Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
SHP2 INHIBITORS AND USES THEREOF
Document Type and Number:
WIPO Patent Application WO/2019/182960
Kind Code:
A1
Abstract:
Compounds of Formula (1) as inhibitors of protein tyrosine phosphatase SHP2 are disclosed. The pharmaceutical compositions comprising compounds of Formula 1, methods of synthesis of these compounds, methods of treatment for diseases associated with the aberrant activity of SHP2 such as cancer using these compounds or compositions containing these compounds are also disclosed.

Inventors:
XIE YINONG (US)
BABISS LEE E (US)
Application Number:
PCT/US2019/022717
Publication Date:
September 26, 2019
Filing Date:
March 18, 2019
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
SYNBLIA THERAPEUTICS INC (US)
International Classes:
C07D401/04; A61K31/4412; A61K31/496; A61K31/499; A61K31/506; A61K31/519; A61P19/02; A61P35/00; C07D213/16; C07D401/12; C07D401/14; C07D403/04; C07D471/04; C07D487/04; C07D491/107; C07D513/04; C07D519/00
Domestic Patent References:
WO2018031658A12018-02-15
WO2018013597A12018-01-18
WO2017156397A12017-09-14
WO2016203405A12016-12-22
WO2016203404A12016-12-22
WO2015175707A12015-11-19
WO2015168466A12015-11-05
WO2015036560A12015-03-19
WO2011022440A22011-02-24
WO2008107398A22008-09-12
WO2007149448A22007-12-27
WO2006060318A22006-06-08
WO2016203405A12016-12-22
Foreign References:
US20050182072A12005-08-18
Other References:
SNELL B.K.: "Pyrimidines . Part II. The Photo-Fries Rearrangement of 2-Dialkyl- amino-4-pyrimidinyl Esters of Alkyl and Arylsulphonic Acids", JOURNAL OF THE CHEMICAL SOCIETY C: ORGANIC, 1968, pages 2367 - 2370, XP055638701
PHILIP J. KOCIENSKI: "Protecting Groups", 1994, GEORG THIEME VERLAG
THEODORA W. GREENEPETER G. M. WUTS: "Protective Groups in Organic Synthesis", 1999, WILEY INTERSCIENCE
Attorney, Agent or Firm:
JOHNSON, Brent A. et al. (US)
Download PDF:
Claims:
CLAIMS

What Is Claimed Is:

1. A compound represented by a formula:

or a pharmaceutically acceptable salt thereof;

wherein X is S, O, N RA, CH RA, SO, S02, CO, or a bond;

Ring A is an optionally substituted aryl, heteroaryl, or bicyclic ring system;

Ring B is an optionally substituted heterocyclic ring system, comprising a mono-cyclic ring, a bicyclic ring system, a tricyclic ring system, or a tetracyclic ring system, wherein the heterocyclic ring system contains heteroaryl and at least 2 ring nitrogen atoms; and

RA is H or Ci-6 hydrocarbyl.

2. The compound of claim 1, wherein Ring A is optionally substituted phenyl.

3. The compound of claim 1, wherein Ring A is optionally substituted naphthalen-l-yl.

4. The compound of claim 1, wherein Ring A is optionally substituted pyridin-3-yl.

5. The compound of claim 1, wherein Ring A is optionally substituted pyridin-4-yl.

6. The compound of claim 1, wherein Ring A is optional ly substituted 2-oxo-l,2-dihydropyridin-4- yi-

7. The compound of claim 1, wherein Ring A is optionally substituted lH-indol-4-yl.

8. The compound of claim 1, wherein Ring A is optionally substituted 2-oxoindolin-4-yl.

9. The compound of claim 1, wherein Ring A is optionally substituted indolin-4-yl.

10. The compound of claim 1, wherein Ring A is optionally substituted 3-(2-oxo-2,5-dihydro-lH- pyrrole-3-carboxamido) phenyl.

11. The compound of claim 1, wherein Ring A is optionally substituted 3-(4-oxo-4H-pyrido[l,2- oc] pyrimidine-3-carboxamido)phenyl.

12. The compound of claim 1, wherein Ring A is optionally substituted 3-(4-oxo-4H-pyrazino[l,2- oc] pyrimidine-3-carboxamido)phenyl.

13. The compound of claim 1, wherein Ring A is optionally substituted 3-(5-oxo-5H-thiazolo[3,2- oc] pyrimidine-6-carboxamido)phenyl.

14. The compound of claim 1, wherein Ring A is optionally substituted 3-(5-oxo-l,5- dihydroimidazo[l,2-oc] pyrimidine-6-carboxamido)phenyl.

15. The compound of claim 1, wherein Ring A is optionally substituted 3-(4-oxo-6,7,8,9-tetrahydro- 4H-pyrido[l,2-oc] pyrimidine-3-carboxamido)phenyl.

16. The compound of claim 1, wherein Ring A is optionally substituted 2,3-dichlorophenyl.

17. The compound of claim 1, wherein Ring A is optionally substituted 2,3-dichloro-pyridin-4-yl.

18. The compound of claim 1, wherein Ring A is optionally substituted 2-amino-3-chloropyridin-4-yl

19. The compound of claim 1, 2, 3, 4, 5, 6, 7 , 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, or 18, wherein Ring

A is unsubstituted.

20. The compound of claim 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, or 18, wherein Ring

A has a Cl substituent.

21. The compound of claim 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, or 18, wherein Ring A has two Cl substituents.

22. The compound of claim 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, or 18, wherein Ring A has a CF3 substituent.

23. The compound of claim 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, or 18, wherein Ring A has an N H2 substituent.

24. The compound of claim 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, or 18, wherein Ring A has a -OCH3 substituent.

25. The compound of claim 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, or 18, wherein Ring

A has an F substituent.

26. The compound of claim 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, or 18, wherein Ring A has an acetyl substituent.

27. The compound of claim 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, or 18, wherein Ring A has a CFI3 substituent.

28. The compound of claim 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, or 18, wherein Ring

A has an OFI substituent.

29. The compound of claim 1, 2, 3, 4, 5, 6, 7 , 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, or 18, wherein Ring A has multiple substituents with any combination of the substituent of claim 20, 21, 22, 23, 24, 25, 26, 27, or 28, at any positions that are chemically permissible.

30. The compound of claim 1, wherein Ring A is any one of the following: phenyl, 2,3-dichlorophenyl, naphthalen-l-yl, 2-(trifluoromethyl)phenyl, 2-(trifluoromethyl)pyridin-3-yl, 5-amino-2-chlorophenyl,

5-amino-2-chloropyridin-3-yl, 3-amino-2-chlorophenyl, 2-amino-3-chloropyridin-4-yl, 2-chloro-3- methoxy phenyl, 3-chloro-2-methoxypyridin-4-yl, 3-fluoro-lH-indol-4-yl, 3,3-difluoro-2-oxoindolin-4- yl, l-acetyl-3,3-difluoroindolin-4-yl, 2-chloro-3-(4-hydroxy-l,5,5-trimethyl-2-oxo-2,5-dihydro-lH- pyrrole-3-carboxamido) phenyl, 2-chloro-3-(2-hydroxy-4-oxo-4H-pyrido[l,2-a]pyrimidine-3- carboxamido)phenyl, 2-chloro-3-(2-hydroxy-4-oxo-4H-pyrazino[l,2-a] pyrimidine-3- carboxamido)phenyl, 2-chloro-3-(7-hydroxy-5-oxo-5H-thiazolo[3,2-a] pyrimidine-6- carboxamido)phenyl, 2-chloro-3-(7-hydroxy-5-oxo-l,5-dihydroimidazo[l,2-a] pyrimidine-6- carboxamido)phenyl, 2-chloro-3-(2-hydroxy-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[l,2-a] pyrimidine-3- carboxamido)phenyl, 2,3-dichloropyridin-4-yl, 2,3-difluorophenyl, 3-chloro-2-fluoropyridin-4-yl, 2,3- difluoropyridin-4-yl, 2-chloro-3-methylphenyl, 3-chloro-2-methylpyridin-4-yl, 3,3-difluoro-l-methyl-

2-oxoindolin-4-yl, 3-chloro-l-methyl-2-oxo-l,2-dihydropyridin-4-yl, or 2-chloro-3-fluorophenyl.

31. The compound of claim 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30, wherein Ring B is optionally substituted 6-oxo-5-(piperidin-l-yl)-l,6- dihydropyrazin-2-yl.

32. The compound of claim 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23,

24, 25, 26, 27, 28, 29, or 30, wherein Ring B is optionally substituted 6-oxo-5-(pyrrolidin-l-yl)-l,6- dihydropyrazin-2-yl.

33. The compound of claim 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30, wherein Ring B is optionally substituted 5-(hexahydropyrrolo[3,4- c] pyrrol-2(lH)-yl)-6-oxo-l,6-dihydropyrazin-2-yl.

34. The compound of claim 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30, wherein Ring B is optionally substituted 5-(3,6-diazabicyclo[3.2.0] heptan-

6-yl)-6-oxo-l,6-dihydropyrazin-2-yl.

35. The compound of claim 1, 2, 3, 4, 5, 6, 1, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30, wherein Ring B is optionally substituted 6-oxo-5-(2-oxa-8- azaspiro[4.5]decan-8-yl)-l,6-dihydropyrazin-2-yl.

36. The compound of claim 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30, wherein Ring B is optionally substituted 6-oxo-5-(piperidin-4-ylamino)- l,6-dihydropyrazin-2-yl.

37. The compound of claim 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30, wherein Ring B is optionally substituted 6-oxo-5- (spiro[bicyclo[3.1.0] hexane-3,4'-piperidin]-l'-yl)-l,6-dihydropyrazin-2-yl.

38. The compound of claim 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23,

24, 25, 26, 27, 28, 29, or 30, wherein Ring B is optionally substituted 6-oxo-5-(8-azaspiro[4.5]decan- 8-yl)-l,6-dihydropyrazin-2-yl.

39. The compound of claim 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30, wherein Ring B is optionally substituted 4-oxo-6-(piperidin-l-yl)-4,5- dihydro-lH-pyrazolo[3,4-d] pyrimidin-3-yl.

40. The compound of claim 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30, wherein Ring B is optionally substituted 4-oxo-6-(pyrrolidin-l-yl)-4,5- dihydro-lH-pyrazolo[3,4-d] pyrimidin-3-yl.

41. The compound of claim 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30, wherein Ring B is optionally substituted 4-oxo-2-(piperidin-l-yl)-3,4- dihydroquinazolin-5-yl.

42. The compound of claim 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30, wherein Ring B is optionally substituted 4-oxo-2-(piperidin-l-yl)-3,4- dihydropyrido[3,4-d] pyrimidin-5-yl.

43. The compound of claim 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23,

24, 25, 26, 27, 28, 29, or 30, wherein Ring B is optionally substituted 7-oxo-2-(piperidin-l-yl)-7,8- dihyd ropy rido[2, 3-d] pyrimidin-5-yl.

44. The compound of claim 1, 2, 3, 4, 5, 6, 1, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30, wherein Ring B is optionally substituted 5-(piperidin-l-yl)-lH- pyrazolo[4,3-d]thiazol-3-yl.

45. The compound of claim 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30, wherein Ring B is optionally substituted 7-oxo-6-(piperidin-4-yl)-6,7- dihydro-lH-pyrazolo[4,3-d] pyrimidin-3-yl.

46. The compound of claim 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23,

24, 25, 26, 27, 28, 29, or 30, wherein Ring B is optionally substituted 6-oxo-8-(piperidin-l-yl)-6,7- dihydro-lH-purin-2-yl.

47. The compound of claim 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23,

24, 25, 26, 27, 28, 29, or 30, wherein Ring B is optionally substituted 8-(piperidin-l-yl)-7H-pu rin-2-y I. 48. The compound of claim 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30, wherein Ring B is optionally substituted 6-oxo-2-(pyrrolidin-l-yl)-l,6- dihydropyrimidin-5-yl.

49. The compound of claim 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23,

24, 25, 26, 27, 28, 29, or 30, wherein Ring B is optionally substituted 6-oxo-2-(piperidin-l-yl)-l,6- dihydropyrimidin-5-yl.

50. The compound of claim 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30, wherein Ring B is optionally substituted 6-oxo-2-(2-oxa-8- azaspiro[4.5]decan-8-yl)-l,6-dihydropyrimidin-5-yl.

51. The compound of claim 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30, wherein Ring B is optionally substituted 2-(3,6-diazabicyclo[3.2.0] heptan- 6-yl)-6-oxo-l,6-dihydropyrimidin-5-yl.

52. The compound of claim 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30, wherein Ring B is optionally substituted 2-(hexahydropyrrolo[3,4- c] pyrrol-2(lH)-yl)-6-oxo-l,6-dihydropyrimidin-5-yl.

53. The compound of claim 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30, wherein Ring B is optionally substituted 2-(3-azabicyclo[3.2.0] heptan-3- yl)-6-oxo-l,6-dihydropyrimidin-5-yl.

54. The compound of claim 1, 2, 3, 4, 5, 6, 1, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30, wherein Ring B is optionally substituted 6-oxo-2-(2-azaspiro[3.4]octan- 2-yl)-l,6-dihydropyrimidin-5-yl.

55. The compound of claim 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30, wherein Ring B is optionally substituted 2-(3-azabicyclo[3.1.0] hexan-3- yl)-6-oxo-l,6-dihydropyrimidin-5-yl.

56. The compound of claim 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23,

24, 25, 26, 27, 28, 29, or 30, wherein Ring B is optionally substituted 6-oxo-2-(2-azaspiro[3.4]octan-

2-yl)-l,6-dihydropyrimidin-5-yl.

57. The compound of claim 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23,

24, 25, 26, 27, 28, 29, or 30, wherein Ring B is optionally substituted 5-oxo-6-(2-oxa-8- azaspiro[4.5]decan-8-yl)-4,5-dihydro-lH-pyrazolo[3,4-b]pyrazin-3-yl.

58. The compound of claim 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23,

24, 25, 26, 27, 28, 29, or 30, wherein Ring B is optionally substituted 5-oxo-6-(piperidin-l-yl)-4,5- dihydro-lH-pyrazolo[3,4-b] pyrazin-3-yl.

59. The compound of claim 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30, wherein Ring B is optionally substituted 6-(piperidin-l-yl)-lH- pyrazolo[3,4-d]pyrimidin-3-yl.

60. The compound of claim 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30, wherein Ring B is optionally substituted 6-oxo-5-(2-azaspiro[3.4]octan-

2-yl)-l,6-dihydropyrazin-2-yl.

61. The compound of claim 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30, wherein Ring B is optionally substituted l-cyclohexyl-2-oxo-l,2- dihydropyridin-4-yl.

62. The compound of claim 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23,

24, 25, 26, 27, 28, 29, or 30, wherein Ring B is optionally substituted 5-(3-azabicyclo[3.1.0] hexan-3- yl)-6-oxo-l,6-dihydropyrazin-2-yl.

63. The compound of claim 1, 2, 3, 4, 5, 6, 1, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30, wherein Ring B is optionally substituted 6-oxo-5-(3H-spiro[benzofuran- 2,4'-piperidin]-l'-yl)-l,6-dihydropy razin-2-y I.

64. The compound of claim 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30, wherein Ring B is optionally substituted 4-oxo-2-(2-oxa-8- azaspiro[4.5]decan-8-yl)-4,7-dihydro-3H-pyrrolo[2,3-d] pyrimidin-5-yl.

65. The compound of claim 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30, wherein Ring B is optionally substituted 5-(5,7- dihydrospiro[cyclopenta[b] pyridine-6,4'-piperidin]-l'-yl)-6-oxo-l,6-dihydropyrazin-2-yl.

66. The compound of claim 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23,

24, 25, 26, 27, 28, 29, or 30, wherein Ring B is optionally substituted 5-(l,3-dihydrospiro[indene- 2,4'-piperidin]-l'-yl)-6-oxo-l,6-dihydropyrazin-2-yl.

67. The compound of claim 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30, wherein Ring B is optionally substituted 5-(4,6- dihydrospiro[cyclopenta[d]thiazole-5,4'-piperidin]-l'-yl)-6-oxo-l,6-dihydropyrazin-2-yl.

68. The compound of claim 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30, wherein Ring B is optionally substituted 6-oxo-5-(spiro[indoline-2,4'- piperidin]-l'-y l)-l,6-dihyd ro py razin-2-yl.

