(3-nitroimidazo[1 ,2-a]pyridin-6-yl)pyridin-2-one (160 mg, 0.56 mmol) in MeOH (10 mL) was added AcOH (0.10 mL, 1.69 mmol) and zinc (368 mg, 5.63 mmol) and the resulting suspension mixture was stirred at rt for 1 h. The mixture was filtered through a pad of Celite™, washed with MeOH and then the filtrate was concentrated under reduced pressure. The residue was triturated with a DCM/MeOH mixture to afford Intermediate 4 (91 mg, 64%) as a solid. MS (ESI) [M+H] + 255.2.

Example 1 : 5-(3-benzylimidazo[1 ,2-a]pyridin-6-yl)-1 ,3-dimethyl-pyridin-2-one (Compound 1)

Step 1: Preparation of (6-bromoimidazo[ 1,2-a]pyridin-3-yl)-phenyl-methanol

To a solution of phenyl magnesium bromide (3 M in THF, 0.40 mL, 1.21 mmol) in dry THF (0.5 mL) was added a solution of 6-bromoimidazo[1 ,2-a]pyridine-3-carbaldehyde (210 mg, 0.933 mmol) in THF (5 mL) at -30 °C. More phenyl magnesium bromide (0.4 mL, 1.21 mmol) was added and the mixture was stirred for 30 min at -30 °C. The mixture was then quenched with water and concentrated under reduced pressure. EtOAc (50 mL) and saturated NH 4 CI (10 mL) were added and the phases were separated. The aqueous phase was then extracted with EtOAc (3 x 30 mL). The combined organic layers were washed with brine, filtered, dried and evaporated under reduced pressure to provide the title compound, which was used in the next step without any further purification (230 mg, 81 %). MS (ESI) [M+H] + 303.1/305.1.

Step 2: Preparation of 3-benzyl-6-bromo-imidazo[1 ,2-a]pyridine

TFA (3.78 mL, 49.3 mmol) was cooled to 0°C and sodium borohydride (168 mg, 4.55 mmol) was added in portion. The mixture was stirred until no more solid was observed (about 20-30 min). The mixture was then added to a solution of (6-bromoimidazo[1 ,2-a]pyridin-3-yl)-phenyl- methanol (230 mg, 0.759 mmol) in DCM (16 mL) and the mixture was stirred for 5 hours. The reaction was quenched with saturated NaHC0 3 (added until the reaction became basic) and EtOAc (30 mL) was added. Phases were separated and aqueous phase was extracted with EtOAc (2 x 20 mL). The combined organic phases were washed with brine, dried and concentrated under reduced pressure. The material was purified by flash chromatography on silica gel using a mixture of EtOAc in hexane as eluent to provide the title compound (0.17 g), which was used in the next step without further purification. MS (ESI) [M+H] + 287.1/289.1.

Step 3: Preparation of Compound 1

3-benzyl-6-bromo-imidazo[1 ,2-a]pyridine (80 mg, 0.279 mmol), 3,5-dimethyl-4-(4,4,5,5- tetramethyl-1 ,3,2-dioxaborolan-2-yl)isoxazole (prepared using the procedure described in US20130053362, 83 mg, 0.334 mmol) in a solution of DME (2 mL) and water (0.1 mL) was degassed by bubbling N 2 for 10 min. Pd(PPh 3 ) 4 (32 mg, 0.028 mmol) and Cs 2 C0 3 (191 mg, 0.585 mmol) were added. The resulting mixture was heated to 85°C for 4 h and then cooled to rt. The mixture was concentrated under reduced pressure and then diluted with saturated NaHC0 3 (10 mL) and EtOAc (10 mL), and the aqueous phase was extracted with EtOAc (3 x 10 mL). The combined organic phases were dried over MgS0 4 , filtered, and concentrated under reduced pressure. The material was purified by flash chromatography on silica gel using a mixture of EtOAc on hexane followed by 10% MeOH in DCM and then by preparative HPLC to provide Compound 1 (15 mg, 16%). 1 H NMR (500 MHz, CDCI 3 ) δ 7.75 (s, 1 H), 7.68 (d, J = 9.4 Hz, 1 H), 7.52 (s, 1 H), 7.39 - 7.33 (m, 2H), 7.30 (dd, J = 6.7, 4.2 Hz, 2H), 7.27 - 7.17 (m, 4H), 4.32 (s, 2H), 3.62 (s, 3H), 2.21 (s, 3H). MS (ESI) [M+H] + 330.3. Example 2: 5-[3-[(4-fluorophenyl)methyl]imidazo[1 ,2-a]pyridin-6-yl]-1 ,3-dimethyl-pyridin-2-one (Compound 2)

Step 1: Preparation of (6-bromoimidazo[1,2-a]pyridin-3-yl)-(4-fluorophenyl) methanol

To a suspension of magnesium (148 mg, 6.18 mmol) in THF (1 mL) was added 1-bromo-4- fluorobenzene (0.25 mL, 2.27 mmol) and the mixture was heated to 50°C for a few seconds. The mixture was then stirred for 5 min and then the rest of 1-bromo-4-fluorobenzene (0.25 mL, 2.27 mmol) was added. The stirring was continued for 15 min and then the mixture was heated to 50°C for about 30 min until most of the solid magnesium disappeared. The mixture was then allowed to cool to rt and 1 mL of this mixture was then treated with a solution of 6- bromoimidazo[1 ,2-a]pyridine-3-carbaldehyde (160 mg, 0.71 1 mmol) in THF (5 mL) at -30°C and the mixture was stirred for 30 min. The reaction was then quenched by addition of a saturated solution of NH 4 CI (5 mL) and then diluted in EtOAc (30 mL). The layers were separated and the aqueous layer was extracted with EtOAc (2 x 20 mL). The combined organic layers were washed with brine, dried and concentrated under reduced pressure. The material was purified by flash chromatography on silica gel using a mixture of EtOAc in hexane as eluent to provide the title compound (61 %). MS (ESI) [M+H] + 321.1/323.1.

Step 2: Preparation of 6-bromo-3-[(4-fluorophenyl)methyl]imidazo[1 ,2-a]pyridine

TFA (2.17 mL, 28.34 mmol) was cooled to 0 °C and sodium borohydride (97 mg, 2.62 mmol) was added in one portion. The mixture was stirred until no more solid observed (about 30 min). Half of this mixture was then added to a solution of (6-bromoimidazo[1 ,2-a]pyridin-3-yl)-phenyl- methanol (140 mg, 0.436 mmol) in DCM (16 mL) and the mixture was stirred for 4 h. Half of the first mixture of TFA/NaBH 4 (kept at 0 °C) was then added to the reaction and the mixture stirred for 2 more hours. The reaction was quenched with saturated NaHC0 3 (5 mL) and EtOAc (30 mL) was added. Phases were separated and aqueous phase was extracted with EtOAc (2 x 20 mL). The combined organic phases were washed with brine, dried and concentrated under reduced pressure. The material was purified by flash chromatography on silica gel using a mixture of EtOAc in hexane to provide the title compound (37 mg, 28%). MS (ESI) [M+H] + 305.1/307.1.

Step 3: Preparation of Compound 2

A mixture of 1 ,3-dimethyl-5-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)pyridin-2-one (prepared using the procedure described in US20130053362, 36 mg, 0.146 mmol), 6-bromo-3-[(4- fluorophenyl)methyl]imidazo[1 ,2-a]pyridine (37 mg, 0.121 mmol) in DME (2 mL) and water (0.1 mL) was degassed by bubbling N 2 for 10 min. Cs 2 C0 3 (83 mg, 0.255 mmol) and Pd(PPh 3 ) 4 (14 mg, 0.012 mmol) were then added and the mixture was degassed for 10 more min. The resulting mixture was heated to 85 °C for 3h and then cooled to rt. The mixture was diluted with saturated NaHC0 3 (10 mL) and EtOAc (10 mL), and the aqueous phase was extracted with EtOAc (3 x 10 mL). The combined organic phases were dried over MgS0 4 , filtered, and concentrated under reduced pressure. The material product was purified by flash chromatography on silica gel using a mixture of EtOAc in hexane as eluent, and followed by preparative HPLC to provide Compound 2 (11 mg, 24%). 1 H NMR (500 MHz, MeOD) δ 8.32 (s, 1 H), 7.86 (d, J = 2.5 Hz, 1 H), 7.71 - 7.59 (m, 3H), 7.40 (s, 1 H), 7.36 - 7.28 (m, 2H), 7.12 - 7.02 (m, 2H), 4.37 (s, 2H), 3.65 (s, 3H), 2.18 (s, 3H). MS (ESI) [M+H] + 348.2. Example 3: 5-[3-[(4-chlorophenyl)methyl]imidazo[1 ,2-a]pyridin-6-yl]-1 ,3-dimethyl-pyridin-2-one (Compound 3)

Step 1: Preparation of (6-bromoimidazo[1,2-a]pyridin-3-yl)-(4-chlorophenyl) methanol

To a solution of 4-chlorophenylmagnesium bromide (1 M in THF, 1.2 mL, 1.2 mmol) in dry THF (1.5 mL) was added 6-bromoimidazo[1 ,2-a]pyridine-3-carbaldehyde (140 mg, 0.62 mmol) at -30 °C and the reaction mixture was stirred for 60 min at -30 °C. More 4-chlorophenylmagnesium bromide (1 M in THF, 1.2 mL, 1.2 mmol) was then added and the mixture was stirred for 2 h. To the mixture, was added saturated NH 4 CI and the solution was concentrated under reduced pressure. To the residue, EtOAc (50 mL) and saturated NaHC0 3 (10 mL) were added and phases were separated. The aqueous phase was then extracted with EtOAc (3 x 30 mL). The combined organic phases were washed with brine, dried over Na 2 S0 , filtered and then evaporated under reduced pressure. The material was purified over by flash chromatography on silica gel using a mixture of EtOAc in hexane as eluent to provide the title compound (130 mg, 62%). MS (ESI) [M+H] + 339.0.

Step 2: Preparation of 6-bromo-3-[(4-chlorophenyl)methyl]imidazo[ 1 ,2-a]pyridine

TFA (3.78 mL, 49.29 mmol) was cooled to 0 °C and sodium borohydride (171 mg, 4.62 mmol) was added in portion. The mixture was stirred until no more solid was observed (about 30 min). The mixture was then added to a solution of (6-bromoimidazo[1 ,2-a]pyridin-3-yl)-(4- chlorophenyl)methanol (130 mg, 0.385 mmol) in DCM (2 mL) and the reaction mixture was stirred for 4 h at rt. To the mixture, saturated NaHC0 3 (5 mL) and EtOAc (30 mL) were added. The phases were separated and the aqueous phase was extracted with EtOAc (2 x 20 mL). The combined organic phases were washed with brine, dried over Na 2 S0 4 , filtered and then concentrated under reduced pressure. The material was purified by flash chromatography on silica gel using a mixture of EtOAc in hexane as eluent to provide the title compound (50 mg, 40%). MS (ESI) [M+H] + 323.0.

Step 3: Preparation of Compound 3

A mixture of 1 ,3-dimethyl-5-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)pyridin-2-one (prepared using the procedure described in US20130053362, 50 mg, 0.202 mmol), 6-bromo-3-[(4- chlorophenyl)methyl]imidazo[1 ,2-a]pyridine (50 mg, 0.155 mmol) in DME (2 mL) and water (0.1 mL) was degassed by bubbling N 2 for 10 min. Cs 2 C0 3 (106 mg, 0.326 mmol) and Pd(PPh 3 ) 4 (18 mg, 0.016 mmol) were then added and the mixture was degassed for 10 more min and the resulting mixture was heated to 85 °C for 4 h. The mixture was cooled to rt and then concentrated under reduced pressure. The mixture was diluted with saturated NaHC0 3 (10 mL) and EtOAc (10 mL), and the aqueous layer was extracted with EtOAc (3 x 10 mL). The combined organic layers were dried over MgS0 4 , filtered, and concentrated under reduced pressure. The material was purified by flash chromatography using a mixture of EtOAc in hexane as eluent, followed by preparative HPLC to provide Compound 3 (11 mg, 17%). 1 H NMR (500 MHz, MeOD) δ 8.29 (s, 1 H), 7.85 (d, J = 2.5 Hz, 1 H), 7.71 - 7.64 (m, 2H), 7.61 (dd, J = 9.4, 1.7 Hz, 1 H), 7.41 (s, 1 H), 7.37 - 7.31 (m, 2H), 7.29 (d, J = 8.6 Hz, 2H), 4.38 (s, 2H), 3.65 (s, 3H), 2.19 (s, 3H). MS (ESI) [M+H] + 364.2.

Example 4: 5-[3-[(4-methoxyphenyl)methyl]imidazo[1 ,2-a]pyridin-6-yl]-1 ,3-dimethyl-pyridin-2- one (Compound 4)

Step 1: Preparation of (6-bromoimidazo[1,2-a]pyridin-3-yl)-(4-methoxyphenyl) methanol

To a solution of 4-methoxyphenylmagnesium bromide (1 M in THF, 2 mL, 1 mmol) in dry THF (1.5 mL) was added a solution of 6-bromoimidazo[1 ,2-a]pyridine-3-carbaldehyde (150 mg, 0.667 mmol) in THF(5 mL) at -30 °C and the reaction mixture was stirred 60 min at -30 °C. Saturated NH 4 CI was added to the mixture and the solution was concentrated under reduced pressure. To the residue, EtOAc (50 mL) and saturated NaHC0 3 (10 mL) were added. The layers were separated and the aqueous layer was extracted with EtOAc (3 x 30 mL). The combined organic layers were washed with brine, dried over Na 2 S0 4 , filtered and evaporated under reduced pressure. The material was purified by flash chromatography on silica gel using a mixture of EtOAc in hexane to provide the title compound (120 mg, 54 %). MS (ESI) [M+H] + 333.0/335.0.

Step 2: Preparation of 6-bromo-3-[(4-methoxyphenyl)methyl]imidazo[1 ,2-a]pyridine

(6-Bromoimidazo[1 ,2-a]pyridin-3-yl)-(4-methoxyphenyl)methanol (40 mg, 0.12 mmol) was suspended in DCM (1.5 mL), BF 3 « Et 2 0 (52 mL, 0.42 mmol) and Et 3 SiH (0.06 mL, 0.3 mmol) were added dropwise, and the reaction mixture was stirred for 10 min. To the mixture, saturated NaHC0 3 (5 mL) was added, followed by DCM (30 mL). The aqueous layer was extracted with DCM (2 x 20 mL). The combined organic layers were dried over Na 2 S0 4 , filtered and concentrated under reduced pressure to afford the title compound (40 mg), which was used in the next step without any further purification. MS (ESI) [M+H] + 317.1/319.1.

Step 3: Preparation of Compound 4

A solution of 6-bromo-3-[(4-methoxyphenyl)methyl]imidazo[1 ,2-a]pyridine (40 mg, 0.126 mmol), 1 ,3-dimethyl-5-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)pyridin-2-one (prepared using the procedure described in US20130053362, 40 mg, 0.164 mmol) in DME (2 mL) and water (0.1 mL) was degassed by bubbling N 2 for 10 min. Cs 2 C0 3 (86 mg, 0.265 mmol) and Pd(PPh 3 ) 4 (15 mg, 0.013 mmol) were then added and the mixture was degassed for another 10 min. The resulting mixture was heated to 85°C overnight and then allowed to cool to rt. To the mixture, saturated NaHC0 3 (10 mL) and EtOAc (10 mL) were added, and the aqueous layer was extracted with EtOAc (3 x 10 mL). The combined organic layers were dried over MgS0 4 , filtered, and evaporated under reduced pressure. The product was purified by flash chromatography on silica gel using a mixture of EtOAc in hexane as eluent, followed by preparative HPLC to provide Compound 4 (11 mg, 24%). 1 H NMR (500 MHz, MeOD) δ 8.26 (d, J = 32.2 Hz, 1 H), 7.83 (d, J = 1.6 Hz, 1 H), 7.74 - 7.58 (m, 3H), 7.41 (s, 1 H), 7.21 (d, J = 8.4 Hz, 2H), 6.89 (d, J = 8.5 Hz, 2H), 4.30 (s, 2H), 3.74 (s, 3H), 3.63 (s, 3H), 2.18 (s, 3H). MS (ESI) [M+H] + 360.2.

