Compound and use thereof

文档序号:816415 发布日期:2021-03-26 浏览:13次 中文

阅读说明:本技术 化合物及其用途 (Compound and use thereof ) 是由 I·弗罗纳 P·蒂维特马海松 D·塔迪夫 B·潘迪亚 K·奥斯博亚 M·卢卡斯 B·L· 于 2019-04-24 设计创作,主要内容包括:本发明的特征在于可用于治疗神经病症的化合物。本发明的化合物单独或与其他药物活性剂组合,可用于治疗或预防神经病症。(The invention features compounds useful for treating neurological disorders. The compounds of the present invention are useful, alone or in combination with other pharmaceutically active agents, for the treatment or prevention of neurological disorders.)

1. A compound having the structure of formula I:

wherein

R1Is H, halo, CN, NO2Hydroxy, optionally substituted amino, optionally substituted C1-C6Alkyl, optionally substituted C1-C6Heteroalkyl, optionally substituted C1-C6Alkenyl, optionally substituted C 1-C6Heteroalkenyl, optionally substituted C3-C10Carbocyclyl, optionally substituted C2-C9Heterocyclyl, optionally substituted C6-C10Aryl or optionally substituted C2-C9(ii) a heteroaryl group, wherein,

R2is H or optionally substituted C1-C6Alkyl, or

R1And R2Together with the atoms to which each is attached to form optionally substituted C3-C10Carbocyclylene, optionally substituted C2-C9Heterocyclylene radical, optionally substituted C6-C10Arylene or optionally substituted C2-C9A heteroarylene group;

L1is optionally substituted C1-C6Alkylene or optionally substituted C1-C6A heteroalkylene group; and is

R3Is optionally substituted C2-C9(ii) a heteroaryl group, wherein,

or a pharmaceutically acceptable salt thereof.

2. The compound of claim 1, wherein R3Is a heteroaryl having the structure of formula Ia:

wherein

X1、X2、X3And X4Is independently O, NR4Or CR5

Wherein

Each R4Independently is H, optionally substituted C1-C6Alkyl, optionally substituted C1-C6Heteroalkyl, optionally substituted C3-C10Carbocyclyl, optionally substituted C2-C9Heterocyclyl, optionally substituted C6-C10Aryl or optionally substituted C2-C9(ii) a heteroaryl group, wherein,

each R5Independently H, CN, optionally substituted C1-C6Alkyl or optionally substituted C1-C6A heteroalkyl group, and

if X is1、X2、X3Or X4Is O, then said adjacent atom is N or CR5(ii) a And is

X5Is N or C, wherein X1、X2、X3、X4Or X51, 2 or 3 of which are O or N.

3. The compound of claim 1 or 2, wherein R3Is that

4. The compound of any one of claims 1 to 3, wherein R3Is that

5. The compound of claim 4, wherein R3Is that

6. The compound of any one of claims 1 to 5, wherein each R5Independently H, CN or optionally substituted C1-C6An alkyl group.

7. The compound of claim 6, wherein each R5Independently H, CN,

Wherein

j is 0, 1, 2, 3, 4 or 5; and is

Each R6Independently is halo, CN, optionally substituted C1-C6Alkyl or optionally substituted C1-C6A heteroalkyl group.

8. The compound of claim 7, wherein each R6Independently F, Cl, Br, I, CN,

9. The compound of claim 7 or 8, wherein j is 0, 1, 2, or 3.

10. The compound of claim 9, wherein j is 2.

11. A compound according to any one of claims 7 to 10Wherein each R is5Independently H, CN,

12. The compound of any one of claims 1 to 11, wherein R4Is H.

13. The compound of any one of claims 1 to 11, wherein R4Is optionally substituted C1-C6Alkyl, optionally substituted C1-C6Heteroalkyl, optionally substituted C3-C10Carbocyclyl, optionally substituted C 2-C9Heterocyclyl, optionally substituted C6-C10Aryl or optionally substituted C2-C9A heteroaryl group.

14. The compound of claim 10, wherein R4Is optionally substituted C1-C6A heteroalkyl group.

15. The compound of claim 11, wherein R4Is that

16. The compound of claim 13, wherein R4Is optionally substituted C3-C10Carbocyclyl, optionally substituted C2-C9Heterocyclyl, optionally substituted C6-C10Aryl or optionally substituted C2-C9A heteroaryl group.

17. The compound of claim 16, wherein R4Is that

18. The compound of claim 13, wherein R4Is optionally substituted C1-C6An alkyl group.

19. The compound of claim 18, wherein R4Is that

20. The compound of claim 18, wherein R4Is that

Wherein

k1 is 0, 1, 2, 3, 4 or 5;

k2 is 0, 1, 2, 3 or 4;

k3 is 0, 1, 2 or 3;

w is O or S;

m is 1 or 2; and is

Each R7Independently halogen, CN, NO2Hydroxy, optionally substituted C1-C6Alkyl or optionally substituted C1-C6A heteroalkyl group.

21. The compound of claim 20, wherein m is 2.

22. The compound of claim 20, wherein m is 1.

23. The compound of any one of claims 20 to 22, wherein each R7Independently halo, CN, optionally Substituted C1-C6Alkyl or optionally substituted C1-C6A heteroalkyl group.

24. The compound of any one of claims 20 to 23, wherein each R7Independently F, Cl, Br, I, CN, -CF3

25. The compound of any one of claims 20 to 24, wherein k1 is 0.

26. The compound of any one of claims 20 to 24, wherein k1 is 1.

27. The compound of claim 26, wherein R4Is that

28. The compound of any one of claims 20 to 24, wherein k1 is 2.

29. The compound of claim 28, wherein R4Is that

30. The compound of any one of claims 20 to 24, wherein k2 is 0.

31. The compound of any one of claims 20 to 24, wherein k2 is 1.

32. The compound of claim 31, wherein R4Is that

33. The compound of any one of claims 20 to 24, wherein k3 is 0.

34. The compound of any one of claims 20 to 24, wherein k3 is 1.

35. The compound of claim 34, wherein R4Is that

36. The compound of claim 35, wherein W is S.

37. The compound of any one of claims 20-36, wherein R4Is that

38. The compound of claim 15, wherein R 4Is that Wherein m is 1 or 2.

39. The compound of claim 38, wherein R4Is that

40. The compound of claim 15, wherein R4Is that

Wherein

k1 is 0, 1, 2, 3, 4 or 5;

each R7Independently halogen, CN, NO2Hydroxy, optionally substituted C1-C6Alkyl or optionally substituted C1-C6A heteroalkyl group;

Rais H, optionally substituted C1-C6Alkyl or optionally substituted C1-C6A heteroalkyl group; and is

RbIs optionally substituted C1-C6Alkyl or optionally substituted C1-C6A heteroalkyl group.

41. The compound of claim 40, wherein each R7Independently is halo, CN, optionally substituted C1-C6Alkyl or optionally substituted C1-C6A heteroalkyl group.

42. Such as rightThe compound of claim 40 or 41, wherein each R7Independently F, Cl, Br, I, CN, -CF3

43. The compound of any one of claims 40 to 42, wherein k1 is 0.

44. The compound of any one of claims 40 to 42, wherein k1 is 1.

45. The compound of claim 44, wherein R4Is that

46. The compound of any one of claims 40 to 42, wherein k1 is 2.

47. The compound of claim 46, wherein R4Is that

48. The compound of any one of claims 40-47, wherein R aIs H or-CH3

49. The compound of any one of claims 40-48, wherein RbIs optionally substituted C1-C6An alkyl group.

50. The compound of claim 49, wherein RbIs that

Wherein

b is 1 or 2; and is

RcIs optionally substituted amino, optionally substituted mercapto, optionally substituted sulfone or optionally substituted sulfoxide.

51. The compound of claim 50, wherein RcIs that

Wherein

Rd、ReAnd RfEach of which is independently H, optionally substituted C1-C6Alkyl or optionally substituted C1-C6An alkyl group.

52. The compound of any one of claims 40-51, wherein R4Is that

53. The compound of any one of claims 1 to 52, wherein R2Is H or optionally substituted C1-C6An alkyl group.

54. The compound of any one of claims 1 to 53, wherein R2Is H,

55. The compound of any one of claims 1 to 54, wherein R2Is H.

56. The compound of any one of claims 1 to 55, wherein R1Is H, halo, CN, NO2Hydroxy, optionally substituted amino, optionally substituted C1-C6Alkyl, optionally substituted C1-C6Heteroalkyl, optionally substituted C1-C6Alkenyl, optionally substituted C1-C6Heteroalkenyl, optionally substituted C3-C10Carbocyclyl, optionally substituted C 2-C9Heterocyclyl, optionally substituted C6-C10Aryl or optionally substituted C2-C9A heteroaryl group.

57. The compound of any one of claims 1 to 56, wherein R1Is H, halo, CN, optionally substituted C1-C6Alkyl, optionally substituted C1-C6Heteroalkyl, optionally substituted C1-C6Alkenyl, optionally substituted C3-C10Carbocyclyl, optionally substituted C2-C9Heterocyclyl, optionally substituted C6-C10Aryl or optionally substituted C2-C9A heteroaryl group.

58. The compound of any one of claims 1-57, wherein R1Is H, halo, CN, optionally substituted C1-C6Alkyl, optionally substituted C1-C6Heteroalkyl or optionally substituted C1-C6An alkenyl group.

59. The compound of any one of claims 1 to 58, wherein R1Is H, F, Cl, Br, I, CN,

60. The compound of any one of claims 1-57, wherein R1Is optionally substituted C3-C10A carbocyclic group.

61. The compound of claim 60, wherein R1Is that

Wherein

c1 is 0, 1, 2, 3, 4 or 5;

c2 is 0, 1, 2, 3 or 4;

c3 is 0, 1, 2, 3, 4, 5 or 6;

c4 is 0, 1, 2, 3, 4 or 5; and is

Each R8Independently is halo, optionally substituted C1-C6Alkyl or optionally substituted C1-C6A heteroalkyl group.

62. The compound of claim 61, wherein c1 is 0.

63. The compound of claim 61, wherein c2 is 0.

64. The compound of claim 61, wherein c3 is 0.

65. The compound of claim 61, wherein c4 is 0.

66. The compound of claim 56 or 57, wherein R1Is optionally substituted C2-C9A heterocyclic group.

67. The compound of claim 66, wherein R1Is that

Wherein

d1 is 0, 1, 2 or 3;

d2 is 0, 1, 2 or 3;

d3 is 0, 1, 2, 3 or 4;

d4 is 0, 1, 2, 3, 4 or 5;

d5 is 0, 1, 2, 3 or 4;

each R9Independently is halo, hydroxy, optionally substituted C1-C6Alkyl or optionally substituted C1-C6A heteroalkyl group; and is

R10Is H, optionally substituted C1-C6Alkyl or optionally substituted C1-C6A heteroalkyl group.

68. The compound of claim 67, wherein d1 is 0.

69. The compound of claim 67, wherein d2 is 0.

70. The compound of claim 67, wherein d3 is 0.

71. The compound of claim 67, wherein d4 is 0.

72. The compound of claim 67, wherein d5 is 0.

73. The compound of claim 67, wherein d3 is 1.

74. The compound of claim 73, wherein R9Is hydroxy or optionally substituted C 1-C6A heteroalkyl group.

75. The compound of claim 74, wherein R9Is a hydroxyl group,

76. The compound of any one of claims 67 to 75, wherein R10Is H,

77. The compound of any one of claims 1-57, wherein R1Is optionally substituted C6-C10And (4) an aryl group.

78. The compound of claim 77, wherein R1Is that

Wherein

e is 0, 1, 2, 3, 4 or 5; and is

Each R11Independently halogen, CN, NO2Hydroxy, optionally substituted C1-C6Alkyl or optionally substituted C1-C6A heteroalkyl group.

79. The compound of claim 78, wherein e is 0.

80. The compound of claim 78, wherein e is 1.

81. The compound of claim 80, wherein R1Is that

82. The compound of claim 78, wherein e is 2.

83. The compound of claim 82, wherein R1Is that

84. The compound of any one of claims 1-57, wherein R1Is optionally substituted C2-C9A heteroaryl group.

85. The compound of claim 84, wherein R1Is that

Wherein

X is O, S or NRN1Wherein R isN1Is H, optionally substituted C1-C6Alkyl or optionally substituted C1-C6A heteroalkyl group;

Y1、Y2and Y3Each of which is independently N or CRC1Wherein R isC1Is H, halo, CN, optionally substituted C 1-C6Alkyl or optionally substituted C1-C6A heteroalkyl group; and is

R12Is H, halo, CN, optionally substituted C1-C6Alkyl or optionally substituted C1-C6A heteroalkyl group.

86. The compound of claim 85, wherein X is S.

87. The compound of claim 85, wherein X is O.

88. The compound of any one of claims 85 to 87, wherein Y1Is N.

89. The compound of any one of claims 85 to 87, wherein Y1Is CRC1

90. The compound of any one of claims 85 to 89, wherein Y2Is N.

91. The compound of any one of claims 85 to 89, wherein Y2Is CRC1

92. The compound of any one of claims 85 to 91, wherein Y3Is N.

93. The compound of any one of claims 85 to 91, wherein Y3Is CRC1

94. The compound of any one of claims 85 to 93, wherein RC1Is H,

95. The compound of any one of claims 85 to 94, wherein R12Is H,

96. The compound of claim 84, wherein R1Is that

Wherein

f1 is 0, 1, 2, 3 or 4;

f2 is 0, 1, 2 or 3; and is

Each R13Independently is halo, CN, optionally substituted C1-C6Alkyl or optionally substituted C1-C6A heteroalkyl group.

97. The compound of claim 96, wherein R 1Is that

98. The compound of claim 97, wherein f1 is 0.

99. The compound of claim 97, wherein f1 is 1.

100. The compound of claim 99, wherein R1Is that

101. The compound of claim 97, wherein f2 is 0.

102. The compound of claim 84, wherein R1Is that

Wherein

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

RN2is H, optionally substituted C1-C6Alkyl or optionally substituted C1-C6A heteroalkyl group; and is

Each R14Is H, halo, CN, optionally substituted C1-C6Alkyl or optionally substituted C1-C6A heteroalkyl group.

103. The compound of claim 102, wherein R1Is that

104. The compound of claim 102 or 103, wherein g is 0.

105. The compound of claim 84, wherein R1Is that

Wherein

i is 0, 1, 2 or 3;

W1and W2Each of which is independently N or CRC2Wherein R isC2Is H, halo, CN, optionally substituted C1-C6Alkyl or optionally substituted C1-C6A heteroalkyl group;

RN3is H, optionally substituted C1-C6Alkyl, optionally substituted C1-C6Heteroalkyl, optionally substituted C3-C10Carbocyclyl, optionally substituted C2-C9Heterocyclyl or optionally substituted C2-C9A heteroaryl group; and is

Each R15Independently is halo, CN, optionally substituted C1-C6Alkyl or optionally substituted C 1-C6A heteroalkyl group.