69. The compound of claim 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30, wherein Ring B is optionally substituted 5-((3S,4S)-4-amino-3-methyl-2- oxa-8-azaspiro[4.5]decan-8-yl)-6-oxo-l,6-dihydropyrazin-2-yl.

70. The compound of claim 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23,

24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50,

51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, or 69, wherein Ring B is unsubstituted.

71. The compound of claim 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23,

24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50,

51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, or 69, wherein Ring B has a -CH3 substituent.

72. The compound of claim 1, 2, 3, 4, 5, 6, 1, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, or 69, wherein Ring B has an - CH2NH2 substituent.

73. The compound of claim 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23,

24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50,

51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, or 69, wherein Ring B has an -N H2 substituent.

74. The compound of claim 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50,

51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, or 69, wherein Ring B has a - CH2CH2NH2 substituent.

75. The compound of claim 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23,

24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, or 69, wherein Ring B has a 1- aminopropan-2-yl substituent.

76. The compound of claim 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23,

24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50,

51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, or 69, wherein Ring B has a -CN substituent.

77. The compound of claim 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23,

24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50,

51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, or 69, wherein Ring B has an -F substituent.

78. The compound of claim 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23,

24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50,

51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, or 69, wherein Ring B has a -Cl substituent.

79. The compound of claim 1, 2, 3, 4, 5, 6, 1, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, or 69, wherein Ring B has a -CH2F substituent.

80. The compound of claim 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23,

24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, or 69, wherein Ring B has an -OH substituent.

81. The compound of claim 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50,

51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, or 69, wherein Ring B has an -OCH3 substituent.

82. The compound of claim 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22,

23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, or 69, wherein Ring B has multiple substituents with any combination of the substituent of claim 72, 73, 74, 75, 76, 77, 78, 79,

80, or 81, at any positions that are chemically permissible.

83. The compound of claim 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23,

24, 25, 26, 27, 28, 29, or 30, wherein Ring B is any one of the following: 5-(4-(aminomethyl)-4- methylpiperidin-l-yl)-6-oxo-l,6-dihydropyrazin-2-yl, 5-(3-(aminomethyl)-3-methylpyrrolidin-l-yl)-6- oxo-l,6-dihydropyrazin-2-yl, 5-(hexahydropyrrolo[3,4-c] pyrrol-2(lH)-yl)-6-oxo-l,6-dihydropyrazin- 2-yl, 5-(3,6-diazabicyclo[3.2.0]heptan-6-yl)-6-oxo-l,6-dihydropyrazin-2-yl, (S)-5-(4-amino-2-oxa-8- azaspiro[4.5]decan-8-yl)-6-oxo-l,6-dihydropyrazin-2-yl, 6-oxo-5-(piperidin-4-ylamino)-l,6- dihydropyrazin-2-yl, 5-(2-a minospiro[bicyclo[3.1.0] hexane-3, 4'-piperidin]-l'-yl)-6-oxo-l,6- dihydropyrazin-2-yl, 5-((lR,3R)-l-amino-3-methyl-8-azaspiro[4.5]decan-8-yl)-6-oxo-l,6- dihydropyrazin-2-yl, (6-(4-(aminomethyl)-4-methylpiperidin-l-yl)-4-oxo-4,5-dihydro-lH- pyrazolo[3,4-d]pyrimidin-3-yl)amino, 6-(3-(aminomethyl)-3-methylpyrrolidin-l-yl)-4-oxo-4,5- dihydro-lH-pyrazolo[3,4-d] pyrimidin-3-yl, 7-amino-2-(4-(aminomethyl)-4-methylpiperidin-l-yl)-4- oxo-3,4-dihydroquinazolin-5-yl, 2-(4-(aminomethyl)-4-methylpiperidin-l-yl)-4-oxo-3,4- dihydropyrido[3,4-d] pyrimidin-5-yl, 2-(4-(aminomethyl)-4-methylpiperidin-l-yl)-7-oxo-7,8- dihydropyrido[2,3-d] pyrimidin-5-yl, 5-(4-(aminomethyl)-4-methylpiperidin-l-yl)-lH-pyrazolo[4,3- d]thiazol-3-y I, 6-(l-(l-aminopropan-2-yl)piperidin-4-yl)-7-oxo-6,7-dihydro-lH-pyrazolo[4,3- d] pyrimidin-3-yl, 8-(4-(aminomethyl)-4-methylpiperidin-l-yl)-6-oxo-6,7-dihydro-lH-purin-2-yl, 6- amino-8-(4-(aminomethyl)-4-methylpiperidin-l-yl)-7H-purin-2-yl, 4-amino-2-(3-(aminomethyl)-3- methylpyrrolidin-l-yl)-6-oxo-l,6-dihydropyrimidin-5-yl, 2-(4-(aminomethyl)-4-methylpiperidin-l-yl)-

4-cyano-l-methyl-6-oxo-l,6-dihydrc>pyrimidin-5-yl, 4-amino-2-(4-amino-2-oxa-8- azaspiro[4.5]decan-8-yl)-6-oxo-l,6-dihydropyrimidin-5-yl, 4-amino-2-(3,6- diazabicyclo[3.2.0] heptan-6-yl)-6-oxo-l,6-dihydropyrimidin-5-yl, 4-amino-2-(hexahydrc>pyrrolo[3,4- c] pyrrol-2(lH)-yl)-6-oxo-l,6-dihydropyrimidin-5-yl, 4-amino-2-(4-(aminomethyl)-4-methylpiperidin- l-yl)-6-oxo-l,6-dihydropyrimidin-5-yl, 4-amino-2-(6-amino-3-azabicyclo[3.2.0] heptan-3-yl)-6-oxo- l,6-dihydropyrimidin-5-yl, 4-amino-2-(6-amino-2-azaspirc>[3.4]octan-2-yl)-l-methyl-6-oxo-l,6- dihydropyrimidin-5-yl, 4-amino-2-(6-amino-3-azabicyclo[3.1.0] hexan-3-yl)-6-oxo-l,6- dihydropyrimidin-5-yl, 4-amino-2-(6-amino-2-azaspiro[3.4]octan-2-yl)-6-oxo-l,6-dihydropyrimidin- 5-yl, 5-(4-(aminomethyl)-4-methylpiperidin-l-yl)-l-methyl-6-oxo-l,6-dihydropyrazin-2-yl, 5-(4- amino-2-oxa-8-azaspiro[4.5]decan-8-yl)-6-oxo-l/6-dihydropyrazin-2-yl/ (S)-5-(4-amino-2-oxa-8- azaspiro[4.5]decan-8-yl)-l-methyl-6-oxo-l,6-dihydrc>pyrazin-2-yl, 5-(2- aminospiro[bicyclo[3.1.0] hexane-3/4'-piperidin]-l'-yl)-l-methyl-6-oxo-l/6-dihydropyrazin-2-yl/ 5- ((3S,4S)-4-amino-3-methyl-2-oxa-8-azaspirc>[4.5]decan-8-yl)-6-oxo-l,6-dihydrc>pyrazin-2-yl, 5- ((3S,4S)-4-amino-3-methyl-2-oxa-8-azaspirc>[4.5]decan-8-yl)-l-methyl-6-oxo-l,6-dihydrc>pyrazin-2-yl,

5-((lR,3R)-l-amino-3-methyl-8-azaspiro[4.5]decan-8-yl)-l-methyl-6-oxo-l,6-dihydropyrazin-2-yl, 5-

(4-(aminomethyl)-4-methylpiperidin-l-yl)-6-oxo-l,6-dihydrc>pyrazin-2-yl, 6-(4-(aminomethyl)-4- methylpiperidin-l-yl)-4-oxo-4,5-dihydrc>-lH-pyrazolo[3,4-d] pyrimidin-3-yl, 6-(4-(aminomethyl)-4- methylpiperidin-l-yl)-4-methyl-5-oxo-4,5-dihydrc>-lH-pyrazolo[3,4-b]pyrazin-3-yl, 6-(4- (aminomethyl)-4-methylpiperidin-l-yl)-5-oxo-4,5-dihydro-lH-pyrazolo[3,4-b] pyrazin-3-yl, 6-(3-

(aminomethyl)-3-methylpyrrolidin-l-yl)-5-methyl-4-oxo-4,5-dihydro-lH-pyrazolo[3,4-d] pyrimidin-3- yl, 6-(4-(aminomethyl)-4-methylpiperidin-l-yl)-5-methyl-4-oxo-4,5-dihydro-lH-pyrazolo[3,4- d] pyrimidin-3-yl, 6-(4-(aminomethyl)-4-methylpiperidin-l-yl)-lH-pyrazolo[3,4-d] pyrimidin-3-yl, 4- amino-2-(hexahydropyrrolo[3/4-c] pyrrol-2(lH)-yl)-l-methyl-6-oxo-l/6-dihydropyrimidin-5-yl/ 4- amino-2-((3S,4S)-4-amino-3-methyl-2-oxa-8-azaspiro[4.5]decan-8-yl)-6-oxo-l,6-dihydropyrimidin-5- yl, 4-amino-2-((3S,4S)-4-amino-3-methyl-2-oxa-8-azaspiro[4.5]decan-8-yl)-l-methyl-6-oxo-l,6- dihydropyrimidin-5-yl, 4-amino-2-(6-amino-3-azabicyclo[3.1.0] hexan-3-yl)-l-methyl-6-oxo-l,6- dihydropyrimidin-5-yl, 4-amino-2-(6-amino-3-azabicyclo[3.2.0] heptan-3-yl)-l-methyl-6-oxo-l,6- dihydropyrimidin-5-yl, 5-(4-(aminomethyl)-4-fluoropiperidin-l-yl)-6-oxo-l,6-dihydropyrazin-2-yl, 5- (4-(aminomethyl)-4-hydroxypiperidin-l-yl)-6-oxo-l,6-dihydropyrazin-2-yl, (R)-5-(6-amino-2- azaspiro[3.4]octan-2-yl)-6-oxo-l,6-dihydropyrazin-2-yl, (S)-5-(6-amino-2-azaspiro[3.4]octan-2-yl)-6- oxo-l,6-dihydropyrazin-2-yl, l-(3-aminocyclohexyl)-2-oxo-l,2-dihydropyridin-4-yl, (R)-5-(4-amino-2- oxa-8-azaspiro[4.5]decan-8-yl)-6-oxo-l,6-dihydropyrazin-2-yl, 5-(4-amino-4- (fluoromethyl)piperidin-l-yl)-6-oxo-l,6-dihydropyrazin-2-yl, 5-((3S,4S)-4-amino-3-methyl-2-oxa-8- azaspiro[4.5]decan-8-yl)-6-oxo-l,6-dihydropyrazin-2-yl, (R)-5-(l-amino-8-azaspiro[4.5]decan-8-yl)-6- oxo-l,6-dihydropyrazin-2-yl, 5-(6-amino-3-azabicyclo[3.1.0] hexan-3-yl)-6-oxo-l,6-dihydropyrazin-2- yl

(R)-5-(3-amino-3H-spirc>[benzofuran-2,4'-pipe din]- -yl)-6-oxo-l,6-dihydrc>pyrazin-2-yl, 2-((3S,4S)- 4-amino-3-methyl-2-oxa-8-azaspiro[4.5]decan-8-yl)-3-methyl-4-oxo-4,7-dihydro-3H-pyrrolo[2,3- d] pyrimidin-5-yl, 2-((3S,4S)-4-amino-3-methyl-2-oxa-8-azaspiro[4.5]decan-8-yl)-4-oxo-4, 7-dihydro-

3H-pyrrolo[2,3-d] pyrimidin-5-yl, (R)-5-( 1-ami no-3, 3-difluoro-8-azaspiro[4.5]decan-8-yl)-6-oxo-l, 6- dihydropyrazin-2-yl, 5-((lR)-l-amino-3-fluoro-8-azaspiro[4.5]decan-8-yl)-6-oxo-l,6-dihydropyrazin- 2-yl, (S)-5-(5-amino-5, 7-dihydrospiro[cyclopenta [b] pyridine-6, 4'-piperidi n]-l'-yl)-6-oxo-l, 6- dihydropyrazin-2-yl, (S)-5-(l-amino-l,3-dihydrospiro[indene-2,4'-piperidin]-l'-yl)-6-oxo-l,6- dihydropyrazin-2-yl, (S)-5-(4-amino-2-ch loro-4, 6-dihydrospi ro[cyclopenta[d]thiazole-5, 4'-piperidin]- l'-yl)-6-oxo-l,6- dihydropyrazin-2-yl, (R)-5-(3-aminospiro[indoline-2,4'-piperidin]-l'-yl)-6-oxo-l,6- dihydropyrazin-2-yl, or (S)-5-(l-amino-4-methoxy-l,3-dihydrospiro[indene-2,4'-piperidin]-l'-yl)-6- oxo-1, 6-dihydropyrazin-2-yl.

84. The compound of claim 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23,

24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77,

78, 79, 80, 81, 82, or 83, wherein X is S.

85. The compound of claim 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50,

51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77,

78, 79, 80, 81, 82, or 83, wherein X is a bond.

86. The compound of claim 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23,

24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50,

51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77,

78, 79, 80, 81, 82, or 83, wherein X is O.

87. The compound of claim 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50,

51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77,

78, 79, 80, 81, 82, or 83, wherein X is N H.

88. The compound of claim 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23,

24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77,

78, 79, 80, 81, 82, or 83, wherein X is CH(CH3).

89. The compound of claim 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23,

24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, or 83, wherein X is CH2.

90. The compound of any preceding claim, wherein RA is H.

91. The compound of any preceding claim, wherein RA is CH3.

92. The compound of any preceding claim, or a pharmaceutically acceptable salt thereof, wherein the compound is optionally substituted at any position that is chemically permissible. 93. The compound of any one of the preceding claims, wherein the compound is an R-enantiomer.

94. The compound of any one of the preceding claims, wherein the compound is an S-enantiomer.

95. The compound of any one of the preceding claims, wherein the compound is deuterated.

96. A compound, or a pharmaceutically acceptable salt thereof, wherein the compound is any one of the following compounds that is optionally substituted: 3-(4-(aminomethyl)-4-methylpiperidin-l- yl)-6-(2,3-dichlorophenyl)pyrazin-2(lH)-one, 3-(4-(aminomethyl)-4-methylpiperidin-l-yl)-6-((2,3- dichlorophenyl)thio)pyrazin-2(lH)-one, 3-(4-(aminomethyl)-4-methylpiperidin-l-yl)-6-((2,3- dichlorophenyl)thio)-l-methylpyrazin-2(lH)-one, 3-(4-(aminomethyl)-4-methylpiperidin-l-yl)-6-

(2,3-dichlorophenoxy)pyrazin-2(lH)-one, 3-(4-(aminomethyl)-4-methylpiperidin-l-yl)-6-((2,3- dichlorophenyl)amino)pyrazin-2(lH)-one, 3-(4-(aminomethyl)-4-methylpiperidin-l-yl)-6-(2,3- dichlorophenoxy)-l-methylpyrazin-2(lH)-one, 3-(3-(aminomethyl)-3-methylpyrrolidin-l-yl)-6-((2,3- dichlorophenyl)thio)pyrazin-2(lH)-one, 6-((2,3-dichlorophenyl)thio)-3-(hexahydropyrrolo[3,4- c] pyrrol-2(lH)-yl)pyrazin-2(lH)-one, 3-(3,6-diazabicyclo[3.2.0]heptan-6-yl)-6-((2,3- dichlorophenyl)thio)pyrazin-2(lH)-one, 3-(4-amino-2-oxa-8-azaspiro[4.5]decan-8-yl)-6-((2,3- dichlorophenyl)thio)pyrazin-2(lH)-one, 6-((2,3-dichlorophenyl)thio)-3-(piperidin-4-ylamino)pyrazin- 2(lH)-one, 3-(4-(aminomethyl)-4-methylpiperidin-l-yl)-6-(naphthalen-l-ylthio)pyrazin-2(lH)-one, 3- (2-aminospiro[bicyclo[3.1.0]hexane-3,4'-piperidin]-l'-yl)-6-((2,3-dichlorophenyl)thio)pyrazin-2(lH)- one, 3-((3S,4S)-4-amino-3-methyl-2-oxa-8-azaspiro[4.5]decan-8-yl)-6-((2,3- dichlorophenyl)thio)pyrazin-2(lH)-one, 3-((3S,4S)-4-amino-3-methyl-2-oxa-8-azaspiro[4.5]decan-8- yl)-6-((2,3-dichlorophenyl)thio)-l-methylpyrazin-2(lH)-one, 3-((lR,3R)-l-amino-3-methyl-8- azaspiro[4.5]decan-8-yl)-6-((2,3-dichlorophenyl)thio)pyrazin-2(lH)-one, 3-(4-(aminomethyl)-4- methylpiperidin-l-yl)-6-((2-(trifluoromethyl)phenyl)thio)pyrazin-2(lH)-one, 3-(4-(aminomethyl)-4- methylpiperidin-l-yl)-6-((2-(trifluoromethyl)pyridin-3-yl)thio)pyrazin-2(lH)-one, 6-((5-amino-2- chlorophenyl)thio)-3-(4-(aminomethyl)-4-methylpiperidin-l-yl)pyrazin-2(lH)-one, 6-((5-amino-2- chloropyridin-3-yl)thio)-3-(4-(aminomethyl)-4-methylpiperidin-l-yl)pyrazin-2(lH)-one, 6-((3-amino-