Example 5: 5-[3-[(3-fluorophenyl)methyl]imidazo[1 ,2-a]pyridin-6-yl]-1 ,3-dimethyl-pyridin-2-one (Compound 5)

Step 1: Preparation of (6-bromoimidazo[1,2-a]pyridin-3-yl)-(3-fluorophenyl) methanol

1-Fluoro-3-iodo-benzene (554 mg, 2.49 mmol) was added dropwise to isopropylmagnesium chloride (1 M solution, 0.995 mL, 0.995 mmol) at rt. After 15 min, the Grignard was ready to use. LaCI 3 « 2LiCI (0.6 M in THF, 1.66 mL, 0.995 mmol) was added to a solution of 6- bromoimidazo[1 ,2-a]pyridine-3-carbaldehyde (224 mg, 0.995 mmol) in THF (4 mL) at rt. The mixture was stirred for 30 min at rt, then the above Grignard solution was added dropwise at rt and the reaction mixture was stirred for 1 h. To the mixture, saturated NH 4 CI (5 mL) was added and the solution was concentrated under reduced pressure. To the residue, EtOAc (30 mL) and saturated NaHC0 3 (10 mL) were added. The phases were separated and the aqueous layer was extracted with EtOAc (2 x 50 mL). The combined organic layers were washed with brine, dried over Na 2 S0 4 , filtered and concentrated under reduced pressure. The material was purified by flash chromatography on silica gel to provide the title compound (0.32 g, 31 %). 1 H NMR (500 MHz, CDCI 3 ) δ 8.38 (d, J = 1.1 Hz, 1 H), 7.47 (d, J = 9.5 Hz, 1 H), 7.42 - 7.33 (m, 1 H), 7.28 - 7.25 (m, 1 H), 7.24 - 7.19 (m, 2H), 7.18 - 7.15 (m, 1 H), 7.12 - 7.00 (m, 1 H), 6.17 (s, 1 H), 3.12 - 2.80 (m, 1 H). MS (ESI) [M+H] + 321.0, 323.0.

Step 2: Preparation of 6-bromo-3-[(3-fluorophenyl)methyl]imidazo[1 ,2-a]pyridine

(6-bromoimidazo[1 ,2-a]pyridin-3-yl)-(3-fluorophenyl)methanol (40 mg, 0.125 mmol) was suspended in DCM (1.5 mL) and BF 3 « Et 2 0 (0.054 mL, 0.436 mmol) and Et 3 SiH (0.058 mL, 0.311 mmol) were added dropwise. The reaction mixture was stirred for 1 h and then saturated NaHC0 3 (5 mL) was added, followed by DCM (30 mL). The aqueous phase was then extracted with DCM (2 x 20 mL). The combined organic layers were dried over Na 2 S0 4 , filtered and evaporated under reduced pressure to afford the title compound (39 mg), which was used in the next step without further purification. MS (ESI) [M+H] + 305.1 , 307.1.

Step 3: Preparation of Compound 5

A solution of 6-bromo-3-[(3-fluorophenyl)methyl]imidazo[1 ,2-a]pyridine (39 mg, 0.128 mmol), 1 ,3-dimethyl-5-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)pyridin-2-one (prepared using the procedure described in US20130053362, 41 mg, 0.166 mmol) in DME (2 mL) and water (0.1 mL) was degassed by bubbling N 2 for 10 min. Cs 2 C0 3 (87 mg, 0.268 mmol) and Pd(PPh 3 ) 4 (14 mg, 0.013 mmol) were then added and the mixture was degassed for another 10 min. The resulting mixture was heated to 85 °C for 3 h and then cooled to rt. To the mixture, saturated NaHC0 3 (10 mL) and EtOAc (10 mL) were added, and the aqueous layer was extracted with EtOAc (3 x 10 mL). The combined organic layers were dried over MgS0 4 , filtered, and concentrated under reduced pressure. The material was purified by flash chromatography on silica gel using a mixture of EtOAc in hexane as eluent, followed by preparative HPLC to provide Compound 5 (13 mg, 29%). 1 H NMR (500 MHz, CDCI 3 ) δ 7.75 - 7.69 (m, 1 H), 7.66 (dd, J = 9.3, 0.7 Hz, 1 H), 7.50 (s, 1 H), 7.34 - 7.27 (m, 2H), 7.25 - 7.17 (m, 2H), 7.02 - 6.99 (m, 1 H), 6.97 (td, J = 8.4, 2.5 Hz, 1 H), 6.90 (dd, J = 9.6, 1.7 Hz, 1 H), 4.28 (s, 2H), 3.60 (s, 3H), 2.20 (s, 3H). MS (ESI) [M+H] + 348.1.

Examples 6-7: 5-[3-[methoxy(phenyl)methyl]imidazo[1 ,2-a]pyridin-6-yl]-1 ,3-dimethyl-pyridin-2- one isomers (Compounds 6 and 7)

Compound 6 (RT: 7.07) - Compound 7 (RT: 9.61)

Step 1: Preparation of 6-bromo-3-[methoxy(phenyl)methyl]imidazo[1 ,2-a]pyridine

To a solution of 3-phenylprop-2-ynal (11 1 mg, 0.85 mmol), 5-bromopyridin-2-amine (147.6 mg, 0.853 mmol) in MeOH (3 mL) was added acetic acid (1 drop) and the reaction mixture was heated to 80 °C for 8 h. The mixture was concentrated under reduced pressure and the material was purified by flash chromatography on silica using a mixture of EtOAc and hexane as eluent to afford the title compound as a mixture of enantiomers (191 mg, 71 %). 1 H NMR (500 MHz, CDCI 3 ) δ 8.25 (dd, J = 1.9, 0.8 Hz, 1 H), 7.52 (dd, J = 9.5, 0.8 Hz, 1 H), 7.40 (d, J = 4.4 Hz, 4H), 7.37 (dd, J = 8.0, 4.1 Hz, 1 H), 7.24 (d, J = 1.9 Hz, 2H), 5.60 (s, 1 H), 3.40 (s, 3H).

Step 2: Preparation of Compounds 6 and 7

To a solution of 6-bromo-3-[methoxy(phenyl)methyl]imidazo[1 ,2-a]pyridine (50 mg, 0.158 mmol) in dioxane (1 ml_) was added Cs 2 C0 3 (77 mg, 0.236 mmol), 1 ,3-dimethyl-5-(4,4,5,5-tetramethyl- 1 ,3,2-dioxaborolan-2-yl)pyridin-2-one (prepared using the procedure described in US20130053362, 59 mg, 0.24 mmol), Pd(PPh 3 ) 4 (18 mg, 0.016 mmol) and water (0.1 ml_). The reaction mixture was degassed by bubbling N 2 for 10 min and the resulting mixture was then heated at 80 °C for 18 h. The mixture was cooled to rt and EtOAc (5 ml_) and water (5 ml_) were added. The organic phase was dried over Na 2 S0 4 , filtered and evaporated under reduced pressure. The material was purified by flash chromatography on silica gel using a mixture of EtOAc and hexane as eluent, followed by semi-preparative SFC (AS 10 x 250 mm, 5um Isocratic 25% MeOH + 0.1 % NH OH, 10 mL/min 100 Bar) to afford Compound 6 (18.6 mg, 35 %) and Compound 7 (23.4 mg, 43 %).

Compound 6: Retention Time = 7.07; 1 H NMR (500 MHz, CDCI 3 ) δ 8.04 (s, 1 H), 7.65 (d, J = 9.3 Hz, 1 H), 7.49 - 7.32 (m, 6H), 7.29 (dd, J = 2.6, 1.2 Hz, 1 H), 7.24 (dd, J = 9.3, 1.8 Hz, 1 H), 7.20 (d, J = 2.4 Hz, 1 H), 5.70 (s, 1 H), 3.61 (s, 3H), 3.44 (s, 3H), 2.22 (s, 3H). MS (ESI) [M+H] + 360.2.

Compound 7: Retention Time = 9.61 ; 1 H NMR (500 MHz, CDCI 3 ) δ 8.04 (s, 1 H), 7.65 (d, J = 9.3 Hz, 1 H), 7.49 - 7.34 (m, 6H), 7.29 (dd, J = 2.5, 1.1 Hz, 1 H), 7.26 - 7.23 (m, 1 H), 7.21 (d, J = 2.4 Hz, 1 H), 5.70 (s, 1 H), 3.61 (s, 3H), 3.44 (s, 3H), 2.22 (s, 3H). MS (ESI) [M+H] + 360.1.

Example 8: 1 ,3-dimethyl-5-[3-(1-piperidylmethyl)imidazo[1 ,2-a]pyridin-6-yl]pyridin-2-one (Compound 8)

Step 1: Preparation of 6-bromo-3-(1-piperidylmethyl)imidazo[1 ,2-a]pyridine

Piperidine (0.053 ml_, 0.533 mmol) was added to a stirred solution of 6-bromoimidazo[1 ,2- a]pyridine-3-carbaldehyde (100 mg, 0.444 mmol) in DCM (4 ml_). To this mixture was then added acetic acid (0.076 mL, 1.33 mmol) and the mixture was stirred for 45 min. Na(OAc) 3 BH (94 mg, 0.444 mmol) was added and the reaction was stirred for 18 h. Another portion of Na(OAc) 3 BH (188 mg, 0.888 mmol) was added and the reaction mixture was stirred for 3 h. To the mixture, water and saturated NaHC0 3 (10 mL) were added followed by DCM (20 mL). The organic layer was separated and the aqueous layer was extracted with DCM (2 x 20 mL). The combined organic layers were washed with brine, dried over Na 2 S0 4 , filtered and concentrated under reduced pressure. The material was purified by flash chromatography on silica gel using a mixture of EtOAc in hexane as eluent to provide the title compound (45 mg, 34%). MS (ESI) [M+H] + : 294.1 , 296.1.

Step 2: Preparation of Compound 8

A solution of 6-bromo-3-(1-piperidylmethyl)imidazo[1 ,2-a]pyridine (45 mg, 0.153 mmol) and 1 ,3- dimethyl-5-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)pyridin-2-one (prepared using the procedure described in US20130053362, 46 mg, 0.184 mmol) in DME (2 mL) and water (0.1 mL) was degassed by bubbling N 2 for 10 min. Cs 2 C0 3 (105 mg, 0.321 mmol) and Pd(PPh 3 ) 4 (18 mg, 0.015 mmol) were then added and the mixture was degassed for another 10 min. The resulting mixture was heated to 85°C for 3 h and then cooled to rt. To the mixture, saturated NaHC0 3 (10 mL) and EtOAc (10 mL) were added, and the aqueous layer was extracted with EtOAc (3 x 10 mL). The combined organic layers were dried over MgS0 4 , filtered, and concentrated under reduced pressure. The material was purified by flash chromatography on silica gel using a mixture of EtOAc in hexane as eluent, followed by preparative HPLC to provide Compound 8 (1 1 mg, 21 %). 1 H NMR (500 MHz, CDCI 3 ) δ 8.44 (d, J = 0.9 Hz, 1 H), 7.62 (dd, J = 9.3, 0.8 Hz, 1 H), 7.49 (s, 1 H), 7.45 (dd, J = 2.6, 1.2 Hz, 1 H), 7.38 (d, J = 2.4 Hz, 1 H), 7.23 (dd, J = 9.3, 1.9 Hz, 1 H), 3.78 (s, 2H), 3.65 (s, 3H), 2.50 - 2.30 (bs, 3H), 2.25 (s, 3H), 1.86 (s, 1 H), 1.62 - 1.48 (m, 4H), 1.50 - 1.36 (m, 2H). MS (ESI) [M+H] + 337.3.

Example 9: 5-[3-(cyclopentoxymethyl)imidazo[1 ,2-a]pyridin-6-yl]-1 ,3-dimethyl-pyridin-2-one (Compound 9)

Step 1: Preparation of 6-bromo-3-(cyclopentoxymethyl)imidazo[1 ,2-a]pyridine

To a solution of cyclopentanol (0.074 mL, 0.815 mmol) and DIPEA (0.212 mL, 1.22 mmol) in THF (1 mL), Intermediate 1 (100 mg, 0.407 mmol) was added and the reaction mixture was heated to 55 °C for 18 h. The mixture was then cooled and concentrated under reduced pressure. To the residue, EtOAc (30 mL) was added and the organic layer was washed with saturated NaHC0 3 (7 mL). The aqueous layer was then extracted with EtOAc (2 x 20 mL). The combined organic layers were washed with brine, dried over MgS0 4 , filtered and concentrated under reduced pressure. The material was purified by flash chromatography on silica gel using a mixture of EtOAc in hexane as eluent to provide the title compound (40 mg, 33%). MS (ESI) [M+H] + 295.0, 297.0.

Step 2: Preparation of Compound 9

A solution of 6-bromo-3-(cyclopentoxymethyl)imidazo[1 ,2-a]pyridine (40 mg, 0.136 mmol) and 1 ,3-dimethyl-5-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)pyridin-2-one (prepared using the procedure described in US20130053362, 40mg, 0.163 mmol) in DME (2 mL) and water (0.1 mL) was degassed by bubbling N 2 for 10 min. Cs 2 C0 3 (93 mg, 0.285 mmol) and Pd(PPh 3 ) (16 mg, 0.014 mmol) were then added and the mixture was degassed for another 10 min. The resulting mixture was heated to 85°C for 3h and then cooled to rt. To the mixture, saturated NaHC0 3 (10 mL) and EtOAc (20 mL) were added. The layers were separated and the aqueous layer was extracted with EtOAc (3 x 10 mL). The combined organic layers were dried over MgS0 4 , filtered, and concentrated under reduced pressure. The product was purified by flash chromatography on silica gel using a mixture of EtOAc in hexane as eluent, followed by preparative HPLC to provide Compound 9 (7 mg, 13%). 1 H NMR (500 MHz, CDCI 3 ) δ 8.22 (s, 1 H), 7.66 (d, J = 9.0 Hz, 1 H), 7.61 (s, 1 H), 7.47 - 7.42 (m, 1 H), 7.38 (d, J = 2.5 Hz, 1 H), 7.28 (dd, J = 9.2, 1.7 Hz, 1 H), 4.82 (s, 2H), 4.09 - 3.89 (m, 1 H), 3.65 (s, 3H), 2.25 (s, 3H), 1.86 - 1.62 (m, 6H), 1.61 - 1.52 (m, 2H). MS (ESI) [M+H] + 338.3. Example 10: 5-[3-(anilinomethyl)imidazo[1 ,2-a]pyridin-6-yl]-1 ,3-dimethyl-pyridin-2-one (Compound 10)

Step 1: Preparation of N-[(6-bromoimidazo[1 ,2-a]pyridin-3-yl)methyl]aniline

To a solution of Intermediate 1 (54 mg, 0.22 mmol) in MeCN (2 mL) was added aniline (0.024 mL, 0.264 mmol) and K 2 C0 3 (85 mg, 0.66 mmol) and the reaction mixture was heated at 60°C for 1 h. The mixture was cooled and then concentrated under reduced pressure. To the residue, EtOAc (30 mL) and saturated NaHC0 3 (10 mL) were added. The layers were separated and the aqueous layer was extracted with EtOAc (2 x 20 mL). The combined organic layers were washed with brine, dried over Na 2 S0 4 , filtered and then concentrated under reduced pressure. The material was purified by flash chromatography on silica gel using a mixture of EtOAc in hexane and then MeOH in EtOAc as eluent to provide the title compound (10 mg, 15%). MS (ESI) [M+H] + 302.1 , 304.1.

