Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
KRAS G12C INHIBITORS
Document Type and Number:
WIPO Patent Application WO/2020/047192
Kind Code:
A1
Abstract:
The present invention relates to compounds that, inhibit KRas G12C, In particular, the present invention relates to compounds that irreversibly inhibit the activity of KRas G12C, pharmaceutical compositions comprising the compounds and methods of use therefor.

Inventors:
MARX MATTHEW (US)
BLAKE JAMES (US)
FELL JAY (US)
FISCHER JOHN (US)
MEJIA MACEDONIO (US)
Application Number:
PCT/US2019/048713
Publication Date:
March 05, 2020
Filing Date:
August 29, 2019
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
MIRATI THERAPEUTICS INC (US)
ARRAY BIOPHARMA INC (US)
International Classes:
C07D223/00; C07D239/00; C07D487/02; C07D487/04
Domestic Patent References:
WO2017201161A12017-11-23
Foreign References:
US20180072723A12018-03-15
US20180155348A12018-06-07
US9828363B22017-11-28
US20190144444A12019-05-16
CN112430234A2021-03-02
Other References:
HAWRYLUK ET AL.: "Discovery and synthesis of 6,7,8,9-tetrahydro-5H-pyrimido-[4,5-d]azepines as novel TRPV1 antagonists", BIOORGANIC & MEDICINAL CHEMISTRY LETTERS, vol. 20, no. 23, 1 December 2010 (2010-12-01), XP027459374, Retrieved from the Internet [retrieved on 20191014], DOI: 10.1016/j.bmcl.2010.09.023
ALAMGEER ET AL., CURRENT OPIN PHARMCOL., vol. 13, 2013, pages 394 - 401
SANTOS ET AL., SCIENCE, vol. 223, 1984, pages 661 - 664
SAMATARPOULIKAKOS, NAT REV DRUG DISC, vol. 13, no. 12, 2014, pages 928 - 942
DOGAN ET AL., CLIN CANCER RES., vol. 18, no. 22, 26 September 2012 (2012-09-26), pages 6169 - 6177
MCCORMICK, CLIN CANCER RES., vol. 21, no. 8, 2015, pages 1797 - 1801
SUN ET AL., AGNEW CHEM INT ED ENGL., vol. 51, no. 25, 2012, pages 6140 - 6143
OSTREM ET AL., NATURE, vol. 503, 2013, pages 548 - 551
See also references of EP 3844151A4
Attorney, Agent or Firm:
POLYAKOV, Mark, V. et al. (US)
Download PDF:
Claims:
CLAIMS

WE CLAIM:

1. A compound of formula (1):

Formula (I)

or a pharmaceutically acceptable salt thereof:

wherein:

X is a 4-12 membered saturated or partially saturated monocyclic, bridged or spirocyclic ring, wherein the saturated or partially saturated monocyclic ring is optionally substituted with one or more R8;

Y is a bond, O, S or NR3;

R2 is hydrogen, alkyl, hydroxyalkyl, dihydroxyalkyl, alkylaminylalkyl, dialkylaminylalkyl, 7 NR^R10, heterocyclyl, heterocyclylalkyl, aryl, heteroaryl, or heteroarylalkyl, wherein each of the Z, heterocyclyl, heterocyclylalkyl, aryl, heteroaryl, and heteroarylalkyl may be optionally substituted with one or more R9; each Z is Cl - C4 alkylene; each R3 is independently C l - C3 alkyl, halogen or -OR5;

L is a bond, -C(O)-, or C l - C3 alkylene; R4 is hydrogen cycloalkyl, heterocyclyl, aryl aralkyl or heteroaryl, wherein each of the cycloalkyl, heterocyclyl, aryl, aralkyl and hcteroaryl may be optionally substituted with one or more R6, R7 or R8; each R3 is independently hydrogen or Cl - C3 alkyl;

R6 is cycloalkyl, heterocyclyl, heterocyclylalkyl, aryl, or heteroaryl, wherein each of the cycloalkyl, heterocyclyl, aryl, or heteroaryl may be optionally substituted with one or more R7; each R7 is independently halogen, hydroxyl, Cl - C6 alkyl, cycloalkyl, alkoxy, haloalkyl, amino, cyano, heteroalkyl, hydroxyalkyl or Q-haloalkyl, wherein Q is O or S;

R8 is oxo, Cl - C3 alkyl, C2 - C4 alkynyl, heteroalkyl, cyano, -C(0)0R5, -C(0)N(R5)2, -N(R5)2, wherein the Cl - C3 alkyl may be optionally substituted with cyano, halogen, -OR3, -N(R5)2, or heteroaryl; each R9 is independently hydrogen, oxo, acyl, hydroxyl, hydroxyalkyl, cyano, halogen, Cl - C6 alkyl, aralkyl, haloalkyl, hetcroalkyl, cycloalkyl, heterocyclylalkyl, alkoxy, dialkylaminyl, dialkylamidoalkyl, or dialkylaminylalkyl, wherein the Cl - C6 alkyl may be optionally substituted with cycloalkyl; each R10 is independently hydrogen, acyl, Cl - C3 alkyl, heteroalkyl or hydroxyalkyl;

R1 1 is haloalkyl;

RA is absent, hydrogen, deuterium, cyano, halogen, Cl - C-3 alkyl, haloalkyl, heteroalkyl, - C(0)N(R5)2, or hydroxyalkyl; each RB is independently hydrogen, deuterium, cyano, Cl - C3 alkyl, hydroxyalkyl, heteroalkyl, Cl - C3 alkoxy, halogen, haloalkyl, -ZNR5Rn, -C(0)N(R5)2, -NHC(0)Cl - C3 alkyl, - CH2NHC(0)C1 C3 alkyl, heteroaryl, heteroarylalkyl, dialkylaminylalkyl, or heterocyclylalkyl wherein the heterocyclyl portion is substituted with one or more substituents independently selected from halogen, hydroxy], alkoxy and Cl - C3 alkyl, wherein the heteroaryl or the heteroaryl portion of the heteroarylalkyl is optionally substituted with one or more R7; when - is a triple bond then RA is absent, RB is present and p equals one,

or when = is a double bond then RA is present, RB is present and p equals two, or RA, RB and the carbon atoms to which they arc attached form a 5-8 membered partially saturated cycloalkyl optionally substituted with one or more R7; m is 0, 1, 2 or 3; and p is one or two.

2. The compound of claim 1, wherein R'-X is:

wherein the piperazinyl ring is optionally substituted with R8.

3. The compound of claim 2, wherein R’ is -C(0)C(RA) :=:::::= C(RB)P.

4. The compound of claim 3, wherein - is a triple bond and RA is absent, p is one and

RB is hydroxyalkyl or Cl - C3 alkoxy.

5. The compound of claim 3, wherein =^= is a double bond and RA is hydrogen, p is two and at least one RB is independently deuterium, cyano, halogen, haloalkyl, hydroxyalkyl, heteroalkyl, heteroaryl, heteroarylalkyl, -ZNR5Rn, -C(0)N(R5)2, -NHC(0)Cl - C3 alkyl or heterocyclylalkyl wherein the heterocyclyl portion is substituted with one or more substituents independently selected from halogen, hydroxyl, alkoxy or Cl - C3 alkyl.

6. The compound of claim 5, wherein the at least one RB is halogen.

7. The compound of claim 5, wherein the at least one RB is haloalkyl.

8. The compound of claim 5, wherein the at least one RB is -ZNR3Rn.

9. The compound of claim 8, wherein Z is methylene, R3 is methyl and R1 1 is

trifluoromethyl.

10. The compound of claim 5, wherein the at least one RB is cyano.

1 1. The compound of claim 5, wherein the at least one RB is hydroxyalkyl.

12. The compound of claim 5, wherein the at least one RB is heteroalkyl.

13. The compound of claim 12, wherein the heteroalkyl is methoxy ethyl.

14. The compound of claim 5, wherein the at least one RB is -C(0)N(R3)2, wherein each R3 is hydrogen.

15. The compound of claim 5, wherein the at least one RB is -C(0)N(R5)2, wherein each R3 is Cl - C3 alkyl.

16. The compound of claim 5, wherein the at least one RB is heteroaryl optionally substituted with one or more R7.

17. The compound of claim 16. wherein the heteroaryl is pyrrolyl, imidazolyl, pyrazolyl, triazolyl, pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl or triazinyl, each substituted with one or more R7.

18. The compound of claim 5, wherein the at least one RB is heteroarylalkyl optionally substituted with one or more R7.

19. The compound of claim 18, wherein the heteroaryl portion of the heteroarylalkyl is pyrrolyl, imidazolyl, pyrazolyl, triazolyl, pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl or triazinyl, each optionally substituted with one or more R7.

20. The compound of claim 5, wherein the at least one RB is heterocyclylalkyl substituted with one or more R7.

21. The compound of claim 20, wherein the heterocyclyl portion of the heterocyclylalkyl is azetindinyl, pyrrolidinyl, piperidinyl piperazinyl or morpholinyl, each substituted with one or more groups independently selected from halogen, hydroxyl, alkoxy or Cl - C3 alkyl.

22. The compound according to any of claims 5-21 , wherein the double bond is in the E configuration.

23. The compound according to any of claims 5-21 , wherein the double bond is in the Z configuration..

24. The compound of claim 5, wherein — is a double bond and p is two, each RB is hydrogen, and RA is deuterium, cyano, halogen, haloalkyl, heteroalkyl, -C(0)N(R5)2, or hydroxy alkyl.

25. The compound of claim 24, wherein RA is halogen.

26. The compound of claim 24, wherein RA is haloalkyl.

27. The compound of claim 24, wherein RA is cyano.

28. The compound of claim 24, wherein RA is heteroalkyl.

29. The compound of claim 28, wherein the heteroalkyl is methoxymethyl.

30. The compound of claim 28, wherein the heteroalkyl is alkoxy.

31. The compound of claim 24, wherein RA is -C(0)N(R3)2, wherein each R5 is hydrogen.

32. The compound of claim 24, wherein RA is hydroxyalkyl.

33. The compound of claim 2, wherein - is a double bond and p is two, one RB is hydrogen, the second RB is dialkylaminylalkyl, and RA is halogen.

34. The compound of claim 2, wherein - is a double bond and p is two, each RB is deuterium, and RA is deuterium.

35. The compound of claim 2, wherein when :=^= is a double bond and p is two, one RB is hydrogen and RA and one RB and the carbon atoms to which they arc attached form a 5-8 membered partially saturated cycloalkyl substituted with oxo.

36. The compound according to any one of claims 2-35, wherein Y is O.

37. The compound according to any one of claims 2-36, wherein R2 is selected from the group consisting of hydroxyalkyl, alkylaminylalkyl, dialkylaminylalkyl, -ZNR3R10 heterocyclyl and heterocyclylalkyl, wherein each of the Z, heterocyclyl or heterocyclylalkyl are independently optionally substituted with R9.

38. The compound of claim 37, wherein R2 is heterocyclylalkyl optionally substituted with one or more R9.

39. The compound of claim 38, wherein the heterocyclyl of the heterocyclylalkyl is independently azetidinyl, methylazetidinyl, difluoroazetidinyl, tetrahydropyran, pyrrolidinyl, methylpyrrolidinyl, diemethylpyrrolidinyl, isopropylpyrrolidinyl, cycloalkylalkylpyrrolidinyl, hydroxypyrrolindinyl, fluoropyrrolidinyl, difluoropyrrolidinyl, (N-methyl)fluoropynOlidinyl, (N- methyl)difluoropyrrolidinyl, methoxyethylpyrrolidinyl, (N-methyl)methoxypynOlidinyl, piperazinyl, dimethylaminylpyrrolidinyl, morpholinyl, methylmorpholinyl, 1,4-oxazepanyl, piperdinyl, methylpiperidinyl acylpiperdinyl, cyanopiperdinyl, cycloalkylpiperdinyl,

halopiperdinyl, dihalopiperdinyl, fluoropiperdinyl, difluoropiperdinyl, alkoxypiperdinyl, pyrrolidonyl, piperidinonyl, thiomorpholiny 1-1,1 -dioxide, 3-azabicyclo[3.1.OJhexanyl, oxa-5- azabicyclo[2.2.1 ]heptan-5-yl, or azabicyclo[2.2.lJheptan-2-yl.

40. The compound of claim 39, wherein the (N-methyl)difluoropyrrolidinyl is 3,3-difluoro-l- methylpyrrolidinyl.

41. The compound of claim 39, wherein the heterocyclyl is N-methylpyrrolidinyl.

42. The compound of claim 37, wherein R2 is dialkylaminylalkyl optionally substituted with one or more R9.

43. The compound according to any one of claims 2-42, wherein R4 is aryl optionally substituted with one or more R7.

44. The compound of claim 43, wherein the aryl is selected from the group consisting of phenyl and naphthyl optionally substituted with one or more R7.

45. The compound of claim 44, wherein the phenyl and the naphthyl are each optionally substituted with one or more R7 selected from the group consisting of halogen, Cl - C3 alkyl, haloalkyl. and hydroxy alkyl.

46. The compound of claim 45, wherein R7 is selected from the group consisting of methyl, isopropyl, chloro, fluoro, and trifluoromethyl.

47. The compound according to any one of claims 2-42, wherein R4 is heteroaryl.

48. The compound according to claim 47, wherein R4 is indazolyl or quinolinyl optionally substituted with C1-C3 alkyl.

48. The compound according to any one of claims 2-42, wherein R4 is aralkyl optionally substituted with one or more R7.

49. The compound according to any one of claims 2-48, wherein m is zero.

50. The compound according to any one of claims 2-49, wherein L is a bond.

51. The compound according to any one of claims 2-50, wherein R8 is heteroalkyl, C2-C4 alkynyl, or Cl - C3alkyl optionally substituted with -OR\ cyano or heteroaryl.

52. The compound of claim 51, wherein R8 is Cl - C3 alkyl optionally substituted with cyano.

53. The compound of claim 51 , wherein R8 is cyanomethyl.

54. The compound according to any one of claims 51-53, wherein X is substituted with one R8.

55. The compound of claim 1 , wherein the compound is:

56. The compound of claim 1, wherein the compound is of Formula I-A:

Formula I-A wherein X is a piperazinyl ring optionally substituted with R8, where R1, R3, R4, R5, R8, R10, L and m are as defined in claim 1 and R1 1 is hydrogen, methyl or hydroxyalkyl.

57. The compound of claim 56, wherein R1 is -C(0)C(RA) ==== C(RB)P and . is a double bond and RA is hydrogen, p is two and at least one RB is independently deuterium, cyano, halogen, haloalkyl, hydroxyalkyl, heteroalkyl, heteroaryl, heteroarylalkyl, -ZNR5Rn, - C(0)N(R5)2, -NHC(0)Cl - C3 alkyl or heterocyclylalkyl wherein the heterocyclyl portion is substituted with one or more substituents independently selected from halogen, hydroxyl, alkoxy or Cl - C3 alkyl.

58. The compound of claim 56, wherein R1 C(RB)P and . is a double bond, each RB is hydrogen, and RA is deuterium, cyano, halogen, Cl - C-3 alkyl, haloalkyl, heteroalkyl, -C(0)N(R5)2, or hydroxyalkyl.

59. The compound according to any of claims 57-58, wherein R3 and R10 are each Cl - C3 alkyl.

60. The compound of claim 59, wherein R] 1 is methyl.

61. The compound of claim 59, w'herein R8 is heteroalkyl, C2-C4 alkynyl, or Cl - C3 alkyl optionally substituted with -OR3, cyano or heteroaryl.

62. The compound according to any one of claims 54-61 , wherein L is a bond, and R4 is aryl or heteroaryl, each optionally substituted with one or more R6, R7 or R8.

63. The compound of claim 62, wherein the aryl or heteroaryl are substituted with one or more R7 independently selected from hydroxyl, amino, halogen, Cl - C3 alkyl, haloalkyl, Q- haloalkyl, cycloalkyl and alkoxy.

64. The compound of claim 1 , wherein the compound is of Formula I-B:

Formula I-B where X is a piperazinyl ring optionally substituted with R8, and R1, R3, R4, R8, L and m are as defined in claim 1.

65. The compound of claim 64, wherein R1 is C(())C( R' ) . C(RB)P and .— is a double bond and RA is hydrogen, p is two and at least one RB is independently deuterium, cyano, halogen, haloalkyl, hydroxyalkyl, heteroalkyl, heteroaryl, heteroarylalkyl, -ZNRTv1 - C(0)N(R5)2, -NHC(0)C1 - C3 alkyl or heterocyclylalkyl wherein the heterocyclyl portion is substituted with one or more substituents independently selected from halogen, hydroxyl, alkoxy and Cl - C3 alkyl.

66. The compound of claim 64, wherein R1 is -C(0)C(RA) C(RB)P and ==:::::1 is a double bond, each RB is hydrogen, and RA is deuterium, cyano, halogen, Cl - C-3 alkyl, haloalkyl, heteroalkyl, -C(0)N(R5)2, or hydroxyalkyl.

67. The compound according to any one of claims 65-66, wherein R2 is heterocyclylalkyl optionally substituted with one or more R9.

68. The compound of claim 67, wherein the heterocyclyl portion of the heterocyclylalkyl is pyrrolidinyl, piperidinyl, piperazinyl, morpholinyl, 1 ,4-oxazepanyl, thiomorpholinyl- 1 ,1 -dioxide, 3-azabicyclo[3.1.OJhexanyl, 2-oxa-5-azabicyclo[2.2.1]heptan-5-yl, or azabicyclo[2.2.1]heptan-2- yl, each optionally substituted with one or more R9.

69. The compound of claim 68, wherein each R9 is independently acyl, oxo, halogen, cyano, C l - C6 alkyl, hydroxyalkyl, heteroalkyl, cycloalkyl, aralkyl or dialkylamidoalkyl.

70. The compound of claim 69, wherein each R9 is independently C1-C6 alkyl, heteroalkyl, or halogen.

71. The compound according to any one of claims 65-70, wherein the heterocyclyl is pyrrolidinyl optionally substituted with one or more R9.

72. The compound of claim 71, wherein the heterocyclyl is 3,3-difluoro-l- methylpyrrolidinyl .

73. The compound of claim 72, wherein R8 is heteroalkyl, C2-C4 alkynyl, or Cl - C3alkyl optionally substituted with -OR3, cyano or heteroaryl.

74. The compound of claim 73, wherein R8 is C1-C3 alkyl optionally substituted with cyano.

75. The compound according to any one of claims 65-74, wherein L is a bond and R4 is aryl or heteroaryl, each optionally substituted with one or more R6, R7 or R8.

76. The compound of claim 75, wherein the aryl or heteroaryl are substituted with one or more R7 independently selected from hydroxyl, amino, halogen, Cl - C3 alkyl, haloalkyl, Q- haloalkyl, cycloalkyl and alkoxy.

77. The compound according to any one of claims 75-76, wherein R4 is aryl optionally substituted with one or more R6 or R7.

78. The compound according to claim 77, wherein R4 is phenyl or naphthyl, each optionally substituted with one or more R6 or R7.

79. The compound according to claim 77, wherein R4 is optionally substituted with one or more R7 independently selected from halogen, C1-C6 alkyl, and haloalkyl.

80. The compound according to any one of claims 65-79, wherein m is zero.

81. A pharmaceutical composition, comprising a therapeutically effective amount of a compound of Formula (1), Formula I-A or Formula I-B according to any one of claims 1-80, and a pharmaceutically acceptable excipient.

82. A method for inhibiting KRas G12C activity in a cell, comprising contacting the cell in which inhibition of KRas G12C activity is desired with an effective amount of a compound of Formula (I), Formula l-A or Formula I-B according to any one of claims 1-80, pharmaceutically acceptable salts thereof or pharmaceutical compositions containing the compound of Formula (I), Formula I-A or Formula I-B, or pharmaceutically acceptable salt thereof.

83. A method for treating cancer comprising administering to a patient having cancer a therapeutically effective amount of a compound of Formula (I), Formula I-A or Formula I-B, or a pharmaceutically acceptable salt thereof according to any one of claims 1 -80, alone or combined with a pharmaceutically acceptable carrier, excipient or diluents.

84. The method of claim 83, wherein the therapeutically effective amount of the compound is between about 0.01 to 100 g/kg per day.

85. The method of claim 84, wherein the therapeutically effective amount of the compound is between about 0.1 to 50 mg/kg per day.

86. The method of claim 83, wherein the cancer is selected from the group consisting of Cardiac: sarcoma (angiosarcoma, fibrosarcoma, rhabdomyosarcoma, liposarcoma), myxoma, rhabdomyoma, fibroma, lipoma and teratoma; Lung: bronchogenic carcinoma (squamous cell, undifferentiated small cell, undifferentiated large cell, adenocarcinoma), alveolar (bronchiolar) carcinoma, bronchial adenoma, sarcoma, lymphoma, chondromatous hamartoma, mesothelioma; Gastrointestinal: esophagus (squamous cell carcinoma, adenocarcinoma, leiomyosarcoma, lymphoma), stomach (carcinoma, lymphoma, leiomyosarcoma), pancreas (ductal

adenocarcinoma, insulinoma, glucagonoma, gastrinoma, carcinoid tumors, vipoma), small bowel (adenocarcinoma, lymphoma, carcinoid tumors, Kaposi's sarcoma, leiomyoma, hemangioma, lipoma, neurofibroma, fibroma), large bowel (adenocarcinoma, tubular adenoma, villous adenoma, hamartoma, leiomyoma); Genitourinary tract: kidney (adenocarcinoma, Wilm's tumor (nephroblastoma), lymphoma, leukemia), bladder and urethra (squamous cell carcinoma, transitional cell carcinoma, adenocarcinoma), prostate (adenocarcinoma, sarcoma), testis

(seminoma, teratoma, embryonal carcinoma, teratocarcinoma, choriocarcinoma, sarcoma, interstitial cell carcinoma, fibroma, fibroadenoma, adenomatoid tumors, lipoma); Liver:

hepatoma (hepatocellular carcinoma), cholangiocarcinoma, hepatoblastoma, angiosarcoma, hepatocellular adenoma, hemangioma; Biliary tract: gall bladder carcinoma, ampullary carcinoma, cholangiocarcinoma; Bone: osteogenic sarcoma (osteosarcoma), fibrosarcoma, malignant fibrous histiocytoma, chondrosarcoma, Ewing's sarcoma, malignant lymphoma (reticulum cell sarcoma), multiple myeloma, malignant giant cell tumor chordoma,

osteochronfroma (osteocartilaginous exostoses), benign chondroma, chondroblastoma, chondromyxofibroma, osteoid osteoma and giant cell tumors; Nervous system: skull (osteoma, hemangioma, granuloma, xanthoma, osteitis deformans), meninges (meningioma,

meningiosarcoma, gliomatosis), brain (astrocytoma, medulloblastoma, glioma, ependymoma, germinoma (pinealoma), glioblastoma multiform, oligodendroglioma, schwannoma, retinoblastoma, congenital tumors), spinal cord neurofibroma, meningioma, glioma, sarcoma); Gynecological: uterus (endometrial 'carcinoma (serous cystadenocarcinoma, mucinous cystadenocarcinoma, unclassified carcinoma), granulosa-thecal cell tumors, Sertoli-Leydig cell tumors, dysgerminoma, malignant teratoma), vulva (squamous cell carcinoma, intraepithelial carcinoma, adenocarcinoma, fibrosarcoma, melanoma), vagina (clear cell carcinoma, squamous cell carcinoma, botryoid sarcoma (embryonal rhabdomyosarcoma), fallopian tubes (carcinoma); Hematologic: blood (myeloid leukemia (acute and chronic), acute lymphoblastic leukemia, chronic lymphocytic leukemia, myeloproliferative diseases, multiple myeloma, myelodysplastic syndrome), Hodgkin's disease, non-Hodgkin's lymphoma (malignant lymphoma); Skin:

malignant melanoma, basal cell carcinoma, squamous cell carcinoma, Kaposi's sarcoma, moles dysplastic nevi, lipoma, angioma, dermatofibroma, keloids, psoriasis; and Adrenal glands: neuroblastoma.

87. The method of claim 86, wherein the cancer wherein the cancer is a KRas G12C- associated cancer.

88. The method of claims 83-87, wherein the cancer is non-small cell lung cancer.

89. A method for treating cancer in a patient in need thereof, the method comprising (a) determining that the cancer is associated with a KRas G12C mutation (e.g., a KRas G12C- associated cancer); and (b) administering to the patient a therapeutically effective amount of a compound of Formula (II), Formula II-A or Formula II-B or a pharmaceutically acceptable salt thereof according to any one of claims 1 -80, or a pharmaceutical composition thereof.

Description:
KRAS G12C INHIBITORS

FIELD OF THE INVENTION

[0001] The present invention relates to compounds that inhibit KRas G12C. In particular, the present invention relates to compounds that irreversibly inhibit the activity of KRas G12C, pharmaceutical compositions comprising the compounds and methods of use therefor.

BACKGROUND OF THE INVENTION

[0002] Kirsten Rat Sarcoma 2 Viral Oncogene Homolog (“KRas”) is a small GTPase and a member of the Ras family of oncogenes. KRas serves a molecular switch cycling between inactive (GDP-bound) and active (GTP-bound) states to transduce upstream cellular signals received from multiple tyrosine kinases to downstream effectors to regulate a wide variety of processes, including cellular proliferation (e.g., see Alamgeer et al., (2013) Current Opin Pharmcol. 13:394-401).

[0003] The role of activated KRas in malignancy was observed over thirty years ago (e.g., see Santos et al., (1984) Science 223:661 -664). Aberrant expression of KRas accounts for up to 20% of all cancers and oncogenic KRas mutations that stabilize GTP binding and lead to constitutive activation of KRas and downstream signaling have been reported in 25 -30% of lung

adenocarcinomas (e.g., see Samatar and Poulikakos (2014) Nat Rev Drug Disc 13(12): 928-942 doi: 10.1038/nrd428). Single nucleotide substitutions that result in missense mutations at codons 12 and 13 of the KRas primary amino acid sequence comprise approximately 40% of these KRas driver mutations in lung adenocarcinoma, with a G12C transversion being the most common activating mutation (e.g., see Dogan et al., (2012) Clin Cancer Res. 18(22):6169-6177, published online 2012 Sep 26. doi: 10.1 158/1078-0432.CCR-11-3265).

[0004] The well-known role of KRAs in malignancy and the discovery of these frequent mutations in KRas in various tumor types made KRas a highly attractable target of the pharmaceutical industry for cancer therapy. Notwithstanding thirty years of large scale discovery efforts to develop inhibitors of KRas for treating cancer, no KRas inhibitor has demonstrated sufficient safety and/or efficacy to obtain regulatory approval (e.g., see McCormick (2015) Clin Cancer Res. 21 (8): 1797- 1801 ).

[0005] Despite many failed efforts to target KRas, compounds that inhibit KRas activity are still highly desirable and under investigation, including those that disrupt effectors such as guanine nucleotide exchange factors (e.g., see Sun et ah, (2012) Agnew Chem Int Ed Engl. 5l(25):6l40- 6143 doi: l0.1002/anie20l20l358) as well target KRas G12C (e.g., see Ostrem et ah, (2013) Nature 503:548-551). Clearly there remains a continued interest and effort to develop inhibitors of KRas, particularly inhibitors of activating KRas mutants, including KRas G12C.

[0006] Thus, there is a need to develop new KRas G12C inhibitors that demonstrate sufficient efficacy, stability and/or safety for treating KRas Gl2C-mediated cancer. The compounds and compositions of the present invention advantageously overcome one or more of the previous oiiui ivuiiini^a uy iO v iu ^ S i^^ii v ^ r ixas VJ I ZV^ ihitiuhui ¾.

SUMMARY OF THE INVENTION

[0007] In one aspect of the invention, compounds are provided that inhibit KRas G12C activity. In certain embodiments, the compounds are represented by formula (I):

Formula (I)

[0008] or a pharmaceutically acceptable salt thereof, wherein: [0010] X is a 4-12 membered saturated or partially saturated monocyclic, bridged or spirocyclic ring, wherein the saturated or partially saturated monocyclic ring is optionally substituted with one or more R 8 ;

[0011] Y is a bond, O, S or NR 5 ;

[0012] R 1 is -C(0)C(R a ) == C(R B ) P or -S0 2 C(R A ) C(R B ) P ;

[0013] R 2 is hydrogen, alkyl, hydroxyalkyl, dihydroxyalkyl, alkylaminylalkyl,

dialkylaminylalkyl, -Z-NR 5 R 10 , heterocyclyl, heterocyclylalkyl, aryl, heteroaryl, or

heteroarylalkyl, wherein each of the Z, heterocyclyl, heterocyclylalkyl, aryl, heteroaryl, and heteroarylalkyl may be optionally substituted with one or more R 9 ;

[0014] Z is Cl - C4 alkylene;

[0015] each R 3 is independently Cl - C3 alkyl, oxo, or haloalkyl;

[0016] L is a bond, -C(O)-, or C 1 - C3 alkylene;

[0017] R 4 is hydrogen, cycloalkyl, heterocyclyl, aryl, aralkyl or heteroaryl, wherein each of the cycloalkyl, heterocyclyl, aryl, aralkyl and heteroaryl may be optionally substituted with one or more R 6 or R 7 ;

[0018] each R 5 is independently hydrogen or Cl - C3 alkyl;

[0019] R 6 is cycloalkyl, heterocyclyl, heterocyclylalkyl, aryl, or heteroaryl, wherein each of the cycloalkyl, heterocyclyl, aryl, or heteroaryl may be optionally substituted with one or more R 7 ;

[0020] each R 7 is independently halogen, hydroxyl, Cl - C6 alkyl, cycloalkyl, alkoxy, haloalkyl, amino, cyano, heteroalkyl, hydroxyalkyl or Q-haloalkyl, wherein Q is O or S;

[0021] R 8 is oxo, Cl - C3 alkyl, C2 - C4 alkynyl, heteroalkyl, cyano, -C(0)OR 5 , -C(0)N(R 5 ) 2 , - N(R ? ) 2 , wherein the Cl— C3 alkyl may be optionally substituted with cyano, halogen, -OR 3 , - N(R 5 ) 2 , or heteroaryl

[0022] each R 9 is independently hydrogen, oxo, acyl, hydroxyl, hydroxyalkyl, cyano, halogen,

Cl - C6 alkyl, aralkyl, haloalkyl, hcteroalkyl, cycloalkyl, heterocyclylalkyl, alkoxy, dialkylaminyl, dialkylamidoalkyl, or dialkylaminylalkyl, wherein the Cl - C6 alkyl may be optionally substituted with cycloalkyl;

[0023] each R 10 is independently hydrogen, acyl, Cl - C3 alkyl, heteroalkyl or hydroxyalkyl;

[0024] R 11 is haloalkyl;

[0025] R A is absent, hydrogen, deuterium, cyano, halogen, Cl - C-3 alkyl, haloalkyl, heteroalkyl, -C(0)N(R : ’)2, or hydroxyalkyl;

[0026] each R B is independently hydrogen, deuterium, cyano, Cl - C3 alkyl, hydroxyalkyl, heteroalkyl, Cl - C3 alkoxy, halogen, haloalkyl, -ZNR 5 R n , -C(0)N(R 5 ) 2 , -NHC(0)C1 - C3 alkyl, -CH 2 NHC(0)C1 - C3 alkyl, heteroaryl, heteroarylalkyl, dialkylaminylalkyl, or

heterocyclylalkyl wherein the heterocyclyl portion is substituted with one or more substituents independently selected from halogen, hydroxyl, alkoxy and Cl— C3 alkyl, wherein the heteroaryl or the heteroaryl portion of the heteroarylalkyl is optionally substituted with one or more R 7 ;

[0027] m is 0, 1, 2 or 3;

[0028] p is one or two; and wherein,

[0029] when . is a triple bond then R A is absent, R B is present and p equals one,

[0030] or when = is a double bond then R A is present, R B is present and p equals two, or R a , R B and the carbon atoms to which they are attached form a 5-8 membered partially saturated cycloalkyl optionally substituted with one or more R 7 .

[0031] Also included are compounds of Formula I having the Formula I-A:

Formula I- A

[0032] wherein R 1 , R 3 , R 4 , R 5 , R 10 , L and m are as defined for Formula I, R 1 1 is hydrogen, Cl C3 alkyl or hydroxyalkyl, and X is a piperazinyl ring which is optionally substituted with R 8 wherein R 8 is as defined for Formula I.

[0033] Also included are compounds of Formula 11 having the Formula l-B:

Formula I-B

[0034] where R 1 , R 3 , R 4 , R 8 , L and m are as defined for Formula II, R 2 is heterocyclylalkyl optionally substituted with one or more R 9 , and X is a piperazinyl ring which is optionally substituted with R 8 , where R 8 is as defined for Formula I.

[0035] In another aspect of the invention, pharmaceutical compositions are provided comprising a therapeutically effective amount of a compound of the present invention or a pharmaceutically acceptable salt thereof and a pharmaceutically acceptable excipient.

[0036] In yet another aspect of the invention, methods for inhibiting KRas G12C activity in a in a cell, comprising contacting the cell with a compound of Formula I, Formula I-A, Formula 1 -B. ln one embodiment, the contacting is in vitro. In one embodiment, the contacting is in vivo.

[0037] Also provided herein is a method of inhibiting cell proliferation, in vitro or in vivo, the method comprising contacting a cell with an effective amount of a compound of Formula I, Formula I-A, Formula 1-B, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof as defined herein.

[0038] Also provided are methods for treating cancer in a patient comprising administering a therapeutically effective amount of a compound or pharmaceutical composition of the present invention or a pharmaceutically acceptable salt thereof to a patient in need thereof.

[0039] Also provided herein is a method of treating a KRas G12C -associated disease or disorder in a patient in need of such treatment, the method comprising administering to the patient a therapeutically effective amount of a compound of Formula I, Formula I-A, Formula 1 -B, or a pharmaceutically acceptable salt or solvate thereof, or a pharmaceutical composition thereof as defined herein.

[0040] Also provided herein is a compound of Formula I, Formula I-A, Formula 1-B, or a pharmaceutically acceptable salt or solvate thereof, or a pharmaceutical composition thereof as defined herein for use in therapy.

[0041] Also provided herein is a compound of Formula I, Formula I-A, Formula 1 -B,or a pharmaceutically acceptable salt or solvate thereof or a pharmaceutical composition thereof as defined herein for use in the treatment of cancer.

[0042] Also provided herein is a compound of Formula I, Formula I-A, Formula 1 -B,or a pharmaceutically acceptable salt or solvate thereof for use in the inhibition of KRas G12C.

[0043] Also provided herein is a compound of Formula I, Formula I-A, Formula 1 -B, or a pharmaceutically acceptable salt or solvate thereof or a pharmaceutical composition thereof as defined herein, for use in the treatment of a KRas G12C-associated disease or disorder. [0044] Also provided herein is the use of a compound of Formula I, Formula I-A, Formula 1 -B, or a pharmaceutically acceptable salt or solvate thereof, as defined herein in the manufacture of a medicament for the treatment of cancer.

[0045] Also provided herein is a use of a compound of Formula I, Formula I-A, Formula 1-B, or a pharmaceutically acceptable salt or solvate thereof, as defined herein in the manufacture of a medicament for the inhibition of activity of KRas G12C.

[0046] Also provided herein is the use of a compound of Formula I, Formula I-A, Formula 1-B, or a pharmaceutically acceptable salt or solvate thereof, as defined herein, in the manufacture of a medicament for the treatment of a KRas Gl2C-associated disease or disorder.

[0047] Also provided herein is a method for treating cancer in a patient in need thereof, the method comprising (a) determining that the cancer is associated with a KRas G12C mutation (e.g., a KRas Gl 2C-associated cancer); and (b) administering to the patient a therapeutically effective amount of a compound of Formula I, Formula I-A, Formula l-B, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof.

[0048] Also provided herein is a process for preparing a compound of Formula I, Formula I-A, Formula 1-B, or a pharmaceutically acceptable salt or solvate thereof.

[0049] Also provided herein is a compound of Formula I, Formula I-A, Formula 1-B, or a pharmaceutically acceptable salt thereof obtained by a process of preparing the compound as defined herein.

DETAILED DESCRIPTION OF THE INVENTION

[0050] The present invention relates to inhibitors of KRas G12C. In particular, the present invention relates to compounds that irreversibly inhibit the activity of KRas G12C,

pharmaceutical compositions comprising a therapeutically effective amount of the compounds and methods of use therefor.

DEFINITIONS

[0051] Unless defined otherwise, all technical and scientific terms used herein have the same meaning as is commonly understood by one of skill in the art to which this invention belongs. All patents, patent applications, and publications referred to herein are incorporated by reference.

[0052] As used herein,“KRas G12C” refers to a mutant form of a mammalian KRas protein that contains an amino acid substitution of a cysteine for a glycine at amino acid position 12. The assignment of amino acid codon and residue positions for human KRas is based on the amino acid sequence identified by UniProtKB/Swiss-Prot P01116: Variant p.Glyl2Cys.

[0053] As used herein, a“KRas G12C inhibitor” refers to compounds of the present invention that are represented by Formula (I) as described herein. These compounds are capable of negatively modulating or inhibiting all or a portion of the enzymatic activity of KRas G12C.

The KRas G12C inhibitors of the present invention interact with and irreversibly bind to KRas G12C by forming a covalent adduct with the sulfhydryl side chain of the cysteine residue at position 12 resulting in the inhibition of the enzymatic activity of KRas G12C.

[0054] A "KRas Gl2C-associated disease or disorder" as used herein refers to diseases or disorders associated with or mediated by or having a KRas G12C mutation. A non-limiting example of a KRas Gl2C-associated disease or disorder is a KRas G12C-associated cancer.

[0055] As used herein, the term“subject,” "individual," or "patient," used interchangeably, refers to any animal, including mammals such as mice, rats, other rodents, rabbits, dogs, cats, swine, cattle, sheep, horses, primates, and humans. In some embodiments, the patient is a human. In some embodiments, the subject has experienced and/or exhibited at least one symptom of the disease or disorder to be treated and/or prevented. In some embodiments, the subject has been identified or diagnosed as having a cancer having a KRas G12C mutation (e.g., as determined using a regulatory agency-approved, e.g., FDA-approved, assay or kit). In some embodiments, the subject has a tumor that is positive for a KRas G12C mutation (e.g., as determined using a regulatory agency-approved assay or kit). The subject can be a subject with a tumor(s) that is positive for a KRas G12C mutation (e.g., identified as positive using a regulatory agency- approved, e.g., FDA-approvcd, assay or kit). The subject can be a subject whose tumors have a KRas G12C mutation (e.g., where the tumor is identified as such using a regulatory agency- approved, e.g., FDA-approved, kit or assay). In some embodiments, the subject is suspected of having a KRas G12C gene-associated cancer. In some embodiments, the subject has a clinical record indicating that the subject has a tumor that has a KRas G12C mutation (and optionally the clinical record indicates that the subject should be treated with any of the compositions provided herein).

[0056] In some embodiments of any of the methods or uses described herein, an assay is used to determine whether the patient has KRas G12C mutation using a sample (e.g., a biological sample or a biopsy sample (e.g., a paraffin-embedded biopsy sample) from a patient (e.g., a patient suspected of having a KRas G12C-associated cancer, a patient having one or more symptoms of a KRas Gl2C-associated cancer, and/or a patient that has an increased risk of developing a KRas G12C-associated cancer) can include, for example, next generation sequencing,

immunohistochemistry, fluorescence microscopy, break apart FISH analysis, Southern blotting, Western blotting, FACS analysis, Northern blotting, and PCR-based amplification (e.g., RT-PCR and quantitative real-time RT-PCR). As is well-known in the art, the assays are typically performed, e.g., with at least one labelled nucleic acid probe or at least one labelled antibody or antigen-binding fragment thereof.

[0057] The term“regulatory agency” is a country’s agency for the approval of the medical use of pharmaceutical agents with the country. For example, a non-limiting example of a regulatory agency is the U.S. Food and Drug Administration (FDA).

[0058] The term“amino” refers to -NII 2 ;

[0059] The term "acyl" refers to -C(0)CH 3.

[0060] The term "alkyl" as employed herein refers to straight and branched chain aliphatic groups having from 1 to 12 carbon atoms, 1-8 carbon atoms 1-6 carbon atoms, or 1-3 carbon atoms which is optionally substituted with one, two or three substituents. Examples of alkyl groups include, without limitation, methyl, ethyl, propyl, isopropyl, butyl, isobutyl, sec-butyl, tert-butyl, pentyl, and hexyl.

[0061 j The term“haloalkyl” refers to an alkyl chain in which one or more hydrogen has been replaced by a halogen. Examples of haloalkyls are trifluoromethyl, difluoromethyl and fluoromethyl.

[0062] The term“haloalkyloxy” refers to -O-haloalkyl. [0063] An "alkylene," group is an alkyl group, as defined hereinabove, that is positioned between and serves to connect two other chemical groups. Exemplary alkylene groups include, without limitation, methylene, ethylene, propylene, and butylene.

[0064] The term“alkoxy” refers to -OC1 - C6 alkyl.

[0065] The term "cycloalkyl" as employed herein includes saturated and partially unsaturated cyclic hydrocarbon groups having 3 to 12 carbons, for example 3 to 8 carbons, and as a further example 3 to 6 carbons, wherein the cycloalkyl group additionally is optionally substituted. Examples of cycloalkyl groups include, without limitation, cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cycloheptyl, and cyclooctyl.

[0066] The term "heteroalkyl" refers to an alkyl group, as defined hereinabove, wherein one or more carbon atoms in the chain are replaced by a heteroatom selected from the group consisting of O, S, and N.

[0067] As used herein, the term“hydroxyalkyl” refers to -alkyl-OH.

[0068] The term“dihydroxyalkyl” refers to an alkyl group as defined herein wherein two carbon atoms are each substituted with a hydroxyl group.

[0069] The term“alkylaminyl” refers to -NR x -alkyl, wherein R x is hydrogen. In one

embodiment, R x is hydrogen.

[0070] The tenn“dialkylaminyl” refers to N(R y ) 2 , wherein each R y is Cl - C3 alkyl.

[0071] The term“alkylaminylalkyl” refers to -alkyl-NR x -alkyl, wherein R x is hydrogen. In one embodiment, R x is hydrogen.

[0072] The term“dialkylaminylalkyl” refers to -alkyl-N(R y ) 2 , wherein each R y is Cl - C4 alkyl, wherein the alkyl of the— alkyl-N(R y ) 2 may be optionally substituted with hydroxy or hydroxy alkyl.

[0073] An "aryl" group is a C 6 -Ci 4 aromatic moiety comprising one to three aromatic rings, which is optionally substituted. As one embodiment, the aryl group is a C 6 -Cio aryl group. Examples of aryl groups include, without limitation, phenyl, naphthyl, anthracenyl, fluorenyl, and dihydrobenzofuranyl.

[0074] An "aralkyl" or "arylalkyl" group comprises an aryl group covalently linked to an alkyl group, either of which may independently be optionally substituted or unsubstituted. An example of an aralkyl group is (Ci- C 6 )alkyl(C 6 -Cio)aryl, including, without limitation, benzyl, phenethyl, and naphthylmethyl. An example of a substituted aralkyl is wherein the alkyl group is substituted with hydroxyalkyl.

[0075] A "heterocyclyl" or "heterocyclic" group is a ring structure having from about 3 to about 12 atoms, for example 4 to 8 atoms, wherein one or more atoms are selected from the group consisting of N, O, and S, the remainder of the ring atoms being carbon. The heterocyclyl may be a monocyclic, a bicyclic, a spirocyclic or a bridged ring system. The heterocyclic group is optionally substituted with R 7 on carbon or nitrogen at one or more positions, wherein R 7 is as defined for Formula I. The heterocyclic group is also independently optionally substituted on nitrogen with alkyl, aryl, aralkyl, alkylcarbonyl, alkylsulfonyl, arylcarbonyl, arylsulfonyl, alkoxycarbonyl, aralkoxy carbonyl, or on sulfur with oxo or lower alkyl. Examples of heterocyclic groups include, without limitation, epoxy, azetidinyl, aziridinyl, tetrahydrofuranyl, tetrahydropyranyl, pyrrolidinyl, pyrrolidinonyl, piperidinyl, piperaziny!, imidazolidinyl, thiazolidinyl, dithianyl, trithianyl, dioxolanyl, oxazolidinyl, oxazolidinonyl,

decahydroquinolinyl, piperidonyl, 4-piperidinonyl, thiomorpholinyl, thiomorpholinyl 1,1 dioxide, morpholinyl, oxazepanyl, azabicyclohexanes, azabicycloheptanes and oxa

azabiocycloheptanes. Specifically excluded from the scope of this term are compounds having adjacent annular O and/or S atoms.

[0076] The term“heterocyclylalkyl” refers to a heterocyclyl group as defined herein linked to the remaining portion of the molecule via an alkyl linker, wherein the alkyl linker of the heterocyclylalkyl may be optionally substituted with hydroxy or hydroxyalkyl.

[0077] As used herein, the term "heteroaryl" refers to groups having 5 to 14 ring atoms, preferably 5, 6, 9, or 10 ring atoms; having 6, 10, or 14 p electrons shared in a cyclic array; and having, in addition to carbon atoms, from one to three hctcroatoms per ring selected from the group consisting ofN, O, and S. Examples of heteroaryl groups include acridinyl, azocinyl, benzimidazolyl, benzofuranyl, benzothiofuranyl, benzothiophcnyl. benzoxazolyl, benzthiazolyl, benztriazolyl, benztetrazolyl, bcnzisoxazolyl, benzisothiazolyl, benzimidazolinyl, carbazolyl, 4aH-carbazolyl, carbolinyl, chromanyl, chromenyl, cinnolinyl, furanyl, furazanyl, imidazolinyl, imidazolyl, lH-indazolyl, indolenyl, indolinyl, indolizinyl, indolyl, 3H-indolyl, isobenzofuranyl, isochromanyl, isoindazolyl, isoindolinyl, isoindolyl, isoquinolinyl, isothiazolyl, isoxazolyl, methylenedioxyphenyl, naphthyridinyl, octahydroisoquinolinyl, oxadiazolyl, l,2,3-oxadiazolyl, l,2,4-oxadiazolyl, 1,2,5-oxadiazolyl, 1,3,4-oxadiazolyl, oxazolidinyl, oxazolyl, oxazolidinyl, pyrimidinyl, phenanthridinyl, phenanthrolinyl, phenazinyl, phenothiazinyl, phenoxathiinyl, phenoxazinyl, phthalazinyl, piperonyl, pteridinyl, purinyl, pyranyl, pyrazinyl, pyrazolidinyl, pyrazolinyl, pyrazolyl, pyridazinyl, pyridooxazole, pyridoimidazole, pyridothiazole. pyridinyl, pyridyl, pyrimidinyl, pyrrolinyl, 2H-pyrrolyl, pyrrolyl, quinazolinyl, quinolinyl, 4H-quinolizinyl, quinoxalinyl, quinuclidinyl, tetrahydroisoquinolinyl, tetrahydroquinolinyl, tetrazolyl, 6H-1 ,2,5- thiadiazinyl, i,2,3-thiadiazolyl, 1 ,2,4-thiadiazolyl, l,2,5-thiadiazolyl, l ,3,4-thiadiazolyl, thianthrenyl, thiazolyl, thienyl, thienothiazolyl, thienooxazolyl, thienoimidazolyl, thiophenyl, triazinyl, 1,2,3-triazolyl, 1 ,2,4-triazolyl, 1 ,2,5-triazolyl, l,3,4-triazolyl, and xanthenyl.

[0078] A "heteroarylalkyl" group comprises a heteroaryl group covalently linked to an alkyl group, wherein the radical is on the alkyl group, either of which is independently optionally substituted or unsubstituted. Examples of heteroarylalkyl groups include a heteroaryl group having 5, 6, 9, or 10 ring atoms bonded to a C1-C6 alkyl group. Examples of heteroaralkyl groups include pyridylmethyl, pyridylethyl, pyrrolylmethyl, pyrrolylethyl, imidazolylmethyl, imidazolylethyl, thiazolylmethyl, thiazolylethyl, benzimidazolylmethyl, benzimidazolylethyl quinazolinylmethyl, quinolinylmethyl, quinolinylethyl, benzofuranylmethyl, indolinylethyl isoquinolinylmethyl, isoinodylmethyl, cinnolinylmethyl, and benzothiophenylethyl. Specifically excluded from the scope of this term are compounds having adjacent annular O and/or S atoms.