2-chlorophenyl)thio)-3-(4-(aminomethyl)-4-methylpiperidin-l-yl)pyrazin-2(lH)-one, 6-((2-amino-3- chloropyridin-4-yl)thio)-3-(4-(aminomethyl)-4-methylpiperidin-l-yl)pyrazin-2(lH)-one, 3-(4- (aminomethyl)-4-methylpiperidin-l-yl)-6-((2-chloro-3-methoxyphenyl)thio)pyrazin-2(lH)-one, 3-(4-

(aminomethyl)-4-methylpiperidin-l-yl)-6-((3-chloro-2-methoxypyridin-4-yl)thio)pyrazin-2(lH)-one,

3-((3S,4S)-4-amino-3-methyl-2-oxa-8-azaspiro[4.5]decan-8-yl)-6-((3-fluoro-lH-indol-4- yl)thio)pyrazin-2(lH)-one, 4-((5-(4-(aminomethyl)-4-methylpiperidin-l-yl)-6-oxo-l,6- dihydropyrazin-2-yl)thio)-3,3-difluoroindolin-2-one, 6-((l-acetyl-3,3-difluoroindolin-4-yl)thio)-3-(4- (aminomethyl)-4-methylpipe din-l-yl)pyrazin-2(lH)-one, N-(3-((5-(4-(aminomethyl)-4- methylpiperidin-l-yl)-6-oxo-l,6-dihydrc>pyrazin-2-yl)thio)-2-chlorc>phenyl)-4-hydrc>xy-l,5,5- trimethyl-2-oxo-2,5-dihydro-lH-pyrrole-3-carboxamide, N-(3-((5-(4-(aminomethyl)-4- methylpiperidin-l-yl)-6-oxo-l,6-dihydropyrazin-2-yl)thio)-2-chlorophenyl)-2-hydroxy-4-oxo-4H- pyrido[l,2-a] pyrimidine-3-carboxamide, N-(3-((5-(4-(aminomethyl)-4-methylpiperidin-l-yl)-6-oxo- l,6-dihydropyrazin-2-yl)thio)-2-chlorophenyl)-2-hydroxy-4-oxo-4H-pyrazino[l,2-a] pyrimidine-3- carboxamide,

N-(3-((5-(4-(aminomethyl)-4-methylpiperidin-l-yl)-6-oxo-l,6-dihydropyrazin-2-yl)thio)-2- chlorophenyl)-7-hydroxy-5-oxo-5H-thiazolo[3,2-a]pyrimidine-6-carboxamide, N-(3-((5-(4- (aminomethyl)-4-methylpiperidin-l-yl)-6-oxo-l,6-dihydropyrazin-2-yl)thio)-2-chlorophenyl)-7- hydroxy-5-oxo-l,5-dihydroimidazo[l,2-a] pyrimidine-6-carboxamide, N-(3-((5-(4-(aminomethyl)-4- methylpiperidin-l-yl)-6-oxo-l,6-dihydropyrazin-2-yl)thio)-2-chlorophenyl)-2-hydroxy-4-oxo-6,7,8,9- tetrahydro-4H-pyrido[l,2-a] pyrimidine-3-carboxamide, 6-(4-(aminomethyl)-4-methylpiperidin-l-yl)-

3-((2,3-dichlorophenyl)amino)-l,5-dihydro-4H-pyrazolo[3,4-d]pyrimidin-4-one, 6-(4-(aminomethyl)- 4-methyl piperidin-l-yl)-3-(2,3-dichlorophenoxy)-l,5-dihydro-4H-pyrazolo[3,4-d]pyrimidin-4-one, 6-

(4-(aminomethyl)-4-methylpiperidin-l-yl)-3-((2,3-dichlorophenyl)amino)-5-methyl-l,5-dihydro-4H- pyrazolo[3,4-d]pyrimidin-4-one, 6-(4-(aminomethyl)-4-methylpiperidin-l-yl)-3-(2,3- dichlorophenoxy)-5-methyl-l,5-dihydro-4H-pyrazolo[3,4-d] pyrimidin-4-one, 6-(4-(aminomethyl)-4- methylpiperidin-l-yl)-3-(2,3-dichlorophenoxy)-4-methyl-l,4-dihydro-5H-pyrazolo[3,4-b]pyrazin-5- one, 6-(4-(aminomethyl)-4-methylpiperidin-l-yl)-3-(2,3-dichlorophenoxy)-l,4-dihydro-5H- pyrazolo[3,4-b]pyrazin-5-one, 6-(4-(aminomethyl)-4-methylpiperidin-l-yl)-3-(2,3-dichlorophenyl)- l,4-dihydro-5H-pyrazolo[3,4-b] pyrazin-5-one, 6-(4-(aminomethyl)-4-methylpiperidin-l-yl)-3-((2,3- dichlorophenyl)thio)-l,4-dihydro-5H-pyrazolo[3,4-b] pyrazin-5-one, 6-(3-(aminomethyl)-3- methylpyrrolidin-l-yl)-3-(2,3-dichlorophenoxy)-l,5-dihydro-4H-pyrazolo[3,4-d]pyrimidin-4-one, 6- (3-(aminomethyl)-3-methylpyrrolidin-l-yl)-3-(2,3-dichlorophenoxy)-5-methyl-l,5-dihydro-4H- pyrazolo[3,4-d]pyrimidin-4-one, 6-(4-(aminomethyl)-4-methylpiperidin-l-yl)-3-(l-(2,3- dichlorophenyl)ethyl)-5-methyl-l,5-dihydro-4H-pyrazolo[3,4-d]pyrimidin-4-one, 6-(4-

(aminomethyl)-4-methylpiperidin-l-yl)-3-(2,3-dichlorobenzyl)-l,5-dihydro-4H-pyrazolo[3,4- d] pyrimidin-4-one, 7-amino-2-(4-(aminomethyl)-4-methylpiperidin-l-yl)-5-((2,3- dichlorophenyl)thio)quinazolin-4(3H)-one, 2-(4-(aminomethyl)-4-methylpiperidin-l-yl)-5-((2,3- dichlorophenyl)thio)pyrido[3,4-d]pyrimidin-4(3H)-one, 2-(4-(aminomethyl)-4-methylpiperidin-l-yl)- 5-((2,3-dichlorophenyl)thio)pyrido[2,3-d]pyrimidin-7(8H)-one, (l-(3-((2,3-dichlorophenyl)thio)-lH- pyrazolo[4,3-d]thiazol-5-yl)-4-methylpiperidin-4-yl)methanamine, 6-(l-(l-aminopropan-2- yl)piperidin-4-yl)-3-((2,3-dichlorophenyl)thio)-l,6-dihydro-7H-pyrazolo[4,3-d] pyrimidin-7-one, 5-(l-

(l-aminopropan-2-yl)piperidin-4-yl)-3-((2,3-dichlorc>phenyl)thio)-l,6-dihydro-7H-pyrazolo[4,3- d] pyrimidin-7-one, 6-amino-2-(3-(aminomethyl)-3-methylpyrrc>lidin-l-yl)-5-((2,3- dichlorophenyl)thio)-3-methylpyrimidin-4(3H)-one, 6-amino-5-((2,3-dichlorophenyl)thio)-2-

(hexahydropyrrolo[3,4-c]pyrrol-2(lH)-yl)-3-methylpyrimidin-4(3H)-one, 6-amino-2-(3- (aminomethyl)-3-methylpyrrolidin-l-yl)-5-((2,3-dichlorc>phenyl)thio)pyrimidin-4(3H)-one, 2-(4-

(aminomethyl)-4-methylpiperidin-l-yl)-5-(2,3-dichlorophenyl)-l-methyl-6-oxo-l,6- dihydropyrimidine-4-carbonitrile, 6-amino-2-((3S,4S)-4-amino-3-methyl-2-oxa-8- azaspiro[4.5]decan-8-yl)-5-(2,3-dichlorophenoxy)pyrimidin-4(3H)-one, 6-amino-2-((3S,4S)-4-amino- 3-methyl-2-oxa-8-azaspiro[4.5]decan-8-yl)-5-(2/3-dichlorophenoxy)-3-methylpyrimidin-4(3H)-one/ 6-amino-2-(3,6-diazabicyclo[3.2.0]heptan-6-yl)-5-((2,3-dichlorophenyl)thio)-3-methylpyrimidin-

4(3H)-one, 6-amino-2-(3,6-diazabicyclo[3.2.0]heptan-6-yl)-5-((2,3-dichlorc>phenyl)thio)pyrimidin- 4(3H)-one, 6-amino-5-((2/3-dichlorophenyl)thio)-2-(hexahydropyrrolo[3/4-c] pyrrol-2(lH)- yl)pyrimidin-4(3H)-one, 6-amino-2-(4-(aminomethyl)-4-methylpiperidin-l-yl)-5-(2,3- dichlorophenoxy)pyrimidin-4(3H)-one, 6-amino-2-(6-amino-3-azabicyclo[3.2.0] heptan-3-yl)-5-((2,3- dichlorophenyl)thio)pyrimidin-4(3H)-one, 6-amino-2-(6-amino-2-azaspiro[3.4]octan-2-yl)-5-((2,3- dichlorophenyl)thio)-3-methylpyrimidin-4(3H)-one, 6-amino-2-(6-amino-3-azabicyclo[3.1.0]hexan- 3-yl)-5-((2,3-dichlorophenyl)thio)-3-methylpyrimidin-4(3H)-one, 6-amino-2-(6-amino-3- azabicyclo[3.2.0] heptan-3-yl)-5-((2,3-dichlorophenyl)thio)-3-methylpyrimidin-4(3H)-one, 6-amino-2- (6-amino-3-azabicyclo[3.1.0]hexan-3-yl)-5-((2,3-dichlorophenyl)thio)pyrimidin-4(3H)-one, 6-amino- 2-(6-amino-2-azaspiro[3.4]octan-2-yl)-5-((2/3-dichlorophenyl)thio)pyrimidin-4(3H)-one/ 6-(3-

(aminomethyl)-3-methylpyrrolidin-l-yl)-3-((2,3-dichlorc>phenyl)amino)-l,5-dihydro-4H- pyrazolo[3,4-d]pyrimidin-4-one, 3-(4-(aminomethyl)-4-fluoropiperidin-l-yl)-6-((2/3- dichlorophenyl)thio)pyrazin-2(lH)-one, 3-(4-(aminomethyl)-4-hydroxypiperidin-l-yl)-6-((2/3- dichlorophenyl)thio)pyrazin-2(lH)-one, (R)-3-(6-amino-2-azaspiro[3.4]octan-2-yl)-6-((2,3- dichlorophenyl)thio)pyrazin-2(lH)-one, (S)-3-(6-amino-2-azaspiro[3.4]octan-2-yl)-6-((2,3- dichlorophenyl)thio)pyrazin-2(lH)-one, (S)-3-(4-amino-2-oxa-8-azaspiro[4.5]decan-8-yl)-6-((2,3- dichlorophenyl)thio)pyrazin-2(lH)-one, l-(3-aminocyclohexyl)-4-(2,3-dichlorophenyl)pyridin-2(lH)- one, (R)-3-(4-amino-2-oxa-8-azaspiro[4.5]decan-8-yl)-6-((2,3-dichlorc>phenyl)thio)pyrazin-2(lH)-one, 3-(4-amino-4-(fluoromethyl)piperidin-l-yl)-6-((2,3-dichlorophenyl)thio)pyrazin-2(lH)-one, 6-((2- amino-3-chloropyridin-4-yl)thio)-3-((3S,4S)-4-amino-3-methyl-2-oxa-8-azaspiro[4.5]decan-8- yl)pyrazin-2(lH)-one, (R)-3-(l-amino-8-azaspiro[4.5]decan-8-yl)-6-((2,3-dichlorophenyl)thio)pyrazin- 2(lH)-one, 3-((3S,4S)-4-amino-3-methyl-2-oxa-8-azaspiro[4.5]decan-8-yl)-6-((2-chloro-3- methoxyphenyl)thio)pyrazin-2(lH)-one, 3-((3S,4S)-4-amino-3-methyl-2-oxa-8-azaspiro[4.5]decan-8- yl)-6-((3-chloro-2-methoxypyridin-4-yl)thio)pyrazin-2(lH)-one, 3-((3S,4S)-4-amino-3-methyl-2-oxa-

8-azaspiro[4.5]decan-8-yl)-6-((2,3-dichloropyridin-4-yl)thio)pyrazin-2(lH)-one, 3-((3S,4S)-4-amino-3- methyl-2-oxa-8-azaspiro[4.5]decan-8-yl)-6-((2,3-difluorophenyl)thio)pyrazin-2(lH)-one, 3-((3S,4S)- 4-amino-3-methyl-2-oxa-8-azaspiro[4.5]decan-8-yl)-6-((3-chloro-2-fluoropyridin-4-yl)thio)pyrazin- 2(lH)-one, 4-((5-(4-(aminomethyl)-4-methylpiperidin-l-yl)-6-oxo-l,6-dihydropyrazin-2-yl)thio)-3,3- difluoroindolin-2-one, 3-((3S,4S)-4-amino-3-methyl-2-oxa-8-azaspiro[4.5]decan-8-yl)-6-((2,3- dichlorophenyl)thio)pyridin-2(lH)-one, 3-(6-amino-3-azabicyclo[3.1.0]hexan-3-yl)-6-((2,3- dichlorophenyl)thio)pyrazin-2(lH)-one, (R)-3-(3-amino-3H-spiro[benzofuran-274'-piperidin]-l'-yl)-6- ((2,3-dichlorophenyl)thio)pyrazin-2(lH)-one, (S)-3-(4-amino-2-oxa-8-azaspiro[4.5]decan-8-yl)-6- ((2,3-dichloropyridin-4-yl)thio)pyrazin-2(lH)-one, 2-((3S,4S)-4-amino-3-methyl-2-oxa-8- azaspiro[4.5]decan-8-yl)-5-(2,3-dichlorophenoxy)-3-methyl-3,7-dihydro-4H-pyrrolo[2,3-d] pyrimidin-