Step 2: Preparation of Compound 10

A solution of N-[(6-bromoimidazo[1 ,2-a]pyridin-3-yl)methyl]aniline (10 mg, 0.033 mmol) and 1 ,3- dimethyl-5-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)pyridin-2-one (prepared using the procedure described in US20130053362, 10.7 mg, 0.043 mmol) in DME (2 mL) and water (0.1 mL) was degassed by bubbling N 2 for 10 min. Cs 2 C0 3 (23 mg, 0.069 mmol) and Pd(PPh 3 ) 4 (4 mg, 0.003 mmol) were then added and the mixture was degassed for another 10 min. The resulting mixture was heated to 85°C for 2 h and then cooled to rt. To the mixture, saturated NaHC0 3 (10 mL) and EtOAc (30 mL) were added. The layers were separated and the aqueous layer was extracted with EtOAc (3 x 10 mL). The combined organic layers were dried over MgS0 4 , filtered, and concentrated under reduced pressure. The material was purified by flash chromatography on silica gel using a mixture of EtOAc in hexane as eluent, followed by preparative HPLC to provide Compound 10 (5 mg, 44%). 1 H NMR (500 MHz, CDCI 3 ) δ 8.10 (s, 1 H), 7.68 (d, J = 9.4 Hz, 1 H), 7.65 (s, 1 H), 7.36 (d, J = 1.3 Hz, 1 H), 7.32 - 7.25 (m, 4H), 6.84 (t, J = 7.4 Hz, 1 H), 6.78 (d, J = 7.7 Hz, 2H), 4.65 (s, 2H), 3.75 (s, 1 H), 3.61 (s, 3H), 2.21 (s, 3H). MS (ESI) [M+H] + 345.2.

Example 11 : 1 ,3-dimethyl-5-(3-(morpholine-4-carbonyl)imidazo[1 ,2-a]pyridin-6-yl) pyridin-2(1 H)- one (Compound 11)

To a stirred solution of Intermediate 2 (0.1 g, 0.35 mmol) in DMF (2 mL) was added HATU (0.2 g, 0.53 mmol) at rt. The mixture was stirred at this temperature for 30 min, and a solution of morpholine (34 mg, 0.39 mmol) and DIPEA (0.2 mL, 1.05 mmol) in DMF (1 mL) was then added dropwise and the reaction mixture was stirred at rt for 16 h. The mixture was poured into water (30 ml) and the aqueous layer was extracted with EtOAc (3 x 20 mL). The combined organic layers were washed with brine (30 mL), dried over Na 2 S0 4 , filtered and concentrated under reduced pressure. The material was purified by flash chromatography on silica gel using a mixture of 3% MeOH in DCM as eluent to afford Compound 11 (15 mg, 12%) as a solid. 1 H NMR (400 MHz, DMSO) δ ppm 9.00 (s, 1 H), 8.06 (d, J = 2.4 Hz, 1 H), 8.04 (s, 1 H), 7.79 (d, J = 9.6 Hz, 1 H), 7.70 (s, 1 H), 7.68 (d, J = 2 Hz, 1 H), 3.76-3.75 (m, 4H), 3.69-3.68 (m, 4H), 3.53 (s, 3H), 2.10 (s, 3H). MS (ESI) [M+H] + 353.6.

Example 12: N-cyclohexyl-6-(1 ,5-dimethyl-6-oxo-1 ,6-dihydropyridin-3-yl)imidazo[1 ,2-a] pyridine-3-carboxamide (Compound 12)

To a stirred solution of Intermediate 2 (0.1 g, 0.35 mmol) in DMF (2 ml_) was added HATU (0.2 g, 0.53 mmol) at rt. The mixture was stirred at this temperature for 30 min, a solution of cyclohexanamine (39 mg, 0.39 mmol) and DIPEA (0.2 ml_, 1.05 mmol) in DMF (1 ml_) was then added dropwise and the reaction mixture was stirred at rt for 16 h. The reaction mixture was poured into water (30 ml_) and the resulting solid was filtered and dried under reduced pressure. The material was purified by flash chromatography on silica gel using a mixture of 3% MeOH in DCM as eluent to afford Compound 12 (70 mg, 54%) as a solid. 1 H NMR (400 MHz, DMSO) δ ppm 9.62 (s, 1 H), 8.37 (s, 1 H), 8.28 (d, J = 8 Hz, 1 H), 8.05 (d, J = 2.4 Hz, 1 H), 7.76 (dd, J = 0.8 and 9.2 Hz, 1 H), 7.69 (d, J = 2 Hz, 1 H), 7.68-7.66 (m, 1 H), 3.82 (bs, 1 H), 3.54 (s, 3H), 2.1 1 (s, 3H), 1.88-1.86 (m, 2H), 1.77-1.76 (m, 2H), 1.64 (d, J = 11.6 Hz, 1 H), 1.38-1.29 (m, 4H), 1.17- 1.16 (m, 1 H). MS (ESI) [M+H] + 365.6. Example 13: 1 ,3-dimethyl-5-(3-(piperidine-1-carbonyl)imidazo[1 ,2-a]pyridin-6-yl)pyridin-2(1 H)- one (Compound 13)

To a stirred solution of Intermediate 2 (0.1 g, 0.35 mmol) in DMF (2 ml_) was added HATU (0.2 g, 0.53 mmol) at rt. The mixture was stirred at that temperature for 30 min, a solution of piperidine (33 mg, 0.39 mmol) and DIPEA (0.2 ml_, 1.05 mmol) in DMF (1 ml_) was then added dropwise and the reaction mixture was stirred at rt for 16 h. The mixture was poured into water (30 mL) and the aqueous layer was extracted with EtOAc (3 x 20 mL). The combined organic layers were washed with brine (30 mL), dried over Na 2 S0 4 , filtered and evaporated under reduced pressure. The material was purified by flash chromatography on silica gel using a mixture of 3% MeOH in DCM as eluent to afford Compound 13 (70 mg, 57%) as a solid. 1 H NMR (400 MHz, DMSO) δ ppm 8.95 (s, 1 H), 8.05 (s, 1 H), 7.96 (s, 1 H), 7.76 (d, J = 9.6 Hz, 1 H), 7.70 (s, 1 H), 7.66 (d, J = 9.2 Hz, 1 H), 3.70 (s, 4H), 3.53 (s, 3H), 2.10 (s, 3H), 1.67-1.62 (m, 6H). MS (ESI) [M+H] + 351.6.

Example 14: 1 ,3-dimethyl-5-(3-(piperidin-1-ylsulfonyl)imidazo[1 ,2-a]pyridin-6-yl)pyridin-2(1 H)- one (Compound 14)

Step 1: Preparation of 6-bromo-3-(piperidin-1-ylsulfonyl)imidazo[1 ,2-a]pyridine

To a solution of piperidine (0.086 g, 1.02 mmol) in DCM (2 mL) was added TEA (0.28 mL, 2.03 mmol) at rt. The mixture was stirred for 5 min, a solution of Intermediate 3 (0.2 g, 0.68 mmol) in DCM (2 mL) was added and the reaction mixture stirred at rt for 2 h. The mixture was diluted with DCM (30 mL) and then washed with water (30 mL) and brine (30 mL). The organic layer was dried over Na 2 S0 , filtered and evaporated under reduced pressure to afford the title compound (0.15 g, 64%). 1 H NMR (400 MHz, DMSO) δ ppm 8.80 (q, J = 0.8 Hz, 1 H), 8.19 (s, 1 H), 7.83 (dd, J = 0.8 and 9.6 Hz, 1 H), 7.72 (dd, J = 2 and 9.6 Hz, 1 H), 3.08 (t, J = 5.2 Hz, 4H), 1.56-1.50 (m, 4H), 1.42-1.39 (m, 2H). MS (ESI) [M+H] + 346.2.

Step 2: Preparation of Compound 14

A stirred solution of 6-bromo-3-(piperidin-1-ylsulfonyl)imidazo[1 ,2-a]pyridine (0.15 g, 0.44 mmol) and 1 ,3-dimethyl-5-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)pyridin-2(1 H)-one (prepared using the procedure described in US20130053362, 0.13 g, 0.52 mmol) in 1 ,4-dioxane (3 ml_) was purged with N 2 for 10 min followed by the addition of a solution of Na 2 C0 3 (0.14 g, 1.31 mmol) in water (1 ml_), which was again purged with N 2 for another 10 min. After 10 min, Pd(PPh 3 ) 4 (25 mg, 0.022 mmol) was added and the resulting mixture was heated at 100°C for 3h. The mixture was concentrated under reduced pressure, water was added and the aqueous layer was extracted with EtOAc (3 x 20 ml_). The combined organic layers were washed with water (20 ml_) and brine (20 ml_), dried over Na 2 S0 4 , filtered and evaporated under reduced pressure. The material was purified by flash chromatography on silica gel using a mixture of 2% methanol in DCM as eluent to afford Compound 14 (0.11 g, 65%) as a solid. 1 H NMR (400 MHz, DMSO) δ ppm 8.74 (s, 1 H), 8.17 (s, 1 H), 8.04 (d, J = 2.4 Hz, 1 H), 7.91 (d, J = 9.6 Hz, 1 H), 7.80 (dd, J = 1.6 and 1.2 Hz, 1 H), 7.67 (s, 1 H), 3.54 (s, 3H), 3.09 (t, J = 5 Hz, 4H), 2.11 (s, 3H), 1.53 (s, 4H), 1.38 (d, J = 4.4 Hz, 2H). MS (ESI) [M+H] + 387.5.

Example 15: 6-(1 ,5-dimethyl-6-oxo-1 ,6-dihydropyridin-3-yl)-N-ethylimidazo[1 ,2-a] pyridine-3- sulfonamide (Compound 15)

Step 1: Preparation of 6-bromo-N-ethylimidazo[1 ,2-a]pyridine-3-sulfonamide

To a solution of ethyl amine (34 mg, 0.76 mmol) in DCM (2 mL) was added TEA (0.2 mL, 1.52 mmol). The mixture was stirred for 5 min, a solution of Intermediate 3 (0.15 g, 0.51 mmol) in DCM (1 mL) was added and the reaction mixture was stirred at rt for 2 h. The mixture was diluted with DCM (25 mL), washed with water (25 mL) and brine (25 mL). The organic layer was dried over Na 2 S0 4 , filtered and evaporated under vacuum to afford the title compound (0.1 g, 65%), which was used in the next step without purification. MS (ESI) [M+H] + 306.5.

Step 2: Preparation of Compound 15

A stirred solution of 6-bromo-N-ethylimidazo[1 ,2-a]pyridine-3-sulfonamide (0.1 g, 0.33 mmol) and 1 ,3-dimethyl-5-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)pyridin-2(1 H)-one (prepared using the procedure described in US20130053362, 0.122 g, 0.49 mmol) in 1 ,4-dioxane (2 mL) was purged with N 2 for 10 min followed by the addition of a solution of Na 2 C0 3 (0.104 g, 0.99 mmol) in water (0.5 mL), which was again purged with N 2 for 10 min. After 10 min, Pd(PPh 3 ) (19 mg, 0.016 mmol) was added and the reaction mixture was heated at 100°C for 3 h. The mixture was diluted with water and the aqueous layer was extracted using EtOAc (3 x 20 mL). The combined organic layers were washed with water (20 mL) and brine (20 mL), dried over Na 2 S0 , filtered and evaporated under reduced pressure. The material was purified by flash chromatography on silica gel using a mixture of 2-3% methanol in DCM as eluent to afford Compound 15 (17 mg, 15%) as a solid. 1 H NMR (400 MHz, DMSO) δ ppm 8.66 (s, 1 H), 8.33 (bs, 1 H), 8.10 (d, J = 2.4 Hz, 1 H), 8.09 (s, 1 H), 7.90 (d, J = 9.2 Hz, 1 H), 7.79 (dd, J = 2.0 and 1.6 Hz, 1 H), 7.77 (d, J = 2.0 Hz, 1 H), 3.55 (s, 3H), 2.83 (q, J = 8.8 Hz, 2H), 2.13 (s, 3H), 0.89 (t, J = 7.2 Hz, 3H). MS (ESI) [M+H] + 347.5.

Example 16: N-cyclopentyl-6-(1 ,5-dimethyl-6-oxo-1 ,6-dihydropyridin-3-yl)imidazo[1 ,2- a]pyridine-3-sulfonamide (Compound 16)

Step 1: Preparation of 6-bromo-N-cyclopentylimidazo[1,2-a]pyridine-3-sulfonamide

TEA (0.28 mL, 2.03 mmol) was added to a rt stirred solution of cyclopentylamine (86 mg, 1.02 mmol) in DCM (2 mL). The mixture was stirred for 5 min, a solution of Intermediate 3 (0.2 g, 0.68 mmol) in DCM (2 mL) was added and the reaction mixture was stirred at rt for another 2 h. The mixture was diluted with DCM (30 mL) and washed with water (30 mL) and brine (30 mL). The organic layer was dried over Na 2 S0 4 , filtered and evaporated under reduced pressure to afford the title compound (0.15 g, 64%) as a solid. 1 H NMR (400 MHz, DMSO) δ ppm 8.88 (q, J = 0.8 Hz, 1 H), 8.1 1 (s, 1 H), 7.83 (dd, J = 0.8 Hz, 1 H), 7.72 (dd, J = 1.6 Hz, 1 H), 3.46-3.39 (m, 1 H), 1.54-1.49 (m, 4H), 1.40-1.34 (m, 2H), 1.30-1.21 (m, 2H), -NH was not visible MS. (ESI) [M+H] + 346.4.

Step 2: Preparation of Compound 16

A stirred solution of 6-bromo-N-cyclopentylimidazo[1 ,2-a]pyridine-3-sulfonamide (0.15 g, 0.44 mmol) and 1 ,3-dimethyl-5-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)pyridin-2(1 H)-one (prepared using the procedure described in US20130053362, 0.13 g, 0.52 mmol) in 1 ,4-dioxane (3 mL) was purged with N 2 for 10 min followed by the addition of a solution of Na 2 C0 3 (0.14 g, 1.31 mmol) in water (1 mL), which was again purged with nitrogen for another 10 min. After 10 min, Pd(PPh 3 ) (25 mg, 0.022 mmol) was added and the resulting mixture was heated at 100 °C for 3h. The mixture was diluted with water and the aqueous layer was extracted using EtOAc (3 x 20 mL). The combined organic layers were washed with water (20 mL) and brine (20 mL), dried over Na 2 S0 4 , filtered and then evaporated under reduced pressure. The material was purified by flash chromatography on silica gel using a mixture of 2% MeOH in DCM as eluent to afford Compound 16 (0.1 1 g, 65%) as a solid. 1 H NMR (400 MHz, DMSO) δ ppm 8.67 (s, 1 H), 8.44 (d, J = 7.2 Hz, 1 H), 8.10 (d, J = 2.8 Hz, 1 H), 8.09 (s, 1 H), 7.90 (d, J = 9.6 Hz, 1 H), 7.82 (dd, J = 1.6 and 9.6 Hz 1 H), 7.77 (s, 1 H), 3.55 (s, 3H), 3.44-3.39 (m, 1 H), 2.13 (s, 3H), 1.49 (s, 4H), 1.39-1.33 (m, 2H), 1.19-1.16 (m, 2H). MS (ESI) [M+H] + 387.5.

Example 17: 6-(1 ,5-dimethyl-6-oxo-1 ,6-dihydropyridin-3-yl)-N-isopropylimidazo [1 ,2-a] pyridines-sulfonamide (Compound 17)

Step 1: Preparation of 6-bromo-N-isopropylimidazo[1 ,2-a]pyridine-3-sulfonamide

To a rt solution of isopropyl amine (0.045 g, 0.76 mmol) in DCM (2 mL) was added TEA (0.2 mL, 1.52 mmol). The mixture was stirred for 5 min and then a solution of Intermediate 3 (0.15 g, 0.51 mmol) in DCM (1 mL) was added. The reaction mixture was stirred at rt for another 2 h. The mixture was then diluted with DCM (25 mL) and washed with water (25 mL) and brine (25 mL). The organic layer was dried over Na 2 S0 4 , filtered and evaporated under reduced pressure to afford the title compound (0.1 g, 62%), which was used in next step without purification. MS (ESI) [M+H] + 320.5.