[0079] As used herein,“an effective amount” of a compound is an amount that is sufficient to negatively modulate or inhibit the activity of KRas G12C. Such amount may be administered as a single dosage or may be administered according to a regimen, whereby it is effective.

[0080] As used herein, a "therapeutically effective amount" of a compound is an amount that is sufficient to ameliorate, or in some manner reduce a symptom or stop or reverse progression of a condition, or negatively modulate or inhibit the activity of KRas G12C. Such amount may be administered as a single dosage or may be administered according to a regimen, whereby it is effective.

[0081 ] As used herein, treatment means any manner in which the symptoms or pathology of a condition, disorder or disease are ameliorated or otherwise beneficially altered. Treatment also encompasses any pharmaceutical use of the compositions herein.

[0082] As used herein, amelioration of the symptoms of a particular disorder by administration of a particular pharmaceutical composition refers to any lessening, whether permanent or temporary, lasting or transient that can be attributed to or associated with administration of the composition.

COMPOUNDS

[0083] In one aspect of the invention, compounds are provided represented by formula (I):

Formula (I)

[0084] or a pharmaceutically acceptable salt thereof, wherein:

[0085] X is a 4-12 membered saturated or partially saturated monocyclic, bridged or spirocyclic ring, wherein the saturated or partially saturated monocyclic ring is optionally substituted with one or more R 8 ;

[0086] Y is a bond, O, S or NR 5 ;

[0087] R 1 i -C(0)C(R a ) C(R B ) p or -S0 2 C(R a ) C(R B ) P ; [0088] R 2 is hydrogen, alkyl, hydroxyalkyl, dihydroxyalkyl, alkylaminylalkyl,

dialkylaminylalkyl, -Z-NR 5 R 10 , heterocyclyl, heterocyclylalkyl, aryl, heteroaryl, or

heteroarylalkyl, wherein each of the Z, heterocyclyl, heterocyclylalkyl, aryl, heteroaryl, and heteroarylalkyl may be optionally substituted with one or more R 9 ;

[0089] Z is Cl - C4 alkylene;

[0090] each R 3 is independently Cl - C3 alkyl, halogen or -OR 5 ;

[0091] L is a bond, -C(O)-, or C l - C3 alkylene;

[0092] R 4 is hydrogen, cycloalkyl, heterocyclyl, aryl, aralkyl or heteroaryl, wherein each of the cycloalkyl, heterocyclyl, aryl, aralkyl and heteroaryl may be optionally substituted with one or more R 6 or R 7 ;

[0093] each R 5 is independently hydrogen or Cl - C3 alkyl;

[0094] R 6 is cycloalkyl, heterocyclyl, heterocyclylalkyl, aryl, or heteroaryl, wherein each of the cycloalkyl, heterocyclyl, aryl, or heteroaryl may be optionally substituted with one or more R 7 ;

[0095] each R 7 is independently halogen, hydroxyl, Cl - C6 alkyl, cycloalkyl, alkoxy, haloalkyl, amino, cyano, heteroalkyl, hydroxyalkyl or Q-haloalkyl, wherein Q is O or S;

[0096] R 8 is oxo, Cl - C3 alkyl, C2 - C4 alkynyl, heteroalkyl, cyano, -C(0)OR ;> , -C(0)N(R :, ) 2 , - N(R¾ wherein the Cl - C3 alkyl may be optionally substituted with cyano, halogen, -OR 5 , - N(R 3 ) 2 , or heteroaryl;

[0097] each R 9 is independently hydrogen, oxo, acyl, hydroxyl, hydroxyalkyl, cyano, halogen,

Cl - C6 alkyl, aralkyl, haloalkyl, heteroalkyl, cycloalkyl, heterocyclylalkyl, alkoxy,

dialkylaminyl, dialkylamidoalkyl, or dialkylaminylalkyl, wherein the Cl - C6 alkyl may be optionally substituted with cycloalkyl;

[0098] each R 10 is independently hydrogen, acyl, Cl - C3 alkyl, heteroalkyl or hydroxyalkyl; [0099] R A is absent, hydrogen, or Cl - C3 alkyl;

[0100] each R B is independently hydrogen, Cl - C3 alkyl, alkylaminylalkyl, dialkylaminylalkyl or heterocyclylalkyl;

[0101] m is 0, 1 , 2 or 3;

[0102] p is one or two; and wherein,

[0103] when . is a triple bond then R A is absent, R B is present and p equals one;

[0104] or when = is a double bond then R A is present, R B is present and p equals two, or R A , R B and the carbon atoms to which they are attached form a 5-8 membered partially saturated cycloalkyl optionally substituted with one or more R 7 .

[0105] in certain embodiments, R’-X is:

[0106] wherein R 1 is are defined for Formula 1 and the piperazinyl ring is optionally substituted with R 8 , where R 8 is as defined for Formula I. In certain embodiments, R 8 is Cl - C3 alkyl wherein the alkyl is optionally substituted with cyano or OR 3 , or -C(0)N(R 3 ) 2, wherein each R 5 is independently hydrogen or Cl - C3 alkyl.

[0107] In particular embodiments, R 1 is -C(0)C(R A ) . C(R B ) P where R A , R B and p are as defined for Formula II. In one embodiment, R 1 is -C(0)C(R A ) . C(R B ) P , wherein = is a triple bond and R A is absent, p is one and R B is hydroxyalkyl.

[0108] In one embodiment, R 1 is -C(0)C(R A ) — C(R B ) P , wherein is a double bond and R A is hydrogen or Cl C3 alkyl, p is two and at least one R B is deuterium, cyano, Cl - C3 alkyl, hydroxyalkyl, heteroalkyl, Cl - C3 alkoxy, halogen, haloalkyl, -ZNR 5 R n , -C(0)N(R 5 ) 2 , - NHC(0)C1 - C3 alkyl, -CH 2 NF1C(0)C1 C3 alkyl, heteroaryl, heteroarylalkyl,

dialkylaminylalkyl, or heterocyclylalkyl wherein the heterocyclyl portion is substituted with one or more substituents independently selected from halogen, hydroxyl, alkoxy and Cl - C3 alkyl, wherein the heteroaryl or the heteroaryl portion of the heteroarylalkyl is optionally substituted with one or more R 7 . In one embodiment, when === is a double bond, the double bond is in the E configuration. In one embodiment, the double bond is in the Z configuration.

[0109] In certain embodiments, one R B is heterocyclylalkyl substituted with one or more substituents independently selected from halogen, hydroxyl, alkoxy or Cl - C3 alkyl and the other R B is hydrogen. In one embodiment, the heterocyclyl portion of the heterocyclylalkyl is azetidinyl substituted with a halogen. In certain embodiments, the halogen is fluorine. In one embodiment, the heterocyclyl portion of the heterocyclylalkyl is pyrrolidinyl substituted with one or more halogen. In certain embodiments, the halogen-substituted pyrrolidinyl is

fluoropyrrolidinyl or difluorpyrrolidinyl.

[0110] In certain embodiments, one R B is halogen and the other R B is hydrogen. In one embodiment, the halogen is chlorine.

[01 1 1] In certain embodiments, one R B is haloalkyl and the other R B is hydrogen ln one embodiment, the haloalkyl is chloromethyl, fluoromethyl, difluoromethyl or trifluoromethyl.

[0112] In certain embodiments, one R B is heteroalkyl and the other R B is hydrogen. In one embodiment, the heteroalkyl is methoxymethyl.

[01 13] In ceratin embodiments, one R B is -ZNR 3 R n , wherein Z is methylene, R 5 is methyl and R 11 is trifluoromethyl or 2,2,2-trifluoroethyl, and the other R B is hydrogen.

[0114] In certain embodiments, one R B is hydroxy alkyl and the other R B is hydrogen.

[0115] In certain embodiments, one R B is heteroaryl optionally substituted with one or more R 7 and the other R B is hydrogen. In one embodiment, the heteroaryl is pyrrolyl, imidazolyl, pyrazolyl, triazolyl, pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl or triazinyl, each substituted with one or more R 7 .

[01 16] In certain embodiments, one R B is heteroarylalkyl optionally substituted with one or more R 7 , and the other R B is hydrogen. In one embodiment, the heteroaryl portion of the

heteroarylalkyl is pyrrolyl, imidazolyl, pyrazolyl, triazolyl, pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl or triazinyl, each optionally substituted with one or more R 7 . In one embodiment, the one or more R 7 is Cl - C3 alkyl.

[0117] In certain embodiments, one R B is -C(0)N(R :> ) 2 and the other R B is hydrogen. In one embodiment, each R 5 is hydrogen. In one embodiment, each R 5 is Cl - C3 alkyl.

[0118] In certain embodiments, one R B is -NHC(0)Cl C3 alkyl or -CH2NHC(0)Cl - C3 alkyl and the other R B is hydrogen. In one embodiment, the Cl - C3 alkyl is methyl.

[01 19] In one embodiment, R 1 is -C(0)C(R A ) =C(R B ) P , wherein R A is deuterium, cyano, halogen, Cl - C-3 alkyl, haloalkyl, heteroalkyl, -C(0)N(R 5 ) 2 , or hydroxyalkyl, p is two, each R B is hydrogen. In one embodiment, R A is halogen ln one embodiment, the halogen is fluorine or chlorine. In one embodiment, R A is haloalkyl. In one embodiment, the haloalkyl is

trifluorom ethyl. In one embodiment, R A is cyano. In one embodiment, R A is heteroalkyl. In one embodiment, the heteroalkyl is methoxy. In one embodiment, R A is hydroxyalkyl.

[0120] In one embodiment, R 1 is -C(0)C(R A ) C(R B ) P , wherein a double bond and R A is deuterium, p is two and at least one R B is deuterium.

[0121] In one embodiment, R 1 is -C(0)C(R A ) . C(R B ) P , wherein = is a double bond and p is two, one R B is hydrogen and R A and one R B and the carbon atoms to which they are attached form a 5-8 membered partially saturated cycloalkyl substituted with oxo.

[0122] In one embodiment, R 1 is -C(0)C(R A ) . C(R B ) P , wherein . is a double bond and p is two, one R B is hydrogen, the second R B is dialkylaminylalkyl, and R A is halogen.

[0123] In one embodiment, Y is O or NR 5 and R 2 is selected from the group consisting of alkyl, hydroxyalkyl, dihydroxyalkyl, alkylaminylalkyl, dialkylaminylalkyl, heterocyclyl,

heterocyclylalkyl, and heteroaryl. In one embodiment, Y is O and R 2 is hydroxyalkyl, dihydroxyalkyl, alkylaminylalkyl, or dialkylaminylalkyl, wherein the alkylaminylalkyl or dialkylaminylalkyl is optionally substituted with one or more R 9 . In one embodiment, the optionally substituted alkylaminylalkyl or dialkylaminylalkyl is independently selected from methylaminylpropan-2-yl, dimethylaminylethyl, methylethylaminylethyl,

dimethylaminylpropanyl, dimethylaminylpropan-2-yl, dimethylaminylbutanyl, climethylaminylbutan-2-yl, 2-dimethylaminylpropanol, or diethylaminylethyl. In one embodiment, Y is O or NR 5 and R 2 is heterocyclyl or heterocyclylalkyl optionally substituted with one or more R 9 . Nonlimiting examples of one or more R 9 when R 2 is heterocyclyl or heterocyclylalkyl include Cl - C3 alkyl, acyl, oxo, cyano, alkoxy, cycloalkyl, cycloalky lmethyl, halogen, and hydroxyl. Nonlimiting examples of R 2 heterocyclyls optionally substituted with one or more R 9 include azetidinyl, Cl-C3alkyl-substituted azetidinyl (e.g., methylazetidinyl), halo- substituted azetidinyl (e.g., difluoroazetidinyl), tetrahydropyran, pyrrolidinyl, C1-C3 alkyl- substituted pyrrolidinyl (e.g., methylpyrrolidinyl, dimethylpyrrolidinyl, and

isopropylpyrrolidinyl), cycloalkylalkylpyrrolidinyl, hydroxypyrrolindinyl, halo-substituted pyrrolidinyl (e.g., fluoropyrrolidinyl and difluoropyrrolidinyl), methoxyethylpyrrolidinyl, (N- methyl)methoxypyrrolidinyl, piperazinyl, dimethylaminylpyrrolidinyl, morpholinyl,

methylmorpholinyl, 1 ,4-oxazepanyl, piperdinyl, C1-C3 alkyl-substituted piperidinyl (e.g., methylpiperidinyl), acylpiperdinyl, cyanopiperdinyl, cycloalkylpiperdinyl, halopiperdinyl (e.g., fluoropiperdinyl), dihalopiperdinyl (e.g., difluoropiperdinyl), alkoxypiperdinyl, pyrrolidonyl, piperidonyl, thiomorpholinyl- 1,1 -dioxide, 3-azabicyclo[3.l .0]hexanyl, oxa-5- azabicyclo[2.2.l]heptan-5-yl, and azabicyclo[2.2.1]heptan-2-yl.

[0124] In one embodiment, Y is O and R 2 is heteroarylalkyl optionally substituted with one or more R 9 . In one embodiment, the heteroaryl portion of the heteroarylalkyl is pyridinyl.

[0125] In one embodiment, Y is O and R 2 is -ZR^R 10 . In one embodiment, R 5 is Cl - C3 alkyl and R 10 is independently selected from acyl, hydroxyalkyl or alkoxy.

[0126] In one embodiment, Y is a bond and R 2 is hydrogen, heterocyclyl or aryl, wherein said heterocyclyl and aryl are optionally substituted with one or more R 9 .

[0127] In one embodiment, Y is a bond and R 2 is hydrogen.

[0128] In one embodiment, Y is a bond and R 2 is heterocyclyl optionally substituted with one or more R 9 . In one embodiment, Y is a bond and R 2 is heterocyclyl optionally substituted with methyl, halogen or dimethylamino. Nonlimiting examples of R 2 heterocyclyls include azetidinyl, piperidinyl, piperazinyl, morpholinyl, and pyrrolidinyl.

[0129] In one embodiment, Y is a bond and R 2 is aryl optionally substituted with one or more R 9 . In one embodiment, the aryl is phenyl substituted with heterocyclylalkyl.

[0130] In certain other embodiments when X is a monocyclic ring, R 4 is aryl. In one

embodiment, R 4 is selected from the group consisting of phenyl and naphthyl and is optionally substituted with one or more R 6 or R 7 . Examples of R 7 substituents include halogen, hydroxyl, Cl - C6 alkyl (e.g., Cl - C3 alkyl), cycloalkyl, haloalkyl, Q-haloalkyl, amino, cyano,

hydroxyalkyl and alkoxy. In one embodiment, the aryl is phenyl substituted with one or more R 7 groups independently selected from halogen, hydroxyl, Cl - C3 alkyl, haloalkyl, Q-haloalkyl, and alkoxy. ln one embodiment, the aryl is phenyl substituted with one or more R 7 groups independently selected from halogen, haloalkyl, methyl, isopropyl, methoxy, Q-haloalkyl and hydroxyl. In one embodiment, the aryl is phenyl substituted with one or more R 7 groups independently selected from methyl, trifluoromethyl, 2,2,2-trifluoroethyl, hydroxyl,

trifluoromethoxy, hydroxyl, fluoro, chloro, isopropyl, cyclopropyl and trill uoromethylthio. In one embodiment, the aryl is phenyl substituted with one to three R 7 groups independently selected from hydroxyl, fluorine and chlorine. In one embodiment, the aryl is phenyl substituted with hydroxyl and Cl - C3 alkyl or two Cl - C3 alkyl. In one embodiment, the aryl is phenyl substituted with Q-haloalkyl and hydroxyl or fluorine.

[0131] In one embodiment, R 4 is aryl wherein aryl is naphthyl optionally substituted with one or more R 7 . In one embodiment, the aryl is naphthyl substituted with one or more R 7 groups independently selected from halogen, hydroxyl, Cl- C3 alkyl, haloalkyl, hydroxyalkyl, Q- haloalkyl, and alkoxy. In one embodiment, the aryl is naphthyl substituted with one or more R 7 groups independently selected from halogen, haloalkyl, methyl, isopropyl, methoxy, Q- haloalkyl, hydroxymethyl and hydroxyl. In one embodiment, R 4 is naphthyl optionally substituted with one or more R 7 substituents independently selected from halogen, Cl - C3 alkyl, haloalkyl and hydroxyalkyl. In one embodiment, R 4 is naphthyl optionally substituted with one to three R 7 substituents independently selected from methyl, isopropyl, chloro, fluoro, and trifluoromethyl.

[0132] in one embodiment, the aryl is naphthyl optionally substituted with one or more halogen. In one embodiment, the aryl is naphthyl substituted with hydroxyl and trifluoromethyl or Cl - C3alkyl. In one embodiment, the aryl is naphthyl substituted with hydroxyl. [0133] In one embodiment, R 4 is heteroaryl optionally substituted with one or more R 7 . In one embodiment, R 4 is heteroaryl optionally substituted with one or more R 7 independently selected from halogen, hydroxyl, Cl - C3 alkyl, haloalkyl, Q-haloalkyl, alkoxy and amino. In one embodiments, R 4 is indoyl, indazolyl, quinolinyl, isoquinolinyl, pyridinyl or benzo[d]thiazolyl optionally substituted with one or more R 7 . In one embodiment, R 4 is indoyl, indazolyl, quinolinyl, isoquinolinyl, pyridinyl or benzo[d]thiazolyl optionally substituted with one or more R 7 independently selected from halogen, hydroxyl, Cl - C3 alkyl, haloalkyl, Q-haloalkyl, alkoxy and amino. In one embodiment, R 4 is indazolyl or quinolinyl optionally substituted with Cl -C3 alkyl.

[0134] In yet other embodiments, R 4 is heteroaryl, optionally an indoyl or an indazolyl, each of which may be substituted with one or more R 7 . In one embodiment, R 4 is heteroaryl optionally substituted with one or more R 7 substituents independently selected from the group consisting of halogen, hydroxyl, Cl - C3 alkyl, haloalkyl, Q-haloalkyl and alkoxy. In one embodiment, the R 4 heteroaryl is indazolyl optionally substituted with one or two R 7 independently selected from alkoxy, haloalkyl, and C1-C6 alkyl. In other embodiments, the R 4 heteroaryl is a quinolinyl or isoquinolinyl, each optionally substituted with one or more R 7 . In one embodiment, the R 4 heteroaryl is a quinolinyl or isoquinolinyl, each optionally substituted with one or more R 7 independently selected from amino, hydroxyl, Cl - C3 alkyl, and hydroxyl. In one

embodiment, the R 4 heteroaryl is a quinolinyl or isoquinolinyl, each optionally substituted with R 7 selected from hydroxyl and amino. In one embodiment, the R 4 heteroaryl is a pyridinyl optionally substituted with one or more R 7 . In one embodiment, the R 4 heteroaryl is pyridinyl optionally substituted with one or more R 7 independently selected from Cl - C3 alkyl, halogen and haloalkyl. In other embodiments, the R 4 heteroaryl is benzo[d]thiazolyl optionally substituted with one or more R 7 , such as hydroxyl, one or two Cl - C3 alkyl, or hydroxyl and one or two Cl - C3 alkyl. In one embodiment, the R 4 heteroaryl is indolyl optionally substituted with one or more R 7 . In one embodiment, the R 4 heteroaryl is indolyl optionally substituted with one or two R 7 independently selected from hydroxyl and Cl - C3alkyl.

[0135] In one embodiment, where X is a monocyclic ring, R 4 is aralkyl. In certain

embodiments, the aralkyl is benzyl. In other embodiments, the alkyl of the benzyl group is optionally substituted with hydroxyalkyl. [0136] In one embodiment, L is a bond.

[0137] In one embodiment, R J is Cl C3 alkyl. In one embodiment, the Cl - C3 alkyl is methyl.

[0138] In one embodiment, R 3 is halogen. In one embodiment, the halogen is fluorine or chlorine.

[0139] In one embodiment, R 8 is heteroalkyl, C2-C4 alkynyl or Cl - C3 alkyl optionally substituted with -OR 5 , cyano or heteroaryl. In one embodiment, R 8 is methyl, cyanomethyl, methoxymethyl, hydroxymethyl. In one embodiment, R 8 is methyl. In one embodiment, R 8 is cyanomethyl. In one embodiment, R 8 is hydroxymethyl.

[0140] In one embodiment, Formula I includes compounds having the Formula I-A:

[0141] wherein R 1 , R 3 , R 4 , R 3 , R 10 , L and m are as defined for Formula 1, R 11 is hydrogen, methyl or hydroxyalkyl, and X is a piperazinyl ring which is optionally substituted with R 8 wherein R 8 is as defined for Formula I. In one embodiment, L is a bond. In one embodiment,

R 4 is aryl or heteroaryl, each of which is optionally substituted with one or more R 6 or R 7 . In one embodiment, R 4 is aryl or heteroaryl, each of which is optionally substituted with one or more R 7 . In one embodiment, each R 7 is independently selected from hydroxyl, amino, halogen, Cl - C3 alkyl, haloalkyl, Q-haloalkyl, cycloalkyl and alkoxy. In one embodiment, R 3 and R 10 arc each Cl C3 alkyl. In one embodiment, the aryl is phenyl substituted with one or more R 7 groups independently selected from halogen, hydroxyl, Cl - C3 alkyl, haloalkyl, Q-haloalkyl, and alkoxy. In one embodiment, the aryl is phenyl substituted with one or more R 7 groups independently selected from halogen, haloalkyl, methyl, isopropyl, methoxy, Q-haloalkyl and hydroxyl. In one embodiment, the aryl is phenyl substituted with one or more R 7 groups independently selected from methyl, trifluoromethyl, 2,2,2-trifluoroethyl, hydroxyl,

trifluoromethoxy, hydroxyl, fluoro, chloro, isopropyl, cyclopropyl and trifluoromethylthio. In one embodiment, the aryl is phenyl substituted with one to three R 7 groups independently selected from hydroxyl, fluorine and chlorine. In one embodiment, the aryl is phenyl substituted with hydroxyl and Cl - C3 alkyl or two Cl - C3 alkyl. In one embodiment, the aryl is phenyl substituted with Q-haloalkyl and hydroxyl or fluorine. In one embodiment, the aryl is naphthyl substituted with one or more R 7 groups independently selected from halogen, hydroxyl, Cl - C3 alkyl, haloalkyl, Q-haloalkyl, and alkoxy. In one embodiment, the aryl is naphthyl substituted with one or more R 7 groups independently selected from halogen, haloalkyl, methyl, isopropyl, methoxy, Q-haloalkyl and hydroxyl. In one embodiment, R 4 is naphthyl optionally substituted with one or more R 7 substituents independently selected from hydroxyl, halogen, Cl - C3 alkyl, amino, and haloalkyl. In one embodiment, R 4 is naphthyl optionally substituted with one to three R 7 substituents independently selected from difluoromethyl, methyl, hydroxyl, amino, fluoro, and chloro. In one embodiment, the aryl is naphthyl optionally substituted with one or more halogen. In one embodiment, the aryl is naphthyl substituted with hydroxyl and trifluoromethyl or Cl - C3alkyl. In one embodiment, the ary] is naphthyl substituted with hydroxyl. In one embodiment, R 4 is heteroaryl, wherein the hetcroaryl is indazolyl optionally substituted with one or two R 7 independently selected from alkoxy, haloalkyl, and C1-C6 alkyl. In one embodiment, R 4 is heteroaryl, wherein the heteroaryl is quinolinyl or isoquinolinyl, each optionally substituted with one or more R 7 . In one embodiment, R 4 is heteroaryl, wherein the heteroaryl is quinolinyl or isoquinolinyl, each optionally substituted with one or more R 7 independently selected from amino, hydroxyl, Cl - CSalkyl, and hydroxyl. In one embodiment, the R 4 heteroaryl is a pyridinyl optionally substituted with one or more R 7 . In one embodiment, the R 4 heteroaryl is pyridinyl optionally substituted with one or more R 7 independently selected from Cl— C3 alkyl, halogen and haloalkyl. In one embodiment, the R 4 heteroaryl is benzo[d]thiazolyl optionally substituted with one or more R 7 , such as hydroxyl, one or two Cl - C3 alkyl, or hydroxyl and one or two Cl - C3 alkyl. In one embodiment, the R 4 hetcroaryl is indolyl optionally substituted with one or more R 7 . In one embodiment, the R 4 heteroaryl is indolyl optionally substituted with one or two R 7 independently selected from hydroxyl and Cl - C3alkyl. In one embodiment, R 1 1 is methyl. In one embodiment, the piperazinyl ring is unsubstituted. In one embodiment, the piperazinyl ring is substituted with R 8 . ln one embodiment, R 8 is Cl - C3 alkyl optionally substituted with cyano or hydroxyl. In one embodiment, R 8 is methyl, cyanomethyl or hydroxymethyl. In one embodiment, R 8 is methyl. In one embodiment, R 8 is cyanomethyl. In one embodiment, R 8 is hydroxymethyl. In another embodiment, R 3 and R 10 are each Cl - C3 alkyl, R 11 is methyl, R 8 is methyl, cyanomethyl or hydroxymethyl, L is a bond, and R 4 is aryl or heteroaryl, each optionally substituted with one or more R 6 or R 7 .

[0142] In particular embodiments, R 1 is -C(0)C(R A ) C(R B ) P where R A , R B and p are as defined for Formula II. In one embodiment, R 1 is -C(0)C(R A ) = C(R B ) P , wherein := is a triple bond and R A is absent, p is one and R B is hydroxy alkyl.

[0143] In one embodiment, R 1 is -C(0)C(R A ) = = C(R B ) P , wherein = is a double bond and R A is hydrogen or Cl - C3 alkyl, p is two and at least one R B is deuterium, cyano, Cl - C3 alkyl, hydroxyalkyl, heteroalkyl, Cl - C3 alkoxy, halogen, haloalkyl, -ZNR 5 R n , -C(0)N(R 5 ) 2 , - NHC(0)Cl - C3 alkyl, -CH 2 NHC(0)Cl - C3 alkyl, heteroaryl, heteroarylalkyl,

dialkylaminylalkyl, or heterocyclylalkyl wherein the heterocyclyl portion is substituted with one or more substituents independently selected from halogen, hydroxyl, alkoxy and Cl - C3 alkyl, wherein the heteroaryl or the heteroaryl portion of the heteroarylalkyl is optionally substituted with one or more R 7 . In one embodiment, when is a double bond, the double bond is in the E configuration. In one embodiment, the double bond is in the Z configuration.

[0144] In certain embodiments, one R B is heterocyclylalkyl substituted with one or more substituents independently selected from halogen, hydroxyl, alkoxy or Cl - C3 alkyl and the other R B is hydrogen in one embodiment, the heterocyclyl portion of the heterocyclylalkyl is azetidinyl substituted with a halogen. In certain embodiments, the halogen is fluorine. In one embodiment, the heterocyclyl portion of the heterocyclylalkyl is pyrrolidinyl substituted with one or more halogen. In certain embodiments, the halogen-substituted pyrrolidinyl is fluoropyrrolidinyl or difluorpyrrolidinyl.

[0145] In certain embodiments, one R B is halogen and the other R B is hydrogen. In one embodiment, the halogen is chlorine.

[0146] In certain embodiments, one R B is haloalkyl and the other R B is hydrogen. In one embodiment, the haloalkyl is chloromethyl, fluoromethyl, difluoromethyl or trifluoromethyl. [0147] In certain embodiments, one R B is heteroalkyl and the other R B is hydrogen in one embodiment, the heteroalkyl is methoxy methyl.

[0148] In ceratin embodiments, one R B is -ZNR 5 R U , wherein Z is methylene, R 5 is methyl and R 1 1 is trifluoromethyl or 2,2,2-trifluoroethyl, and the other R B is hydrogen.

[0149] In certain embodiments, one R B is hydroxyalkyl and the other R B is hydrogen.

[0150] In certain embodiments, one R B is heteroaryl optionally substituted with one or more R 7 and the other R B is hydrogen. In one embodiment, the heteroaryl is pyrrolyl, imidazolyl, pyrazolyl, triazolyl, pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl or triazinyl, each substituted with one or more R 7 .

[0151] In certain embodiments, one R B is heteroarylalkyl optionally substituted with one or more R 7 , and the other R B is hydrogen. In one embodiment, the heteroaryi portion of the

heteroarylalkyl is pyrrolyl, imidazolyl, pyrazolyl, triazolyl, pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl or triazinyl, each optionally substituted with one or more R 7 . In one embodiment, the one or more R 7 is C 1 - C3 alkyl.

[0152] In certain embodiments, one R B is -C(0)N(R 5 ) 2 and the other R B is hydrogen. In one embodiment, each R 5 is hydrogen. In one embodiment, each R 5 is Cl - C3 alkyl.

[0153] In certain embodiments, one R B is -NHC(0)Cl - C3 alkyl or -CH 2 NHC(0)C1 - C3 alkyl and the other R B is hydrogen ln one embodiment, the Cl - C3 alkyl is methyl.

[0154] In one embodiment, R 1 is -C(0)C(R A ) =C(R B ) P , wherein R A is deuterium, cyano, halogen, Cl - C-3 alkyl, haloalkyl, heteroalkyl, -C(0)N(R 3 ) 2 , or hydroxyalkyl, p is two, each R B is hydrogen. In one embodiment, R A is halogen. In one embodiment, the halogen is fluorine or chlorine. In one embodiment, R A is haloalkyl. In one embodiment, the haloalkyl is

trifluoromethyl. In one embodiment, R A is cyano. In one embodiment, R A is heteroalkyl. In one embodiment, the heteroalkyl is methoxy. In one embodiment, R A is hydroxyalkyl.

[0155] In one embodiment, R 1 is -C(0)C(R A ) = C(R B ) P , wherein .— is a double bond and R A is deuterium, p is two and at least one R B is deuterium. [0156] ln one embodiment, R 1 is -C(0)C(R A ) === C(R B ) P , wherein =^= is a double bond and p is two, one R B is hydrogen and R A and one R B and the carbon atoms to which they are attached form a 5-8 membered partially saturated cycloalkyl substituted with oxo.

[0157] In one embodiment, R 1 is -C(0)C(R A ) C(R B ) P , wherein is a double bond and p is two, one R B is hydrogen, the second R B is dialkylaminylalkyl, and R A is halogen.

[0158] In one embodiment, Formula I includes compounds having the Formula l-B:

Formula I-B and R 1 , R J , R 4 , R 9 , L and m are as defined for Formula I, R 2 is heterocyclylalkyl optionally substituted with one or more R 9 , and X is a piperazinyl ring which is optionally substituted with R s , where R 8 is as defined for Formula I. In one embodiment, the heterocyclyl portion of the R 2 heterocyclylalkyl is a monocyclic, bicyclic, or bridged ring system having one or two ring heteroatoms independently selected from N and O. In one embodiment, R 2 heterocyclyl is pyrrolidinyl, piperidinyl, piperazinyl, morpholinyl, 1,4-oxazepanyl, thiomorpholinyl- 1,1 -dioxide, 3-azabicyclo[3.l .0]hexanyl, 2-oxa-5-azabicyclo[2.2.l]heptan-5-yl, and azabicyclo[2.2.1 ]heptan- 2-yl, optionally substituted with one or more R 9 . In one embodiment, each R 9 is selected from acyl, oxo, halogen, cyano, Cl - C3 alkyl, alkoxy, hydroxyalkyl, heteroalkyl, cycloalkyl, aralkyl and di alkyl ami doalkyl. In one embodiment, L is a bond. In one embodiment, R 4 is aryl or heteroaryl, each of which is optionally substituted with one or more R 6 or R 7 . In one

embodiment, R 4 is aryl or heteroaryl, each of which is optionally substituted with one or more R 7 . In one embodiment, each R 7 is independently selected from hydroxyl, amino, halogen, Cl - C3 alkyl, haloalkyl, Q-haloalkyl, cycloalkyl and alkoxy. In one embodiment, the aryl is phenyl substituted with one or more R 7 groups independently selected from halogen, hydroxyl, Cl - C3 alkyl, haloalkyl, Q-haloalkyl, and alkoxy. In one embodiment, the aryl is phenyl substituted with one or more R 7 groups independently selected from halogen, haloalkyl, methyl, isopropyl, methoxy, Q-haloalkyl and hydroxyl. In one embodiment, the aryl is phenyl substituted with one or more R 7 groups independently selected from methyl, trifluoromethyl, 2,2,2-trifluoroethyl, hydroxyl, trifluoromethoxy, hydroxyl, fluoro, chloro, isopropyl, cyclopropyl and

trifluoromethylthio. ln one embodiment, the aryl is phenyl substituted with one to three R 7 groups independently selected from hydroxyl, fluorine and chlorine. In one embodiment, the aryl is phenyl substituted with hydroxyl and Cl - C3 alkyl or two Cl - C3 alkyl. In one embodiment, the aryl is phenyl substituted with Q-haloalkyl and hydroxyl or fluorine. In one embodiment, the aryl is naphthyl substituted with one or more R 7 groups independently selected from halogen, hydroxyl, C l - C3 alkyl, haloalkyl, Q-haloalkyl, and alkoxy. In one embodiment, the aryl is naphthyl substituted with one or more R' groups independently selected from halogen, haloalkyl, methyl, isopropyl, methoxy, Q-haloalkyl and hydroxyl. In one embodiment, R 4 is naphthyl optionally substituted with one or more R 7 substituents independently selected from hydroxyl, halogen, Cl - C3 alkyl, amino, and haloalkyl. In one embodiment, R 4 is naphthyl optionally substituted with one to three R 7 substituents independently selected from

difluoromethyl, methyl, hydroxyl, amino, fluoro, and chloro. In one embodiment, the aryl is naphthyl optionally substituted with one or more halogen. In one embodiment, the aryl is naphthyl substituted with hydroxyl and trifluoromethyl or Cl - C3alkyl. In one embodiment, the aryl is naphthyl substituted with hydroxyl. In one embodiment, R 4 is heteroaryl, wherein the heteroaryl is indazolyl optionally substituted with one or two R 7 independently selected from alkoxy, haloalkyl, and C1-C6 alkyl. In one embodiment, R 4 is heteroaryl, wherein the heteroaryl is quinolinyl or isoquinolinyl, each optionally substituted with one or more R 7 . In one embodiment, R 4 is hetcroaryl, wherein the heteroaryl is quinolinyl or isoquinolinyl, each optionally substituted with one or more R 7 independently selected from amino, hydroxyl, Cl - C3 alkyl, and hydroxyl. In one embodiment, the R 4 heteroaryl is a pyridinyl optionally substituted with one or more R'. In one embodiment, the R 4 heteroaryl is pyridinyl optionally substituted with one or more R 7 independently selected from Cl - C3 alkyl, halogen and haloalkyl. In one embodiment, the R 4 heteroaryl is benzo[d]thiazolyl optionally substituted with one or more R 7 , such as hydroxyl, one or two Cl - C3 alkyl, or hydroxyl and one or two Cl - C3 alkyl. In one embodiment, the R 4 heteroaryl is indolyl optionally substituted with one or more R 7 . In one embodiment, the R 4 heteroaryl is indolyl optionally substituted with one or two R 7 independently selected from hydroxyl and Cl - C3 alkyl. In one embodiment, R 1 1 is methyl. In one embodiment, the piperazinyl ring is unsubstituted. In one embodiment, the piperazinyl ring is substituted with R 8 . In one embodiment, the piperazinyl ring is unsubstituted. In one embodiment, the piperazinyl ring is substituted with R 8 . In one embodiment, R 8 is Cl - C3 alkyl optionally substituted with cyano, hydroxyl or methoxy. In one embodiment, R 8 is methyl, cyanomethyl, hydroxymethyl or methoxymethyl.

[0159] In particular embodiments, R 1 is -C(0)C(R A ) C(R B ) P where R A , R B and p are as defined for Formula II. In one embodiment, R 1 is -C(0)C(R A ) . C(R B ) P , wherein = is a triple bond and R A is absent, p is one and R B is hydroxyalkyl.

[0160] In one embodiment, R 1 is -C(0)C(R A ) — C(R B ) P , wherein = is a double bond and R A is hydrogen or Cl - C3 alkyl, p is two and at least one R B is deuterium, cyano, Cl - C3 alkyl, hydroxyalkyl, heteroalkyl, Cl - C3 alkoxy, halogen, haloalkyl, -ZNR 5 R n , -C(0)N(R 5 ) 2 , - NHC(0)C1 - C3 alkyl, -CH 2 NHC(0)Cl - C3 alkyl, heteroaryl, heteroarylalkyl,

dialky laminylalkyl, or heterocyclylalkyl wherein the heterocyclyl portion is substituted with one or more substituents independently selected from halogen, hydroxyl, alkoxy and Cl - C3 alkyl, wherein the heteroaryl or the heteroaryl portion of the heteroarylalkyl is optionally substituted with one or more R 7 . In one embodiment, when == is a double bond, the double bond is in the E configuration. In one embodiment, the double bond is in the Z configuration.

[0161] In certain embodiments, one R B is heterocyclylalkyl substituted with one or more substituents independently selected from halogen, hydroxyl, alkoxy or Cl C3 alkyl and the other R B is hydrogen. In one embodiment, the heterocyclyl portion of the heterocyclylalkyl is azetidinyl substituted with a halogen. In certain embodiments, the halogen is fluorine. In one embodiment the heterocyclyl portion of the heterocyclylalkyl is pyrrolidinyl substituted with one or more halogen. In certain embodiments, the halogen-substituted pyrrolidinyl is fluoropyrrolidinyl or difluorpyrrolidinyl.

[0162] In certain embodiments, one R B is halogen and the other R B is hydrogen in one embodiment, the halogen is chlorine. [0163] In certain embodiments, one R B is haloalkyl and the other R B is hydrogen. In one embodiment, the haloalkyl is chloromethyl, fluoromethyl, difluoromethyl or trifluoromethyl.

[0164] In certain embodiments, one R B is heteroalkyl and the other R B is hydrogen. In one embodiment, the heteroalkyl is methoxymethyl.

[0165] In ceratin embodiments, one R B is -ZNR 3 R n , wherein Z is methylene, R 5 is methyl and R 11 is trifluoromethyl or 2,2,2-trifluoroethyl, and the other R B is hydrogen.

[0166] In certain embodiments, one R B is hydroxy lkyl and the other R B is hydrogen.

[0167] In certain embodiments, one R B is heteroaryl optionally substituted with one or more R 7 and the other R B is hydrogen. In one embodiment, the heteroaryl is pyrrolyl, imidazolyl, pyrazolyl, triazolyl, pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl or triazinyl, each substituted with one or more R 7 .

[0168] In certain embodiments, one R B is heteroarylalkyl optionally substituted with one or more R 7 , and the other R B is hydrogen. In one embodiment, the heteroaryl portion of the

heteroarylalkyl is pyrrolyl, imidazolyl, pyrazolyl, triazolyl, pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl or triazinyl, each optionally substituted with one or more R 7 . In one embodiment, the one or more R 7 is Cl - C3 alkyl.

[0169] In certain embodiments, one R B is -C(0)N(R 5 ) 2 and the other R B is hydrogen. In one embodiment, each R 5 is hydrogen. In one embodiment, each R 5 is Cl - C3 alkyl.

[0170] In certain embodiments, one R B is -NHC(0)Cl - C3 alkyl or -CH 2 NHC(0)C1 - C3 alkyl and the other R B is hydrogen. In one embodiment, the Cl - C3 alkyl is methyl.

[0171] In one embodiment, R 1 is -C(0)C(R A ) =C(R B ) P , wherein R A is deuterium, cyano, halogen, Cl - C-3 alkyl, haloalkyl, heteroalkyl, -C(0)N(R 5 ) 2 , or hydroxyalkyl, p is two, each R B is hydrogen. In one embodiment, R A is halogen. In one embodiment, the halogen is fluorine or chlorine. In one embodiment, R A is haloalkyl. In one embodiment, the haloalkyl is

trifluoromethyl. In one embodiment, R A is cyano. In one embodiment, R A is heteroalkyl. In one embodiment, the heteroalkyl is methoxy. In one embodiment, R A is hydroxyalkyl. [0172] In one embodiment, R 1 is -C(0)C(R A ) - C(R B ) P , wherein - is a double bond and R A is deuterium, p is two and at least one R B is deuterium.

[0173] In one embodiment, R 1 is -C(0)C(R A ) C(R B ) P , wherein .— is a double bond and p is two, one R B is hydrogen and R A and one R B and the carbon atoms to which they are attached form a 5-8 membered partially saturated cycloalkyl substituted with oxo.

[0174] In one embodiment, R 1 is -C(0)C(R A ) . C(R B ) P , wherein : = is a double bond and p is two, one R B is hydrogen, the second R B is dialkylaminylalkyl, and R A is halogen.

[0175] In one embodiment, X is a saturated bridged ring system. Nonlimiting examples of bridged ring systems include diazabicycloheptancs and diazabicyclooctanes. In certain embodiments, when X is a saturated bridged ring system, R 1 is -C(0)CH=CH 2 . ln one embodiment, the bridged ring system is substituted with one or two groups independently selected from R 8 , where R 8 is as defined for Formula I. In one embodiment, the bridged ring system is unsubstituted ln one embodiment, the bridged ring system is

diazabicy clo [3.2.1 ]octan- 8 -y 1 or diazabicyclo [3.2.1 ] octan-3 -y 1.

[0176] In one embodiment, R'-X is:

[0177] wherein A and B are a spirocyclic ring system, wherein A and B are the same or different and independently represent a 4-6 membered saturated ring systems, wherein the rings are optionally substituted with one or more R 8 , wherein R 8 is as defined for Formula I. In certain embodiments, R 1 is -C(0)CH=CH 2 . In certain embodiments, rings A and B are unsubstituted.

[0178] In one embodiment, the spirocyclic ring system is unsubstituted. Non-limiting examples of spirocyclic ring systems include:

[0179] In certain embodiments when A and B represent a spirocyclic ring system, R 1 is

C(0)CH=CH 2 .

[0180] In one embodiment of Formula I, R 2 is selected from the group consisting of

hydroxyalkyl, dialkylaminylalkyl, heterocyclyl and heterocyclylalkyl, wherein each of the heterocyclyl or heterocyclylalkyl are independently optionally substituted with R 9 . In another embodiment, R 2 is heterocyclyl and heterocyclylalkyl, wherein each of the heterocyclyl or heterocyclylalkyl are independently optionally substituted with one or more R 9 . In certain embodiments, R 2 is dialkylaminylalkyl optionally substituted with one or more R 9 . Non-limiting examples include dimethylaminylethyl, dimethylaminylpropanyl, dimethylaminylpropan-2-yl, dimethylaminylbutanyl, dimethylaminylbutan-2-yl, 2-dimethylaminylpropanol, or

diethylaminylethyl.

[0181] In one embodiment, Y is O and R 2 is selected from the group consisting of hydroxyalkyl, dialkylaminylalkyl, heterocyclyl, heterocyclylalkyl, and -ZR 5 R 10 , wherein R 3 and R 10 are as defined for Formula 1.

[0182] In one embodiment, Y is O and R 2 is selected from the group consisting of hydroxy alkyl, dialkylaminylalkyl, heterocyclyl and heterocyclylalkyl, wherein each of the heterocyclyl or heterocyclylalkyl are independently optionally substituted with R 9 . In another embodiment, R 2 is heterocyclyl and heterocyclylalkyl, wherein each of the heterocyclyl or heterocyclylalkyl are independently optionally substituted with one or more R 9 . Non-limiting examples of R 9 include acyl, oxo, halogen, cyano, Cl - C6 alkyl, alkoxy, hydroxyalkyl, heteroalkyl, cycloalkyl, aralkyl or dialkylamidoalkyl. In certain embodiments, R 2 is dialkylaminylalkyl optionally substituted with one or more R 9 . Non-limiting examples include dimethylaminylethyl,

dimethylaminylpropanyl, dimethylaminylpropan-2-yl, dimethylaminylbutanyl,

dimethylaminylbutan-2-yl, 2-dimethylaminylpropanol, or diethylaminylethyl.

[0183] In one embodiment of Formula I, R 4 is aryl optionally substituted with one or more R 6 or R 7 . In one embodiment, R 4 is phenyl or naphthyl optionally substituted with one or more R 6 or R 7 . In one embodiment, R 4 is phenyl or naphthyl optionally substituted with one or more R 7 . In one embodiment, R 4 is phenyl or naphthyl optionally substituted with one or more R 7 substituents independently selected from halogen, hydroxyl, Cl - C3alkyl, cycloalkyl, alkoxy, haloalkyl, or Q-haloalkyl wherein Q is O or S. In one embodiment, R 4 is phenyl or naphthyl optionally substituted with one or more R 7 substituents independently selected from methyl, triiluoromethyl, hydroxyl, trifluoromethoxy, hydroxyl, fluoro, chloro, isopropyl, cyclopropyl and methylthio.

[0184] In one embodiment, R 4 is isoquinolinyl which is optionally substituted with amino. In one embodiment, R 4 is aralkyl. In certain embodiments, the aralkyl is benzyl. In one embodiment, the aralkyl is benzyl wherein the alkyl portion is substituted with hydroxyl or hydroxyalkyl.

[0185] Nonlimiting examples of compounds of Formula (I), Formula I-A and Formula I-B are selected from the group consisting of:

[0186] and pharmaceutically acceptable salts thereof.

[0187] In one embodiment, the compounds of Formula I include trifluoroacetic acid salts of the above compounds. The compounds of Formula (I), Formula I-A, Formula I-B, may be formulated into pharmaceutical compositions.

PHARMACEUTICAL COMPOSITIONS

[0188] In another aspect, the invention provides pharmaceutical compositions comprising a KRas G12C inhibitor according to the invention and a pharmaceutically acceptable carrier, excipient, or diluent. Compounds of the invention may be formulated by any method well known in the art and may be prepared for administration by any route, including, without limitation, parenteral, oral, sublingual, transdermal, topical, intranasal, intratracheal, or intrarectal. In certain embodiments, compounds of the invention are administered intravenously in a hospital setting.

In one embodiment, administration may be by the oral route.

[0189] The characteristics of the carrier will depend on the route of administration. As used herein, the term "pharmaceutically acceptable" means a non-toxic material that is compatible with a biological system such as a cell, cell culture, tissue, or organism, and that does not interfere with the effectiveness of the biological activity of the active ingredient(s). Thus, compositions according to the invention may contain, in addition to the inhibitor, diluents, fillers, salts, buffers, stabilizers, solubilizers, and other materials well known in the art. The preparation of pharmaceutically acceptable formulations is described in, e.g., Remington's Pharmaceutical Sciences, 18th Edition, ed. A. Gennaro, Mack Publishing Co., Easton, Pa., 1990.

[0190] As used herein, the term pharmaceutically acceptable salt refers to salts that retain the desired biological activity of the above -identified compounds and exhibit minimal or no undesired toxicological effects. Examples of such salts include, but are not limited to acid addition salts formed with inorganic acids (for example, hydrochloric acid, hydrobromic acid, sulfuric acid, phosphoric acid, nitric acid, and the like), and salts formed with organic acids such as acetic acid, oxalic acid, tartaric acid, succinic acid, malic acid, ascorbic acid, benzoic acid, tannic acid, pamoic acid, alginic acid, polyglutamic acid, naphthalenesulfonic acid,

naphthalenedisulfonic acid, and polygalacturonic acid. The compounds can also be administered as pharmaceutically acceptable quaternary salts known by those skilled in the art, which specifically include the quaternary ammonium salt of the formula— NR+Z-, wherein R is hydrogen, alkyl, or benzyl, and Z is a counterion, including chloride, bromide, iodide, -O-alkyl, toluenesulfonate, methylsulfonate, sulfonate, phosphate, or carboxylate (such as benzoate, succinate, acetate, glycolate, maleate, malate, citrate, tartrate, ascorbate, benzoate, cinnamoate, mandeloate, benzyloate, and diphenylacetate).