106. The compound of claim 105, wherein i is 0.

107. The compound of claim 105 or 106, wherein W1Is N.

108. The compound of claim 105 or 106, wherein W1Is CRC2

109. The compound of any one of claims 105 to 108, wherein W2Is N.

110. The compound of any one of claims 105 to 108, wherein W2Is CRC2

111. The compound of any one of claims 105-110, wherein RN3Is H, optionally substituted C1-C6Alkyl, optionally substituted C1-C6Heteroalkyl or optionally substituted C2-C9Heterocyclic radical。

112. The compound of claim 111, wherein RN3Is that

113. The compound of any one of claims 1 to 112, wherein R1Is F, Cl, Br, I, CN, NO2、NH2

114. The compound of any one of claims 1 to 52, wherein R1And R2Together with the atoms to which each is attached to form optionally substituted C3-C10Carbocyclylene, optionally substituted C2-C9Heterocyclylene radical, optionally substituted C6-C10Arylene or optionally substituted C2-C9A heteroarylene group.

115. The compound of claim 114, wherein R1And R2Together with the atoms to which each is attached to form optionally substituted C 3-C10A carbocyclylene group.

116. The compound of claim 115, wherein the compound has the structure of formula Ib:

or a pharmaceutically acceptable salt thereof.

117. The compound of any one of claims 1 to 116, wherein L1Is optionally substituted C1-C6A heteroalkylene group.

118. The compound of any one of claims 1 to 117, wherein L1Is that

119. The compound of any one of claims 1 to 118, wherein L1Is that

120. A compound having the structure of formula II,

wherein

R1Is H, halo, CN, NO2Hydroxy, optionally substituted amino, optionally substituted C1-C6Alkyl, optionally substituted C1-C6Heteroalkyl, optionally substituted C1-C6Alkenyl, optionally substituted C1-C6Heteroalkenyl, optionally substituted C3-C10Carbocyclyl, optionally substituted C2-C9Heterocyclyl, optionally substituted C6-C10Aryl or optionally substituted C2-C9A heteroaryl group; and R is2Is H or optionally substituted C1-C6An alkyl group; or R1And R2Together with the atoms to which each is attached to form optionally substituted C3-C10Carbocyclylene, optionally substituted C2-C9Heterocyclylene radical, optionally substituted C6-C10Arylene or optionally substituted C2-C9A heteroarylene group;

L1is optionally substituted C1-C6Alkylene or optionally substituted C1-C6A heteroalkylene group; and is

R3Is optionally substituted C 2-C9A heterocyclic group,

or a pharmaceutically acceptable salt thereof.

121. The compound of claim 120, wherein R3Is a heterocyclic group having the structure of formula IIa:

wherein

n is 0, 1, 2, 3, 4, 5 or 6; and is

Each R17Is that

Wherein

Each L2Independently is deletion, O, S, optionally substituted amino, optionally substituted C1-C6Alkyl, optionally substituted C1-C6Heteroalkyl, optionally substituted C3-C10Carbocyclyl or optionally substituted C2-C9A heterocyclic group; and is

Each R18Independently is halo, optionally substituted C1-C6Alkyl, optionally substituted C1-C6Heteroalkyl, optionally substituted C3-C10Carbocyclyl, optionally substituted C2-C9Heterocyclyl, optionally substituted C6-C10Aryl or optionally substituted C2-C9A heteroaryl group.

122. The compound of claim 121, wherein each L is2Independently is deletion, O, S, optionally substituted amino, optionally substituted C1-C6Alkyl or optionally substituted C1-C6A heteroalkyl group.

123. The compound of claim 121 or 122, wherein each R is18Independently is halo, optionally substituted C3-C10Carbocyclyl, optionally substituted C2-C9Heterocyclyl, optionally substituted C6-C10Aryl or optionally substituted C2-C9A heteroaryl group.

124. The compound of any one of claims 121 to 123, wherein each R18Independently is halo, optionally substituted C 6-C10Aryl or optionally substituted C2-C9A heteroaryl group.

125. The compound of any one of claims 121 to 124, wherein each R is18Independently is F, Cl, Br, I or

Wherein

r is 0, 1, 2, 3, 4 or 5; and is

Each R19Independently is halo, CN, hydroxy, optionally substituted C1-C6Alkyl or optionally substituted C1-C6A heteroalkyl group.

126. The compound of any one of claims 121 to 125, wherein q is 0, 1, 2, or 3.

127. The compound of any one of claims 121 to 126, wherein q is 1.

128. Such asThe compound of any one of claims 121 to 127, wherein L2Are missing.

129. The compound of any one of claims 121-127, wherein L2Is O.

130. The compound of any one of claims 121-127, wherein L2Is thatWherein R isN4Is H, optionally substituted C1-C6Alkyl or optionally substituted C1-C6A heteroalkyl group.

131. The compound of claim 130, wherein RN4Is H or-CH3

132. The compound of any one of claims 121-127, wherein L2Is that

133. The compound of any one of claims 121-132, wherein R3Is that

134. The compound of any one of claims 126 to 133, wherein R 17Is that

135. The compound of claim 120, wherein R3Is a heterocyclic group having the structure of formula IIb:

wherein

RgAnd RhTogether with the atoms to which each is attached to form optionally substituted C3-C10Carbocyclylene or optionally substituted C2-C9A heterocyclylene group.

136. The compound of claim 135, wherein R3Is that

Wherein

n is 1, 2, 3 or 4;

s is 0, 1, 2, 3, 4, 5, 6 or 7;

z is O, S,Or NRN5Wherein R isN5Is H, optionally substituted C1-C6Alkyl, optionally substituted C1-C6Heteroalkyl, optionally substituted C3-C10Carbocyclyl, optionally substituted C2-C9Heterocyclyl, optionally substituted C6-C10Aryl or optionally substituted C2-C9A heteroaryl group; and R is21aAnd R21bEach of which is independently H, halo, optionally substituted C1-C6Alkyl, optionally substituted C1-C6Heteroalkyl, optionally substituted C3-C10Carbocyclyl, optionally substituted C2-C9Heterocyclyl, optionally substituted C6-C10Aryl or optionally substituted C2-C9A heteroaryl group; and is

Each R20Independently is halo, optionally substituted C1-C6Alkyl, optionally substituted C1-C6Heteroalkyl, optionally substituted C3-C10Carbocyclyl, optionally substituted C2-C9Heterocyclyl, optionally substituted C6-C10Aryl or optionally substituted C2-C9A heteroaryl group.

137. The compound of claim 136, wherein each R is 20Independently is optionally substituted C1-C6Alkyl, optionally substituted C1-C6Heteroalkyl, optionally substituted C6-C10Aryl or optionally substituted C2-C9A heteroaryl group.

138. The compound of claim 136 or 137, wherein RN5Is H, optionally substituted C1-C6Alkyl, optionally substituted C1-C6Heteroalkyl, optionally substituted C6-C10Aryl or optionally substituted C2-C9A heteroaryl group.

139. The compound of any one of claims 135 to 138, wherein n is 1.

140. The compound of any one of claims 135 to 138, wherein n is 2.

141. The compound of any one of claims 135 to 140, wherein s is 0.

142. The compound of any one of claims 135 to 140, wherein s is 1.

143. The compound of any one of claims 135 to 140, wherein s is 2.

144. The compound of any one of claims 135 to 143, wherein Z is O.

145. The compound of any one of claims 135 to 143, wherein Z is NRN5

146. The compound of any one of claims 135 to 143, wherein Z is

147. The compound of any one of claims 135 to 146, wherein R21aAnd R21bEach of which is independently H, optionally substituted C1-C6Alkyl or optionally substituted C 1-C6A heteroalkyl group.

148. The compound of claim 147, wherein R21aIs H and R21bIs H.

149. The compound of any one of claims 135-148, wherein R20Is that

Wherein

e is 0, 1, 2, 3, 4 or 5; and is

Each R22Independently halogen, CN, NO2Hydroxy, optionally substituted C1-C6Alkyl or optionally substituted C1-C6A heteroalkyl group.

150. The compound of claim 149, wherein e is 0.

151. The compound of claim 149, wherein e is 1.

152. As claimed in claim151, wherein R20Is that

153. The compound of claim 149, wherein e is 2.

154. The compound of claim 153, wherein R20Is that

155. The compound of any one of claims 135-154, wherein R3Is that

156. The compound of any one of claims 135-155, wherein R3Is that

157. The compound of claim 120, wherein R3Is a heterocyclic group having the structure of formula IIc, formula IId, or formula IIe:

wherein

s1 is 0, 1, 2, 3, 4, 5 or 6;

s2 is 0, 1, 2, 3 or 4; and is

Each R23Is that

Wherein

Each L2Independently is deletion, O, S, optionally substituted amino, optionally substituted C1-C6Alkyl, optionally substituted C 1-C6Heteroalkyl, optionally substituted C3-C10Carbocyclyl or optionally substituted C2-C9A heterocyclic group; and is

Each R18Independently is halo, optionally substituted C1-C6Alkyl, optionally substituted C1-C6Heteroalkyl, optionally substituted C3-C10Carbocyclyl, optionally substituted C2-C9Heterocyclyl, optionally substituted C6-C10Aryl or optionally substituted C2-C9A heteroaryl group.

158. The compound of claim 157, wherein each L is2Independently is deletion, O, S, optionally substituted amino, optionally substituted C1-C6Alkyl or optionally substituted C1-C6A heteroalkyl group.

159. The compound of claim 157 or 158, wherein each R is18Independently is halo, optionally substituted C3-C10Carbocyclyl, optionally substituted C2-C9Heterocyclyl, optionally substituted C6-C10Aryl or optionally substituted C2-C9A heteroaryl group.

160. The compound of any one of claims 157 to 159, wherein each R is18Independently is halo, optionally substituted C6-C10Aryl or optionally substituted C2-C9A heteroaryl group.

161. The compound of any one of claims 157 to 160, wherein each R is18Independently F, Cl, Br, I,

Wherein

r1 is 0, 1, 2, 3, 4 or 5;

r2 is 0, 1, 2, 3 or 4; and is

Each R19Independently is halo, hydroxy, optionally substituted C1-C6Alkyl or optionally substituted C 1-C6A heteroalkyl group.

162. The compound of any one of claims 157 to 161, wherein L is2Are missing.

163. The compound of any one of claims 157 to 161, wherein L is2Is O.

164. The compound of any one of claims 157 to 161, wherein L is2Is thatWherein R isN4Is H, optionally substituted C1-C6Alkyl or optionally substituted C1-C6A heteroalkyl group.

165. The compound of claim 164, wherein RN4Is H or-CH3

166. The compound of any one of claims 157 to 161, wherein L is2Is that

167. The compound of any one of claims 157 to 166, wherein s1 is 0 or 1.

168. The compound of any one of claims 157 to 166, wherein s2 is 0 or 1.

169. The compound of any one of claims 157 to 168, wherein R3Is that

170. The compound of any one of claims 157 to 169, wherein R23Is that

171. The compound of claim 120, wherein R3Is a heterocyclic group having the structure of formula IIf:

wherein

RgAnd RhTogether with the atoms to which each is attached to form optionally substituted C3-C10Carbocyclylene or optionally substituted C2-C9A heterocyclylene group.

172. As set forth in claim 171The compound of (1), wherein R gAnd RhTogether with the atoms to which each is attached to form optionally substituted C3-C10A heterocyclylene group.

173. The compound of claim 172, wherein R3Is that

Wherein

n is 1, 2, 3, 4 or 5;

t1 is 0, 1, 2, 3, 4, 5, 6 or 7;

t2 is 0, 1, 2, 3 or 4;

Z1is O, S or NRN5Wherein R isN5Is H, optionally substituted C1-C6Alkyl or optionally substituted C1-C6A heteroalkyl group; and is

Each R20Independently is halo, optionally substituted C1-C6Alkyl, optionally substituted C1-C6Heteroalkyl, optionally substituted C3-C10Carbocyclyl, optionally substituted C2-C9Heterocyclyl, optionally substituted C6-C10Aryl or optionally substituted C2-C9A heteroaryl group.

174. The compound of claim 173, wherein each R is20Independently is halo, optionally substituted C1-C6Alkyl, optionally substituted C1-C6Heteroalkyl, optionally substituted C6-C10Aryl or optionally substituted C2-C9A heteroaryl group.

175. The compound of claim 173 or 174, wherein R isN5Is H, optionally substituted C1-C6Alkyl, optionally substitutedSubstituted C1-C6Heteroalkyl, optionally substituted C6-C10Aryl or optionally substituted C2-C9A heteroaryl group.

176. The compound of any one of claims 173 to 175, wherein n is 2.

177. The compound of any one of claims 173 to 175, wherein n is 3.

178. The compound of any one of claims 173 to 177, wherein t1 is 0.

179. The compound of any one of claims 173 to 177, wherein t1 is 1.

180. The compound of any one of claims 173 to 177, wherein t2 is 1.

181. The compound of any one of claims 173 to 180, wherein Z is1Is O.

182. The compound of any one of claims 173 to 180, wherein R is3Is that

183. The compound of claim 120, wherein R3Is a heterocyclic group having the structure of formula IIg or IIh:

wherein

u1 is 0, 1, 2, 3, 4 or 5;

u2 is 0, 1, 2, 3 or 4;

Z2is O, S or NR24

Wherein

R24Is H, halo, optionally substituted C1-C6Alkyl, optionally substituted C1-C6Heteroalkyl, optionally substituted C3-C10Carbocyclyl, optionally substituted C2-C9Heterocyclyl, optionally substituted C6-C10Aryl or optionally substituted C2-C9A heteroaryl group; and is

Each R23Is that

Wherein

Each L2Independently is deletion, O, S, optionally substituted amino, optionally substituted C1-C6Alkyl, optionally substituted C1-C6Heteroalkyl, optionally substituted C3-C10Carbocyclyl or optionally substituted C2-C9A heterocyclic group; and is

Each R18Independently is halo, optionally substituted C1-C6Alkyl, optionally substituted C 1-C6Heteroalkyl, optionally substituted C3-C10Carbocyclyl, optionally substituted C2-C9Heterocyclyl, optionally substituted C6-C10Aryl or optionally substituted C2-C9A heteroaryl group.