Step 2: Preparation of Compound 17

A stirred solution of 6-bromo-N-isopropylimidazo[1 ,2-a]pyridine-3-sulfonamide (0.1 g, 0.31 mmol) and 1 ,3-dimethyl-5-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)pyridin-2(1 H)-one (prepared using the procedure described in US20130053362, 0.117 g, 0.47 mmol) in 1 ,4- dioxane (2 mL) was purged with N 2 for 10 min, followed by the addition of a solution of Na 2 C0 3 (0.1 g, 0.93 mmol) in water (0.5 mL). The mixture was then purged again with N 2 for another 10 min. After 10 min, Pd(PPh 3 ) 4 (18 mg, 0.02 mmol) was added and the resulting mixture was heated at 100 °C for 3 h. The mixture was diluted with water and the aqueous layer was extracted using ethyl acetate (3 x 20 mL). The combined organic layers were washed with water (20 mL) and brine (20 mL), dried over Na 2 S0 and then evaporated under reduced pressure. The material was purified by flash chromatography on silica gel using a mixture of 2-3% MeOH in DCM as eluent to afford Compound 17 (16 mg, 14%) as a solid. 1 H NMR (400 MHz, DMSO) δ ppm 8.66 (s, 1 H), 8.41 (d, J = 7.6 Hz, 1 H), 8.10 (d, J = 2.8 Hz, 1 H), 8.09 (s, 1 H), 7.90 (d, J = 9.6 Hz, 1 H), 7.82 (dd, J = 2.0 and 9.6 Hz, 1 H), 7.78 (d, J = 1.2 Hz, 1 H), 3.55 (s, 3H), 3.27-3.22 (m, 1 H), 2.13 (s, 3H), 0.88 (d, J = 6.8 Hz, 6H). MS (ESI) [M+H] + 361.5.

Example 18: N-cyclohexyl-6-(1 ,5-dimethyl-6-oxo-1 ,6-dihydropyridin-3-yl)imidazo[1 ,2-a]pyridine- 3-sulfonamide (Compound 18)

Step 1: Preparation of 6-bromo-N-cyclohexylimidazo[1 ,2-a]pyridine-3-sulfonamide

To a solution of cyclohexylamine (38 mg, 0.37 mmol) in DCM (1 mL) was added TEA (0.1 mL, 0.75 mmol) at rt. The mixture was stirred for 5 min and then a solution of Intermediate 3 (0.075 g, 0.25 mmol) in DCM (1 mL) was added. The reaction mixture was stirred at rt for another 2 h. The mixture was then diluted with DCM (15 mL) and washed with water (15 mL) and brine (15 mL). The organic layer was dried over Na 2 S0 4 , filtered and evaporated under reduced pressure to afford the title compound (40 mg, 44%), which was used in the next step without purification. MS (ESI) [M+H] + 360.4.

Step 2: Preparation of Compound 18

A stirred solution of 6-bromo-N-cyclohexylimidazo[1 ,2-a]pyridine-3-sulfonamide (40 mg, 0.11 mmol) and 1 ,3-dimethyl-5-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)pyridin-2(1 H)-one (prepared using the procedure described in US20130053362, 0.033 g, 0.13 mmol) in 1 ,4- dioxane (1 mL) was purged with N 2 for 10 min. This was followed by the addition of a solution of Na 2 C0 3 (43 mg, 0.33 mmol) in water (0.5 mL), and the reaction mixture was again purged with N 2 for 10 min. After 10 min, Pd(PPh 3 ) 4 (8 mg g, 0.006 mmol) was added and the resulting mixture was heated at 100°C for 3 h. The mixture was diluted with water and the aqueous layer was extracted using EtOAc (3 x 10 mL). The combined organic layers were washed with water (10 mL) and brine (10 mL), dried over Na 2 S0 , filtered and then evaporated under reduced pressure. The material was purified by flash chromatography on silica gel using a mixture of 2- 3% MeOH in DCM as eluent to afford Example 18 (20 mg, 44%) as a solid. 1 H NMR (400 MHz, DMSO) δ ppm 8.67 (s, 1 H), 8.45 (d, J = 7.6 Hz, 1 H), 8.09 (d, J = 2.4 Hz, 1 H), 8.08 (s, 1 H), 7.89 (d, J = 9.6 Hz, 1 H), 7.82 (dd, J = 1.6 and 9.2 Hz, 1 H), 7.78 (s, 1 H), 3.55 (s, 3H), 2.96-2.93 (m, 1 H), 2.13 (s, 3H), 1.54-1.52 (m, 2H), 1.41-1.39 (m, 3H), 1.1 1-1.01 (m, 5H). MS (ESI) [M+H] + 401.4.

Example 19: 1 ,3-dimethyl-5-(3-(pyrrolidin-1-ylsulfonyl)imidazo[1 ,2-a]pyridin-6-yl)pyridin-2(1 H)- one (Compound 19)

Step 1: Preparation of 6-bromo-3-(pyrrolidin-1-ylsulfonyl)imidazo[1,2-a]pyridine

To a rt solution of pyrrolidine (72 mg, 1.02 mmol) in DCM (2 mL) was added TEA (0.28 mL, 2.03 mmol). The mixture was stirred for 5 min and then a solution of Intermediate 3 (0.2 g, 0.68 mmol) in DCM (2 mL) was added. The resulting mixture was stirred at rt for another 2h. The mixture was diluted with DCM (30 mL) and washed with water (30 mL) and brine (30 mL). The organic layer was dried over Na 2 S0 4 , filtered and evaporated under reduced pressure to afford the title compound (0.15 g, 67%) as a solid. 1 H NMR (400 MHz, DMSO) δ ppm 8.88 (s, 1 H), 8.23 (s, 1 H), 7.83 (d, J = 9.2 Hz, 1 H), 7.72 (dd, J = 1.6 and 9.6 Hz, 1 H), 3.24 (t, J = 6.6 Hz, 4H), 1.74 (t, J = 6.6 Hz, 4 H). MS (ESI) [M+H] + 332.4.

Step 2: Preparation of Compound 19

A stirred solution of 6-bromo-3-(pyrrolidin-1-ylsulfonyl)imidazo[1 ,2-a]pyridine (0.15 g, 0.45 mmol) and 1 ,3-dimethyl-5-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)pyridin-2(1 H)-one

(prepared using the procedure described in US20130053362, 0.136 g, 0.54 mmol) in 1 ,4- dioxane (3 mL) was purged with N 2 for 10 min. A solution of Na 2 C0 3 (0.145 g, 1.36 mmol) in water (1 mL) was added and the resulting mixture was purged with N 2 for another 10 min. After 10 min, Pd(PPh 3 ) 4 (26 mg, 0.02 mmol) was added and the mixture was heated at 100°C for 3 h. The solvent was evaporated under vacuum and the aqueous layer was extracted using EtOAc (3 x 20 mL). The combined organic layers were washed with water (20 mL) and brine (20 mL), dried over Na 2 S0 4 , filtered and then evaporated under reduced pressure. The material was purified by flash chromatography on silica gel using a mixture of 2% MeOH in DCM as eluent to afford Compound 19 (0.1 1 g, 41 %) as a solid. 1 H NMR (400 MHz, DMSO) δ ppm 8.80 (s, 1 H), 8.21 (s, 1 H), 8.04 (d, J = 2.4 Hz, 1 H), 7.90 (d, J = 9.6 Hz, 1 H), 7.79 (dd, J = 1.6 and 2 Hz, 1 H), 7.65 (d, J = 1.6 Hz, 1 H), 3.54 (s, 3H), 3.25 (t, J = 6.8 Hz, 4H), 2.11 (s, 3H), 1.75-1.71 (m, 4H). MS (ESI) [M+H] + 373.5. Example 20: 5-[3-[(4,4-difluoro-1-piperidyl)methyl]imidazo[1 ,2-a]pyridin-6-yl]-1 ,3-dimethyl- pyridin-2-one (Compound 20)

Step 1: Preparation of 6-bromo-3-[(4,4-difluoro- 1-piperidyl)methyl]imidazo[ 1 ,2-a]pyridine

To a solution of Intermediate 1 (75 mg, 0.31 mmol) in DMF (5 ml_) was added 4,4- difluoropiperidine hydrochloride (72 mg, 0.46 mmol) and DIPEA (0.19 ml_, 1.07 mmol). The reaction mixture was stirred at rt for 18 h. The resulting mixture was diluted with EtOAc (10 ml_) and water (10 ml_) and the organic layer was separated, dried over MgS0 4 , filtered and concentrated under reduced pressure. The material was purified by flash chromatography on silica gel using a mixture of MeOH in DCM as eluent to afford the title compound (46 mg, 46%) as a solid. MS (ESI) [M+H] + 330.1/332.1.

Step 2: Preparation of Compound 20

To a solution of 6-bromo-3-[(4,4-difluoro-1-piperidyl)methyl]imidazo[1 ,2-a]pyridine (46 mg, 0.14 mmol) in DME (5 ml_) and water (0.5 ml_) was added 1 ,3-dimethyl-5-(4,4,5,5-tetramethyl-1 ,3,2- dioxaborolan-2-yl)pyridin-2-one (prepared using the procedure described in US20130053362, 65 mg, 0.167 mmol), Cs 2 C0 3 (113.5 mg, 0.35 mmol) and Pd(PPh 3 ) 4 (16 mg, 0.014 mmol). The resulting mixture was degassed for 5 min with N 2 and then heated in a sealed tube at 90°C for 18 h. The mixture was cooled to rt, diluted with EtOAc (10 ml_) and water (5 ml_). The organic layer was separated, dried over MgS0 4 , filtered and concentrated under reduced pressure. The material was purified by flash chromatography on silica gel using a mixture of MeOH in DCM as eluent, followed by preparative HPLC purification to afford Compound 20 (14.6 mg, 28%) as a solid. 1 H NMR (500 MHz, CDCI 3 ) δ 8.27 (s, 1 H), 7.66 (d, J = 9.1 Hz, 1 H), 7.55 (s, 1 H), 7.43 (dd, J = 2.5, 1.1 Hz, 1 H), 7.36 (d, J = 2.4 Hz, 1 H), 7.32 - 7.24 (m, 2H), 3.89 (s, 1 H), 3.66 (s, 3H), 2.60 (d, J = 5.4 Hz, 4H), 2.26 (s, 3H), 2.05 - 1.89 (m, 4H). MS (ESI) [M+H] + 373.1.

Example 21 : 1 ,3-dimethyl-5-[3-(morpholinomethyl)imidazo[1 ,2-a]pyridin-6-yl]pyridin-2-one (Compound 21)

Step 1: Preparation of 4-[(6-bromoimidazo[1,2-a]pyridin-3-yl)methyl]morpholine

To a solution of Intermediate 1 (75 mg, 0.305 mmol) in DMF (3 mL) was added morpholine (67 μΙ_, 0.76 mmol) and the reaction mixture was stirred at rt for 18 h. The mixture was diluted with EtOAc (10 mL) and water (10 mL). The organic layer was separated, dried over MgS0 4 , filtered and concentrated under reduced pressure. The material was purified by flash chromatography on silica gel using a mixture of EtOAc and hexane as eluent to afford the title compound (40 mg, 44%) as a solid.

Step 2: Preparation of Compound 21

To a suspension of 4-[(6-bromoimidazo[1 ,2-a]pyridin-3-yl)methyl]morpholine (40 mg, 0.16 mmol) in DME (3 mL) was added 1 ,3-dimethyl-5-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2- yl)pyridin-2-one (prepared using the procedure described in US20130053362, 40 mg, 0.16 mmol), Cs 2 C0 3 (110 mg, 0.34 mmol) and Pd(PPh 3 ) (16 mg, 0.014 mmol). The reaction mixture was degassed for 5 min and then heated in a sealed tube at 90°C for 18 h. The mixture was cooled to rt and diluted with EtOAc (10 mL) and water (10 mL). The organic layer was separated, dried over MgS0 , filtered and concentrated under reduced pressure. The material was purified by flash chromatography on silica gel using a mixture of MeOH in DCM as eluent, followed by preparative HPLC purification to afford Compound 21 (10.6 mg, 23%) as a solid. 1 H NMR (500 MHz, CDCI 3 ) δ 8.34 (s, 1 H), 7.64 (d, J = 9.2 Hz, 1 H), 7.53 (s, 1 H), 7.44 (dd, J = 2.5, 1.2 Hz, 1 H), 7.36 (d, J = 2.4 Hz, 1 H), 7.28 - 7.21 (m, 1 H), 3.84 (s, 2H), 3.71 - 3.66 (m, 4H), 3.65 (s, 3H), 2.47 (d, J = 4.3 Hz, 4H), 2.25 (s, 3H). MS (ESI) [M+H] + 339.3.

Example 22: 5-[3-(cyclopentylamino)imidazo[1 ,2-a]pyridin-6-yl]-1 ,3-dimethyl-pyridin-2-one (Compound 22)

Cyclopentanone (0.13 mL, 1.4 mmol) was added to a solution of Intermediate 4 (178 mg, 0.7 mmol) in DCM (5 mL) and the resulting mixture was stirred at rt for 18 h. The solution was evaporated under reduced pressure. The residue was dissolved in MeOH (5 mL) and then NaBH 4 (79 mg, 2.1 mmol) was added and the reaction mixture was stirred at rt for another 1 h. The mixture was concentrated and the residue was diluted in EtOAc (10 mL) and water (10 mL). The organic layer was separated, dried over MgS0 4 , filtered and concentrated under reduced pressure. The material was purified by preparative HPLC to afford Compound 22 (4 mg, 2%) as a solid. 1 H NMR (500 MHz, CDCI 3 ) δ 7.97 (s, 1 H), 7.57 (d, J = 9.4 Hz, 1 H), 7.47 (s, 1 H), 7.42 (s, 1 H), 7.20 - 7.15 (m, 2H), 3.76 (s, 1 H), 3.66 (d, J = 8.0 Hz, 3H), 2.25 (s, 3H), 2.02 - 1.93 (m, 2H), 1.80 (s, 2H), 1.66 - 1.48 (m, 5H). MS (ESI) [M+H] + 323.3.

Example 23: 5-[3-(cyclopentylmethylamino)imidazo[1 ,2-a]pyridin-6-yl]-1 ,3-dimethyl-pyridin-2- one (Compound 23)

To a solution of Intermediate 4 (300 mg, 1.18 mmol) in DCM (5 mL) was added cyclopentane carbaldehyde (231 mg, 2.36 mmol) and the mixture was stirred at rt for 18 h. The solution was concentrated under reduced pressure and the residue was dissolved in MeOH (5 mL). NaBH (134 mg, 3.54 mmol) was added and the reaction mixture was stirred at rt for 1 h. The mixture was concentrated and the residue was diluted in EtOAc (10 mL) and water (10 mL). The organic layer was separated, dried over MgS0 4 , filtered and concentrated under reduced pressure. The material was purified by preparative HPLC to afford Compound 23 (12 mg, 3%) as a solid. 1 H NMR (500 MHz, CDCI 3 ) δ 7.91 (dd, J = 1.7, 0.9 Hz, 1 H), 7.58 - 7.52 (m, 1 H), 7.47 (dd, J = 2.5, 1.2 Hz, 1 H), 7.39 (d, J = 2.5 Hz, 1 H), 7.18 (s, 1 H), 7.12 (dd, J = 9.3, 1.8 Hz, 1 H), 3.64 (s, 3H), 3.09 (d, J = 6.5 Hz, 2H), 2.24 (d, J = 8.0 Hz, 3H), 2.23 - 2.16 (m, 1 H), 1.93 - 1.81 (m, 2H), 1.66 (dt, J = 8.9, 7.7 Hz, 3H), 1.63 - 1.56 (m, 2H), 1.38 - 1.24 (m, 2H). MS (ESI) [M+H] + 337.4. Example 24: 5-(3-benzyl-2-methyl-imidazo[1 ,2-a]pyridin-6-yl)-1 ,3-dimethyl-pyridin-2-one (Compound 24)

Step 1: Preparation of 5-(3-benzoyl-2-methyl-imidazo[1,2-a]pyndin-6-yl)-1,3-dim

one

To a solution of (6-bromo-2-methyl-imidazo[1 ,2-a]pyridin-3-yl)-phenyl-methanone (prepared using the procedure described in WO2015086498, 279 mg, 0.88 mmol) in DME (3 mL) and water (0.3 mL) was added 1 ,3-dimethyl-5-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)pyridin-2- one (prepared using the procedure described in US20130053362, 264 mg, 1.06 mmol), Cs 2 C0 3 (721 mg, 2.21 mmol) and Pd(PPh 3 ) 4 (102 mg, 0.089 mmol). The reaction mixture was degassed for 5 min and then heated in a sealed tube at 80 °C for 18 h. The mixture was cooled to rt, diluted with EtOAc (10 mL) and water (10 mL). The organic layer was separated, dried over MgS0 4 , filtered and concentrated under reduced pressure. The material was purified by flash chromatography on silica gel using a mixture of MeOH in DCM as eluent to afford the title compound (200 mg, 63%) as a solid. 1 H NMR (500 MHz, CDCI 3 ) δ 9.47 (dd, J = 1.8, 0.8 Hz, 1 H), 8.07 (d, J = 2.5 Hz, 1 H), 7.81 (ddd, J = 10.0, 9.3, 1.3 Hz, 2H), 7.74 - 7.61 (m, 4H), 7.57 (t, J = 7.5 Hz, 2H), 3.53 (s, 3H), 2.10 (s, 3H), 2.04 (s, 3H). MS (ESI) [M+H] + 358.2.