[0191] The active compound is included in the pharmaceutically acceptable carrier or diluent in an amount sufficient to deliver to a patient a therapeutically effective amount without causing serious toxic effects in the patient treated. In one embodiment, a dose of the active compound for all of the above-mentioned conditions is in the range from about 0.01 to 300 mg/kg, for example 0.1 to 100 mg/kg per day, and as a further example 0.5 to about 25 mg per kilogram body weight of the recipient per day. A typical topical dosage will range from 0.01-3% wt/wt in a suitable carrier. The effective dosage range of the pharmaceutically acceptable derivatives can be calculated based on the weight of the parent compound to be delivered. If the derivative exhibits activity in itself, the effective dosage can be estimated as above using the weight of the derivative, or by other means known to those skilled in the art.

[0192] The pharmaceutical compositions comprising compounds of the present invention may be used in the methods of use described herein.

METHODS OF USE

[0193] In yet another aspect, the invention provides for methods for inhibiting KRas G12C activity in a cell, comprising contacting the cell in which inhibition of KRas G12C activity is desired with an effective amount of a compound of Formula (II), Formula II-A, or Formula II-B, pharmaceutically acceptable salts thereof or pharmaceutical compositions containing the compound or pharmaceutically acceptable salt thereof ln one embodiment, the contacting is in vitro. In one embodiment, the contacting is in vivo.

[0194] As used herein, the term "contacting" refers to the bringing together of indicated moieties in an in vitro system or an in vivo system. For example, "contacting" a KRas G12C with a compound provided herein includes the administration of a compound provided herein to an individual or patient, such as a human, having KRas G12C, as well as, for example, introducing a compound provided herein into a sample containing a cellular or purified preparation containing the KRas G12C.

10195] In one embodiment, a cell in which inhibition of KRas G12C activity is desired is contacted with an effective amount of a compound of Formula (II), Formula II -L, or Formula II- B, to negatively modulate the activity of KRas G12C. In other embodiments, a therapeutically effective amount of pharmaceutically acceptable salt or pharmaceutical compositions containing the compound of Formula (II), Formula II-A, or Formula II-B, may be used.

[0196] By negatively modulating the activity of KRas G12C, the methods described herein are designed to inhibit undesired cellular proliferation resulting from enhanced KRas G12C activity within the cell. The cells may be contacted in a single dose or multiple doses in accordance with a particular treatment regimen to effect the desired negative modulation of KRas G12C. The degree of covalent modification of KRas GI2C may be monitored in vitro using well known methods, including those described in Example A below ln addition, the inhibitory activity of exemplary compounds in cells may be monitored, for example, by measuring the inhibition of KRas G12C activity of the amount of phosphylated ERK, including those described in Example B below, to assess the effectiveness of treatment and dosages may be adjusted accordingly by the attending medical practitioner.

[0197] In another aspect, methods of treating cancer in a patient in need thereof, comprising administering to said patient a therapeutically effective amount of a compound of Formula (I), Formula II-A, or Formula II-B, pharmaceutically acceptable salts thereof or pharmaceutical compositions comprising the compound or pharmaceutically acceptable salts thereof are provided.

[0198] The compositions and methods provided herein may be used for the treatment of a KRas G12C-associated cancer in a patient in need thereof, comprising administering to said patient a therapeutically effective amount of a compound of Formula (II), Formula II-A, or Formula II-B, pharmaceutically acceptable salts thereof or pharmaceutical compositions comprising the compound or pharmaceutically acceptable salts thereof are provided. In one embodiment, the KRas G12C-associated cancer is lung cancer. [0199] The compositions and methods provided herein may be used for the treatment of a wide variety of cancers including tumors such as lung, prostate, breast, brain, skin, cervical carcinomas, testicular carcinomas, etc. More particularly, cancers that may be treated by the compositions and methods of the invention include, but are not limited to tumor types such as astrocytic, breast, cervical, colorectal, endometrial, esophageal, gastric, head and neck, hepatocellular, laryngeal, lung, oral, ovarian, prostate and thyroid carcinomas and sarcomas. More specifically, these compounds can be used to treat: Cardiac: sarcoma (angiosarcoma, fibrosarcoma, rhabdomyosarcoma, liposarcoma), myxoma, rhabdomyoma, fibroma, lipoma and teratoma; Lung: bronchogenic carcinoma (squamous cell, undifferentiated small cell,

undifferentiated large cell, adenocarcinoma), alveolar (bronchiolar) carcinoma, bronchial adenoma, sarcoma, lymphoma, chondromatous hamartoma, mesothelioma; Gastrointestinal: esophagus (squamous cell carcinoma, adenocarcinoma, leiomyosarcoma, lymphoma), stomach (carcinoma, lymphoma, leiomyosarcoma), pancreas (ductal adenocarcinoma, insulinoma, glucagonoma, gastrinoma, carcinoid tumors, vipoma), small bowel (adenocarcinoma, lymphoma, carcinoid tumors, Kaposi's sarcoma, leiomyoma, hemangioma, lipoma, neurofibroma, fibroma), large bowel (adenocarcinoma, tubular adenoma, villous adenoma, hamartoma, leiomyoma); Genitourinary tract: kidney (adenocarcinoma, Wilm's tumor (nephroblastoma), lymphoma, leukemia), bladder and urethra (squamous cell carcinoma, transitional cell carcinoma, adenocarcinoma), prostate (adenocarcinoma, sarcoma), testis (seminoma, teratoma, embryonal carcinoma, teratocarcinoma, choriocarcinoma, sarcoma, interstitial cell carcinoma, fibroma, fibroadenoma, adenomatoid tumors, lipoma); Liver: hepatoma (hepatocellular carcinoma), cholangiocarcinoma, hepatoblastoma, angiosarcoma, hepatocellular adenoma, hemangioma; Biliary tract: gall bladder carcinoma, ampullary carcinoma, cholangiocarcinoma; Bone:

osteogenic sarcoma (osteosarcoma), fibrosarcoma, malignant fibrous histiocytoma,

chondrosarcoma, Ewing's sarcoma, malignant lymphoma (reticulum cell sarcoma), multiple myeloma, malignant giant cell tumor chordoma, osteochronfroma (osteocartilaginous exostoses), benign chondroma, chondroblastoma, chondromyxofibroma, osteoid osteoma and giant cell tumors; Nervous system: skull (osteoma, hemangioma, granuloma, xanthoma, osteitis deformans), meninges (meningioma, meningiosarcoma, gliomatosis), brain (astrocytoma, medulloblastoma, glioma, ependymoma, germinoma (pinealoma), glioblastoma multiform, oligodendroglioma, schwannoma, retinoblastoma, congenital tumors), spinal cord neurofibroma, meningioma, glioma, sarcoma); Gynecological: uterus (endometrial carcinoma), cervix (cervical carcinoma, pre -tumor cervical dysplasia), ovaries (ovarian carcinoma (serous

cystadenocarcinoma, mucinous cystadenocarcinoma, unclassified carcinoma), granulosa-thecal cell tumors, Sertoli-Leydig cell tumors, dysgerminoma, malignant teratoma), vulva (squamous cell carcinoma, intraepithelial carcinoma, adenocarcinoma, fibrosarcoma, melanoma), vagina (clear cell carcinoma, squamous cell carcinoma, botryoid sarcoma (embryonal

rhabdomyosarcoma), fallopian tubes (carcinoma); Hematologic: blood (myeloid leukemia (acute and chronic), acute lymphoblastic leukemia, chronic lymphocytic leukemia, myeloproliferative diseases, multiple myeloma, myelodysplastic syndrome), Hodgkin's disease, non-Hodgkin's lymphoma (malignant lymphoma); Skin: malignant melanoma, basal cell carcinoma, squamous cell carcinoma, Kaposi's sarcoma, moles dysplastic nevi, lipoma, angioma, dermatofibroma, keloids, psoriasis; and Adrenal glands: neuroblastoma. In certain embodiments, the cancer is non-small cell lung cancer.

[0200] The concentration and route of administration to the patient will vary depending on the cancer to be treated. The compounds, pharmaceutically acceptable salts thereof and

pharmaceutical compositions comprising such compounds and salts also may be co-administered with other anti -neoplastic compounds, e.g., chemotherapy, or used in combination with other treatments, such as radiation or surgical intervention, either as an adjuvant prior to surgery or post-operatively.

[0201] Also provided herein is a compound of Formula 1, Formula I-A, Formula 1 -B, or a pharmaceutically acceptable salt or solvate thereof, or a pharmaceutical composition thereof as defined herein for use in therapy.

[0202] Also provided herein is a compound of Formula I, Formula I-A, Formula 1-B, or a pharmaceutically acceptable salt or solvate thereof or a pharmaceutical composition thereof as defined herein for use in the treatment of cancer.

[0203] Also provided herein is a compound of Formula I, Formula I-A, Formula 1 -B, or a pharmaceutically acceptable salt or solvate thereof for use in the inhibition of KRas G12C. [0204] Also provided herein is a compound of Formula I, Formula I-A, Formula l-B, or a pharmaceutically acceptable salt or solvate thereof or a pharmaceutical composition thereof as defined herein, for use in the treatment of a KRas Gl2C-associated disease or disorder.

[0205] Also provided herein is the use of a compound of Formula I, Formula I-A, Formula l-B, or a pharmaceutically acceptable salt or solvate thereof, as defined herein in the manufacture of a medicament for the treatment of cancer.

[0206] Also provided herein is a use of a compound of Formula I, Formula I-A, Formula l-B, or a pharmaceutically acceptable salt or solvate thereof, as defined herein in the manufacture of a medicament for the inhibition of activity of KRas G12C.

[0207] Also provided herein is the use of a compound of Formula I, Formula I-A, Formula l-B, or a pharmaceutically acceptable salt or solvate thereof, as defined herein, in the manufacture of a medicament for the treatment of a KRas Gl2C-associated disease or disorder.

[0208] Also provided herein is a method for treating cancer in a patient in need thereof, the method comprising (a) deteimining that cancer is associated with a KRas G12C mutation (e.g., a KRas Gl2C-associated cancer) (e.g., as determined using a regulatory agency-approved, e.g., FDA-approved, assay or kit); and (b) administering to the patient a therapeutically effective amount of a compound of Formula I, Formula I-A, Formula l-B, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof.

[0209] One skilled in the art will recognize that, both in vivo and in vitro trials using suitable, known and generally accepted cell and/or animal models are predictive of the ability of a test compound to treat or prevent a given disorder.

[0210] One skilled in the art will further recognize that human clinical trials including first-in human, dose ranging and efficacy trials, in healthy patients and/or those suffering from a given disorder, may be completed according to methods well known in the clinical and medical arts.

REACTION SCHEMES AND EXAMPLES

[021 1 ] The compounds of the present invention may be prepared from commercially available reagents using the synthetic methods and reaction schemes described herein, or using other reagents and conventional methods well known to those skilled in the art.

[0212] For instance, compounds of the present invention may be prepared according to the General Reaction Schemes I & II.

GENERAL REACTION SCHEMES

SCHEME I

[0213] Compounds of Formula I wherein L is a bond, -Y-R 2 is other than hydrogen and R 4 is aryl or heteroaryl can be prepared according to Scheme 1. In step A, an appropriately

functionalized dihydropyridopyrimidine (6) is coupled to a heterocycle containing one nucleophilic amine species, with the other bound to a protecting group to provide compound (7). This coupling proceeds in a solvent such as dimethylacetamide in the presence of a base such as triethylamine or Hunig’s base in step B, the substituent -Y-R 2 is introduced by substitution of the activated sulfoxide by a nucleophile, for example (S)-l-(dimethylamino-propan-2-ol in a polar solvent such as dioxane to provide compound (8). ln step C, the Boc group is removed using conditions known in the art, for example with trifluoroacetic acid in a solvent such as dichloromethane to provide compound (9). In step D, the substituent R 4 is introduced with a palladium coupling, using a suitable functionalized aryl or heteroaryl system, for example an aryl triflate, in the presence of a palladium catalyst such as Pd 2 DBA3/BINAP in a solvent such as toluene with a base such as sodium tert-butoxide to provide compound (10). In step E, the protecting group of ring X is removed, for example hydrogenolysis by Pd/C in the presence of ¾ in a polar solvent such as EtOII/THF to provide compound (11). In step F, R 1 is introduced to provide a compound of Formula 1, for example by treating with an acid chloride having the formula C1-C(0)C(R A ) C(R B ) P or C1-S0 2 C(R A ) === C(R B ) P or an anhydride having the formula are as defined for Formula I. For example, in the case where R 1 is an acryloyl group, this reaction proceeds, for example, in a solvent such as methylene chloride in the presence of acryloyl chloride acryloyl anhydride and a base such as Ilunig’s base. In some cases, the species R 4 and R 2 may also contain protecting groups, which can be removed at a subsequent step in the synthetic sequence.

[0214 j Compounds (1), (2), (3), (4), (5) and (6) as shown and described above for Scheme I are useful as intermediates for preparing compounds of Formula I, Formula I-A or Formula I-B and are provided as further aspects of the invention.

SCHEME II [0215 ] Compounds of Formula I wherein L and Y are bonds, R 2 is hydrogen and R 4 is aryl or heteroaryl can be prepared according to Scheme II. In step A, an appropriately functionalized bicycle (1) is coupled to a heterocycle containing one nucleophilic amine species, with the other bound to a protecting group to provide compound (2). This coupling proceeds in a solvent such as dichloromethane in the presence of a base such as triethylamine or Hunig’s base. In step B, the Boc group of compound (2) is removed using conditions known in the art, for example with trifluoroacetic acid in a solvent such as dichloromethane, to provide compound (3). In step C, the substituent R 4 is introduced with a palladium coupling, using a suitable functionalized aryl or heteroaryl system, for example an aryl triflate, in the presence of a palladium catalyst such as Pd2DBA 3 /Xantphos in a solvent such as toluene with a base such as sodium tert-butoxide to provide compound (4). In step D, the protecting group of ring X compound (4) is removed, for example hydrogenolysis by Pd/C in the presence of H 2 in a polar solvent such as EtOII/THF to provide compound (5). In the final step, E, R' is introduced to provide a compound of Formula

1, for example by treating with an acid chloride having the formula C1-C(C))C(R A ) .— C(R B ) P or C1-S0 2 C(R A ) = C(R B ) P, or an anhydride having the formula C(R B ) P - are as defined for Formula I. For example, in the case where R 1 is an acryloyl group, this reaction proceeds, for example, in a solvent such as methylene chloride in the presence of acryloyl chloride or an acryloyl anhydride and a base such as Hunig’s base. In some cases, the species R 4 will also contain a protecting group, which can be removed at a subsequent step in the synthetic sequence.

[0216] Compounds (7), (8), (9), (10) and (1 1 ) as shown and described above for Scheme II are useful as intermediates for preparing compounds of Formula I and are provided as further aspects of the invention.

[0217] Accordingly, also provide is a process for preparing a compound of Formula I, comprising: for a compound of Formula I where -Y-R 2 is other than hydrogen, reacting a compound of formula 5

5

[0218] where X, R’. R 4 , L and ra are as defined for Formula I with an acid chloride having the formula C1-C(0)C(R A ) = C(R B ) P or C1-S0 2 C(R A ) = C(R B ) P or an anhydride having the formula C(R B ) P == C(R A )C(0)0C(0)C(R A ) == C(R B ) P where R A , R B and p are as defined for Formula I, in the presence of a base; and

[0219] optionally forming a salt thereof.

[0220] The compounds of the present invention may have one or more chiral center and may be synthesized as stereoisomeric mixtures, isomers of identical constitution that differ in the arrangement of their atoms in space. The compounds may be used as mixtures or the individual components/isomers may be separated using commercially available reagents and conventional methods for isolation of stereoisomers and enantiomers well-known to those skilled in the art, e.g., using CFIIRALPAK® (Sigma- Aldrich) or CHIRALCEL® (Diacel Corp) chiral chromatographic HPLC columns according to the manufacturer's instructions. Alternatively, compounds of the present invention may be synthesized using optically pure, chiral reagents and intermediates to prepare individual isomers or enantiomers. Unless otherwise indicated, all chiral (enantiomeric and diastereomeric) and racemic forms are within the scope of the invention. Unless otherwise indicated, whenever the specification, including the claims, refers to compounds of the invention, the term“compound” is to be understood to encompass all chiral (enantiomeric and diastereomeric) and racemic forms.

[0221] The following Examples are intended to illustrate further certain embodiments of the invention and are not intended to limit the scope of the invention. Intermediate 1

3-(methoxymethoxy)naphthalen- 1 -yl trifluoromethanesulfonate

[0222] 3-Hydroxynaphthalen-l-yl trifluoromethanesulfonate (13.101 g, 44.831 mmol) was dissolved in dichloromethane (100 mL) and stirred at 0 °C. To this solution was added chloro(methoxy)methane (3.7456 ml, 49.315 mmol) and Hunig's base (1 1.745 mL, 67.247 mmol). The reaction was stirred at 0 °C for 4 hrs. The reaction was partitioned with 1M HC1 and washed with saturated sodium bicarbonate. The combined organic layers were dried over magnesium sulfate and concentrated under vacuum. The concentrated material was loaded onto a 120 g RediSep® gold silica gel column with dichloromethane and purified by normal phase chromatography (CombiFlash®, 0%-20% ethyl acetate/hexanes as the eluent) to give 3- (methoxymethoxy)naphthalen- 1 -yl trifluoromethanesulfonate (1 1.785 g, 35.045 mmol, 78.171 % yield).

Intermediate 2

2-bromo-7-(methoxymethoxy)naphthalenc

[0223] To a solution of 7-bromonaphthalen-2-ol (2.0 g, 9.0 mmol) in dimethyl acetamide (40 mL) was added chloro(methoxy)methane (1.4 g, 18 mmol) and cesium carbonate (5.8 g, 18 mmol) and the reaction mixture was stirred overnight at room temperature. The reaction was diluted with water and the aqueous layer washed with ethyl acetate. The combined organic layers were washed with water and brine, dried over magnesium sulfate and concentrated under vacuum. The crude material was purified by normal phase chromatography using 5-50% ethyl acetate/hexanes as the eluent to give 2-bromo-7-(methoxymethoxy)naphthalene (1.0 g, 3.7 mmol, 42 % yield). lntermediate 3

[0224] To a stirred solution of 2-bromo-3-fluorophenol (1422 mg, 7.445 mmol) in 22 mL tetrahydrofuran at room temperature under nitrogen was added NaH (327.6 mg, 8.190 mmol) neat as a solid portion wise. After 15 minutes, a solution had formed. Chloro(methoxy)methane (678.6 pL, 8.934 mmol) was added by syringe. After stirring for 2 hours, the reaction was quenched with saturated ammonium chloride solution and then partitioned between ethyl acetate (30 mL) and water (30 mL). The combined organic layers were isolated, washed with brine, dried over MgS0 4 , filtered and concentrated. The crude product was loaded in a minimum of dichloromethane onto a 40 gram RediSep® column pre-wet with hexanes and eluted with an ethyl acetate/hexanes gradient (0% to 20% ethyl acetate). Fractions containing the product were combined and concentrated to provide the product as a clear oil (1.45g, 83%).

Intermediate 4

2-bromo- 1 -fluoro-4-(methoxymethoxy)benzene

[0225] To a stirred solution of 3-bromo-4-fluorophenol (327 mg, 1.71 mmol) in 5.1 mL tetrahydrofuran at room temperature under nitrogen was added NaH (75.3 mg, 1.88 mmol) neat as a solid portion wise. After 15 minutes, a solution had formed. Chloro(methoxy)methane (156 qL, 2.05 mmol) was added by syringe. After stirring for 2 hours, the reaction was quenched with saturated ammonium chloride solution and partitioned between ethyl acetate and water. The combined organic layers were washed with brine, dried over MgS0 4 , filtered and concentrated. The crude product was loaded in a minimum of dichloromethane onto a 24 gram RediSep® column pre-wet with hexanes and eluted with an ethyl acetate/hexanes gradient (0% to 20% ethyl acetate). Fractions containing the product were combined and concentrated to provide the product as a clear oil (120 mg, 29.8%)

Intermediate 5

4-bromo-5-methyl-l-((2-(trimethylsilyl)ethoxy)methyl)-lH-ind azole

[0226] To a solution of 4-bromo-5 -methyl- lH-indazole (0.7 g, 3.3 mmol) in dimethyl acetamide (30 mL) cooled to 0 °C was added NaH (0.19 g, 4.6 mmol) in portions and the reaction mixture was purged with nitrogen. The reaction was stirred for 20 minutes, and then (2- (chloromethoxy)ethyl)trimethylsilane (0.83 g, 5.0 mmol) was added and the reaction was stirred for 2 hours while warming to room temperature. The reaction was quenched by pouring into water and the aqueous layer was extracted into ethyl acetate. The combined organic layers were washed with water and brine, dried over MgS0 4 and concentrated under vacuum. The crude material was purified by chromatography using 10-50% ethyl acetate/hexanes as the eluent to give 4-bromo-5 -methyl- 1 -((2-(trimethylsilyl)ethoxy)methyl)-lH-indazole (0.87 g, 79%).

Intermediate 6

(R)- 1 -(pyrrolidin- 1 -yl)propan-2-ol

[0227] In a sealed tube, R-(+)-Propylene oxide (3.69 mL, 52.7 mmol) was cooled to -78°C and then sparged with anhydrous dimethyl amine for a few minutes. The reaction mixture was heated to 70°C for 16 hours. The reaction was cooled and concentrated in vacuo for 20 minutes to provide (R)-l -(pyrrolidin- l-yl)propan-2-ol (5.35 g, 41.4 mmol, 98.2% yield).

Intermediate 7

(R)- 1 -morpholinopropan-2-ol

[0228] In a sealed tube, R-(+)-Propylene oxide (2.1 11 mL, 30.13 mmol) and morpholine (1.490 mL, 17.22 mmol) were heated to 70°C for 20 hours. The reaction was cooled and concentrated in vacuo to provide (R)-l -morpholinopropan-2-ol (2.47 g, 17.01 mmol, 98.80 % yield).

Intermediate 8

(R)- 1 -(dimethylamino)butan-2-ol [0229] In a sealed tube, R-(+)-Propylene oxide (4.00 g, 55.5 mmol) and dimethylamine (1.00 g, 22.2 mmol), were heated to 65°C for 18 hours. The reaction was cooled and concentrated in vacuo. The resulting residue was purified by silica gel (0-12% MeOH in DCM) to provide (R)- l-(dimethylamino)butan-2-ol (1.38 g, 1 1 .8 mmol, 53.1 % yield). lntermediate 9

(R)-l-((R)-3-methoxypyrrolidin-l -yl)propan-2-ol

[0230] In a sealed tube, (R)-3-methoxypyrrolidine hydrochloride (1.00 g, 7.27 mmol), TEA (2.03 mL, 14.5 mmol) and R-(+)-Propylene oxide (1.27 mL, 18.2 mmol) were heated to 65°C for 18 hours. The reaction was cooled and concentrated in vacuo. The resulting residue was purified by silica gel (0-12% MeOTI in DCM) to provide (R)-l-((R)-3-methoxypyrrolidin-l- yl)propan-2-ol (775 mg, 4.87 mmol, 67.0 % yield). lntermediate 10

(R)- 1 -((S)-3 -methoxypyrrolidin- 1 -yl)propan-2-ol

[0231] In a sealed tube, (S)-3-methoxypyrrolidine hydrochloride (1.00 g, 7.27 mmol), TEA (2.03 mL, 14.5 mmol) and R-(+)-Propylene oxide ( 1.27 mL, 1 8.2 mmol) were heated to 65°C for 18 hours. The reaction was cooled and concentrated in vacuo. The resulting residue was purified by silica gel (0-12% MeOH in DCM) to provide (R)-l -((S)-3-methoxypyrrolidin-l- yl)propan-2-ol (781 mg, 4.90 mmol, 67.5 % yield)

Intermediate 1 1

(R)-l -((S)-3-((tert-butyldimethylsilyl)oxy)pyrrolidin-l-yl)propan -2-ol

[0232] In a sealed tube, R-(+)-Propylene oxide (0.609 mL, 8.69 mmol) and (S)-3-((tert- butyldimethylsilyl)oxy)pyrrolidine (1.00 g, 4.97 mmol) were heated to 70°C for 20 hours. The reaction was cooled and concentrated in vacuo to provide (R)-l-((S)-3-((tert- butyldimethylsilyl)oxy)pyrrolidin-l-yl)propan-2-ol (1.29 g, 4.20 mmol, 84.6 % yield).

Intermediate 12

Boc tert-butyl 2-(hydroxymethyl)-4-methylpiperazine- 1 -carboxylate

[0233] To a suspension of lithium chloride (246 mg, 5.81 mmol) and Lithium Borohydride (126 mg, 5.81 mmol) in ethanol (9 mL), at 0°C under nitrogen, a solution of 1 -(tert-butyl) 2-methyl 4- methylpiperazine-1 ,2-dicarboxylate (750 mg, 2.90 mmol) in dry THF (6 mL) was added dropwise. The reaction was stirred overnight forming a white precipitate. The precipitate was filtered and washed with ethanol. The combined filtrate and organic extracts were concentrated to provide a white residue which was extracted with ethyl acetate. The combined organic layers were washed with saturated sodium chloride solution, dried over sodium sulfate and

concentrated in vacuo. The residue was purified by chromatography with isocratic 10% MeOH in DCM with 0.2% NLLOH to provide tert-butyl 2-(hydroxymethyl)-4-methylpiperazine-l- carboxylate (104 mg, 0.452 mmol, 15.6 % yield).

Intermediate 13

(S)-2-(2-metbylpiperidin- 1 -yl)ethan-l -ol [0234] A mixture of (S)-2-methylpiperidine (100 mg, 1.01 mmol), 2-bromoethanol (78 pL, 139 mg, 1.11 mmol, 1.1 eq.), sodium iodide (151 mg, 1 eq.), potassium carbonate (418 g, 3 eq.) and acetonitrile (1 mL) in a 4-mL vial was purged with nitrogen, sealed and stirred at room temperature for 2 days. The reaction mixture was partitioned between diethyl ether (15 mL) and water (2 mL). The ether layer was washed with brine (2 mL), acidified with TFA and dried under high vacuum for 2 days. The residue was washed with ether (3 mL), diluted with water (0.5 mL) and 10M NaOH was added (0.2 mL). The layers were separated and the upper layer was carefully dried over NaOH. The ether solution was evaporated under nitrogen to yield crude (S)- 2-(2-methylpiperidin-l-yl)ethan-l-ol (100 mg, 0.698 mmol, 69.24% yield) as colorless oil.

Intermediate 14

(R)-2-(2-methylpiperidin-l -yl)ethan-l -ol

[0235] Synthesized according to the method of Intermediate 13, using (R)-2-methylpiperidine (99 mg, 1 mmol) in place of (S)-2-methylpiperidine.

Intermediate 15

(S)-2-(3-methoxypiperidin-l-yl)ethan-l-ol

[0236] Synthesized according to the method of Intermediate 13, using (S)-3-methoxypiperidine (173 mg, 1.50 mmol) in place of (S)-2-methylpiperidine.

Intermediate 16

(R)-2-(3-methoxypiperidin- l -yl)ethan-l -ol

[0237] Synthesized according to the method of Intermediate 13, using R-3-methoxypiperidine (173 mg, 1.50 mmol) in place of (S)-2-methylpiperidine.

Intermediate 17

3 -( 1 ,4-oxazepan-4-y l)propan- 1 -ol

[0238] To a vial was added homomorpholine (0.250 g, 2.472 mmol), Acetonitrile (4.943 mL, 2.472 mmol) and 3 -Bromo- 1 -propanol (0.2459 mL, 2.719 mmol). Potassium carbonate (0.6832 g, 4.943 mmol) was added and the mixture was warmed to 50 °C and stirred for 6 hours. The mixture was cooled to ambient temperature, diluted with DCM, filtered and the collected solids were washed with DCM. The filtrate was concentrated in vacuo and the crude oil was purified via column chromatography (Biotage Isolera, 12g Isco RediSep Gold, 10-20% MeOH/DCM with 0.2% NH 4 OH) to afford 3-(l ,4-oxazepan-4-yl)propan- l -ol (0.272 g, 1 .708 mmol) as a colorless oil.

Intermediate 1 8

3-((l S,4S)-2-oxa-5-azabicyclo[2.2. l jheptan-5-yl)propan- l -ol [0239] Synthesized according to the method of Intermediate 17, using (1 S,4S)-2-Oxa-5- azabicyclo[2.2.1]heptane (0.250 g, 2.522 mmol) in place of homomorpholine.

Intermediate 19

2-(4-methoxypiperidin- 1 -yl)ethan- 1 -ol

[0240] Synthesized according to the method of Intermediate 13, using 4-methoxypiperidine (173 mg, 1.50 mmol) in place of (S)-2-methylpiperidine.

Intermediate 20

2-(4,4-difluoropiperidin- 1 -yl)ethan- 1 -ol

[0241] Synthesized according to the method of Intermediate 13, using 4,4-difluoropiperidine hydrochloride (173 mg, 1.50 mmol) in place of (S)-2-methylpiperidine.

Intermediate 21

(S)-2-(3 -fluoropiperidin- 1 -yl)ethan- 1 -ol

[0242] Synthesized according to the method of Intermediate 13, using S-3-fluoropiperidine hydrochloride (209 mg, 1.50 mmol) in place of (S)-2-methylpiperidine. lntermediate 22

(R)- 1 -(4-(2-hydroxypropyl)piperazin- 1 -yl)ethan- 1 -one

[0243] Step A: 1 -|4-[(2// )-2- [pipcrazin-l-yl lcthanonc: (2i?)-2-methyloxirane

(1.00 g, 17.2 mmol, 1.20 mL, 1.00 eq) and 1-piperazin-l-ylethanone (8.00 g, 62.4 mmol, 3.62 eq ) were taken up into a microwave tube. The sealed tube was heated at 150 °C for 1 hour under microwave. The mixture was dissolved in DCM (80.0 mL), added (Boc) 2 0 (3.62 eq,l3.6 g) and stirred at 20 °C for 1 hour. The residue was purified by column chromatography (DCM/MeOH 100/1 to 10/1) to give l-[4-[(2/?)-2-hydroxypropyl]piperazin-l-yl]ethanone (3.80 g, 13.5 mmol, 78.2 % yield, 66.0 % purity) as a yellow oil.

Intermediate 23

l -(benzyloxy)-3-bromo-5-cyciopropylbenzene

[0244] Step A: l -benzyloxy-3,5-dibromo-benzene: To a mixture of 3,5-dibromophenol (1.50 g, 5.95 mmol, 1 .00 eq) and K 2 CO 3 (2.47 g, 17.9 mmol, 3.00 eq) in MeCN (30.0 mL) was added benzyl bromide (1 .07 g, 6.25 mmol, 742 pL, 1 .05 eq ), the reaction mixture was stirred at 80 °C for 2 hours. The reaction mixture was filtered and concentrated. The residue was purified by column chromatography (S1O 2 , Petroleum ether/Ethyl acetate— 1 : 1 to give l -benzyloxy-3,5- dibromobenzene (1.60 g, 4.68 mmol, 78.6 % yield) as colorless oil.

[0245] Step B: 1 -benzyloxy -3 -bromo-5 -cyclopropy lbenzene : To a mixture of l-benzyloxy-3,5- dibromobenzene (1.20 g, 3.51 mmol, 1.00 eq) and cyclopropylboronic acid (392 mg, 4.56 mmol, 1.30 eq) in H 2 0 (4.00 mL) and dioxane (20.0 mL) w r as added Pd(dppf)Cl2 (513 mg, 702 pmol, 0.20 eq) and CS 2 CO 3 (2.29 g, 7.02 mmol, 2.00 eq). The reaction mixture was stirred at 90 °C for 12 hours under N 2 . The reaction mixture was added to water (20 mL) and extracted with ethyl acetate (2 x 15 mL). The combined organic layers were dried over Na 2 S0 4 , filtered and concentrated. The residue was purified by column chromatography (Si0 2 , Petroleum ether/Ethyl acetate = 1 : 1 to give l -benzyloxy-3-bromo-5-cyclopropyl -benzene (270 mg, 890 pmol, 25.4 % yield) as colorless oil. lntermediate 24

4-(benzyloxy)-2-bromo- 1 -fluorobenzene

[0246] To a solution of 3-bromo-4-fluorophcnol (4.00 g, 20.9 mmol, 1.00 eq) and K 2 C0 3 (8.68 g, 62.8 mmol, 3.00 eq) in ACN (80.0 mL) was added benzyl bromide (3.65 g, 21.4 mmol, 2.54 mL, 1.02 eq) and the reaction mixture was stirred at 60 °C for 2 hrs. The reaction mixture was filtered and concentrated in vacuum. The residue was purified by silica gel chromatography (petroleum ethenethyl acetate; gradient from 1 :0 to 10: 1) to give 4-benzyloxy-2-bromo-l- fluoro-benzene (5.02 g, 17.0 mmol, 81.0 % yield, 95 % purity) was obtained as white solid.

Intermediate 25

2-(3-fluoropyrrolidin-l -yl)ethan-l-ol

[0247] Step A: fc/7-biityl 3-fluoropyrrolidine-l -carboxylate: To a solution of /er/-butyl 3- hydroxypyrrolidine-l -carboxylate (10.0 g, 53.4 mmol, 1.00 eq) in DCM ( 150.00 mL) was added diethylaminosulfur trifluoride (DAST) (12.9 g, 80.1 mmol, 10.6 mL, 1.50 eq) at -40 °C under a nitrogen atmosphere. After stirring at - 40 °C for 2 hours, the mixture was warmed to 20 °C and stirred for 16 hours. The mixture was poured into 5% aqueous sodium bicarbonate (200 mL) and extracted with dichloromethane (2 x 100 mL). The organic layer was dried over sodium sulfate, filtered and concentrated under vacuum. The residue was purified by column chromatography over silica gel (petroleum ether/ethyl acetate 100:1 to 5:1). The desired fractions were collected and concentrated under vacuum to give te/7-butyl 3-fluoropyrrolidine-l-carboxylate (4.30 g, 22.7 mmol, 42.6 % yield) as a colorless oil. 'll NMR (400 MIIz, Chloroform-d) d = 5.27 (t, J— 3.6 Hz, 0.5H), 5.13 (t, j= 3.6 Hz, 0.5H), 3.77 - 3.38 (m, 4H), 2.26 - 2.15 (m, 1H), 2.08 - 1.85 (m, 1H), 1.46 (s, 9H).

[0248] Step B: 3 -fluoropyrrolidine : To a solution of /e/7-butyl 3-fluoropyrrolidine-l-carboxylate (4.30 g, 22.7 mmol, 1.00 eq) in DCM (50.00 mL) was added HCl/dioxane (4 M, 35.0 mL, 6.16 eq) drop wise at 0 °C. The mixture was warmed to 20 °C and stirred for 1 hour. The mixture was concentrated under vacuum. The residue was triturated with diisopropyl ether (20 mL) and the precipitate was filtered and dried under vacuum to provide 3 -fluoropyrrolidine (2.70 g, 21.5 mmol, 94.6 % yield, HC1) as a white solid. 'H NMR (400 MHz, Methanol-d 4 ) 5 = 5.51 (t, J- 3.6 Hz, 0.5H), 5.38 (t, 7=3.6 Hz, 1H), 3.66 - 3.27 (m, 5H), 2.45 - 2.12 (m, 2H).

[0249] Step C: methyl 2-(3-fluoropyrrolidin-l-yl)acetate: A suspension of 3 -fluoropyrrolidine (2.70 g, 21.5 mmol, 1.00 eq, HC1) in DCM (27.00 mL) was cooled to 0° C. Triethylamine (5.44 g, 53.8 mmol, 7.45 mL, 2.50 eq) and methyl 2-bromoacetate (3.62 g, 23.7 mmol, 2.23 mL, 1.10 eq) were added and the reaction mixture was stirred at 20 °C for 16 h. The reaction mixture was diluted with CH2CI2 (100 mL) and water (50 mL). The organic layer was washed with 5% aqueous citric acid solution (1 c 50 mL). To the water layer, saturated aqueous sodium carbonate solution was added (20 mL) and extracted with ethyl acetate (3 x 100 mL). The combined organic layers were dried over sodium sulfate and concentrated in vacuo to give methyl 2-(3- fluoropyrrolidin-l -yl)acetate (2.20 g, 13.7 mmol, 63.5 % yield). 'H NMR (400 MHz,

Chloroform-d) d = 5.22 - 5.02 (m, 1H), 3.66 (s, 3H), 3.35 (s, 2H), 3.07 - 2.93 (m, 1H), 2.91 - 2.77 (m, 2H), 2.67 (dt, j= 5.2, 8.4 Hz, 1H), 2.21 - 1.93 (m, 2H).

[0250] Step D: 2-(3-fluoropyrrolidin-l-yl)ethanol: To a solution of L1AIH4 (706 mg, 18.6 mmol, 1.50 eq) in THF (20 mL) was added a solution of methyl 2-(3-fluoropyrrolidin-l-yl)acetate (2.00 g, 12.4 mmol, 1.00 eq) in THF (10 mL) dropwise at 0 °C. The mixture was warmed up to 20 °C and stirred for 3 hours. The mixture was quenched with saturated aqueous sodium sulfate solution (1 mL). The mixture was filtered and the filtrate was concentrated under vacuum. The product was purified by silica gel chromatography using 5% MeOH in DMC. The desired fractions were collected and concentrated under vacuum to give 2-(3-fluoropyrrolidin-l- yl)ethanol (1.20 g, 9.01 mmol, 72.6 % yield) as a colorless oil. 'H NMR (400 MHz, Chloroform- d) d = 5.28 - 5.05 (m, 1H), 3.68 - 3.61 (m, 2H), 2.99 - 2.73 (m, 4H), 2.72 - 2.67 (m, 2H), 2.58 - 2.45 (m, 1H), 2.28 - 1.97 (m, 2H).

Intermediate 26

1 -(tert-butyl) 3-methyl piperazine- 1 ,3 -dicarboxylate

[0251 ] Step A: methyl piperazine-2-carboxylate: To a mixture of 1 -tert-butyl 2-methyl piperazine- l,2-dicarboxylate (5.0 g, 22.6 mmol, 1.00 eq) in MeOII (50.0 mL) was added HCl/dioxane (4.0 M, 134 mL). The reaction mixture was degassed and purged with nitrogen 3 times, and the mixture was stirred at 25 °C for 12 hours under a nitrogen atmosphere. The reaction mixture was concentrated under reduced pressure to dryness to give methyl piperazine- 2-carboxylate (4.89 g, 2HC1, crude) as a white solid, which was used directly in the next step without further purification.

[0252] Step B: 1 -(tert-butyl) 3-methyl piperazine-l ,3-dicarboxylate: To a solution of methyl piperazine-2-carboxylate (4.30 g, crude) and TEA (8.02 g, 79.2 mmol, 1 1.0 mL) in MeOH (50.0 mL) was added di-tert-butyl dicarbonate (4.32 g, 19.8 mmol, 4.55 mL). After stirring at 25 °C for 12 hours, the reaction mixture was filtered and concentrated under reduced pressure to dryness. The residue was purified by column chromatography (Si0 2 , DCM / MeOH = 1 :0 to 20: 1) to give 1 -(tert-butyl) 3-methyl piperazine- l ,3-dicarboxylate (4.80 g, 19.7 mmol, two steps, 99.0 % yield) as a colorless oil.'H NMR (400 MHz, chloroform-d) 5 = 4.10 - 3.85 (m, 1H), 3.73 (s, 3H), 3.71 - 3.65 (m, 1H), 3.47 - 3.38 (m, 1 H), 3.10 - 2.98 (m, 211), 2.78 - 2.66 (m, 1H), 2.17 (s, 1H), 1.46 (s, 9H).

Intermediate 27

4-brom onaphthalen-2 -ol

[0253] Step A: 2,4-dibromonaphthalen-l -amine: To a solution of Br 2 (246 g, 1.54 mol, 79.3 mL, 2.18 eq) in AcOH (750 mL) was added a solution of naphthalen-l -amine (101 g, 705 mmol, 99.0 mL, 1.00 eq) in AcOH (500 mL) at ambient temperature, and the reaction was stirred at 70 °C for 1 hour. The reaction mixture was cooled at room temperature and filtered. The filter cake was washed with AcOH (300 mL), then added to 20 % aqueous of NaOH (1.2 L). The mixture was stirred for 20 min and filtered. The isolated solid was washed with water (1 L) and dried under vacuum to provide 2,4-dibromonaphthalen-l -amine (200 g, 664 mmol, 94.2% yield) as gray solid. ESI MS m/z 301. 9 [M+H] + .

[0254] Step B: 4-bromo- 1 -diazonio-naphthalen-2-olate: To a solution of 2,4-dibromonaphthalen- 1 -amine (60.0 g, 199 mmol, 1.00 eq) in AcOH (900 mL) and propionic acid (150 mL) was added NaN0 2 (16.5 g, 239 mmol, 13.0 mL, 1.20 eq) portionwise at 5-8 °C over 30 min, and then the reaction mixture was stirred at 5-8 °C for 30 min. The reaction mixture was poured into ice- water (4000 mL), and the resulting solid was collected and washed with water (2 50 mL) to provide 4-bromo- l-diazonio-naphthalen-2-olate (150 g, wet crude) as gray solid which was used directly in the next step. Tl NMR (400 MHz, CDC1 3 ) d 8.12 - 8.10 (d, .7=8.4 Hz, 1H), 7.62 - 7.58 (t, J=7.6 Hz, 1H), 7.41 - 7.37 (t, J=7.6 Hz, 1H), 7.31 - 7.29 (d, ,7=8.0 Hz, 1H), 7.20 (s, 1H).

[0255] Step C: 4-bromonaphthalen-2-ol: To a solution of 4-bromo- l-diazonio-naphthalen-2- olate (100 g, 402 mmol, 1.00 eq) in EtOH (2.00 L) was added portionwise NaBHi (30.4 g, 803 mmol, 2.00 eq) at 13-15 °C over 1 h, and the reaction mixture was stirred at 15-18 °C for 3 hrs. The reaction was filtered and concentrated to dryness. The residue was dissolved in DCM (1000 mL) and washed with water (500 mL c 2). The organic phase was dried over Na 2 S0 4 and concentrated to dryness. The residue was purified by silica gel column chromatograph, eluting with diethyl ether/ethyl acetate (60: 1 to 10:1). The isolated product was further purified by reverse phase HPLC to provide 4-bromonaphthalen-2-ol (40.0 g, 139 mmol, 17.3 % yield, 77.4% purity) as a gray solid. Ή NMR (400 MHz, CDCl 3 ) d 8.07 - 8.05 (d, .7=8.0 Hz, 1H), 7.60 - 7.58 (d, ,7=7.6 Hz, 1H), 7.41 - 7.36 (m, 3H), 7.07 (s, 1H).

[0256] Step D: 3-benzyloxy-l-bromo-naphthaIene: A mixture of 4-bromonaphthalen-2-ol (30.0 g, 134 mmol, 1.00 eq ), benzyl bromide (25.3 g, 148 mmol, 17.6 mL, 1.10 eq) and K2CO3 (55.7 g, 403 mmol, 3.00 eq) in MeCN (500 mL) was heated at 80 °C for 1 hr. The reaction mixture was filtered and concentrated to dryness. The residue was purified by silica gel column

chromatography, eluting with diethyl ether/ethyl acetate (100:1 to 60:1) to provide 3-benzyloxy- 1 -bromo-naphthalene (40.0 g, 128 mmol, 95 % yield) as yellow oil. *H NMR (400 MHz, CDCI3) d 8.19 - 8.17 (d, .7=8.0 Hz, 1H), 7.75 - 7.32 (d, .7=8.8 Hz, 1H), 7.64 - 7.63 (d, .7=2.4 H z ,IH), 7.52 - 7.37 (m, 7H), 7.23 - 7.21 (d, J=2.0 H z ,IH), 5.2 (s, 2H).

Intermediate 28

3-methoxynaphthalen-l -yl trifluoromethanesulfonate

[0257] Step A: 3-methoxynaphthalen-l -ol : To a solution of naphthalene- 1 ,3 -diol (3.00 g, 18.7 mmol, 1.00 eq) in MeOH (60.0 mL) was added HCl/MeOH (4 M, 60.0 mL, 12.8 eq) at 0 °C. The mixture was stirred at 25 °C for 60 hours. The solvent was removed under vacuum. The residue was purified by silica gel chromatography (diethyl etherethyl acetate=10:l to 5:1) to give 3- methoxynaphthalen-l-ol (2.10 g, 12.1 mmol, 64.4% yield) as a brown solid. 'H NMR (400 MHz, CDCl 3 -d 6 ) d = 8.10 - 8.08 (d, J=8.4 Hz, 1H).7.73 - 7.71 (d, J=8.4 Hz, 1H), 7.47 - 7.45(m, 1 H), 7.38 - 7.35(m, 1H), 6.80 - 6.79 (d, J=2.0 Hz, 1H), 6.56 - 6.55 (d, .7=2.4 Hz, 1H), 3.92 (s, 3H).

[0258] Step B: (3-methoxy- 1 -naphthyl) trifluoromethanesulfonate: To a solution of 3- methoxynaphthalen-l -ol (2.10 g, 12.0 mmol, 1.00 eq) in DCM (40.0 mL) was added DIEA (7.79 g, 60.3 mmol, 10.5 mL, 5.00 eq) and trifluoromethanesulfonic anhydride (5.10 g, 18.1 mmol, 2.98 mL, 1.50 eq) at 0 °C. The mixture was stirred at 25 °C for 1 hour. The mixture was diluted with DCM (30 mL) and water (10 mL) and extracted with DCM (20 mL). The combined organic layers were washed with brine (5 mL), dried over Na 2 S0 4 and concentrated under vacuum. The residue was purified by silica gel chromatography (diethyl ether :ethyl acetate=20: l to 10 : l) to give (3 -methoxy-1 -naphthyl) trifluoromethanesulfonate (3.00 g, 8.52 mmol, 70.7 % yield, 87.0 % purity) as a brown oil. ESI MS m/z 307.1 [M+H] f . lntermediate 29

tert-butyl (1 -bromoisoquinolin-3-yl)carbamate

[0259] Step A: A mixture of l-bromoisoquinolin-3-amine (400 mg, 1.79 mmol, 1.00 eq) and e/-Z-butoxycarbonyl tert-butyl carbonate (3.91 g, 17.9 mmol, 4.12 mL, 10.0 eq) was stirred at 70 °C for 16 hours. The residue was purified by column chromatography (Si0 2 , diethyl ether/ethyl acetate = 5: 1) to give /er -butyl N-(l -bromo-3-isoquinolyl) carbamate (400 mg, 1.24 mmol, 69.2 % yield) as a yellow solid. ESI MS m/z 322.1 , 324.1 [M+H] + .

Intermediate 30

3-methoxy-6-methylnaphthalen- 1 -yl trifluoromethanesulfonate

[0260] Step A: 3-methoxynaphthalen-l -ol: To a solution of naphthalene- 1 ,3-diol (40.0 g, 250 mmol, 1.00 eq) in MeOH (800 mL) was added HC1 (4 M, 750 mL, 12.0 eq, 4 M in MeOH) at 0 °C. The mixture was warmed up to 1 8 °C and stirred for 30 hours. The mixture was concentrated under vacuum. The residue was purified by column chromatography over silica gel (petroleum ether/ethyl acetate 100/1 to 1/1 ). The desired fractions were collected and concentrated under vacuum to give 3-methoxynaphthalen-l -ol (17.7 g, 96.5 mmol, 38.6 % yield, 95 % purity) as a red oil. ] H NMR (400MHz, Chloroform-d) d = 8.17 (d, J= 8.4 Hz, 1H), 7.74 (d, j = 8.0 Hz, 1 H), 7.50 (ddd, J = 1.2, 6.8, 8.0 Hz, 1H), 7.38 (ddd, .7=1.2, 6.8, 8.0 Hz, 1H), 6.81 (d, J=2.0 Hz, 1H), 6.76 (br s, 1H), 6.62 (d, J=2.4 Hz, 1H), 3.91 (s, 3H).