4-one, 2-((3S,4S)-4-amino-3-methyl-2-oxa-8-azaspiro[4.5]decan-8-yl)-5-(2,3-dichlorophenoxy)-3,7- dihydro-4H-pyrrolo[2,3-d]pyrimidin-4-one, 6-((3S,4S)-4-amino-3-methyl-2-oxa-8- azaspiro[4.5]decan-8-yl)-3-((2,3-dichlorophenyl)amino)-l,4-dihydro-5H-pyrazolo[3,4-b] pyrazin-5- one, 6-((3S,4S)-4-amino-3-methyl-2-oxa-8-azaspiro[4.5]decan-8-yl)-3-((2,3-dichlorophenyl)amino)- 4-methyl-l,4-dihydro-5H-pyrazolo[3,4-b] pyrazin-5-one, 2-((3S,4S)-4-amino-3-methyl-2-oxa-8- azaspiro[4.5]decan-8-yl)-5-((2,3-dichlorophenyl)amino)-3,7-dihydro-4H-pyrrolo[2,3-d]pyrimidin-4- one, 2-((3S,4S)-4-amino-3-methyl-2-oxa-8-azaspiro[4.5]decan-8-yl)-5-((2,3-dichlorophenyl)amino)- 3-methyl-3,7-dihydro-4H-pyrrolo[2,3-d] pyrimidin-4-one, (R)-3-(l-amino-3,3-difluoro-8- azaspiro[4.5]decan-8-yl)-6-((2,3-dichlorophenyl)thio)pyrazin-2(lH)-one, 3-((lR)-l-amino-3-fluoro-8- azaspiro[4.5]decan-8-yl)-6-((2,3-dichlorophenyl)thio)pyrazin-2(lH)-one, 3-((3S,4S)-4-amino-3- methyl-2-oxa-8-azaspiro[4.5]decan-8-yl)-6-((2-chloro-3-methylphenyl)thio)pyrazin-2(lH)-one, 3- ((3S,4S)-4-amino-3-methyl-2-oxa-8-azaspiro[4.5]decan-8-yl)-6-((3-chloro-2-methylpyridin-4- yl)thio)pyrazin-2(lH)-one, N-(3-((5-((3S,4S)-4-amino-3-methyl-2-oxa-8-azaspiro[4.5]decan-8-yl)-6- oxo-l,6-dihydropyrazin-2-yl)thio)-2-chlorophenyl)-2-hydroxy-4-oxo-4H-pyrido[l,2-a] pyrimidine-3- carboxamide, 4-((5-((3S,4S)-4-amino-3-methyl-2-oxa-8-azaspiro[4.5]decan-8-yl)-6-oxo-l,6- dihydropyrazin-2-yl)thio)-3,3-difluoro-l-methylindolin-2-one, 3-((3S,4S)-4-amino-3-methyl-2-oxa-8- azaspiro[4.5]decan-8-yl)-6-((3-chloro-l-methyl-2-oxo-l,2-dihydropyridin-4-yl)thio)pyrazin-2(lH)- one, (S)-3-(5-amino-5,7-dihydrospiro[cyclopenta[b]pyridine-6,4'-piperidin]-l'-yl)-6-((2,3- dichlorophenyl)thio)pyrazin-2(lH)-one, (S)-3-(l-amino-l,3-dihydrospiro[indene-2,4'-piperidin]-l'- yl)-6-((2,3-dichloropyridin-4-yl)thio)pyrazin-2(lH)-one, (S)-3-(4-amino-2-chloro-4,6- dihydrospiro[cyclopenta[d]thiazole-5,4'-piperidin]-l'-yl)-6-((2,3-dichlorophenyl)thio)pyrazin-2(lH)- one, (R)-3-(3-aminospiro[indoline-2,4'-piperidin]-l'-yl)-6-((2,3-dichlorophenyl)thio)pyrazin-2(lH)- one, (S)-3-(l-amino-4-methoxy-l,3-dihydrospiro[indene-2,4'-piperidin]-l'-yl)-6-((2,3- dichlorophenyl)thio)pyrazin-2(lH)-one, 3-((3S,4S)-4-amino-3-methyl-2-oxa-8-azaspiro[4.5]decan-8- yl)-6-((3-chloro-2-oxo-l,2-dihydropyridin-4-yl)thio)pyrazin-2(lH)-one, (S)-3-(l-amino-l,3- dihydrospiro[indene-2,4'-piperidin]-l'-yl)-6-((2,3-dichlorophenyl)thio)pyrazin-2(lH)-one, 3-((3S,4S)- 4-amino-3-methyl-2-oxa-8-azaspiro[4.5]decan-8-yl)-6-((2-chloro-3-fluorophenyl)thio)pyrazin-2(lH)- one, (l-(3-((2,3-dichlorophenyl)thio)-lH-pyrazolo[3,4-d] pyrimidin-6-yl)-4-methylpiperidin-4- yl)methanamine, 3-(4-amino-2-oxa-8-azaspiro[4.5]decan-8-yl)-6-((2,3-dichlorophenyl)thio)-l- methylpyrazin-2(lH)-one, 3-(2-aminospiro[bicyclo[3.1.0] hexane-3,4'-piperidin]-l'-yl)-6-((2,3- dichlorophenyl)thio)-l-methylpyrazin-2(lH)-one, 3-((lR,3R)-l-amino-3-methyl-8- azaspiro[4.5]decan-8-yl)-6-((2,3-dichlorophenyl)thio)-l-methylpyrazin-2(lH)-one, or 6-((3S,4S)-4- amino-3-methyl-2-oxa-8-azaspiro[4.5]decan-8-yl)-3-(2,3-dichlorophenoxy)-l,4-dihydro-5H- pyrazolo[3,4-b]pyrazin-5-one.

97. The compound of claim 1, wherein any substituent of the compound has a molecular weight of about 15 g/mol to about 200 g/mol.

98. A pharmaceutical composition comprising a compound of any preceding claims or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable vehicle, diluent, or carrier.

99. A method of treating a mammal, including a human, having a disease, disorders, or condition associated with the aberrant activity of SHP2, including cancer and autoimmune disorders, comprising administering to a mammal in need thereof a therapeutically effective amount of the compound of any preceding claim.

100. A medicament comprising a composition comprising a therapeutically effective amount of the compound of any preceding claim. 101. A kit comprising a medicament of claim 100 and a label indicating that the medicament is for treating a disease, disorders, or condition associated with the aberrant activity of SHP2.

Description:
SHP2 INHIBITORS AND USES THEREOF

CROSS-REFERENCE TO RELATED APPLICATION

This application claims the benefit of U.S. Provisional Application No. 62/646,107, filed March 21, 2018; which is incorporated by reference by its entirety.

FIELD

The present disclosure relates to inhibitors of protein tyrosine phosphatase SHP2 (Src Homolgy-2 phosphatase) and their use in treating SHP2 mediated disorders. More specifically, this disclosure is directed to compounds that inhibit SHP2 and compositions comprising these compounds, methods of treating diseases associated with the aberrant activity of SHP2, and methods of synthesizing these compounds.

BACKGROUND

Tyrosyl phosphorylation regulates human cellular processes from cell differentiation to growth and apoptosis etc. Tyrosyl phosphorylation is regulated by protein-tyrosine kinases (PTK) and protein-tyrosine phosphatases (PTP). The imbalance of regulation governed by PTK and PTP activity leads to various diseases.

SHP2 is a non-receptor protein tyrosine phosphatase (PTP) encoded by the Protein-tyrosine phosphatase non-receptor type 11 (PTPN11) gene. It contains two N-terminal Src homology 2 domains (N-SH2 and C-SH2), a catalytic domain, and a C-terminal tail. The protein exists in an inactive, auto-inhibited basal conformation that blocks the active site. This self-inhibition state is stabilized by a binding network involving residues from both the N-SH2 and catalytic domains. Stimulation by, for example, cytokines or growth factors results in enzymatic activation of SHP2 and makes the active site available for dephosphorylation of PTPN11 substrates.

SHP2 is widely expressed in most tissues and contributes to various cellular functions including proliferation, differentiation, cell cycle maintenance and migration. It is involved in signaling through the Ras-mitogen-activated protein kinase, the JAK-STAT, EGFR, or the phosphoinositol 3-kinase-AKT pathways. Mutations in the PTPN11 gene and subsequently in SHP2 lead to hyperactivation of SHP2 catalytic activity, and have been identified in several human diseases, such as Noonan Syndrome, Leopard Syndrome, juvenile myelomonocytic leukemias, neuroblastoma, melanoma, acute myeloid leukemia and cancers of the breast, lung, melanoma, neuroblastoma, hepatocellular carcinoma, and colon. These mutations disrupt the auto-inhibition between the N-SH2 domains and the catalytic site allowing constitutive access of substrates to the catalytic site of the enzyme.

Additionally, there is growing evidence that PTPN11/SHP2 may be implicated in immune evasion during tumorigenesis, and hence a SHP2 inhibitor could stimulate the immune response in cancer patients. Furthermore, SHP2 plays an important role in JAK/STAT3 pathway, with clear correlation between its phosphatase activity and systemic autoimmunity, thus a SHP2 inhibitor could be used to treat autoimmune diseases such as Lupus and Rheumatoid Arthritis.

Therefore, SHP2 represents a highly attractive target for the development of novel therapies for the treatment of various diseases associated with the aberrant activity of SHP2. The compounds of the present disclosure that are capable of inhibiting the activity of SHP2, possess great potential as novel small molecule therapies for the treatment of various diseases mentioned above.

SUMMARY

This disclosure relates to compounds represented by Formula 1: (Formula 1)

or a pharmaceutically acceptable salt thereof; wherein X is S, O, NR A , CHR A , SO, S0 2 , CO, or a bond; Ring A is an optionally substituted aryl, heteroaryl, or bicyclic ring system; Ring B is an optionally substituted heterocyclic ring system, including non-aromatic ring system and heteroaryl, comprising a mono-cyclic ring, a bicyclic ring system, a tricyclic ring system, or a tetracyclic ring system, wherein the heterocyclic ring system contains at least 2 ring nitrogen atoms; and R A is H or Ci- 6 hydrocarbyl.

Some embodiments include a method of treating diseases, disorders, or conditions associated with the aberrant activity of SHP2, such as but not limited to, cancer, and autoimmune disorders, comprising administering a therapeutically effective amount of a compound described herein, or any optionally substituted compound represented in Table I below, or a pharmaceutically acceptable salt thereof (referred to collectively herein as a "subject compound"), to a patient in need thereof.

Some embodiments include use of a compound described herein, such as a compound of Formula 1, a subject compound described herein in the manufacture of a medicament for the treatment of cancer, autoimmune diseases, inflammatory diseases, autoinflammatory conditions, and other SHP2 mediated disorders in a mammal.

Some embodiments include a pharmaceutical composition comprising a therapeutically effective amount of a subject compound described herein, or a pharmaceutically acceptable salt thereof, in combination with at least one pharmaceutically acceptable vehicle, diluent, or carrier.

Some embodiments include a process for making a pharmaceutical composition comprising combining a subject compound described herein and at least one pharmaceutically acceptable carrier.

Some embodiments include a medicament comprising a composition comprising a therapeutically effective amount of a subject compound. Some embodiments include a kit comprising a medicament of above and a label indicating that the medicament is for treating a disease, disorders, or condition associated with the aberrant activity of SHP2.

DESCRIPTION

Unless otherwise indicated, any reference to a compound herein by structure, name, or any other means, includes pharmaceutically acceptable salts, such as sodium, potassium, and ammonium salts, or HCI, H2SO4, HCO2H, and CF3CO2H salts; prodrugs, such as ester prodrugs; alternate solid forms, such as polymorphs, solvates, hydrates, etc.; tautomers; or any other chemical species that may rapidly convert to a compound described herein under conditions in which the compounds are used as described herein.

If stereochemistry is not indicated, a name or structural depiction includes any stereoisomer or any mixture of stereoisomers.

Unless otherwise indicated, when a compound or chemical structural feature such as aryl is referred to as being "optionally substituted," it includes a feature that has no substituents (i.e. unsubstituted), or a feature that is "substituted," meaning that the feature has one or more substituents. The term "substituent" is broad, and includes a moiety that occupies a position normally occupied by one or more hydrogen atoms attached to a parent compound or structural feature. In some embodiments, a substituent may be an ordinary organic moiety known in the art, which may have a molecular weight (e.g. the sum of the atomic masses of the atoms of the substituent) of 15 g/mol to 50 g/mol, 15 g/mol to 100 g/mol, 15 g/mol to 150 g/mol, 15 g/mol to 200 g/mol, 15 g/mol to 300 g/mol, or 15 g/mol to 500 g/mol. In some embodiments, a substituent may be an ordinary organic moiety known in the art, which may have a molecular weight of 15 g/mol to 200 g/mol. In some embodiments, a substituent comprises, or consists of: 0-30, 0-20, 0-10, or 0-5 carbon atoms; and 0-30, 0-20, 0-10, or 0-5 heteroatoms, wherein each heteroatom may independently be: N, O, S, P, Si, F, Cl, Br, or I; provided that the substituent includes one C, N, O, S, P, Si, F, Cl, Br, or I atom. Examples of substituents include, but are not limited to, alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, aryl, heteroaryl, hydroxy, alkoxy, aryloxy, acyl, acyloxy, alkylcarboxylate, thiol, al kylthio, cyano, halo, thiocarbonyl, O-carbamyl, N-carbamyl, O-thiocarbamyl, N-thiocarbamyl, C-amido, N-amido, S-sulfonamido, N-sulfonamido, isocyanato, thiocyanato, isothiocyanato, nitro, silyl, sulfenyl, sulfinyl, sulfonyl, haloalkyl, haloalkoxyl, trihalomethanesulfonyl, trihalomethanesulfonamido, amino, phosphonic acid, etc.

For convenience, the term "molecular weight" is used with respect to a moiety or part of a molecule to indicate the sum of the atomic masses of the atoms in the moiety or part of a molecule, even though it may not be a complete molecule.

The term "treating" or "treatment" includes the diagnosis, cure, mitigation, treatment, or prevention of disease in man or other animals, or any activity that otherwise affects the structure or any function of the body of man or other animals.

A hydrogen atom in any position of a compound of Formula 1 may be replaced by a deuterium. In some embodiments, a compound of Formula 1 contains a deuterium atom or multiple deuterium atoms.

With respect to Formula 1, in some embodiments Ring A is: optionally substituted phenyl, optionally substituted naphthalen-l-yl, optionally substituted pyridin-3-yl, optionally substituted pyridin-4-yl, optionally substituted 2-oxo-l,2-dihydropyridin-4-yl, optionally substituted lH-indol-4- yl, optionally substituted 2-oxoindolin-4-yl, optionally substituted indolin-4-yl, optionally substituted 3-(2-oxo-2,5-dihydro-lH-pyrrole-3-carboxamido)phenyl, optionally substituted 3-(4-oxo-4H- pyrido[l,2-a]pyrimidine-3-carboxamido)phenyl, optionally substituted 3-(4-oxo-4H-pyrazino[l,2- a]pyrimidine-3-carboxamido)phenyl, optionally substituted 3-(5-oxo-5H-thiazolo[3,2-a]pyrimidine-6- carboxamido)phenyl, optionally substituted 3-(5-oxo-l,5-dihydroimidazo[l,2-a]pyrimidine-6- carboxamido)phenyl, or optionally substituted 3-(4-oxo-6,7,8,9-tetrahydro-4H-pyrido[l,2- a]pyrimidine-3-carboxamido)phenyl. For example, Ring A may be 2,3-dichlorophenyl, 2,3- dichloro pyridin-4-yl, or 2-amino-3-chloropyridin-4-yl.

phenyl naphthalen-l-yl pyridin-3-yl pyridin- 4 -yl 2-oxo-l,2-dihydropyridin-4-yl

lH-lndol-4-yl 2-Oxoindolin-4-yl lndolin-4-yl

3-(2-oxo-2 5-dihydro-l/-/-pyrrole-3-carboxamido)phenyl

3-(4-oxo-4/-/-pyrido[l,2-o]pyrimidine-3-carboxamido)phenyl

3-(4-oxo-4/-/-pyrazino[l,2-a]pyrimidine-3-carboxamido)phenyl

3-(5-oxo-5/-/-thiazolo[3,2-a]pyrimidine-6-carboxamido)phenyl

3-(5-oxo-l,5-dihydroimidazo[l,2-a]pyrimidine-6-carboxamido)p henyl

3-(4-oxo-6,7,8,9-tetrahydro-4/-/-pyrido[l,2-a]pyrimidine-3-c arboxamido)phenyl

In some embodiments, Ring A is unsubstituted.

In some embodiments, Ring A has a Cl substituent.

In some embodiments, Ring A has two Cl substituents.

In some embodiments, Ring A has two Cl substituents at 2- and 3-positions; for example, Ring A is 2,3-dichlorophenyl.

In some embodiments, Ring A has a CF3 substituent.

In some embodiments, Ring A has a CF3 substituent at 2-position.

In some embodiments, Ring A has an N FI2 substituent.

In some embodiments, Ring A has an N FI2 substituent and a Cl substituent.

In some embodiments, Ring A has an NFI2 substituent and a Cl substituent with Cl at 2-position and N FI2 at 5-position. In some embodiments, Ring A has an NH2 substituent and a Cl substituent with Cl at 2-position and N H2 at 3-position.

In some embodiments, Ring A has an NH2 substituent and a Cl substituent with Cl at 3-position and N H2 at 2-position.

In some embodiments, Ring A has an -OCH 3 substituent.

In some embodiments, Ring A has an -OCH 3 substituent and a Cl substituent.

In some embodiments, Ring A has an -OCH 3 substituent and a Cl substituent with Cl at 2- position and -OCH 3 at 3-position.

In some embodiments, Ring A has an F substituent.