Step 2: Preparation of Compound 24

A solution of 5-(3-benzoyl-2-methyl-imidazo[1 ,2-a]pyridin-6-yl)-1 ,3-dimethyl-pyridin-2-one (50 mg, 0.14 mmol), hydrazine hydrate (0.40 mL, 4.57 mmol) and KOH (63 mg, 1.12 mmol) in ethylene glycol (1 mL) was heated in a sealed vial at 150°C for 20 h. The mixture was then cooled to rt and diluted with DCM (10 mL) and H 2 0 (10 mL). The aqueous layer was separated and extracted with DCM (3 x 10 mL). The combined organic layers were dried over Na 2 S0 4 , filtered and concentrated under reduced pressure. The material was purified by preparative HPLC to provide Compound 24 (13 mg, 27 %) as a solid. 1 H NMR (500 MHz, CDCI 3 ) δ 7.61 (dd, J = 1.7, 0.9 Hz, 1 H), 7.56 (dd, J = 9.2, 0.9 Hz, 1 H), 7.34 - 7.22 (m, 4H), 7.14 (ddd, J = 6.8, 5.7, 3.1 Hz, 4H), 4.29 (s, 2H), 3.58 (s, 3H), 2.52 (s, 3H), 2.19 (s, 3H). MS (ESI) [M+H] + 344.2.

Example 25: 5-[3-(cyclobutoxymethyl)imidazo[1 ,2-a]pyridin-6-yl]-1 ,3-dimethyl-pyridin-2-one (Compound 25)

Step 1: Preparation of 6-bromo-3- cyclobutoxymethyl)imidazo[ 1,2-a]pyridine

Intermediate 1 (100 mg, 0.41 mmol) was added to a solution of cyclobutanol (64 μί, 0.815 mmol) and DIPEA (0.212 mL, 1.22 mmol) in THF (1 mL) and the reaction mixture was heated at 55°C for 3 d. The mixture was cooled and then diluted with EtOAc (30 mL) and saturated NaHC0 3 (20 mL). The aqueous layer was separated and extracted with EtOAc (3 x 20 mL). The combined organic layers were washed with brine, dried over Na 2 S0 , filtered and then concentrated under reduced pressure. The material was purified by flash chromatography on silica gel using a mixture of EtOAc in hexane as eluent to provide the title compound (22 mg, 19 %). 1 H NMR (500 MHz, CDCI 3 ) δ 8.33 (dd, J = 1.8, 0.8 Hz, 1 H), 7.55 (s, 1 H), 7.50 (dt, J = 9.2, 2.6 Hz, 1 H), 7.30 - 7.25 (m, 1 H), 4.69 (s, 2H), 4.00 - 3.90 (m, 1 H), 2.21 - 2.10 (m, 2H), 1.97 - 1.85 (m, 2H), 1.76 - 1.65 (m, 1 H), 1.57 - 1.42 (m, 1 H). MS (ESI) [M+H] + 281.2.

Step 2: Preparation of Compound 25

Pd(PPh 3 ) 4 (9 mg, 0.008 mmol) was added to a degassed solution of 6-bromo-3- (cyclobutoxymethyl)imidazo[1 ,2-a]pyridine (22 mg, 0.078 mmol), 1 ,3-dimethyl-5-(4,4,5,5- tetramethyl-1 ,3,2-dioxaborolan-2-yl)pyridin-2-one (prepared using the procedure described in US20130053362, 25 mg, 0.102 mmol), and Cs 2 C0 3 (64 mg, 0.196 mmol) in DME (3 mL) and water (0.3 mL) under N 2 . The resulting mixture was heated at 80°C for 20 h and then cooled to rt. The mixture was filtered through a pad of Celite™ and washed with EtOAc (100 mL). The organic layer was washed with saturated NaHC0 3 (100 mL) and the aqueous layer was re- extracted with EtOAc (3 x 50 mL). The combined organic layers were dried over Na 2 S0 4 , filtered, and concentrated under reduced pressure. The material was purified by flash chromatography on silica gel using a mixture of EtOAc in hexane as eluent, followed by preparative HPLC to provide Compound 25 (3.8 mg, 15%) as a solid. 1 H NMR (500 MHz, CDCI 3 ) δ 8.20 (s, 1 H), 7.66 (d, J = 9.4 Hz, 1 H), 7.59 (s, 1 H), 7.46 (s, 1 H), 7.39 (d, J = 2.4 Hz, 1 H), 7.29 (dd, J = 9.4, 1.6 Hz, 1 H), 4.78 (s, 2H), 4.01 (s, 1 H), 3.65 (s, 3H), 2.25 (s, 3H), 2.19 - 2.09 (m, 2H), 1.90 (qd, J = 10.0, 2.7 Hz, 2H), 1.72 - 1.67 (m, 1 H), 1.49 (dq, J = 10.7, 8.1 Hz, 1 H). MS (ESI) [M+H] + 324.2.

Example 26: 1 ,3-dimethyl-5-[3-[(2-oxo-1-piperidyl)methyl]imidazo[1 ,2-a]pyridin-6-yl]pyridin-2- one (Compound 26)

Step 1: Preparation of 1-[(6-bromoimidazo[1,2-a]pyridin-3-yl)methyl]piperidin-2-one To a solution of piperidin-2-one (38 mg, 0.383 mmol) in DMF (1 mL) was added NaH (60 % dispersion in mineral oil, 24 mg, 0.61 1 mmol) and the mixture was stirred for 20 min. Intermediate 1 (30 mg, 0.122 mmol) was then added and the reaction mixture was stirred for 2 h at rt. The mixture was quenched with water and concentrated under reduced pressure. The residue was diluted with saturated NaHC0 3 (10 mL) and EtOAc (30 mL), and the aqueous layer was extracted with EtOAc (3 x 10 mL). The combined organic layers were dried over Na 2 S0 4 , filtered, and concentrated under reduced pressure to afford the title compound, which was used in the next step without further purification. MS (ESI) [M+H] + 308.1/310.1.

Step 2: Preparation of Compound 26

A solution of 1-[(6-bromoimidazo[1 ,2-a]pyridin-3-yl)methyl]piperidin-2-one (38 mg, 0.122 mmol) and 1 ,3-dimethyl-5-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)pyridin-2-one (prepared using the procedure described in US20130053362, 39.5 mg, 0.159 mmol) in DME (2 mL) and water (0.1 mL) was degassed for 10 min. Cs 2 C0 3 (83 mg, 0.256 mmol) and Pd(PPh 3 ) 4 (14 mg, 0.012 mmol) were then added and the mixture was degassed for another 10 min. The resulting mixture was then heated at 85°C for 2 h. The mixture was cooled to rt and was diluted with saturated NaHC0 3 (10 mL) and EtOAc (30 mL). The layers were separated and the aqueous layer was extracted with EtOAc (3 x 10 mL). The combined organic layers were dried over MgS0 , filtered, and concentrated under reduced pressure. The material was purified by flash chromatography on silica gel using a mixture of EtOAc in hexane as eluent, followed by preparative HPLC to provide Compound 26 (7 mg, 16%) as a solid. 1 H NMR (500 MHz, CDCI 3 ) δ 8.89 (s, 1 H), 7.77 (d, J = 9.3 Hz, 1 H), 7.66 (s, 1 H), 7.51 (d, J = 20.4 Hz, 1 H), 7.48 (d, J = 2.4 Hz, 1 H), 7.41 (dd, J = 9.3, 1.6 Hz, 1 H), 4.95 (s, 2H), 3.67 (s, 3H), 3.33 - 3.19 (m, 2H), 2.50 - 2.36 (m, 2H), 2.26 (s, 3H), 1.90 - 1.58 (m, 4H). MS (ESI) [M+H] + 351.2. Example 27: 5-(3-anilinoimidazo[1 ,2-a]pyridin-6-yl)-1 ,3-dimethyl-pyridin-2-one (Compound 30)

Step 1: Preparation of 6-bromo-N-phenyl-imidazo[1 ,2-a]pyridin-3-amine

To a solution of 5-bromopyridin-2-amine (1.0 g, 5.78 mmol) in DCE (20 ml) and MeOH (10 ml_) was added glyoxylic acid monohydrate (638.4 mg, 6.94 mmol) and the mixture was stirred at rt for 30 min. To the solution, phenyl isonitrile (1.16 g, 11.28 mmol) was added and the reaction mixture was stirred at rt for 48 h. The mixture was diluted with DCM (20 ml_), treated with solid NaHC0 3 (2.5 g), then activated charcoal and stirred at rt for 15 min. The mixture was filtered through a plug of Celite™ and the filtrate was concentrated under reduced pressure. The material was purified by flash chromatography on silica using a mixture of MeOH in DCM as eluent and then further triturated with Et 2 0 and hexane to afford the title compound (935 mg, 56%) as a solid. 1 H NMR (500 MHz, DMSO) δ 8.17 (dd, J = 1.9, 0.9 Hz, 1 H), 8.05 (s, 1 H), 7.57 (dd, J = 9.5, 0.8 Hz, 1 H), 7.51 (d, J = 0.6 Hz, 1 H), 7.33 (dd, J = 9.5, 1.9 Hz, 1 H), 7.16 (dd, J = 8.5, 7.4 Hz, 2H), 6.80 - 6.72 (m, 1 H), 6.64 - 6.57 (m, 2H). MS (ESI) [M+H] + 288.1/290.1.

Step 2: Preparation of Compound 30

To a solution of 6-bromo-N-phenyl-imidazo[1 ,2-a]pyridin-3-amine (102.3 mg, 0.355 mmol) in a mixture of DME (2 ml_) and water (0.2 ml_), were added 1 ,3-dimethyl-5-(4,4,5,5-tetramethyl- 1 ,3,2-dioxaborolan-2-yl)pyridin-2-one (US2013/0053362; 106.13 mg, 0.426 mmol), Cs 2 C0 3 (289 mg, 0.888 mmol) and Pd(PPh 3 ) 4 (41.2 mg, 0.035 mmol) and the resulting mixture was degassed with N 2 for 5 min and then heated to 100 °C in a sealed tube for 18 h. The mixture was cooled to rt and diluted with EtOAc (10 ml_) and water (10 ml_). The organic phase was separated, dried over MgS0 , filtered and concentrated under reduced pressure. The material was purified by flash chromatography on silica gel using a mixture MeOH in DCM as eluent, followed by preparative HPLC purification to afford Compound 30 (18.8 mg, 16%) as a solid. 1 H NMR (500 MHz, CDCI 3 ) δ 7.90 (dd, J = 1.8, 0.9 Hz, 1 H), 7.67 (dd, J = 9.3, 0.9 Hz, 1 H), 7.56 (d, J = 0.7 Hz, 1 H), 7.37 (dd, J = 2.6, 1.2 Hz, 1 H), 7.32 (s, 1 H), 7.29 (dd, J = 9.3, 1.9 Hz, 1 H), 7.24 - 7.19 (m, 2H), 6.88 (m, 1 H), 6.62 - 6.57 (m, 2H), 5.56 (s, 1 H), 3.60 (s, 3H), 2.18 (s, 3H). MS (ESI) [M+H] + 331.2.

Example 28: 1 ,3-dimethyl-5-[3-(2,2,2-trifluoroethoxymethyl)imidazo[1 ,2-a]pyridin-6-yl]pyridin-2- one (Compound 34)

Step 1: Preparation of 6-bromo-3-(2,2 2-trifluoroethoxymethyl)imidazo[1,2-a]pyridine

To a solution of 2,2,2-trifluoroethanol (408.95 mg, 4.07 mmol) in DMF (5 mL) at 0 °C was added NaH (162.9 mg, 4.07 mmol) and the mixture was stirred for 10 min. To the mixture, Intermediate 1 (100.0 mg, 0.407 mol) and DIPEA (0.142 mL, 0.815 mmol) in DMF (2 mL) were added and the reaction mixture was stirred at rt for 4 h. The mixture was poured into water (20 mL) and the aqueous layer was extracted with EtOAc (3 x 10 mL). The combined organic layers were dried over MgS0 4 , filtered and concentrated under reduced pressure. The material was purified by flash chromatography on silica gel using a mixture of EtOAc in hexane as eluent to afford the title compound (102.2 mg, 81 %) as a solid. 1 H NMR (500 MHz, CDCI 3 ) δ 8.32 (dd, J = 1.8, 0.8 Hz, 1 H), 7.63 (s, 1 H), 7.55 (dd, J = 9.5, 0.8 Hz, 1 H), 7.33 (dd, J = 9.5, 1.9 Hz, 1 H), 4.97 (s, 2H), 3.79 (q, J = 8.6 Hz, 2H). MS (ESI) [M+H] + 309.1/31 1.1.

Step 2: Preparation of Compound 34

Pd(PPh 3 ) (9 mg, 0.008 mmol) was added to a degassed solution of 6-bromo-3-(2,2,2 trifluoroethoxymethyl)imidazo[1 ,2-a]pyridine (22 mg, 0.078 mmol), and 1 ,3-dimethyl-5-(4,4,5,5 tetramethyl-1 ,3,2-dioxaborolan-2-yl)pyridin-2-one (prepared as in US2013/0053362; 25 mg, 0.102 mmol) and Cs 2 C0 3 (64 mg, 0.196 mmol) in a mixture of DME (3 mL) and water (0.3 mL) under N 2 . The resulting reaction mixture was heated to 80 °C for 20 h and then cooled to rt. The mixture was filtered through Celite™ and washed with EtOAc (100 mL). To the filtrate, saturated NaHC0 3 (25 mL) was added and the aqueous phase was extracted with EtOAc (3 x 50 mL). The combined organic layers were dried over Na 2 S0 4 , filtered, and concentrated under reduced pressure. The material was purified by flash chromatography on silica gel using a mixture of EtOAc in hexane as eluent, followed by preparative HPLC to provide Compound 34 (3.8 mg, 15 %) as a solid. 1 H NMR (500 MHz, CDCI 3 ) δ 8.21 (s, 1 H), 7.74 - 7.65 (m, 2H), 7.45 (d, J = 1.3 Hz, 1 H), 7.39 (d, J = 2.5 Hz, 1 H), 7.35 (dd, J = 9.3, 1.8 Hz, 1 H), 5.04 (s, 2H), 3.83 (q, J = 8.7 Hz, 2H), 3.65 (s, 3H), 2.25 (s, 3H). MS (ESI) [M+H] + 352.3.