[0261] Step B: /e/ /-hutyl-[( ' 3- ethoxy-l-naphthyl)oxy 1-dimcthyl-silane: To a solution of 3- methoxynaphthalen- 1 -ol (20.0 g, 1 15 mmol, 1.00 eq) and imidazole (23.5 g, 344 mmol, 3.00 eq) in THF (400 mL) was added TBSC1 (26.0 g, 172 mmol, 21.1 mL, 1.50 eq) dropwise at 0 °C. The mixture was warmed up to 25 °C and stirred for 16 hours. The mixture was diluted with petroleum ether (600 mL) and ethyl acetate (200 mL), and then washed with water (l x 200 mL) and brine (l x 200 mL). The separated organic layer was dried over sodium sulfate, filtered and concentrated under vacuum. The residue was purified by column chromatography over silica gel (petroleum ether/ethyl acetate 100/1 to 10/1). /e/v-bii tyl-[(3-methoxy-l -naphthyl )oxy |-dimethyl- silane (28.0 g, 97.1 mmol, 84.6 % yield) was obtained as a colorless oil. ] H NMR (400MHz, Chloroform-d) 5 = 8.01 (d, .7= 8.4 Hz, 1H), 7.61 (d, j = 8.0 Hz, 1H), 7.35 (dt, j= 1.2, 7.6 Hz, 1H), 7.24 (dt, J= 1.2, 7.6 Hz, 1H), 6.71 (d, J= 2.0 Hz, 1H), 6.48 (d, j= 2.4 Hz, 1H), 3.82 (s, 3H), 1.02 (s, 9H), 0.23 (s, 6H).

[0262] Step C: /er/-butyl-[[3-methoxy-6-(4.4,5,5-tetramcthyl-L3.2-dioxaboro lan-2-yl)-l- naphthyl]oxy]-dimethyl-silane and /er/-butyl((3-methoxy-7-(4,4,5,5-tetramethyl-L3,2- dioxaborolan-2-yl)naphthalen-l-yl)oxy)dimethylsilane: A mixture of tert- b u ty 1 - [(3 - m e tli o x y - I - naphthyl) oxy]-dimethyl-silane (26.0 g, 90.1 mmol, 1.00 eq), 4,4,5,5-tetramethyl-2-(4,4,5,5- tetramethyl-l,3,2-dioxaborolan-2-yl)-l ,3,2-dioxaborolane (45.8 g, 180 mmol, 2.00 eq), (IZ,5Z)- cycloocta-l,5-diene;2,4-dimethyl-BLAHbicyclo[1.1.0]butane (2.39 g, 3.61 mmol, 0.04 eq) and 4-/cr/-butyl-2-(4-/£'rt-butyl-2-pyridyl)pyridine (1.45 g, 5.41 mmol, 0.06 eq) in hexane (500 mL) was stirred at 100 °C under nitrogen atmosphere for 16 hours. The mixture was diluted with water (500 mL) and ethyl acetate (1000 mL). The separated organic layer was washed with brine (l x 500 mL), dried over sodium sulfate, filtered and concentrated under vacuum. The residue was purified by column chromatography over silica gel (petroleum etehr/ethyl acetate 100/1 to 10/1). The desired fractions were collected and concentrated under vacuum to give a mixture of /er/-butyl-[[3-methoxy-6-(4,4,5,5-tetramethyl-l ,3,2-dioxaborolan-2-yl)-l-naphthyl]oxy]- dimethyl-silane and /<?r/-butyl((3-methoxy-7-(4,4,5,5-tetramethyl-l,3,2- dioxaborolan-2- yl)naphthalen-l-yl)oxy)dimethylsilane (38.0 g, 85.3 mmol, 94.6 % yield, 93 % purity) as a light yellow oil. ESI MS m/z 415.5 [M+H] + [0263] Step D: 8- butyl(dimethyl)silyl]oxy-6-methoxy- naphthalen-2-ol: To a solution of

mixture (36.0 g, 86.9 mmol, 1.00 eq) of /er/-butyl-[[3-methoxy-6-(4,4,5,5- tetramethyl-l,3,2- dioxaborolan-2-yl)-l- naphthyl]oxy]-dimethyl-silane and /er/-butyl((3-methoxy-7-(4,4,5,5- tetramethyl-1,3, 2-dioxaborolan-2-yl)naphthalen-l-yl)oxy)dimethylsilanein in acetone (400 mL) was added a solution of Oxone (58.7 g, 95.6 mmol, 1.10 eq) in H 2 0 (400 mL) at 0 °C. The mixture was stirred at 0 °C for 1 hour. The mixture was quenched with 5% aqueous sodium thiosulfate solution (50 mL) and extracted with ethyl acetate (2 x 300 mL). The extracts were combined and washed with water (l x 200 mL), brine (l x 200 mL), dried over magnesium sulfate, filtered and the filtrate was concentrated under vacuum. The residue was purified by column chromatography over silica gel (petroleum ether/ethyl acetate 200/1 to 20/1). The desired fractions were collected and concentrated under vacuum to give 8-[/er/-butyl(dimethyl)silyl]oxy- 6-methoxy- naphthalen-2-ol (9.00 g, 28.4 mmol, 32.7 % yield, 96 % purity) as a colorless oil and 5-[/er/-butyl(dimethyl)silyl]oxy-7-methoxy-naphthalen-2-ol (9.00 g, 29.0 mmol, 33.4 % yield,

98 % purity) as a white solid. ESI MS m/z 305.2 [M+H] +

[0264] Step E: [5-[fer/-butyl(dimethyl)silyl |oxy-7-methoxy-2-naphthyl]

trifluoromethanesulfonate : To a solution of 5-[/er/-butyl(dimethyl)silyl]oxy-7- methoxy- naphthalen-2-ol (11.0 g, 36.1 mmol, 1.00 eq) and DIEA (14.0 g, 108 mmol, 18.9 mL, 3.00 eq) in DCM (150 mL) was added Tf 2 0 (12.2 g, 43.4 mmol, 7.15 mL, 1.20 eq) dropwise at - 40 °C. The mixture was stirred for 1 hour. The mixture was diluted with dichloromethane (200 mL) and washed with water (1 c 200 mL) and brine (l x 200 mL). The separated organic layer was dried over sodium sulfate, filtered and concentrated under vacuum. The residue was purified by column chromatography over silica gel (petroleum ether/ethyl acetate 100/1 to 10/1). The desired fractions were collected and concentrated under vacuum to give [5 -[tert- butyl(dimethyl)silyl]oxy-7-methoxy-2-naphthyl] trifluoromethanesulfonate (13.0 g, 29.8 mmol, 82.4 % yield, 100 % purity) as a white solid. ESI MS m/z 436.9 [M+H]

[0265] Step F butyl-[(3-methoxy-6-methyl-l-naphthyl)oxy]-dimethyl-silane: To a solution of [5-[/er/-butyl(dimethyl)silyl]oxy-7-methoxy-2- naphthyl]trifluoromethanesulfonate (12.5 g, 28.6 mmol, 1.00 eq) and K 2 CO 3 (1 1.9 g, 85.9 mmol, 3.00 eq) in dioxane (160 mL) was added Pd(PPh 3 )4 (3.31 g, 2.86 mmol, 0.10 eq) and trimethylboroxine (14.4 g, 57.3 mmol, 16.0 mL, 2.00 eq) under nitrogen atmosphere. The reaction was heated to 100 °C for 16 hours. The mixture was diluted with ethyl acetate (200 mL) and then washed with water (1 x 200 mL) and brine (1 c 200 mL). The separated organic layer was dried over sodium sulfate, filtered and concentrated under vacuum. The residue was purified by column chromatography over silica gel (petroleum ether/ethyl acetate 100/1 to 5/1). The desired fractions were collected and concentrated under vacuum to give /cr/-butyl-[(3-methoxy-6-methyl-l -naphthyl)oxy]-dimethyl-silane (8.00 g, 24.6 mmol, 85.9 % yield, 93 % purity) as a colorless oil as red solid. ESI MS rn/z 303.2 [M+H] +

[0266] Step G: 3-methoxy-6-methyl-naphthalen-l-ol: To a solution of /er/-butyl-[(3-methoxy-6- methyl-l -naphthyl) oxy]-dimethyl-silane (8.00 g, 26.5 mmol, 1.00 eq) in THF (100 mL) was added TBAF (10.4 g, 39.7 mmol, 1.50 eq) at 0 °C. The mixture was stirred at 0 °C for 3 hours. The mixture was diluted with water (100 mL) and ethyl acetate (200 mL). The separated organic layer was washed with brine (1 c 100 mL), dried over sodium sulfate, filtered and concentrated under vacuum. The residue was purified by column chromatography over silica gel (petroleum ether/ethyl acetate 50/1 to 5/1). The desired fractions were collected and concentrated under vacuum to give 3-methoxy-6-methyl-naphthalen-l-ol (4.70 g, 25.0 mmol, 94.4 % yield) as a red solid. ESI MS m/z 188.4 [M+H] +

[0267] Step H: 3-methoxy-6-methyl-l -naphthyl trifluoromethanesulfonate: To a solution of 3- methoxy-6-methyl-naphthalen-l-ol (4.70 g, 25.0 mmol, 1.00 eq) and DIEA (9.68 g, 74.9 mmol, 13.1 mL, 3.00 eq) in DCM (3.00 mL) was added Tf 2 0 (8.45 g, 30.0 mmol, 4.94 mL, 1.20 eq) dropwise at - 40 °C. The mixture was stirred for 1 hour. The mixture was diluted with dichloromethane (200 mL) and washed with water (1 c 200 mL) and brine (1 x 200 mL). The separated organic layer was dried over sodium sulfate, filtered and concentrated under vacuum. The residue was purified by column chromatography over silica gel (petroleum ether/ethyl acetate 100/1 to 10/1). 3 -methoxy-6-methyl-l -naphthyl trifluoromethanesulfonate (7.70 g, 24.0 mmol, 96.2 % yield, 99.9 % purity) was obtained as a colorless oil. ESI MS m/z 320.7 [M+H] + .

[0268] The following intermediates were prepared according to the preparation for Intermediate 3, substituting the appropriate phenol for 2-bromo-3-fluorophenol.

Intermediate 40

2-bromo-3-fluoro- 1 -(methoxymethoxy)-4-methylbenzene

[0269] Step 1 : 3-fluoro-4-methylphenol (1.016 g, 8.055 mmol) was placed in Cs 2 (3.9 mL, 64.44 mmol) and was cooled to 0°C. Br 2 (0.4150 mL, 8.055 mmol) was added and the mixture was stirred at room temperature for 2 hrs. 10% Na 2 S 2 0 2 was added and the mixture was extracted with DCM. The organic layers were combined, dried and filtered to provide 2-bromo-3-fluoro- 4-methylphenol (1.389 g, 6.775 mmol, 84.10 % yield) which was used directly in the next step.

[0270] Step 2: 2-bromo-3-fluoro-l-(methoxymethoxy)-4-methylbenzene was prepared according to the procedure for Intermediate 8 using 2-bromo-3-fluoro-4-methyiphenol in place of 2-bromo-3-fluorophenol.

Intermediate 41

2-bromo-l-isopropoxy-4-(methoxymethoxy)benzene

[0271] Step 1 : 4-isopropoxyphenol (1.00 g, 6.57 mmol) and TEA (1.83 mL, 13.1 mmol) were placed in DCM (25 mL). Acetyl chloride (7.56 mL, 7.56 mmol) was added dropwise and the reaction was stirred at room temperature for 2hr. Water was added and the mixture was extracted with DCM. The organic layer was dried, filtered and concentrated to provide 4- isopropoxyphenyl acetate (1.24 g, 6.38 mmol, 97.2 % yield) which was directly in the next step.

[0272] Step2: 4-Isopropoxyphenyl acetate (1.24 g, 6.585 mmol) was placed in ACN (20 mL) and N-bromosuccinimide (1.173 g, 6.590 mmol) was added. The mixture was stirred for 18 hr.

Water was added and the mixture was extracted with ether. The organic layers were combined, dried, and concentrated to provide 3-bromo-4-isopropoxyphenyl acetate (1.584 g, 5.800 mmol, 88.00 % yield) which was directly in the next step.

[0273] Step 3: 3-Bromo-4-isopropoxyphenyl acetate (500 mg, 1.83 mmol) was placed in MeOH (7 mL). A solution of KOH (111 mg, 1.98 mmol) in water (2 mL) was added to mixture and was stirred for 1 hr at room temperature. The reaction mixture was adjusted to pH 3 by the addition of IN HC1. The mixture was extracted with DCM. The extracts were combined, dried, filtered and concentrated to provide crude 3-bromo-4-isopropoxyphenol which was used directly the next reaction.

[0274] Step 4: 2-Bromo-l-isopropoxy-4-(methoxymethoxy)benzene was prepared according to the procedure for Intermediate 8 using 3-bromo-4-isopropoxyphenol in place of 2-bromo-3- fluorophenol

Intermediate 42

l -bromo-3-chloro-2-isopropyl-5-(methoxymethoxy)benzene

[0275] Step 1 : l-bromo-3-chloro-2-isopropyl-5-methoxybenzene (952 mg, 3.61 mmol) was placed in DCM (3 mL) and was cooled to 0°C. BBr3 (9030 pL, 9.03 mmol) was added and the reaction was stirred at 0°C for 2 lir. Water was added and the mixture was extracted with DCM. The extracts were combined and concentrated. The resulting residue was purified by silica gel (0-20% EtOAc in hexane) to provide 3-bromo-5-chloro-4-isopropylphenol (575 mg, 2.30 mmol, 63.8 % yield)

[0276] Step 2: l-bromo-3-chloro-2-isopropyl-5-(methoxymethoxy)benzene was prepared according to the procedure for Intermediate 8 using 3-bromo-5-chloro-4-isopropylphenol in place of 2-

Intermediate 43

l-iodo-3-(methoxymethoxy)naphthalene

[0277] To a solution of 4-iodonaphthalen-2-ol (0.80 g, 3.0 mmol) in DCM (20 mL) was added N-ethyl-N-isopropylpropan-2-amine (1.1 mL, 5.9 mmol) and chloro(methoxy)methane (0.29 g, 3.6 mmol) and the reaction stirred at room temperature for 4 hours, with additional

chloro(methoxy)methane (0.15 g) being added after 2 hours. The reaction was washed with brine and concentrated in vacuo. The material was purified by chromatography using a gradient of 0 to 10% EtOAc/hexanes as the eluent to give l-iodo-3-(methoxymethoxy)naphthalene (0.80 g, 2.5 mmol, 86 % yield).

Intermediate 44

3-benzyloxy-l-bromo-naphthalene

[0278] Step A: 2,4-dibromonaphthalen-l -amine: To a solution of Br 2 (246 g, 1.54 mol, 79.3 mL) in AcOH (750 mL) was added a solution of naphthalen-1 -amine (101 g, 705 mmol, 99.0 mL) in AcOH (500 mL) at room temperature and the reaction stirred at 70 °C for 1 hour. The reaction mixture was cooled to room temperature and filtered. The filter cake was washed with AcOH (300 mL). The solid was next suspended in 20 % aqueous ofNaOH (1.2 L). The mixture was stirred for 20 minutes and filtered. The solid was washed with water (1 L) and dried under vacuum to give 2,4-dibromonaphthalen-l -amine (200 g, 664 mmol, 94.2% yield) as gray solid. ES+APCI MS m/z 301.9 [M+H] + .

[0279] Step B: 4-bromo- 1 -diazonio-naphthalen-2-olate: To a solution of 2,4-dibromonaphthalen- 1 -amine (60.0 g, 199 mmol) in AcOH (900 mL) and propionic acid (150 mL) was added NaN0 2 (16.5 g, 239 mmol, 13.0 mL) portionwise at 5-8 °C over 30 minutes and the reaction mixture stirred at 5-8 °C for 30 minutes. The reaction mixture was poured into ice-water (4000 mL), the slurry filtered and the solid washed with water (2 x 50 mL) to give 4-bromo- l-diazonio- naphthalen-2-olate (150 g, wet crude) which was used crude in the next step immediately.’H NMR (400 MHz, CDC1 3 ) 5 8.12 - 8.10 (d, .7=8.4 Hz, 1H), 7.62 - 7.58 (t, .7=7.6 Hz, 1H), 7.41 - 7.37 (t, J=7.6 Hz, 1H), 7.31 - 7.29 (d, .7=8.0 Hz, 1H), 7.20 (s, 1H).

[0280] Step C: 4-bromonaphthalen-2-ol: To a solution of 4-bromo-l -diazonio-naphthalen-2- olate (100 g, 402 mmol) in EtOH (2.00 L) was added portion-wise NaBH 4 (30.4 g, 803 mmol) at 13-15 °C over 1 hour and the reaction stirred at 15-18 °C for 3 hours. The reaction was filtered and concentrated to dryness. The residue was dissolved in DCM (1000 mL) and washed with water (500 mL x 2). The organics were dried over Na 2 S0 4 and concentrated to dryness. The residue was purified by chromtography eluting with petroleum ether/EtOAc (60/1 10/1) and material re-purified by reverse phase HPLC to give 4-bromonaphthalen-2-ol (40.0 g, 139 mmol, 17.3 % yield, 77.4% purity) as a gray solid. *H NMR (400 MHz, CDC1 3 ) d 8.07 - 8.05 (d, ,7=8.0 Hz, 1H), 7.60 - 7.58 (d, J=7.6 Hz, 1H), 7.41 - 7.36 (m, 3H), 7.07 (s, 1H).

[0281] Step D: 3-benzyloxy-l-bromo-naphthalene: A mixture of 4-bromonaphthalen-2-ol (30.0 g, 134 mmol), BnBr (25.3 g, 148 mmol, 17.6 mL) and K 2 CO 3 (55.7 g, 403 mmol) in MeCN (500 mL) was heated at 80 °C for 1 hr. The reaction mixture was filtered and concentrated to dryness. The residue was purified by silica gel column eluting with PE/E A (100/1 to 60/1) to give 3- benzyloxy-l -bromo-naphthalene (40.0 g, 128 mmol, 95 % yield). NMR (400 MHz, CDCI 3 ) d 8.19 - 8.17 (d, ,7=8.0 Hz, 1H), 7.75 - 7.32 (d, ,7=8.8 Hz, 1H), 7.64 - 7.63 (d, ,7=2.4 H z , I H), 7.52 - 7.37 (m, 7H), 7.23 - 7.21 (d, ,7=2.0 Hz,IH), 5.2 (s, 2H).

Intermediate 45

4-bromo-5-methyl-l-tetrahydropyran-2-yl-indazole

[0282] Step A: 4-bromo-5-methyl-l-tetrahvdropyran-2-yl-indazole: To a mixture of 4-bromo-5- methyl-1 H-indazole (3 g, 14.2 mmol) and 3,4-dihydro-2H-pyran (2.39 g, 28.4 mmol, 2.60 mL) in DCM (30 mL) was added TsOH*H 2 0 (270 mg, 1.42 mmol) and the mixture stirred at 15 °C for 2 hours. After completion, the reaction mixture was concentrated under vacuum and the residue purified by column chromatography using 5- 20& EtO Ac/Petroleum Ether as eluent to give 4-bromo-5-methyl-l-tetrahydropyran-2-yl-indazole (4 g, 13.6 mmol, 95.3% yield) as white solid. Ή NMR (400 MHz, chloroform-d) d 8.01 (s, 1H), 7.47 (d, .7=8.4 Hz, HI), 7.25 (d, 7=8.4 Hz, 1H), 5.70 (dd, 7=2.8, 9.2 Hz, 1H), 4.05 - 3.96 (m, 1H), 3.79 - 3.70 (m, 1 H), 2.66 - 2.44 (m, 4H), 2.25 - 2.04 (m, 2H), 1.84 - 1.56 (m, 3H).

Intermediate 46

4-bromo-5-methoxy- 1 -(tetrahydro-2H-pyran-2-yl)- 1 H-indazole

[0283] 4-bromo-5-methoxy-l-(tetrahydro-2H-pyran-2-yl)-l H-indazole was prepared following Intermediate 51 substituting 4-bromo-5-methoxy-l H-indazole for 4-broiuo-5-methyl- 1 II- indazole in Step A. NMR (400 MHz, chloroform-d) d 8.00 (s, 1H), 7.53 (d, .7=9.2 Hz, 1H), 7.16 (d, .7=9.2 Hz, 1H), 5.70 (dd, 7=2.8, 9.2 Hz, 1H), 4.04 - 3.98 (m, 1H), 3.96 (s, 3H), 2.55 - 2.49 (m, HI), 2.23 - 2.05 (m, 2H), 1.83 - 1.69 (m, 3H).

Intermediate 47

3-(benzyloxy)- 1 -bromo-2-methylnaphthalene

[0284] Step A: ethyl 2-methyl-3-oxo-4-phenyl-butanoate. To a dried 250 ml three-necked flask was added ethyl 3-oxo-4-phenyl-butanoate (4.00 g, 19.4 mmol.), THF (50.0 mL), sodium hydride (931 mg, 23.3 mmol) and the reaction stirred for 0.5 hours at 0°C. A solution of methyl iodide (3.03 g, 21.3) was next added drop-wise. After addition was completed, the reaction mixture was warmed to 20 °C and stirred for two hours at 20°C. The reaction mixture was quenched by addition of water (10.0 mL) at 20 °C and then diluted with ethyl acetate (20.0 mL) and the layers separated. The aqueous layer was next extracted with ethyl acetate (20.0 mL c 3). The combined organic layers were washed with brine (30.0 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (Si02, Petroleum ether : Ethyl acetate 20: 1 to 10: 1) to give ethyl 2-methyl-3-oxo-4-phenyl-butanoate (3.60 g, 16.3 mmol, 84.3% yield) as a colorless oil. 'H NMR (400 MHz, CDCL) d = 7.38 - 7.28 (m, 3II), 7.25 - 7.19 (m, 2H), 4.22 - 4.15 (m, 2H), 3.87 (d, J = 2.0 Hz, 2H), 3.65 (q, ,7= 7.2 Hz, 1H), 1.34 (d, ,7= 7.2 Hz, 3H), 1.30 - 1.26 (m, 3H).

[0285] Step B: 2-methylnaphthalene-L3-diol. A solution of ethyl 2-methyl-3-oxo-4-phenyl- butanoate (3.60 g, 16.3 mmol) in concentrated sulfuric acid (19.9 g, 203 mmol) was stirred at 15 °C for 12 hours. The reaction mixture was poured into ice-water (30.0 mL) and the resulting solid collected by filtration and dried under vacuum to afford 2-methylnaphthalene-l ,3-diol (1.80 g, 10.3 mmol, 63.2% yield) as a red solid 'll NMR (400 MHz, CDC ) d = 8.02 (d, J= 8.0 Hz, 1H), 7.65 - 7.54 (m, 1 H), 7.41 (t, .7= 7.2 Hz, 1H), 7.36 - 7.31 (m, HI), 6.80 (s, 1H), 4.29 - 4.20 (s, 2H), 2.41 - 2.24 (s, 3H).

[0286] Step C: 3-methoxy-2-methyl-naphthalen-l-ol. 2-methylnaphthalene-l,3-diol (1.70 g, 9.76 mmol) was added to HCl/MeOH (2 M, 35.0 mL) and the result mixture was stirred at 30 °C for 3 days. The reaction was concentrated in vacuo and the residue purified by Prep-TLC (Petroleum ether : Ethyl acetate 1 :1) to give 3-methoxy-2-methyl-naphthalen-l-ol (800 mg, 4.25 mmol, 43.5% yield) as a white solid. Ή NMR (400 MHz, CDC1 3 ) d = 8.02 (d, J= 8.4 Hz, HI), 7.69 (d, j= 8.4 Hz, III), 7.44 - 7.38 (m, 1H), 7.37 - 7.31 (m, 1H), 6.79 (s, 1H), 5.14 (s, 1H), 3.94 (s, 3H), 2.29 (s, 3H).

[0287] Step D: (3-methoxy-2-methyl-l-naphthyl)trifluoromethanesulfonate. To a mixture of 3- methoxy-2-methyl-naphthalen-l-ol (800 mg, 4.25 mmol.) and pyridine (504 mg, 6.38 mmol) in DCM (10.0 mL) was added trifluoro acetic anhydride (1.44 g, 5.10 mmol) dropwise at 0°C under N 2 atmosphere. The mixture was warmed to 20°C and stirred for an additional 5 hours. The solvent was removed under vacuum and the residue purified by Prep-TLC (Petroleum ether : Ethyl acetate 1 : 1) to give (3-methoxy-2-methyl-l -naphthyl)trifluoiOmethanesu]fonate (1.30 g, 4.06 mmol, 95.5% yield) as a white solid. 'H NMR (400 MHz, CDCI3) d = 7.97 (d, J = 7.6 Hz, 1H), 7.79 - 7.74 (m, 1H), 7.52 - 7.43 (m, 2H), 7.14 (s, 1H), 3.99 (s, 3H), 2.42 (s, 3H)

[0288] Step E: l-bromo-3-methoxy-2-methyl-naphthalene : In a sealed tube was added (3- methoxy-2-methyl-l-naphthyl)trifluoromethanesulfonate (466 mg, 1.45 mmol), t-Bu-Brettphos (154 mg, 290 pmol), potassium bromide (259 mg, 2.17 mmol), PEG-200 (175 mg), 2-butanone (157 mg, 2.17 mmol) and Pd 2 (dba) 3 (133 mg, 145 pmol) in toluene (10.0 mL) and the mixture de-gassed with N2 for 5 minutes. Next, triisobutylaluminum (431 mg, 2.17 mmol) was added drop-wise at 20 °C. The mixture was heated to 100 °C for 24 hrs. The reaction mixture was poured into water (30.0 mL) and the aqueous layer extracted with ethyl acetate (20.0 mL c 3). The combined organics were washed with brine (30.0 mL), dried over anhydrous sodium sulfate and concentrated in vacuo to give a residue which was pre-purified by column chromatography (Petroleum ether:Ethyl acetate 10: 1) and then by Prep-TLC (Petroleum ether : Ethyl acetate 10: 1) to give l-bromo-3-methoxy-2-methyl-naphthalene (700 mg, 2.79 mmol, 64.1 % yield) as a white solid. l H NMR (400 MHz, CDCb) d = 8.26 - 8.17 (m, 1H), 7.73 - 7.69 (m, 1H), 7.47 - 7.40 (m, 2 II), 7.09 (s, 1H), 3.98 - 3.95 (m, 311), 2.56 (s, 3H).

[0289] Step F: 4-bromo-3-methyl-naphthalen-2-ol: To a solution of l-bromo-3-methoxy-2- methyl-naphthalene (580 mg, 2.31 mmol) and tetrabutylammonium iodide (2.13 g, 5.78 mmol) in DCM (11.0 mL) cooled to -78 °C was added a solution of BCl 3 (1 M, 5.78 mL) dropwise over a period of 10 minutes while under N 2. The reaction mixture was warmed to 0 °C and stirred for 2 hours at room temperature. Next the solvent was removed under vacuum and the residue was purified by Prep-TLC (Petroleum ether : Ethyl acetate 5:1) to give 4-bromo-3- methyl-naphthalen-2-ol (500 mg, 2.1 1 mmol, 91.3% yield) as a white solid. 'H NMR (400 MHz, CDCb) d = 8.26 - 8.15 (m, 1H), 7.63 (dd, .7= 3.6, 6.0 Hz, 1H), 7.45 - 7.38 (m, 2H), 7.11 (s, 1H), 5.09 (s, 1H), 2.60 (s, 3H), 1.56 (s, 3H).

[0290] Step G: 3-benzyloxy-l-bromo-2-methyl-naphthalene. To a mixture of 4-bromo-3- methyl-naphthalen-2-ol (265 mg, 1.12 mmol) and benzyl bromide (201 mg, 1.18 mmol) in acetonitrile (3.00 mL) was added potassium carbonate (310 mg, 2.24 mmol) in one portion at 20 °C under N 2 . The mixture was next stirred at 60°C for two hours. The solvent was removed under vacuum and the residue purified by Prep-TLC /Petroleum ether : Ethyl acetate 5:1) to give the 3-benzyloxy-l-bromo-2-methyl-naphthalene (250 mg, 695 pmol, 31.0% yield, 91.0% purity) as a white solid. ES+APCI MS m/z 327.0, 329.0 [M+H] + .

Intermediate 48

/er/-butyl-2-(cyanomethyl)-4-[2-[[(25)-l-methylpyrrolidin -2-yl]methoxy]-5, 6,7,8- tetrahydropyrido[3 ,4-d]pyrimidin-4-yl]piperazine- 1 -carboxylate

[0291] Step A: (4-bromo-2-naphthyl) 2,2-dimethylpropanoate. To a solution of 4- bromonaphthalen-2-ol (10 g, 44.8 mmol) and TEA (9.07 g, 89.7 mmol) in DCM (200 mL) was added 2,2-dimethylpropanoyl chloride (8.1 1 g, 67.2 mmol) at 0°C. The reaction mixture was stirred at 0 °C for 10 min. T reaction mixture was quenched by addition of water (50 mL) and the layers separated. The organic layer was washed with brine (30 mL), dried over Na 2 S0 4 filtered and concentrated under vacuum. The residue was purified by silica gel chromatography (PE: EA =1 :0 to 100:1) to give (4-bromo-2-naphthyl) 2,2-dimethylpropanoate (9 g, 29.3 mmol, 65.4% yield) as a red oil. l U NMR (400MHz, CHLOROFORM-d) d = 8.22 (d, .7=8.0 Hz, 1H), 7.83 - 7.77 (m, 1H), 7.63 - 7.49 (m, 4H), 1.41 (s, 9H).

Intermediate 49

[0292] Naphthalen-l -yl trifluoromethanesulfonate. alpha-Naphthol (4 g, 27.74 mmol) was dissolved in DCM (200 mL) in a 3 neck flask. The reaction was cooled to l0°C in a water bath. N-ethyl-N-isopropylpropan-2-amine (4.846 ml, 27.74 mmol) and trifluoromethanesulfonic anhydride (4.668 ml, 27.74 mmol) were added to the solution dropwise. The reaction was stirred at 10°C for 2 hours. TLC (25% EtOAc, UV vis) showed reaction complete. The organics were with water (2X) and brine (2X). The organics were dried over MgS04 and concentrated in vacuo. The concentrate was purified using normal phase chromatography on the CombiFlash (0%-l2% EtOAc:Hexanes). All fractions containing clean product were combined and conentrated in vacuo to give naphthalen-l -yl trifluoromethanesulfonate (6.77 g, 24.51 mmol, 88.34 % yield).

Intermediate 50

Tert-butyl (S)-2-(hydroxymethyl)-4-methylpiperazine-l-carboxylate

[0293] To a solution of (S)-l-Boc-2-hydroxymethylpiperazine (1.0 g, 4.62 mmol) in DCE (92.47 ml, 4.624 mmol) was added formaldehyde (3.474 ml, 46.24 mmol) (37% in water) followed by sodium triacetoxyborohydride (4.9 g, 23.12 mmol). The mixture was stirred vigorously at room temperature for 2.5hours. The mixture was treated with saturated sodium bicarbonate (30 mL), stirred for 10 min then extracted with DCM (3 x 10 mL). The combined organic phases were dried over sodium sulfate, filtered and concentrated. ES+APC1 MS m/z 231.1 [M+H] + .

Intermediate 51

Tert-butyl (R)-2-(hydroxymethyl)-4-methylpiperazine- 1 -carboxylate

[0294] Title compound was prepared as in Intermediate 57, substituting tert-butyl (R)-2- (hydroxymethyl)piperazine-l -carboxylate for (S)-l-Boc-2-hydroxymethylpiperazine. ES+APCI MS m/z 231.1 [M+Hf

Intermediate 52

l-bromo-3-chloro-2-fluoro-5-(methoxymethoxy)benzene

[0295] To a round bottom flask was added THF (8.87 ml, 4.44 mmol) followed by sodium hydride, 60 % dispersion in mineral oil (0.213 g, 5.32 mmol). The mixture was cooled to 0 °C then 3-bromo-5-chloro-4-fluorophenol (1.0 g, 4.44 mmol) was added portionwise. Once the bubbling had ceased the resulting dark mixture was stirred at 0 °C for 30 min. Then

chloromethyl methyl ether (0.421 ml, 5.54 mmol) was added and the mixture was warmed to ambient temperature where it was stirred for 2 hr. A saturated aqueous ammonium chloride solution was added and the mixture was extracted with DCM. The organic layer was dried over sodium sulfate, filtered and concentrated. Crude material was chromatographed (0-15% EtOAc in hexanes) to provide product as clear oil.

Intermediate 53

4-bromo-l-tetrahydropyran-2-yl-5-(trifluoromethyl)indazol e

[0296] Step A: 4-bromo-l-tetrahvdropyran-2-yl-5-(trifluoromethyl)indazole: To a solution of 4- bromo-5-(trifluoromethyl)-lH-indazole (500 mg, 1.89 mmol, 1 eq ) in DCM (10 mL) was added 3,4-dihydro-2H-pyran (476 mg, 5.66 mmol, 517 uL, 3 eq) and TsOH HhO (35.9 mg, 188 pmol, 0.1 eq). The mixture was stirred at 15 °C for 1 hour. The mixture was concentrated. The residue was purified by column chromatography (Si0 2 , PE:EA=10: 1 to 1 :1) to give 4-bromo-l- tetrahydropyran-2-yl-5-(trifluoromethyl)indazole (480 mg, 1.37 mmol, 72.9% yield) as yellow oil.1H NMR (400 MHz, chloroform-d) d 8.20 (s, 1H), 7.69 - 7.63 (m, 2H), 5.70 (dd, .7=2.8, 8.8 Hz, 1H), 4.05 - 3.96 (m, 1H), 3.79 - 3.70 (m, 1H), 2.56 - 2.50 (m, 1H), 2.27 - 2.04 (m, 2H), 1.80 - 1.74 (m, 2H), 1.60 - 1.54 (m, 1 H).

Intermediate 54

[0297] 8 -bromo-6-(methoxymethoxy)q uinoline : A stirred suspension of 8-bromoquinolin-6-ol (1.00 g, 4.46 mmol) in DCM (20 mL) was cooled to 0°C and diisopropylethylamine (1 .2 mL, 6.7 mmol, 1.5 eq.) was added followed by chloro(methoxy)methane (0.41 mL, 5.4 mmol, 1.2 eq.) dropwise and the reaction mixture was warmed to room temperature overnight. Concentrated aqueous ammonia (0.5 mL, ~5 mmol) was next added and the resulted mixture was stirred for lhour at room temperature. The mixture was evaporated in vacuo and chromatographed on silica gel, Redisep 40g, using 20% EtO Ac/hexane as eluent to give a colorless powder (0.52 g, 44%). ES+APCi MS m/z 268.0, [M+H] + .

Intermediate 55

[0298] To a solution of but-3-enenitrile (80.0 g, 1.19 mol, 96.4 mL, 1.00 eq) in /er/-butanol (130 mL) and petroleum ether (480 mL) was added a solution ofBr 2 (19l g, 1.19 mol, 61.5 mL, 1.00 eq) in tert- butanol (130 mL). The mixture was stirred at 10 °C for 4 hours. The mixture was used into next step without any workup.

[0299] To the above mixture (274 mL) was added a solution of AC/V-dibenzylethane-l ,2-diamine (160 g, 445 mmol, 157 mL, 2 HOAc) and Et 3 N (178 g, 1.76 mol, 245 mL) in toluene (300 mL). After was stirred at 110 °C for 2 hours, the mixture was filtered and the filtrate was concentrated under vacuum. The residue was purified by column chromatography (Si0 2 , petroleum ether/ethyl acetate = 3/1) to give 2-(l, 4-dibenzylpiperazin-2-yl)acetonitrile (75.0 g, 246 mmol, two steps 55.7 % yield) as a yellow solid. LCMS [ESI, M+l]: 306. [0300] ‘H NMR (400MHz, chloroform-d) d = 7.37 - 7.23 (m, 10H), 3.80 (d, J= 13.2 Hz, 1H), 3.60 - 3.42 (m, 3H), 3.06 - 2.96 (m, 1H), 2.95 - 2.83 (m, 1H), 2.69 - 2.53 (m, 4H), 2.52 - 2.35 (m, 3H).

[0301] To a solution of 2-(l,4-dibenzylpiperazin-2-yl)acetonitrile (160 g, 524 mmol, 1.00 eq) in dichloroethane (1.50 L) was added 1 -chloroethyl carbonochloridate (300 g, 2.10 mol, 4.00 eq) at 15 °C. After stirred at 85 °C for 48 h, the mixture was concentrated under vacuum. The residue was then taken up into methanol (1.50 L) and heated to reflux for 1 hour. The mixture was concentrated. The solid was treated with methyl tert-butyl ether (1.00 L), 2-piperazin-2- ylacetonitrile (Intermediate 62, 90.0 g, 454 mmol, 86.7 % yield, 2HC1) was obtained as a white solid and used for next step without further purification.

[0302] Ή NMR (400MHz, DMSO-d6) d = 10.19 (br s, 2H), 4.01 - 3.73 (m, 1H), 3.69 - 3.41 (m, 4H), 3.32 (dt, ./= 2.8, 13.2 Hz, 1H), 3.27 - 3.10 (m, 3H).

Intermediate 56

Cbz Cbz

t wo steps yield: 72% Boc y e :

[0303] To a solution of /ert-butyl (3f?)-3-(hydroxymethyl)piperazine-l -carboxylate (80.0 g, 370 mmol, 1.0 eq) in Ethyl acetate (1400 mL) was added NaHCOa (93.2 g, 1.1 1 mol, 43.2 niL, 3.0 eq), H 2 0 (700 mL) and benzyl carbonochloridate (82.0 g, 481 mmol, 68.4 mL, 1.30 eq). The mixture was stirred at 25 °C for 12 hour. After completion, the organic phase was separated, washed with water (500 mL x 2) dried over Na 2 S0 4 and filtered. The solvent was removed under vacuum to give a residue. The residue was purified by column chromatography (Si0 2 , Petroleum ether/Ethyl acetate=40/l to 1/1). The product 1 -benzyl 4-/ert-butyI ( 2R)-2 - (hydroxymethyl)piperazine-l,4-dicarboxylate (85.0 g, 235 mmol, 64% yield, 96% purity) was obtained as a yellow oil. LCMS [ESI, M-99]: 251.

[0304] To a solution of 1 -benzyl 4-tert-butyl (27?)-2-(hydroxymethyl)piperazine-l,4- dicarboxylate (20.0 g, 57.1 mmol, 1.0 eq ) in 2-Methyltetrahydrofuran (240 mL) was added TEA (17.3 g, 171.23 mmol, 23.8 mL, 3.0 eq) and methanesulfonyl chloride (7.74 g, 67.6 mmol, 5.23 mL, 1.18 eq). The mixture was stirred at 20 °C for 1 hour. The reaction mixture was quenched by addition TI2O 150 mL at 20 °C. The reaction mixture was extracted with Ethyl acetate (300 mL x 2). The organic layers were washed with H 2 0 (100 mL), dried over Na 2 S0 4 , and filtered. The solvent was removed under vacuum. 1 -benzyl 4-tert-butyl (2R)-2-

(methylsulfonyloxymethyl)piperazine-l ,4-dicarboxylate (22.0 g, crude) was obtained as a yellow oil. The crude product was used directly to the next step without further purification.

[0305] To a solution of 1 -benzyl 4-tert-butyl (27i;)-2-(mcthylsulfonyloxy methyl (piperazine-! ,4- dicar boxy late (22.0 g, 51.3 mmol) in DMA (150 mL) was added NaCN (10.4 g, 21 1 mmol). The mixture was stirred at 60 °C for 12 hour. The solvent was removed under vacuum to give a oil residue. The residue was diluted with H 2 0 (40.0 mL) and extracted with Ethyl acetate (50.0 mL x 3). The combined organic layers were washed with saturated brine (80.0 mL), dried over Na 2 S0 4 , filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (S1O2, Petroleum ether/Ethyl acetate=40/l to 5:1) The product 1 -benzyl 4-tert-butyl (2ri)-2-(cyanomethyl)piperazine-l,4-dicarboxylate (18.5 g, 46.4 mmol, two steps yield 72%) was obtained as a yellow oil. LCMS [ESI, M+l]: 360.

[0306] To a solution of 1-benzyl 4-tert-butyl (25)-2-(cyanomethyl)piperazine-l,4-dicarboxylate (18.5 g, 43.3 mmol, 1.00 eq) in dioxane (40.0 mL) was added HCbdioxane (4 M, 54.1 mL, 5.0 eq). The mixture was stirred at 20 °C for 1 hour. Then the reaction mixture was added NaHC0 3 to pH>7, and concentrated under reduced pressure to remove dioxane. The residue was diluted with II 2 0 (50.0 mL) and extracted with Ethyl acetate (50.0 mL c 3). The combined organic layers were washed with H 2 0 (20.0 mL) , dried over Na 2 S0 4 , filtered and concentrated under reduced pressure to give a residue. The product benzyl (25)-2-(cyanomethyl)piperazine-l- carboxylate (Intermediate 63, 1 1.5 g, 91.8% purity, 95% yield) was obtained as a yellow oil. LCMS [ESI, M+l ]: 260. [0307] Ή NMR (400MHz, CHLOROFORM-d) d = 7.37 - 7.31 (m, 5H), 5.14 (s, 2H), 4.49 (br, s, 1H), 3.93 (br, s, 1H), 3.07 - 2.81 (m, 5II), 2.78 - 2.54 (m, 2H).

Intermediate 57 rTV Br LiMe, THF, Mel

k^-k ^ Br - ! - : - ^

T T 0 - 25 °C, 3.5 h

43%

1 -bromo-8-methylnaphthalene

[0308] Step A: 1 -bromo-8-methyl-naphthalene. To a solution of 1,8-dibromonaphthalene (1 g, 3.50 mmol, 1 eq ) in THF (20 ml.) was added MeLi (1.6 M in diethyl ether, 2.62 mL, 1.2 eg) at 0°C dropwise. After stirring for 30 minutes at 0°C, iodomethane (3.38 g, 23.8 mmol, 1.48 mL, 6.81 eq) was added dropwise. The mixture was warmed up to 25°C and stirred for another 3 hours. The reaction mixture was quenched with water (20 mL) and extracted with ethyl acetate (20 mL x 3). The combined organic layers were washed with brine (20 mL), dried over Na 2 S04, filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (column: Phenomenex Gemini Cl 8 250*50mm* 10 um; mobile phase: [water (0.05% ammonium hydroxide v/v) - ACN]; B%: 45% - 70%, 28 MIN; 40% min). Ttitle compound l-bromo-8-methyl -naphthalene (340 mg, 1.49 mmol, 43% yield, 97% purity) was obtained as a yellow solid after lyophilisation.

[0309] Ή NMR (400MHz, chloroform-d) d = 7.75 (dd, J = 0.8, 7.2 Hz, 1H), 7.69 (dd, J = 0.8, 8.0 Hz, 1H), 7.66 - 7.59 (m, 1 H), 7.30 - 7.22 (m, 2H), 7.13 (t, j= 8.0 Hz, HI), 3.05 (s, 3H).

Intermediate 58

79%

1 -bromo-8-chloronaphthalene [0310] Step A: lZ/-naphtho[ 1 ,8-deli 1 ,2.3]triazine. To a solution of naphthalene- 1,8-diamine (100 g, 632 mmol, 1 eq ) in AcOH (200 mL) and EtOH (1000 mL) was added isoamyl nitrite (72.6 g, 619 mmol, 83.4 mL, 0.98 eq) dropwise over a period of 2 h with temperature controlled between 18 and 21 °C under a cold-water bath. After the addition, the resulting red suspension was stirred at 25 °C for 16 hours. The solid was collected by filtration, washed with ethanol (2 x 500 mL) and dried under vacuum. Compound lZ7-naphtho[l,8-de][l,2,3]triazine (84 g, 496 mmol, 79% yield) was obtained as a red crystalline solid and directly used next step without purification. LCMS [ESl, M+l]: 170.

[031 1] Step B: 8-chloronaphthalen-l -amine. To a solution of 1 Z/-naphtho[l,8-de][l,2,3]triazine (84 g, 496 mmol, 1 eq) in HC1 (1.5 L) was added Cu (2.10 g, 33.1 mmol, 234 uL, 0.0665 eq).

The mixture was stirred at 25 °C for 12 hours. The resulting mixture was diluted with water (500 mL) and heated at 85 °C for 30 mins. The resulting almost clear aqueous solution was filtered, cooled, treated with aqueous ammonia (until blue to litmus paper) and the solution was extracted with ether acetate (2 x 1000 mL). The combined extracts were dried over Na 2 S0 4, filtered and concentrated under vacuum. The residue was purified by column chromatography (Si0 2 , Petroleum ether/Ethyl acetate = 200/1 to 5/1). Compound 8-chloronaphthalen-l -amine (57 g, 259 mmol, 52% yield, 81% purity) was obtained as a red solid. LCMS [ESI, M+l]: 178.

[0312] Step C: 1 -bromo-8-chloro-naphthalene. To a solution of 8-chloronaphthalen-l -amine (57 g, 320 mmol, 1 eq) and TsOH*H 2 0 (219 g, 1.16 mol, 3.6 eq) in MeCN (1000 mL) was added a solution ofNaN0 2 (39.8 g, 577 mmol, 1.8 eq) and CuBr (138 g, 963 mmol, 29.3 mL, 3 eq) in H 2 0 (120 ml,) at - 5 °C, then the reaction mixture was stirred at 25 °C for 12 hours. The reaction mixture was added saturated Na 2 S0 3 solution (100 mL) and stirred for 15 mins, then extracted with ethyl acetate (1000 mL><3). The combined organic layers were washed with brine (500 mL), dried over Na 2 S0 4 , filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (Si0 2 , Petroleum ether). Title compound 1- bromo-8-chloro-naphthalene (56 g, 229 mmol, 72% yield, 99% purity) was obtained as white solid.

[0313] Ή NMR (400MHz, chloroform-d) 5 = 7.93 (dd, J= 1.2, 7.6 Hz, 1H), 7.82 (dd, J= 1.2, 8.4, 1H), 7.79 (dd, ./= 1.2, 8.4, HI), 7.67 (dd, ./ = 1.2, 7.6 Hz, 1H), 7.37 (t, J= 8.0 Hz, 1 H), 7.28 (t, .7= 8.0 Hz, III). EXAMPLE 1

2-[(2S)-4-[8-(8-methyl-l-naphthyl)-2-[[(2.S)-l-Methylpyrr olidin-2-yl]niethoxy]-5,6,7,9- tetrahydropyrimido[4,5-c]azepin-4-yl]-l -prop-2-enoyl-piperazin-2-yl]acetonitrile

Cbz Cbz

i

[0314] Compound 1-2: To a solution of tert- butyl 3-oxopiperidine-l-carboxylate (20.0 g, 100 mmol, 1.0 eq) in isopropyl ether (160 mL) was added dropwise BF 3* Et 2 0 (17.1 g, 120 mmol, 15.0 mL, 1.2 eq) at 0 °C and then ethyl 2-diazoacetate (15.0 g, 130 mmol, 1.3 eq) dropwise over 0.5 hour at 0 °C. The resulting mixture was stirred at 0 °C for 1 hour. After completion, the reaction mixture was quenched by saturated aqueous NaHCCfi (200 mL) and the solution was stirred for 1 hour, then extracted with ethyl acetate (2 x 150 mL). The combined organic layers were washed with saturated brine (1 c 200 mL), dried and concentrated to give a residue. The residue was purified by column chromatography (Sifb, petroleum ether: ethyl acetate = 30:1 to 20:1) to give Ol-tert-butyl 04-ethyl 3-oxoazepane-l ,4-dicarboxylate (3.2 g, 11.2 mmol, 1 1% yield) as yellow oil.

[0315] Compound 1-3: Na (774 g, 33.6 mmol, 3.0 eq) was dissolved in methyl alcohol (45.0 mL) in portions at 0 °C for 0.5 hour, then Ol-tert-butyl 04-ethyl 3-oxoazepane-l,4- dicarboxylate (3.2 g, 11.2 mmol, 1.0 eq) and 2-mcthylisothiourea (2.02 g, 14.5 mmol, 1.3 eq , 0.5 H2SO4) was added to the reaction mixture, the reaction mixture was stirred at 15 °C for 12 hours. After completion, the reaction mixture was adjusted with IN HC1 to pH ~ 6, then the precipitated solid w r as filtered and washed with methyl alcohol (20.0 mL), the filtrate w r as dried and concentrated to give /e/7 -butyl 4-hydroxy-2-methylsulfanyl-5,6,7,9-tetrahydropyrimido[4,5- cjazepine -8-carboxylate (3.4 g, crude) as yellow solid which was used for the next step without further purification.