In some embodiments, Ring A has two F substituents.

In some embodiments, Ring A has two F substituents at same position.

In some embodiments, Ring A has two F substituents at 2- and 3-positions.

In some embodiments, Ring A has an F substituent and a Cl substituent.

In some embodiments, Ring A has an F substituent and a Cl substituent with Cl at 2-position and F at 3-position.

In some embodiments, Ring A has an acetyl substituent.

In some embodiments, Ring A has a CH 3 substituent.

In some embodiments, Ring A has two CH 3 substituents that are at same position.

In some embodiments, Ring A has a CH 3 substituent and a Cl substituent.

In some embodiments, Ring A has a CH3 substituent and a Cl substituent with Cl at 2-position and CH3 at 4-position.

In some embodiments, Ring A has a CH3 substituent and a Cl substituent with Cl at 2-position and CH3 at 3-position.

In some embodiments, Ring A has a CH 3 substituent and two F substituents.

In some embodiments, Ring A has a CH 3 substituent and two F substituents with two F at same position.

In some embodiments, Ring A has an OFI substituent.

In some embodiments, Ring A has an OFI substituent and a Cl substituent. In some embodiments, Ring A has multiple substituents with any combination of the above substituents.

With respect to Formula 1, in some embodiments Ring B is: optionally substituted 6-oxo-5- (piperidin-l-yl)-l,6-dihydropyrazin-2-yl, optionally substituted 6-oxo-5-(pyrrolidin-l-yl)-l,6- dihydropyrazin-2-yl, optionally substituted 5-(hexahydropyrrolo[3,4-c]pyrrol-2(lH)-yl)-6-oxo-l,6- dihydropyrazin-2-yl, optionally substituted 5-(3,6-diazabicyclo[3.2.0] heptan-6-yl)-6-oxo-l,6- dihydropyrazin-2-yl, optionally substituted 6-oxo-5-(2-oxa-8-azaspiro[4.5]decan-8-yl)-l,6- dihydropyrazin-2-yl, optionally substituted 6-oxo-5-(piperidin-4-ylamino)-l,6-dihydropyrazin-2-yl, 6- oxo-5-(spiro[bicyclo[3.1.0] hexane-3,4'-piperidin]- -yl)-l,6-dihydropyrazin-2-yl, optionally substituted 6-oxo-5-(8-azaspiro[4.5]decan-8-yl)-l,6-dihydropyrazin-2-yl, optionally substituted 4- oxo-6-(piperidin-l-yl)-4,5-dihydro-lH-pyrazolo[3,4-d] pyrimidin-3-yl, 4-oxo-6-(pyrrolidin-l-yl)-4,5- dihydro-lH-pyrazolo[3,4-d]pyrimidin-3-yl, optionally substituted 4-oxo-2-(piperidin-l-yl)-3,4- dihydroquinazolin-5-yl, optionally substituted 4-oxo-2-(piperidin-l-yl)-3,4-dihydropyrido[3,4- d] pyrimidin-5-yl, optionally substituted 7-oxo-2-(piperidin-l-yl)-7,8-dihydropyrido[2,3-d] pyrimidin-5- yl, optionally substituted 5-(piperidin-l-yl)-lH-pyrazolo[4,3-d]thiazol-3-yl, optionally substituted 7- oxo-6-(piperidin-4-yl)-6,7-dihydro-lH-pyrazolo[4,3-d] pyrimidin-3-yl, optionally substituted 6-oxo-8- (piperidin-l-yl)-6,7-dihydro-lH-purin-2-yl, optionally substituted 8-(piperidin-l-yl)-7H-purin-2-yl, optionally substituted 6-oxo-2-(pyrrolidin-l-yl)-l,6-dihydropyrimidin-5-yl, optionally substituted 6- oxo-2-(piperidin-l-yl)-l,6-dihydropyrimidin-5-yl / optionally substituted 6-oxo-2-(2-oxa-8- azaspiro[4.5]decan-8-yl)-l,6-dihydropyrimidin-5-yl, optionally substituted 2-(3,6- diazabicyclo[3.2.0]heptan-6-yl)-6-oxo-l,6-dihydropyrimidin-5 -yl, optionally substituted 2- (hexahydropyrrolo[3,4-c] pyrrol-2(lH)-yl)-6-oxo-l / 6-dihydropyrimidin-5-yl / optionally substituted 2- (3-azabicyclo[3.2.0]heptan-3-yl)-6-oxo-l,6-dihydropyrimidin- 5-yl, optional ly substituted 6-oxo-2-(2- azaspiro[3.4]octan-2-yl)-l,6-dihydropyrimidin-5-yl, optionally substituted 2-(3- azabicyclo[3.1.0]hexan-3-yl)-6-oxo-l,6-dihydropyrimidin-5-yl , optionally substituted 6-oxo-2-(2- azaspiro[3.4]octan-2-yl)-l,6-dihydropyrimidin-5-yl, optionally substituted 5-oxo-6-(2-oxa-8- azaspiro .Sldecan-S-ylJ^S-dihydro-lH-pyrazololS^-bl pyrazin-S-yl, optionally substituted 5-oxo-6- (piperidin-l-ylJ^S-dihydro-lH-pyrazololS^-bl pyrazin-S-yl, optionally substituted 6-( pi peridin-l-yl)- lH-pyrazolo[3,4-d]py midin-3-yl, optionally substituted 6-oxo-5-(2-azaspiro[3.4]octan-2-yl)-l,6- dihydropyrazin-2-yl, optionally substituted l-cyclohexyl-2-oxo-l,2-dihydropyridin-4-yl, optionally substituted 5-(3-azabicyclo[3.1.0]hexan-3-yl)-6-oxo-l,6-dihydropyrazin-2 -yl, optionally substituted 6- oxo-5-(3H-spiro[benzofuran-2 / 4'-piperidin]-l'-yl)-l / 6-dihydropyrazin-2-yl / optionally substituted 4- oxo-2-(2-oxa-8-azaspiro[4.5]decan-8-yl)-4 / 7-dihydro-3H-pyrrolo[2 / 3-d]pyrimidin-5-yl / optionally substituted 5-(5,7-dihydrospiro[cyclopenta[b]pyridine-6,4'-pipendin]- -yl)-6-oxo-l,6- dihydropyrazin-2-yl, optionally substituted 5-(l 7 3-dihydrospiro[indene-2 7 4'-piperidin]-l'-yl)-6- oxo-l,6-dihydropyrazin-2-yl, optionally substituted 5-(4 7 6-dihydrospiro[cyclopenta[d]thiazole-5 7 4'- piperidin]-l'-yl)-6-oxo-l / 6-dihydropyrazin-2-yl / or optionally substituted 6-oxo-5-(spiro[indoline-2,4'- piperidin]-l'-yl)-l 7 6-dihydropyrazin-2-yl. The core structures for some suitable Ring B groups are listed in Table 1A below.

Table 1A.

In some embodiments, Ring B is unsubstituted.

In some embodiments, Ring B has a -CH 3 substituent.

In some embodiments, Ring B has a -CH 2 NH 2 substituent.

In some embodiments, Ring B has a -NH 2 substituent.

In some embodiments, Ring B has a -CH 2 CH 2 NH 2 substituent.

In some embodiments, Ring B has a l-aminopropan-2-yl substituent.

In some embodiments, Ring B has a -CN substituent.

In some embodiments, Ring B has an -F substituent.

In some embodiments, Ring B has a -Cl substituent.

In some embodiments, Ring B has a -CH2F substituent.

In some embodiments, Ring B has an -OFI substituent.

In some embodiments, Ring B has an -OCFI 3 substituent.

In some embodiments, Ring B has multiple substituents with any combination of the above substituents.

With respect to Formula 1, in some embodiments, X is S, O, NR A , CFIR A , SO, SO 2 , CO, or a bond.

In some embodiments, X is S. In some embodiments, X is a bond. In some embodiments, X is O. In some embodiments, X is N H. In some embodiments, X is— CH(CH 3 ). In some embodiments, X is CH 2 .

With respect to Formula 1, in some embodiments, R A is FI or C 1-6 hydrocarbyl. In some embodiments, R A is FI. In some embodiments, R A is CFI3.

In Appendix A below, various possibilities for Ring A are depicted. Ring A could be any core structure in any of these depicted possibilities, wherein these core structures are optionally substituted.

Appendix A:

Phenyl,

2.3-dichlorophenyl,

naphthalen-l-yl,

2-(trifluoromethyl) phenyl,

2-(trifluoromethyl)pyridin-3-yl,

5-amino-2-chlorophenyl,

5-amino-2-chloropyridin-3-yl,

3-amino-2-chlorophenyl,

2-amino-3-chloropyridin-4-yl,

2-chloro-3-methoxy phenyl,

3-chloro-2-methoxypyridin-4-yl,

3-fluoro-lH-indol-4-yl,

3,3-difluoro-2-oxoindolin-4-yl,

1-acetyl-3,3-difluoroindolin-4-yl,

2-chloro-3-(4-hydroxy-l,5,5-trimethyl-2-oxo-2,5-dihydro-lH-p yrrole-3-carboxamido) phenyl, 2-chloro-3-(2-hydroxy-4-oxo-4H-pyrido[l,2-a]pyrimidine-3-car boxamido)phenyl,

2-chloro-3-(2-hydroxy-4-oxo-4H-pyrazino[l,2-a] pyrimidine-3-carboxamido)phenyl,

2-chloro-3-(7-hydroxy-5-oxo-5H-thiazolo[3,2-a]pyrimidine- 6-carboxamido)phenyl,

2-chloro-3-(7-hydroxy-5-oxo-l,5-dihydroimidazo[l,2-a] pyrimidine-6-carboxamido)phenyl,

2-chloro-3-(2-hydroxy-4-oxo-6,7,8,9-tetrahydro-4H-pyrido[l,2 -a] pyrimidine-3-carboxamido)phenyl,

2.3-dichloropyridin-4-yl,

2.3-difluorophenyl,

3-chloro-2-fluoropyridin-4-yl,

2.3-difluoropyridin-4-yl,

2-chloro-3-methy I phenyl,

3-chloro-2-methylpyridin-4-yl,

3.3-difluoro-l-methyl-2-oxoindolin-4-yl, 3-chloro-l-methyl-2-oxo-l,2-dihydropyridin-4-yl, or

2-chloro-3-fluorophenyl.

In Appendix B below, various possibilities for Ring B are depicted. Ring B could be any core structure in any of these depicted possibilities, wherein these core structures are optionally substituted.

Appendix B:

5-(4-(aminomethyl)-4-methylpiperidin-l-yl)-6-oxo-l,6-dihydro pyrazin-2-yl,

5-(3-(aminomethyl)-3-methylpyrrolidin-l-yl)-6-oxo-l,6-dihydr opyrazin-2-yl,

5-(hexahydropyrrolo[3,4-c] pyrrol-2(lH)-yl)-6-oxo-l,6-dihydropyrazin-2-yl,

5-(3,6-diazabicyclo[3.2.0]heptan-6-yl)-6-oxo-l,6-dihydrop yrazin-2-yl,

(S)-5-(4-amino-2-oxa-8-azaspiro[4.5]decan-8-yl)-6-oxo-l,6-di hydropyrazin-2-yl,

6-oxo-5-(piperidin-4-ylamino)-l,6-dihydropyrazin-2-yl,

5-(2-aminospiro[bicyclo[3.1.0] hexane-3,4'-piperidin]-l'-yl)-6-oxo-l,6-dihydropyrazin-2-yl,

5-((lR,3R)-l-amino-3-methyl-8-azaspiro[4.5]decan-8-yl)-6-oxo -l,6-dihydropyrazin-2-yl,

(6-(4-(aminomethyl)-4-methylpiperidin-l-yl)-4-oxo-4,5-dih ydro-lH-pyrazolo[3,4-d] pyrimidin-3- yl)amino,

6-(3-(aminomethyl)-3-methylpyrrolidin-l-yl)-4-oxo-4,5-dihydr o-lH-pyrazolo[3,4-d]pyrimidin-3-yl,

7-amino-2-(4-(aminomethyl)-4-methylpiperidin-l-yl)-4-oxo- 3,4-dihydroquinazolin-5-yl,

2-(4-(aminomethyl)-4-methylpiperidin-l-yl)-4-oxo-3,4-dihy dropyrido[3,4-d]pyrimidin-5-yl,

2-(4-(a minomethy l)-4-methyl piperidin-l-y l)-7-oxo-7,8-dihyd ropy rido[2, 3-d] pyrimidin-5-yl,

5-(4-(aminomethyl)-4-methylpiperidin-l-yl)-lH-pyrazolo[4,3-d ]thiazol-3-yl,

6-(l-(l-aminopropan-2-yl)piperidin-4-yl)-7-oxo-6,7-dihydro-l H-pyrazolo[4,3-d]pyrimidin-3-yl,

8-(4-(a minomethy l)-4-methyl piperidin-l-y l)-6-oxo-6,7-dihydro-lH-purin-2-y I,

6-a mino-8-(4-(a minomethy l)-4-methy I piperidin-l-y l)-7H-purin-2-y I,

4-a mino-2-(3-(a minomethy l)-3-methy I pyrrolidin-l-yl)-6-oxo-l,6-dihydropyrimidin-5-yl,

2-(4-(a minomethy l)-4-methyl piperidin-l-y l)-4-cyano-l-methyl-6-oxo-l,6-dihydropyrimidin-5-yl,

4-amino-2-(4-amino-2-oxa-8-azaspiro[4.5]decan-8-yl)-6-oxo -l,6-dihydropyrimidin-5-yl,

4-amino-2-(3,6-diazabicyclo[3.2.0] heptan-6-yl)-6-oxo-l,6-dihydropyrimidin-5-yl,

4-amino-2-(hexahydropyrrolo[3,4-c] pyrrol-2(lH)-yl)-6-oxo-l,6-dihydropyrimidin-5-yl, 4-amino-2-(4-(aminomethyl)-4-methylpiperidin-l-yl)-6-oxo-l,6 -dihydrc>pyrimidin-5-yl,

4-amino-2-(6-amino-3-azabicyclo[3.2.0] heptan-3-yl)-6-oxo-l,6-dihydrc>pyrimidin-5-yl,

4-amino-2-(6-amino-2-azaspiro[3.4]octan-2-yl)-l-methyl-6-oxo -l / 6-dihydropyrimidin-5-yl /

4-amino-2-(6-amino-3-azabicyclo[3.1.0] hexan-3-yl)-6-oxo-l,6-dihydropyrimidin-5-yl,

4-amino-2-(6-amino-2-azaspiro[3.4]octan-2-yl)-6-oxo-l / 6-dihydropyrimidin-5-yl /

5-(4-(aminomethyl)-4-methylpiperidin-l-yl)-l-methyl-6-oxo-l, 6-dihydropyrazin-2-yl,

5-(4-amino-2-oxa-8-azaspiro[4.5]decan-8-yl)-6-oxo-l / 6-dihydropyrazin-2-yl /

(S)-5-(4-amino-2-oxa-8-azaspiro[4.5]decan-8-yl)-l-methyl- 6-oxo-l,6-dihydrc>pyrazin-2-yl,

5-(2-aminospiro[bicyclo[3.1.0] hexane-3 / 4'-piperidin]-l'-yl)-l-methyl-6-oxo-l / 6-dihydropyrazin-2-yl /

5-((3S,4S)-4-amino-3-methyl-2-oxa-8-azaspirc>[4.5]deca n-8-yl)-6-oxo-l,6-dihydrc>pyrazin-2-yl,

5-((3S,4S)-4-amino-3-methyl-2-oxa-8-azaspirc>[4.5]deca n-8-yl)-l-methyl-6-oxo-l,6- dihydropyrazin-2- yi,

5-((lR,3R)-l-amino-3-methyl-8-azaspiro[4.5]decan-8-yl)-l-met hyl-6-oxo-l,6-dihydrc>pyrazin-2-yl,

5-(4-(aminomethyl)-4-methylpiperidin-l-yl)-6-oxo-l,6-dihy dropyrazin-2-yl,

6-(4-(aminomethyl)-4-methylpiperidin-l-yl)-4-oxo-4,5-dihydro -lH-pyrazolo[3,4-d] pyrimidin-3-yl,

6-(4-(aminomethyl)-4-methylpiperidin-l-yl)-4-methyl-5-oxo -4,5-dihydro-lH-pyrazolo[3,4-b] pyrazin-