Example 29: 6-(1 ,5-dimethyl-6-oxo-1 ,6-dihydropyridin-3-yl)-N-(tetrahydro-2H-pyran-4- yl)imidazo[1 ,2-a]pyridine-3-sulfonamide (Compound 42)

Step 1: Preparation of 6-bromo-N-(tetrahydro-2H-pyran-4-yl)imidazo[1,2-a]pyridine-3 - sulfonamide

TEA (0.71 mL, 5.0754 mmol) was added to a room temperature stirred solution of tetrahydro- 2H-pyran-4-amine hydrochloride (0.349 g, 2.5377 mmol) in DCM (5 mL), followed by the addition of a solution of Intermediate 3 (0.5 g, 1.6918 mmol) in DCM (5 mL). The reaction mixture was stirred for 2 h and, after completion, was concentrated under reduced pressure. The product obtained was purified by silica gel column chromatography using a gradient of 50- 70% ethyl acetate in hexane as eluent. The fractions were combined and concentrated to afford the title compound (0.2 g, 33%) as a semisolid (MH + 360.04).

Step 2: Preparation of Compound 42

A stirred solution of 6-bromo-N-(tetrahydro-2H-pyran-4-yl)imidazo[1 ,2-a]pyridine-3-sulfonarnide (0.2 g, 0.5552 mmol) and 1 ,3-dimethyl-5-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)pyridin- 2(1 H)-one (0.208 g, 0.8328 mmol) in 1 ,4-dioxane (5 mL) was purged with nitrogen for 20 min, followed by the addition of sodium carbonate (0.177 g, 1.6656 mmol) in water (0.5 mL). The reaction mixture was purged again with nitrogen for 20 min. Pd(PPh 3 ) 4 (0.032 g, 0.0277 mmol) was then added and the reaction mixture was heated at 100°C for 6 h. The reaction mixture was concentrated under reduced pressure and the crude product was purified by silica gel chromatography using 2-3% methanol in DCM as eluent. Fractions were combined and concentrated to afford Compound 42 (0.060 g, 27%) as a solid. 1 H NMR (400 MHz, DMSO) δ 8.67 (s, 1 H), 8.61 (d, J = 7.6 Hz, 1 H), 8.1 1 (s, 1 H), 8.09 (s, 1 H), 7.9 (d, J = 9.6 Hz, 1 H), 7.83 (dd, J = 1.6 and 9.6 Hz, 1 H), 7.78 (d, J = 1.2 Hz, 1 H), 3.69-3.66 (m, 2H), 3.56 (s, 3H), 3.22-3.16 (m, 3H), 2.14 (s, 3H), 1.39-1.27 (m, 4H), MH + 403.29.

Example 30: 6-(1 ,5-dimethyl-6-oxo-1 ,6-dihydropyridin-3-yl)-N-(tetrahydro-2H-pyran-3- yl)imidazo[1 , 2-a]pyridine-3-sulfon )

Compound 43 (RT 9.02) - Compound 44 (RT 1 1.09)

Step 1: Preparation of 6-bromo-N-(tetrahydro-2H^yran-3-yl)imidazo[1,2-a]pyridine-3- sulfonamide

TEA (1.3 mL, 9.1358 mmol) was added to a room temperature stirred solution of tetrahydro-2H- pyran-3-amine hydrochloride (0.629 g, 4.5679 mmol) in DCM (5 mL), followed by the addition of a solution of Intermediate 3 (0.9 g, 3.0453 mmol) in DCM (5 mL). The reaction mixture was stirred at rt for 2 h and, after completion of reaction, was concentrated under reduced pressure. The product obtained was purified by silica gel chromatography using a gradient of 50-70% ethyl acetate in hexane as eluent. Fractions were combined and concentrated to afford the title compound (0.32 g, 27%) as a semisolid. MH + 362.45.

Step 2: Preparation of Compounds 43 and 44

A stirred solution of 6-bromo-N-(tetrahydro-2H-pyran-3-yl)imidazo[1 ,2-a]pyridine-3-sulfonamide (0.32 g, 0.8883 mmol) and 1 ,3-dimethyl-5-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)pyridin- 2(1 H)-one (0.332 g, 1.3325 mmol) in 1 ,4-dioxane (5 mL) was purged with nitrogen for 20 min, followed by the addition of sodium carbonate (0.283 g, 2.665 mmol) in water (0.5 mL). The reaction mixture was purged with nitrogen for another 20 min. Pd(PPh 3 ) 4 (0.051 g, 0.0444) was the added and the reaction mixture was heated at 100°C for 6 h. The reaction mixture was concentrated under reduced pressure and the crude product was purified by silica gel chromatography using 2-3% methanol in DCM as eluent. Fractions were combined and concentrated to afford the title compound in racemic form (0.125 g), which was further purified by chiral preparative HPLC (Chiralpak™ IC (250*21) mm, 5μ Column, flow 15.0 mL/min, isocratic 30% n-Hexane in I PA: Methanol (70:30)) to afford two isomers in separate fractions: Fraction 1 (Compound 43): 0.025 g and Fraction 2 (Compound 44): 0.030 g.

Compound 43: Retention Time = 9.02: 1 H NMR (400 MHz, DMSO) δ ppm 8.69 (s, 1 H), 8.65 (bs, 1 H), 8.11 (s, 2H), 7.90 (d, J = 9.6 Hz, 1 H), 7.83 (dd, J = 1.6 and 9.6 Hz, 1 H), 7.79 (d, J = 1.2 Hz, 1 H), 3.55-3.53 (m, 4H), 3.48-3.42 (m, 1 H), 3.26-3.21 (m, 1 H), 3.06-3.00 (m, 2H), 2.14 (s, 3H), 1.56-1.53 (m, 1 H), 1.49-1.46 (m, 1 H), 1.37-1.26 (m, 2H), M+1 : 403.29.

Compound 44: Retention Time = 11.09: 1 H NMR (400 MHz, DMSO) δ ppm 8.69 (s, 1 H), 8.65 (bs, 1 H), 8.1 1 (s, 1 H), 8.10 (s, 1 H), 7.90 (d, J = 9.2 Hz, 1 H), 7.83 (dd, J = 1.6 and 9.2 Hz, 1 H), 7.79 (d, J = 1.6 Hz, 1 H), 3.56-3.53 (m, 4H), 3.45-3.42 (m, 1 H), 3.26-3.21 (m, 1 H), 3.04-3.02 (m, 2H), 2.14 (s, 3H), 1.56-1.53 (m, 1 H), 1.49-1.46 (m, 1 H), 1.35-1.26 (m, 2H), MH + : 403.29.

Example 31 : (R)-1 ,3-dimethyl-5-(3-((2-methylpyrrolidin-1-yl)sulfonyl)imidazo[ 1 ,2-a] pyridin-6- yl)pyridin-2(1 H)-one (Compound 45)

Step 1: Preparation of (R)-6-bromo-3-((2-methylpyrrolidin-1-yl)sulfonyl)imidazo[ 1,2-a] pyridine

TEA (0.42 mL, 3.0453 mmol) was added to a room temperature stirred solution of (R)-2- methylpyrrolidine hydrochloride (0.185 g, 1.5226 mmol) in DCM (5 mL), followed by the addition of a solution of Intermediate 3 (0.3 g, 1.0151 mmol) in DCM (5 mL). The reaction mixture was stirred at rt for 2 h and, after completion, was concentrated under reduced pressure. The product obtained was purified by silica gel chromatography using a gradient of 50-70% ethyl acetate in hexane as eluent. The fractions were combined and concentrated to afford the title compound (0.07 g, 20%). MH + 346.19.

Step 2: Preparation of Compound 45

A stirred solution of (R)-6-bromo-3-((2-methylpyrrolidin-1-yl)sulfonyl)imidazo[1 ,2-a]pyridine (0.070 g, 0.2033 mmol) and 1 ,3-dimethyl-5-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)pyridin- 2(1 H)-one (0.076 g, 0.3050 mmol) in 1 ,4-dioxane (5 mL) was purged with nitrogen for 20 min, followed by the addition of sodium carbonate (0.065 g, 0.61 mmol) in water (0.5 mL). The reaction mixture was purged with nitrogen for another 20 min. Pd(PPh 3 ) 4 (0.012 g, 0.0101 mmol) was then added and the reaction mixture was heated at 100°C for 6 h. The reaction mixture was concentrated under reduced pressure and the crude product was purified by silica gel chromatography using 2-3% methanol in DCM as eluent. Fractions were combined and concentrated to afford Compound 45 (0.025 g, 16%). 1 H NMR (400 MHz, DMSO) δ 8.77 (s, 1 H), 8.23 (s, 1 H), 8.05 (d, J = 2.4 Hz, 1 H), 7.90 (d, J = 9.2 Hz, 1 H), 7.80 (dd, J = 1.6 and 2 Hz, 1 H), 7.63 (d, J = 1.2 Hz, 1 H), 3.84-3.80 (m, 1 H), 3.55 (s, 3H), 3.43-3.37 (m, 1 H), 3.24-3.17 (m, 1 H), 2.11 (s, 3H), 1.84-1.75 (m, 2H), 1.58-1.48 (m, 2H), 1.24 (d, J = 6.4 Hz, 3H). MH + 387.34.

Example 32: (S)-1 ,3-dimethyl-5-(3-((2-methylpyrrolidin-1-yl)sulfonyl)imidazo[ 1 ,2-a] pyridin-6- yl)pyridin-2(1 H)-one (Compound

Step 1: Preparation o -6-bromo-3-((2-methylpyrrolidin- 1-yl)sulfonyl)imidazo[ 1,2-a] pyridine

TEA (0.71 mL, 5.0754 mmol) was added to a room temperature stirred solution of (S)-2- methylpyrrolidine hydrochloride (0.309 g, 2.5377 mmol) in DCM (5 mL), followed by the addition of a solution of Intermediate 3 (0.5 g, 1.6918 mmol) in DCM (5 mL). The reaction mixture was stirred at rt for 2 h and, after completion, was concentrated under reduced pressure. The product obtained was purified by silica gel chromatography using a gradient of 50-70% ethyl acetate in hexane as eluent. Fractions were combined and concentrated to afford the title compound (0.1 g, 17%) as a semisolid. MH + 346.0.

Step 2: Preparation of Compound 46

A stirred solution of (S)-6-bromo-3-((2-methylpyrrolidin-1-yl)sulfonyl)imidazo[1 ,2-a]pyridine (0.1 g, 0.2905 mmol) and 1 ,3-dimethyl-5-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)pyridin-2(1 H)- one (0.109 g, 0.4357 mmol) in 1 ,4-dioxane (5 mL) was purged with nitrogen for 20 min, followed by the addition of sodium carbonate (0.092 g, 0.8715 mmol) in water (0.5 mL). The reaction mixture was again purged with nitrogen for 20 min. Pd(PPh 3 ) 4 (0.017 g, 0.0145 mmol) was then added and the reaction mixture was heated at 100 °C for 6 h. The reaction mixture was concentrated under reduced pressure and the crude product was purified by silica gel chromatography using 2-3% methanol in DCM as eluent. Fractions were combined and concentrated to dryness to afford 0.1 g, which product was repurified by preparative HPLC using 30% acetonitrile in water with ammonium acetate to afford Compound 46 (0.015 g, 13%) as a solid. 1 H N MR (400 MHz, DMSO) δ 8.79 (s, 1 H), 8.23 (s, 1 H), 8.05 (d, J = 2.8 Hz, 1 H), 7.90 (dd, J = 5.6 Hz and 0.8 Hz, 1 H), 7.80 (dd, J = 9 9.6 Hz and 2 Hz, 1 H), 7.63 (dd, J = 1.2 and 2.4 Hz, 1 H), 3.84-3.80 (m, 1 H), 3.55 (s, 3H), 3.43-3.37 (m, 1 H), 3.24-3.17 (m, 1 H), 2.1 1 (s, 3H), 1.87-1.73 (m, 2H), 1.58-1.48 (m, 2H), 1.24 (d, J = 6.4 Hz, 3H). MH + 387.34. Example 33: 6-(1 ,5-dimethyl-6-oxo-1 ,6-dihydropyridin-3-yl)-N-methyl-N-(tetrahydrofuran -3- yl)imidazo[1 , 2-a]pyridine-3-sulfonamide (Compounds 47 and 48)

Compound 47 (RT 8.43) - Compound 48 (RT 10.17)

Step 1: Preparation of 6-bromo-N-methyl-N-(tetrahydrofuran-3-yl)imidazo[1,2-a]pyrid ine sulfonamide

TEA (1.5 mL, 10.963 mmol) was added to a room temperature stirred solution of N- methyltetrahydrofuran-3-amine hydrochloride (0.754 g, 5.4815 mmol) in DCM (5 mL), followed by the addition of a solution of Intermediate 3 (1.08 g, 3.6543 mmol) in DCM (5 mL). The reaction mixture was stirred at rt for 20 h and, after completion, was concentrated under reduced pressure. The product obtained was purified by silica gel chromatography using a gradient of 50-70% ethyl acetate in hexane as eluent. The fractions were combined and concentrated to dryness to afford the title compound (0.24 g, 18%) as a semisolid. MH + 360.14.

Step 2: Preparation of Compounds 47 and 48

A stirred solution of 6-bromo-N-methyl-N-(tetrahydrofuran-3-yl)imidazo[1 ,2-a]pyridine-3- sulfonamide (0.24 g, 0.6662 mmol) and 1 ,3-dimethyl-5-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan- 2-yl)pyridin-2(1 H)-one (0.249 g, 0.9993 mmol) in 1 ,4-dioxane (5 mL) was purged with nitrogen for 20 min, followed by the addition of sodium carbonate (0.212 g, 1.9987 mmol) in water (0.5 mL). The reaction mixture was again purged with nitrogen for 20 min. Pd(PPh 3 ) 4 (0.038 g, 0.0333) was then added and the reaction mixture was heated at 100°C for 6 h. The reaction mixture was concentrated under reduced pressure and the crude product was purified by silica gel chromatography using 2-3% methanol in DCM as eluent. Fractions were combined and concentrated to dryness to afford the title compound in racemic form (0.070 g) which was further purified by chiral preparative HPLC (ChiralCEL™ OJ-H (250*20)mm, 5μ Column, flow 70.0 mL/min, isocratic 80% liquid C0 2 and 20% I PA) to afford two isomers in separate fractions: Fraction 1 (Compound 47): 0.018 g and Fraction 2 (Compound 48): 0.025 g.

Compound 47: Retention Time = 8.43: 1 H NMR (400 MHz, DMSO) δ ppm 8.66 (s, 1 H), 8.22 (s, 1 H), 8.05 (d, J = 2.0 Hz, 1 H), 7.92 (d, J = 9.2 Hz, 1 H), 7.80 (dd, J = 1.6 and 2 Hz, 1 H), 7.68 (d, J = 1.2 Hz, 1 H), 4.79-4.74 (m, 1 H), 3.81-3.75 (m, 1 H), 3.55 (s, 3H), 3.54-3.51 (m, 2H), 3.47-3.41 (m, 1 H), 2.75 (s, 3H), 2.12 (s, 3H), 1.90-1.81 (m, 1 H), 1.59-1.51 (m, 1 H) , M+1 : 403.2

Compound 48: Retention Time = 10.17: 1 H NMR (400 MHz, DMSO) δ ppm 8.67 (s, 1 H), 8.22 (s, 1 H), 8.04 (d, J = 2.8 Hz, 1 H), 7.92 (d, J = 9.6 Hz, 1 H), 7.80 (dd, J = 1.6 and 2 Hz, 1 H), 7.67 (d, J = 1.6 Hz, 1 H), 4.79-4.73 (m, 1 H), 3.80-3.75 (m, 1 H), 3.54 (s, 3H), 3.53-3.51 (m, 2H), 3.46- 3.39 (m, 1 H), 2.75 (s, 3H), 2.1 1 (s, 3H), 1.89-1.81 (m, 1 H), 1.59-1.50 (m, 1 H), M+1 : 403.2

Example 34: 5-(3-(cyclopentylsulfonyl)imidazo[1 ,2-a] pyridi n-6-yl)- 1 ,3-dimethylpyridin-2(1 H)-one (Compound 49)

Step 1: Preparation of 6-bromo-3-(cyclopentylthio)imidazo[1 ,2-a]pyridine

Copper(l) iodide (0.024 g, 0.13 mmol) and 6-bromoimidazo[1 ,2-a]pyridine (0.5 g, 2.53 mmole) were charged to an oven dried vial. The vial was evacuated and back-filled with oxygen. Under a counter-flow of oxygen, cyclopentanethiol (0.42 g, 4.06 mmol) was added, followed by DMSO (3 mL) using a syringe. The vial was placed in a pre-heated oil bath at 120 °C and the reaction mixture was stirred vigorously for 28 h at this temperature under oxygen. After completion, the reaction mixture was allowed to cool to room temperature and was then diluted with water (50 mL) and extracted with ethyl acetate (30 mL x 3). The combined ethyl acetate layer was washed with brine (30 mL), dried over anhydrous Na 2 S0 4 and concentrated under vacuum to afford the title compound (0.3 g, 14%) as a semisolid. This product was used in the next step without further purification. MH + 297.0.