184. The compound of claim 183, wherein each L2Independently is deletion, O, S, optionally substituted amino, optionally substituted C1-C6Alkyl or optionally substituted C1-C6A heteroalkyl group.

185. Such as rightThe compound of claim 183 or 184, wherein each R is18Independently is halo, optionally substituted C3-C10Carbocyclyl, optionally substituted C2-C9Heterocyclyl, optionally substituted C6-C10Aryl or optionally substituted C2-C9A heteroaryl group.

186. The compound of any one of claims 183 to 185, wherein Z is2Is O or NR24

187. The compound of any one of claims 183 to 186, wherein u1 is 0 or 1.

188. The compound of any one of claims 183 to 186, wherein u2 is 0 or 1.

189. The compound of claim 187 or 188, wherein R3Is that

190. The compound of any one of claims 183-189, wherein R24Is that

Wherein

r1 is 0, 1, 2, 3, 4 or 5;

r2 is 0, 1, 2, 3 or 4; and is

Each R19Independently is halo, optionally substituted C1-C6Alkyl or optionally substituted C1-C6A heteroalkyl group.

191. The method of claims 183 to 1 90, wherein R24Is that

192. The compound of claim 120, wherein R3Is a heterocyclic group having the structure of formula Iii, formula IIj or formula IIk:

wherein

v1 is 0, 1, 2, 3, 4, 5 or 6;

v2 is 0, 1, 2, 3 or 4; and is

Each R25Is that

Wherein

Each L2Independently is deletion, O, S, optionally substituted amino, optionally substituted C1-C6Alkyl, optionally substituted C1-C6Heteroalkyl, optionally substituted C3-C10Carbocyclyl or optionally substituted C2-C9A heterocyclic group; and is

Each R18Independently is halo, optionally substituted C1-C6Alkyl, optionally substituted C1-C6Heteroalkyl, optionally substituted C3-C10Carbocyclyl, optionally substituted C2-C9Heterocyclyl, optionally substituted C6-C10Aryl or optionally substituted C2-C9A heteroaryl group.

193. The compound of claim 192, wherein each L2Independently is deletion, O, S, optionally substituted amino, optionally substituted C1-C6Alkyl or optionally substituted C1-C6A heteroalkyl group.

194. The compound of claim 192 or 193, wherein each R18Independently is halo, optionally substituted C3-C10Carbocyclyl, optionally substituted C2-C9Heterocyclyl, optionally substituted C6-C10Aryl or optionally substituted C2-C9A heteroaryl group.

195. The compound of any one of claims 192 to 194, wherein each R is 18Independently is halo, optionally substituted C6-C10Aryl or optionally substituted C2-C9A heteroaryl group.

196. The compound of any one of claims 192-195, wherein each R is18Is F, Cl, Br, I,

Wherein

r1 is 0, 1, 2, 3, 4 or 5;

r2 is 0, 1, 2, 3 or 4; and is

Each R19Independently is halo, optionally substituted C1-C6Alkyl or optionally substituted C1-C6A heteroalkyl group.

197. The compound of any one of claims 192 to 196, wherein v1 is 0 or 1.

198. The compound of any one of claims 192 to 196, wherein v2 is 0 or 1.

199. The compound of any one of claims 192-198, wherein L2Are missing.

200. The compound of any one of claims 192-198, wherein L2Is O.

201. The compound of any one of claims 192-198, wherein L2Is thatWherein R isN4Is H, optionally substituted C1-C6Alkyl or optionally substituted C1-C6A heteroalkyl group.

202. The compound of claim 201, wherein RN4Is H or-CH3

203. The compound of any one of claims 192-198, wherein L2Is that

204. The compound of any one of claims 192-203, wherein R3Is that

205. The compound of any one of claims 157 to 204, wherein R 25Is that

206. The compound of claim 120, wherein R3Is provided withA heterocyclyl group of the structure of formula IIm:

wherein

RgAnd RhTogether with the atoms to which each is attached to form optionally substituted C3-C10Carbocyclylene or optionally substituted C2-C9A heterocyclylene group.

207. The compound of claim 206, wherein RgAnd RhTogether with the atoms to which each is attached to form optionally substituted C3-C10A heterocyclylene group.

208. The compound of claim 207, wherein R3Is thatWherein

n is 1, 2, 3, 4 or 5;

t1 is 0, 1, 2, 3, 4, 5, 6 or 7;

Z1is O, S or NRN5Wherein R isN5Is H, optionally substituted C1-C6Alkyl or optionally substituted C1-C6A heteroalkyl group; and is

Each R20Independently is halo, optionally substituted C1-C6Alkyl, optionally substituted C1-C6Heteroalkyl, optionally substituted C3-C10Carbocyclyl, optionally substituted C2-C9Heterocyclyl, optionally substituted C6-C10Aryl or optionally substituted C2-C9A heteroaryl group.

209. The compound of claim 208, wherein each R20Independently is optionally substituted C1-C6Alkyl, optionally substitutedC of (A)1-C6Heteroalkyl, optionally substituted C6-C10Aryl or optionally substituted C2-C9A heteroaryl group.

210. The compound of claim 208 or 209, wherein RN5Is H, optionally substituted C 1-C6Alkyl, optionally substituted C1-C6Heteroalkyl, optionally substituted C6-C10Aryl or optionally substituted C2-C9A heteroaryl group.

211. The compound of any one of claims 208 to 210, wherein n is 2.

212. The compound of any one of claims 208 to 210, wherein n is 3.

213. The compound of any one of claims 208 to 212, wherein t1 is 0.

214. The compound of any one of claims 208 to 212, wherein t1 is 1.

215. The compound of any one of claims 208 to 212, wherein t1 is 2.

216. The compound of any one of claims 208 to 215, wherein Z is1Is O.

217. The compound of any one of claims 120 to 216, wherein R2Is H or optionally substituted C1-C6An alkyl group.

218. The compound of any one of claims 120 to 217, wherein R2Is H or-CH3

219. Any one of claims 120 to 218The compound of (1), wherein R2Is H.

220. The compound of any one of claims 120 to 219, wherein R1Is H, halo, CN, NO2Hydroxy, optionally substituted amino, optionally substituted C1-C6Alkyl, optionally substituted C1-C6Heteroalkyl, optionally substituted C1-C6Alkenyl, optionally substituted C 1-C6Heteroalkenyl, optionally substituted C3-C10Carbocyclyl, optionally substituted C2-C9Heterocyclyl, optionally substituted C6-C10Aryl or optionally substituted C2-C9A heteroaryl group.

221. The compound of any one of claims 120 to 220, wherein R1Is H, halo, CN, optionally substituted C1-C6Alkyl, optionally substituted C1-C6Heteroalkyl, optionally substituted C1-C6Alkenyl, optionally substituted C3-C10Carbocyclyl, optionally substituted C2-C9Heterocyclyl, optionally substituted C6-C10Aryl or optionally substituted C2-C9A heteroaryl group.

222. The compound of any one of claims 120 to 221, wherein R1Is H, halo, CN, optionally substituted C1-C6Alkyl, optionally substituted C1-C6Heteroalkyl or optionally substituted C1-C6An alkenyl group.

223. The compound of any one of claims 120 to 222, wherein R1Is H, F, Cl, Br, I, CN,

224. The compound of any one of claims 120 to 221, wherein R1Is optionally substituted C3-C10A carbocyclic group.

225. The compound of claim 224, wherein R1Is that

Wherein

c1 is 0, 1, 2, 3, 4 or 5;

c2 is 0, 1, 2, 3 or 4;

c3 is 0, 1, 2, 3, 4, 5 or 6;

c4 is 0, 1, 2, 3, 4 or 5; and is

Each R8Independently is halo, optionally substituted C1-C6Alkyl or optionally substituted C 1-C6A heteroalkyl group.

226. The compound of claim 225, wherein c1 is 0.

227. The compound of claim 225, wherein c2 is 0.

228. The compound of claim 225, wherein c3 is 0.

229. The compound of claim 225, wherein c4 is 0.

230. The compound of any one of claims 120 to 221, wherein R1Is optionally substituted C2-C9A heterocyclic group.

231. The compound of claim 230, wherein R1Is that

Wherein

d1 is 0, 1, 2 or 3;

d2 is 0, 1, 2 or 3;

d3 is 0, 1, 2, 3 or 4;

d4 is 0, 1, 2, 3, 4 or 5;

d5 is 0, 1, 2, 3 or 4;

each R9Independently is halo, hydroxy, optionally substituted C1-C6Alkyl or optionally substituted C1-C6A heteroalkyl group; and is

R10Is H, optionally substituted C1-C6Alkyl or optionally substituted C1-C6A heteroalkyl group.

232. The compound of claim 231, wherein d1 is 0.

233. The compound of claim 231, wherein d2 is 0.

234. The compound of claim 231, wherein d3 is 0.

235. The compound of claim 231, wherein d4 is 0.

236. The compound of claim 231, wherein d5 is 0.

237. The compound of claim 231, wherein d3 is 1.

238. As set forth in claim 237The compound of (1), wherein R9Is hydroxy or optionally substituted C1-C6A heteroalkyl group.

239. The compound of claim 238, wherein R9Is a hydroxyl group,

240. The compound of any one of claims 231 to 239, wherein R10Is H,

241. The compound of any one of claims 120 to 221, wherein R1Is optionally substituted C6-C10And (4) an aryl group.

242. The compound of claim 241, wherein R1Is that

Wherein

e is 0, 1, 2, 3, 4 or 5; and is

Each R11Independently halogen, CN, NO2Hydroxy, optionally substituted C1-C6Alkyl or optionally substituted C1-C6A heteroalkyl group.

243. The compound of claim 242, wherein e is 0.

244. The compound of claim 242, wherein e is 1.

245. The compound of claim 244, wherein R1Is that

246. The compound of claim 242, wherein e is 2.

247. The compound of claim 246, wherein R1Is that

248. The compound of any one of claims 120 to 221, wherein R1Is optionally substituted C2-C9A heteroaryl group.

249. The compound of claim 248, wherein R1Is thatOr

Wherein

X is O, S or NRN1Wherein R isN1Is H, optionally substituted C 1-C6Alkyl or optionally substituted C1-C6A heteroalkyl group;

Y1、Y2and Y3Each of which is independently N or CRC1Wherein R isC1Is H, halogen, CN,Optionally substituted C1-C6Alkyl or optionally substituted C1-C6A heteroalkyl group; and is

R12Is H, halo, CN, optionally substituted C1-C6Alkyl or optionally substituted C1-C6A heteroalkyl group.

250. The compound of claim 249, wherein X is S.

251. The compound of claim 249, wherein X is O.

252. The compound of any one of claims 249-251, wherein Y is Y1Is N.

253. The compound of any one of claims 249-251, wherein Y is Y1Is CRC1

254. The compound of any one of claims 249-253, wherein Y is Y2Is N.

255. The compound of any one of claims 249-253, wherein Y is Y2Is CRC1

256. The compound of any one of claims 249-255, wherein Y is Y3Is N.

257. The compound of any one of claims 249-255, wherein Y is Y3Is CRC1

258. The compound of any one of claims 249-257, wherein RC1Is H,

259. The compound of any one of claims 249-257, wherein R12Is H,

260. The compound of claim 248, wherein R 1Is that

Wherein

f1 is 0, 1, 2, 3 or 4;

f2 is 0, 1, 2 or 3; and is

Each R13Independently is halo, CN, optionally substituted C1-C6Alkyl or optionally substituted C1-C6A heteroalkyl group.

261. The compound of claim 260, wherein R1Is that

262. The compound of claim 261, wherein f1 is 0.

263. The compound of claim 261, wherein f1 is 1.

264. The compound of claim 263, wherein R is1Is that

265. The compound of claim 261, wherein f2 is 0.

266. The compound of claim 248, wherein R1Is that

Wherein

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

RN2is H, optionally substituted C1-C6Alkyl or optionally substituted C1-C6A heteroalkyl group; and is

Each R263Is H, halo, CN, optionally substituted C1-C6Alkyl or optionally substituted C1-C6A heteroalkyl group.

267. The compound of claim 266, wherein R1Is that

268. The compound of claim 266 or 267, wherein g is 0.

269. The compound of claim 248, wherein R1Is that

Wherein

i is 0, 1, 2 or 3;

W1and W2Each of which is independently N or CRC2Wherein R isC2Is H, halo, CN, optionally substituted C1-C6Alkyl or optionally substituted C1-C6A heteroalkyl group;

RN3is H, optionally substituted C 1-C6Alkyl, optionally substituted C1-C6Heteroalkyl, optionally substituted C3-C259Carbocyclyl, optionally substituted C2-C9Heterocyclyl or optionally substituted C2-C9A heteroaryl group; and is

Each R15Independently is halo, CN, optionally substituted C1-C6Alkyl or optionally substituted C1-C6A heteroalkyl group.

270. The compound of claim 269, wherein i is 0.

271. The compound of claim 269 or 270, wherein W1Is N.

272. The compound of claim 269 or 270, wherein W1Is CRC2

273. The compound of any one of claims 269 to 272, wherein W is W2Is N.

274. The compound of any one of claims 269 to 272, wherein W is W2Is CRC2

275. The compound of any one of claims 269 to 274, wherein R isN3Is H, optionally substituted C1-C6Alkyl, optionally substituted C1-C6Heteroalkyl or optionally substituted C2-C9A heterocyclic group.

276. The compound of claim 275, wherein RN3Is that

277. The compound of any one of claims 120 to 276, wherein R1And R2Together with the atoms to which each is attached to form optionally substituted C3-C10Carbocyclylene, optionally substituted C2-C9Heterocyclylene radical, optionally substituted C6-C10Arylene or optionally substituted C2-C9A heteroarylene group.

278. The compound of claim 277, wherein R1And R2Together with the atoms to which each is attached to form optionally substituted C3-C10A carbocyclylene group.

279. The compound of claim 278, wherein the compound has the structure of formula Ib:

or a pharmaceutically acceptable salt thereof.

280. The compound of any one of claims 120 to 279, wherein L1Is optionally substituted C1-C6A heteroalkylene group.

281. The compound of any one of claims 120 to 280, wherein L1Is that

282. The compound of any one of claims 120-281, wherein L1Is that

283. The compound of claim 282, wherein L1Is that

284. A compound having the structure of any one of compounds 1-464 of table 1, or a pharmaceutically acceptable salt thereof.

285. A pharmaceutical composition comprising a compound of any one of claims 1 to 284, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient.

286. A method of treating a neurological disorder in a subject in need thereof, comprising administering an effective amount of a compound of any one of claims 1 to 284, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition of claim 285.