3-yl,

6-(4-(aminomethyl)-4-methylpiperidin-l-yl)-5-oxo-4,5-dihydro -lH-pyrazolo[3,4-b] pyrazin-3-yl,

6-(3-(aminomethyl)-3-methylpyrrc>lidin-l-yl)-5-methyl- 4-oxo-4,5-dihydrc>-lH-pyrazolo[3,4- d] pyrimidin-3-yl,

6-(4-(aminomethyl)-4-methylpiperidin-l-yl)-5-methyl-4-oxo-4, 5-dihydro-lH-pyrazolo[3,4- d] py rimidin-3-yl,

6-(4-(aminomethyl)-4-methylpiperidin-l-yl)-lH-pyrazolo[3,4-d ]pyrimidin-3-yl,

4-amino-2-(hexahydropyrrolo[3 / 4-c] pyrrol-2(lH)-yl)-l-methyl-6-oxo-l / 6-dihydropyrimidin-5-yl /

4-amino-2-((3S / 4S)-4-amino-3-methyl-2-oxa-8-azaspiro[4.5]decan-8-yl)- 6-oxo-l / 6-dihydropyrimidin-

5-yl,

4-amino-2-((3S,4S)-4-amino-3-methyl-2-oxa-8-azaspirc>[4.5 ]decan-8-yl)-l-methyl-6-oxo-l,6- dihydropyrimidin-5-yl,

4-amino-2-(6-amino-3-azabicyclo[3.1.0] hexan-3-yl)-l-methyl-6-oxo-l,6-dihydrc>pyrimidin-5-yl, 4-amino-2-(6-amino-3-azabicyclo[3.2.0] heptan-3-yl)-l-methyl-6-oxo-l,6-dihydropyrimidin-5-yl,

5-(4-(aminomethyl)-4-fluoropiperidin-l-yl)-6-oxo-l,6-dihy dropyrazin-2-yl,

5-(4-(aminomethyl)-4-hydroxypiperidin-l-yl)-6-oxo-l,6-dih ydropyrazin-2-yl,

(R)-5-(6-amino-2-azaspiro[3.4]octan-2-yl)-6-oxo-l,6-dihyd ropyrazin-2-yl,

(S)-5-(6-amino-2-azaspiro[3.4]octan-2-yl)-6-oxo-l,6-dihyd ropyrazin-2-yl,

1-(3-aminocyclohexyl)-2-oxo-l,2-dihydropyridin-4-yl,

(R)-5-(4-amino-2-oxa-8-azaspiro[4.5]decan-8-yl)-6-oxo-l,6-di hydropyrazin-2-yl,

5-(4-amino-4-(fluoromethyl)piperidin-l-yl)-6-oxo-l,6-dihydro pyrazin-2-yl,

5-((3S,4S)-4-amino-3-methyl-2-oxa-8-azaspiro[4.5]decan-8-yl) -6-oxo-l,6-dihydropyrazin-2-yl, (R)-5-(l-amino-8-azaspiro[4.5]decan-8-yl)-6-oxo-l,6-dihydrop yrazin-2-yl,

5-(6-amino-3-azabicyclo[3.1.0]hexan-3-yl)-6-oxo-l,6-dihyd ropyrazin-2-yl,

(R)-5-(3-amino-3H-spiro[benzofuran-2,4'-piperidin]- -yl)-6-oxo-l,6-dihydropyrazin-2-yl,

2-((3S,4S)-4-amino-3-methyl-2-oxa-8-azaspiro[4.5]decan-8-yl) -3-methyl-4-oxo-4,7-dihydro-3H- pyrrolo[2,3-d]pyrimidin-5-yl,

2-((3S,4S)-4-amino-3-methyl-2-oxa-8-azaspiro[4.5]decan-8- yl)-4-oxo-4,7-dihydro-3H-pyrrolo[2,3- d] py rimidin-5-yl,

(R)-5-( 1-ami no-3, 3-difluoro-8-azaspiro[4.5]decan-8-yl)-6-oxo-l,6-dihyd ropy razin-2-y I,

5-((lR)-l-amino-3-fluoro-8-azaspiro[4.5]decan-8-yl)-6-oxo -l,6-dihydropyrazin-2-yl,

(S)-5-(5-a mino-5, 7-dihydrospiro[cyclopenta[b] pyridine-6, 4'-piperidin]- -yl)-6-oxo-l, 6- dihydropyrazin-2-yl,

(S)-5-(l-amino-l,3-dihydrospiro[indene-2,4'-piperidin]- -yl)-6-oxo-l,6-dihydropyrazin-2-yl,

(S)-5-(4-amino-2-chloro-4,6-dihydrospiro[cyclopenta[d]thi azole-5,4'-piperidin]- -yl)-6-oxo-l,6- dihydropyrazin-2-yl,

(R)-5-(3-aminospiro[indoline-2,4'-piperidin]- -yl)-6-oxo-l,6-dihydropyrazin-2-yl, or

(S)-5-(l-amino-4-methoxy-l,3-dihydrospiro[indene-2,4'-pip eridin]- -yl)-6-oxo-l,6-dihydropyrazin-2- yi-

Some embodiments include one of the compounds in Table 1, wherein any of the compounds in Table 1 below may be optionally substituted. Table 1

An example, not as an attempt to limit the scope of the disclosure, of a useful composition for a dosage form containing about 10-1000 mg of compound 83 is shown in Table IB below:

Table IB. Example of dosage form of compound 83

A pharmaceutical composition comprising a compound of Formula 1 may be adapted for oral, or parental, such as intravenous, intramuscular, topical, intraperitoneal, nasal, buccal, sublingual, or subcutaneous administration, or for administration via respiratory tract in the form of, for example, an aerosol or an air-suspended fine powder. The dosage of a compound of Formula 1 may vary depending on the route of administration, body weight, age, the type and condition of the disease being treated. A pharmaceutical composition provided herein may optionally comprise two or more compounds of the Formula 1 without an additional therapeutic agent, or may comprise an additional therapeutic agent (i.e., a therapeutic agent other than a compound provided herein). For example, the subject compounds can be administered simultaneously, sequentially, or separately in combination with at least one other therapeutic agent. The other therapeutic agent can be a small molecule, an antibody-drug conjugate, or a biologic. Therapeutic agents suitable for combination with a subject compound include, but are not limited to antibiotics, antiemetic agents, antidepressants, and antifungal agents, anti-inflammatory agents, antiviral agents, and anticancer agents that are known in the art. In some embodiments, the other therapeutic agents are chemotherapy agents, for example, mitotic inhibitors such as a taxane, a vinca alkaloid, paclitaxel; or tyrosine kinase inhibitors, for example Erlotinib; ALK inhibitors such as Crizotinib; BRAF inhibitors such as Vemurafanib; MEK inhibitors such as trametinib; or other anticancer agents, i.e. cisplatin, flutamide, gemcitabine, CTLA-4 inhibitors, PD-1 inhibitors and PD-L1 inhibitors. Such combination may offer significant advantages, including synergistic activity, in therapy. The pharmaceutical composition may be used for the treatment of cancer, autoimmune diseases, inflammatory diseases, autoinflammatory conditions, and other SHP2 mediated disorders in patients. The term "patient" herein means a mammal (e.g., a human or an animal). In some embodiments, the patient has cancer.

The pharmaceutical composition described herein can be prepared by combining a compound of Formula 1 with at least one pharmaceutical acceptable inert ingredient, such as a carrier, excipient, filler, lubricant, flavoring agent, buffer, etc., selected on the basis of the chosen route of administration and standard pharmaceutical practice as described, for example, in Remington's Pharmaceutical Sciences, 2005, the disclosure of which is hereby incorporated herein by reference, in its entirety. The relative proportions of active ingredient and carrier may be determined, for example, by the solubility and chemical nature of the compounds, chosen route of administration and standard pharmaceutical practice.

Some embodiments include a method of treating a SHP2 mediated disease or disorder comprising administering a therapeutically effective amount of a compound of Formula 1, or any compound described herein, or a pharmaceutically acceptable salt thereof ("subject compound"), or a pharmaceutical composition comprising a subject compound to a patient in need thereof. The term a "therapeutically effective amount" herein refers to an amount of a subject compound, or a pharmaceutical composition containing a subject compound, sufficient to be effective in inhibiting SH P2 and thus providing a benefit in the treatment of cancer, autoimmune diseases, inflammatory diseases, autoinflammatory conditions, and other SH P2 mediated disorders in patients, such as to delay or minimize symptoms associated with cancer, autoimmune, inflammatory diseases, and autoinflammatory conditions, or to ameliorate a disease or infection or cause thereof, or to prevent the further development of a disorder, or reducing the severity of symptoms that are otherwise expected to develop without treatment.

EXPERIMENTAL:

GENERAL SYNTHETIC METHODS: The compounds of the present invention, or their pharmaceutically acceptable salts, can be synthesized using the methods described below in schemes 1-6. It will be understood that all proposed reaction conditions, including choice of solvent, reaction atmosphere, reaction temperature, duration of the experiment and work up procedures, are chosen to be the conditions standard for that reaction, which should be readily recognized by one skilled in the art. It is understood by one skilled in the art of organic synthesis that optimum reaction conditions may vary with the particular reactants or solvents used, but such conditions can be determined by the person skilled in the art, using routine optimization procedures. Additionally, one skilled in the art will recognize that in many cases, these compounds will be mixtures of stereoisomers that may be separated at various stages of the synthetic schemes using conventional techniques, such as, but limited to, crystallization, normal-phase chromatography, reversed phase chromatography and chiral chromatography, to afford single enantiomers. For all the protection and deprotection methods, see Philip J. Kocienski, in "Protecting Groups", Georg Thieme Verlag Stuttgart, New York, 1994 and, Theodora W. Greene and Peter G. M. Wuts in "Protective Groups in Organic Synthesis", Wiley Interscience, 3rd Edition 1999. The schemes 1-6 are representative of methods useful in synthesizing the compounds of the present invention. They are not to constrain the scope of the invention in any way.

Scheme 1

int-1 Formula 1-1 Formula 1 -2 Scheme 1 illustrates a method for preparing compounds of Formula 1. In which L is S, O, N or a bond; Ri is H, C1-C6 alkyl, NH2, or CN; R 2 is aryl, heterocycloalkyl or heteroaryl; R3 is C1-C6 alkyl, ORi; R5 is alkyl, H; R6 is alkyl, H; R5 and R6 together with the atom or atoms to which they are attached, can combine to form a monocyclic or polycyclic C3-C12 cycloalkyl or heterocycle.

Compound int-1 is treated with aryl or alkyl boronic acids or esters or salts (where L is a bond) under suitable metal catalysts (such Pd2(dba)3, or the like), suitable ligands (such as dppf, or the like), suitable bases (such as Cs 2 C0 3 , or the like), suitable solvents (such as DMF, or the like) to provide a product of int-2. In other cases, where the L of Formula 1 is O, N or S, compound int-1 was reacted with corresponding phenols, thiophenols, thioalcohols or amines under suitable metal catalysts (such as Cul, Pd2(dba) 3 ), suitable ligands (such as TMEDA, XPFIOS, Xantphos, or the like), suitable salts or bases (such as Cs 2 C0 3 , K 3 P0 4 or the like), suitable solvents (such as DMF, dioxane or the like) to provide a product of int-2. The reaction temperature is ranged from 80°C to 140°C, and the reaction takes 1 - 24 hours to complete. Compound int-2 reacted with amines with or without bases (such as DIPEA), in suitable solvents (such as DMF, NMP or the like) under the temperature range from 80°C to 140°C to offer compound of Formula 1. If desired, further transformations may be performed to provide a product of Formula 1-2. For example, the compound of Formula 1-1 wherein R 3 = OFI may be subjected to an alkylation reaction to provide a compound of Formula 1-2.

Scheme 2

3

Scheme 2 illustrates a method for preparing compounds of Formula 1-3. Compound lnt-1 was first halogenated to provide intermediate lnt-2. The activated chlorine in lnt-2 was displaced by an amine to afford lnt-4. lnt-4 reacted with an aryl amine or phenol to provide target compounds of Formula 1-3 (R 3 = FI).

For the compounds of Formula 1-3 where R 3 was OFI, lnt-6 was regioselectively hydrolyzed to provide lnt-3, and then followed by substitutions and couplings to offer target compounds.

Deprotection steps can be incorporated either before or after the coupling reactions. Scheme 3

Formula 1-3

Additionally, the compounds of Formula 1-3 can be synthesized using an alternative way, as illustrated in Scheme 3. The order of reaction steps may alter. Coupling reactions may occur before the amine displacement.

Scheme 4

lnt-1 lnt-2 Formula 1-4

Scheme 4 illustrates a method for preparing compounds of Formula I-4. Int-l was reacted with an aryl amine or phenol in the presence of a suitable metal catalyst (such as Cul, or the like), a suitable ligand (such as TMEDA, TMFID, or the like), a suitable salt (such as K3PO4, or the like) and a suitable solvent (such as dioxane or the like). The reaction proceeds at a temperature ranged 80 °C to 140 °C, with the reaction time from 1 - 24 hours lnt-2 reacted with an amine in the presence of a suitable coupling reagent (such as BOP-CI, or the like), a suitable base (such DIEPA, DBU, or the like), and a suitable solvent (such as DMF, TH F or the like). The reaction proceeded at temperature 80°C to 130°C with 1 - 24 hours to finish.

Scheme 5

lnt-1 lnt-2 Formula 1-4

Alternatively, as illustrated in Scheme 5, the order of reactions can be modified to change the overall synthesis to allow for variations at different positions of the molecule at different stages of the preparation. For example, in Scheme 5, compound of Formula lnt-1 is activated and reacted with an amine to provide lnt-2 first, and then followed with the coupling reaction, to provide compound of Formula 1-4.

Scheme 6

Formula 1-5 (R 3 = H) Formula 1-5

Scheme 6 illustrates a method for preparing compounds of Formula I-5.

Bromine of lnt-1 was displaced with methoxy group to afford lnt-2, and then lnt-2 was halogenated with N BS or N IS to offer lnt-3. From lnt-3, after the substitution, coupling and deprotection compounds of formula 1-5 (R3 = H) were synthesized. The compound of Formula 1-5 wherein R 3 = H may be subjected to an alkylation reaction to provide another series compound of Formula 1-5.

EXPERIMENTAL PROCEDURES AND EXAMPES

Experiments were generally carried out under inert atmosphere (nitrogen or argon), particularly in cases where oxygen-or moisture-sensitive reagents or intermediates were employed. Commercial solvents and reagents were generally used without further purification, including anhydrous solvents where appropriate. Products were generally dried under vacuum before being carried on to further reactions or submitted for biological testing. Mass spectrometry data is reported from liquid chromatography-mass spectrometry (LCMS) instrumentation. Mass spectra, MS (m/z), were recorded using either electrospray ionization (ESI) or atmospheric pressure chemical ionization (APCI). Where relevant and unless otherwise stated the m/z data provided are for isotopes 19F, 35CI, 79Br and 1271. Chemical shifts for nuclear magnetic resonance (NMR) data are expressed in parts per million (ppm, d) referenced to residual peaks from the deuterated solvents employed, using conventional abbreviations for designation of major peaks: e.g. s, singlet; d, doublet; t, triplet; q, quartet; m, multiplet; br, broad. The following abbreviations have been used for common solvents: CDCI 3 , deuterochloroform; d6-DMSO, derterodimethylsulphoxide; and CD 3 OD, deuteromethanol.

In general, reactions were followed by thin layer chromatography (TLC) and/or liquid chromatography-mass spectrometry (LCMS) and subjected to work-up when appropriate.

Purification was carried out by chromatographic and/or H PLC.

Unless noted otherwise, all reactants were obtained commercially.

Example 1

Preparation of 2-(4-(aminomethyl)-4-methylpiperidin-l-yl)-5-(2,3-dichloroph enyl)-l-methyl-6-oxo- l,6-dihydropyrimidine-4-carbonitrile

NH 2

Step 1: preparation of tert-butyl ((l-(4,5-dichloro-l-methyl-6-oxo-l,6-dihydropyrimidin-2-yl)- 4- methylpiperidin-4-yl)methyl)carbamate (3)

To a mixture of compound 1 (1.4 g, 1.0 eq) and compound 2 (1.5 g, 1.0 eq) in DMF (25 mL) was added DIEA (3.2 mL, 2.0 eq). The mixture was degassed and protected with nitrogen. The reaction was stirred at 80 °C for 2 h. EtOAc (150 mL) was added to the mixture under r.t. After standard work up procedure, the residue was purified by column chromatography to give compound 3 as a white solid (1.9 g, 71%).