Compound 1-4: To a solution of tert- butyl 4-hydroxy-2-methylsulfanyl-5, 6,7,9- tetrahydropyrimido[4,5-c]azepine-8-carboxylate (3.4 g, 10.9 mmol, 1.0 eq) and TEA (3.87 g, 38.2 mmol, 5.3 niL, 3.5 eq) in dichloromethane (35.0 mL) was added Tf 2 0 (6.16 g, 21.8 mmol, 3.6 mL, 2.0 eq) at -40 °C and stirred for 0.5 hour. After completion, the reaction mixture was quenched by addition of water (30.0 mL) at -40 °C and extracted with ethyl acetate (3 x 30 mL). The combined organic layers were dried over Na 2 S0 4 , filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (S1O2, petroleum ether: ethyl acetate= 15:1 to 10:1) to give /er/-butyl 2-methylsulfanyl-4- (trifluoromethylsulfonyloxy)-5,6,7,9-tetrahydropyrimido[4,5- c]azepine-8-carboxylate (3.7 g,

8.13 mmol, 74% yield, two steps, 98% purity) as yellow oil. LCMS [ESI, M-55]: 388.

[0316] Compound 1-5: A mixture of tert- butyl 2-methylsulfanyl-4-

(trifluoromethylsulfonyloxy)-5,6,7,9-tetrahydropyrimido[4 ,5-c]azepine-8-carboxylate (3.7 g,

8.34 mmol, 1.0 eq), benzyl (2S)-2-(cyanomethyl)piperazine-l-carboxylate (2.27 g, 8.76 mmol, 1.05 eq) and DIEA (3.24 g, 25.0 mmol, 4.36 mL, 3.0 eq) in DMAC (35.0 mL) was degassed and purged with N 2 for 3 times, and then the mixture was stirred at 15 °C for 3 hours under N 2 atmosphere. After completion, the reaction mixture was quenched by addition II 2 0 (100.0 mL), and then extracted with ethyl acetate (3 x 30 mL). The combined organic layers were washed with saturated brine (4 x 30 mL), dried over Na 2 SQi, filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (Si0 2 , petroleum ether: ethyl acetate = 5:1 to 1 :1) to give /er/-butyl 4-[(35 -4-benzyloxycarbonyl-3-(cyanomethyl) piperazin-1 -yl]-2-methylsulfanyl-5,6, 7,9-tetrahydropyrimido[4,5-c]azepine-8-carboxylale (3.0 g, 5.27 mmol, 63% yield, 97% purity) as white solid. LCMS [ES1, M+l]: 553.

[0317] 'll NMR (400 MHz, chloroform-d) d 7.43 - 7.34 (m, 5H), 5.24 - 5.17 (m, 211), 4.69 - 4.35 (m, 3H), 4.13 - 4.1 1 (m, 1H), 3.81 - 3.74 (m, 2H), 3.57 - 3.50 (m, 2H), 3.28 - 3.20 (m, 2H), 2.94 - 2.85 (m, 2H), 2.77 - 2.65 (m, 3H), 2.52 (s, 3H), 1.99 - 1.95 (m, 1H), 1.36 (s, 9H).

Compound 1-6: To a solution of tert- butyl 4-[(J5 , )-4-benzyloxycarbonyl-3- (cyanomethyl)piperazin-l -yl]-2-methylsulfanyl-5,6,7,9-tetrahydropyrimido[4,5-c]azepi ne-8- carboxylate (3.0 g, 5.43 mmol, 1.0 eq) in ethyl acetate (10.0 mL) was added m-CPBA (1.10 g, 5.43 mmol, 1.0 eq). The mixture was stirred at 0 °C for 0.5 hour. After completion, the reaction mixture was quenched with saturated aq. Na 2 S0 3 (15.0 mL) and diluted with H 2 0 (20.0 mL).

The crude mixture was extracted with ethyl acetate (3 c 30 mL). The combined extracts were washed with saturated aq. NaHCCL (30.0 mL), dried with Na 2 S0 4 the solvent was then removed under vacuum. The residue was purified by column chromatography (Si0 2 , Ethyl acetate:

Methanol 30: 1 to 10: 1). Compound /ert-butyl d-f /T -benzyloxycarbonyl-S- (cyanomethyl)piperazin-l-yl]-2-methy]sulfinyl-5,6,7,9-tetrah ydropyrimido[4,5-c]azepine-8- carboxylate (2.9 g, 5.05 mmol, 93% yield, 99% purity) was obtained as white solid. LCMS [ESI, M+l]: 569.

[0318] Compound 1-7: To a solution of [(2S)-l-methylpyrrolidin-2-yl]methanol (705 mg, 6.12 mmol, 727 uL, 1.2 eq ) and /-BuONa (833 mg, 8.67 mmol, 1.7 eq) in toluene (20.0 mL) was added a solution of tert- butyl 4-[(35)-4-benzyloxycarbonyl-3- (cyanomethyl)piperazin-l-yl]-2- methylsulfinyl-5,6,7,9-tetrahydropyrimido[4,5-c]azepine-8-ca rboxylate (2.9 g, 5.10 mmol, 1.0 eq) in toluene (10.0 mL) dropwise at 0 °C. The reaction mixture was stirred at 0 °C for 0.5 hour. After completion, the reaction was quenched with water (40.0 mL). The crude mixture was extracted with ethyl acetate (2 x 50 mL). The combined extracts were washed with saturated brine (100 mL), dried with Na 2 S0 4 , the solvent was removed under vacuum. The residue was purified by column chromatography (A1 2 0 3 , Petroleum ether: Ethyl acetate = 3:1 to 0: 1).

Compound tert- butyl 4-[(35 r )-4-benzyloxycarbonyl-3-(cyanomethyl)piperazin-l-yl]-2 - [[(2,S')- 1 - methylpyrrolidin-2-yl]methoxy]-5,6,7,9-tetrahydropyrimido[4, 5-c-]azepine-8-carboxylate (2.4 g, 3.84 mmol, 75% yield, 99% purity) was obtained as white solid. LCMS [ESI, M+l ]: 620.

[0319] Compound 1-8: To a solution of /er/-butyl 4-[(3S -4-benzyloxycarbonyl-3- (cy anomethyl)piperazin-l-yl]-2-[[(25 -l-methylpynolidin-2-yl]methoxy]-5, 6,7,9- tetrahydropyrimido[4,5-c]azepine-8-carboxylate (400 mg, 645.42 pmol, 1.0 eq) in dioxane (5.0 mL) was added HChdioxane (4.0 M, 5.0 mL, 31.0 eq). The mixture was stirred at 20 °C for 0.5 hour under N 2 atmosphere. After completion, the reaction mixture was quenched by addition saturated aq. Na 2 C0 3 at 0 °C until pH ~ 8, and then extracted with ethyl acetate (3 c 20 mL). The combined organic layers were dried over Na 2 S0 4 , filtered and concentrated under reduced pressure to give a residue. Benzyl(25)-2-(cyanomethyl)-4-[2-[[(2S , )-l- methylpyrrolidin-2- yl]methoxy]-6,7,8,9-tetrahydro-57/-pyrimido[4,5-c]azepin-4-y l]piperazine-l-carboxylate (330 mg, crude) was obtained as white solid which was used to the next step without further purification. LCMS [ESI, M+l]: 520.

[0320] Compound 1-9: A mixture of benzyl (25)-2-(cyanomethyl)-4-[2-[[(25 r )-l- methylpyriOlidin-2-yl]methoxy]-6,7,8,9-tetrahydro-577-pyrimi do[4,5-c]azepin-4-yl]piperazine-l- carboxylate (330 mg, 540 pmol, 1.0 eq ), l-bromo- 8 -methyl -naphthalene (239 mg, 1.08 mmol,

2.0 eq), Xantphos (93.7 mg, 162 pmol, 0.3 eq), Pd 2 (dba) 3 (74.2 mg, 81.0 pmol, 0.15 eq) and CS 2 CO 3 (528 mg, 1.62 mmol, 3.0 eq) in toluene (3.0 mL) was degassed and purged with N 2 for 3 times, and then the mixture was stirred at 90 °C for 12 hours under N 2 atmosphere. After completion, the reaction was washed with HC1 (1 N, 2 x 5.0 mL). The aqueous phase was treated with solid NaHC0 3 to pH ~ 7 and extracted with ethyl acetate (3 x 5.0 mL). The organic layer was dried over Na 2 S0 4 , filtered and concentrated. The residue was purified by column chromatography (basic A1 2 0 3 , Petroleum ether: Ethyl acetate = 3: 1 to Ethyl acetate : Methanol = 50:1) and then residue was purified by reverse phase flash [C18, 0.1% FA in water , 0-65% MeCN] and was treated with NallC'Ch solid to pH ~ 7, and extracted with ethyl acetate (2 c 50 mL). The organic layers were dried over Na 2 S0 4 , filtered and concentrated under vacuum.

Benzyl(2S)-2-(cyanomethyl)-4-[8-(8-methyl-l-naphthyl)-2-[[(2 5)-l-methylpyrrolidin-2- yl]methoxy]-5, 6, 7, 9-tetrahydropyrimido[4,5-c]azepin-4-yl]piperazine-l -carboxylate (60.0 mg, 91.0 pmol, 17% yield, 100% purity) was obtained as white solid. LCMS [ESI, M+l]: 660.

[0321] ¾ NMR (400 MHz, chloroform-d) d 7.55 - 7.46 (m, 2LI), 7.31 - 7.27 (m, 2H), 7.27 - 7.10 (m, 6H), 7.06 (t , J= 5.8 Hz, 1H), 5.09 (s, 2H), 4.58 (br s, 1H), 4.28 - 4.20 (m, 2H), 4.19 - 4.1 1 (m, 1H), 3.99 - 3.92 (m, 1H), 3.71 - 3.59 (m, 1H), 3.49 (t, J= 12.6 Hz, 1H), 3.36 - 3.03 (m, 4H), 2.98 - 2.76 (m, 4H), 2.73 - 2.47 (m, 7H), 2.31 (d, J= 3.2 Hz, 3H), 2.18 - 2.10 (m, 1H), 1.92 - 1.79 (m, 3 FI), 1.73 - 1.54 (m, 3H).

[0322] Compound 1-10: To a solution of benzyl (25)-2-(cyanomethyl)-4-[8-(8-methyl-l- naphthyl)-2-[[(25)-l -methylpyrrolidin-2-yl]methoxy]-5,6,7,9-tetrahydropyrimido[4 ,5-c]azepin- 4-yl]piperazine-l -carboxylate (60 mg, 90.9 pmol, 1.0 eq) in MeOH (1.0 mL) and NH3*MeOH (1.0 mL, 15% purity) was added Pd/C (20 mg, 10% purity) under N 2 atmosphere. The suspension was degassed and purged with H 2 for 3 times and the mixture was stirred under H 2 (15 Psi) at 20 °C for 0.5 hour. After completion, the crude mixture was filtered through a pad of celite. The cake was washed with MeOH (2 c 5.0 mL) and the filtrate was concentrated.

Compound 2-[(25>4-[8-(8-methyl -l-naphthyl)-2-[[(26)-l-methylpyrrolidin-2-yl]methoxy]- 5,6,7,9-tetrahydropyrimido[4,5-c]azepin-4-yl]piperazin-2-yl] acetonitrile (45.0 mg, 75.3 pmol, 83% yield, 88% purity) was obtained as yellow solid and used into the next step without further purification. LCMS [ESI, M+1 J: 526. [0323] Example 1: To a solution of2-[(25)-4-[8-(8-methyl-l-naphthyl)-2-[[(25)-l- methylpyrrolidin-2-yl]methoxy]-5,6,7,9-tetrahydropyrimido[4, 5-c]azepin-4-yl]piperazin-2- yljacetonitrile (45.0 mg, 75.3 pmol, 1.0 eq ) in dichloromethane (1.0 mL) was added TEA (30.5 mg, 301 pmol, 41.9 pL, 4.0 eq) and prop-2-enoyl prop-2-enoate (9.50 mg, 75.3 pmol, 1.0 eq) at 0 °C. The mixture was stirred at 0 °C for 0.5 hour under N 2 atmosphere. After completion, the organic solvent was washed with water (5.0 mL). The aqueous phase was extracted with dichloromethane (3 x 5.0 mL). The combined extracts were washed with brine (10.0 mL), dried with Na 2 S0 4 the solvent was then removed under vacuum. The residue was purified by column chromatography (basic AI 2 O 3 , Petroleum ether : Ethyl acetate = 3:1 to Ethyl acetate : Methanol - 50:1) and then purified by prep-HPLC (column: Xtimate Cl 8 lOp 250 mm x 50 mm; mobile phase: [water (0.05% ammonium hydroxide v/v)-ACN]; B%: 63% - 93%, 10 min) and lyophilization to give 2-[(2S)-4-[8-(8-methyl-l -naphthyl) -2-[[(25)-l -Methylpyrrolidin-2- yI]methoxy]-5,6,7,9-tetrahydropyrimido[4,5-c]azepin-4-yl]-l- prop-2-enoyl-piperazin-2- yljacetonitrile (10.5 mg, 17.8 pmol, 24% yield, 99% purity) as yellow solid. LCMS [ESI, M+l]: 580.

[0324] *H NMR (400 MHz, chloroform-d) d 7.71 - 7.60 (m, 2H), 7.41 - 7.28 (m, 3H), 7.23 - 7.17 (m, 1H), 6.67 - 6.53 (m, 1H), 6.45 - 6.36 (m, 1H), 5.83 (br d, J= 10.4 Hz, 1H), 5.25 - 4.97 (m, 1H), 4.43 - 4.23 (m, 3H), 4.18 - 4.08 (m, 1H), 4.05 - 3.54 (m, 3H), 3.51 - 3.36 (m, 1H), 3.35 - 3.16 (m, 2H), 3.14 - 2.89 (m, 4H), 2.87 - 2.59 (m, 7H), 2.44 (br d, J= 2.4 Hz, 3H), 2.34 - 2.21 (m, 1H), 2.1 1 - 1.91 (m, 311), 1.89 - 1.67 (m, 3H).

EXAMPLE 2

2-[(2 L S)-l -(2-iluoroprop-2-enoyl)-4-[8-(8-methyl-l-naphthyl)-2-[[(25)- l-methylpyrrolidin-2- yl]methoxy]-5,6,7,9-tetrahydropyrimido[4,5-c]azcpin-4-yl]pip erazin-2-yl]acetonitrile

[0325] Example 2: To a solution of 2-[(2ri)-4-[8-(8-methyl-l-naphthyl)-2-[[(2X)-l - methylpynOlidin-2-yl]methoxy]-5,6,7,9-tetrahydropyrimido[4,5 -c]azepin-4-yl]piperazin-2- yljacetonitrile (120 mg, 201 pmol, 1.0 eq), 2-fluoroprop-2-enoic acid (41.1 mg, 457 pmol, 2.27 eq ) in dichloromethane (2.0 mL) was added D1EA (1 18 mg, 913 pmol, 159 uL, 4.55 eq) and HATU (174 mg, 457 pmol, 2.27 eq) at 0 °C. The mixture was stirred at 15 °C for 1 hour under N 2 atmosphere. After completion, the water was added (10.0 mL). The aqueous phase was extracted with dichloromethane (2 >< 10 mL). The combined extracts were washed with brine (15.0 mL), dried with Na 2 S0 4 , the filtrate was removed under vacuum. The residue was purified by column chromatography (Base A1 2 0 3 , Petroleum ether: Ethyl acetate = 3:1 to Ethyl acetate: Methanol = 50:1) and then purified by prep-HPLC (column: Waters Xbridge 150 c 255p; mobile phase: [water (0.05% ammonium hydroxide v/v)-ACN]; B%: 60%-84%,l0 min) and lyophilized to give 2-[(2S -l-(2-fluoroprop-2-enoyl)- 4-[8-(8-methyl-l -naphthyl)-2-[[(25)-l- methylpyiTolidin-2-yl]methoxy]-5,6,7,9-tetrahydropyrimido[4, 5-c]azepin-4-yl]piperazin-2- yl]acelonitrile (26.6 mg, 44.2 pmol, 22% yield, 99% purity) as white solid. LCMS [ESI, M+l]: 598.

[0326] 1H NMR (400 MHz, chloroform-d) d 7.69 - 7.60 (m, 2H), 7.41 - 7.28 (m, 3H), 7.22 - 7.18 (m, 1H), 5.51 - 5.33 (m, 1H), 5.26 (dd, J = 3.6, 16.8 Hz, 1H), 5.05 - 4.65 (m, 1H), 4.42 - 4.25 (m, 3H), 4.17 - 4.09 (m, 1H), 3.87 - 3.78 (m, 1H), 3.69 (br t, J= 13.8 Hz, 1H), 3.55 - 3.15 (m, 4H), 3.13 - 2.92 (m, 4H), 2.89 - 2.54 (m, 7H), 2.44 ( d, J= 2.4 Hz, 3H), 2.32 - 2.20 (m, 1H), 2.13 - 1.93 (m, 3H), 1.83 - 1 .64 (m, 3H).

EXAMPLE 3

2-[(2S)-4-[8-(2-methyl-l-naphthyl)-2-[[(2S)-l-methylpyrro lidin-2-yl]methoxy]-5,6,7,9- tetrahydropyrimido[4,5-c]azepin-4-yl]-l -prop-2-enoyl-piperazin-2-yl]acetonitrile

Example 3

[0327] Compound 3-1: To a mixture of benzyl (25)-2-(cyanomethyl)-4-[2-[[(25)-1 - methylpyrrolidin-2-yl]methoxy]-6,7,8,9-tetrahydro-5f7-pyrimi do[4,5-c]azcpin-4-yl]piperazine-l- carboxylate (300 mg, 577 pmol, 1.0 eq) 1 -bromo-2-methyl-naphthalene (191 mg, 866 pmol, 1.5 eq), CS 2 CO 3 (564 mg, 1.73 mmol, 3.0 eq) and RuPhos (108 mg, 231 pmol, 0.4 eq) in toluene (9 niL) was added Pd 2 (dba) 3 (106 mg, 115 pmol, 0.2 eq) under N 2 . The suspension was degassed under vacuum and purged with N 2 several times. The mixture was stirred under N 2 at 90 °C for 8 hours. Water (20 mL) was added into the mixture. The mixture was diluted with EtOAc (10 mL) and extracted with EtOAc (3 >< 15 mL). The combined organic layers were washed with brine (20 mL). The combined organic layers were dried over anhydrous Na 2 S0 4 , filtered and concentrated under vacuum. The residue was purified by reverse-phase flash [water (0.1 % FA)/acetonitrile] to give benzyl (2S)-2-(cyanomethyl)-4-[8-(2-methyl-l-naphthyl)-2-[[(2S , )-l- methylpyrrolidin-2-yl]methoxy]-5,6,7,9-tetrahydropyrimido[4, 5-c]azepin-4-yl]piperazine-l- carboxylate (250 mg, 360 pmol, 62 % yield, 95 % purity) as a yellow solid. LCMS [ESI, M+l]: 660.

[0328] *H NMR (400 MHz, chloroform-d) d = 8.14 - 7.96 (m, 1H), 7.82 - 7.75 (m, 1H), 7.60 (d, J = 8.4 Hz, 1H), 7.45 - 7.34 (m, 7H), 7.30 (s, 1H), 5.27 - 5.16 (m, 2H), 4.73 (br s, HI), 4.63 - 4.52 (m, 1H), 4.49 - 4.38 (m, 1H), 4.35 (br d, J= 12.8 Hz, 1H), 4.1 1 - 4.04 (m, 1 H), 3.84 (br s, HI), 3.78 - 3.64 (m, 1H), 3.62 - 3.48 (m, 1H), 3.43 - 3.23 (m, 3H), 3.13 - 2.88 (m, 5H), 2.84 - 2.74 (m, III), 2.63 (br s, 1H), 2.49 - 2.36 (m, 6H), 2.26 (br s, III), 2.15 - 2.08 (m, 1H), 2.04 - 1.93 (m, 1H), 1.86 - 1.67 (m, 3H).

[0329] Compound 3-2: To a solution of benzyl (25)-2-(cyanomethyl)-4-[8-(2-methyl-l- naphthy l)-2- [[(2, S')- 1 -methyl pyrrol idin-2-yl]methoxy]-5,6,7,9-tetrahydropyrimi do [4,5-c]azepin- 4-yl]piperazine-l -carboxylate (250 mg, 379 pmol, 1 eq) and NH3 » MeOH (2 mL, 20 % purity) in MeOH (4 mL) was added Pd/C (50 mg, 10 % purity) under N 2 . The suspension was degassed under vacuum and purged with H 2 several times. The mixture was stirred under H 2 (15 psi) at 15 °C for 0.5 hour. The reaction mixture was filtered and the filtrate was concentrated under vacuum to give 2-[(2X)-4-[8-(2-methyl-l -naphthyl)-2-[[(2X)- 1 -methylpyrrolidin-2-yl]methoxyj- 5,6,7,9-tetrahydiOpyrimido[4,5-c]azepin-4-yl]piperazin-2-yl] acetonitrile (190 mg, 325 mihoΐ, 86 % yield, 90 % purity) as a yellow solid which was used for next step without further purification.

[0330] Example 3: To a solution of 2-[(25’)-4-[8-(2-methyl-l-naphthyl)-2-[[(25)-l - methylpyrrolidin-2-yl]methoxy]-5,6,7,9-tetrahydropyrimido[4, 5-c]azepin-4-yl]piperazin-2- yljacetonitrile (190 mg, 361 pmol, 1 eq) and DIEA (93.4 mg, 723 mihoΐ, 126 mE, 2 eq) in DCM (4 mL) was added prop-2-enoyl chloride (49.1 mg, 542 mihoΐ, 44.2 uL, 1.5 eq) at -40 °C. The reaction mixture was stirred at -40 °C for 0.5 hour. The reaction mixture was quenched by water (5 mL). The mixture was extracted with DCM (3 x 10 mL). The combined organic layers were dried over anhydrous Na 2 S0 4 , filtered and concentrated under vacuum. The residue was purified by prep-HPLC (column: Xtimate Cl 8 150*25mm*5pm;mobile phase: [water (0.05 %

ammonium hydroxide v/v)-ACN];B %: 55 % - 85 %, 8 min) to give 2-[(2S)-4-[8-(2-methyl-l - naphthyl)-2-[[(2S)-l-methylpyrrolidin-2-yl]methoxy]-5,6,7,9- tetrahydropyrimido[4,5-c]azepin- 4-yl]-l -prop-2-enoyl-piperazin-2-yl]acetonitrile (65.0 mg, 1 1 1 mhioΐ, 31 % yield, 98.8 % purity) as a white solid. LCMS [ESI, M+l ]: 580.

[0331] 'H NMR (400 MHz, chloroform-d) d = 8.13 - 7.97 (m, 1 H), 7.83 - 7.76 (m, 1H), 7.60 (d, J= 8.4 Hz, 1H), 7.45 - 7.35 (m, 2H), 7.29 (d, J= 8.4 Hz, 1 H), 6.69 - 6.55 (m, 1 H), 6.40 (br d, J = 16.8 Hz, 1 H), 5.84 (br d, ./ = 10.4 Hz, 1H), 5.15 (br s, 1H), 4.63 - 4.52 (m, 1 H), 4.50 - 4.36 (m,

1 H), 4.33 (br dd, ./ = 4.4, 10.0 Hz, 1H), 4.16 - 3.87 (m, 3H), 3.86 - 3.71 (m, 1H), 3.55 (ddd, = 5.6, 1 1.6, 17.2 Hz, 2H), 3.46 - 3.36 (m, 111), 3.34 - 3.25 (m, HI), 3.14 - 2.90 (m, 5H), 2.83 (br s, 1 H), 2.61 (br d, J= 3.2 Hz, HI), 2.50 - 2.38 (m, 6H), 2.29 - 2.19 (m, 1 H), 2.17 - 2.05 (m, 211), 2.04 - 1 .94 (m, 1H), 1.86 - 1.71 (m, 3H).

EXAMPLE 4 2-[(2.S)-4-[2-[[(2S)-l-methylpyrrolidin-2-yl]methoxy]-8-(o-t olyl)-5,6,7,9- tetrahydropyrimido[4,5-c]azepin-4-yl]-l-prop-2-enoyl-piperaz in-2-yl]acetonitrile

Example 4

[0332] Compound 4-1: To a mixture of benzyl (2S)-2-(cyanomethyl)-4-[2-[[(25 -l - methylpyrrolidin-2-yr]methoxy]-6,7,8,9-tetrahydro-5//-pyrimi do[4,5-c]azepin-4-yl]piperazine- 1 - carboxylate (300 mg, 577 mhioΐ, 1.0 eq), l-bromo-2-methyl-benzene (148 mg, 866 mpioΐ, 104 uL, 1.5 eq), CS2CO3 (564 mg, 1.73 mmol, 3.0 eq) and RuPhos (108 mg, 231 mihoΐ, 0.4 eq) in toluene (9 mL) was added Pd 2 (dba) 3 (106 mg, 115 mhioΐ, 0.2 eq) under N 2 . The suspension was degassed under vacuum and purged with N 2 several times. The mixture was stirred under N 2 at 90 °C for 7 hours. Water (20 mL) was added into the mixture. The mixture was diluted with EtOAc (10 mL) and extracted with EtOAc (2 x 15 mL). The combined organic layers were washed with brine (20 mL). The combined organic layers were dried over anhydrous Na 2 S0 4 , filtered and concentrated under vacuum. The residue was purified by reverse-phase flash [water (0.1 % FA)/acetonitrile] to give benzyl (25 , )-2-(cyanomethyl)-4-[2-[[(25 , )-l-methylpyrrolidin-2- yl]methoxy]-8-(o-tolyl)-5,6,7,9-tetrahydropyrimido[4,5- ]azepin-4-yl]piperazine-l -carboxylate (230 mg, 358 mihoΐ, 62 % yield, 95 % purity) as a white solid. LCMS [ESI, M+l]: 610. [0333] Ή NMR (400 MHz, chloroform-d) d = 7.45 - 7.32 (m, 5H), 7.14 (br t , J= 6.0 Hz, 2H), 7.07 - 7.02 (m, 1H), 6.98 - 6.92 (m, 1H), 5.20 (s, 2H), 4.68 (br s, 1H), 4.39 (dd, J= 4.8, 10.8 Hz, 1H), 4.20 - 4.08 (m, 4H), 3.82 (br d, J= 13.2 Hz, 1H), 3.65 (br d, J= 12.4 Hz, 1H), 3.39 - 3.18 (m, 411), 3.1 1 (br t, J= 7.6 Hz, 1H), 2.99 - 2.64 (m, 6H), 2.48 (s, 3H), 2.35 - 2.24 (m, 1 H), 2.14 (s, 3H), 2.11 - 2.06 (m, 1 H), 2.05 - 1.93 (m, 2H), 1.87 - 1.71 (m, 3H).

[0334] Compound 4-2: To a solution of benzyl (25)-2-(cyanomethyl)-4-[2-[[(25 -l- methylpyrrolidin-2-yl]methoxy]-8-(o-tolyl)-5,6,7,9-tetrahydr opyrimido[4,5-c]azepin-4- yl]piperazine-l-carboxylate (230 mg, 377 pmol, 1.0 eq) and NH 3 ·MeOH (1 mL, 20 % purity) in MeOH (4 mL) was added Pd/C (50 mg, 10 % purity) under N 2 . The suspension was degassed under vacuum and purged with ¾ several times. The mixture was stirred under ¾ (15 psi) at 15 °C for 0.5 hour. The reaction mixture was filtered and the filtrate was concentrated under vacuum. 2-[(25 r )-4-[2-[[(2,S -l-methylpyrrolidin-2-yl]methoxy]-8-(o-tolyl)-5, 6,7,9- tetrahydropyrimido[4,5-c]azepin-4-yl]piperazin-2-yl]acetonit rile (180 mg, 341 pmol, 90 % yield, 90 % purity) was obtained as a yellow solid which was used for the next step without further purification.

[0335] Example 4: To a solution of 2-[(2S -4-[2-[[(25 -l -methylpyrrolidin-2-yl]methoxy]-8-(o- tolyl)-5,6,7,9-tetrahydropyrimido[4,5-c]azepin-4-yl]piperazi n-2-yl]acetonitrile (180 mg, 378 mihoΐ, 1 eq) and DIEA (97.8 mg, 757 mpioΐ, 132 uL, 2 eq) in DCM (4 mL) was added prop-2- enoyl chloride (51.4 mg, 567 mihoΐ, 46.3 uL, 1.5 eq) at -40 °C. The reaction mixture was stirred at -40 °C for 0.5 hour. The reaction mixture was quenched by water (5 mL). The mixture was extracted with DCM (3 x 10 mL). The combined organic layers were dried over anhydrous Na 2 S0 4 , filtered and concentrated under vacuum. The residue was purified by prep-HPLC (column: Xtimatc C18 150*25mm*5pm;mobile phase: [water (0.05 % ammonium hydroxide v/v)-ACN];B %: 51 % - 81 %, 8 min) to give 2-[(25)-4-[2-[[(25)-l-methylpyrrolidin-2- yl]methoxy]-8-(o-tolyl)-5,6,7,9-tetrahydropyrimido[4,5-c]aze pin-4-yl]-l -prop-2-enoyl-piperazin- 2-yl]acetonitrile (56.6 mg, 104 pmol, 28 % yield, 97.5 % purity) as a white solid. LCMS [ES1, M+l ]: 530.

[0336] Ή NMR (400 MHz, chloroform-d) d = 7.18 - 7.1 1 (m, 211), 7.04 (d, J= 7.6 Hz, 1H), 6.99 - 6.91 (m, 1H), 6.67 - 6.52 (m, 1H), 6.38 (dd, ./= 1.6, 16.8 Hz, 1H), 5.82 (br d, J= 10.8 Hz, III), 5.09 (br s, 1H), 4.38 (dd, J= 4.8, 10.8 Hz, 1H), 4.24 - 4.09 (m, 3H), 3.87 (br d, J= 13.6 Hz, 2H), 3.73 (br d, J= 12.4 Hz, 1H), 3.57 (br s, 1EI), 3.39 - 3.31 (m, 1H), 3.31 - 3.16 (m, 2H), 3.09 (br t, ./= 7.6 Hz, 1H), 2.94 (br dd, J= 8.4, 16.4 Hz, 2H), 2.82 (br d , J= 4.4 Hz, 3H), 2.67 (td, J= 6.8, 13.2 Hz, 1H), 2.47 (s, 3H), 2.33 - 2.22 (m, 1H), 2.15 (s, 3H), 2.10 - 1.92 (m, 3H), 1.89 - 1.75 (m, 3H).

EXAMPLE 5

2-[(2S)-4-[2-[[(2S -l-methylpyrrolidin-2-yl]methoxy]-8-phenyl-5,6,7,9-tetrahydr opyrimido[4,5- c]azepin-4-yl]-l-prop-2-enoyl-piperazin-2-yl]acetonitrile

Example 5

[0337] Compound 5-1 : To a mixture of benzyl (25)-2-(cyanomethyl)-4-[2-[[(25)-l- methylpyrrolidin-2-yl]methoxy]-6,7,8,9-tetrahydro-5//-pyrimi do[4,5-c]azepin-4-yl]piperazine-l- carboxylate (300 mg, 577 pmol, 1.0 eq ), iodobenzene (177 mg, 866 pmol, 96.5 pL, 1.5 eq), CS2CO3 (564 mg, 1.73 mmol, 3.0 eq) and RuPhos (108 mg, 231 pmol, 0.4 eq) in toluene (9 mL) was added Pd 2 (dba)3 (106 mg, 1 15 pmol, 0.2 eq) under N 2 . The suspension was degassed under vacuum and purged with N 2 several times. The mixture was stirred under N 2 at 90 °C for 7 hours. Water (20 ml.) was added into the mixture. The mixture was diluted with EtOAc (10 mL) and extracted with EtOAc (2 x 15 mL). The combined organic layers were washed with brine (20 mL). The combined organic layers were dried over anhydrous Na 2 SC>4, filtered and concentrated under vacuum. The residue was purified by reverse-phase flash [water (0.1 % FA)/ acetonitrile] to give benzyl (2.S')-2-(cyanomethyl )-4-[2-| | (2.V)- 1 -methylpyrrolidin-2-yl |methoxy |- 8-phenyl-5,6,7,9-tetrahydropyrimido[4,5-c]azepin-4-yl]pipera zinc-l-carboxylate (220 mg, 351 prnol, 61 % yield, 95 % purity) as a yellow solid. LCMS [ESI, M+l]: 596.

[0338] ‘H NMR (400 MHz, chloroform-d) d = 7.41 - 7.31 (m, 5H), 7.20 - 7.14 (m, 2H), 6.82 (d, J= 8.0 Hz, 2H), 6.68 (t, J= 7.2 Hz, 1H), 5.22 - 5.12 (m, 2H), 4.62 (hr s, 1H), 4.57 (s, 2H), 4.41 (br s, 1H), 4.22 - 4.15 (m, 1H), 4.07 (hr s, 1H), 3.83 - 3.59 (m, 3H), 3.46 (br d, J= 1 1.2 Hz, 1H), 3.24 (br s, 1H), 3.12 (br dd, J= 3.6, 13.6 Hz, 2H), 2.91 - 2.62 (m, 7H), 2.51 (s, 3H), 2.3 l (br d, J = 8.0 Hz, 1H), 2.09 (br d, J= 8.4 Hz, 1H), 1.97 (br s, 2H), 1.86 - 1.70 (m, 3H).

[0339] Compound 5-2: To a solution of benzyl (25)-2-(cyanomethyl)-4-[2-[[(2<S)-l - methylpynOlidin-2-y]]methoxy]-8-phenyl-5,6,7,9-tetrahydropyr imido[4,5-c]azepin-4- yl]piperazine-l -carboxylate (220 mg, 369 mihoΐ, 1.0 eq) and NIL'MeOH (1 mL, 20 % purity) in MeOH (4 mL) was added Pd/C (50 mg, 10 % purity) under N 2 . The suspension was degassed under vacuum and purged with H 2 several times. The mixture was stirred under H 2 (15 psi) at 15 °C for 0.5 hour. The reaction mixture was filtered and the filtrate was concentrated under vacuum to. 2-[(25)-4-[2-[[(25)-l -methylpyrrolidin-2-yl]methoxy]-8-phenyl-5, 6,7,9- tetrahydropyrimido[4,5-c]azepin-4-yl]piperazin-2-yl]acetonit rile (160 mg, 312 pmol, 84 % yield, 90 % purity) was obtained as a white solid which was used for next step without further purification.

[0340] Example 5: To a solution of 2-[(25)-4-[2-[[(25)-l -methylpyrrolidin-2-yl]methoxy]-8- phenyl-5,6,7,9-tetrahydropyrimido[4,5-c]azepin-4-yl]piperazi n-2-yl]acetonitrile (160 mg, 347 pmol, 1.0 eq) and DIEA (89.6 mg, 693 mhioΐ, 121 mT, 2.0 eq) in DCM (4 mL) was added prop-2- enoyl chloride (47.1 mg, 520 mihoΐ, 42.4 mT, 1 .5 eq) at -40 °C. The reaction mixture was stirred at -40 °C for 0.5 hour. The reaction mixture was quenched by water (5 mL). The mixture was extracted with DCM (3 x 10 mL). The combined organic layers were dried over anhydrous Na 2 SC>4, filtered and concentrated under vacuum. The residue was purified by prep-HPLC (column: Xtimate Cl 8 150*25mm*5pm;mobile phase: [water (0.05 % ammonium hydroxide v/v)-ACN];B%: 40 %-70 %, 8 min) to give 2-[(2S)-4-[2-[[(2S)-l -rnethylpyrrolidin-2- yl]methoxy]-8-phenyl-5,6,7,9-tetrahydropyrimido[4,5-c]azepin -4-yl]-l -prop-2-enoyl-piperazin- 2-yl]acetonitrile (38.6 mg, 73.4 pmol, 21 % yield, 98.1 % purity) as a white solid. LCMS [ESI, M+l]: 516.

[0341] Ή NMR (400 MHz, chloroform-d) d = 7.18 (t, J= 8.0 IIz, 2H), 6.85 - 6.81 (m, 2H), 6.69 (t, 7 = 7.2 Hz, 1H), 6.54 (br d, J= 10.4 Hz, 1H), 6.36 (dd, J= 1.6, 16.8 Hz, 1H), 5.80 (br d, J = 10.4 Hz, 1H), 5.03 (br s, 1H), 4.58 (s, 2H), 4.41 (dd, ,7= 4.8, 10.4 Hz, 1 H), 4.17 (dd, J= 7.2,

10.4 Hz, 1H), 4.03 - 3.66 (m, 4H), 3.55 (br d, 7= 1 1.2 Hz, 2H), 3.19 - 3.06 (m, 2H), 2.88 (br dd, J= 8.0, 16.4 Hz, 2H), 2.82 - 2.63 (m, 4H), 2.50 (s, 3H), 2.35 - 2.26 (m, 1H), 2.14 - 2.04 (m, 1H), 2.02 - 1 .89 (m, 2H), 1.89 - 1.72 (m, 3H).

EXAMPLE 6

2- [(2,S')-4- [2- [ [ (2.S')- 1 -methylpyrrolidin-2-yl]mcthoxy |-8-( 1 -naphthvl )-5,6.7,9- tetrahydropyrimido[4,5-c]azepin-4-yl]-l -prop-2-enoyl-piperazin-2-yl]acetonitrile

Example 6 [0342] Compound 6-1: To a mixture of benzyl (25)-2-(cyanomethyl)-4-[2-[[(25)-l- methylpyrrolidin-2-yl]methoxy]-6,7,8,9-tetrahydro-5//-pyrimi do[4,5-c]azepin-4-yl]piperazine-l- carboxylale (200 mg, 385 pmol, 1.0 eq), 1 -bromonaphthalene (120 mg, 577 pmol, 80.2 uL, 1.5 eq), CS 2 CO 3 (376 mg, 1.15 mmol, 3.0 eq) and RuPhos (71.8 mg, 154 pmol, 0.4 eq) in toluene (6 mL) was added Pd 2 (dba) 3 (70.5 mg, 77.0 pmol, 0.2 eq) under N 2 . The suspension was degassed under vacuum and purged with N 2 several times. The mixture was stirred under N 2 at 90 °C for 8 hours. Water (15 mL) was added into the mixture. The mixture was diluted with EtOAc (10 mL) and extracted with EtOAc (2 x 15 mL). The combined organic layers were washed with brine (20 mL). The combined organic layers were dried over anhydrous Na 2 S0 4 , filtered and concentrated under vacuum. The residue was purified by reverse-phase flash [water (0.1 % FA)/acetonitrile] to give benzyl (2,S')-2-(cyanomcthyl)-4-|2-| | (2L ' )- 1 -methylpyrrolidin-2- yl]methoxy]-8-(l-naphthyl)-5,6,7,9-tetrahydropyrimido[4,5-c] azepin-4-yl]piperazine-l- carboxylate (140 mg, 206 pmol, 54 % yield, 95 % purity) as a yellow solid. LCMS [ESI, M+l]: 646.

[0343] ‘H NMR (400 MHz, chloroform-d) 6 = 7.95 (d, J= 8.4 Hz, 1H), 7.81 (d, J= 8.0 Hz, 1H), 7.54 (d, .7= 8.4 Hz, 1H), 7.46 - 7.34 (m, 8H), 7.13 (d, J= 7.6 Hz, 1H), 5.21 (s, 2H), 4.70 (br s, 1H), 4.44 - 4.35 (m, 3H), 4.20 - 4.15 (m, 1H), 3.87 (br d, J= 12.4 Hz, 1H), 3.70 (br d, J= 12.4 Hz, 1H), 3.57 - 3.41 (m, 2H), 3.40 - 3.23 (m, 2H), 3.09 (br s, 1H), 2.99 (dt, .7= 3.2, 12.4 Hz, 1H), 2.91 (br s, 3H), 2.81 - 2.73 (m, 1H), 2.68 (br s, 1H), 2.47 (s, 3H), 2.28 (br d , J= 8.0 Hz, 1H),

2.19 - 2.06 (m, 2H), 2.04 - 1.99 (m, 1H), 1.89 - 1.69 (m, 4H).

[0344] Compound 6-2: To a solution of benzyl (2S)-2-(cyanomethyl)-4-[2-[[(2S')-l - methylpyrrolidin-2-yl]methoxy]-8-(l-naphthyl)-5,6,7,9-tetrah ydropyrimido[4,5-c]azepin-4- yl]piperazine-l-carboxylate (140 mg, 217 pmol, 1.0 eq) and NH 3 » MeOH (0.5 mL, 30 % purity) in MeOH (4 mL) was added Pd/C (50 mg, 10 % purity) under N 2 . The suspension was degassed under vacuum and purged with H 2 several times. The mixture was stirred under PI 2 (15 psi) at 15 °C for 0.5 hour. The reaction mixture was filtered and the filtrate was concentrated under vacuum to give 2-[(26)-4-[2-[[(2.S)- 1 -methylpyrrolidin-2-yl]methoxy]-8-( 1 -naphthyl)-5, 6,7,9- tetrahydropyrimido[4,5-c]azepin-4-yl]piperazin-2-yljacetonit rile (105 mg, 185 pmol, 85 % yield, 90 % purity) as a yellow solid which was used for next step without further purification. [0345] Example 6: To a solution of 2-[(2C)-4-[2-[[(25)-1 -methylpyrrolidin-2-yl]methoxy]-8-(l - naphthyl)-5,6,7,9-tetrahydropyrimido[4,5-c]azepin-4-yl]piper azin-2-yl]acetonitrile (105 mg, 205 pmol, 1.0 eq) and D1EA (53.0 mg, 410 pmol, 71.5 pL, 2.0 eq) in DCM (2 mL) was added prop- 2-enoyl chloride (27.9 mg, 308 pmol, 25.1 pL, 1.5 eq) at -40 °C. The reaction mixture was stirred at -40 °C for 0.5 hour. The reaction mixture was quenched with water (8 mL). The mixture was extracted with DCM (3 x 10 mL). The combined organic layers were dried over anhydrous Na ?. S() i, filtered and concentrated under vacuum. The residue was purified by reverse-phase flash [water (0.1% FA)/acetonitrile] The residue was purified by prep-HPLC (column: Xtimate C18 150*25mm*5pm;mobile phase: [water (0.05 % ammonium hydroxide v/v)-ACNj;B %: 54 % - 84 %, 10 min) to give 2-[(2S)-4-[2-[[(25 -l-methylpyrrolidin-2- yl]methoxy]-8- (l -naphthyl)-5,6,7,9-tetrahydropyrimido[4,5-c]azepin-4-yl]-l-p rop-2-enoyl- piperazin-2-yl]acetonitrile (27.9 mg, 49.4 pmol, 24 % yield, 100 % purity) as a white solid.

I C MS [ES1, M+lj: 566.

[0346] *H NMR (400 MHz, chloroform-d) d = 7.95 (d, J= 8.4 Hz, 1H), 7.81 (d, J= 8.0 Hz, 1H), 7.54 (d, J= 8.4 Hz, 1H), 7.47 - 7.34 (m, 3H), 7.13 (d, J- 6.8 Hz, 1H), 6.59 (br d, J= 1 1.2 Hz, 1H), 6.40 (dd, J= 1.6, 16.8 Hz, 1 H), 5.83 (br d, /= 10.8 Hz, 1H), 5.1 1 (br s, 1H), 4.45 - 4.32 (m, 3H), 4.17 (dd, J = 6.8, 10.8 Hz, 1H), 3.93 (br d, J= 13.6 Hz, 2LI), 3.78 (br d , j= 12.0 Hz, 1 H), 3.69 - 3.39 (m, 3H), 3.30 (dd, J= 4.0, 13.6 Hz, 1H), 3.13 - 2.74 (m, 6H), 2.72 - 2.61 (m, 1H), 2.46 (s, 3H), 2.33 - 2.23 (m, 1H), 2.21 - 1.97 (m, 3H), 1.89 - 1.70 (m, 3H).

EXAMPLE 7

2-((5)-l -acryloyl-4-(8-(8-chloronaphthalen-l-yl)-2-(((S r )-l-methylpyrrolidin-2-yl)methoxy)-

6,7,8,9-tetrahydro-57/-pyrimido[4,5-c]azepin-4-yl)piperaz in-2-yl)acetonitrile

Cbz

[0347] Compound 7-1: To a solution of benzyl (2S)-2-(cyanomethyl)-4-[2-[[(2,S)-l - methylpyrrolidin-2-yl]methoxy]-6,7,8,9-tetrahydro-5/7-pyrimi do[4,5-c]azepin-4-yl]piperazine-l - carboxylate (250 mg. 481 pmol, 1.0 eq) and l-bromo-8-chloro- naphthalene (232 mg, 962 pmol, 2.0 eq) in toluene (4.0 mL) was added RuPhos (89.8 mg, 192 pmol, 0.4 eq), CS 2 CO3 (392 mg, 1.20 mmol, 2.5 eq) and Pd 2 (dba) 3 (88.1 mg, 96.2 pmol, 0.2 eq), the reaction mixture was stirred at 90 °C for 10 hours under N 2 . After completion, the reaction mixture was filtered through a celite, the filter cake was washed with ethyl acetate (10 mL), and adjusted with 1N HC1 aqueous to pH~3, the organic layer was separated, and the aqueous was adjusted with Na 2 C03 solid to pi 1 8. extracted with ethyl acetate (2 x 10 mL), the organic layer was washed with saturated brine (1 c 20 mL), dried over Na 2 S0 4 , filtered and concentrated. The residue was purified by reverse phase flash (Cl 8, 0.1 %FA in water, 30%-50% MeCN). The product benzyl (25 -4-[8-(8- chloro-l-naphthyl)-2-[[(25)-l -methylpyrrolidin-2-yl] methoxy]-5,6,7,9-tetrahydropyrimido[4,5- c]azepin-4-yl]-2-(cyanomethyl)piperazine-l-carboxylate (130 mg, 185 pmol, 38% yield, 97% purity) was obtained as yellow solid. LCMS [ES1, M+l]: 680.

[0348] Ή NMR (400 MHz, chloroform-d) d 7.74 - 7.68 (m, 1H), 7.59 - 7.51 (m, 1H), 7.48 - 7.15 (m, 9H), 5.21 (s, 2H), 4.74 - 4.63 (m, 1H), 4.40 - 4.29 (m, 211), 4.21 - 4.05 (m, 211), 3.84 - 3.71 (m, 1 H), 3.69 - 3.46 (m, 2H), 3.44 - 3.15 (m, 3LI), 3.13 - 2.60 (m, 8H), 2.46 (s, 3H), 2.33 - 2.21 (m, 1 H), 2.17 - 2.02 (m, 2H), 1.96 - 1.72 (m, 4H). LCMS [ESI, M+l]: 546.