287. A method of inhibiting protein-related cytotoxicity comprising administering an effective amount of a compound of any one of claims 1-284 or a pharmaceutical composition of claim 285.

288. The method of claim 287, wherein the toxicity is α -synuclein-associated toxicity.

289. The method of claim 287, wherein the toxicity is ApoE 4-related toxicity.

290. The method of any one of claims 287-289, wherein the cell is a mammalian neural cell.

291. A method of treating a stearoyl-CoA desaturase (SCD) -associated disorder in a subject in need thereof, comprising administering an effective amount of a compound of any one of claims 1 to 284, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition of claim 285.

292. The method of claim 291, wherein said SCD-related disorder is SCD 5-related disorder.

293. A method of inhibiting SCD5, the method comprising contacting a cell with an effective amount of a compound of any one of claims 1 to 284, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition of claim 285.

294. A method of inhibiting SCD1, the method comprising contacting a cell with an effective amount of a compound of any one of claims 1 to 284, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition of claim 285.

Background

The lack of a thorough understanding of the molecular perturbations that cause Disease, and the limited pool of robust model systems, have led to the failure to produce successful Disease modifying therapies for common and progressive neurological disorders such as Parkinson's Disease (PD) and Alzheimer's Disease (AD). Progress is being made in many ways to find agents that can prevent the progression of these conditions. However, the present therapy provides very little, if any, relief for most, if not all, of these diseases. Thus, there is a need to develop therapies that can alter the progression of neurodegenerative diseases. More generally, there is a need for better methods and compositions for treating neurodegenerative diseases in order to improve the quality of life of people afflicted with such diseases.

Disclosure of Invention

In one aspect, the present disclosure provides compounds having the structure of formula I:

wherein

R1Is H, halo, CN, NO2Hydroxy, optionally substituted amino, optionally substituted C 1-C6Alkyl, optionally substituted C1-C6Heteroalkyl, optionally substituted C1-C6Alkenyl, optionally substituted C1-C6Heteroalkenyl, optionally substituted C3-C10Carbocyclyl, optionally substituted C2-C9Heterocyclyl, optionally substituted C6-C10Aryl or optionally substituted C2-C9(ii) a heteroaryl group, wherein,

R2is H or optionally substituted C1-C6Alkyl, or

R1And R2Together with the atoms to which each is attached to form optionally substituted C3-C10Carbocyclylene, optionally substituted C2-C9Heterocyclylene radical, optionally substituted C6-C10Arylene or optionally substituted C2-C9A heteroarylene group;

L1is optionally substituted C1-C6Alkylene or optionally substituted C1-C6A heteroalkylene group; and is

R3Is optionally substituted C2-C9(ii) a heteroaryl group, wherein,

or a pharmaceutically acceptable salt thereof.

In some embodiments, R3Is a heteroaryl having the structure of formula Ia:

wherein

X1、X2、X3And X4Is independently O, NR4Or CR5

Wherein

Each R4Independently is H, optionally substituted C1-C6Alkyl, optionally substituted C1-C6Heteroalkyl, optionally substituted C3-C10Carbocyclyl, optionally substituted C2-C9Heterocyclyl, optionally substituted C6-C10Aryl or optionally substituted C2-C9(ii) a heteroaryl group, wherein,

each R5Independently H, CN, optionally substituted C1-C6Alkyl or optionally substituted C1-C6A heteroalkyl group, and

if X is1、X2、X3Or X4Is O, then the adjacent atom is N or CR 5(ii) a And X5Is N or C, whereinX1、X2、X3、X4Or X51, 2 or 3 of (A) is O or N.

In some embodiments, R3Is that

In some embodiments, R3Is that

In some embodiments, R3Is that

In some embodiments, each R is5Independently H, CN or optionally substituted C1-C6An alkyl group.

In some embodiments, each R is5Independently H, CN,

Wherein

j is 0, 1, 2, 3, 4 or 5; and is

Each R6Independently is halo, CN, optionally substituted C1-C6Alkyl or optionally substituted C1-C6A heteroalkyl group.

In some embodiments, each R is6Independently F, Cl, Br, I, CN,

In some embodiments, j is 0, 1, 2, or 3.

In some embodiments, j is 2.

In some embodiments, each R is5Independently H, CN,

In some embodiments, R4Is H.

In some embodiments, R4Is optionally substituted C1-C6Alkyl, optionally substituted C1-C6Heteroalkyl, optionally substituted C3-C10Carbocyclyl, optionally substituted C2-C9Heterocyclyl, optionally substituted C6-C10Aryl or optionally substituted C2-C9A heteroaryl group.

In some embodiments, R4Is optionally substituted C1-C6A heteroalkyl group.

In some embodiments, R4Is that

In some embodiments, R4Is optionally substituted C 3-C10Carbocyclyl, optionally substituted C2-C9Heterocyclyl, optionally substituted C6-C10Aryl or optionally substituted C2-C9A heteroaryl group.

In some embodiments, R4Is that

In some embodiments, R4Is optionally substituted C1-C6An alkyl group.

In some embodiments, R4Is that

In some embodiments, R4Is that

Wherein

k1 is 0, 1, 2, 3, 4 or 5;

k2 is 0, 1, 2, 3 or 4;

k3 is 0, 1, 2 or 3;

w is O or S;

m is 1 or 2; and is

Each R7Independently halogen, CN, NO2Hydroxy, optionally substituted C1-C6Alkyl or optionally substituted C1-C6A heteroalkyl group.

In some embodiments, m is 2. In some embodiments, m is 1.

In some embodiments, each R is7Independently is halo, CN, optionally substituted C1-C6Alkyl or optionally substituted C1-C6A heteroalkyl group.

In some embodiments, each R is7Independently F, Cl, Br, I, CN, -CF3

In some embodiments, k1 is 0. In some embodiments, k1 is 1.

In some embodiments, R4Is that

In some embodiments, k1 is 2.

In some embodiments, R4Is that

In some embodiments, k2 is 0. In some embodiments, k2 is 1.

In some embodiments, R4Is that

In some embodiments, k3 is 0. In some embodiments, k3 is 1.

In some embodiments, R4Is that

In some embodiments, W is S.

In some embodiments, R4Is that

In some embodiments, R4Is that Wherein m is 1 or 2.

In some embodiments, m is 1. In some embodiments, m is 2.

In some embodiments, R4Is that

In some embodiments, R4Is that

Wherein

k1 is 0, 1, 2, 3, 4 or 5;

each R7Independently halogen, CN, NO2Hydroxy, optionally substituted C1-C6Alkyl or optionally substituted C1-C6A heteroalkyl group;

Rais H, optionally substituted C1-C6Alkyl or optionally substituted C1-C6A heteroalkyl group; and is

RbIs optionally substituted C1-C6Alkyl or optionally substituted C1-C6A heteroalkyl group.

In some embodiments, each R is7Independently is halo, CN, optionally substituted C1-C6Alkyl or optionally substituted C1-C6A heteroalkyl group.

In some embodiments, each R is7Independently F, Cl, Br, I, CN, -CF3

In some embodiments, k1 is 0. In some embodiments, k1 is 1.

In some embodiments, R4Is that

In some embodiments, k1 is 2.

In some embodiments, R4Is that

In some embodiments, RaIs H or-CH3

In some embodiments, RbIs optionally substituted C1-C6An alkyl group.

In some embodiments, RbIs that

Wherein

b is 1 or 2; and is

RcIs optionally substituted amino, optionally substituted mercapto, optionally substituted sulfone or optionally substituted sulfoxide.

In some embodiments, RcIs that

Wherein

Rd、ReAnd RfEach of which is independently H, optionally substituted C1-C6Alkyl or optionally substituted C1-C6An alkyl group.

In some embodiments, R4Is that

Or in some embodiments, R2Is H or optionally substituted C1-C6An alkyl group.

In some embodiments, R2Is H,

In some embodiments, R2Is H.

In some embodiments, R1Is H, halo, CN, NO2Hydroxy, optionally substituted amino, optionally substituted C1-C6Alkyl, optionally substituted C1-C6Heteroalkyl, optionally substituted C1-C6Alkenyl, optionally substituted C1-C6Heteroalkenyl, optionally substituted C3-C10Carbocyclyl, optionally substituted C2-C9Heterocyclyl, optionally substituted C6-C10Aryl or optionally substituted C2-C9A heteroaryl group.

In some embodiments, R1Is H, halo, CN, optionally substituted C1-C6Alkyl, optionally substituted C1-C6Heteroalkyl, optionally substituted C1-C6Alkenyl, optionally substituted C3-C10Carbocyclyl, optionally substituted C2-C9Heterocyclyl, optionally substituted C6-C10Aryl or optionally substituted C2-C9A heteroaryl group.

In some embodiments, R 1Is H, halo, CN, optionally substituted C1-C6Alkyl, optionally substituted C1-C6Heteroalkyl or optionally substituted C1-C6An alkenyl group.

In some embodiments, R1Is H, F, Cl, Br, I, CN,

In some embodiments, R1Is optionally substituted C3-C10A carbocyclic group.

In some embodiments, R1Is that

Wherein

c1 is 0, 1, 2, 3, 4 or 5;

c2 is 0, 1, 2, 3 or 4;

c3 is 0, 1, 2, 3, 4, 5 or 6;

c4 is 0, 1, 2, 3, 4 or 5; and is

Each R8Independently is halo, optionally substituted C1-C6Alkyl or optionally substituted C1-C6A heteroalkyl group.

In some embodiments, c1 is 0.

In some embodiments, c2 is 0.

In some embodiments, c3 is 0.

In some embodiments, c4 is 0.

In some embodiments, R1Is optionally substituted C2-C9A heterocyclic group.

In some embodiments, R1Is that

Wherein

d1 is 0, 1, 2 or 3;

d2 is 0, 1, 2 or 3;

d3 is 0, 1, 2, 3 or 4;

d4 is 0, 1, 2, 3, 4 or 5;

d5 is 0, 1, 2, 3 or 4;

each R9Independently is halo, hydroxy, optionally substituted C1-C6Alkyl or optionally substituted C1-C6A heteroalkyl group; and is

R10Is H, optionally substituted C1-C6Alkyl or optionally substituted C1-C6A heteroalkyl group.

In some embodiments, d1 is 0 or 1. In some embodiments, d1 is 0. In some embodiments, d1 is 1.

In some embodiments, d2 is 0, 1, or 2. In some embodiments, d2 is 0. In some embodiments, d2 is 1.

In some embodiments, d3 is 0, 1, or 2. In some embodiments, d3 is 0. In some embodiments, d3 is 1.

In some embodiments, d4 is 0, 1, or 2. In some embodiments, d4 is 0. In some embodiments, d4 is 1.

In some embodiments, d5 is 0, 1, or 2. In some embodiments, d5 is 0. In some embodiments, d5 is 1.

In some embodiments, R9Is hydroxy or optionally substituted C1-C6A heteroalkyl group.

In some embodiments, R9Is a hydroxyl group,

In some embodiments,R10Is H,

In some embodiments, R1Is optionally substituted C6-C10And (4) an aryl group.

In some embodiments, R1Is that

Wherein

e is 0, 1, 2, 3, 4 or 5; and is

Each R11Independently halogen, CN, NO2Hydroxy, optionally substituted C1-C6Alkyl or optionally substituted C1-C6A heteroalkyl group.

In some embodiments, e is 0. In some embodiments, e is 1.

In some embodiments, R1Is that

In some embodiments, e is 2.

In some embodiments, R1Is that

In some embodiments, R1Is optionally substituted C2-C9A heteroaryl group.

In some embodiments, R1Is that

Wherein

X is O, S or NRN1Wherein R isN1Is H, optionally substitutedC of (A)1-C6Alkyl or optionally substituted C1-C6A heteroalkyl group;

Y1、Y2and Y3Each of which is independently N or CRC1Wherein R isC1Is H, halo, CN, optionally substituted C1-C6Alkyl or optionally substituted C1-C6A heteroalkyl group; and R is12Is H, halo, CN, optionally substituted C1-C6Alkyl or optionally substituted C1-C6A heteroalkyl group.

In some embodiments, X is S. In some embodiments, X is O.

In some embodiments, Y is1Is N. In some embodiments, Y is1Is CRC1

In some embodiments, Y is2Is N. In some embodiments, Y is2Is CRC1

In some embodiments, Y is3Is N. In some embodiments, Y is3Is CRC1

In some embodiments, RC1Is H,

In some embodiments, R12Is H,

In some embodiments, R1Is that

Wherein

f1 is 0, 1, 2, 3 or 4;

f2 is 0, 1, 2 or 3; and is

Each R13Independently is halo, CN, optionally substituted C1-C6Alkyl or optionally substituted C1-C6A heteroalkyl group.

In some embodiments, R1Is that

In some embodiments, f1 is 0. In some embodiments, f1 is 1.

In some embodiments, R1Is that

In some embodiments, f2 is 0.

In some embodiments, R1Is that

Wherein

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

RN2is H, optionally substituted C1-C6Alkyl or optionally substituted C1-C6A heteroalkyl group; and is

Each R14Is H, halo, CN, optionally substituted C1-C6Alkyl or optionally substituted C1-C6A heteroalkyl group.

In some embodiments, R1Is that

In some embodiments, g is 0.

In some embodiments, R1Is that

Wherein

i is 0, 1, 2 or 3;

W1and W2Each of which is independently N or CRC2Wherein R isC2Is H, halo, CN, optionally substituted C1-C6Alkyl or optionally substituted C1-C6A heteroalkyl group;

RN3is H, optionally substituted C1-C6Alkyl, optionally substituted C1-C6Heteroalkyl, optionally substituted C3-C10Carbocyclyl, optionally substituted C2-C9Heterocyclyl or optionally substituted C2-C9A heteroaryl group; and is

Each R15Independently is halo, CN, optionally substituted C1-C6Alkyl or optionally substituted C1-C6A heteroalkyl group.

In some embodiments, i is 0.

In some embodiments, W1Is N. In some embodiments, W1Is CRC2

In some embodiments, W2Is N. In some embodiments, W2Is CRC2

In some embodiments, RN3Is H, optionally substituted C1-C6Alkyl, optionally substituted C 1-C6Heteroalkyl or optionally substituted C2-C9A heterocyclic group.

In some embodiments, RN3Is that

In some embodiments, R1Is F, Cl, Br, I, CN, NO2、NH2

In some embodiments, R1And R2Together with the atoms to which each is attached to form optionally substituted C3-C10Carbocyclylene, optionally substituted C2-C9Heterocyclylene radical, optionally substituted C6-C10Arylene or optionally substituted C2-C9A heteroarylene group.