Step 2: preparation of tert-butyl ((l-(5-chloro-4-cyano-l-methyl-6-oxo-l,6-dihydropyrimidin-2- yl)-4- methylpiperidin-4-yl)methyl)carbamate (4)

A mixture of compound 3 (200 mg, 1.0 eq), Zn(CN) 2 (57.9 g, 1.0 eq) and Pd(PPh 3 ) 4 (56 mg, 0.1 eq) in DMF was stirred at 150 °C for 2 h, and the mixture was cooled to rt. EtOAc was added, after standard work up procedure the residue was purified by prep-TLC to give compound 4 as a white solid (60 mg, 31%).

Step 3: preparation of tert-butyl ((l-(4-cyano-5-(2,3-dichlorophenyl)-l-methyl-6-oxo-l,6- dihydropyrimidin-2-yl)-4-methylpiperidin-4-yl)methyl)carbama te (6)

To a mixture of compound 4 (60 mg, 1.0 eq), K2CO3 (42 mg, 2 eq) and compound 5 (86 mg, 3.0 eq) in DMF (10 mL) was added Pd(dppf)Cl2 (11 mg, 0.1 eq). The resulting mixture was stirred at 95 °C overnight. The mixture was diluted with EtOAc. After standard work up procedure, the residue was purified by prep-TLC to give compound 6 as a white solid (20 mg, 26%).

Step 4: preparation of 2-(4-(aminomethyl)-4-methylpiperidin-l-yl)-5-(2,3-dichloroph enyl)-l-methyl- 6-oxo-l,6-dihydropyrimidine-4-carbonitrile

HCI/dioxane (0.5 mL, 10.0 eq) was added to a solution of compound 6 (19 mg, 1.0 eq) in DCM (5.0 mL), and then the reaction was stirred at rt for 1 h. The mixture was concentrated and neutralized with ammonium hydroxide, purified by prep-H PLC to give the desired compound as a white solid (2.5 mg, 17%). LC-MS: [M+H] + : 406.1. X H NMR (400 MHz, CDBOD) <5 7.65 (dd, J = 8.0, 1.6 Hz, 1H), 7.40 (t, J

= 8.0 Hz, 1H), 7.33 (dd, J = 8.0, 1.2 Hz, 1H), 3.55 - 3.48 (m, 4H), 3.34 - 3.31 (m, 2H), 2.66 (s, 1H), 1.71 - 1.66 (m, 2H), 1.55 - 1.52 (m, 2H), 1.10 (s, 3H).

Example 2

Preparation of 3-(4-(aminomethyl)-4-methylpiperidin-l-yl)-6-(2,3-dichloroph enyl) pyrazin-2(lH)-one

Step 1: Preparation of 3-(benzyloxy)-5-bromopyrazin-2-amine (2)

To a solution of benzyl alcohol (13.5 g, 1.05 eq) in TH F (300 mL) was added NaH (60%, 5.7 g, 1.2 eq) at rt. The mixture was stirred at rt for 30 min. Compound 1 (30.0 g, 118 mmol, 1.0 eq) was added to the mixture. The mixture was heated to 70 °C and stirred overnight. The reaction was quenched by water, worked up under standard operation to give compound 2 as a yellow solid (25.0 g, 75%).

Step 2: preparation of 3-(benzyloxy)-5-bromo-2-chloropyrazine (4)

To a solution of compound 2 (9.0 g, 1.0 eq) in TH F (30 mL) was added CuCh (12.96 g, 3.0 eq) and CuCI (5.75 g, 2.0 eq) at rt. Compound 3 (12.9 mL, 3.0 eq) was added dropwise to the mixture 10 min later. The reaction was worked up under standard operation to give compound 4 as a yellow oil (4.0 g, 41%). Step 3: preparation of 3-(benzyloxy)-2-chloro-5-(2,3-dichlorophenyl)pyrazine (6)

To a solution of compound 4 (3.0 g, 1.0 eq) in DMF (30 mL) was added K2CO3 (4.14 g, 3.0 eq), compound 5 (1.9 g, 1.0 eq) and Pd(dppf)Cl2 (0.73 g, 0.1 eq). The resulting mixture was stirred at 95 °C under N2 atmosphere overnight. The reaction was quenched by water and worked up under standard operation to give compound 6 as a yellow solid (1.5 g, 40%).

Step 4: preparation of tert-butyl ((l-(3-(benzyloxy)-5-(2,3-dichlorophenyl)pyrazin-2-yl)-4- methylpiperidin-4-yl)methyl)carbamate (8)

To a solution of compound 6 (500 mg, 1.0 eq) in DMF (5 mL) were added compound 7 (342 mg, 1.1 eq) and K2CO3 (660 mg, 3.5 eq). The mixture was stirred at 60 °C under N2 atmosphere for 5 h. The reaction was quenched by water and worked up under standard operation to give compound 8 as a white solid (400 mg, 53%).

Step 5: preparation of 3-(4-(aminomethyl)-4-methylpiperidin-l-yl)-6-(2,3-dichloroph enyl)- pyrazin-2(lH)-one

To a solution of compound 8 (100 mg, 1.0 eq) in DCM (5 mL) was added BCI3 (1M in DCM, 5.0 eq). The mixture was stirred at rt for 1.5 h. The reaction was quenched by MeOH (5 mL) and the mixture was concentrated and purified by prep-HPLC to give target compound as a white solid (7 mg, 10%). LC-MS: [M+H] + = 367. X H NMR (400 MHz, DMSO-dg) <5 7.67 (d, J 7.4 Hz, 1H), 7.52 - 7.34 (m, 2H), 6.91 (s, 1H), 4.10 - 4.03 (m, 2H), 3.45 - 3.32 (m, 2H), 2.88 (d, J 6.0 Hz, 1H), 2.39 (s, 1H), 1.50 - 1.41 (m, 2H), 1.26 - 1.24 (m, 2H), 0.90 (d , J 9.2 Hz, 3H).

Example 3

Preparation of 3-(4-(aminomethyl)-4-methylpiperidin-l-yl)-6-((2,3-dichlorop henyl)thio)-l- methylpyrazin-2(lH)-one

Step 1: preparation of 3-(benzyloxy)-2-chloro-5-((2,3-dichlorophenyl)thio)pyrazine (3)

To a mixture of compound 1 (3.0 g, 1.0 eq), compound 2 (1.79 g, 1.0 eq), Pd2(dba)3 (300 mg) and Xantphos (300 mg) in dioxane (50 mL) was added DIEA (3.0 mL, 1.8 eq), and then the mixture was degassed and protected with nitrogen. EtOAc (150 mL) was added to the mixture 2 hours later. After standard work up procedure the residue was purified by column chromatography to give compound 3 as a colorless oil (2.9 g, 73%).

Step 2: preparation of 3-chloro-6-((2,3-dichlorophenyl)thio)pyrazin-2(lH)-one (4)

BCI3 (10.0 mL, 1M in DCM, 1.99 eq) was added to a solution of compound 3 (2.0 g, 1.0 eq) in DCM at rt and then the reaction was stirred at rt overnight. The reaction was quenched by sat. NaHCC>3 and stirred for 1 h. The suspension was then filtered and the solid was washed with DCM and water. The solid was dried to give the crude product 4 as a yellow solid. (3.0 g, 78%).

Step 3: preparation of 3-chloro-6-((2,3-dichlorophenyl)thio)-l-methylpyrazin-2(lH)- one (5)

Mel (1 mL) was added to a mixture of crude 4 (2.0 g, 1.0 eq) and K2CO3 (2.0 g, 5.6 eq) in DMF (10 mL) and the resulting mixture was stirred at 25 °C for 1 h. The mixture was diluted with EtOAc and then was washed with brine. The organic solution was dried over Na 2 S04 and concentrated. The residue was purified by column chromatography to give compound 5 as a white solid (505 mg, 60%). Step 4: preparation of tert-butyl ((l-(5-((2,3-dichlorophenyl)thio)-4-methyl-3-oxo-3,4- dihydropyrazin-2-yl)-4-methylpiperidin-4-yl)methyl)carbamate (7)

A mixture of compound 5 (96 mg, 1.0 eq), compound 6 (73 mg, 1.07 eq) and DIEA (0.7 mL, 1.8 eq) in NMP (2 mL) was stirred at 140 °C for 1 h. The mixture was diluted with EtOAc and then washed with brine. The organic solution was dried over Na 2 S04 and concentrated. The residue was purified by column chromatography to give compound 7 as an off-shite solid (140 mg, 91%)

Step 3: preparation of 3-(4-(aminomethyl)-4-methyl piperidin-l-yl)-6-((2,3-dichlorophenyl)thio)-l- methylpyrazin-2(lH)-one

HCI/MeOH (1.0 mL, 10.0 eq) was added to the solution of compound 7 (51 mg, 1.0 eq) in EtOAc (2.0 mL) and then the reaction was stirred at rt for 1 h. The reaction solution was kept for 2 days and the obtained suspension was filtered. The solid was washed with EtOAc and the solid was dried to give the desired compound as HCI salt, white solid (24 mg, 53%). LC-MS: [M+H] + : 413.1. X H NMR (400 MHz, CD 3 OD) d 7.45 (d, J = 8.0 Hz, 1H), 7.30-7.25 (m, 2H), 7.09 (d, J = 8.0 Hz, 1H), 4.48-4.43 (m, 2H), 3.88-3.81 (m, 2H), 3.46 (s, 3H), 2.96 (s, 2H), 1.81-1.67 (m, 4H), 1.23 (s, 3H).

The compounds in Table A below were synthesized in the similar manner using appropriate reagents and conditions. The compounds listed in Table A are merely non-limiting examples. Other subject compounds could also be made using similar methods.

Table A

Example 4

Preparation of 6-(4-(aminomethyl)-4-methyl piperidin-l-yl)-3-((2, 3-dichlorophenyl) a mino)-l, 5- dihydro-4H-pyrazolo [3, 4-d] pyrimidin-4-one

Cl

Step 1: prepa ration of 2, 4, 6-trichloropyrimidine-5-ca rba ldehyde (2) A mixture of compound 1 (25.6 g, 1.0 eq) with POCI 3 (100 mL) and DMF (30 mL) was heated at 120 °C for 15 h, and then DMF was evaporated. Ice water was added to the residue and the solid formed was collected and dried to give compound 2 (9.2 g, 21%).

Step 2: preparation of 4, 6-dichloro-lFI-pyrazolo [3, 4-d] pyrimidine (3)

To a solution of compound 2 (16.0 g, 1.0 eq) in methanol (80 mL) was added dropwise a solution of hydrazine monohydrate (4.55 g, 1.2 eq) in methanol at 0 °C, and then a solution of triethylamine (15.28 g, 2.0eq) in methanol was added dropwise at 0 °C. The mixture was stirred at 0 °C for 2 h, then evaporated in vacuo. The residue was suspended in hot isopropyl alcohol and the insoluble materials were removed by filtration. The combined filtrate was concentrated in vacuo to give the title compound 3 as a yellow solid (9.0 g, 62%).

Step 3: preparation of 6-chloro-4-methoxy-lH-pyrazolo [3, 4-d] pyrimidine (4)

To a solution of compound 3 (480 mg, 2.54 mmol, 1.0 eq) in TH F was added cesium carbonate (1.65 g, 2.0 eq) and methanol (3.0 mL), and the mixture was heated to 60 °C. The reaction was quenched with water (10 mL) and worked up under sta ndard procedure to afford compound 4 as a brown solid (468 mg, 99%).

Step 4: preparation of 6-chloro-3-iodo-4-methoxy-lH-pyrazolo [3, 4-d] pyrimidine (5)

To a solution of compound 4 (468 mg, l.Oeq) in dry DMF (12 mL) was added N- iodosuccinimide (857mg, 1.5eq) and the reaction mixture was heated to 80 °C under stirring for 3 h. The reaction was quenched with water (10 mL) and worked up under standard process to afford compound 5 as a brown solid (522 mg, 66%).

Step 5: preparation of 6-chloro-3-iodo-4-methoxy-l-((2-(trimethylsilyl) ethoxy) methyl)-lH-pyrazolo [3, 4-d] pyrimidine (6)

To a solution of compound 5 (2.6 g, 1.0 eq) in DCM (20 mL) was added DIEA (2.37 g, 2.2 eq) at 0 °C, and then SEMCI (1.67 g, 1.2 eq) was added dropwise. The reaction was quenched with water (10 mL) and worked up under standard process to afford compound 6 as a brown solid (1.55 g, 42%).

Step 6: preparation of tert-butyl ((l-(3-iodo-4-methoxy-l-((2-(trimethylsilyl) ethoxy) methyl)-lH- pyrazolo [3, 4-d] pyrimidin-6-yl)-4-methylpiperidin-4-yl) methyl) carbamate (8)

To a solution of compound 6 (520 mg, 1.0 eq) in N MP (10 mL) was added compound 7 (296 mg, 1.1 eq) at RT, and then the mixture was stirred at 75 °C for 2 h. The reaction was quenched with water (10 mL) and worked up under standard process to afford compound 8 as a white solid (450 mg, 60%).

Step 7: preparation of tert-butyl ((l-(3-((2,3-dichlorophenyl)amino)-4-methoxy-l-((2- (trimethylsilyl)ethoxy)methyl)-lH-pyrazolo[3,4-d] pyrimidin-6-yl)-4-methylpiperidin-4- yl)methyl)carbamate (10)

To a solution of compound 8 (200 mg, 1.0 eq) in toluene was added compound 9 (61.5 mg, 1.2 eq), cesium carbonate (123.7 mg, 1.2 eq) and xa ntphos (183.1 mg, 1.0 eq). After that, Pd (dba (5.0 mg) was added under the atmosphere of Ar. The mixture was stirred at 120 °C for 16 h. The reaction was quenched with water and worked up under standard process to give compound 10 as a white solid (42 mg, 20%).

Step 8: preparation of 6-(4-(aminomethyl)-4-methylpiperidin-l-yl)-3-((2, 3-dichlorophenyl) amino)-l, 5-dihydro-4H-pyrazolo [3, 4-d] pyrimidin-4-one

To a mixture of compound 10 (97.0 mg, 1.0 eq) in H 2 0 (4 mL) was added H Br (40%, 2 mL), and then the mixture was stirred at 90 °C for 16 h. The reaction was quenched by water followed by standard work up process. The crude product was purified by prep-H PLC to give desired compound, as HCOOH salt (white solid, 9.79 mg, 16%). LC-MS: [M+H] + = 422.2. X H NMR (400 MHz, CD 3 OD): <5 8.53 (brs, 1H), 8.37 (d, J = 8.8 Hz, 1H), 7.24 (t, J = 8.4 Hz, 1H), 7.05 (d, J = 8.0 Hz, 1H), 4.03 - 3.98 (m, 2H), 3.50-3.43 (m, 2H), 2.87 (s, 2H), 1.64 - 1.56 (m, 4H), 1.17 (s, 3H).

The compounds in Table B below were synthesized in the similar manner using appropriate reagents and conditions. The compounds listed in Table B are merely non-limiting examples. Other subject compounds could also be made using similar methods.

Table B

Example 5

Preparation of (l-(3-((2,3-dichlorophenyl)thio)-lH-pyrazolo[3,4-d] pyrimidin-6-yl)-4- methylpiperidin-4-yl)methanamine

Step 1: preparation of 6-chloro-3-iodo-lH-pyrazolo[3,4-d] pyrimidine (2)

To a solution of compound 1 (780 mg, 1.0 eq) in DMF was added NIS (1300 mg, 1.2 eq) in portion at rt. The mixture was heated to 80 °C and stirred for 4 h. The reaction was quenched by water and worked up under standard operation to afford compound 2 as yellow solid (560 mg, 68%). Step 2: preparation of 6-chloro-3-((2,3-dichlorophenyl)thio)-lH-pyrazolo[3,4-d] pyrimidine (4)

To a solution of compound 2 (280 mg, 1.0 eq) in dioxane were added Pd (dba (46 mg, 0.05 eq), XantPhos (58 mg, 0.1 eq) and DIEA (200 mg, 1.5 eq) at rt. The mixture was heated to 90 °C and stirred for 14 h. The reaction was quenched by water and worked up under standard operation to afford compound 4 as yellow solid (140 mg, 48%).