[0349] Compound 7-2: To a solution of TMSC1 (239 mg, 2.21 mmol, 280 pL, 15.0 eq) in MeCN (2 mL) containing 4A molecular sieves (100 mg) at 0 °C was added Nal (353 mg, 2.35 mmol, 16.0 eq) in portions. Stirring was continued for a period of 1 hour at 15 °C. Then a solution of benzyl (2A)-4-[8-(8-chloro-l-naphthyl)-2-[[(25)-l- methylpyrrolidin-2-yl]methoxy]-5, 6,7,9- tetrahydropyrimido[4,5-c]azepin-4-yl]-2-(cyanomethyl)piperaz ine-l-carboxylate (100 mg, 147 pmol, 1.0 eq) in MeCN (1 mL) was added to the reaction, and the reaction mixture was stirred at 15 °C for 1 1 hours. After completion, the reaction mixture was concentrated, then the reaction mixture was added IN HC1 aqueous (8 mL), extracted with methyl tert- butyl ether (2 x 5 mL), the organic layer was discarded, and the aqueous phase was adjusted to pH~8 with saturated Na 2 C0 3 aqueous, and then extracted with ethyl acetate (2 x 8 mL), the organic layer was dried over Na 2 S04, filtered and concentrated. The product 2-[(25)-4-[8-(8-chloro-l-naphthyl)-2-[[(25)- l-methylpyrrolidin-2-yl]methoxy]-5,6,7,9-tetrahydropyrimido[ 4,5-c]azepin-4-yl]piperazin-2- yl] acetonitrile (45 mg, 81.0 pmol, 55% yield, 98% purity) was obtained as brown oil.

methylpyrrolidin-2-yl]methoxy]-5,6,7,9-tetrahydropyrimido[4, 5-c]azepin-4-yl]piperazin-2- yljacetonitrile (45 mg, 82.4 pmol, 1.0 eq) in dichloromethane (1.0 mL) was added DIEA (42.6 mg, 329 pmol, 57.4 uL, 4.0 eq) and prop-2-enoyl chloride (1 1.2 mg, 124 pmol, 10.1 pL, 1.5 eq) in portions at -40 °C, the reaction mixture was stirred at -40 °C for 0.5 hour. After completion, the reaction mixture was quenched with water (1.0 mL) at -40 °C, the reaction mixture was warmed to 20 °C, and diluted with water (5 mL), then extracted with dichloromethane (2 x 5 mL), the combined organic layer was dried over Na 2 SC>4, filtered and concentrated. The residue was purified column chromatography (base AI 2 O 3 , petroleum ether/ethyl acetate=3/l to petroleum ether/ethyl acetate/methanol=3/l/0.1), the crude product was repurified by prep-HPLC (column: Xtimate C18 150*25mm*5pm; mobile phase: [water (0.05% ammonium hydroxide v/v)-ACN]; B%: 56%-86%, 10 min). The product 2-(( 1 S)-l- acryloyl-4-(8-(8-chloronaphthalen-l- yl)-2-((( L S')-l-methylpyrrolidin-2-yl)methoxy)-6,7,8,9-tctrahydr o-5//-pyrimido[4,5-c]azepin-4- yl)piperazin-2-yl)acetonitrile (14.8 mg, 24.1 pmol, 29 % yield, 97% purity) was obtained as white solid. LCMS [ESI, M+l j: 600.

[0351] ‘H NMR (400 MIIz, chloroform-d) d 7.74 - 7.68 (m, 1H), 7.58 - 7.52 (m, HI), 7.47 - 7.43 (m, 1H), 7.41 - 7.33 (m, 111), 7.32 - 7.16 (m, 2H), 6.71 - 6.53 (m, 1H), 6.39 (d, J= 16.8 IIz, HI), 5.83 (br d, J - 10.4 Hz, 1H), 5.18 - 5.05 (m, HI), 4.52 - 4.29 (m, 3H), 4.16 - 4.08 (m, 1 H), 4.02 - 3.78 (m, 2H), 3.75 - 3.47 (m, 3H), 3.43 - 3.32 (m, HI), 3.30 - 3.16 (m, 1H), 3.08 (br t, ./= 7.6 Hz, 1H), 3.04 - 2.87 (m, 3H), 2.84 - 2.60 (m, 3H), 2.45 (s, 3H), 2.34 - 2.21 (m, 1H), 2.19 - 1.97 (m, 2H), 1.96 - 1.72 (m, 4H).

EXAMPLE 8

2-[(25)-4-[8-(3-isopropylphenyl)-2-[[(25)-l-methylpyrroli din-2-yl]methoxy]-5, 6,7,9- tetrahydropyrimido[4,5-c]azepin-4-yl]-l-prop-2-enoyl-piperaz in-2-yl]acetonitrile

[0352] Compound 8-1: To a mixture of benzyl (25 -2-(cyanomethyl)-4-[2-[[(2S’)-l- methylpyrrolidin-2-yl]methoxy]-6,7,8,9-tetrahydro-5/7-pyrimi do[4,5-c]azepin-4-yl]piperazine-l- carboxylate (150 mg, 289 pmol, 1.0 eg), l-bromo-3-isopropyl-benzene (86.2 mg, 433 pmol, 12.9 pL, 1.5 eq ), CS 2 CO 3 (282 mg, 866 pmol, 3.0 eg) and RuPhos (53.9 mg, 1 15 pmol, 0.4 eg) in toluene (4 mL) was added Pd 2 (dba)3 (52.9 mg, 57.7 pmol, 0.2 eq) under N 2 . The suspension was degassed under vacuum and purged with N 2 several times. The mixture was stirred under N 2 at 90 °C for 8 hours. Water (15 mL) was added into the mixture. The mixture was diluted with EtOAc (10 mL) and extracted with EtOAc (2 x 15 mL). The combined organic layers were washed with brine (20 mL). The combined organic layers were dried over anhydrous Na 2 S0 4 , filtered and concentrated under vacuum. The residue was purified by reverse-phase flash [water (0.1 % FA)/acetonitrile] to give benzyl (2S)-2-(cyanomethyl)-4-[8-(3-isopropylphenyl)-2-[[(2.S)-

1-methylpyrrolidin-2-yl]methoxy]-5,6,7,9-tetrahydropyrimi do[4,5-c]azepin-4-yl]piperazine-l- carboxylate (120 mg, 179 pmol, 62 % yield, 95 % purity) as a yellow solid. LCMS [ESI, M+l]: 638.

[0353] 'H NMR (400 MHz, chloroform-d) d = 7.42 - 7.32 (m, 5H), 7.1 1 (t, J - 8.0 Hz, 1H), 6.72 (s, 1H), 6.65 (dd, J= 2A, 8.0 Hz, 1H), 6.58 (d, 7= 7.6 Hz, HI), 5.24 - 5.13 (m, 2H), 4.71 - 4.55 (m, 3H), 4.41 (dd, J- 4.8, 10.8 Hz, 1H), 4.21 - 3.99 (m, 2H), 3.84 - 3.60 (m, 3H), 3.46 (br d, 7 =

12.8 Hz, 1H), 3.24 (br s, 1H), 3.12 (br dd, 7- 3.6, 13.2 Hz, 2H), 2.93 - 2.63 (m, 7H), 2.50 (s, 3H), 2.34 - 2.24 (m, 1H), 2.13 - 2.06 (m, 1H), 1.96 (br s, 2H), 1.88 - 1.74 (m, 3H), 1.21 (d, 7-

6.8 Hz, 6H).

[0354] Compound 8-2: To a solution of benzyl (2S)-2-(cyanomethyl)-4-[8-(3-isopropyiphenyl)-

2-| [(2, S')- 1 -methylpyrrolidin-2-yl]mcthoxy |-5,6,7,9-tctrahydropyrimido| 4,5-c|azepin-4- yl]piperazine-l-carboxylate (120 mg, 188 pmol, 1.0 eq) and NHi ' MeOH (1 mL, 20 % purity) in MeOH (4 mL) was added Pd/C (50 mg, 10 % purity) under N 2 . The suspension was degassed under vacuum and purged with H 2 several times. The mixture was stirred under 112 (15 psi) at 15 °C for 0.5 hour. The reaction mixture was filtered and the filtrate was concentrated under vacuum to give 2-[(25)-4-[8-(3-isopropylphenyl)-2-[[(27)-l-methylpyrrolidin -2-yl]methoxy]- 5,6,7,9-tetrahydropyrimido[4,5-c]azepin-4-yl]piperazin-2-yl] acetonitrile (60 mg, 107 pmol, 57 % yield, 90 % purity) as a yellow solid which was used for next step without further purification.

[0355] Example 8: To a solution of 2-[(25)-4-[8-(3-isopropylphenyl)-2-[[(27)- 1 - methylpyrrolidin-2-yl]methoxy]-5,6,7,9-tetrahydropyrimido[4, 5-c]azepin-4-yl]piperazin-2- yl]acetonitrile (60 mg, 119 pmol, 1.0 eq) and DIEA (30.8 mg, 238 pmol, 41.5 pL, 2.0 eq) in DCM (1.5 mL) was added prop-2-enoyl chloride (16.2 mg, 179 pmol, 14.6 pL, 1.5 eq) at -40 °C. The reaction mixture was stirred at -40 °C for 0.5 hour. The reaction mixture was quenched by water (8 mL). The mixture was extracted with DCM (3 c 10 mL). The combined organic layers were dried over anhydrous Na 2 S0 4 , filtered and concentrated under vacuum. The residue was purified by reverse-phase flash [water (0.1 % FA)/acetonitrile] The residue was purified by prep-IIPLC (column: Waters Xbridge 150*25 5u;mobile phase: [water (0.05 % ammonium hydroxide v/v)-ACN];B %: 55 % - 76 %, 10 min) to give 2-|(27)-4-[8-(3-isopropylphenyl)-2- [[(2S)-l-methylpynOlidin-2-yl]methoxy]-5,6,7 ; 9-tetrahydropyrimido[4.5-c]azepin-4-yl]-l -prop- 2-enoyl-piperazin-2-yl]acetonitrile (28.8 mg, 51.3 pmol, 43 % yield, 99.4 % purity) as a white solid. LCMS [ESI, M+l]: 558.

[0356] Ή NMR (400 MHz, chloroform-d) d = 7.1 1 (t, J= 8.0 Hz, 1H), 6.72 (s, 1H), 6.66 (dd, J = 2.4, 8.4 Hz, 1H), 6.59 (d, J= 7.6 Hz, 1H), 6.53 (br s, 1H), 6.40 - 6.32 (m, 1H), 5.80 (br d, J = 10.4 Hz, 1H), 5.05 (br s, 1H), 4.58 (s, 2H), 4.42 (br s, 1H), 4.18 (br dd, J= 7.2, 10.4 Hz, 1H), 4.07 - 3.65 (m, 4H), 3.55 (br d, J= 13.2 Hz, 2H), 3.14 (br dd, J= 3.6, 13.6 PIz, 2H), 2.98 - 2.62 (m, 7H), 2.52 (s, 3H), 2.32 (br d, j= 8.8 Hz, 1H), 2.16 - 1.91 (m, 3H), 1.89 - 1.70 (m, 3H), 1.21 (d, J= 6.8 Hz, 6H).

EXAMPLE 9

2-((A)-l-acryloyl-4-(8-(3-fluoro-2-methylphenyl)-2-(((S )-l-methylpyrrolidin-2-yl)methoxy)-

6,7,8,9-tetrahydro-57/-pyrimido[4,5-c]azepin-4-yl)piperaz in-2-yl)acetonitrile

Cbz

i

[0357] Compound 9-1 : To a solution of benzyl (25)-2-(cyanomethyl)-4-[2-[[(25) -1 - methy lpyrrolidin-2 -y l]methoxy ] -6,7, 8 ,9-tetrahy dro-577-pyrimido [4,5-c] azepin-4-y 1] pi perazine- 1 - carboxylate (150 mg, 289 pmol, 1.0 eq) and l-bromo-3-fluoro-2- methyl-benzene (109 mg, 577 pmol, 2.0 eq) in toluene (3.0 mL) was added RuPhos (53.9 mg, 1 15 pmol, 0.4 eq), CS 2 CO 3 (235 mg, 722 pmol, 2.5 eq) and Pd 2 (dba) 3 (52.9 mg, 57.7 pmol, 0.2 eq), the reaction mixture was stirred at 90 °C for 12 hours under N 2 . After completion, the reaction mixture was filtered through a celite, the filter cake was washed with ethyl acetate (10 mL), and adjusted with 1N HC1 aqueous to pH~3, then the organic layer was separated, and the aqueous was adjusted with Na 2 C0 3 solid to pH~8, extracted with ethyl acetate (2 x 10 mL), the organic layer was washed with saturated brine (1 x 20 mL), dried over Na 2 S0 4 , filtered and concentrated. The residue was purified by reverse phase flash (Cl 8, 0.l%FA in water, 30%-50% MeCN). The product benzyl (2S)-2-(cyanomethyl)-4-[8-(3-fluoro-2-methyl-phenyl)-2-[[(2S )-l - methylpyrrolidin-2- yl]methoxy]-5,6,7,9-tetrahydropyrimido[4,5-c]azepin-4-yl]pip erazine-l -carboxylate (1 10 mg, 175 pmol, 61% yield, 100% purity) was obtained as brown oil. LCMS [ESI, M+l]: 628.

[0358] Ή NMR (400 MHz, chloroform-d) d 7.44 - 7.31 (m, 5H), 7.1 1 - 7.02 (m, I II), 6.81 (d, J = 8.4 Hz, 1H), 6.73 (t, J= 8.6 Hz, 1H), 5.20 (s, 2H), 4.73 - 4.61 (m, 1H), 4.45 - 4.32 (m, III), 4.18 - 4.13 (m, 4H), 3.83 (br d, J= 13.2 Hz, 1H), 3.65 (br d, J- 12.4 Hz, 1H), 3.41 - 3.1 8 (m, 4H), 3.14 - 3.05 (m, 1H), 3.00 - 2.63 (m, 6H), 2.48 (s, 3H), 2.36 - 2.21 (m, 1H), 2.13 - 1.94 (m, 6H), 1.89 - 1.73 (m, 3H).

[0359] Compound 9-2: To a solution of benzyl (2S)-2-(cyanomethyl)-4-[8-(3- fluoro-2-methyl- phenyl)-2-[[(2A)-l-methylpyrrolidin-2-yl]methoxy]-5,6,7,9-te trahydropyrimido[4,5-c]azepin-4- yl]piperazine-l -carboxylate (1 10 mg, 175 pmol, 1.0 eq) in methanol (1.5 mL) was added Pd/C (30 mg, 175 pmol, 10% purity, 1.0 eq) and NH 3 » MeOH (1.5 mL, 20% purity, 1.0 eq), the suspension was degassed under vacuum and purged with H 2 several times. The mixture was stirred under H 2 (15 psi) at 20 °C for 1 hour. After completion, the reaction mixture was filtered through a pad of celite, and the filter cake was washed with dichloromethane (2 x 5 mL), the filtrate was concentrated. The product 2-[(2S)-4-[8-(3-fluoro-2-methyl-phenyl)-2-[[(2S)-l - methylpyrrohdin-2-yl]methoxy]-5,6,7,9-tetrahydropyrimido[4,5 -c]azepin-4-yl]piperazin-2- yl]acetonitrile (70 mg, 140 pmol, 80% yield, 99% purity) was obtained as white solid which was used for the next step without further purification. LCMS [ESI, M+l]: 494. [0360] Example 9: To a solution of 2-[(2S)-4-[8-(3-fluoro-2-methyl-phenyl) -2-[[(2S)-l- methylpyrrolidin-2-yl]methoxy]-5,6,7,9-tetrahydropyrimido[4, 5-c]azepin-4-yl]piperazin-2- yljacetonitrile (70 mg, 142 pmol, 1.0 eq) in dichloromethane (1.5 mL) was added DIEA (73.3 g, 567 pmol, 98.8 pL, 4.0 eq) and prop-2-enoyl chloride (19.3 mg, 213 pmol, 17.3 pL, 1.5 eq) in portions at -40 °C, the reaction mixture was stirred at -40 °C for 0.5 hour. After completion, the reaction mixture was quenched with water (1.0 mL) at -40 °C, then the reaction mixture was warmed to 20 °C, and diluted with water (5 mL), extracted with dichloromethane (2 x 5 mL), the combined organic layer was dried over Na 2 S04, filtered and concentrated. The residue was purified column chromatography (base AI 2 O 3 , petroleum ether/ethyl acetate=3/l to petroleum ether/ethyl acetate/methanol=3/l/0.l), the crude product was repurified by prep-IIPLC (column: Xtimate C18 l50*25mm*5pm; mobile phase: [water (0.05% ammonium hydroxide v/v)-ACN]; B%: 51%-81%, 10 min). The product 2-((S)-l- acryloyl-4-(8-(3-fhroro-2-methylphenyl)-2-(((S)-

1-methylpyrrolidin-2-yl)methoxy)-6,7,8,9-tetrahydro-5//-p yrimido[4,5-c]azepin-4-yl)piperazin-

2-yl)acetonitrile (19.7 mg, 35.7 pmol, 25% yield, 99% purity) was obtained as white solid. LCMS [ESI, M+l]: 548.

[0361] 1 II NMR (400 MHz, chloroform-d) d 7.12 - 7.03 (m, 1H), 6.81 (d, J= 8.0 Hz, 1H), 6.73 (t, J= 8.6 Hz, 1H), 6.65 - 6.51 (m, 1H), 6.40 (dd, J= 1.6, 16.8 Hz, 1H), 5.83 (br d, 10.8 Hz, 1H), 5.16 - 5.07 (m, 1H), 4.38 (dd, J= 4.4, 10.4 Hz, 1H), 4.21 - 4.08 (m, 3H), 4.03 - 3.85 (m,

2H), 3.82 - 3.46 (m, 2H), 3.41 - 3.19 (m, 3H), 3.16 - 2.90 (m, 3H), 2.87 - 2.61 (m, 4H), 2.48 (s,

3H), 2.35 - 2.23 (m, 1H), 2.17 - 1.93 (m, 6H), 1.89 - 1.70 (m, 3H).

EXAMPLE 10

2-((5)-l-acryloyl-4-(8-(2-fluoronaphthalen-l-yl)-2-(((S)- l-methylpyrro]idin-2-yl)methoxy)-

6,7,8,9-tetrahydro-5fT-pyrimido[4,5-c]azepin-4-yl)piperaz in-2-yl)acetonitrile

[0362] Compound 10-1 : A mixture of benzyl (2 < S)-2-(cyanomethyl)-4-[2-[[(2S) -1- methylpyrrolidin-2-yl]methoxy]-6,7,8,9-tetrahydro-5ff-pyrimi do[4,5-c]azepin-4-yl]piperazine-l- carboxylate (300 mg, 577 mhioΐ, 1 eq ), (2-fluoro-l -naphthyl) trifluoromethanesulfonate (339 mg, 1.15 mmol, 2 eg), RuPhos (107 mg, 231 pmol, 0.4 eq), Pd 2 (dba)3 (105 mg, 1 15 mthoI, 0.2 eq) and CS2CO3 (564 mg, 1.73 mmol, 3 eq) in toluene (5 mL) was degassed and purged with N 2 for 3 times, and then the mixture was stirred at 90 °C for 12 hours under N 2 atmosphere. The reaction mixture was diluted with water (20 mL) and extracted with ethyl acetate (20 mL x 3). The combined organic layers were washed with brine (20 mL), dried over Na 2 S0 4 , filtered and concentrated under reduced pressure to give a residue. The residue was purified by reverse phase flash [water (0.1% formic acid)/acetonitrile)]. The collected desired fractions were neutralized with saturated NaHCCb aqueous solution to pH = 7 and extracted with ethyl acetate (20 mL x 3). The combined organic layers were washed with brine (20 mL), dried over Na 2 S0 4 , filtered and concentrated under reduced pressure to give the product benzyl (25 -2-(cyanomethyl)-4-[8-(2- fluoro-1 -naphthyl)- 2-[[(2<5')-l-methylpyrrolidin-2-yl]methoxy]-5,6,7,9-tetra hydropyrimido[4,5- c]azepin-4-yl]piperazine-l -carboxylate (140 mg, 192 pmol, 33% yield, 91% purity) was obtained as a yellow oil. LCMS [ESI, M+l]: 664. Compound 10-2: To a solution of benzyl (2 S)- 2-(cyanomethyl)-4-[8-(2-fIuoro- l-naphthyl)-2-[[(25 , )-l-methylpyrrolidin-2-yl]methoxy]-5,6,7,9- tetrahydropyrimido[4,5-c]azepin-4-yl]piperazine-l -carboxylate (140 g, 210 pmol, 1 eq) in methanol (5 mL) was added NH 3* MeOH (2 mL, 20% purity) and Pd/C (50 mg, 10% purity) under N 2 . The suspension was degassed under vacuum and purged with H 2 several times. The mixture was stirred under H 2 (15 psi) at 25°C for 0.5 hour. The Pd/C was filtered off and the filtrate was concentrated under vacuum. 2-[(2S')-4-[8-(2-fluoro-l-naphthyl)-2-[[(25 -l- methylpyrrolidin-2-yl]mcthoxyJ-5,6,7,9-tetrahydropyrimido[4, 5-c]azepin-4-yl]piperazin-2- yl] acetonitrile (80 mg, 151 mihoΐ, 72% yield) was obtained as a yellow oil and used into next step without further purification. LCMS fESl, M+l]: 530.

[0363] Example 10: To a solution of 2-[(25 -4-[8-(2-fluoro-l-naphthyl)-2- [[(2S)-l- methylpyrrolidin-2-yl]methoxy]-5,6,7,9-tetrahydropyrimido[4, 5-c]azepin-4-yl]piperazin-2- yljacetonitrile (60 mg, 1 13 pmol, 1 eq ) and DIEA (73.2 mg, 566 pmol, 98.7 pL, 5 eq) in DCM (1 mL) was added a solution of prop-2-enoyl chloride (15.4 mg, 169 pmol, 13.9 pL, 1.5 eq) in DCM (1 mL) at - 40 °C. After stirred at - 40 °C for 0.5 hour, the reaction mixture was quenched with water (20 mL) and extracted with DCM (20 mL c 3). The combined organic layers were washed with brine (10 mL), dried over Na 2 S0 4 , filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (Si0 2 , Ethyl

acetate/Methanol = 100/1 to 10/1), followed by prep-HPLC (column: Waters Xbridge 150*25 5p; mobile phase: [water (0.05% ammonium hydroxide v/v) - ACN]; B%: 52% - 76%, 10 min). The desired fraction was collected and lyophilized. 2-[(2X)-4-[8-(2-fluoro-l-naphthyl)-2-[[(2S - l-methylpyrrolidin-2-yl]methoxy]-5,6,7,9-tetrahydropyrimido[ 4,5-c]azepin-4-yl]-l-prop-2- enoyl-piperazin-2-yl]acetonitrile (21 mg, 35.1 pmol, 31% yield, 97% purity) was obtained as a white solid. LCMS [ESI, M+l]: 584.

[0364] Ή NMR (400MHz, chloroform-d) d = 8.18 (d, J= 7.6 Hz, 1H), 7.80 (dd, J= 1.6, 7.6 Hz, 1H), 7.65 (dd, J= 5.2, 8.8 Hz, 1H), 7.49 - 7.38 (m, 2H), 7.27 - 7.20 (m, 1H), 6.75 - 6.53 (m, 1H), 6.41 (dd, ./ = 1.6, 16.8 Hz, 1H), 5.84 (d, J= 10.4 Hz, HI), 5.31 - 4.47 (m, 2H), 4.42 - 4.16 (m, 2H), 4.20 - 4.08 (m, III), 4.05 - 3.80 (m, 2H), 3.79 - 3.48 (m, 2H), 3.45 - 2.54 (m, 1011), 2.44 (s, 3H), 2.34 - 2.19 (m, 1H), 2.19 - 1.92 (m, 3H), 1.90 - 1.72 (m, 3H).

EXAMPLE 1 1

2-((X)- 1 -acryloyl-4-(8-(5-methylnaphthalen- 1 -yl)-2-(((5')- 1 -methylpyrrolidin-2-yl)mcthoxy)- 6,7,8,9-tetrahydro-5//-pyrimido[4,5-c]azepin-4-yl)piperazin- 2-yl)acetonitrile

11-A 11-B

[0365] Compound 11-B: To a mixture of l,5-dibromonaphthalene (1.00 g, 3.50 mmol, 1.00 eq) in THF (10.0 mL) was added n-BuLi (2.5 M, 1.82 mL, 1.3 eq) in portion at -78 °C under N 2 . The mixture was stirred at -78 °C for 30 min, then CH3I (4.58 g, 32.3 mmol, 2.01 mL, 9.23 eq) was added dropwise and warmed to 25 °C and stirred for 1 hour. The reaction mixture was quenched with water (15.0 mL), then extracted with ethyl acetate(30.0 mL c 3). The combined organic layers were washed with brine (50.0 mL c 1), dried over Na 2 S0 4 , filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (Si0 2 , Petroleum ether/Ethyl acetate- 1/0 to 10/1). Compound l-bromo-5-methy 1-naphthalene (450 mg, 2.01 mmol, 58% yield) was obtained as a yellow solid.

[0366] Ή NMR (400MHz, chloroform-d) d = 8.15 (d, J= 8.4 Hz, 1H), 7.99 (d, J= 8.4 Hz, 1H), 7.81 (dd, J= 0.8, 7.2 Hz, 1H), 7.49 (dd, J= 7.2, 8.8 Hz, III), 7.41 - 7.33 (m, 2H), 2.72 (s, 3H).

[0367] Compound 11-1: To a mixture of benzyl (2S)-2-(cyanomethyl)-4-[2-[[(25)-l- methylpyrrolidin-2-yl]methoxy]-6,7,8,9-tetrahydro-5//-pyrimi do[4,5-c]azepin-4-yl]piperazine-l- carboxylate (400 mg, 770 mihoΐ, 1.00 eq) and 1 -bromo- 5 -methyl- naphthalene (204 mg, 924 mihoΐ, 1.20 eq) in toluene (10.0 mL) was added Pd 2 (dba) 3 (141 mg, 154 mihoΐ, 0.20 eq), RuPhos (144 mg, 308 pmol, 0.4 eq), Cs 2 C0 3 (752 mg, 2.31 mmol, 3.00 eq) under N 2 . The mixture was degassed and purged with N 2 for 3 times, then heated to 90 °C and stirred for 8 hours. The reaction mixture was diluted with water (20.0 mL) and extracted with ethyl acetate (30 mL x 3). The combined organic layers were washed with brine (50 mL x 1), dried over sodium sulfate, filtered and concentrated under reduced pressure to give a residue. The residue was purified by reverse phase flash [water (0.1% FA)/acetonitrile]. The desired fractions were collected and neutralized with saturated NaHCCfi solution and extracted with ethyl acetate (50 mL x 3). The separated organic layer was dried over sodium sulfate, filtered and concentrated under vacuum. Compound benzyl (2£)-2-(cyanomethyl)-4- [8-(5-methyl-l-naphthyl)-2-[[(25 -l- methylpyrrolidin-2-yl]methoxy]-5,6,7,9-tetrahydropyrimido[4, 5-c]azepin-4-yl]piperazine-l - carboxylate (310 mg, 470 pmol, 61% yield, 100% purity) was obtained as a yellow solid. LCMS [ESI, M+l]: 660.

[0368] l H NMR (400MHz, chloroform-d) 6 - 7.86 (d, J= 7.6 Hz, HI), 7.71 (d, J= 8.4 Hz, 1H), 7.46 - 7.34 (m, 6H), 7.31 - 7.27 (m, 1H), 7.26 - 7.22 (m, III), 7.15 (d, J= 7.2 Hz, 1H), 5.27 - 5.14 (m, 2H), 4.70 (hr s, 1H), 4.43 - 4.33 (m, 3H), 4.22 - 4.06 (m, 2H), 3.87 (br d, J= 11.6 Hz, 1H), 3.70 (br d , J= 12.8 Hz, 1H), 3.57 - 3.39 (m, 2H), 3.39 - 3.20 (m, 2H), 3.09 (br t, J= 7.6 Hz, 1H), 3.05 - 2.83 (m, 4LI), 2.82 - 2.73 (m, 1H), 2.68 (s, 4H), 2.46 (s, 311), 2.33 - 2.23 (m, 1H),

2.17 - 2.00 (m, 3H), 1.90 - 1.72 (m, 3H).

[0369] Compound 11-2: To a solution of benzyl (2S -2-(cyanomcthyl)-4-[8-(5-methyl-l- naphthyl)-2-[[(25)-l-methylpyrrolidin-2-yl]methoxy]-5,6,7,9- tetrahydropyrimido[4,5-c]azepin- 4-yl]piperazine-l-carboxylate (160 mg, 242 pmol, 1.00 eq) in methanol (3.00 mL) was added Pd/C (50.0 mg, 10% purity), NH 3* MeOH (3.00 mL, 20% purity) under N 2 . The suspension was degassed under vacuum and purged with II 2 several times. The mixture was stirred under H 2 (15 psi) at 25 °C for 2 hours. The reaction mixture was concentrated under reduced pressure to give a residue. The crude product was used in the next step directly without further purification.

Compound 2-[(25)-4-[8-(5-methyl-l-naphthyl)-2-[[(2S)-l -methylpyrrolidin-2-yl]methoxy]- 5,6,7,9-tetrahydropyrimido[4,5-c]azepin-4-yl]piperazin-2-yl] acetonitrile (150 mg, 282 pmol,

99% yield, 99% purity) was obtained as a white solid. LCMS [ESI, M+l]: 526.

[0370] Example 11: To a mixture of 2-[(25 -4-[8-(5-methyl-l -naphthyl)-2-[[(25)-l - methylpyrrolidin-2-yl]methoxy]-5,6,7,9-tetrahydropyrimido[4, 5-c]azcpin-4-yl]piperazin-2- yl]acetonitrile (120 mg, 228 mmol, 1.00 eq) in dichloromethane (3.00 mL) was added TEA (1 15 mg, 1.14 mmol, 159 m , 5.00 eq) and prop-2-enoyl chloride (31.0 mg, 342 pmol, 27.9 pL, 1.50 eq) in portion at -40 °C under N 2 . Then the mixture was stirred at -40 °C for 0.5 hours. The reaction mixture was diluted with ice-water (5.00 mL) and extracted with dichloromethane (30.0 mL x 2). The combined organic layers were washed with brine (10.0 mL x 1), dried over sodium sulfate, filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (column: Xtimate Cl 8 150 x 25 mm x 5 pnpmobile phase: [water (0.05% ammonium hydroxide v/v)-ACN];B%: 55%-85%,10 min). Compound 2-[(25)-4-[8-(5- methyl- 1 -naphthyl)-2-[[(25)- 1 -methylpyrrolidin-2-yl]methoxyj-5,6,7,9-tetrahydropyrimido[4 ,5- c]azepin-4-yl]-l -prop-2-enoyl-piperazin-2-yl]acetonitrile (22.3 mg, 38.4 mhioΐ, 17% yield,

99.8% purity) was obtained as a off-white solid. LCMS [ESI, M+l]: 580.

[0371 ] Ή NMR (400MHz, chloroform-d) d = 7.86 (br d, J= 12 Hz, III), 7.72 (d. J = 8.4 Hz, 1H), 7.43 (t, ./ = 7.6 Hz, 1H), 7.30 - 7.28 (m, 1LI), 7.27 - 7.23 (m, 1H), 7.16 (d , J= 12 Hz, 1H), 6.59 (br d, 1 1.2 Hz, 1H), 6.40 (dd, ./ = 1.6, 16.4 Hz, 1H), 5.83 (br d , J = 10.6 Hz, HI), 5.1 1 (br s, 1H), 4.45 - 4.30 (m, 3H), 4.16 (dd, J= 6.8, 10.4 Hz, 1H), 3.92 (br d , J= 13.6 Hz, 1H), 3.78 (br d, J = 12.0 Hz, lH), 3.70 - 3.38 (m, 3LI), 3.29 (dd, J= 4.0, 13.6 Hz, 1H), 3.16 - 2.72 (m, 7H), 2.71 - 2.59 (m, 4H), 2.45 (s, 3H), 2.27 (dt, .7= 7.2, 9.2 Hz, 1H), 2.18 - 1.96 (m, 3H), 1.89 - 1.73 (m, 3 LI).

EXAMPLE 12

2-((X)- 1 -acryloy l-4-(8-(5-fluoronaphthalen- 1 -yl)-2-(((S -l-methylpyrrolidin-2-yl)methoxy)- 6,7,8,9-tetrahydro-5i/-pyrimido[4,5-c]azepin-4-yl)piperazin- 2-yl)acetonitrile

[0372] Compound 12-B: To a mixture of 5-bromonaphthalen-l -amine (2.00 g, 9.01 mmol, 1.00 eq) in pyridine * hydrofluoride (29.8 g, 180 mmol, 27.1 mL, 20.0 eq) was added NaN0 2 (2.49 g, 36.0 mmol, 4.00 eq) at 0 °C under N 2 . The mixture was stirred at 25 °C for 30 min, and then heated to 60 °C and stirred for 2 hours. The reaction mixture was diluted with water (20.0 mL) and extracted with ethyl acetate (30.0 mL x 3). The combined organic layers were washed with brine (50.0 mL c 1), dried over sodium sulfate, filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (Si() 2 , Petroleum ether/Ethyl acetate = 1/0 to 1/0). Compound l-bromo-5-fluoro-naphthalene (1.30 g, 4.45 mmol, 49% yield) was obtained as a yellow oil.

[0373] Compound 12-1 : To a mixture of benzyl (25 -2-(cyanomethyl)-4-[2-[[(25 r )-l- methylpyrrolidin-2-yl]methoxy]-6,7,8,9-tetrahydro-577-pyrimi do[4,5-c]azepin-4-yl]piperazine-l - carboxylate (500 mg, 962 pmol, 1.00 eq) and l-bromo-5-fluoro- naphthalene (433 mg, 1.92 mmol, 2.00 eq) in toluene (15.0 mL) was added Pd 2 (dba) 3 (176 mg, 192 pmol, 0.20 eq), RuPhos (180 mg, 385 pmol, 0.40 eq) and CS2CO3 (941 mg, 2.89 mmol, 3.00 eq) under N 2 . The mixture was degassed and purged with N 2 for 3 times, then heated to 90 °C and stirred for 8 hours. The reaction mixture was diluted with water (20.0 mL) and extracted with ethyl acetate (30.0 mL c 3). The combined organic layers were washed with brine (50.0 mL x 1), dried over sodium sulfate, filtered and concentrated under reduced pressure to give a residue. The residue was purified by reverse phase flash [water (0.1 % TA)/acetonitrile]. The desired fractions were collected and neutralized with saturated NaHC0 3 solution (12 mL), and then extracted with ethyl acetate (50.0 mL x 3). The separated organic layer was dried over sodium sulfate, filtered and concentrated under vacuum. Compound benzyl ( 2S)-2 - (cyanomethyl)-4-[8-(5-fluoro-l - naphthyl)-2-[[(25)-l -methylpyrrolidin-2-yl]methoxy]-5,6,7,9-tetrahydropyrimido[4 ,5-c]azepin- 4-yl]piperazine-l -carboxylate (480 mg, 723 pmol, 75% yield, 100% purity) was obtained as a yellow solid. LCMS [ESI, M+l ]: 664.

[0374] J H NMR (400MHz, chloroform-d) d = 7.80 (d, J= 8.4 Hz, 1H), 7.71 (d, J= 8.4 Hz, 1H), 7.46 - 7.33 (m, 6H), 7.30 (s, 1H), 7.17 (d, = 7.2 Hz, 1H), 7.14 - 7.07 (m, 1H), 5.25 - 5.17 (m, 2H), 4.70 (br s, 1H), 4.43 - 4.33 (m, 3H), 4.17 (d, J= 6.8 Hz, 1H), 3.87 (br d, J= 12.0 Hz, I II), 3.70 (br d, J= 12.8 Hz, 1H), 3.58 - 3.50 (m, 1H), 3.47 - 3.22 (m, 3H), 3.09 (br t, J= 7.6 Hz, 1H), 3.05 - 2.83 (m, 4H), 2.81 - 2.72 (m, 1H), 2.72 - 2.60 (m, 1H), 2.46 (s, 3H), 2.28 (dt, J= 7.2 , 9.2 Hz, 1H), 2.20 - 2.06 (m, 2II), 2.04 - 1.94 (m, 1H), 1.92 - 1.78 (m, 4H). LCMS [ESI, M+l]: 530.

[0375] Compound 12-2: To a solution of benzyl (2.S)-2-(cyanomethyl)-4-[8-(5-fluoro- l - naph thy l )-2-[[(2, S)- 1 -methy [pyrrol idin-2-yl]methoxy |-5, 6,7.9-tetrahydropyrimido [4, 5-0 |azcpin- 4-yl]piperazine-l -carboxylate (330 mg, 497 pmol, 1.00 eq) in methanol (6.00 mL) was added Pd/C (80.0 mg, 10% purity) and NH * McOH (3.00 mL, 20% purity) under N 2 . The suspension was degassed under vacuum and purged with H 2 several times. The mixture was stirred under H 2 (15 psi) at 25 °C for 1 hour. The reaction mixture was concentrated under reduced pressure to give a residue. 2-[(25)-4-[8-(5-fluoro-l-naphthyl)-2-[[(25)-l-methylpyrrolid in-2-yl]methoxyJ- 5,6,7,9-tetrahydropyrimido[4,5-c]azepin-4-yl]piperazin-2-yl] acetonitrile (262 mg, 490 pmol, 99% yield, 99% purity) was obtained as a yellow solid and used in the next step directly without further purification.

[0376] Example 12: To a mixture of 2-[(25)-4-[8-(5-fluoro-l -naphthyl)-2-[[(25 -l - methylpyrrolidin-2-yl]methoxy]-5,6,7,9-tetrahydropyrimido[4, 5-c]azepin-4-yl]piperazin-2- yl]acetonitrile ( 150 mg, 283 pmol, 1.00 eq) in dichloromethane (3.00 mL) was added TEA (143 mg, 1.42 mmol, 197 mE, 5.00 eq), prop-2-enoyl chloride (38.5 mg, 425 pmol, 34.6 pL, 1.50 eq) in portion at -40 °C under N 2 . The mixture was stirred at -40 °C for 30 min. The reaction mixture was quenched by adding water (3.00 mL) at -40 °C, and then extracted with dichloromethane (10.0 mL x 3). The combined organic layers were washed with brine (10.0 mL x 1), dried over sodium sulfate, filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (column: Xtimate Cl 8 150 x 25 mm x 5 pnpmobile phase: [water (0.05% ammonium hydroxide v/v)-ACN];B%: 53%-83%,10 min). Compound 2-[(25)-4-[8-(5- fluoro-l-naphthyl)-2-[[(25 -l-methylpyrrolidin-2-yl]methoxy]-5,6,7,9-tetrahydropyrimido [4,5- c]azepin-4-yl]-l-prop-2-enoyl-piperazin-2-yl]acetonitrile (34.2 mg, 58.5 pmol, 21% yield) was obtained as a white solid. LCMS [ESI, M+1J: 584.

[0377] ! H NMR (400MHz, chloroform-d) 5 = 7.81 (d, J= 8.0 Hz, 1H), 7.72 (d, ./ = 8.4 Hz, 1H), 7.44 (t, J= 7.6 Hz, 1H), 7.32 - 7.27 (m, 1H), 7.18 (d, J= 7.2 Hz, 1H), 7.14 - 7.07 (m, 1H), 6.69 - 6.53 (m, 1H), 6.40 (dd, J= 2.0, 16.8 Hz, 1H), 5.83 (br d, .7= 10.4 Hz, 1H), 5.10 (br s, 1H), 4.44 - 4.32 (m, 3H), 4.17 (dd, J = 6.8, 10.4 Hz, 1H), 3.93 (br d, ./= 14.0 Hz, 2H), 3.78 (br d, J= 12.0 Hz, 1H), 3.68 - 3.40 (m, 3H), 3.30 (dd, J = 3.6, 13.6 Hz, 1H), 3.18 - 2.86 (m, 5H), 2.80 (br s,

HI), 2.71 - 2.61 (m, HI), 2.46 (s, 311), 2.35 - 2.23 (m, 111), 2.22 - 1.97 (m, 3H), 1.92 - 1.72 (m, 3H).

EXAMPLE 13

2-[(2S)-4-[2-[[(2S)-l-methylpyrrolidin-2-yl]methoxy]-8-[2 -(trifluoromethyl)phenyl]-5,6,7,9- tetrahydropyrimido[4,5-c]azepin-4-yl]-l -prop-2-enoyl-piperazin-2-yl]acetonitrile

Ill

Example 13

[0378] Compound 13-1: To a mixture of benzyl (2S)-2-(cyanomethyl)-4-[2-[[(25)-l- methylpyrrolidin-2-yl]methoxy]-6,7,8,9-tetrahydro-577-pyrimi do[4,5-c]azepin-4-yl]piperazine-l- carboxylate (200 mg, 385 pmol, 1.0 eq), l-bromo-2-(trifluoromethyl)benzene (173 mg, 770 mihoΐ, 105 pL, 2.0 eq), Cs 2 C0 3 (376 mg, 1.15 mmol, 3.0 eq) and RuPhos (71.8 mg, 154 pmol, 0.4 eq) in toluene (6 L) was added Pdjfdbab (70.5 mg, 77.0 pmol, 0.2 eq) under N 2 . The suspension was degassed under vacuum and purged with N 2 several times. The mixture was stirred under N 2 at 110 °C for 13 hours. Water (15 mL) was added into the mixture. The mixture was diluted with EtOAc (10 mL) and extracted with EtOAc (2 x 15 mL). The combined organic layers were washed with brine (20 mL). The combined organic layers were dried over anhydrous Na 2 S0 4 , filtered and concentrated under vacuum. The residue was purified by reverse-phase flash [water (0.1 % FA)/acetonitrile] to give benzyl (2S)-2-(cyanomethyl)-4-[2-[[(2S)-l- methylpyrrolidin-2-yl]methoxy]-8- [2-(trifluoromethyl)phenyl]-5,6,7,9-tetrahydropyrimido[4,5- c]azepin-4-yl]piperazine-l -carboxylate (140 mg, 194 pmol, 50 % yield, 92 % purity) as a yellow solid. LCMS [ESI, M+l]: 664.

[0379] Ή NMR (400 MHz, chloroform-d) d = 7.63 - 7.58 (m, 1H), 7.55 - 7.49 (m, 1H), 7.42 -

7.33 (m, 611), 7.22 (t, J= 7.6 Hz, 1H), 5.26 - 5.16 (m, 2H), 4.70 (br s, 1H), 4.37 (br dd, J= 4.8, 10.4 Hz, 1H), 4.16 - 4.08 (m, 2H), 3.81 (br d, J= 10.0 Hz, III), 3.66 (br d, J= 12.4 Hz, 1H),

3.52 - 3.14 (m, 5H), 3.09 (br t, J= 7.6 Hz, 1H), 3.02 - 2.72 (m, 6H), 2.66 (br s, 1H), 2.47 (s, 3H),

2.33 - 2.22 (m, 1H), 2.05 - 1.91 (m, 3H), 1.89 - 1.74 (m, 3H)..

[0380] Compound 13-2: To a solution of benzyl (25 r )-2-(cyanomethyl)-4-[2-[[(25)-l- methylpyrrolidin-2-yl]methoxyj-8-[2-(trilluoromethyl)phenyl] -5,6,7,9-tetrahydropyrimido[4,5- c]azepin-4-yl]piperazine-l -carboxylate (140 mg, 21 1 pmol, 1.0 eq) and NTL'MeOH (2 mL, 15 % purity) was added Pd/C (60 mg, 10 % purity) under N 2. The suspension was degassed under vacuum and purged with H 2 several times. The mixture was stirred under Eh (15 psi) at 15 °C for 0.5 hour. The reaction mixture was filtered and the filtrate was concentrated under vacuum to give 2-[(25)-4-[2-[[(25')-l-methylpyrrolidin-2-yl]methoxy]-8-[2-( trifluoromethyl) phenyl]- 5,6,7,9-tetrahydropyrimido[4,5-c]azepin-4-yl]piperazin-2-yl] acetonitrile (75 mg, 127 pmol, 60 % yield, 90 % purity) as a yellow solid which was used for next step without further purification.

[0381] Example 13: To a solution of 2-[(250-4-[2-[[(25)-l-methylpyrrolidin-2-yl]methoxy]-8- [2-(trifluoromethyl)phenyl]-5,6,7,9-tetrahydropyrimido[4,5-c ]azepin-4-yl]piperazin-2- yl]acetonitrile (75 mg, 142 pmol, 1.0 eq ) and DIEA (36.6 g, 283 pmol, 49.3 uL, 2.0 eq) in DCM (1.5 mL) was added prop-2-enoyl chloride (19.2 mg, 212 pmol, 17.3 pL, 1.5 eq) at -40 °C. The reaction mixture was stirred at -40 °C for 0.5 hour. The reaction mixture was quenched by water (8 mL). The mixture was extracted with DCM (3 >< 10 mL). The combined organic layers were dried over anhydrous Na 2 S0 4 , filtered and concentrated under vacuum. The residue was purified by reverse-phase flash [water (0.1 % FA)/acetonitrile] The residue was purified by prep-HPLC (column: Waters Xbridge 150*25 5p;mobile phase: [water (0.05 % ammonium hydroxide v/v)-ACN];B %: 55 % - 73 %, 10 min) to give 2-[(2X)-4-[2-[[(25)-l-methylpyrrolidin- 2-yl]methoxy]-8-[2- (trifluoromethyl)phenyl]-5,6,7,9-tetrahydropyrimido[4,5-6]az epin-4-yl]-l- prop-2-enoyl-piperazin-2-yl]acetonitrile (10.3 mg, 17.4 pmol, 12 % yield, 98.8 % purity) as a white solid. LCMS [ESI, M+lj: 584.

[0382] *H NMR (400 MHz, chloroform-d) d = 7.61 (d, 7 = 7.6 Hz, 1H), 7.56 - 7.49 (m, 1H), 7.35 (d, 7= 8.0 Hz, 1H), 7.23 (t, 7= 7.6 Hz, 1 H), 6.58 (br d, ./= 10.0 Hz, 1H), 6.39 (dd, j= 1.6, 16.8 Hz, III), 5.82 (br d, J= 10.4 Hz, 1H), 5.13 (br s, 1H), 4.36 (dd, 7 = 4.8, 10.6 Hz, 1H), 4.22 (q, 7 = 17.2 Hz, 2H), 4.1 1 (dd, 7= 6.8, 10.4 Hz, 1H), 4.04 - 3.81 (m, 2H), 3.80 - 3.38 (m, 2H), 3.34 - 3.17 (m, 3H), 3.08 (br t, 7= 7.6 Hz, 1H), 3.04 - 2.72 (m, 5H), 2.71 - 2.60 (m, 1H), 2.46 (s, 3H), 2.32 - 2.22 (m, 1H), 2.12 - 1.90 (m, 3H), 1.89 - 1.72 (m, 3H).

EXAMPLE 14

2-[(25)-4-[8-[3-fluoro-2-(trifluoromethyl)phenyl]-2-[[(2X )-l -methylpyrrolidin-2-ylJmethoxy]-

5,6,7,9-tetrahydropyrimido[4,5-c]azepin-4-yl]-l-prop-2-en oyl-piperazin-2-yl]acetonitrile

[0383] Compound 14-1: To a solution of benzyl (2S)-2-(cyanomethyl)-4-[2-[[(2S)-l - methylpyrrolidin-2-yl]methoxy]-6,7,8,9-tetrahydro-5//-pyrimi do[4,5-c]azepin-4-yl]piperazine-l - carboxylate (250 mg, 481 pmol, 1.0 eq ), l-bromo-3-fluoro-2- (trifluoromethyl)benzene (234 mg, 962 pmol, 2.0 eq), RuPhos (89.8 mg, 192 pmol, 0.40 eq) and CS2CO3 (392 mg, 1.20 mmol, 2.50 eq) in toluene (3.0 mL) was added Pd 2 (dba) 3 (88.1 mg, 96.2 pmol, 0.20 eq). The mixture was stirred at 1 10 °C for 12 hours. After completion, the reaction mixture was added water (10.0 mL) and extracted with ethyl acetate (3 x 10.0 mL). The organic layer was dried over Na 2 S0 4 , filtered and concentrated. The residue was purified by reverse phase flash (Cl 8, 0.1% FA in water, 0-60 % MeCN) to give the compound benzyl (25 -2-(cyanomethyl)-4-[8-[3-fluoro-2- (trifluoromethyl)phenyl]-2-[[(25)-l -methylpyrrolidin-2-yl]methoxy]-5, 6,7,9- tetrahydropyrimido[4,5-c]azepin-4-yl]piperazine-l -carboxylate (130 mg, 190 pmol, 39% yield, 99% purity) as yellow solid. LCMS [ESI, M+l]: 682.

[0384] Ή NMR (400 MHz, chloroform-d) d 7.42 - 7.36 (m, 5H), 6.94 (d, J= 8.4 Hz, III), 6.88 - 6.81 (m, 1H), 5.20 (s, 2H), 4.74 - 4.62 (m, 1H), 4.40 - 4.35 (m, 1 H), 4.17 - 4.10 (m, 4H), 3.85 - 3.75 (m, 1H), 3.66 - 3.56 (m, 1H), 3.43 - 3.17 (m, 4H), 3.13 - 3.05 (m, 1H), 2.99 - 2.81 (m, 2H), 2.79 - 2.54 (m, 5H), 2.47 (s, 3H), 2.33 - 2.24 (m, 1H), 2.04 - 1.94 (m, 3H), 1.87 - 1.73 (m, 3H).