In some embodiments, R1And R2Together with the atoms to which each is attached to form optionally substituted C3-C10A carbocyclylene group.

In some embodiments, the compound has the structure of formula Ib:

or a pharmaceutically acceptable salt thereof.

In some embodiments, L is1Is optionally substituted C1-C6A heteroalkylene group.

In some embodiments, L is1Is that

In some embodiments, L is1Is that

In one aspect, the present disclosure provides a compound having the structure of formula II:

wherein

R1Is H, halo, CN, NO2Hydroxy, optionally substituted amino, optionally substituted C1-C6Alkyl, optionally substituted C1-C6Heteroalkyl, optionally substituted C1-C6Alkenyl, optionally substituted C1-C6Heteroalkenyl, optionally substituted C3-C10Carbocyclyl, optionally substituted C2-C9Heterocyclyl, optionally substituted C6-C10Aryl or optionally substituted C 2-C9A heteroaryl group; and is

R2Is H or optionally substituted C1-C6An alkyl group; or

R1And R2Together with the atoms to which each is attached to form optionally substituted C3-C10Carbocyclylene, optionally substituted C2-C9Heterocyclylene radical, optionally substituted C6-C10Arylene or optionally substituted C2-C9A heteroarylene group;

L1is optionally substituted C1-C6Alkylene or optionally substituted C1-C6A heteroalkylene group; and is

R3Is optionally substituted C2-C9A heterocyclic group,

or a pharmaceutically acceptable salt thereof.

In some embodiments, R3Is a heterocyclic group having the structure of formula IIa:

wherein

n is 0, 1, 2, 3, 4, 5 or 6; and is

Each R17Is that

Wherein

Each L2Independently is deletion, O, S, optionally substituted amino, optionally substituted C1-C6Alkyl, optionally substituted C1-C6Heteroalkyl, optionally substituted C3-C10Carbocyclyl or optionally substituted C2-C9A heterocyclic group; and is

Each R18Independently is halo, optionally substituted C1-C6Alkyl, optionally substituted C1-C6Heteroalkyl, optionally substituted C3-C10Carbocyclyl, optionally substituted C2-C9Heterocyclyl, optionally substituted C6-C10Aryl or optionally substituted C2-C9A heteroaryl group.

In some embodiments, each L is2Independently is deletion, O, S, optionally substituted amino, optionally substituted C1-C6Alkyl or optionally substituted C1-C6A heteroalkyl group.

In some embodiments, each R is18Independently is halo, optionally substituted C3-C10Carbocyclyl, optionally substituted C2-C9Heterocyclyl, optionally substituted C6-C10Aryl or optionally substituted C2-C9A heteroaryl group.

In some embodiments, each R is18Independently is halo, optionally substituted C6-C10Aryl or optionally substituted C2-C9A heteroaryl group.

In some embodiments, each R is18Independently is F, Cl, Br, I or

Wherein

r is 0, 1, 2, 3, 4 or 5; and is

Each R19Independently is halo, CN, hydroxy, optionally substituted C1-C6Alkyl or optionally substituted C1-C6A heteroalkyl group.

In some embodiments, each R is19Independently F, Cl, Br, I, CN, hydroxy,

In some embodiments, r is 0, 1, or 2. In some embodiments, r is 0. In some embodiments, r is 1. In some embodiments, r is 2.

In some embodiments, R18Is that

In some embodiments, R18Is that

In some embodiments, q is 0, 1, 2, or 3. In some embodiments, q is 1.

In some embodiments, L is2Is absent. In some embodiments, L is2Is O. In some embodiments, L is2Is thatWherein R isN4Is H, optionally substituted C1-C6Alkyl or optionally substituted C 1-C6A heteroalkyl group. In some embodiments, L is2Is that

In some embodiments, RN4Is H or-CH3

In some embodiments, R3Is that

In some embodiments, R17Is that

In some embodiments, R3Is a heterocyclic group having the structure of formula IIb:

wherein

RgAnd RhTogether with the atoms to which each is attached to form optionally substituted C3-C10Carbocyclylene or optionally substituted C2-C9A heterocyclylene group.

In some embodiments, RgAnd RhTogether with the atoms to which each is attached to form optionally substituted C3-C10A carbocyclylene group.

In some embodiments, RgAnd RhTogether with the atoms to which each is attached to form optionally substituted C2-C9A heterocyclylene group.

In some embodiments, R3Is that

Wherein

n is 1, 2, 3 or 4;

s is 0, 1, 2, 3, 4, 5, 6 or 7;

z is O, S,Or NRN5Wherein R isN5Is H, optionally substituted C1-C6Alkyl, optionally substituted C1-C6Heteroalkyl, optionally substituted C3-C10Carbocyclyl, optionally substituted C2-C9Heterocyclyl, optionally substituted C6-C10Aryl or optionally substituted C2-C9A heteroaryl group; and R is21aAnd R21bEach of which is independently H, halo, optionally substituted C1-C6Alkyl, optionally substituted C1-C6Heteroalkyl, optionally substituted C3-C10Carbocyclyl, optionally substituted C 2-C9Heterocyclyl, optionally substituted C6-C10Aryl or optionally substituted C2-C9A heteroaryl group; and is

Each R20Independently is halo, optionally substituted C1-C6Alkyl, optionally substituted C1-C6Heteroalkyl, optionally substituted C3-C10Carbocyclyl, optionally substituted C2-C9Heterocyclyl, optionally substituted C6-C10Aryl or optionally substituted C2-C9A heteroaryl group.

In some embodiments, each R is20Independently is optionally substituted C1-C6Alkyl, optionally substituted C1-C6Heteroalkyl, optionally substituted C6-C10Aryl or optionally substituted C2-C9A heteroaryl group.

In some embodiments, RN5Is H, optionally substituted C1-C6Alkyl, optionally substituted C1-C6Heteroalkyl, optionally substituted C6-C10Aryl or optionally substituted C2-C9A heteroaryl group.

In some embodiments, n is 1. In some embodiments, n is 2.

In some embodiments, s is 0. In some embodiments, s is 1. In some embodiments, s is 2.

In some embodiments, Z is O. In some embodiments, NRN5. In some embodiments, Z is

In some embodiments, R21aAnd R21bEach of which is independently H, optionally substituted C1-C6Alkyl or optionally substituted C1-C6A heteroalkyl group.

In some embodiments, R21aIs H.

In some embodiments, R 21bIs H.

In some embodiments, R21aIs H and R21bIs H.

In some embodiments, R20Is that

Wherein

e is 0, 1, 2, 3, 4 or 5; and is

Each R22Independently halogen, CN, NO2A hydroxyl groupRadical, optionally substituted C1-C6Alkyl or optionally substituted C1-C6A heteroalkyl group.

In some embodiments, e is 0. In some embodiments, e is 1.

In some embodiments, R20Is that

In some embodiments, e is 2.

In some embodiments, R20Is that

In some embodiments, R3Is that

In some embodiments, R3Is that

In some embodiments, R3Is a heterocyclic group having the structure of formula IIc, formula IId, or formula IIe:

wherein

s1 is 0, 1, 2, 3, 4, 5 or 6;

s2 is 0, 1, 2, 3 or 4; and is

Each R23Is that

Wherein

Each L2Independently is deletion, O, S, optionally substituted amino, optionally substituted C1-C6Alkyl, optionally substituted C1-C6Heteroalkyl, optionally substituted C3-C10Carbocyclyl or optionally substituted C2-C9A heterocyclic group; and is

Each R18Independently is halo, optionally substituted C1-C6Alkyl, optionally substituted C1-C6Heteroalkyl, optionally substituted C3-C10Carbocyclyl, optionally substituted C2-C9Heterocyclyl, optionally substituted C6-C10Aryl or optionally substituted C2-C9A heteroaryl group.

In some embodiments, each L is2Independently is deletion, O, S, optionally substituted amino, optionally substituted C1-C6Alkyl or optionally substituted C1-C6A heteroalkyl group.

In some embodiments, each R is18Independently is halo, optionally substituted C3-C10Carbocyclyl, optionally substituted C2-C9Heterocyclyl, optionally substituted C6-C10Aryl or optionally substituted C2-C9A heteroaryl group.

In some embodiments, each R is18Independently F, Cl, Br, I, Wherein

r1 is 0, 1, 2, 3, 4 or 5;

r2 is 0, 1, 2, 3 or 4; and is

Each R19Independently is halo, hydroxy, optionally substituted C1-C6Alkyl or optionally substituted C1-C6A heteroalkyl group.

In some embodiments, each R is19Independently F, Cl, Br, I, CN, hydroxy,

In some embodiments, r1 is 0, 1, or 2. In some embodiments, r1 is 0. In some embodiments, r1 is 1. In some embodiments, r1 is 2.

In some embodiments, R18Is that

In some embodiments, R18Is that

In some embodiments, r2 is 0, 1, or 2. In some embodiments, r2 is 0. In some embodiments, r2 is 1.

In some embodiments, R18Is that

In some embodiments, R18Is that

In some embodiments, L is2Are missing. In some embodiments, L is2Is O. In some embodiments, L is2Is thatWherein R isN4Is H, optionally substituted C1-C6Alkyl or optionally substituted C1-C6A heteroalkyl group. In some embodiments, L is2Is that

In some embodiments, RN4Is H or-CH3

In some embodiments, s1 is 0 or 1. In some embodiments, s1 is 0. In some embodiments, s1 is 1.

In some embodiments, s2 is 0 or 1. In some embodiments, s2 is 0. In some embodiments, s2 is 1.

In some embodiments, R3Is that

In some embodiments, R23Is that

In some embodiments, R3Is a heterocyclic group having the structure of formula IIf:

wherein

RgAnd RhTogether with the atoms to which each is attached to form optionally substituted C3-C10Carbocyclylene or optionally substituted C2-C9A heterocyclylene group.

In some embodiments, RgAnd RhTogether with the atoms to which each is attached to form optionally substituted C3-C10A heterocyclylene group.

In some embodiments, R3Is that

Wherein

n is 1, 2, 3, 4 or 5;

t1 is 0, 1, 2, 3, 4, 5, 6 or 7;

t2 is 0, 1, 2, 3 or 4;

Z1is O, S or NRN5Wherein R isN5Is H, optionally substituted C 1-C6Alkyl or optionally substituted C1-C6A heteroalkyl group; and is

Each R20Independently is halo, optionally substituted C1-C6Alkyl, optionally substituted C1-C6Heteroalkyl, optionally substituted C3-C10Carbocyclyl, optionally substituted C2-C9Heterocyclyl, optionally substituted C6-C10Aryl or optionally substituted C2-C9A heteroaryl group.

In some embodiments, each R is20Independently is halo, optionally substituted C1-C6Alkyl, optionally substituted C1-C6Heteroalkyl, optionally substituted C6-C10Aryl or optionally substituted C2-C9A heteroaryl group.

In some embodiments, each R is20Is F, Cl, Br, I, CN, hydroxyl,

In some embodiments, each R is20Is F, Cl, Br, I or CN.

In some embodiments, RN5Is H, optionally substituted C1-C6Alkyl, optionally substituted C1-C6Heteroalkyl, optionally substituted C6-C10Aryl or optionally substituted C2-C9A heteroaryl group.

In some embodiments, n is 1, 2, or 3.

In some embodiments, n is 1. In some embodiments, n is 2. In some embodiments, n is 3.

In some embodiments, t1 is 0 or 1. In some embodiments, t1 is 0. In some embodiments, t1 is 1.

In some embodiments, t2 is 0 or 1. In some embodiments, t2 is 0. In some embodiments, t2 is 1.

In some embodiments, Z1Is O.

In some embodiments, R3Is that

In some embodiments, R3Is a heterocyclic group having the structure of formula IIg or IIh:

wherein

u1 is 0, 1, 2, 3, 4 or 5;

u2 is 0, 1, 2, 3 or 4;

Z2is O, S or NR24

Wherein

R24Is H, halo, optionally substituted C1-C6Alkyl, optionally substituted C1-C6Heteroalkyl, optionally substituted C3-C10Carbocyclyl, optionally substituted C2-C9Heterocyclyl, optionally substituted C6-C10Aryl or optionally substituted C2-C9A heteroaryl group; and is

Each R23Is that

Wherein

Each L2Independently is deletion, O, S, optionally substituted amino, optionally substituted C1-C6Alkyl, optionally substituted C1-C6Heteroalkyl, optionally substituted C3-C10Carbocyclyl or optionally substituted C2-C9A heterocyclic group; and is

Each R18Independently is halo, optionally substituted C1-C6Alkyl, optionally substituted C1-C6Heteroalkyl, optionally substituted C3-C10Carbocyclyl, optionally substituted C2-C9Heterocyclyl, optionally substituted C6-C10Aryl or optionally substituted C2-C9A heteroaryl group.

In some embodiments, each L is2Independently is deletion, O, S, optionally substituted amino, optionally substituted C1-C6Alkyl or optionally substituted C1-C6A heteroalkyl group.

In some embodiments, each R is18Independently is halo, optionally substituted C 3-C10Carbocyclyl, optionally substituted C2-C9Heterocyclyl, optionally substituted C6-C10Aryl or optionally substituted C2-C9A heteroaryl group.

In some embodiments, Z2Is O or NR24. In some embodiments, Z2Is O. In some embodiments, Z2Is NR24

In some embodiments, u1 is 0 or 1. In some embodiments, u1 is 0. In some embodiments, u1 is 1.

In some embodiments, u2 is 0 or 1. In some embodiments, u2 is 0. In some embodiments, u2 is 1.

In some embodiments, R3Is that

In some embodiments, R24Is that

Wherein

r1 is 0, 1, 2, 3, 4 or 5;

r2 is 0, 1, 2, 3 or 4; and is

Each R19Independently is halo, optionally substituted C1-C6Alkyl or optionally substituted C1-C6A heteroalkyl group.

In some embodiments, each R is19Independently F, Cl, Br, I, CN, hydroxy,

In some embodiments, r1 is 0, 1, or 2. In some embodiments, r1 is 0. In some embodiments, r1 is 1. In some embodiments, r1 is 2.

In some embodiments, R24Is that

In some embodiments, R24Is that

In some embodiments, r2 is 0, 1, or 2. In some embodiments, r2 is 0. In some embodiments, r2 is 1.