Step 3: preparation of tert-butyl ((l-(3-((2,3-dichlorophenyl)thio)-lH-pyrazolo[3,4-d] pyrimidin-6-yl)- 4-methylpiperidin-4-yl)methyl)carbamate (6) To a solution of compound 4 (145mg, 1.0 eq) in DIEA was added compound 5 (300 mg, 3.0 eq) at rt. The mixture was heated at 140 °C for 24 h. The reaction was quenched by water and worked up under standard operation to give compound 6 as a yellow oil (110 mg, 48%).

Step 4: preparation of (l-(3-((2,3-dichlorophenyl)thio)-lH-pyrazolo[3,4-d]pyrimidin -6-yl)-4- methylpiperidin-4-yl)methanamine

To a solution of compound 6 (110 mg, 1.0 eq) in DCM was added TFA (1 mL). The mixture was stirred at rt for 2 h. The mixture was concentrated and neutralized with ammonium hydroxide (pH = 7-8), purified by prep-H PLC to give the target compound as a white solid (HCOOH salt, 6 mg, 7%). LC- MS: [M+H] + = 423.2. X H NMR (400 MHz, CD 3 OD) <5 8.54 (s, 1H), 7.41 (dd, J = 8.0, 1.2 Hz, 1H), 7.14 (t, J

= 8.0 Hz, 1H), 6.92 (dd, J = 8.0, 1.2 Hz, 1H), 4.62 (br, 1H), 4.42 - 4.36 (m, 2H), 3.61 - 3.54 (m, 2H), 2.89 (s, 2H), 1.60 - 1.50 (m, 4H), 1.20 (s, 3H).

Example 6

Preparation of 6-amino-5-((2,3-dichlorophenyl)thio)-2-(hexahydropyrrolo[3,4 -c] pyrrol-2(lH)-yl)-3- methylpyrimidin-4(3H)-one

To a solution of compound 1 (31.08 g, 1.0 eq) in H MDS (150 mL) was added cone. H2SO4 (0.6 mL) at room temperature. The mixture was heated to reflux for 3 h. Then the mixture was concentrated in vacuo. The residue was dissolved in DMF (150 mL) and Mel (115.8 g, 3.4 eq) was added at rt. The mixture was stirred at rt for 16 h. The reaction was quenched by water and worked up under standard operation to give compound 2 as a white solid (16 g, 47%).

Step 2: preparation of 6-amino-5-bromo-3-methylpyrimidine-2,4(lH,3H)-dione (3)

To a solution of compound 2 (5.0 g, 1.0 eq) in DMF (50 mL) was added N BS (7.9 g, 1.25 eq) and the mixture was stirred at rt overnight. The reaction was quenched by water and worked up under standard operation to give compound 3 as a white solid (4.7 g, 60%).

Step 3: preparation of 6-amino-5-((2,3-dichlorophenyl)thio)-3-methylpyrimidine-2,4( lH,3H)-dione (5) To a solution of compound 3 (500 mg, 1.0 eq) in dioxane (10 mL) was added compound 4 (813 mg, 2.0 eq), K3PO4 (1445 mg, 3.0 eq), TMEDA (105 mg, 0.4 eq) and Cul (86 mg, 0.2 eq). The resulting mixture was stirred at 100 °C under N 2 atmosphere for 2 h. The reaction was quenched by water and worked up under standard operation to give compound 5 as a brown solid (210 mg, 29%).

Step 4: preparation of tert-butyl 5-(4-amino-5-((2,3-dichlorophenyl)thio)-l-methyl-6-oxo-l,6- dihydropyrimidin-2-yl)hexahydropyrrolo[3,4-c]pyrrole-2(lH)-c arboxylate (7)

To a solution of compound 5 (60 mg, 1.0 eq) in DMF (2 mL) was added compound 6 (60 mg, 1.5 eq), BOP (250 mg, 3.0 eq) and DBU (143 mg, 5.0 eq). The reaction was quenched by water and worked up under standard operation to give compound 7 as a white solid (55 mg, 57%).

Step 5: preparation of 6-amino-5-((2,3-dichlorophenyl)thio)-2-(hexahydropyrrolo[3,4 -c]pyrrol-2(lH)- yl)-3-methylpyrimidin-4(3H)-one

A solution of compound 7 (50 mg, 1.0 eq) in HCI/MeOH (3 M, 3 mL) was stirred at rt for 4 h. The mixture was concentrated and purified by pre-H PLC to give the desired compound as HCOOH salt (white solid, 21 mg, 53%). LC-MS: [M+H] + = 412. X H NMR (400 MHz, CD3OD) <5 8.52 (br s, 1H), 7.24 (dd, J = 8.0, 1.2 Hz, 1H), 7.09 (t, J = 8.0 Hz, 1H), 6.75 (dd, J = 8.0, 1.2 Hz, 1H), 3.74-3.61 (m, 4H), 3.60- 3.50 (m, 2H), 3.45 (s, 3H), 3.30-3.13 (m, 4H).

The compounds in Table C below were synthesized in the similar manner using appropriate reagents and conditions. The compounds listed in Table C are merely non-limiting examples. Other subject compounds could also be made using similar methods. Table C

Example 7:

Preparation of N-(3-((5-(4-(aminomethyl)-4-methylpiperidin-l-yl)-6-oxo-l,6- dihydropyrazin-2- yl)thio)-2-chlorophenyl)-4-hydroxy-l,5,5-trimethyl-2-oxo-2,5 -dihydro-lH-pyrrole-3-carboxamide

Step 1: preparation of tert-butyl ((l-(3-(benzyloxy)-5-((2-chloro-3-(4-hydroxy-l,5,5-trimethyl -2- oxo-2,5-dihydro-lH-pyrrole-3-carboxamido)phenyl)thio)pyrazin -2-yl)-4-methylpiperidin-4- yl)methyl)carbamate (3)

To a mixture of compound 1 (80 mg, 1.0 eq) in bromobenzene (5 mL) was added compound 2

(107 mg, 0.5 eq) at r.t. and the mixture was degassed and protected with nitrogen. The reaction was stirred at 160 °C for 3 h. Solvent was removed to give crude compou nd 3 as an oil (140 mg, 100%). Step 2: preparation of N-(3-((5-(4-(aminomethyl)-4-methylpiperidin-l-yl)-6-oxo-l,6- dihydropyrazin- 2-yl)thio)-2-chlorophenyl)-4-hydroxy-l,5,5-trimethyl-2-oxo-2 ,5-dihydro-lH-pyrrole-3-carboxamide To a solution of compound 3 (140 mg, 1.0 eq) in DCM (3 mL) was added BCIB (1.9 mL, 1.0 mol/L in DCM, 10.0 eq) at room temperature. The mixture was stirred at r.t. for 1 h. The mixture was quenched by MeOH (3 mL) followed by standard work up process. Desired compound was obtained after prep-HPLC as a slightly red solid (25 mg, 12%). LC-MS: [M+H] + = 547. X H NMR (400 MHz, DMSO) <5 11.37 (br s, 1 H), 8.46 (d, J = 8.0 Hz, 1H), 8.30 (s, 1H), 7.18 (s, 1H), 7.09 (s, 1H), 6.49 (d, J = 8.4 Hz, 1H), 4.24-4.21 (m, 2H), 3.55-3.48 (m, 2H), 2.74 (s, 3H), 2.65 (s, 2H), 1.55-1.50 (m, 2H), 1.38-1.33 (m,

2H), 1.12 (s, 6H), 1.02 (s, 3H).

The compounds in Table D below were synthesized by similar manners using appropriate reagents and conditions. The compounds listed in Table D are merely non limiting examples. Other subject compounds could also be made using similar methods. Table D

Example 8:

Preparation of intermediate tert-butyl ((l-(3-(benzyloxy)-5-bromopyrazin-2-yl)-4-methylpiperidin-4- yl)methyl)carbamate

To a solution of compound 1 (320 mg, 1.0 eq) in DMF (5 mL) was added compound 2 (292 mg,

1.2 eq) and K2CO3 (441 mg, 3.0 eq) at rt. The reaction mixture was stirred at 60 °C under Ar atmosphere for 2 hours before it was quenched by water. The desired compound was obtained after standard work up process as a white solid (140 mg, 26%).

Example 9:

Preparation of l-(3-aminocyclohexyl)-4-(2,3-dichlorophenyl)pyridin-2(lH)-on e

Step 1: Preparation of 4-(2,3-dichlorophenyl)-2-methoxypyridine (3)

To a solution of compound 1 (2.0g, leq) in DMSO were added compound 2 (1 g, leq), PdChidppf) (0.77 g, O.leq) and K2C03(2.9g, 2eq). The mixture was stirred at 95 °C overnight. The reaction was quenched by water, and worked up under standard procedure. The residue was purified by flash column chromatography to give the title compound (2.1 g, yield: 78.9%).

Step 2: Preparation of 4-(2,3-dichlorophenyl)pyridin-2(lH)-one (4)

The mixture of compound 3 (l.Og, leq) in cone. HCI (20 mL) was stirred at 150 °C overnight. Ice-water was added into the mixture and pH was adjusted to 7. After standard work up procedure, title compound (0.94 g, yield: 100%) was obtained.

Step 3: Preparation of 3-((tert-butoxycarbonyl)amino)cyclohexyl methanesulfonate (6)

To a solution of compound 5 (l.Og, 1.0 eq), EtsN (1 ml, 1.5 eq) in DCM (20 mL) was added MsCI.

The mixture was quenched with water (30 mL) 2 hours later. After standard work up procedure, the residue was purified by flash column chromatography to give the title compound (1.0 g, yield: 73.5%). Step 4: Preparation of tert-butyl (3-(4-(2,3-dichlorophenyl)-2-oxopyridin-l(2H)- yl)cyclohexyl)carbamate (7)

To a solution of compound 4 (1.0 g, 1.0 eq) in DMF (10 mL) was added K 2 CO 3 (1.7 g, 3.0eq) and compound 6 (1.7 g, 1.2eq), and the mixture was stirred at 110 °C overnight. The reaction was quenched with water (30 mL), followed by standard work up procedure. The residue was purified by flash column chromatography to give the title compound (170 mg, yield: 9.34%).

Step 5: Synthesis of l-(3-aminocyclohexyl)-4-(2,3-dichlorophenyl)pyridin-2(lH)-on e

The solution of compound 7 (170 mg, 1.0 eq) in 4M HCI/dioxane(5mL) was stirred at room temperature for 30 min. NaOH solution was added to the mixture at 0 °C , and the mixture was extracted with EA (3 x 30 mL). The organic layer was dried over Na 2 S0 4 , filtered and concentrated. The final compound was obtained after Pre-HPLC as HCOOH salt (lOOmg, yield: 76.3%). LC-MS:

[M+H] + = 338.2. X H NMR (400 MHz, CD 3 OD) <5 8.22-8.21 (d, J = 4.8 Hz 1H), 7.63-7.61(d, J = 7.6 Hz, 1H), 7.4- (m, 1H), 7.33-7.31 (m, 2H), 6.85(s, 1H), 5.54(s, 1H), 3.53 (m, 1H), 2.48-2.44 (d, J = 13.2 Hz, 1H), 2.082-2.01(m, 2H), 1.79 (m, 3H), 1.66 (m 3H).

Pharmacological Testing

Some of the compounds disclosed herein were assessed for their ability to selectively inhibit SH P2 activity. The inhibitory properties of the compounds described herein can be evidenced by testing in the following assays. SHP2 Phosphatase Assays

IC50 values were determined at room temperature in 384-well black polystyrene plate, using a final reaction volume of 15 pL and the following assay buffer conditions: 60 mM Hepes (pH = 7.2), 75 mM NaCI, 75 mM KCI, and 1 mM EDTA, 0.05% P-20, 5 mM dithiothreitol (DTT). Full length SHP2 enzyme (diluted to 0.1 nM in reaction buffer) were co-incubated with 1 uM IRS-1 peptide and 0.01 nM to 10 mM compounds of the disclosure for 60 min. The surrogate substrate DiFM UP (5 pL, 100 mM) was added, and incubated at rt for 60 min. The reaction was then quenched by the addition of 5 pL of a 40 mM solution of bpV(Phen). The fluorescence signal was monitored using a microplate reader (Envision, Perkin-Elmer) using excitation and emission wavelengths of 360 nm and 450 nm, respectively. The inhibitor dose-response curves were analyzed using normalized IC50 regression curve fitting with control-based normalization. The inhibitory activity results of the compounds of the disclosure is shown in Table 2.

Table 2. IC50: +++: < 50 nM; ++: < 100 nM; +: < 1 mM;

p-ERK/total ERK Cellular Assay

Cells were seeded in 384-well cell culture plate and incubated overnight. Test compounds were added to the cell plate and the plate was incubated for 2-6 hours. To detect p-ERK, the cell plate was used AlphaLISA SureFire Ultra p-ERK kit; to detect total ERK, the cell plate was used total ERK HTRF kit. The plate was read on the Envision. The inhibitory activity results of the compounds of the disclosure is shown in Table 3.

Table 3. IC50: +++: < 0.1 mM; ++: < 0.5 mM; +: < 1 mM;

Cell Proliferation Assay

Firstly, the cells were seeded in 384-well cell culture plate and incubate overnight. The test compounds were added to the cell plate and incubated for 3-5 days. The cell plate was then detected using CellTiter Glo reagents. The inhibitory activity results of the compounds of the disclosure is shown in Table 4. Table 4. IC50: +++: < 1 mM; ++: < 5 mM; +: < 10 mM

KYSE-520 Xenograft Model

KYSE-520 cells were expanded in culture, harvested and injected subcutaneously into 6-8 weeks old female BALB/c nude mice (5 x 10 s cell/each mouse, supplemented with Matrigel (1:1) for tumor development, n = 9 per group). Subsequent administration of a compound by oral gavage started when the mean tumor size reached approximately 150-200 mm 3 . During the treatment (once a day for 4 weeks), the tumor volumes were measured using a caliper. Statistical analysis of difference in tumor volume among the groups were evaluated using a one-way ANOVA. Vehicle alone was the negative control.

Many of the compounds described herein is very potent and selective, with enzymatic IC50 less than 10 nM. The compounds tested also displayed superior anti-tumor activities in the in vivo animal models. I n some embodiments, the dose amount per day falls within the range of 3-100 mg/kg to achieve the tumor regression or >80% tumor growth inhibition.

Unless otherwise indicated, all numbers expressing quantities of ingredients, properties such as molecular weight, reaction conditions, and etc. used in herein are to be understood as being modified in all instances by the term "a bout." 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 may be modified according to the desired properties sought to be achieved, and should, therefore, be considered as part of the disclosure. At the very least, the examples shown herein are for illustration only, not as an attempt to limit the scope of the disclosure.

The terms "a," "an," "the" and similar referents used in the context of describing embodiments of the present disclosure (especially in the context of the following claims) are to be construed to cover both the singular and the plural, unless otherwise indicated herein or clearly contradicted by context. All methods described herein may be performed in any suitable order unless otherwise indicated herein or otherwise clearly contradicted by context. The use of any and all examples, or exemplary language (e.g., "such as") provided herein is intended merely to better illustrate embodiments of the present disclosure and does not pose a limitation on the scope of any claim. No language in the specification should be construed as indicating any non-claimed element essential to the practice of the embodiments of the present disclosure.

Groupings of alternative elements or embodiments disclosed herein are not to be construed as limitations. Each group member may be referred to and claimed individually or in any combination with other members of the group or other elements found herein. It is anticipated that one or more members of a group may be included in, or deleted from, a group for reasons of convenience and/or patentability.

Certain embodiments are described herein, including the best mode known to the inventors for carrying out the embodiments. Of course, variations on these described embodiments will become apparent to those of ordinary skill in the art upon reading the foregoing description. The inventor expects skilled artisans to employ such variations as appropriate, and the inventors intend for the embodiments of the present disclosure to be practiced otherwise than specifically described herein. Accordingly, the claims include all modifications and equivalents of the subject matter recited in the claims as permitted by applicable law. Moreover, any combination of the above- described elements in all possible variations thereof is contemplated unless otherwise indicated herein or otherwise clearly contradicted by context.

In closing, it is to be understood that the embodiments disclosed herein are illustrative of the principles of the claims. Other modifications that may be employed are within the scope of the claims. Thus, by way of example, but not of limitation, alternative embodiments may be utilized in accordance with the teachings herein. Accordingly, the claims are not limited to embodiments precisely as shown and described.