[0385] Compound 14-2: To a solution of benzyl (2 < S)-2-(cyanomethyl)-4-[8-[3-fluoro- 2- (tri fluoromethyl)phenyl]-2-[[(25 -l-methylpyrrolidin-2-yl]methoxy]-5, 6,7,9- tetrahydropyrimido[4,5-c]azepin-4-yl]piperazine-l-carboxylat e (130 mg, 191 pmol, 1.0 eq) in MeOII (2.0 mL) and NH MeOH (2.0 mL, 20% purity) was added Pd/C (40.0 mg, 10% purity). The mixture was stirred under ¾ (15 Psi) atmosphere at 15 °C for 0.5 hour. After completion, the reaction mixture was filtered and washed with tetrahydrofuran (20.0 mL). The filtrate was concentrated to give the compound 2- [(2A ' )-4- [8- [3 - P uoro-2-( tri ll uoromcthy l)pheny4 |-2- [ [ (2.S )- 1 - methylpyrrolidin-2-yl]methoxy]-5,6,7,9-tetrahydropyrimido[4, 5-c]azepin-4-yl]piperazin-2- yljacetonitrile (100 mg, 183 pmol, 96% yield) as yellow solid. The product was used for the next step without further purification. LCMS [ESI, M+l]: 548.

[0386] Example 14: To a solution of 2-[(2 1 S)-4-[8-[3-fluoro-2- (trifluoromethyl) phenyl]-2- [[(2X)-l -methylpyrrolidin-2-yl]methoxy]-5,6,7,9-tetrahydropyrimido[4 ,5-c]azepin-4- yl]piperazin-2-yl]acetonitrile (90.0 mg, 164 pmol, 1.0 eq) and DIEA (42.5 mg, 329 pmol, 57.3 pL, 2.0 eq) in dichloromethane (2.0 mL) was added prop-2-enoyl chloride (22.3 mg, 247 pmol, 20.1 pL, 1.50 eq) at -40 °C. The mixture was stirred at -40 °C for 0.5 hour. After completion, the reaction mixture was quenched with H 2 0 (2.50 mL) at -40 °C, then warmed to 15 °C and extracted with dichloromethane (2 >< 10 mL). The combined organic layers were dried over Na 2 S0 4 , filtered and concentrated under reduced pressure to give the residue. The residue was purified by prep-IIPLC (column: Xtimate Cl 8 150*25mm*5um; mobile phase: [water (0.05% ammonium hydroxide v/v)-ACN]; B%: 51%-81%, 10 min) to give the compound 2-[(2S -4-[8- [3-fluoro-2-(triiluoromethyl)phenyl]-2-[[(2S)-l -methylpyrrolidin-2-yl]methoxy]-5, 6,7,9- tetrahydropyrimido[4,5-c]azepin-4-yl]-l-prop-2-enoyl-piperaz in-2-yl]acetonitrile (30.2 mg, 50.1 pmol, 30% yield, 99% purity) as white solid. LCMS [ESI, M+1 J: 602.

[0387] 1H NMR (400 MIIz, Chloroform-d) d 7.40 - 7.33 (m, 1H), 6.95 (d, J = 8.4 Hz, 1 H), 6.89 - 6.82 (m, 1 H), 6.67 - 6.50 (m, 1H), 6.38 (dd, J = 1.6, 16.8 Hz, 1 H), 5.82 (br d, J = 10.8 Hz, HI),

5.20 - 4.90 (m, 1H), 4.42 - 4.26 (m, 3H), 4.18 - 4.1 1 (m, III), 4.08 - 3.77 (m, 2H), 3.75 - 3.50 (m,

2H), 3.42 - 3.21 (m, 3H), 3.14 - 3.05 (m, 1H), 3.03 - 2.87 (m, 2H), 2.81 - 2.62 (m, 4H), 2.48 (s,

3H), 2.33 - 2.24 (m, 1H), 2.10 - 1.94 (m, 3H), 1.88 - 1 .68 (m, 3H). EXAMPLE 15

2-[(2,S)-4-[2-[[(2S)-l-methylpyrrolidin-2-yl]methoxy]-8-[ 3-(trifluoromethyl)-2-pyridyl]-5,6,7,9- tetrahydropyrimido[4,5-c]azepin-4-yl]-l-prop-2-enoyl-piperaz in-2-yl]acetonitrilc

[0388] Compound 15-1: A mixture of benzyl (25)-2-(cyanomethyl)-4-[2-[[(25 -l- methylpyrrolidin-2-yl]methoxy]-6,7,8,9-tetrahydro-5i/-pyrimi do[4,5- < :]azepin-4-yl]piperazine-l - carboxylate (300 mg, 577 pmol, 1.0 eg) and 2-fluoro-3-(trifluoromethyl) pyridine (953 mg, 5.77 mmol, 10 eq) was heated to 120 °C and stirred for 14 hours. The mixture was diluted with EtOAc (10 mL) and CuS04 (4 %, 15 mL). Then the mixture was extracted with EtOAc (2 x 20 mL). The combined organic layers were washed with CuS04 (4 %, 3 x 15 mL) and brine (30 mL). The combined organic layers were dried over anhydrous Na 2 S0 4 , filtered and concentrated under vacuum. The residue was purified by reverse-phase flash [water (0.1% FA)/acetonitrile] to give benzyl (25)-2-(cyanomethyl)-4-[2-[[(25)-l-methylpyrrolidin-2-yl]met hoxy]-8-[3- (trifluoromethyl)-2-pyridyl]-5,6,7,9-tetrahydropyrimido[4,5- c]azepin-4-yl]piperazine-l- carboxylate (140 mg, 200 pmol, 35 % yield, 95 % purity) as a yellow solid. LCMS [ESI, M+l]: 665. [0389] 'H NMR (400 MHz, chloroform-d) d = 8.29 (dd , J= 1.6, 4.8 Hz, 1H), 7.83 (dd, J= 1.6, 7.6 Hz, 1H), 7.42 - 7.32 (m, 5H), 6.89 (dd, J= 4.8, 7.6 Hz, 1H), 5.24 - 5.13 (m, 2H), 4.65 (br s, 1H), 4.62 - 4.45 (m, 2H), 4.41 (dd, ./ = 4.8, 10.4 Hz, 1H), 4.16 - 4.00 (m, 2H), 3.75 (br d, J =

13.2 Hz, 1H), 3.67 - 3.51 (m, 3H), 3.27 (br s, 1H), 3.19 (dd, J= 3.6, 13.6 Hz, 1H), 3.1 1 (br t, J = 7.6 Hz, 1H), 2.88 (dt, J= 3.6, 12.4 Hz, 1H), 2.82 - 2.64 (m, 4H), 2.62 - 2.54 (m, 1H), 2.49 (s, 3H), 2.35 - 2.25 (m, 1H), 2.15 - 2.06 (m, 2H), 2.04 - 1.97 (m, 1H), 1.93 - 1.76 (m, 3H).

[0390] Compound 15-2: To the solution of benzyl (25)-2-(cyanomethyl)-4-[2-[[(25 -l- methylpyrro]idin-2-yl]methoxy]-8-[3-(trifluoromethyl)-2-pyri dyl]-5,6,7,9- tetrahydropyrimido[4,5-c]azepin-4-yl]piperazine-l-carboxylat e (140 mg, 21 1 pmol, 1 .0 eq) and NH 3 ·MeOH (1 mL, 20 % purity) was added Pd/C (50 mg, 10 % purity) under N 2 . The suspension was degassed under vacuum and purged with ¾ several times. The mixture was stirred under ¾ (15 psi) at 15 °C for 0.5 hour. The reaction mixture was filtered and the filter cake was washed with MeOH (3 c 8 mL). The filtrate was concentrated under vacuum to give 2- [(2A)-4-[2-[[(2S> l -methylpyrrolidin-2-yl] methoxy]-8-[3-(trifluoromethyl)-2-pyridyl]-5, 6,7,9- tetrahydropyrimido[4,5-c]azepin-4-yl]piperazin-2-yl]acetonit rile (45 mg, 76.3 pmol, 36 % yield, 90 % purity) as a yellow solid which was used for next step without further purification.

[0391] Example 15: To a solution of 2-[(2S * )-4-[2-[[(2X)-l-methylpyrro]idin-2-y]]methoxy]-8- [3-(trifluoromethyl)-2-pyridyl]-5,6,7,9-tetrahydropyrimido[4 ,5-c]azepin-4-yl]piperazin-2- yljacetonitrile (45 mg, 84.8 pmol, 1.0 eq) and DIEA (32.9 mg, 254 pmol, 44.3 pL, 3.0 eq) in DCM (1 mL) was added 2-oxoacetyl chloride (1 1.8 mg, 127 pmol, 1.5 eq) at -40 °C, and the mixture wus stirred at -40 °C for 0.5 hour. Water (8 mL) was added into the mixture. The mixture was diluted with EtOAc (8 mL) and extracted with EtOAc (2 x 10 mL). The combined organic layers were washed with brine (20 mL). The combined organic layers were dried over anhydrous Na 2 S0 4 , filtered and concentrated under vacuum. The residue was purified by reverse-phase flash [water (0.1 % FA)/acetonitrile] The residue was purified by prep-HPLC (column: Waters Xbridge 150*25 5p;mobile phase: [water (0.05 % ammonium hydroxide v/v)- ACN]; B %: 45 % - 69 %, 10 min) to give 2-[(2X)-4-[2-[[(2S -l-methy]pyrrolidin-2- yl]methoxy]-8-[3-(trifluoromethyl)-2- pyridyl]-5,6,7,9-tetrahydropyrimido[4,5-c]azepin-4-ylj-l- prop-2-enoyl-piperazin-2-yl]acetonitrile (10.9 mg, 18.5 pmol, 22 % yield, 99.7 % purity) as a white solid. LCMS [ESI, M+l ]: 585. [0392] (400 MHz, chloroform-d) d = 8.29 (dd, ,/ = 1 .6, 4.8 Hz, 1H), 7.84 (dd, J= 1.6, 7.6 Hz, III), 6.90 (dd, .7 = 4.8, 7.6 Hz, 1 H), 6.55 (br d , J= 10.4 Hz, I II), 6.37 (dd, J= 1.6, 16.8 Hz, 1 H), 5.80 (br d, J= 10.4 Hz, 1H), 5.07 (br s, 1 H), 4.63 - 4.46 (m, 2H), 4.39 (dd, J= 4.8, 10.8 Hz, 1H), 4.14 (dd, J = 7.2, 10.4 Hz, 1 H), 4.03 - 3.74 (m, 2H), 3.72 - 3.42 (m, 4H), 3.21 (dd, J = 3.6, 13.6 Hz, 1H), 3.09 (br t, J = 7.6 Hz, HI), 3.00 - 2.82 (m, 2H), 2.80 - 2.56 (m, 4H), 2.48 (s, 3H), 2.34 - 2.23 (m, 1H), 2.17 - 1.99 (m, 3H), 1 .90 - 1.76 (m, 3H).

EXAMPLE 16

2- 1 ( 2,S , )-4-[S- [2-(hy d roxyniethy 1) phenyl] -2- 1 | ( 2.S ' }- 1 -methy lpyrrolidin-2-yl]methoxy]-5,6,7.9- tetrahydropyrimido[4,5-c]azepin-4-yl]-l -prop-2-enoyl-piperazin-2-yl]acetonitrile

[0393] Compound 16-B: To a solution of (2-bromophenyl)methanol (1 g, 5.35 mmol, 1.0 eq) and TSOH.H 2 0 (102 mg, 535 pmol, 0.1 eq) in DCM (20 ml.) was added DHP (899 mg, 10.7 mmol, 978 pL, 2.0 eq), the mixture was stirred at 1 8 °C for 14 hours. To the mixture was added saturated NaHCCb (20 mL). The mixture was extracted with DCM (2 x 15 mL). The combined organic layers were dried over anhydrous Na 2 S0 4 , filtered and concentrated under vacuum. The residue was purified by silica gel chromatography (Petroleum ether: Ethyl acetate = 150:1) to give 2-[(2- bromophenyl)methoxy] tetrahydropyran (1.15 g, 4.03 mmol, 75 % yield, 95 % purity) as a colorless oil.

[0394] 'H NMR (400 MHz, chloroform-d) d = 7.57 - 7.50 (m, 2H), 7.33 (dt, J= 1.2, 7.6 Hz,

1H), 7.18 - 7.12 (m, 1H), 4.84 (d, j= 13.2 Hz, 1H), 4.79 (t, .7 = 3.6 Hz, 1H), 4.59 (d, .7- 13.2 Hz, 1H), 3.99 - 3.90 (m, 1H), 3.63 - 3.53 (m, 1H), 1.98 - 1.85 (m, 1H), 1.84 - 1.59 (m, 4H), 1.58 - 1.53 (m, 1H).

[0395] Compound 16-1: To a mixture of benzyl (25 -2-(cyanomelhyl)-4-[2-[[(25 -l- methylpyrrolidin-2-yl]methoxy]-6,7,8,9-tetrahydro-577-pyrimi do[4,5-c]azepin-4-yl]piperazine-l- carboxylate (400 mg, 770 pmol, 1.0 eq), 2-[(2-bromophenyl)methoxy]tetrahydropyran (313 mg, 1.15 mmol, 1.5 eq), Cs 2 C0 3 (752 mg, 2.31 mmol, 3.0 eq) and RuPhos (144 mg, 308 pmol, 0.4 eq) in toluene (8 mL) was added Pd 2 (dba) 3 (141 mg, 154 pmol, 0.2 eq) under N 2 . The suspension was degassed under vacuum and purged with N 2 several times. The mixture was stirred under N 2 at 90 °C for 14 hours. Water (20 mL) was added into the mixture. The mixture was diluted with EtOAc (10 mL) and extracted with EtOAc (2 x 20 mL). The combined organic layers were washed with brine (20 mL). The combined organic layers were dried over anhydrous Na 2 S0 4 , filtered and concentrated under vacuum. The residue was purified by reverse-phase flash [water (0.1% FA)/acetonitrile] to give benzyl (25 -2-(cyanomethyl)-4-[2-[[(25 -l-methylpyrrolidin- 2- yl]methoxy]-8-[2-(tetrahydropyran-2-yloxymethyl)phenyl]-5,6, 7,9-tetrahydropyrimido[4,5- c]azepin-4-yl]piperazine-l-carboxylate (360 mg, 482 p ol, 62 % yield, 95 % purity) as a yellow solid. LCMS [ESI, M+l]: 710.

[0396] Ή NMR (400 MHz, chloroform-d) d = 7.46 (dd, j= 2.8, 7.6 Hz, 1TI), 7.42 - 7.32 (m,

5H), 7.26 - 7.20 (m, 1 H), 7.1 1 - 7.05 (m, 2H), 5.27 - 5.16 (m, 2TI), 4.67 (br dd, .7= 4.4, 12.4 Hz, 2H), 4.54 - 4.41 (m, 2H), 4.37 (ddd, j= 2.0, 4.8, 10.4 Hz, 1H), 4.28 - 4.15 (m, 2H), 4.12 - 4.07 (m, 1H), 3.88 - 3.74 (m, 2H), 3.65 (br d, ,7- 12.4 Hz, 1H), 3.45 - 3.16 (m, 5H), 3.08 (br t, j= 7.2 Hz, 1H), 2.99 - 2.71 (m, 5H), 2.69 - 2.60 (m, 1 H), 2.46 (s, 3H), 2.27 (dt, .7= 7.2, 9.2 Hz, 1H), 2.04 - 1.89 (m, 2H), 1.88 - 1.69 (m, 5H), 1.66 - 1.43 (m, 5H).

[0397] Compound 16-2: To a solution ofbcnzyl (25 2-(cyanomethyl)-4-[2-[[(25')-l- methylpyrrolidin-2-yl]methoxy]-8-[2-(tetrahydiOpyran-2-yloxy methyl)phenyl]-5, 6,7,9- tetrahydropyrimido[4,5-c]azepin-4-yl]piperazine-l-carboxyiat e (200 mg, 282 pmol, 1.0 eq) and NHh ' MeOH (2 mL, 20 % purity) was added Pd/C (50 mg, 10 % purity) under N 2 . The suspension was degassed under vacuum and purged with Th several times. The mixture was stirred under II 2 (15 psi) at 15 °C for 0.5 hour. The reaction mixture was filtered and the filter cake was washed with MeOH (3 x 8 mL). The filtrate was concentrated under vacuum to give 2- [(2S)-4 - [2- [ [(25)- 1 -methylpyrrolidin-2 -y ljmethoxy] -8 - [2-(tetrahydropyran-2 - yloxymethyl)phenyl]-5,6,7,9-tetrahydropyrimido[4,5-c]azepin- 4-yl]piperazin-2-yl]acetonitrile (160 mg, 250 pmol, 89 % yield, 90 % purity) as a yellow solid which was used for next step without further purification.

[0398] Compound 16-3: To a solution of 2-[(25)-4-[2-[[(25)-l-methylpyrrolidin-2-yl]methoxy]- 8-[2-(tetrahydropyran-2-yloxymethyl)phenyl]-5,6,7,9-tetrahyd ropyrimido[4,5-c]azepin-4- yl]piperazin-2-yl]acetonitrile (160 mg, 278 pmol, 1.0 eq) and DIEA (71.8 mg, 556 pmol, 96.8 pL, 2.0 eq) in DCM (3 mL) was added prop-2-enoyl chloride (37.7 mg, 417 pmol, 34.0 pL, 1 .5 eq) at -40 °C, the mixture was stirred at -40 °C for 0.5 hour. The reaction mixture was quenched with water (10 mL). The mixture was diluted with EtOAc (10 mL) and extracted with EtOAc (2 x 15 mL). The combined organic layers were washed with brine (20 mL). The combined organic layers were dried over anhydrous Na 2 S0 4 , filtered and concentrated under vacuum. The residue was purified by reverse-phase flash [water (0.1% FA)/acetonitrile] to give 2-[(25)-4-[2-[[(25)-l- methylpyrrolidin-2- yl]methoxy]-8-[2-(tetrahydropyran-2-yloxymethyl)phenyl]-5,6, 7,9- tetrahydropyrimido[4,5-c]azepin-4-yl]-l-prop-2-enoyl-piperaz in-2-yl]acetonitrile (120 mg,

181.01 pmol, 65.14% yield, 95% purity) as a yellow solid. LCMS [ESI, M+l]: 630.

[0399] Example 16: To the solution of 2-[(25)-4-[2-[[(25)-l-methylpyrrolidin-2-yl]methoxy]-8- [2-(tetrahydropyran-2-yloxymethyl)phenyl]-5,6,7,9-tetrahydro pyrimido[4,5-c]azepin-4-yl]-l- prop-2-enoyl-piperazin-2-yl]acetonitrile (100 mg, 159 pmol, 1.0 eq) in DCM (0.1 mL) was added TFA (308 mg, 2.70 mmol, 0.2 mL, 17 eq), the mixture was stirred at 15 °C for 1 hour. The reaction mixture was concentrated under vacuum. The residue was diluted with EtOAc (10 mL) and saturated NalTC03 (10 mL). Then the mixture was extracted with EtOAc (2 x 15 mL). The combined organic layers were washed with brine (15 mL). The combined organic layers were dried over anhydrous Na 2 S0 4 , filtered and concentrated under vacuum. The residue was purified by reverse-phase flash [water (0.1 % FA)/acetonitrile] The residue was purified by prep-HPLC (column: Xtimate C18 150*25mm*5pm;mobile phase: [water (0.05 % ammonium hydroxide v/v)-ACN];B %: 32 % - 62 %,l0 min) to give 2-[(2S)-4-[8-[2-(hydroxymethyl) phenyl]-2-[[(25)- l-methylpyrrolidin-2-yl]methoxy]-5,6,7,9-tetrahydropyrimido[ 4,5-c]azepin-4-yl]-l-prop-2- enoyl-piperazin-2-yl]acetonitrile (12.0 mg, 22.0 pmol, 14 % yield, 99.7 % purity) as a white solid. LCMS [ESI, M+l]: 546.

[0400] Ή NMR (400 MEIz, chloroform-d) d = 7.32 (dd, J -1.2, 7.6 Hz, 1H), 7.30 - 7.27 (m, 1H), 7.18 (d, J= 7.2 Hz, 1H), 7.10 (dt, J= 1.2, 7.6 Hz, 1H), 6.66 - 6.52 (m, III), 6.39 (dd, J= 1.6,

16.8 Hz, 1H), 5.82 (br d, J= 11.6 Hz, 1H), 5.07 (br s, HI), 4.65 (d, J= 1.2 Hz, 2H), 4.37 (dd, J= 5.2, 10.4 Hz, 1H), 4.29 - 4.12 (m, 3H), 3.87 (br d , J= 13.6 Hz, 2H), 3.82 - 3.46 (m, 2H), 3.44 - 3.23 (m, 3H), 3.14 - 2.98 (m, 2H), 2.97 - 2.89 (m, 1H), 2.88 - 2.79 (m, 2H), 2.78 - 2.60 (m, 2H), 2.47 (s, 3H), 2.28 (dt, J= 7.2, 9.2 Hz, 1H), 2.13 - 1.92 (m, 3H), 1.90 - 1.75 (m, 3H).

EXAMPLE 17

4-[(3 S)-3-(cyanomethyl)piperazin-l-yl]-7-(3-hydroxy-l-naphthyl)-2 -[[(2S)-l-methylpyrrolidin-

2-yl]methoxy]-6,8-dihydro-5//-l,7-naphthyridine-3-carboni trile

[0401] Compound 17-B: To a mixture of 8-bromoisoquinoline (1.00 g, 4.81 mmol, 1.00 eq) in dichloromethane (10.0 mL) was added m-CPBA (1.17 g, 5.77 mmol, 85% purity, 1.20 eq) at 0 °C. After stirring at 0 °C for 0.5 h and 20 °C for 1 hour, the mixture was washed with saturated Na 2 C0 3 (3 x 10.0 mL) and brine (1 x 10.0 mL), dried over Na 2 S0 4 , filtered and concentrated under vacuum to give 8-bromo-2-oxido-isoquinolin-2-ium (1.20 g, crude) as a yellow solid and used into next step without further purification. LCMS [ESI, M+l]: 226.

[0402] Compound 17-C: A mixture of 8-bromo-2-oxido-isoquinolin-2-ium (2.20 g, crude) in POCI 3 (20.0 g, 130 mmol, 12.1 mL) was stirred at 100 °C for 2 hours. The mixture was concentrated under vacuum. The residue was diluted with EA (10.0 mL) and adjusted pH > 7 by saturated Na 2 C(L. The organic layer was washed brine (1 x 5.00 mL), dried over Na 2 S0 4 , filtered and concentrated under vacuum. The residue was purified by column chromatography (Si0 2 , PE/EA=5/1) to give 8-bromo-l-chloro-isoquinoline (550 mg, 2.22 mmol, 23 yield, 98% purity) as a yellow solid. LCMS [ESI, M+l]: 244.

[0403] Ή NMR (400MHz, chloroform-d) d = 8.35 - 8.27 (m, 1H), 8.09 - 7.99 (m, 1H), 7.80 (br d, .7=8.4 Hz, 1 H), 7.64 - 7.58 (m, 1H), 7.53 - 7.44 (m, 1H).

[0404] Compound 17-1: A mixture of benzyl (25)-2-(cyanomethyl)-4-[2-[[(25 -l - methylpyrrolidin-2-yl]methoxy]-6,7,8,9-tetrahydro-5/7-pyrimi do[4,5-c]azepin-4-yl]piperazine-l- carboxylate (400 mg, 770 pmol, 1.00 eq), 8-bromo-l-chloro-isoquinoline (224 mg, 924 pmol,

1.2 eq), KF (89.4 mg, 1.54 mmol, 36.1 pL, 2.00 eq) in DMSO (4.00 mL) was stirred at 100 °C for 12 hours. The mixture was diluted with ethyl acetate (10.0 mL), washed with brine (3 c 10.0 mL), the organic layer was dried over Na 2 S0 4 , filtered and concentrated under vacuum. The residue was purified by reverse phase flash [water (FA, 0.1 %)/acetonitrile]. The desired fraction was collected and treated with NaHCCL (3.00 g). The mixture was concentrated under vacuum to removed acetonitrile. The mixture was extracted with ethyl acetate (3 xlO.O mL), the organic layers were washed brine (1 c 10.0 mL), dried over Na 2 S0 4 , filtered and concentrated under vacuum to give benzyl (25')-4-[8-(8-bromo-l-isoquinolyl)-2- [[(2S)-l-methylpyrrolidin-2- yl]methoxy]-5,6,7,9-tetrahydropyrimido[4,5-c]azepin-4-yl]-2- (cyanomethyl)piperazine-l- carboxylate (240 mg, 278 pmol, 36% yield, 84% purity) as a yellow solid. LCMS [ESI, M+l]: 725. [0405] ! II NMR (400MHz, chloroform-d) 5 - 7.92 - 7.76 (m, 1H), 7.71 (d, J= 7.6 Hz, 1H), 7.65

- 7.57 (t, J= 8.8 Hz 1H), 7.41 - 7.30 (m, 6H), 7.09 - 6.97 (dt, J= 5.2 Hz, 7= 30.4 Hz, 1H), 5.24 - 5.14 (m, 2H), 4.92 - 4.69 (m, HI), 4.66 (br s, 1H), 4.58 - 4.49 (m, 1H), 4.40 - 4.30 (m, 1H), 4.08

- 3.45 (m, 511), 3.41 - 3.03 (m, 4H), 3.00 - 2.82 (m, 1H), 2.81 - 2.50 (m, 5H), 2.48 - 2.43 (d, J = 8.4 Hz, 3H), 2.33 - 2.22 (m, 1H), 2.12 - 2.06 (m, 1H), 2.03 - 1.93 (m, 2H), 1.89 - 1.75 (m, 3H).

[0406] Compound 17-2: A mixture of benzyl (2S)-4-[8-(8-bromo-l-isoquinolyl)-2-[[(2S)-l- methylpyrrolidin-2-yl]methoxy]-5,6,7,9-tetrahydropyrimido[4, 5-c]azepin-4-yl]-2- (cyanomethyl)piperazine-l-carboxylate (0.10 g, 138 pmol, 1.00 eq), methylboronic acid (165 mg, 2.76 mmol, 11.6 pL, 20.0 eq), Pd(PPh 3 ) 4 (15.9 mg, 13.8 pmol, 0.10 eq) and K3PO4 (87.8 mg, 413 pmol, 3.00 eq) in DMF (3.00 mL) was stirred at 110 °C for 10 h under N 2 . The mixture was diluted with ethyl acetate (5.00 mL), washed with brine (3 c 3.00 mL). The combined organic layer dried over Na 2 S0 4 , filtered and concentrated under vacuum. The residue was purified by reverse phase flash [water (FA, 0.1 %)/acetonitrile]. The desired fraction was collected and solid NaHC0 3 (1.00 g) was added. The mixture was concentrated under vacuum to remove acetonitrile. The residue was extracted with ethyl acetate (3 x5.00 mL), the organic layers were washed brine (1 x 5.00 mL), dried over Na 2 SOi, filtered and concentrated under vacuum to give benzyl (25)-2-(cyanomethyl)-4-[8-(8-methyl-l-isoquinolyl)-2-[[(25 )-l- methylpyrrolidin-2-yl]methoxy]-5,6,7,9-tetrahydropyrimido[4, 5-c]azepin-4-yl]piperazine-l- carboxylate (60.0 mg, 64.5 pmol, 47% yield, 71% purity) as a yellow oil. LCMS [ES1, M+l j: 661.

[0407] Compound 17-3: A mixture of benzyl (25 -2-(cyanomethyl)-4-[8-(8-methyl-l- isoquinolyl)-2-[[(25)-l-methylpyrrolidin-2-yl]methoxy]-5,6,7 ,9-tetrahydropyrimido[4,5- c]azepin-4-yl]pipcrazine-l-carboxylate (0.05 g, 75.7 pmol, 1.0 eq) and Pd/C (5.0 mg, 10% purity) in NH:, * MeOH (1.0 mL, 20% purity) and methanol (1.0 mL) was stirred at 25 °C for 1 hour under II 2 at 15 psi. The mixture was filtered and concentrated under vacuum. The residue was purified by prep-HPLC (column: Waters Xbridge 150*25 5p; mobile phase: [water (0.05% ammonium hydroxide v/v) - ACN]; B%: 40% - 64%, 10 min). The desired fractions were collected. The mixture was concentrated under vacuum to remove acetonitrile. The residue was lyophilized to give 2-[(25)-4-[8-(8-methyl-l -isoquinolyl)-2-[[(25 r )-l -methylpyrrolidin -2- yl]methoxy]-5,6,7,9-tetrahydropyrimido[4,5-c]azepin-4-yl]pip erazin-2-yl]acetonitrile (18.7 mg, 35.5 pmol, 47% yield, 100% purity) as a white solid. LCMS [ESI, M+l]: 527.

[0408] Ή NMR (400MHz, CHLOROFORM-d) 5 = 7.92 (dd, .7=5.6, 6.4 Hz, 1H), 7.56 - 7.51 (d, J=8.0 Hz, 1H), 7.44 (t, J=8.0 Hz, 1FI), 7.25 (m, 1H), 7.16 (t, =5.6 Hz, 1H), 4.60 - 4.43 (m, 2H), 4.37 (m, 1H), 4.12 (dd, .7=6.8, 10.4 Hz, 1H), 3.72 - 3.62 (m, 2H), 3.62 - 3.53 (m, 1H), 3.49 - 3.37 (m, 1FI), 3.32 - 3.19 (m, 1H), 3.13 - 2.76 (m, 6H), 2.74 (d, J=2.0 Hz, 3H), 2.72 - 2.58 (m, 2H), 2.51 (d, J=6.4 Hz, 2H), 2.45 (d, J=1.6 Hz, 3H), 2.43 - 2.32 (m, 1H), 2.31 - 2.22 (m, 1H), 2.10 - 1.90 (m, 5H).

[0409] Example 17: To a solution of 2-[(25')-4-[8-(8-methyl-l-isoquinolyl)-2-[[(2S)-l- methylpyrrolidin-2-yl]methoxy]-5,6,7,9-tetrahydropyrimido[4, 5-c]azepin-4-yl]piperazin-2- yljacetonitrile (50.0 mg, 94.9 pmol, 1.0 eq) and TEA (38.4 mg, 380 pmol, 52.9 pL, 4.0 eq) in dichloromethane (1.0 mL) was added prop-2-enoyl chloride (8.59 mg, 94.9 pmol, 7.74 pL, 1.0 eq) at -40 °C. After stirring at -40 °C for 0.5 h, the mixture was diluted with water (3.00 mL) dichloromethane (3.00 mL), the mixture was separated. The organic layer was dried over Na 2 Ot, filtered and concentrated under vacuum. The residue was purified by prep-HPLC (column: Waters Xbridge 150*25 5p; mobile phase: [water (0.05% ammonium hydroxide v/v) - ACN]; B%: 40% - 67%, lOmin). The desired fractions were collected and concentrated under vacuum to remove acetonitrile. The residue was lyophilized to give 2-[(2 > S , )-4-[8-(8-methyl-l- isoquinolyl)-2-[[(25 -l-methylpyrrolidin-2-yl]methoxy]-5,6,7,9-tetrahydropyrimido [4,5- c]azepin-4-yl]-l-prop-2-enoyl-piperazin-2-yl]acetonitrile (15.3 mg, 25.5 pmol, 27% yield, 97% purity) as white solid. LCMS [ESI, M+l]: 581 .

[0410] ] H NMR (400MHz, CHLOROFORM-d) d = 7.96 (dd, ,7=5.2 Hz, J=48.4 Hz, III), 7.57 - 7.51 (t, 7=4.0 Hz, 1H), 7.45 (t, .7=7.6 Hz, 1H), 7.28 - 7.25 (m, 1H), 7.16 (dd, 7=5.6, .7=31.2 Hz, 1H), 6.57 (m, 1H), 6.42 - 6.33 (m, 1H), 5.81 (m, 1H), 5.09 (br s, 1H), 4.71 - 4.54 (m, 1H), 4.51 - 4.48 (m, 1H), 4.36 (m, 1 H), 4.14 (m, 1H), 3.86 (br s, HI), 3.78 - 3.50 (m, 5H), 3.21 (m, 1H), 3.09 (m, 211), 2.92 - 2.81 (m, 2H), 2.79 - 2.71 (m, 4H), 2.69 - 2.51 (m, 2FI), 2.46 (d, J= 5.6 Hz, 3H), 2.34 - 2.21 (m, 2H), 2.15 - 1.92 (m, 3H), 1.90 - 1.82 (m, 2H).

EXAMPLE 18 2-[(25}-4-[8-(2-fluoro-6-methyl-phcnyl)-2-[[(25)-l-methylpyr rolidin-2-yl]methoxy]-5,6,7,9- tetrahydropyrimido[4,5-c]azepin-4-yl]-l -prop-2 -enoyl-piperazin-2-yl]acetonitrile

Example 18

[0411] Example 18 was prepared from compound Example 1-8.

[0412] Compound 18-1: To a solution of benzyl (2,S , )-2-(cyanomethyl)-4-[2-[[(2S')-l -methyl pyrrolidin-2-yl]methoxy]-6,7,8,9-tetrahydro-5//-pyrimido[4,5 -c]azepin-4-yl]piperazine-l- carboxylate (3.0 g, 5.77 mmol, 1.0 eq ) and l ,2-difluoro-3-nitro-benzene (1.29 g, 8.08 mmol, 1.4 eq) in MeCN (60 mL)was added DIEA (1.49 g, 1 1.6 mmol, 2.01 mL, 2.0 eq) and the reaction was stiiTcd for 12 hours at 80 °C. Upon completion, the mixture was concentrated under vacuum, diluted with water (40 mL) and extracted with Ethyl acetate (2 x 60 mL). The organic layers were dried over Na 2 S04 and concentrated under vacuum. The residue was purified by reversed phase flash [water (0.1% FA)/acetonitrile] The desired fractions were collected and neutralized with solid NallCCb, concentrated under vacuum to remove MeCN and extracted with ethyl acetate (2 c 80 mL). The organic layers were dried over Na 2 S0 4 and concentrated under vacuum to give benzyl (2S -2-(cyanomethyl)-4-[8-(2-fluoro-6-nitro- phenyl)-2-[[(25’)-l-methylpyrrolidin- 2-yl]methoxy]-5,6,7,9-tetrahydropyrimido[4,5-c]azepin-4-yl]p iperazine-l -carboxylate (2.3 g, 3.42 mmol, 59% yield, 98% purity) as a yellow solid.

[0413] ‘H NMR (400MHz, chloroform-d) 5 = 7.48 - 7.32 (m, 6H), 7.26 - 7.22 (m, 1H), 7.19 - 7.1 1 (m, 1 H), 5.25 - 5.15 (m, 211), 4.68 (br s, 1H), 4.40 - 4.30 (m, 2H), 4.28 - 4.18 (m, 1 H), 4.18

- 4.05 (m, 2H), 3.79 (br d, =12.0 Hz, 1H), 3.67 (br d, J= 1 1.6 IIz, III), 3.42 (td, J=5.2, 1 1.2 Hz, 1H), 3.37 - 3.18 (m, 3H), 3.08 (br t, =7.2 Hz, 1H), 2.99 - 2.79 (m, 2H), 2.79 - 2.69 (m, 3H), 2.68

- 2.60 (m, 1H), 2.46 (s, 3H), 2.27 (dt, .7=7.2, 9.2 Hz, 1H), 2.10 - 1 .92 (m, 3H), 1 .89 - 1.63 (m,

3H).

[0414] Compound 18-2: To a solution of benzyl (25)-2-(cyanomethyl)-4-[8-(2-fluoro-6-nitro- phenyl)-2-[[(25)-l-methylpyrrolidin-2-yl]methoxy]-5,6,7,9-te trahydropyrimido[4,5-c]azepin-4- yl]piperazine-l-carboxylate (2.0 g, 3.04 mmol, 1.0 eq), (Boc) 2 0 (1.33 g, 6.07 mmol, 1.40 mL, 2.0 eq) in MeOH (40 mL) was added Pd/C (1.0 g, 10% purity) under N 2 . The suspension was degassed under vacuum and purged with H 2 several times. The mixture was stirred under H 2 (15 psi) at 25 °C for 12 hours. Upon completion, the catalyst was removed by filtering through a plug of celite. The solvent was removed under reduced pressure to give tert- butyl ( 2S)-4-[&-(2 - amino-6-fluoro-phenyl)-2-[[(25 , )-l-methylpyrrolidin-2-yl]methoxy]-5,6,7,9- tetrahydropyrimido[4,5-c]azepin-4-yl]-2-(cyanomethyl)piperaz ine-l -carboxylate (1.55 g, 1.82 mmol, 60% yield, 70% purity) as a yellow solid which was used directly in the next step without further purification.

[0415] Compound 18-3: tert- butyl (25)-4-[8-(2-amino-6-fluoro-phenyl)-2-[[(25)-l-methyl pyrrolidin-2-yl]methoxy]-5,6,7,9-tetrahydropyrimido[4,5-c]az epin-4-yl]-2- (cyanomethyl)piperazine-l -carboxylate (1 .45 g, 2.44 mmol, 1.0 eq) was dissloved in MeCN (14.5 mL) and H 2 0 (7.25 mL) followed by the addition of TsOH*H 2 0 (1.86 g, 9.75 mmol, 4.0 eq). A solution of KI (1.21 g, 7.31 mmol, 3.0 eq) and NaN0 2 (336 mg, 4.88 mmol, 2.0 eq) in H 2 0 (3 mL) was added to the reaction mixture of slowly at 0 °C. The mixture was stirred at 0 °C for 1 hour. Upon completion, the mixture was concentrated under vacuum to remove acetonitrile, added water (10 mL) and extracted with Ethyl acetate (2 x 30 mL). Combined organic layers were dried over Na 2 S0 4 , filtered and concentrated under vacuum. The residue was purified by silica gel chromatography (Ethyl acetate/Methanol 50/1 to 5/1) followed by prep-HPLC

(column: Phenomenex luna Cl 8 250*50mm* 10 um; mobile phase: [water (0.225%FA)-ACN]; B%: 30%-60%, 23MIN; 30%min). The desired fractions were collected and neutralized with solid NaHCCfi, concentrated under vacuum to remove MeCN and extracted with ethyl acetate (2 x 50 mL). The organic layers were dried over Na 2 S0 4 and concentrated under vacuum to give /er/-butyl(25)-2-(cyanomethyl)-4-[8-(2-fluoro-6-iodo-phenyl) -2-[[(25)-l -methyl pyrrolidin-2- yl]methoxy]-5,6,7,9-tetrahydropyrimido[4,5-c]azepin-4-yl]pip erazine-l -carboxylate (420 mg, 587 pmol, 24% yield, 98.6% purity) as a yellow solid.

[0416] Compound 18-4: A mixture of tert- butyl (25)-2-(cyanomethyl)-4-[8-(2-fluoro-6-iodo- phenyl)-2-[[(25)-l-methylpyrrolidin-2-yl]methoxy]-5,6,7,9-te trahydropyrimido[4,5-c]azepin-4- yl]piperazine-l -carboxylate (320 mg, 454 pmol, 1.0 eq ), methylboronic acid (543 mg, 9.07 mmol, 20.0 eq ), Pd(PPh 3 ) 4 (52.4 mg, 45.4 pmol, 0.1 eq) and K 3 P0 4 (289 mg, 1.36 mmol, 3.0 eq) in DMF (6 mL) was stirred at 90 °C for 10 hours under N 2 . Upon completion, the mixture was diluted with ethyl acetate (10 mL) and extracted with water (3 x 5 mL). The organic layers were dried over Na 2 S0 4 and concentrated under vacuum. The residue was purified by reversed phase flash [water (0.1% NH H 2 0)/acetonitrile]. The desired fractions were collected and

concentrated under vacuum. The residue was further purified by prep-HPLC (column: Xtimate Cl 8 150*25mm*5um; mobile phase: [water (0.05% ammonium hydroxide v/v)-ACN];

B%:68%-98%). The desired fractions were collected and concentrated under vacuum to remove MeCN, extracted with ethyl acetate (2 x 10 mL). The organic layers were dried over Na 2 S0 4 and concentrated under vacuum to give tert- butyl (25)-2-(cyanomethyl)-4- [8-(2-fluoro-6-methyl- phenyl)-2-[[(25 -l-methylpyrrolidin-2-yl]methoxy]-5,6,7,9-tetrahydropyrimido [4,5-c]azepin-4- yl]piperazine-l -carboxylate (115 mg, 194 pmol, 43% yield, 100% purity) as a yellow oil.

[0417] ] H NMR (400MHz, chloroform-d) 6 = 7.03 - 6.95 (m, 1 H), 6.94 - 6.85 (m, 2H), 4.62 (br s, 1 H), 4.38 - 4.23 (m, 2H), 4.22 - 3.95 (m, 3H), 3.77 (br d, ,7=13.6 Hz, 1H), 3.62 (br d, J=12.8 Hz, 1H), 3.45 - 3.12 (m, 4H), 3.08 (br t, .7=7.2 Hz, 1H), 2.98 - 2.71 (m, 5H), 2.64 (td, .7=6.4, 13.2 Hz, 1H), 2.45 (s, 3H), 2.33 - 2.24 (m, III), 2.23 (s, 3H), 2.04 - 1.90 (m, 3FI), 1.88 - 1.76 (m, 3H), 1.52 (s, 9H). [0418] Compound 18-5: To a solution of /er/-butyl (2S -2-(cyanomethyl)-4-[8-(2-fluoro-6- methyl-phenyl)-2-[[(2S)-l-methylpyrrolidin-2-yl]methoxy]-5,6 ,7,9-tetrahydropyrimido[4,5- c]azepin-4-yl] piperazine- 1-carboxy late (30.0 mg, 50.5 pmol, 1.0 eq) in dichloromethane (0.03 mL) was added TFA (86.4 mg, 758 pmol, 56.1 pL, 15.0 eq). The mixture was stirred at 25 °C for 1 hour. Upon completion, the mixture was diluted with dichloromethane (1 mL) and neutralized with saturated NaHC0 3 solution at 0 °C. The separated aqueous layer was extracted with dichloromethane (3 x 2 mL). Combined organic layers were dried over Na 2 S0 4 and concentrated under vacuum. The residue was purified by prep-HPLC (column: Xtimate Cl 8 l50*25mm*5pm; mobile phase: [water (0.05% ammonium hydroxide v/v)-ACN]; B%: 49%-79%, lmin). The desired fractions were collected and lyophilized to give 2-[(2S -4-[8-(2-fluoro-6-methyl-phenyl)- 2-[[(2S0-l-methylpyrrolidin-2-yl]methoxy]-5,6,7,9-tetrahydro pyrimido[4,5-c]azepin-4- yl]piperazin-2-yl]acetonitrile (8.18 mg, 16.5 pmol, 33% yield, 99.8% purity) as a yellow solid. LCMS [ESI, M+l]: 494.

[0419] Ή NMR (400MHz, chloroform-d) d = 7.02 - 6.95 (m, 1H), 6.94 - 6.84 (m, 2H), 4.46 - 4.03 (m, 4H), 3.74 (br d, .7=12.4 Hz, 1H), 3.56 (br d, J=9.6 Hz, 1H), 3.28 (br s, 3H), 3.15 - 2.97 (m, 4H), 2.91 - 2.73 (m, 3H), 2.69 - 2.59 (m, 1H), 2.54 (d, J=6.8 Hz, 2LI), 2.45 (s, 3H), 2.30 - 2.24 (m, 1H), 2.23 (s, 3H), 2.09 - 2.00 (m, 1H), 1.99 - 1.91 (m, 2H), 1.78 - 1.67 (m, 3H).

[0420] SFC condition: Column: Cellucoat 50 x 4.6mm I.D., 3pm, Mobile phase: Phase A for C0 2 , and Phase B for MeOH (0.05% DEA); Gradient elution: MeOFI (0.05% DEA) in CO2 from 5% to 40%, Flow rate: 3mL/min; Wavelength: 220nm, Column Temp: 35C; Back Pressure: lOOBar.

[0421] Example 18: To a solution of 2-[(25)-4-[8-(2-fluoro-6-methyl-phenyl)-2-[[(2,S -l- methylpyrrolidin-2-yl]methoxyJ-5,6,7,9-tetrahydropyrimido[4, 5-c]azepin-4-yl]piperazin-2- yl]acetonitrile (66 mg, 109 pmol, 1.0 eq, TFA) and DIEA (562 mg, 4.34 mmol, 757 uL, 40.0 eq) in dichloromethane (1.5 mL) was added prop-2-enoyl chloride (19.7 mg, 217 pmol, 17.7 uL, 2.0 eq) dropwise at -40 °C. The mixture was stirred at -40 °C for 10 minutes. Upon completion, the mixture was quenched with saturated aqueous sodium bicarbonate (0.5 mL) and layers were separated. The aqueous phase was extracted with dichloromethane (5 mL). Combined organic layers were dried over Na 2 S0 4 , filtered and concentrated under vacuum. The residue was purified by prep-HPLC (column: Waters Xbridge 150*25 5u; mobile phase: [water (0.05% ammonium hydroxide v/v)-ACN]; B%: 55%-74%, lOmin). The desired fractions were collected and lyophilized to give 2-[(2S)-4-[8-(2-fluoro-6-methyl-phenyl)-2-[[(2X)-l- methylpyrrolidin-2- yl]methoxy]-5,6,7,9-tetrahydropyrimido[4,5-c]azepin-4-yl]-l- prop-2-enoyl-piperazin-2- yljacetonitrile (7.15 mg, 13.0 pmol, 12% yield, 99.5% purity) as a white solid. LCMS [ESI, M+l]: 548.

[0422] J H NMR (400MHz, chloroform-d) d = 7.03 - 6.96 (m, 1H), 6.95 - 6.84 (m, 2H), 6.59 (br s, 1H), 6.39 (dd, .7=1.6, 16.8 IIz, III), 5.83 (br d, J=10.4 Hz, 1H), 5.11 (br s, 1H), 4.46 - 4.08 (m, 4H), 4.08 - 3.79 (m, 2H), 3.79 - 3.44 (m, 2H), 3.43 - 3.14 (m, 3H), 3.09 (br t, J=7.6 Hz, 1H), 3.04 - 2.72 (m, 5H), 2.70 - 2.60 (m, 1H), 2.46 (s, 3H), 2.34 - 2.25 (m, 1H), 2.23 (s, 3H), 2.09 - 1.94 (m, 3H), 1.89 - 1 .73 (m, 3H).

[0423] SFC condition: Column: Cellucoat 50><4.6mm I.D., 3um, Mobile phase: Phase A for C0 2 , and Phase B for MeOH (0.05%DEA); Gradient elution: MeOH (0.05% DEA) in CO2 from 5% to 40%, Flow rate: 3mL/min; Wavelength: 220nm, Column Temp: 35C; Back Pressure: lOOBar.

EXAMPLE 19

2-[(2S)-4-[8-(5-methyl-17/-indazol-4-yl)-2-[[(25)-l-methy lpyrrolidin-2-yl]methoxy]-5,6,7,9- tetrahydropyrimido[4,5-c]azepin-4-yl]-l -prop-2 -enoyl-piperazin-2-yl]acetonitrile

[0424] Compound 19-1 : To a mixture of benzyl (25 -2-(cyanomethyl)-4-[2-[[(25)-l- methylpyrrolidin-2-yl]methoxy]-6,7,8,9-tetrahydro-5//-pynmid o[4,5-c]azepin-4-yl]piperazine-l- carboxylate (800 mg, 1.54 mmol, 1.0 eg), 4-bromo-5-methyl-l- tetrahydropyran-2-yl-indazole (545 mg, 1.85 mmol, 6.31 pL, 1.2 eg), CS 2 CO 3 (1.50 g, 4.62 mmol, 3.0 eg) an RuPhos (287 mg, 616 pmol, 0.4 eg) in toluene (20 mL) was added Pd 2 (dba) 3 (282 mg, 308 pmol, 0.2 eg) under N 2 . The suspension was degassed under vacuum and purged with N 2 several times. The mixture was stirred under N 2 at 90 °C for 8 hours. The reaction mixture was filtered and the filtrate was concentrated under vacuum. The residue was purified by reverse-phase flash [water (0.1% FA)/acetonitrile]. The desired fractions were collected and NaHC0 3 added to pH ~7,

concentrated under vacuum to remove MeCN and extracted with ethyl acetate (2 x 40 mL). The organic layers were dried over Na 2 S() i and concentrated under vacuum to give benzyl (2S)-2- (cyanomethyl)-4-[2-[[(2,S)-l-methylpyrrolidin-2- yl]methoxy]-8-(5-methyl-l-tetrahydropyran-2- yl-indazol-4-yl)-5, 6, 7, 9-tetrahydropyrimido[4,5-c]azepin-4-yl]piperazine-l -carboxylate (700 mg, 935 pmol, 61% yield, 98% purity) as a yellow solid. LCMS [ESI, M+l]: 734.