In some embodiments, R24Is that

In some embodiments, R24Is that

In some embodiments, R3Is a heterocyclic group having the structure of formula Iii, formula IIj or formula IIk:

wherein

v1 is 0, 1, 2, 3, 4, 5 or 6;

v2 is 0, 1, 2, 3 or 4; and is

Each R25Is that

Wherein

Each L2Independently is deletion, O, S, optionally substituted amino, optionally substituted C1-C6Alkyl, optionally substituted C1-C6Heteroalkyl, optionally substituted C3-C10Carbocyclyl or optionally substituted C2-C9A heterocyclic group; and is

Each R18Independently is halo, optionally substituted C1-C6Alkyl, optionally substituted C1-C6Heteroalkyl, optionally substituted C3-C10Carbocyclyl, optionally substituted C2-C9Heterocyclyl, optionally substituted C6-C10Aryl or optionally substituted C2-C9A heteroaryl group.

In some embodiments, L is2Independently is deletion, O, S, optionally substituted amino, optionally substituted C1-C6Alkyl or optionally substituted C1-C6A heteroalkyl group.

In some embodiments, each R is18Independently is halo, optionally substituted C3-C10Carbocyclyl, optionally substituted C2-C9Heterocyclyl, optionally substituted C6-C10Aryl or optionally substituted C2-C9A heteroaryl group.

In some embodiments, each R is18Independently is halo, optionally substituted C6-C10Aryl or optionally substituted C 2-C9A heteroaryl group.

In some embodiments, each R is18Is F, Cl, Br, I,

Wherein

r1 is 0, 1, 2, 3, 4 or 5;

r2 is 0, 1, 2, 3 or 4; and is

Each R19Independently is halo, optionally substituted C1-C6Alkyl or optionally substituted C1-C6A heteroalkyl group.

In some embodiments, each R is19Independently F, Cl, Br, I, CN, hydroxy,

In some embodiments, r is 0, 1, or 2. In some embodiments, r is 0. In some embodiments, r is 1. In some embodiments, r is 2.

In some embodiments, R18Is that

In some embodiments, R18Is that

In some embodiments, v1 is 0 or 1. In some embodiments, v1 is 0. In some embodiments, v1 is 1.

In some embodiments, v2 is 0 or 1. In some embodiments, v2 is 0. In some embodiments, v2 is 1.

In some embodiments, L is2Are missing.

In some embodiments, L is2Is O.

In some embodiments, L is2Is that Wherein R isN4Is H, optionally substituted C1-C6Alkyl or optionally substituted C1-C6A heteroalkyl group.

In some embodiments, RN4Is H or-CH3

In some embodiments, L is2Is that

In some embodiments, R 3Is that

In some embodiments, R25Is that

In some embodiments, R3Is a heterocyclic group having the structure of formula IIm:

wherein

RgAnd RhTogether with the atoms to which each is attached to form optionally substituted C3-C10Carbocyclylene or optionally substituted C2-C9A heterocyclylene group.

In some embodiments, RgAnd RhTogether with the atoms to which each is attached to form optionally substituted C3-C10A heterocyclylene group.

In some embodiments, R3Is that

Wherein

n is 1, 2, 3, 4 or 5;

t1 is 0, 1, 2, 3, 4, 5, 6 or 7;

Z1is O, S or NRN5Wherein R isN5Is H, optionally substituted C1-C6Alkyl radicalOr optionally substituted C1-C6A heteroalkyl group; and is

Each R20Independently is halo, optionally substituted C1-C6Alkyl, optionally substituted C1-C6Heteroalkyl, optionally substituted C3-C10Carbocyclyl, optionally substituted C2-C9Heterocyclyl, optionally substituted C6-C10Aryl or optionally substituted C2-C9A heteroaryl group.

In some embodiments, each R is20Independently is optionally substituted C1-C6Alkyl, optionally substituted C1-C6Heteroalkyl, optionally substituted C6-C10Aryl or optionally substituted C2-C9A heteroaryl group.

In some embodiments, RN5Is H, optionally substituted C1-C6Alkyl, optionally substituted C1-C6Heteroalkyl, optionally substituted C6-C10Aryl or optionally substituted C 2-C9A heteroaryl group.

In some embodiments, n is 1, 2, or 3. In some embodiments, n is 2. In some embodiments, n is 3.

In some embodiments, t1 is 0, 1, or 2. In some embodiments, t1 is 1. In some embodiments, t1 is 2.

In some embodiments, Z1Is O.

Or in some embodiments, R2Is H or optionally substituted C1-C6An alkyl group.

In some embodiments, R2Is H or-CH3

In some embodiments, R2Is H.

In some embodiments, R1Is H, halo, CN, NO2Hydroxy, optionally substituted amino, optionally substituted C1-C6Alkyl, optionally substituted C1-C6Heteroalkyl, optionally substituted C1-C6Alkenyl, optionally substituted C1-C6Heteroalkenyl, optionally substituted C3-C10Carbocyclyl, optionally substituted C2-C9Heterocyclyl, optionally substituted C6-C10Aryl or optionally substituted C2-C9A heteroaryl group.

In some embodiments, R1Is H, halo, CN, optionally substituted C1-C6Alkyl, optionally substituted C1-C6Heteroalkyl, optionally substituted C1-C6Alkenyl, optionally substituted C3-C10Carbocyclyl, optionally substituted C2-C9Heterocyclyl, optionally substituted C6-C10Aryl or optionally substituted C2-C9A heteroaryl group.

In some embodiments, R1Is H, halo, CN, optionally substituted C 1-C6Alkyl, optionally substituted C1-C6Heteroalkyl or optionally substituted C1-C6An alkenyl group.

In some embodiments, R1Is H, F, Cl, Br, I, CN,

In some embodiments, R1Is optionally substituted C3-C10A carbocyclic group.

In some embodiments, R1Is that

Wherein

c1 is 0, 1, 2, 3, 4 or 5;

c2 is 0, 1, 2, 3 or 4;

c3 is 0, 1, 2, 3, 4, 5 or 6;

c4 is 0, 1, 2, 3, 4 or 5; and is

Each R8Independently is halo, optionally substituted C1-C6Alkyl or optionally substituted C1-C6A heteroalkyl group.

In some embodiments, c1 is 0.

In some embodiments, c2 is 0.

In some embodiments, c3 is 0.

In some embodiments, c4 is 0.

In some embodiments, R1Is optionally substituted C2-C9A heterocyclic group.

In some embodiments, R1Is that

Wherein

d1 is 0, 1, 2 or 3;

d2 is 0, 1, 2 or 3;

d3 is 0, 1, 2, 3 or 4;

d4 is 0, 1, 2, 3, 4 or 5;

d5 is 0, 1, 2, 3 or 4;

each R9Independently is halo, hydroxy, optionally substituted C1-C6Alkyl or optionally substituted C1-C6A heteroalkyl group; and is

R10Is H, optionally substituted C1-C6Alkyl or optionally substituted C1-C6A heteroalkyl group.

In some embodiments, d1 is 0 or 1. In some embodiments, d1 is 0. In some embodiments, d1 is 1.

In some embodiments, d2 is 0, 1, or 2. In some embodiments, d2 is 0. In some embodiments, d2 is 1.

In some embodiments, d3 is 0, 1, or 2. In some embodiments, d3 is 0. In some embodiments, d3 is 1.

In some embodiments, d4 is 0, 1, or 2. In some embodiments, d4 is 0. In some embodiments, d4 is 1.

In some embodiments, d5 is 0, 1, or 2. In some embodiments, d5 is 0. In some embodiments, d5 is 1.

In some embodiments, R9Is hydroxy or optionally substituted C1-C6A heteroalkyl group.

In some embodiments, R9Is a hydroxyl group,

In some embodiments, R10Is H,

In some embodiments, R1Is optionally substituted C6-C10And (4) an aryl group.

In some embodiments, R1Is that

Wherein

e is 0, 1, 2, 3, 4 or 5; and is

Each R11Independently halogen, CN, NO2Hydroxy, optionally substituted C1-C6Alkyl or optionally substituted C1-C6A heteroalkyl group.

In some embodiments, e is 0. In some embodiments, e is 1.

In some embodiments, R1Is that

In some embodiments, e is 2.

In some embodiments, R 1Is that

In some embodiments, R1Is optionally substituted C2-C9A heteroaryl group.

In some embodiments, R1Is that

Wherein

X is O, S or NRN1Wherein R isN1Is H, optionally substituted C1-C6Alkyl or optionally substituted C1-C6A heteroalkyl group;

Y1、Y2and Y3Each of which is independently N or CRC1Wherein R isC1Is H, halo, CN, optionally substituted C1-C6Alkyl or optionally substituted C1-C6A heteroalkyl group; and R is12Is H, halo, CN, optionally substituted C1-C6Alkyl or optionally substituted C1-C6A heteroalkyl group.

In some embodiments, X is S. In some embodiments, X is O.

In some embodiments, Y is1Is N. In some embodiments, Y is1Is CRC1

In some embodiments, Y is2Is N. In some embodiments, Y is2Is CRC1

In some embodiments, Y is3Is N. In some embodiments, Y is3Is CRC1

In some embodiments, RC1Is H,

In some embodiments, R12Is H,

In some embodiments, R1Is that

Wherein

f1 is 0, 1, 2, 3 or 4;

f2 is 0, 1, 2 or 3; and is

Each R13Independently is halo, CN, optionally substituted C1-C6Alkyl or optionally substituted C1-C6A heteroalkyl group.

In some embodiments, R1Is that

In some embodiments, f1 is 0. In some embodiments, f1 is 1.

In some embodiments, R1Is that

In some embodiments, f2 is 0.

In some embodiments, R1Is that

Wherein

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

RN2is H, optionally substituted C1-C6Alkyl or optionally substituted C1-C6A heteroalkyl group; and is

Each R14Is H, halo, CN, optionally substituted C1-C6Alkyl or optionally substituted C1-C6A heteroalkyl group.

In some embodiments, R1Is that

In some embodiments, g is 0.

In some embodiments, R1Is that

Wherein

i is 0, 1, 2 or 3;

W1and W2Each of which is independently N or CRC2Wherein R isC2Is H, halo, CN, optionally substituted C1-C6Alkyl or optionally substituted C1-C6A heteroalkyl group;

RN3is H, optionally substituted C1-C6Alkyl, optionally substituted C1-C6Heteroalkyl, optionally substituted C3-C10Carbocyclyl, optionally substituted C2-C9Heterocyclyl or optionally substituted C2-C9A heteroaryl group; and is

Each R15Independently is halo, CN, optionally substituted C1-C6Alkyl or optionally substituted C1-C6A heteroalkyl group.

In some embodiments, i is 0.

In some embodiments, W1Is N. In some embodiments, W1Is CRC2

In some embodiments, W2Is N. In some embodiments, W2Is CRC2

In some embodiments, RN3Is H, optionally substituted C1-C6Alkyl, optionally substituted C 1-C6Heteroalkyl or optionally substituted C2-C9A heterocyclic group.

In some embodiments, RN3Is that

In some embodiments, R1Is F, Cl, Br, I, CN, NO2、NH2

In some embodiments, R1And R2Together with the atoms to which each is attached to form optionally substituted C3-C10Sub-carbonCyclic radical, optionally substituted C2-C9Heterocyclylene radical, optionally substituted C6-C10Arylene or optionally substituted C2-C9A heteroarylene group.

In some embodiments, R1And R2Together with the atoms to which each is attached to form optionally substituted C3-C10A carbocyclylene group.

In some embodiments, the compound has the structure of formula Ib:

or a pharmaceutically acceptable salt thereof.

In some embodiments, L is1Is optionally substituted C1-C6A heteroalkylene group.

In some embodiments, L is1Is that

In some embodiments, L is1Is that

In one aspect, the present disclosure provides a compound, or a pharmaceutically acceptable salt thereof, having the structure of any one of compounds 1-464 of table 1.

TABLE 1 Compounds of the invention

In one aspect, the present disclosure provides a pharmaceutical composition comprising a compound of any of the foregoing compounds, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient.

In some embodiments, the pharmaceutical composition comprises a compound of formula I or formula II and a pharmaceutically acceptable excipient.

In one aspect, the present disclosure provides a method of treating a neurological disorder in a subject in need thereof, comprising administering an effective amount of any of the foregoing compounds or a pharmaceutical composition thereof.

In one aspect, the present disclosure provides a method of inhibiting toxicity in a cell associated with a protein, the method comprising administering an effective amount of any of the foregoing compounds or a pharmaceutical composition thereof.

In some embodiments, the toxicity is alpha-synuclein-associated toxicity. In some embodiments, the toxicity is ApoE 4-related toxicity.

In some embodiments, the cell is a mammalian neural cell.

In one aspect, the present disclosure provides a method of treating a stearoyl-CoA desaturase (SCD) -associated disorder in a subject in need thereof, comprising administering an effective amount of any of the foregoing compounds, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof.

Non-limiting exemplary SCD-related disorders include, but are not limited to, metabolic disorders (e.g., diabetes (e.g., type I diabetes and type II diabetes), hyperglycemia, metabolic Syndrome, obesity, lipid disorders, fatty liver, nonalcoholic steatohepatitis (NASH), nonalcoholic fatty liver disease (NAFLD), and hypertension), cancer, cardiovascular disease, cerebrovascular disease, renal disease, liver disease, skin disorders (e.g., acne vulgaris)), Central Nervous System (CNS) disorders, dementia, multiple sclerosis, schizophrenia, mild cognitive impairment, alzheimer's disease, cerebral amyloid angiopathy, and dementia associated with Down Syndrome (Down Syndrome).

In some embodiments, the SCD-related disorder is an SCD 1-related disorder.

In some embodiments, the SCD-related disorder is an SCD 5-related disorder.

In one aspect, the present disclosure provides a method of inhibiting SCD5, the method comprising contacting a cell with an effective amount of any of the foregoing compounds, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof.

In one aspect, the present disclosure provides a method of inhibiting SCD1, the method comprising contacting a cell with an effective amount of any of the foregoing compounds, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof.

Chemical terminology

It is to be understood that the terminology used herein is for the purpose of describing particular embodiments and is not intended to be limiting.

One skilled in the art will appreciate that certain compounds described herein can exist in one or more different isomeric (e.g., stereoisomeric, geometric, tautomeric) and/or isotopic (e.g., where one or more atoms have been replaced with a different isotope of an atom, such as hydrogen substituted with deuterium). Unless otherwise indicated or clear from context, the depicted structures are understood to represent any such isomeric or isotopic form, either individually or in combination.