Step 2: Preparation of 6-bromo-3-(cyclopentylsulfonyl)imidazo[1 ,2-a]pyridine

To a suspension of 6-bromo-3-(cyclopentylthio)imidazo[1 ,2-a]pyridine (0.3 g, 1.01 mmol) in DCM (6 mL) was added m-CPBA (0.35 g, 2.02 mmol) and the resulting reaction mixture was stirred at room temperature for 3 h. The reaction mixture was then diluted with DCM (50 mL), washed with water (40 mL X 2) and brine (30 mL), dried over anhydrous Na 2 S0 4 and concentrated under reduced pressure to afford the title compound (0.21 g, 29%) as an oil: MH + : 331.0. The product was used for the next step without further purification.

Step 3: Preparation of Compound 49

A stirred solution of 6-bromo-3(cyclopentylsulfonyl)imidazo[1 ,2-a]pyridine (0.21 g, 0.64 mmol) and 1 ,3-dimethyl-5-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)pyridin-2(1 H)-one (0.21 g, 0.83 mmol) in 1 ,4-dioxane (4 mL) was purged with nitrogen for 10 minutes, followed by the addition of sodium carbonate (0.21 g, 1.92 mmol) in water (0.5 mL). The reaction mixture was again purged with nitrogen for 10 min. Pd(PPh 3 ) 4 (0.037 g, 0.032 mmol) was then added and the reaction mixture was heated at 90°C for 6 h. The solvent was evaporated under reduced pressure and water (80 mL) was added. The product was extracted using ethyl acetate (40 mL x 3). The combined organic layer was washed with brine (50 mL), dried over anhydrous Na 2 S0 4 and concentrated under reduced pressure. The product obtained was purified by silica gel flash chromatography using 1.5 to 2% methanol in DCM as eluent. Fractions were combined and concentrated to give 0.1 1 g of product, which was triturated with a methanol:diethyl ether (0.5 mL : 8 mL) and with diethyl ether (5 mL x 2) to afford Compound 49 (0.032 g, 29%) as a solid. 1 H NMR (400 MHz, DMSO) δ 8.76 (s, 1 H), 8.24 (s, 1 H), 8.08 (d, J = 2 Hz, 1 H), 7.94 (d, J = 9.2 Hz, 1 H), 7.83 (dd, J = 1.2 and 9.6 Hz, 1 H), 7.71 (s, 1 H), 4.10-4.02 (m, 1 H), 3.55 (s, 3H), 2.12 (s, 3H), 1.94-1.87 (m, 4H), 1.67-1.58 (m, 4H).; MH + 372.29.

Example 35: (S)-5-(3-((3-methoxypyrrolidin-1-yl)sulfonyl)imidazo[1 ,2-a]pyridin-6-yl)-1 ,3- dimethylpyridin-2(1 H)-one (Comp

Step 1: Preparation -6-bromo-3-((3-methoxypyrrolidin- 1-yl)sulfonyl)imidazo[ 1,2-a]pyridine

TEA (0.57 mL, 4.0606 mmol) was added to a room temperature stirred solution of (S)-3- methoxy pyrrolidine hydrochloride (0.279 g, 2.0303 mmol) in DCM (5 mL), followed by the addition of a solution of Intermediate 3 (0.4 g, 1.3535 mmol) in DCM (5 mL). The reaction mixture was stirred at rt for 2 h and, after completion, was evaporated under reduced pressure. The product obtained was purified by silica gel chromatography using a gradient of 50-70% ethyl acetate in hexane as eluent. The fractions were combined and concentrated to afford the title compound (0.2 g, 41 %) as a semisolid. MH + 360.14.

Step 2: Prepa

A stirred solution of (S)-6-bromo-3-((3-methoxypyrrolidin-1-yl)sulfonyl)imidazo[1 ,2-a]pyridine (0.2 g, 0.5552 mmol) and 1 ,3-dimethyl-5-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)pyridin- 2(1 H)-one (0.207 g, 0.8328 mmol) in 1 ,4-dioxane (5 mL) was purged with nitrogen for 20 min, followed by the addition of sodium carbonate (0.177 g, 1.6656 mmol) in water (0.5 mL). The reaction mixture was purged again with nitrogen for 20 min. Pd(PPh 3 ) 4 (0.032 g, 0.0278 mmol) was then added and the reaction mixture was heated at 100°C for 6 h. The reaction mixture was concentrated under reduced pressure and the crude product was purified by silica gel chromatography using 2-3% methanol in DCM as eluent. Fractions were combined and concentrated to afford Compound 50 (0.038 g, 17%) as a solid. 1 H NMR (400 MHz, DMSO) δ 8.79 (s, 1 H), 8.20 (s, 1 H), 8.05 (d, J = 2.8 Hz, 1 H), 7.90 (d, J = 9.2 Hz, 1 H), 7.79 (dd, J = 1.6 and 9.6 Hz, 1 H), 7.66 (d, J = 1.2 Hz, 1 H), 3.85-3.84 (m, 1 H), 3.55 (s, 3H), 3.44-3.35 (m, 3H), 3.25-3.21 (m, 1 H), 2.93 (s, 3H), 2.12 (s, 3H), 1.89-1.86 (m, 2H). MH + 403.29.

Example 36: (R)-5-(3-((3-methoxypyrrolidin-1-yl) sulfonyl) imidazo [1 , 2-a] pyridin-6-yl)-1 , 3- dimethylpyridin-2(1 H)-one (Compound 51)

Step 1: Preparation of (R)-6-bromo-3-((3-methoxypyrrolidin- 1-yl)sulfonyl)imidazo[ 1 ,2-a]pyridine

TEA (0.71 ml_, 5.0755 mmol) was added to a room temperature stirred solution of (S)-3- methoxy pyrrolidine hydrochloride (0.349 g, 2.5377 mmol) in DCM (5 ml_), followed by the addition of a solution of Intermediate 3 (0.5 g, 1.6918 mmol) in DCM (5 ml_). The reaction mixture was stirred at rt for 2 h and, after completion, was concentrated under reduced pressure. The product obtained was purified by silica gel chromatography using a gradient of 50-70% ethyl acetate in hexane as eluent. The fractions were combined and concentrated to afford the title compound (0.2 g, 33%) as a semisolid. MH + 360.14

Step 2: Prepa

A stirred solution of (R)-6-bromo-3-((3-methoxypyrrolidin-1-yl)sulfonyl)imidazo[1 ,2-a]pyridine (0.2 g, 0.5552 mmol) and 1 ,3-dimethyl-5-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)pyridin- 2(1 H)-one (0.207 g, 0.8328 mmol) in 1 ,4-dioxane (5 ml_) was purged with nitrogen for 20 min, followed by addition of sodium carbonate (0.177 g, 1.6656 mmol) in water (0.5 ml_). The reaction mixture was again purged with nitrogen for 20 min. Pd(PPh 3 ) 4 (0.032 g, 0.0278 mmol) was then added and the reaction mixture was heated at 100°C for 6 h. The reaction mixture was concentrated under reduced pressure and the crude product was purified by silica gel chromatography using 2-3% methanol in DCM as eluent. Fractions were combined and concentrated to dryness to afford Compound 51 (0.045 g, 20%) as a solid. 1 H NMR (400 MHz, DMSO) δ 8.79 (s, 1 H), 8.20 (s, 1 H), 8.05 (d, J = 2 Hz, 1 H), 7.90 (d, J = 9.6 Hz, 1 H), 7.80-7.78 (m, 1 H), 7.67 (s, 1 H), 3.84-3.83 (m, 1 H), 3.55 (s, 3H), 3.44-3.35 (m, 3H), 3.25-3.19 (m, 1 H), 2.93 (s, 3H), 2.12 (s, 3H), 1.89-1.86 (m, 2H). MH + 403.34.

Example 37: 6-(1 ,5-dimethyl-6-oxo-1 ,6-dihydropyridin-3-yl)-N-(tetrahydrofuran-3-yl) imidazo[1 ,2-a]pyridine-3-sulfonamide (Compounds 52 and 53)

Compound 52 (RT 4.17) - Compound 53 (RT 4.79)

Step 1: Preparation -bromo-N-(tetrahydrofuran-3-yl)imidazo[ 1 ,2-a]pyridine-3-sulfonamide

DIPEA (0.7 mL, 4.06 mmol) was added to a room temperature stirred solution of 3-amino tetrahydrofuran hydrochloride (0.251 g, 2.03 mmol) in DMSO (5 mL), followed by the addition of a solution of Intermediate 3 (0.4 g, 1.35 mmol) in DMSO (5 mL). The reaction mixture was heated at 100°C under microwave irradiation for 40 min. After completion, the reaction mixture was allowed to cool to rt and was diluted with ethyl acetate (250 mL) and washed with water (150 mL X 3). The organic layer was dried over anhydrous Na 2 S0 4 , filtered and concentrated under reduced pressure. The product obtained was purified by silica gel chromatography using a gradient of 70-100% ethyl acetate in hexane as eluent. The fractions were combined and concentrated to afford the title compound (0.15 g, 32%) as a semisolid. M + 348.14.

Step 2: Preparation of Compounds 52 and 53

A stirred solution of 6-bromo-N-(tetrahydrofuran-3-yl)imidazo[1 ,2-a]pyridine-3-sulfonamide (0.15 g, 0.43 mmol) and 1 ,3-dimethyl-5-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)pyridin-2(1 H)-one (0.162 g, 0.65 mmol) in 1 ,4-dioxane (5 mL) was purged with nitrogen for 20 min, followed by the addition of sodium carbonate (0.138 g, 1.30 mmol) in water (0.5 mL). The reaction mixture was again purged with nitrogen for 20 min. Pd(PPh 3 ) (0.025 g, 0.02) was then added and the reaction mixture was heated at 100°C for 6 h. The reaction mixture was concentrated under reduced pressure and the crude product was purified by silica gel chromatography using 2-3% methanol in DCM as eluent. Fractions were combined and concentrated to afford the title compound in racemic form (0.080 g), which was further purified by chiral preparative HPLC (Chiralpak™ AD-H (250*21) mm, 5μ Column, flow 70.0 ml /min, isocratic (A) liquid C0 2 (B) 0.3% DEA in MeOH, (A):(B) = 60:40) to afford two isomers in separate fractions: Fraction 1 (Compound 52): 0.015 g and Fraction 2 (Compound 53): 0.040 g for a total of 33 % yield.

Compound 52: Retention Time = 4.17: 1 H NMR (400 MHz, Methanol-d 4 ) δ ppm 8.79 (s, 1 H), 8.14 (s, 1 H), 8.00 (d, J = 2.4 Hz, 1 H), 7.87-7.81 (m, 3H), 3.92-3.87 (m, 1 H), 3.81 (q, J = 7.6 Hz, 1 H), 3.70 (s, 3H), 3.68-3.65 (m, 2H), 3.43 (dd, J = 3.6 and 9.2 Hz, 1 H), 2.25 (s, 3H), 2.08-1.99 (m, 1 H), 1.67-1.59 (m, 1 H), M+1 : 389.24

Compound 53: Retention Time = 4.79: 1 H NMR (400 MHz, Methanol-d ) δ ppm 8.79 (s, 1 H), 8.14 (s, 1 H), 8.00 (d, J = 2.4 Hz, 1 H), 7.87-7.81 (m, 3H), 3.93-3.87 (m, 1 H), 3.81 (q, J = 7.6 Hz, 1 H), 3.70 (s, 3H), 3.68-3.65 (m, 2H), 3.43 (dd, J = 3.6 and 9.2 Hz, 1 H), 2.25 (s, 3H), 2.08-1.99 (m, 1 H), 1.67-1.59 (m, 1 H), M+1 : 389.24

Example 38: 1 ,3-dimethyl-5-[2-methyl-3-(2-pyridylmethyl)imidazo[1 ,2-a]pyridin-6-yl] pyridin-2- one (Compound 54)

Step 1: Preparation of (6-bromo-2-meth l-imidazo[1 ,2-a]pyridin-3-yl)-(2-pyridyl) methanone

To a suspension of 5-bromopyridin-2-amine (357.0 mg, 2.06 mmol) in MeOH (7 mL) was added Ν,Ν-dimethylacetamide dimethyl acetal (305.36 mg, 2.06 mmol) and the reaction mixture was heated to 80 °C for 18 h. The mixture was cooled to rt and concentrated under reduced pressure. The residue was diluted with EtOAc (10 mL) and then washed with saturated NaHC0 3 (10 mL). The organic layer was dried over MgS0 , filtered and concentrated under reduced pressure. The material was dissolved in toluene (7 mL) and 2-bromo-1-(2-pyridyl)ethanone hydrobromide (644.1 mg, 2.06 mmol) was added followed with DIPEA (0.36 mL, 2.06 mmol) and the reaction mixture was heated in a sealed tube at 140 °C for 30 min. The mixture was cooled to rt, diluted with EtOAc (10 mL) and washed with water (2 x 30 mL). The organic layer was dried over MgS0 4 , filtered and concentrated under reduced pressure. The material was triturated with Et 2 0 and hexane to afford the title compound (1 13.0 mg, 17%) as a solid. 1 H NMR (500 MHz, CDCI 3 ) δ 9.85 - 9.73 (m, 1 H), 8.83 (dd, J = 4.3, 1.6 Hz, 2H), 7.61 (dt, J = 20.0, 5.6 Hz, 2H), 7.47 (dd, J = 4.3, 1.6 Hz, 2H), 2.14 (s, 3H). MS (ESI) [M+H] + 316.1/318.1.

Step 2: Preparation of 1,3-dimethyl-5-[2-methyl-3-(pyridine-2-carbonyl)imidazo[1,2- a] pyridin-6- yl]pyridin-2-one

To a solution of (6-bromo-2-methyl-imidazo[1 ,2-a]pyridin-3-yl)-(2-pyridyl)methanone (327.0 mg, 1.034 mmol) in a mixture of DME (3 mL) and water (0.3 mL) were added 1 ,3-dimethyl-5- (4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)pyridin-2-one (prepared as in US20130053362, 309.2 mg, 1.24 mmol), Cs 2 C0 3 (842.5 mg, 2.59 mmol) and Pd(PPh 3 ) 4 (1 19.9 mg, 0.103 mmol). The reaction mixture was degassed with N 2 for 5 min and then heated to 100 °C for 18 h. The mixture was cooled to rt and diluted with EtOAc (10 mL) and water (10 mL). The phases were separated and the organic layer was dried over MgS0 4 , filtered and concentrated under reduced pressure. The material was purified by flash chromatography on silica gel using a gradient (0-20%) of MeOH in DCM as eluent to afford the title compound (213 mg, 57%) as a solid. 1 H NMR (500 MHz, CDCI 3 ) δ 9.66 (dd, J = 1.9, 0.9 Hz, 1 H), 8.72 (ddd, J = 4.8, 1.7, 1.0 Hz, 1 H), 7.94 (td, J = 7.7, 1.7 Hz, 1 H), 7.81 (dt, J = 7.8, 1.1 Hz, 1 H), 7.69 (dd, J = 9.2, 0.9 Hz, 1 H), 7.59 - 7.42 (m, 4H), 3.64 (s, 3H), 2.24 (s, 3H), 2.15 (s, 3H). MS (ESI) [M+H] + 359.3.