[0425] 'H NMR (400MHz, chloroform-d) d = 8.04 (d, 7=2.4 Hz, 1H), 7.43 - 7.33 (m, 5H), 7.26 - 7.16 (m, 2H), 5.67 (dd, 7=2.4, 9.6 Hz, 1H), 5.26 - 5.17 (m, 2H), 4.70 (br s, III), 4.54 - 4.38 (m, 2H), 4.35 (dd, 7=4.8, 10.8 Hz, 1 H), 4.20 - 4.08 (m, 2H), 4.03 (br d, .7=10.0 Hz, 1H), 3.82 (br d, 7=12.4 IIz, 1H), 3.77 - 3.70 (m, 1H), 3.66 (br d, 7=13.2 Hz, 1H), 3.61 - 3.53 (m, 1H), 3.50 - 3.40 (m, 1 H), 3.33 (br s, HI), 3.23 (br d, 7=1 1 .6 Hz, 1 H), 3.07 (br t, 7=7.6 Hz, 1H), 3.01 - 2.82 (m, 4H), 2.81 - 2.73 (m, HI), 2.68 - 2.51 (m, 2H), 2.45 (s, 3H), 2.26 (d, 7=1.2 Hz, 4H), 2.19 - 2.13 (m, 1H), 2.08 (br s, 1 H), 2.04 - 1.96 (m, 2H), 1.86 - 1.78 (m, 2H), 1.73 - 1.60 (m, 3H). [0426] Compound 19-2: To the solution of benzyl (2S)-2-(cyanomethyl)-4-[2-[[(2<S)-l- methylpyrrolidin-2-yl]methoxy]-8-(5-methyl-l-tetrahydropyran -2-yl-indazol-4-yl)-5,6,7,9- tetrahydropyrimido[4,5-c]azepin-4-yl]piperazine-l-carboxylat e (670 mg, 913 pmol, 1.0 eq) and NH 3* MeOH (5 mL, 30% purity) in MeOH (10 mL) was added Pd/C (300 mg, 10% purity) under N 2 . The suspension was degassed under vacuum and purged with H 2 several times. The mixture was stirred under 1 I 2 (15 psi) at 25 °C for 40 mins. The reaction mixture was filtered and the filtrate was concentrated under vacuum to give 2-[(25)-4-[2-[[(2S)-l-methylpyrrolidin-2- yl]methoxy]-8-(5 -methyl-1- tetrahydropyran-2-yl-indazol-4-yl)-5,6,7,9-tetrahydropyrimid o[4,5- c]azepin-4-yl]piperazin-2-yl]acetonitrile (480 mg, 720 pmol, 79% yield, 90% purity) as a white solid which was used for next step without further purification. LCMS [ESI, M+l]: 600.

[0427] Compound 19-3: To a solution of 2-[(25)-4-[2-[[(2S)-l-methylpyrrolidin-2- yljmethoxy]- 8-(5-methyl-l -tetrahydropyran-2-yl-indazol-4-yl)-5,6,7,9-tetrahydropyrimi do[4,5-c]azepin-4- yl]piperazin-2-yl]acetonitrile (40 mg, 66.7 pmol, 1.0 eq) in DCM (0.05 mL) was added TFA (308 mg, 2.70 mmol, 0.2 mL, 40 eq). the mixture was stirred at 25 °C for 0.5 hour. The reaction mixture was concentrated under vacuum. The residue was diluted with DCM (4 mL) and saturated aqueous Nal lCCL was added to pH = 7 ~ 8. Then the mixture was extracted with DCM (2 x 5 mL). The combined organic layers were dried over anhydrous Na 2 S0 4 , filtered and concentrated under vacuum. The residue was purified by prep-HPLC (column: Waters Xbridge 150*25 5p;mobile phase: [water (0.05% ammonium hydroxide v/v)-ACN];B%: 25% - 53%,l0min). The desired fractions were collected and lyophilized to give 2-[(25 -4-[8-(5- methyl-l//-indazol-4-yl)-2-[[(25)-l -methylpyrrolidin-2-yl]methoxy]-5, 6,7,9- fetrahydropyrimido[4,5-c]azepin-4-yl]piperazin-2-yl]acetonit rile (8.46 mg, 16.4 pmol, 25% yield, 100% purity) as a white solid. LCMS [ESI, M+l]: 516.

[0428] *H NMR (400MHz, chloroform-d) d = 8.1 1 (d, ,7=0.8 Hz, HI), 7.21 - 7.11 (m, 2H), 4.93 - 4.56 (m, 1H), 4.46 (s, 2H), 4.38 (dd, 7=4.8, 10.4 Hz, 1H), 4.14 (dd, 7=6.8, 10.4 Hz, 1H), 3.78 (br d, ,7=12.0 Hz, 1H), 3.65 - 3.47 (m, 311), 3.34 - 3.26 (m, 1H), 3.17 - 2.99 (m, 4H), 2.92 - 2.81 (m, 3H), 2.66 (td, .7=6.8, 13.2 Hz, 1H), 2.54 (d, J=6.4 Hz, 2H), 2.46 (s, 3H), 2.31 - 2.24 (m, 4H), 2.10 - 1.99 (m, 3H), 1.89 - 1.81 (m, 2H).

[0429] Example 19: To the solution of 2-[(2S)-4-[8-(5-methyl-177-indazol-4-yl)-2-[[(2S -l- methylpyn O lidin-2-yi]methoxy]-5,6,7,9-tetrahydropyrimido[4,5-c]a zepin-4-yl]piperazin-2- yl] acetonitrile (100 mg, 194 mihoΐ, 1.0 eq ) and TEA (58.9 mg, 582 mipoΐ, 81.0 uL, 3.0 eq) in DCM (2 mL) was added prop-2-enoyl chloride (17.6 mg, 194 mihoΐ, 15.8 pL, 1.0 eq) at -40 °C, the mixture was stirred at -40 °C for 10 mins. The reaction mixture was quenched by water (4 mL). The mixture was extracted with DCM (3 x 5 mL). The combined organic layers were dried over anhydrous Na 2 S0 4 , filtered and concentrated under vacuum. The residue was purified by reverse-phase flash [water (0.1% FA)/acetonitrile] Then the residue was purified by prep-HPLC (column: Xtimate C18 150*25mm*5pm;mobile phase: [water (0.05% ammonium hydroxide v/v)-ACN];B%: 40%-70%,lmin). The desired fractions were collected and lyophilized to give 2- [(25 -4-[8-(5-methyl- l/7-indazol-4-y l)-2-[[(25 -l -methylpyrrolidin-2-yl]inethoxy]-5, 6,7,9- tetrahydropyrimido[4,5-c]azepin-4-yl]-l -prop-2-enoyl-piperazin-2-yl]acetonitrile (16.8 mg, 29.1 mihoΐ, 12% yield, 98.6% purity) as a white solid. LCMS [ESI, M+l]: 570.

[0430] Ή NMR (400 MHz, chloroform-d) d = 10.34 (br s, 1H), 8.1 1 (s, 1H), 7.21 - 7.1 1 (m,

2H), 6.67 - 6.52 (m, 1H), 6.39 (dd, 7=1.6, 16.8 IIz, 1H), 5.82 (br d, 7=10.4 Hz, 1H), 5.1 1 (br s, 1H), 4.55 - 4.43 (m, 2H), 4.36 (dd, 7=4.8, 10.4 Hz, III), 4.14 (dd, 7=6.8, 10.8 Hz, 1H), 4.07 - 3.82 (m, 2H), 3.75 (br d, 7=1 1.6 Hz, 1H), 3.66 - 3.41 (m, 3H), 3.26 (dd, 7=3.9, 13.6 Hz, 1H),

3.08 (br t, 7=8.0 Hz, 1H), 3.04 - 2.88 (m, 4H), 2.80 (br s, 1H), 2.69 - 2.61 (m, III), 2.46 (s, 3H), 2.33 - 2.22 (m, 4H), 2.17 - 1.98 (m, 3H), 1.90 - 1.78 (m, 3H).

EXAMPLE 20

2-[(25)-l-(2-fluoiOprop-2-enoyl)-4-[8-(5-methyl-l//-indaz ol-4-yl)-2-[[(2S -l-methylpyrrolidin-

2-yl]methoxy]-5,6,7,9-tetrahydropyrimido[4,5-c]azepin-4-y l]piperazin-2-yl]acetonitrile

[0431] Example 20: To a solution of 2-[(25)-4-[8-(5-methyl-l /-indazol-4-yl)-2- [[(25}-l - methylpyrrolidin-2-yl]methoxy]-5,6,7,9-tetrahydropyrimido[4, 5-c]azepin-4-yl]piperazin-2- yl]acetonitrile (100 mg, 194 pmol, 1.0 eq), 2-fluoroprop-2-enoic acid (26.2 mg, 291 pmol, 3.16 pL, 1.5 eq) and TEA (118 mg, 1.16 mmol, 162 pL, 6.0 eq) in DMF (2 ml.) was added T3P (617 mg, 970 pmol, 577 pL, 50% purity, 5.0 eq) at -40 °C, the mixture was stirred at -40 °C for 10 mins. The mixture was stirred at 0 °C for 20 mins. Water (20 mL) was added into the mixture. The mixture was diluted with DCM (10 mL) and extracted with DCM (3 x 10 mL). The combined organic layers were dried over anhydrous Na 2 S0 4 , filtered and concentrated under vacuum. The residue was purified by prep-FIPLC (column: Xtimate Cl 8

150 * 25 m m * 5 p ;m obi 1 e phase: [water (0.05% ammonium hydroxide v/v)-ACN];B%: 44%- 74%,lmin). The desired fractions were collected and lyophilized to give 2-[(25 -l-(2-fluoroprop- 2- enoyl)-4-[8-(5-methyl-l/f-indazol-4-yl)-2-[[(25 -l -methylpyrrolidin-2-yl]methoxy]-5, 6,7,9- tetrahydropyrimido[4,5-c]azepin-4-yl]piperazin-2-yl]acetonit rile (13.2 mg, 22.2 pmol, 1 1% yield, 98.5% purity) as an off- white solid. LCMS [ESI, M+l]: 588.

[0432] *H NMR (400MHz, chloroform-d) d = 10.10 (br s, 1H), 8.1 1 (s, 1H), 7.21 - 7.14 (m, 2H), 5.52 - 5.32 (m, 1H), 5.25 (dd, 7=3.6, 16.8 Hz, III), 4.88 (br s, III), 4.56 - 4.42 (m, 2H), 4.35 (dd, 7=4.8, 10.4 Hz, 1H), 4.13 (dd, 7=6.8, 10.4 Hz, 1H), 3.88 (br d, .7=13.6 Hz, 1 H), 3.74 (br d,

7=13.6 Hz, 1 II), 3.64 - 3.42 (m, 3H), 3.29 (dd, 7=3.6, 13.6 Hz, 1H), 3.1 1 - 2.78 (m, 7H), 2.70 - 2.59 (m, 1H), 2.45 (s, 3H), 2.31 - 2.23 (m, 4H), 2.15 - 1.99 (m, 3H), 1.84 - 1.69 (m, 3H).

EXAMPLE 21

2-((S)-l-acryloyl-4-(2-(((5)-l-methylpyrrolidin-2-yl)meth oxy)-8-(8-(trifluoromethyl)naphthalen- l -yl)-6,7,8,9-tetrahydro-57/-pyrimido[4,5-c]azepin-4-yl)piper azin-2-yl)acetonitrile

Example 21

[0433] Compound 21-B: l,8-dibromonaphthalene (5 g, 17.5 mmol, 1.0 eq) was dissolved in THF (40 mL). The mixture was cooled down to - 70 °C and n-BuLi (2.5 M in hexane, 6.99 mL, 1.0 eq) was added dropwise. After 15 minutes, I 2 (4.44 g, 17.5 mmol, 3.52 mL, 1.0 eq) dissolved in THF (40 mL) was added. The mixture was allowed to reach 25 °C and stirred for 1 hour. The reaction was quenched with 30 mL of 1 M sodium thiosulfate solution in water. The reaction mixture was diluted with ethyl acetate (50 mL) and washed with water (20 mL). The organic layer was washed with brine (20 mL), dried over Na 2 S0 4 , filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (Si0 2 , Petroleum ether/Ethyl acetate=100/l to 10:1). l-bromo-8-iodo-naphthalene (3 g, 8.74 mmol,

50% yield, 97% purity) was obtained as a yellow solid.

[0434] Tl NMR (400MHz, chloroform-d) d = 8.43 (dd, = 1.2, 7.6 Hz, 1H), 7.96 (dd, J= 1.2, 7.6 Hz, 1H), 7.89 - 7.77 (m, 2H), 7.28 (t, J= 8.0 Hz, 1FI), 7.08 (t, J = 7.6 Hz, 1 H).

[0435] Compound 21-C: A mixture of Cul (1.57 g, 8.26 mmol, 1.1 eq) and KF (479 mg, 8.26 mmol, 193 pL, 1.1 eq) was thoroughly mixed and heated to 150 °C under vacuum by using oil pump with heat gun with gentle shaking until an homogemeous greenish color was obtained. To the mixture was added DMSO (50 mL), trimethyl(trifluoromethyl)silane (3.20 g, 22.5 mmol, 3.0 eq) and l-bromo-8-iodo-naphthalene (2.5 g, 7.51 mmol, 1.0 eq) was added and the slurry was heated to 25 °C for 16 h. The reaction mixture was diluted with ethyl acetate (50 mL) and washed with water (3 c 30 mL). The combined organic layers were washed with brine (20 mL), dried over Na 2 S0 4 , filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (column: Phenomenex Synergi C18 150*25* lOpm; mobile phase: [water (0.225% FA) - ACN]; B%: 66%- 86%, lOmin). l-bromo-8- (trifluoromethyl)naphthalene (900 mg, 3.14 mmol, 42% yield, 96% purity) was obtained as a white solid.

[0436] *H NMR (400MHz, chloroform-d) d = 8.12 (d, J= 7.6 Hz, 1H), 8.10 - 8.02 (m, 2H), 7.90 (dd, J = 1.2, 8.0 Hz, 1H), 7.54 (t, J= 7.6 Hz, 1H), 7.37 (t, .7= 7.6 Hz, 1H).

[0437] Compound 21-1: A mixture of benzyl (2S)-2-(cyanomethyl)-4-[2-[[(2S) -1 - methylpyrrolidin-2-yl]methoxy]-6,7,8,9-tetrahydro-5//-pyrimi do[4,5-c]azepin-4-yl]piperazine-l- carboxylate (500 mg, 962 pmol, 1.0 eq), l-bromo-8- (trifluoromethyl)naphthalene (529 mg, 1.92 mmol, 2.0 eq), Cs 2 C0 3 (940 mg, 2.89 mmol, 3.0 eq), BINAP-Pd-G3 (95.5 mg, 96.2 mhioΐ, 0.1 eq) in dioxane (10 mL) was degassed and purged with N 2 for 3 times, and then the mixture was stirred at 90 °C for 12 hours under N 2 atmosphere. The reaction mixture was diluted with water (20 mL) and extracted with ethyl acetate (3 c 20 mL). The combined organic layers were washed with brine (20 mL), dried over Na 2 S0 4 , filtered and concentrated under reduced pressure to give a residue. The residue was purified by reverse phase flash [water (0.1% formic

acid)/acetonitrile)]. The mixture was adjusted pH ~ 7 with saturated NaHCCfi aqueous solution and extracted with ethyl acetate (3 x 20 mL). The combined organic layers were washed with brine (20 mL), dried over Na 2 S0 4 , filtered and concentrated under reduced pressure to give the product benzyl (2S , )-2-(cyanomethyl)-4-[2-[[(25)-l-methylpyrrolidin-2-yl] methoxy]-8-[8- (trifluoromethyl)-l-naphthyl]-5,6,7,9-tetrahydropyrimido[4,5 -c]azepin-4-yl]piperazine-l- carboxylate (80 mg, 105 pmol, 11% yield, 94% purity) was obtained as a yellow· solid. LCMS [ESI, M+l]: 714.

[0438] Compound 21-2: To a solution of benzyl (25)-2-(cyanomethyl)-4-[2-[[(2.S)- l - methylpyrrolidin-2-yl]methoxy]-8-[8-(trifluoromethyl)-l -naphthyl]-5, 6,7,9- tetrahydropyrimido[4,5-c]azepin-4-yl]piperazine-l-carboxylat e (70 mg, 98.1 mhioΐ, 1.0 eq ) in MeOH (5 mL) was added NEb/MeOH (2 mL, 20% purity) and Pd/C (20 mg, 10% purity) under N 2 atmosphere. The suspension was degassed under vacuum and purged with H 2 several times. The mixture was stirred under H 2 (15 psi) at 25 °C for 0.5 hour. The reaction mixture was concentrated under vacuum. 2-[(2X)-4-[2-[[(25)-l -methylpyrrolidin-2-yl]methoxy]-8-[8- (trifluoromethyl) -l -naphthyl]-5,6,7,9-tetrahydropyrimido[4,5-c]azepin-4-yl]pipe razin-2- yl] acetonitrile (40 mg, 69.0 mihoΐ, 70% yield) was obtained as a yellow oil and used next step without purification. LCMS [ESI, M+l]: 580.

[0439] Example 21: To a solution of 2-[(26T4-[2-[[(2S -l-methylpynOlidin-2-yl]methoxy]-8- [8-(trifluoromethyl)-l-naphthyl]-5,6,7,9-tetrahydropyrimido[ 4,5-c]azepin-4-yl]piperazin-2- yljacetonitrile (40 mg, 69.0 mihoΐ, 1.0 eq) and DIEA (44.6 mg, 345 pmol, 60.1 pL, 5.0 eq) in dichloromethane (1 mL) was added a solution of prop-2-enoyl chloride (9.37 mg, 103 mihoΐ, 8.44 pL, 1.5 eq) in dichloromethane (1 mL) at - 40 °C. The mixture was stirred at - 40 °C for 0.5 hour. The reaction mixture was quenched with water (20 mL) and extracted with

dichloromethane (3 c 20 mL). The combined organic layers were washed with brine (10 mL), dried over Na 2 S0 4 , filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (Si0 2 , Ethyl acetate/Methanol= 100/1 to 10:1) and further purification by prep-IIPLC (column: Waters Xbridge 150 * 25 5m; mobile phase: [water (0.05% ammonia hydroxide v/v) - ACN]; B%: 55% - 79%, lOmin). The desired fraction was collected and lyophilizcd. 2-[(2S')-4-[2-[[(2S)-l-methylpyrrolidin-2-yl]methoxy]-8-[8- (trifluoiOmethyl)-l-naphthyl]-5,6,7,9-tctrahydropyrimido[4,5 -c]azepin-4-yl]-l-prop-2-enoyl- piperazin-2 -yljacetonitrile (18 mg, 28,4 mihoΐ, 41% yield, 99.9% purity) was obtained as a white solid. LCMS [ESI, M+l]: 634.

[0440] Ή NMR (400MHz, chloroform-d) d = 8.07 - 7.88 (m, 2H), 7.83 - 7.70 (m, 1H), 7.62 - 7.40 (m, 3H), 6.71 - 6.51 (m, 1H), 6.39 (dd, ./= 1 .6, 16.8 Hz, III), 5.83 (d, J= 10.8 Hz, 1 H),

5.30 - 4.47 (m, 1H), 4.43 - 4.27 (m, 2H), 4.22 - 3.89 (m, 3H), 3.88 - 3.45 (m, 3H), 3.42 - 3.16 (m, 3H), 3.14 - 2.70 (m, 6H), 2.69 - 2.57 (m, 1H), 2.42 (d, J= 5.2 Hz, 3H), 2.32 - 2.19 (m, 1H), 2.09 - 1.90 (m, 3H), 1.88 - 1.75 (m, 3H). 14).

EXAMPLE 22 2-((S)-l -(2-fluoroacryloyl)-4-(2-(((5)-l-methylpyrrolidin-2-yl)metho xy)-8-(naphthalen-l -y])-

6,7,8,9-tetrahydro-57/-pyrimido[4,5-c]azepin-4-yl)piperaz in-2-yl)acetonitrile

Cbz

Cbz i

[0441] Compound 22-1 : To a mixture of benzyl (2S)-2-(cyanomethyl)-4-[2-[[(25 -l- methylpyrrolidin-2-yl]methoxy]-6,7,8,9-tetrahydro-5 /-pyrimido[4,5-c]azepin-4-yl]piperazine-l - carboxylate (300 mg, 577 pmol, 1 .00 eq) and 1 -bromonaphthalene (239 mg, 1.15 mmol, 160 pL, 2.00 eq) in toluene (25.0 mL) was added Pd 2 (dba) 3 (52.9 mg, 57.7 pmol, 0.10 eq), RuPhos (53.9 g, 1 15 pmol, 0.20 eq), Cs 2 C(¾ (564 mg, 1.73 mmol, 3.00 eq) in one portion under N 2 . The mixture was degassed and purged with N 2 for 3 times and stirred at 90 °C for 5 hours. The reaction mixture was diluted with water (20.0 ml) and extracted with ethyl acetate (30 mL x 3). The combined organic layers were washed with brine (30 mL c 1), dried over sodium sulfate, filtered and concentrated under reduced pressure to give a residue. The residue was purified by reverse phase flash [water (0.1 % FA)/acetonitrile]. The desired fractions were collected and neutralized with saturated NaHCCfi solution (5.00 mL) and extracted with ethyl acetate (50.0 mL x 2). The separated organic layer was dried over sodium sulfate, filtered and concentrated under vacuum. Compound of benzyl (2S)-2- (cyanomcthyl)-4-|2-| | (2. S’)- 1 -methylpyrrolidin-2- yl]methoxy]-8-(l-naphthyl)-5,6,7,9-tetrahydropyrimido[4,5-c] azepin-4-yl|piperazine-l- carboxylate (260 mg, 403 pmol, 70% yield) as a yellow solid was obtained.

[0442] 'H NMR (400MHz, chloroform-d) d = 7.95 (d, ./ = 8.4 Hz, 1H), 7.81 (d, J = 8.0 Hz, 1H), 7.54 (d, J= 8.4 Hz, 1H), 7.46 - 7.33 (m, 8H), 7.12 (dd, J= 0.4, 7.2 Hz, 1H), 5.29 - 5.14 (m, 2H), 4.70 (br s, 1H), 4.45 - 4.31 (m, 3H), 4.21 - 4. l4 (m, 2H), 3.87 (br d, J = 13.2 Hz, 1H), 3.70 (br d, J= 13.2 Hz, 1H), 3.56 - 3.42 (m, 2H), 3.40 - 3.21 (m, 2H), 3.08 (br t, J= 7.2 Hz, 1H), 3.04 - 2.84 (m, 4H), 2.83 - 2.74 (m, 1 H), 2.72 - 2.60 (m, 1H), 2.45 (s, 3H), 2.33 - 2.23 (m, 1H), 2.21 - 2.06 (m, 2H), 2.05 - 1.96 (m, 2H), 1.89 - 1.76 (m, 2H).

[0443] Compound 22-2: To a solution of benzyl (25)-2-(cyanomethyl)-4-[2-[[(25 r )-l- methylpyrrolidin-2-yl]methoxy]-8-(l -naphthyl)-5,6,7,9-tetrahydropyrimido[4,5-c]azepin-4- yl]piperazine-l-carboxylate (150 mg, 232 pmol, 1.00 eq) in MeOH (3.00 mL) was added Pd/C (50.0 mg, 10% purity), NH 3* MeOH (1 .00 mL, 20% purity) under N 2 . The suspension was degassed under vacuum and purged with H? several times. The mixture was stirred under H 2 (15 psi) at 25 °C for 2 hours. The reaction mixture was filtered and concentrated under reduced pressure to give a residue. Compound of 2-[(2S r )-4-[2-| [(2Sd-l -methylpyrrolidin-2-yl]methoxy]- 8-(l-naphthyl)-5,6,7,9-tetrahydropyrimido[4,5-c]azepin-4-yl] piperazin-2-yl]acetonitrile (86.0 mg, 155 pmol, 67% yield, 92% purity) as a yellow solid was obtained, which was used in the next step directly without further purification. LCMS [ESI, M+l]: 512.

[0444] Example 22: To a mixture of 2-[(2S -4-[2-[[(2X)-Tmcthylpyrrolidin-2- yl]methoxy]-8- (l-naphthyl)-5,6,7,9-tetrahydropyrimido[4,5-c]azepin-4-yl]pi perazin-2-yl]acetonitrile (76.0 mg, 149 pmol, 1.00 eq) in ethyl acetate (2 mL) was added TEA (120 mg, 1.19 mmol, 165 pL, 8.00 eq), 2-fluoroprop-2-enoic acid (26.8 mg, 297 pmol, 2.00 eq), T3P (284 mg, 446 pmol, 265 pL, 50% purity, 3.00 eq) in portion at 0 °C under N 2 . The mixture was stirred at 25 °C for 30 min. The reaction mixture was quenched by addition of w r ater (2.00 mL) at 0 °C, and then diluted with water (3 mL) and extracted with ethyl acetate (20 mL c 3). The combined organic layers were washed with brine (20 mL x 1), dried over sodium sulfate, filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (column: Waters Xbridge 150 x 25 5p; mobile phase: [water (0.05% ammonia hydroxide v/v)-ACN]; B%: 55%- 82%, 10 min). Compound of 2-[(2S -l -(2-fluoroprop -2-enoyl)-4-[2-[[(25 r )-l -methylpyrrolidin-2- yl]methoxy]-8-(l -naphthyl)-5,6,7,9-tetrahydropyrimido[4,5-c]azepin-4-yl]pipe razin-2- yl] acetonitrile (23.7 mg. 39.9 pmol, 27% yield, 98.3% purity) as a white solid was obtained. LCMS [ESI, M+l]: 584.

[0445] ] H NMR (400MHz, chloroform-d) d = 7.95 (d, J = 8.4 Hz, 1H), 7.81 (d, J= 8.0 Hz, 1H), 7.54 (d, J= 8.0 Hz, 1H), 7.47 - 7.41 (m, 1 H), 7.40 - 7.33 (m, 2H), 7.13 (d, J= 6.8 Hz, 1H), 5.53 -

5.31 (m, 1H), 5.25 (dd, J= 3.6, 16.8 Hz, 1H), 4.88 (br s, 1H), 4.44 - 4.33 (m, 3H), 4.17 (dd, J = 6.8, 10.8 Hz, 2H), 3.91 (br d, ./ - 13.6 Hz, 1H), 3.76 (br d, J= 12.8 Hz, 1H), 3.59 - 3.38 (m, 3H),

3.31 (dd, J= 3.6, 14.0 Hz, 1H), 3.12 - 2.80 (m, 6H), 2.73 - 2.61 (m, 1 H), 2.46 (s, 3H), 2.32 - 2.22 (m, 1H), 2.20 - 1.93 (m, 5H), 1.89 - 1.77 (m, 1 H).

EXAMPLE 23

2-[(2S)-l-(2-fluoroprop-2-enoyl)-4-[8-(8-methyl-l-naphthy l)-5,6,7,9-tetrahydropyrimido[4,5- c]azepin-4-yl]piperazin-2-yl]acetonitrile

Example 23

[0446] Compound 23-1: To a mixture of /ert-butyl (25)-2-(cyanomethyl)-4-(6,7,8,9-tetrahydro - 5//-pyrimido[4,5-c]azepin-4-yl)piperazine-l -carboxylate (2 g, 5.37 mmol, 1 eq) and l -bromo-8- methyl-naphthalene (1 .78 g, 8.05 mmol, 1.5 eq) in toluene (30 mL) was added Pd 2 (dba)3 (983 mg, 1.07 mmol, 0.2 eq ), Xantphos (1.24 g, 2.15 mmol, 0.4 eq CS2CO3 (5.25 g, 16.1 mmol, 3 eq ) in one portion under N 2 . The mixture was stirred at 110 °C for 4 hours. Upon completion, the mixture was diluted with water (30 mL) and extracted with ethyl acetate (1 c 30 mL). The organic layer was separated, dried over sodium sulfate, filtered and concentrated under vacuum. The residue was purified by column chromatography (S1O2, Dichloromethane/Methanol = 100/1 to 10/1). After that, the residue was purified by reverse-phase HPLC (0.1% FA condition). The residue was basified with saturated aqueous NalfCOs solution to pH = 8 and extracted with ethyl acetate (3 x 50 mL). The organic layers were separated, dried over sodium sulfate, filtered and concentrated under vacuum. /er/-butyl (2X)-2-(cyanomethyl)-4-[8-(8-methyl-l-naphthyl)-5,6,7,9- tetrahydropyrimido[4,5-c]azepin-4-yl]piperazine-l-carboxylat e (1 g, 1.80 mmol, 34% yield, 92.4% purity) as a yellow oil was obtained. LCMS [ESI, M+l]: 513.

[0447] l H NMR (400MHz, chloroform-d) d = 8.58 (d, J=7.0 Hz, 1H), 7.69 - 7.60 (m, 2H), 7.42 -

7.35 (m, 1H), 7.35 - 7.31 (m, 1FI), 7.30 - 7.27 (m, 1H), 7.22 - 7.16 (m, 1H), 4.65 (br s, 1H), 4.52

- 4.42 (m, 1H), 4.40 - 4.29 (m, 1H), 3.85 - 3.71 (m, 1H), 3.65 - 3.53 (m, 1H), 3.50 - 3.39 (m, 1H),

3.35 - 3.25 (m, 2H), 3.23 - 3.14 (m, 1H), 3.11 - 2.85 (m, 5H), 2.83 - 2.71 (m, 5H), 2.03 - 1.97 (m, 1H), 1.53 (d, .7=1.4 Hz, 9H).

[0448] Compound 23-2: To a solution of tert- butyl (2N)-2-(cyanomethyl)-4-[8-(8-methyl-l - naphthyl)-5,6,7,9-tetrahydropyrimido[4,5-c]azepin-4-yl]piper azine-l-carboxylate (lOOmg, 195 mihoΐ, 1 eq) in MeCN (1 mL) was added HCl/dioxane (4 M, 2 mL, 41.0 eq) at 25 °C. The mixture was stirred at 25 °C for 0.5 hour. Upon completion, the mixture was concentrated under vacuum. The residue was diluted with saturated NaHCCfi solution (3 mL) and extracted with ethyl acetate (2 x 3 mL). The combined organic layers were washed with brine (1 c 5 mL), dried over sodium sulfate, filtered and concentrated under vacuum. The residue was purified by prep- HPLC (column: Waters Xbridge 150 * 25 5m; mobile phase: [water (0.05% ammonia hydroxide v / v) - ACN]; B%: 38% - 68%, lOmin). The residue was concentrated under reduced pressure to remove ACN, and then lyophilization. 2-[(25')-4-[8-(8-methyl-l - naphthyl)-5,6,7,9- tetrahydropyrimido[4,5-c]azepin-4-yl]piperazin-2-yl]acetonit rile (14.9 mg, 35.3 mhioΐ, 18% yield, 97.5% purity) as a yellow solid was obtained. LCMS [ESI, M+l]: 413.

[0449] 1H NMR (400MHz, chloroform-d) d = 8.55 (br s, 1H), 7.72 - 7.58 (m, 2H), 7.47 - 7.14 (m, 411), 4.77 - 4.20 (m, 2H), 3.91 - 2.72 (m, 13H), 2.56 (br d, J=5.0 Hz, 2H), 2.02 (br s, 3H). [0450] Example 23: To a solution of 2-[(25)-4-[8-(8-methyl-l-naphthyl)-5, 6,7,9- tetrahydropyrimido[4,5-c]azepin-4-yl]piperazin-2-yl]acetonit rile (150 mg, 364 pmol, 1 eq), 2- fluoroprop-2-enoic acid (98.2 mg, 1.09 mmol, 3 eq) and Et 3 N (331 mg, 3.27 mmol, 455 pL, 9 eq) in DMF (15 mL) was added T3P (926 mg, 1.45 mmol, 865 pL, 50% purity, 4 eq) at 0 °C.

The mixture was stirred at 0 °C for 0.5 hour. The residue was diluted with water (15 mL) and extracted with ethyl acetate (2 x 20 mL). The combined organic layers were washed with brine (1 x 20 mL), dried over sodium sulfate, filtered and concentrated under vacuum. The mixture was purified by column chromatography (Si0 2 , Dichloromethane/Methanol = 10/1). After that, the residue was purified by prep-HPLC (column: Waters Xbridge 150 * 25 5p; mobile phase: [water (0.05% ammonia hydroxide v / v) - ACN]; B%: 48% - 78%, lOmin). The residue was concentrated under reduced pressure to remove ACN, and then lyophilization. 2-[(2S)-l-(2- fluoroprop-2-enoyl)-4-[8-(8-methyl-l-naphthyl)-5,6,7,9-tetra hydropyrimido[4,5-c]azepin-4- yl]piperazin-2-y]]acetonitrile (33.8 mg, 68.3 pmol, 19% yield, 98.0% purity) as a white solid was obtained. LCMS [ESI, M+l]: 485.5.

[0451] Ή NMR (400MHz, chloroform-d) d = 8.59 (d, .7=7.0 Hz, 1H), 7.71 - 7.61 (m, 2H), 7.45 - 7.27 (m, 3H), 7.23 - 7.14 (m, 1H), 5.56 - 5.32 (m, 1H), 5.26 (dd, J=3.6, 16.9 Hz, 1H), 4.56 - 4.44 (m, 1H), 4.42 - 4.31 (m, 1H), 4.22 - 3.96 (m, 1H), 3.86 (br t, J=13.2 Hz, HI), 3.68 (br t, .7=1 1.4 Hz, 1H), 3.54 - 3.20 (m, 4H), 3.19 - 2.72 (m, 9H), 2.17 - 1.94 (m, 2H).

EXAMPLE 24

2-((5)-4-(8-(l//-indazol-4-yl)-2-(((5)-l -methylpyrrolidin-2-yl)methoxy)-6,7,8,9-tetrahydro-5//- pyrimido[4,5-c]azepin-4-yl)-l-acryloylpiperazin-2-yl)acetoni trile

Example 24

[0452] Example 24: To a mixture of 2-[(2,S)-4-[8-(177-indazol-4-yl)-2-[[(2S')-l - methylpyrrolidin-2-yl]methoxy]-5,6,7,9-tetrahydropyrimido[4, 5-c]azepin-4-yl]piperazin-2- yljacetonitrile (20.0 mg, 39.9 mpioΐ, 1.0 eq) in dichloromethane (0.50 mL) was added TEA (12.1 mg, 120 mihoΐ, 16.7 pL, 3.0 eq) and prop-2-enoyl prop-2-enoate (5.03 mg, 39.9 pmol, 1.0 eq) in portion at -40 °C under N 2 . The mixture was stirred at -40 °C for 10 min. The reaction mixture was quenched by addition of methanol (0.05 mL) at -40 °C, and then diluted with water (0.50 mL) and extracted with dichloromethane (3.0 mL x 3). The combined organic layers were washed with brine (2.0 mL x 1), dried over sodium sulfate, filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (column: Waters Xbridge 150 c 25 5 m; mobile phase: [water (0.05% ammonia hydroxide v/v)-ACN]; B%: 27%- 57%, 10 min). Compound 2-[(2S)-4-[8-(17/-indazol-4-yl)-2-[[(2 > S)-l- methylpyrrolidin-2- yl]methoxy]-5,6,7,9-tetrahydropyrimido[4,5-c]azepin-4-yl]-l- prop-2-enoyl-piperazin-2- yl] acetonitrile (1.60 mg, 2.56 pmol, 6% yield, 89% purity) as a white solid was obtained. LCMS [ESI, M+l]: 556.

[0453] Ή NMR (400MHz, chloroform-d) d = 10.61 - 9.47 (m, 1H), 8.04 (s, 1H), 7.20 (t, 7=8.0 Hz, 1H), 6.90 (d, .7=8.0 Hz, 1 H), 6.56 (br s, 1H), 6.43 - 6.29 (m, 2H), 5.80 (br d, 7=10.8 Hz, 1H), 5.21 - 4.63 (m, 3H), 4.47 (dd, 7=4.8, 10.6 Hz, 1H), 4.24 (dd, 7=6.8, 10.8 Hz, 1H), 3.94 - 3.73 (m, 3H), 3.69 - 3.32 (m, 2H), 3.23 - 3.08 (m, 2H), 2.91 (br dd, 7=8.0, 16.4 Hz, 2H), 2.84 - 2.66 (m, 4H), 2.53 (s, 3H), 2.40 - 2.26 (m, 1 H), 2.24 - 2.03 (m, 3H), 1.95 - 1.73 (m, 4H).

EXAMPLE 25

2-[(2S -4-[8-(8-methyl-l -naphthyl)-5,6,7,9-tetrahydropyrimido[4,5-c]azepin-4-yl]-l-p rop-2- enoyl-piperazin-2-yl]acetonitrile

23-2 Example 25

[0454] Example 25: To a mixture of 2-[(2S)-4-[8-(8-methyl-l -naphthyl)-5, 6,7,9- tetrahydropyrimido[4,5-c]azepin-4-yl]piperazin-2-yl]acetonit rile (100 mg, 150 pmol, 1 eq , HC1) and DIEA (194 mg, 1.50 mmol, 261 pL, 10 eq) in DCM (10 mL) was added prop-2-enoyl prop- 2-enoate (22.7 mg, 180 pmol, 1.2 eq) in portion at - 40 °C. The mixture was stirred at - 40 °C for 30 min. Upon completion, the mixture was diluted with water (5 mL). The organic layer was separated, washed with brine (1 x 10 mL), dried over sodium sulfate, filtered and concentrated under vacuum. The mixture was purified by column chromatography (Si0 2 ,

Dichloromethane/Methanol = 10/1). After that the residue was purified by prep-HPLC (column: Waters Xbridge 150 * 25 5m; mobile phase: [water (0.05% ammonia hydroxide v / v) - ACN]; B%: 42% - 72%, lOmin). The residue was concentrated under reduced pressure to remove ACN, and then lyophilization. 2-|(2,S')-4-[8-(8-methyl -l-naphthyl)-5,6,7,9-tetrahydropyrimido[4,5- c]azepin-4-yl]-l-prop-2-enoyl-piperazin-2-yl]acetonitrile (6.2 mg, 12.9 pmol, 9% yield, 96.9% purity) as a white solid was obtained. LCMS [ESI, M+l]: 467.

[0455] 'Ll NMR (400MHz, chloroform-d) 5 = 8.59 (d, .7=6.2 Hz, 1H), 7.75 - 7.57 (m, 2II), 7.46 - 7.28 (m, 3H), 7.20 (br d, .7=4.2 Hz, 1H), 6.61 (br s, 1H), 6.40 (br d, J=l6.4 Hz, 1 H), 5.84 (br d, .7=10.6 Hz, 1H), 4.58 - 4.42 (m, HI), 4.41 - 4.30 (m, 1H), 3.98 (br s, 1H), 3.92 - 3.79 (m, 1H), 3.70 (br s, 1H), 3.47 (br dd, 7=7.2, 13.0 Hz, 1H), 3.37 - 2.62 (m, 12H), 2.04 (br d, /=5.2 Hz, 2H).

EXAMPLE 26

2-((S , )-4-(8-(l7/-indazol-4-yl)-2-(((5)-l-methylpyrrolidin-2 -yl)methoxy)-6,7,8,9-tetrahydro-5/7- pyrimido[4,5-c]azepin-4-yl)-l -(2-fluoroacryloyl)piperazin-2-yl)acetonitrile

Example 26

[0456] Example 26: To a mixture of 2-[(25)-4-[8-(li/-indazol-4-yl)-2-[[(25)-l - methylpyrrolidin-2-yl]methoxy]-5,6,7,9-tetrahydropyrimido[4, 5-cjazepin-4-yl]piperazin-2- yl]acetonitrile (30.0 mg, 59.8 mhioΐ, 1.00 eq) in ethyl acetate (0.20 mL) was added TEA (96.8 mg, 957 pmol, 133 pL, 16.0 eq), 2-lluoroprop-2-enoic acid (5.39 mg, 59.8 pmol, 1.00 eq) and T3P (228 mg, 359 pmol, 213 pL, 50% purity, 6.00 eq) in portion at 0 °C under N 2 . The mixture was stirred at 25 °C for 0.5 hour. The reaction mixture was quenched by addition of water (1.00 mL) at 0 °C, and then extracted with ethyl acetate (5 mL c 3). The combined organic layers were washed with brine (2 mL x 1), dried over sodium sulfate, filtered and concentrated under reduced pressure to give a residue. The residue was purified by prep-HPLC (column: Waters Xbridge 150 x 25 5 p;mobile phase: [water (0.05% ammonia hydroxide v/v)-ACN];B%: 30%- 60%, 10 min). Compound 2-[(25)-l-(2-fhroroprop-2-enoyl)-4-[8-(l//-indazol-4- yl)-2-[[(25)-l- methylpyrrolidin-2-yl]methoxy]-5,6,7,9-tetrahydropyrimido[4, 5-c]azepin-4-yl]piperazin-2- yl]acetonitrile (5.75 mg, 9.82 pmol, 16% yield, 98% purity) as a gray solid was obtained. LCMS [ESI, M+l]: 574.

[0457] l U NMR (400MHz, chloroform-d) d = 10.69 - 9.24 (m, 1H), 8.04 (s, 1H), 7.20 (t, J-8.0 Hz, 1H), 6.91 (d, .7=8.4 Hz, 1H), 6.34 (d, J= 7.6 Hz, 1H), 5.52 - 5.29 (m, 1H), 5.23 (dd, .7-3.6, 16.8 Hz, 1H), 5.01 - 4.54 (m, 4H), 4.44 (dd, .7=4.8, 10.8 Hz, 1H), 4.23 (dd, J-7.2, 10.8 Hz, 1H), 3.87 - 3.74 (m, 3H), 3.63 (br d, .7=13.2 Hz, 1H), 3.43 (br s, 1H), 3.21 (dd, .7-4.0, 14.0 Hz, 1H), 3.12 (br t, 7=7.2 Hz, 1H), 3.02 - 2.88 (m, 2H), 2.85 - 2.68 (m, 4H), 2.51 (s, 3H), 2.38 - 2.26 (m, 1H), 2.21 - 2.06 (m, 3H), 1.95 - 1.78 (m, 3H).

EXAMPLE A

KRas G12C Modification Assay

[0458] This Example illustrates that exemplary compounds of the present invention may be assayed using a LCMS assay to detect a covalent adduct of the exemplary compound and KRAS G12C.

[0459] The protein concentration of GDP-loaded K-Ras (1-169) G12C, C51 S,C80L,C1 18S and GTP-loaded K-Ras (1-169) G12C,C51 S,C80L,C118S,Q6lH are adjusted to 2 mM in K-Ras Assay Buffer (25 mM HEPES,150 mM NaCl. 5 mM MgCl 2 , and 10 mM Octyl b- glucopyranoside at pH 7.5). A 10 pL aliquot of each protein solution is transferred to a 384 well microtiter plate. Initial compound stocks are generated at fifty times their desired final assay concentration in DMSO. [0460] Exemplary compounds of Formula (I) are diluted 25-fold into K-Ras Assay Buffer to a final of two times their final concentration. A 10 pL aliquot of each diluted compound solution is then added to each of the protein solutions in the microtiter plate to initiate reaction. Typical final compound concentrations are 3.0, 5.0 and 25.0 mM. At each time point, the reactions are quenched with 20 pL of a 25 mM acetic acid solution. Usual assay endpoints are 15, 180 and 1440 minutes. Once all reactions are quenched, the plates are heat sealed and the samples are injected into a LC/MS system for data acquisition.

[0461] Data collection may take place on an Agilent 6520 Q-TOF Accurate Mass Spectrometer. Samples are injected in their liquid phase onto a C-3 reverse phase column to remove assay buffer and prepare the samples for mass spectrometer. The proteins are eluted from the column using an acetonitrile gradient and fed directly into the mass analyzer. Initial raw data analysis may take place using Agilent MassFIunter software immediately post data acquisition.

[0462] Raw data analysis of the intact protein is exclusively a deconvolution of the multiple charge states of each protein in solution using a maximum entropy deconvolution provided in Mass Hunter. To minimize complexity, only the data over limited mass ranges are considered for analysis, with a minimum of one Dalton mass step intervals. The heights of all masses identified during raw data analysis are exported to be further analyzed in Spotfire® data analysis software.

[0463] Final data analysis is a multistep process in the Spotfire® data analysis software package. Briefly, each protein mass is calculated as a percent of the total signal of that sample, that percentage is then normalized to the percentage of signal of the protein in the absence of reactive compounds. Those normalized signals are reported as normalized percent of control (POC). An increased POC value indicates a compound that displays a higher degree of modification at a given condition compared to other compounds under the same conditions.

EXAMPLE B

Inhibition of KRas G12C-dependent Cell Growth

[0464] This Example illustrates that exemplary compounds of the present invention inhibit the growth of tumor cell lines that express KRas G12C. [0465] The cellular inhibition of KRAs G12C by exemplary compounds of the present invention was determined by measuring the amount of a downstream marker of KRas activity,

phosphorylated ERK (“Phospho-ERK”).

[0466] NC1-H358 cells (ATCC CRL-5807) express KRas G12C and were grown in RPMI medium supplemented with 10% fetal bovine serum, penicillin/streptomycin and 10 mM

HEPES. Cells were plated in poly-D-Lysine coated 96-well plates at a concentration of 50,000 cells/well and allowed to attach for 8-12 hours. Diluted compounds were then added at a final concentration of 0.5 % DMSO. After 3 hours, the medium was removed, 150 uL of 4% formaldehyde was added and the plates were incubated for 20 minutes. The plates were washed with PBS, and permeabilized using 150 pL of ice cold 100% methanol for 10 minutes. Non specific antibody binding to the plates was blocked using 100 pL Licor Blocking Buffer (Li-Cor Biotechnology, Lincoln NE) for 1 hour at room temperature. Positive control samples and samples lacking cells were parallel processed with test samples as standards.

[0467] The amount Phospho-ERK was determined using an antibody specific for the

phosphorylated form of ERK and compared to the amount of GAPDH. Primary antibodies used for detection were added as follows: Phospho-ERK (Cell Signaling cs9101) diluted 1 :500 and GAPDH (Millipore MAB374) diluted 1 :5000 in Licor block + 0.05% Tween 20. The plates were incubated for 2 hours at room temperature. The plates were washed with PBS + 0.05% Tween 20.

[0468] Secondary antibodies used to visualize primary antibodies were added as follows: Anti- rabbit-680 diluted 1 : 1000 and Anti-mouse-800 diluted 1 :1000 in Licor Block + 0.05% Tween 20 and incubated for 1 hour at room temperature. The plates were washed with PBS + 0.05% Tween 20. A 100 qL aliquot of PBS was added to each well and the plates were read on a L1COR AERIUS plate reader.

[0469] The pERK(Thr202/Tyr204) signal was normalized with the GAPDH signal and percent of DMSO control values were calculated. IC50 values were generated using a 4 parameter fit of the dose response curve. The results for exemplary compounds of Formula (I) arc shown in Table 2. Key:“A” < 500 nM;“B” > 500 nM and ND = not determined.

Table 1 Inhibition of KRas Gl2C-mediated Cell Proliferation by Exemplary Compounds

[0470] While the invention has been described in connection with specific embodiments thereof, it will be understood that it is capable of further modifications and this application is intended to cover any variations, uses, or adaptations of the invention following, in general, the principles of the invention and including such departures from the present disclosure as come within known or customary practice within the art to which the invention pertains and as may be applied to the essential features hereinbefore set forth, and as follows in the scope of the appended claims.