In some embodiments, one or more compounds depicted herein may exist in different tautomeric forms. It is clear from the context that, unless explicitly excluded, reference to such compounds encompasses all such tautomeric forms. In some embodiments, tautomeric forms result from the exchange of single bonds with adjacent double bonds and the concomitant migration of protons. In certain embodiments, a tautomeric form can be a prototropic tautomer, which is an isomeric protonation state having the same empirical formula and total charge as the reference form. Examples of moieties having prototropic tautomeric forms are keto-enol pairs, amide-imide pairs, lactam-lactam pairs, amide-imide pairs, enamine-imide pairs, and cyclic forms in which protons may occupy two or more positions of the heterocyclic ring system, such as 1H-imidazole and 3H-imidazole, 1H-1, 2, 4-triazole, 2H-1, 2, 4-triazole and 4H-1, 2, 4-triazole, 1H-isoindole and 2H-isoindole, and 1H-pyrazole and 2H-pyrazole. In some embodiments, tautomeric forms can be in equilibrium or locked into one form by appropriate substitution spaces. In certain embodiments, tautomeric forms result from acetal interconversion (e.g., interconversion as shown in the following schemes):

One skilled in the art will appreciate that, in some embodiments, isotopes of compounds described herein can be made and/or utilized in accordance with the present invention. "isotopic" refers to atoms having the same atomic number but differing mass numbers due to the difference in the number of neutrons in the core. For example, isotopes of hydrogen include tritium and deuterium. In some embodiments, isotopic substitution (e.g., replacement of hydrogen with deuterium) can alter the physicochemical properties of the molecule, such as the rate of racemization at metabolic and/or chiral centers.

As is known in the art, many chemical entities (particularly many organic molecules and/or many small molecules) can take a variety of different solid forms, such as, for example, amorphous and/or crystalline forms (e.g., polymorphs, hydrates, solvates, etc.). In some embodiments, such entities may be utilized in any form, including in any solid form. In some embodiments, such entities are utilized in a particular form (e.g., in a particular solid form).

In some embodiments, the compounds described and/or depicted herein may be provided and/or utilized in the form of a salt.

In certain embodiments, the compounds described and/or depicted herein may be provided and/or utilized in the form of hydrates or solvates.

Throughout this specification, substituents of the compounds of the present disclosure are disclosed in groups or ranges. The disclosure is expressly intended to include each and every individual subcombination of the members of such groups and ranges. For example, the term "C1-C6Alkyl "is expressly intended to disclose methyl, ethyl, C individually3Alkyl radical, C4Alkyl radical, C5Alkyl and C6An alkyl group. Furthermore, unless otherwise indicated, when a compound comprises a plurality of positions where substituents are disclosed in groups or ranges, this disclosure is intended to encompass individual compounds and groups of compounds (e.g., genera and subgenera) containing each and every single subcombination of members at each position.

Herein, the phrase of the form "optionally substituted X" (e.g., optionally substituted alkyl) is intended to be equivalent to "X, wherein X is optionally substituted" (e.g., "alkyl, wherein the alkyl is optionally substituted"). This is not intended to mean that the feature "X" (e.g. alkyl) itself is optional.

The term "acyl" as used herein, means hydrogen or alkyl as defined herein attached to the parent molecular group through a carbonyl group, as defined herein, and is exemplified by formyl (i.e., carboxyaldehyde group), acetyl, trifluoroacetyl, propionyl, and butyryl. Exemplary unsubstituted acyl groups include 1 to 6, 1 to 11, or 1 to 21 carbons.

The term "alkyl" as used herein refers to a branched or straight chain monovalent saturated aliphatic hydrocarbon group having 1 to 20 carbon atoms (e.g., 1 to 16 carbon atoms, 1 to 10 carbon atoms, or 1 to 6 carbon atoms). Alkylene is a divalent alkyl group.

The term "alkenyl" as used herein, alone or in combination with other groups, refers to a straight or branched chain hydrocarbon residue having a carbon-carbon double bond and having 2 to 20 carbon atoms (e.g., 2 to 16 carbon atoms, 2 to 10 carbon atoms, 2 to 6, or 2 carbon atoms).

The term "alkynyl", as used herein, alone or in combination with other groups, refers to a straight or branched chain hydrocarbon residue having a carbon-carbon triple bond and having from 2 to 20 carbon atoms (e.g., 2 to 16 carbon atoms, 2 to 10 carbon atoms, 2 to 6, or 2 carbon atoms).

The term "amino" as used herein denotes-N (R)N1)2Wherein each R isN1Independently H, OH, NO2、N(RN2)2、SO2ORN2、SO2RN2、SORN2N-protecting groups, alkyl, alkoxy, aryl, arylalkyl, cycloalkyl, acyl (e.g., acetyl, trifluoroacetyl, or other acyl groups described herein), wherein these are listed RN1Each of the groups may be optionally substituted; or two RN1Combine to form an alkylene or heteroalkylene group, and wherein each R N2Independently H, alkyl or aryl. The amino group of the present invention may be an unsubstituted amino group (i.e., -NH)2) Or substituted amino (i.e., -N (R)N1)2)。

The term "aryl" as used herein refers to an aromatic mono-or poly-carbocyclic group of 6 to 12 carbon atoms having at least one aromatic ring. Examples of such groups include, but are not limited to, phenyl, naphthyl, 1, 2, 3, 4-tetrahydronaphthyl, 1, 2-dihydronaphthyl, indanyl, and 1H-indenyl.

The term "arylalkyl" as used herein denotes an alkyl group substituted with an aryl group. Exemplary unsubstituted arylalkyl groups are 7 to 30 carbons (e.g., 7 to 16 or 7 to 20 carbons, such as C)1-C6Alkyl radical C6-10Aryl radical, C1-C10Alkyl radical C6-10Aryl or C1-C20Alkyl radicalC6-10Aryl) such as benzyl and phenethyl. In some embodiments, each of the alkyl and aryl groups can be further substituted with 1, 2, 3, or 4 substituents as defined herein for the respective group.

The term "azido" as used herein denotes-N3A group.

The term "cyano" as used herein denotes a CN group.

The term "carbocyclyl" as used herein refers to a non-aromatic C formed from carbon atoms3-C12Monocyclic, bicyclic or tricyclic structures. Carbocyclyl structures include cycloalkyl and unsaturated carbocyclyl.

The term "cycloalkyl" as used herein refers to a saturated, non-aromatic, monovalent mono-or multicyclic group of 3 to 10, preferably 3 to 6, carbon atoms. This term is further exemplified by groups such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, norbornyl, and adamantyl.

The term "halo" as used herein refers to a fluoro (fluoro), chloro (chloro), bromo (bromo) or iodo (iodo) group.

The term "heteroalkyl," as used herein, refers to an alkyl group, as defined herein, in which one or more of the constituent carbon atoms has been replaced with nitrogen, oxygen, or sulfur. In some embodiments, heteroalkyl groups may be further substituted with 1, 2, 3, or 4 substituents as described herein for alkyl groups. An example of heteroalkyl is "alkoxy," which as used herein refers to alkyl-O- (e.g., methoxy and ethoxy). Heteroalkylidene is a divalent heteroalkyl group.

The term "heteroalkenyl" as used herein refers to an alkenyl group, as defined herein, in which one or more of the constituent carbon atoms has been replaced with nitrogen, oxygen, or sulfur. In some embodiments, a heteroalkenyl group can be further substituted with 1, 2, 3, or 4 substituents as described herein for the alkenyl group. An example of a heteroalkenyl group is "alkenyloxy," which, as used herein, refers to alkenyl-O-. Heteroalkenylene is a divalent heteroalkenyl group.

The term "heteroalkynyl" as used herein refers to an alkynyl group, as defined herein, in which one or more of the constituent carbon atoms has been replaced by nitrogen, oxygen or sulfur. In some embodiments, heteroalkynyl may be further substituted with 1, 2, 3, or 4 substituents as described herein for alkynyl groups. An example of a heteroalkynyl group is "alkynyloxy," which, as used herein, refers to alkynyl-O-. Heteroalkynylene is divalent heteroalkynyl.

The term "heteroaryl" as used herein refers to an aromatic mono-or polycyclic group of 5 to 12 atoms having at least one aromatic ring containing 1, 2 or 3 ring heteroatoms selected from N, O and S, wherein the remaining ring atoms are C. One or two ring carbon atoms of a heteroaryl group may be replaced by a carbonyl group. Examples of heteroaryl groups are pyridyl, pyrazolyl, benzoxazolyl, benzimidazolyl, benzothiazolyl, imidazolyl, oxazolyl and thiazolyl.

The term "heteroarylalkyl" as used herein, denotes an alkyl group substituted with a heteroaryl group. Exemplary unsubstituted heteroarylalkyl groups are 7 to 30 carbons (e.g., 7 to 16 carbons or 7 to 20 carbons, such as C)1-C6Alkyl radical C2-C9Heteroaryl group, C1-C10Alkyl radical C2-C9Heteroaryl or C1-C20Alkyl radical C2-C9Heteroaryl). In some embodiments, each of the alkyl and heteroaryl groups can be further substituted with 1, 2, 3, or 4 substituents as defined herein for the respective group.

The term "heterocyclyl" as used herein denotes a mono-or polycyclic group of 3 to 12 atoms having at least one ring containing 1, 2, 3 or 4 ring heteroatoms selected from N, O or S, wherein no ring is aromatic. Examples of heterocyclyl groups include, but are not limited to, morpholinyl, thiomorpholinyl, furanyl, piperazinyl, piperidinyl, pyranyl, pyrrolidinyl, tetrahydropyranyl, tetrahydrofuranyl, and 1, 3-dioxanyl.

The term "heterocyclylalkyl" as used herein refers to an alkyl group substituted with a heterocyclyl. Exemplary unsubstituted heterocyclylalkyl groups are 7 to 30 carbons (e.g., 7 to 16 or 7 to 20 carbons, such as C)1-C6Alkyl radical C2-C9Heterocyclic group, C1-C10Alkyl radical C2-C9Heterocyclyl or C1-C20Alkyl radical C2-C9A heterocyclic group). In some embodiments, each of the alkyl and heterocyclyl groups may be further substituted by 1, 2, 3, or 4 substituents as defined herein for the respective group.

The term "hydroxy" as used herein denotes an-OH group.

The term "N-protecting group" as used herein denotes those groups intended to protect amino groups from undesired reactions during the synthetic process. Commonly used N-protecting Groups are disclosed in Greene, "Protective Groups in Organic Synthesis," 3 rd edition (John Wiley & Sons, New York, 1999). N-protecting groups include acyl, aroyl or carbamoyl groups such as formyl, acetyl, propionyl, pivaloyl, t-butylacetyl, 2-chloroacetyl, 2-bromoacetyl, trifluoroacetyl, trichloroacetyl, phthaloyl, o-nitrophenoxyacetyl, α -chlorobutyryl, benzoyl, 4-chlorobenzoyl, 4-bromobenzoyl, 4-nitrobenzoyl, and chiral auxiliaries such as protected or unprotected D, L or D, L-amino acids such as alanine, leucine and phenylalanine; sulfonyl-containing groups such as benzenesulfonyl and p-toluenesulfonyl; a carbamate-forming group such as benzyloxycarbonyl, p-chlorobenzyloxycarbonyl, p-methoxybenzyloxycarbonyl, p-nitrobenzyloxycarbonyl, 2-nitrobenzyloxycarbonyl, p-bromobenzyloxycarbonyl, 3, 4-dimethoxybenzyloxycarbonyl, 3, 5-dimethoxybenzyloxycarbonyl, 2, 4-dimethoxybenzyloxycarbonyl, 4-methoxybenzyloxycarbonyl, 2-nitro-4, 5-dimethoxybenzyloxycarbonyl, 3, 4, 5-trimethoxybenzyloxycarbonyl, 1- (p-biphenylyl) -1-methylethoxycarbonyl, α -dimethyl-3, 5-dimethoxybenzyloxycarbonyl, benzhydryloxycarbonyl, t-butoxycarbonyl, diisopropylmethoxycarbonyl, isopropoxycarbonyl, ethoxycarbonyl, p-chlorobenzyloxycarbonyl, p-methoxybenzyloxycarbonyl, p-nitrobenzyloxycarbonyl, p-nitrobenzyloxy, Methoxycarbonyl, allyloxycarbonyl, 2, 2, 2-trichloroethoxycarbonyl, phenoxycarbonyl, 4-nitrophenoxycarbonyl, fluorenyl-9-methoxycarbonyl, cyclopentyloxycarbonyl, adamantyloxycarbonyl, cyclohexyloxycarbonyl and phenylthiocarbonyl; arylalkyl groups such as benzyl, triphenylmethyl, and benzyloxymethyl; and silyl groups, such as trimethylsilyl. Preferred N-protecting groups are allyloxycarbonyl, formyl, acetyl, benzoyl, pivaloyl, t-butylacetyl, alanyl, phenylsulfonyl, benzyl, t-butoxycarbonyl (Boc) and benzyloxycarbonyl (Cbz).

The term "nitro" as used herein denotes NO2A group.

The term "mercapto" as used herein denotes the-SH group.

The alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, carbocyclyl (e.g., cycloalkyl), aryl, heteroaryl, and heterocyclyl groups may be substituted or unsubstituted. When substituted, typically 1 to 4 substituents will be present unless otherwise indicated. Substituents include, for example: aryl (e.g., substituted and unsubstituted phenyl), carbocyclyl (e.g., substituted and unsubstituted cycloalkyl), halo (e.g., fluoro), hydroxy, heteroalkyl (e.g., substituted and unsubstituted methoxy, ethoxy, or thioalkoxy), heteroaryl, heterocyclyl, amino (e.g., NH2Or mono or dialkylamino), azido, cyano, nitro or mercapto. Aryl, carbocyclyl (e.g., cycloalkyl), heteroaryl, and heterocyclyl may also be substituted with alkyl (unsubstituted and substituted, such as arylalkyl (e.g., substituted and unsubstituted benzyl)).