Step 3: Preparation of Compound 54

A solution of 1 ,3-dimethyl-5-[2-methyl-3-(pyridine-2-carbonyl)imidazo[1 , 2- a] py r i d i n-6-y I ] py ri d i n- 2-one (213.0 mg, 0.59 mmol), hydrazine hydrate (1.72 mL, 19.39 mmol) and KOH (266.7 mg, 4.75 mmol) in ethylene glycol (4 mL) was heated to 150 °C in a sealed vial for 20 h. The solution was cooled to rt and diluted with DCM (10 mL) and H 2 0 (10 mL). The aqueous layer was extracted with DCM (3 x 10 mL). The combined organic layers were dried over MgS0 4 , filtered and concentrated under reduced pressure. The material was purified by preparative HPLC to afford Compound 54 (55 mg, 27 %) as a solid. 1 H NMR (500 MHz, CDCI 3 ) δ 8.56 (d, J = 4.4 Hz, 1 H), 8.10 (s, 1 H), 7.56 (m, 2H), 7.36 (s, 1 H), 7.28 (d, J = 2.2, Hz, 1 H), 7.19 - 7.15 (m, 2H), 7.00 (d, J = 7.8 Hz, 1 H), 4.43 (s, 2H), 3.75 (s, 3H), 3.61 (s, 3H), 2.55 (s, 3H). MS (ESI) [M+H] + 345.2.

Example 39: 1 ,3-dimethyl-5-[2-methyl-3-(3-pyridylmethyl)imidazo[1 ,2-a]pyridin-6-yl] pyridin-2- one (Compound 55)

Step 1: Preparation of (6-bromo-2-meth l-imidazo[1 ,2-a]pyridin-3-yl)-(3-pyridyl) methanone

To a suspension of 5-bromopyridin-2-amine (362 mg, 2.092 mmol) in MeOH (5 mL) was added Ν,Ν-dimethylacetamide dimethyl acetal (309.6 mg, 2.09 mmol) and the reaction mixture was heated to 80 °C for 18 h. The mixture was cooled to rt and concentrated under reduced pressure. The residue was diluted with EtOAc (10 mL) and the organic layer was washed with saturated NaHC0 3 (10 mL). The organic layer was dried over MgS0 4 , filtered and concentrated under reduced pressure. The residue (500 mg, 2.07 mmol) was dissolved in toluene (10 mL) and 2-bromo-1-(3-pyridyl)ethanone hydrobromide (580.2 mg, 2.07 mmol) was added, followed by DIPEA (0.37 mL, 2.07 mmol). The reaction mixture was heated at 140 °C for 10 min and then was cooled to rt in the oil bath. The mixture was then concentrated under reduced pressure and the residue was dissolved in EtOAc (20 mL) with a minimum of MeOH. The organic layer was washed with saturated NaHC0 3 (3 x 20 mL). The combined organic layers were dried over Na 2 S0 , filtered and concentrated under reduced pressure. The material was purified by flash chromatography on silica gel using a mixture of MeOH in DCM as eluent to afford the title compound (138 mg, 21 %) as a solid. 1 H NMR (500 MHz, CDCI 3 ) δ 9.73 (dd, J = 1.7, 1.0 Hz, 1 H), 8.90 - 8.79 (m, 2H), 8.02 - 7.93 (m, 1 H), 7.63 - 7.54 (m, 2H), 7.49 (ddd, J = 7.8, 4.9, 0.8 Hz, 1 H), 2.20 (s, 3H). MS (ESI) [M+H] + 316.1/318.1.

Step 2: Preparation of 1,3-dimethyl-5-[2-methyl-3-(pyridine-3-carbonyl)imidazo[1,2- a] pyridin-6- yl]pyridin-2-one

To a solution of (6-bromo-2-methyl-irnidazo[1 ,2-a]pyridin-3-yl)-(3-pyridyl)methanone (138 mg, 0.436 mmol) in a mixture of DME (5 mL) and water (0.5 mL) were added 1 ,3-dimethyl-5- (4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)pyridin-2-one (prepared as in US20130053362, 130.5 mg, 0.524 mmol), Cs 2 C0 3 (355 mg, 1.09 mmol) and Pd(PPh 3 ) 4 (50.62 mg, 0.044 mmol). The reaction mixture was degassed with N 2 for 5 min and then heated in a sealed tube at 100 °C for 18 h. The mixture was cooled to rt, and then diluted with EtOAc (10 mL) and water (10 mL). The organic layer was dried over MgS0 4 , filtered and concentrated under reduced pressure. The material was triturated with Et 2 0 to afford the title compound (129 mg, 83%), which was used in the next step without further purification. 1 H NMR (500 MHz, CDCI 3 ) δ 9.67 (dd, J = 1.9, 0.9 Hz, 1 H), 8.87 (ddd, J = 6.6, 3.5, 1.2 Hz, 2H), 8.05 - 7.98 (m, 1 H), 7.76 - 7.69 (m, 1 H), 7.61 (dd, J = 9.2, 1.9 Hz, 1 H), 7.54 - 7.45 (m, 3H), 3.66 (s, 3H), 2.25 (d, J = 14.2 Hz, 6H). MS (ESI) [M+H] + 359.3.

Step 3: Preparation of Compound 55

A solution of 1 ,3-dimethyl-5-[2-methyl-3-(pyridine-3-carbonyl)imidazo[1 , 2- a] py r i d i n-6-y I ] py ri d i n- 2-one (129.0 mg, 0.36 mmol), hydrazine hydrate (1.04 mL, 11.75 mmol) and KOH (161.5 mg, 2.88 mmol) in ethylene glycol (1 mL) was heated to 150 °C in a sealed vial for 20 h. The solution was cooled to rt and diluted with DCM (10 mL) and water (10 mL). The phases were separated and the aqueous layer was extracted with DCM (3 x 10 mL). The combined organic layers were dried over MgS0 , filtered and concentrated under reduced pressure. The material was purified by preparative HPLC to afford Compound 55 (45 mg, 36%) as a solid. 1 H NMR (500 MHz, CDCI 3 ) δ 8.55 - 8.46 (m, 2H), 7.64 - 7.54 (m, 2H), 7.35 (d, J = 7.9 Hz, 1 H), 7.29 (m, 1 H), 7.25 - 7.14 (m, 3H), 4.29 (s, 2H), 3.59 (s, 3H), 2.52 (s, 3H), 2.20 (s, 3H). MS (ESI) [M+H] + 345.3. Example 40: 1 ,3-dimethyl-5-[2-methyl-3-(4-pyridylmethyl)imidazo[1 ,2-a]pyridin-6-yl] pyridin-2- one (Compound 56)

Step 1: Preparation of (6-bromo-2-meth l-imidazo[1 ,2-a]pyridin-3-yl)-(4-pyridyl) methanone

To a suspension of 5-bromopyridin-2-amine (357 mg, 2.06 mmol) in MeOH (7 mL) was added Ν,Ν-dimethylacetamide dimethyl acetal (305.4 mg, 2.06 mmol) and the reaction mixture was heated to 80 °C for 18 h. The mixture was cooled to rt and concentrated under reduced pressure. The residue was diluted in EtOAc (10 ml) and the organic layer was washed with saturated NaHC0 3 . The organic layer was dried over MgS0 4 , filtered and concentrated under reduced pressure. The residue was diluted in toluene (7 mL) and 2-bromo-1-(4-pyridyl)ethanone hydrobromide (644.1 mg, 2.06 mmol) was added followed by DIPEA (0.36 mL, 2.06 mmol) and the reaction mixture was heated in a sealed tube at 140 °C for 30 min. The mixture was cooled to rt, diluted with EtOAc (10 mL) and the organic layer was washed with water (2 x 30 mL). The organic layer was dried over MgS0 , filtered and concentrated under reduced pressure. The material was triturated with Et 2 0 and hexane to afford the title compound (113.0 mg, 17%) as a solid. 1 H NMR (500 MHz, CDCI 3 ) δ 9.85 - 9.73 (m, 1 H), 8.83 (dd, J = 4.3, 1.6 Hz, 2H), 7.61 (dt, J = 20.0, 5.6 Hz, 2H), 7.47 (dd, J = 4.3, 1.6 Hz, 2H), 2.14 (s, 3H). MS (ESI) [M+H]+ 316.1/318.1.

Step 2: Preparation of 1,3-dimethyl-5-[2-methyl-3-(pyridine-4-carbonyl)imidazo[1,2- a] pyridin-6- yl]pyridin-2-one

To a solution of (6-bromo-2-methyl-imidazo[1 ,2-a]pyridin-3-yl)-(4-pyridyl)methanone (150 mg, 0.474 mmol) in a mixture of DME (5 mL) and water (0.5 mL) were added 1 ,3-dimethyl-5- (4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)pyridin-2-one (prepared as in US20130053362, 141.8 mg, 0.569 mmol), Cs 2 C0 3 (386.5 mg, 1.19 mmol) and Pd(PPh 3 ) 4 (54.8 mg, 0.047 mmol). The reaction mixture was degassed with N 2 for 5 min and then heated in a sealed tube at 100 °C for 18 h. The mixture was cooled to rt and diluted with EtOAc (10 ml_) and water (10 ml_). The organic layer was separated, dried over MgS0 4 , filtered and concentrated under reduced pressure. The material was triturated with a mixture of EtOAc and hexane to afford the title compound (166 mg, 98%) as a solid. 1 H NMR (500 MHz, CDCI 3 ) δ 9.74 (dd, J = 1.8, 0.8 Hz, 1 H), 8.86 (d, J = 4.8 Hz, 2H), 7.75 (dd, J = 9.2, 0.8 Hz, 1 H), 7.65 (dd, J = 9.2, 1.9 Hz, 1 H), 7.56 - 7.48 (m, 4H), 3.68 (s, 3H), 3.51 (d, J = 5.5 Hz, 2H), 2.28 (s, 3H), 2.19 (s, 3H). MS (ESI) [M+H] + 359.3.

Step 3: Preparation of Compound 56

A solution of 1 ,3-dimethyl-5-[2-methyl-3-(pyridine-4-carbonyl)imidazo[1 ,2-a]pyridin-6-yl]pyridin- 2-one (166.0 mg, 0.46 mmol), hydrazine hydrate (1.34 ml, 15.12 mmol) and KOH (207.9 mg, 3.7 mmol) in ethylene glycol (3 ml_) was heated to 150 °C in a sealed vial for 20 h. The solution was cooled to rt and diluted with DCM (10 ml_) and water (10 ml_). The aqueous layer was extracted with DCM (3 x 10 ml_). The combined organic layers were dried over Na 2 S0 4 , filtered and concentrated under reduced pressure. The material was purified by preparative HPLC to afford Compound 56 (52.0 mg, 33%) as a solid. 1 H NMR (500 MHz, CDCI 3 ) δ 8.46 (d, J = 5.4 Hz, 2H), 7.55 (d, J = 9.3 Hz, 1 H), 7.48 (s, 1 H), 7.20 - 7.11 (m, 3H), 6.97 (d, J = 5.8 Hz, 2H), 4.22 (s, 2H), 3.52 (s, 3H), 2.44 (s, 3H), 2.13 (s, 3H). MS (ESI) [M+H] + 345.3.

Example 41 : Biological activity

a) In vitro bromodomain inhibition

To measure activity of bromodomain inhibitors, a His-epitope tagged BRD4 BD149-170 is purchased from BPS Bioscience. BRD4 binding and inhibition is assessed by monitoring the engagement of biotinylated H4-tetraacetyl peptide (H4K5/8/12/16; AnaSpec #64989-025) with the target using the AlphaLISA technology (Perkin-Elmer). Specifically, in a 384 well OptiPIate, BRD4(BD1 ) (200 nM final) is pre- incubated with either DMSO (final 1.0% DMSO) or a compound dilution series in DMSO. All reagents are diluted in assay buffer containing 50 mM HEPES (pH 7.4), 100 mM NaCI, 0.1 % (w/v) BSA, and 0.05% (w/v) CHAPS. After a 30 minute incubation at room temperature, H4 peptide is added (200 nM final) and the reaction is incubated an additional 30 minutes at room temperature. Alpha streptavidin donor beads and AlphaLISA nickel chelate acceptor beads are then added to a final concentration of 10 Mg/mL each. After one hour, equilibration plates are read on an Envision instrument and IC 50 s calculated using a four parameter non-linear curve fit. The results are shown in Table 1 (BRD4 alpha).

b) Transcription of human C-Myc in human leukemic MV-4- 11 cells:

The effect of compounds on transcription of human C-Myc gene is monitored in human B- myelomonocytic leukemia cell line MV-4-1 1 (from American Type Culture Collection (ATCC), Manassas, Virginia, USA) using QuantiGene 2.0 assay kit (Affymetrix, Santa Clara, California, USA).

Typically, 8,000 MV-4-11 cells are plated in sterile 96-well plates (Costar #3598 from Fisher Scientific Canada, Ottawa, Ontario, Canada) in Iscove's medium supplemented with 10% fetal bovine serum, glutamine (2 mM), and penicillin (100 I.U.) and streptomycin (100 μg/mL) (all from Wisent Inc., St. Bruno, Quebec, Canada). Compounds are dissolved in DMSO at 30 mM. A series of 1 :3 dilutions are first made in DMSO, and further 1 : 100 dilutions are made in serum- containing cell culture media. The final concentration of DMSO is 0.1 % in cell culture media. After cells are treated with various concentrations of test compound for 4 hours, cells are lysed using Quantigene 2.0 sample processing kit (#QS0100). C-Myc mRNA is detected using a QuantiGene 2.0 assay kit (#QS0009) with gene-specific probe to human C-Myc (#SA-50182) following the manufacturer's recommendations. Luminescene signals are read on Flexstation II microplate reader (Molecular Devices, Sunnyvale, California, USA). Percentage of inhibition of C-Myc transcription is analyzed using EXCEL (2010 version). The results are shown in Table 1 (MV-4-1 1 ).

Table 1.

Compound No BRD4a (ICso, μΜ) MV-4-11 (ICso, μΜ)

1 0.122 0.058

2 0.09946 0.0592

3 0.1432 0.0863

4 0.3125 0.2758

5 0.1 13 0.0939

6 N/A 0.0913

7 N/A 0.0928

8 0.195 0.8444

9 0.1531 0.1523

10 0.3177 0.8373

1 1 N/A 2.7293 Compound No BRD4a (ICso, μΜ) MV-4-11 (ICso, μΜ)

12 N/A 0.912

13 N/A 2.6578

14 N/A 0.508

15 N/A 2.5243

16 N/A 0.0813

17 N/A 0.7583

18 N/A 0.0937

19 N/A 0.1699

20 N/A 0.11 18

21 N/A 0.8159

22 N/A 0.0098

23 N/A 0.0165

24 N/A 0.0322

25 N/A 0.1742

26 N/A 0.4919

30 N/A 0.0249

34 N/A 0.2344

42 N/A 0.8647

43 N/A 0.2534

44 N/A 0.5967

45 N/A 0.5071

46 N/A 0.1776

47 N/A 0.731

48 N/A 0.5367

49 N/A 0.8514

50 N/A 2.3697

51 N/A 0.6147

52 N/A 0.8906

53 N/A 0.8983

54 N/A 0.1508

55 N/A 0.0845

56 N/A 0.1533

N/A: not available

Although the invention has been illustrated and described with respect to one or more implementations, equivalent alterations and modifications will occur to others skilled in the art upon the reading and understanding of this specification. In addition, while a particular feature of the invention may have been disclosed with respect to only one of several implementations, such feature may be combined with one or more other features of the other implementations as may be desired and advantageous for any given or particular application. Accordingly, it is understood that the examples and embodiments described herein are for illustrative purposes only and that various modifications or changes in light thereof will be suggested to persons skilled in the art and are to be included within the spirit and purview of this application and scope of the appended claims. Any publication, document, patent, patent application or publication referred to herein should be construed as incorporated by reference each in their entirety for all purposes.