The compounds of the invention may have one or more asymmetric carbon atoms and may exist in the form of optically pure enantiomers, mixtures of enantiomers (such as, for example, racemates), optically pure diastereomers, mixtures of diastereomers, diastereomer racemates or mixtures of diastereomer racemates. Optically active forms can be obtained, for example, by resolution of the racemate, by asymmetric synthesis or asymmetric chromatography (chromatography using chiral adsorbents or eluents). That is, certain disclosed compounds may exist in various stereoisomeric forms. Stereoisomers are compounds that differ only in their spatial arrangement. Enantiomers are paired stereoisomers, the mirror images of which are non-superimposable, the most common reason being that they contain asymmetrically substituted carbon atoms which act as chiral centers. "enantiomer" means one of a pair of molecules that are mirror images of each other and that do not overlap. Diastereomers are stereoisomers that are unrelated to mirror images, the most common reason being that they contain two or more asymmetrically substituted carbon atoms and represent the configuration of the substituent around one or more chiral carbon atoms. Enantiomers of compounds can be prepared, for example, by separating the enantiomers from the racemate using one or more well-known techniques and methods, such as, for example, chiral chromatography and separation methods based thereon. One skilled in the art can readily determine the appropriate technique and/or method to isolate an enantiomer of a compound described herein from a racemic mixture. "racemate" or "racemic mixture" means a compound containing two enantiomers, wherein such mixture does not exhibit optical activity; i.e. they do not rotate the plane of the polarized light. "geometric isomers" refer to isomers that differ in the orientation of the substituent atoms associated with a carbon-carbon double bond, cycloalkyl ring, or bridged bicyclic ring system. The atoms (other than H) on each side of the carbon-carbon double bond may be in the E (substituents on opposite sides of the carbon-carbon double bond) or Z (substituents oriented on the same side) configuration. "R", "S", "R", "E", "Z", "cis" and "trans" indicate the configuration relative to the core molecule. Some of the disclosed compounds may exist as atropisomeric forms. Atropisomers are stereoisomers that arise due to the hindered rotation about a single bond, wherein the rotating steric strain barrier is high enough to allow separation of conformers. The compounds of the present invention can be prepared as individual isomers or resolved from mixtures of isomers by isomer-specific synthesis. Conventional resolution techniques include the formation of a free base salt of each isomer of the isomer pair using an optically active acid (followed by fractional crystallization and regeneration of the free base), the formation of an acid form salt of each isomer of the isomer pair using an optically active amine (followed by fractional crystallization and regeneration of the free acid), the formation of an ester or amide of each isomer of the isomer pair using an optically pure acid, amine or alcohol (followed by chromatographic separation and removal of the chiral auxiliary), or the resolution of the isomeric mixtures of the starting materials or the final product using various well-known chromatographic methods. When the disclosed compounds are named or depicted by structure stereochemistry, the named or depicted stereoisomer is at least 60, 70, 80, 90, 99, or 99.9 wt% relative to the other stereoisomers. When a single enantiomer is named or depicted by structure, the depicted or named enantiomer is at least 60, 70, 80, 90, 99, or 99.9% optically pure. When a single diastereomer is named or depicted by structure, the depicted or named diastereomer is at least 60, 70, 80, 90, 99, or 99.9% weight pure. Percent optical purity is the ratio of the weight of an enantiomer relative to the weight of the enantiomer plus the weight of its optical isomer. Diastereoisomeric purity by weight is the ratio of the weight of one diastereomer to the weight of all diastereoisomers. When the disclosed compounds are named or depicted by structure stereochemistry, the named or depicted stereoisomer is at least 60%, 70%, 80%, 90%, 99%, or 99.9% pure by mole fraction relative to the other stereoisomers. When a single enantiomer is named or depicted by structure, the depicted or named enantiomer is at least 60%, 70%, 80%, 90%, 99%, or 99.9% pure by mole fraction. When a single diastereomer is named or depicted by structure, the depicted or named diastereomer is at least 60%, 70%, 80%, 90%, 99%, or 99.9% pure by mole fraction. Percent purity in terms of mole fraction is the ratio of moles of an enantiomer to moles of enantiomer plus the moles of its optical isomer. Similarly, percent purity by mole fraction is the ratio of moles of a diastereomer to moles of a diastereomer plus moles of its isomer. When the disclosed compounds are named or depicted by structure without indicating stereochemistry and have at least one chiral center, it is to be understood that the name or structure encompasses any enantiomer of the compound that is free of the corresponding optical isomer, racemic mixtures of the compound, or mixtures that are enriched in one enantiomer relative to its corresponding optical isomer. When the disclosed compounds are named or depicted by structure without indication of stereochemistry and have two or more chiral centers, it is understood that the name or structure encompasses a diastereomer free of other diastereomers, a number of diastereomers free of other diastereomeric pairs, a mixture of diastereomers, a mixture of diastereomeric pairs, a mixture of diastereomers in which one diastereomer is enriched relative to the other diastereomer or diastereomers. The present invention encompasses all of these forms.

Definition of

In this application, unless otherwise clear from the context, (i) the term "a" may be understood to mean "at least one"; (ii) the term "or" may be understood to mean "and/or"; (iii) the terms "comprising" and "including" can be understood to encompass a listing of components or steps on an item-by-item basis, whether presented by themselves or in combination with one or more other components or steps; and (iv) the terms "about" and "approximately" can be understood to allow for standard variations as would be understood by one of ordinary skill in the art; and (v) where a range is provided, an endpoint is included.

The term "administering" as used herein refers to administering a composition (e.g., a compound, complex, or formulation comprising a compound or complex as described herein) to a subject or system. Administration to an animal subject (e.g., to a human) can be by any suitable route. For example, in some embodiments, administration can be bronchial (including by bronchial instillation), buccal, enteral, intradermal, intramedullary (intradermal), intraarterial, intradermal, intragastric, intramedullary, intramuscular, intranasal, intraperitoneal, intrathecal, intravenous, intraventricular, mucosal, nasal, oral, rectal, subcutaneous, sublingual, topical, tracheal (including by intratracheal instillation), transdermal, vaginal, and vitreous.

The term "animal" as used herein refers to any member of the kingdom animalia. In some embodiments, "animal" refers to a human at any stage of development. In some embodiments, "animal" refers to a non-human animal at any stage of development. In some embodiments, the non-human animal is a mammal (e.g., a rodent, a mouse, a rat, a rabbit, a monkey, a dog, a cat, a sheep, a cow, a primate, and/or a pig). In some embodiments, the animal includes, but is not limited to, a mammal, a bird, a reptile, an amphibian, a fish, and/or a worm. In some embodiments, the animal can be a transgenic animal, a genetically engineered animal, and/or a clone.

The terms "about" and "about" as used herein are each intended to encompass normal statistical variations as would be understood by one of ordinary skill in the art to be appropriate for the relevant context. In certain embodiments, the terms "about" or "about" each refer to a range of values that fall within 25%, 20%, 19%, 18%, 17%, 16%, 15%, 14%, 13%, 12%, 11%, 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, 1%, or less of the stated value in either direction (greater or less) unless otherwise stated or otherwise evident from the context (e.g., where the number would exceed 100% of the possible values).

When the term "related" is used herein, two events or entities are "related" to each other if the presence, level, and/or form of one event or entity is associated with the presence, level, and/or form of the other event or entity. For example, a particular entity (e.g., polypeptide) is considered to be associated with a particular disease, disorder, or condition if its presence, level, and/or form is associated with the incidence and/or susceptibility (e.g., in a relevant population) of the disease, disorder, or condition.

In the practice of the methods of the present invention, an "effective amount" of any one of the compounds of the present invention or any combination of the compounds of the present invention or a pharmaceutically acceptable salt thereof, is administered, alone or in combination, via any conventional and acceptable method known in the art.

The term "combination therapy" as used herein refers to those situations in which a subject is exposed to two or more therapeutic agents simultaneously. In some embodiments, two or more compounds may be administered simultaneously; in some embodiments, such compounds may be administered sequentially; in some embodiments, such compounds are administered in overlapping dosing regimens.

The term "dosage form" as used herein refers to a physically discrete unit of active compound (e.g., therapeutic or diagnostic agent) for administration to a subject. Each unit containing a predetermined amount of active agent. In some embodiments, such amounts are unit dose amounts (or whole fraction thereof) suitable for administration according to a dosing regimen that has been determined to correlate with a desired or beneficial result when administered to a relevant population (i.e., by a therapeutic dosing regimen). One of ordinary skill in the art understands that the total amount of a therapeutic composition or compound administered to a particular subject is determined by one or more attending physicians and may involve the administration of multiple dosage forms.

The term "dosing regimen" as used herein refers to a group of unit doses (typically more than one) administered individually to a subject, typically separated by a period of time. In some embodiments, a given therapeutic compound has a recommended dosing regimen, which may involve one or more doses. In some embodiments, the dosing regimen comprises a plurality of doses, each dose separated from each other by a time period of the same length; in some embodiments, the dosing regimen comprises multiple doses and at least two different time periods separated by a single dose. In some embodiments, all doses within a dosing regimen are of the same unit dose amount. In some embodiments, different doses within a dosing regimen have different amounts. In some embodiments, a dosing regimen comprises a first dose of a first dosage amount followed by one or more additional doses of a second dosage amount different from the first dosage amount. In some embodiments, the dosing regimen comprises a first dose of a first dosage amount followed by one or more additional doses of a second dosage amount that is the same as the first dosage amount. In some embodiments, the dosing regimen is associated with a desired or beneficial result when administered in a relevant population (i.e., is a therapeutic dosing regimen).

The term "pharmaceutical composition" as used herein means a composition containing a compound described herein formulated with a pharmaceutically acceptable excipient and manufactured or sold according to the approval of a governmental regulatory agency as part of a therapeutic regimen for the treatment of disease in a mammal. The pharmaceutical composition can be formulated, for example, for oral administration in a unit dosage form (e.g., a tablet, capsule, caplet, soft capsule, or syrup); for topical administration (e.g., in the form of a cream, gel, lotion, or ointment); for intravenous administration (e.g., as a sterile solution in a microparticle-free plug and in a solvent system suitable for intravenous use); or in any other pharmaceutically acceptable formulation.

As used herein, "pharmaceutically acceptable excipient" refers to any ingredient other than the compounds described herein (e.g., a vehicle capable of suspending or dissolving an active compound) and has substantially non-toxic and non-inflammatory properties in a patient. Excipients may include, for example: antiadherents, antioxidants, binders, coating agents, compression aids, disintegrants, dyes (colorants), softeners, emulsifiers, fillers (diluents), film formers or coating agents, flavors, fragrances, glidants (flow enhancers), lubricants, preservatives, printing inks, adsorbents, suspending or dispersing agents, sweeteners, and water of hydration. Exemplary excipients include, but are not limited to: butylated Hydroxytoluene (BHT), calcium carbonate, calcium phosphate (dibasic), calcium stearate, croscarmellose, cross-linked polyvinylpyrrolidone, citric acid, crospovidone, cysteine, ethylcellulose, gelatin, hydroxypropyl cellulose, hydroxypropyl methylcellulose, lactose, magnesium stearate, maltitol, mannitol, methionine, methylcellulose, methyl paraben, microcrystalline cellulose, polyethylene glycol, polyvinylpyrrolidone, povidone, pregelatinized starch, propyl paraben, retinyl palmitate, shellac, silicon dioxide, sodium carboxymethylcellulose, sodium citrate, sodium starch glycolate, sorbitol, starch (corn), stearic acid, sucrose, talc, titanium dioxide, vitamin a, vitamin E, vitamin C, and xylitol.

The term "pharmaceutically acceptable salt" as used herein means any pharmaceutically acceptable salt of the compound of formula (I). For example, pharmaceutically acceptable salts of any of the compounds described herein include those salts which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and animals without excessive toxicity, irritation, allergic response, and commensurate with a reasonable benefit/risk ratio. Pharmaceutically acceptable salts are well known in the art. For example, pharmaceutically acceptable Salts are described in Berge et al, J.pharmaceutical Sciences 66:1-19, 1977 and Pharmaceutical Salts: Properties, Selection, and Use, (edited by P.H.Stahl and C.G.Wermuth), Wiley-VCH, 2008. Salts may be prepared in situ during the final isolation and purification of the compounds described herein, or separately by reacting the free base group with a suitable organic acid.

The compounds of the present invention may have ionizable groups so as to be capable of being prepared as pharmaceutically acceptable salts. These salts may be acid addition salts involving inorganic or organic acids, or they may be prepared from inorganic or organic bases in the case of the acidic forms of the compounds of the invention. Typically, the compounds are prepared or used as pharmaceutically acceptable salts prepared as addition products of pharmaceutically acceptable acids or bases. Suitable pharmaceutically acceptable acids and bases and methods for preparing suitable salts are well known in the art. Salts can be prepared from pharmaceutically acceptable non-toxic acids and bases, including inorganic and organic acids and bases.

The term "pure" means substantially pure or free of undesired components (e.g., other compounds and/or other components of a cell lysate), material offset, mixture, or defect.

Representative acid addition salts include acetate, adipate, alginate, ascorbate, aspartate, benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphorate, camphorsulfonate, citrate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, fumarate, glucoheptonate, glycerophosphate, hemisulfate, heptanoate, hexanoate, hydrobromide, hydrochloride, hydroiodide, 2-hydroxy-ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, malate, maleate, malonate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oleate, oxalate, palmitate, pamoate, pectate, persulfate, 3-phenylpropionate, phosphate, sulfate, salicylate, and the like, Picrate, pivalate, propionate, stearate, succinate, sulfate, tartrate, thiocyanate, tosylate, undecanoate, and valerate. Representative alkali or alkaline earth metal salts include sodium, lithium, potassium and magnesium salts, as well as non-toxic ammonium, quaternary ammonium and amine cation salts, including but not limited to ammonium, tetramethylammonium, tetraethylammonium, methylamine, dimethylamine, trimethylamine, triethylamine and ethylamine salts.

The term "stearoyl-CoA desaturase (SCD) -associated disorder" as used herein refers to an undesirable physiological condition, disorder or disease associated with and/or mediated at least in part by an SCD protein. In some cases, an SCD-related disorder is associated with excessive SCD levels and/or activity. SCD introduces a double bond at position C9-C10 of saturated fatty acids (such as palmitoyl-CoA and stearoyl-CoA), which are converted to palmitoyl-CoA and oleoyl-CoA, respectively. One SCD gene (SCD1) has been characterized in humans, and for humans there are two subtypes (SCD1 and SCD 5). SCD-related diseases may be associated with SCD1 and/or SCD5 and/or mediated at least in part by SCD1 and/or SCD 5. Exemplary SCD-related disorders include SCD-related disorders, including, but not limited to, metabolic disorders (e.g., diabetes (e.g., type I diabetes and type II diabetes), hyperglycemia, metabolic syndrome, obesity, lipid disorders, fatty liver, nonalcoholic steatohepatitis (NASH), nonalcoholic fatty liver disease (NAFLD), and hypertension), cancer, cardiovascular disease, cerebrovascular disease, nephropathy, liver disease, skin disorders (e.g., acne vulgaris)), Central Nervous System (CNS) disorders, dementia, multiple sclerosis, schizophrenia, mild cognitive disorders, alzheimer's disease, cerebral amyloid angiopathy, and dementia associated with down's syndrome. Additional SCD-related disorders are described herein or known in the art.

The term "subject" as used herein refers to any organism to which a composition of the invention may be administered, e.g., for experimental, diagnostic, prophylactic and/or therapeutic purposes. Typical subjects include any animal (e.g., mammals such as mice, rats, rabbits, non-human primates, and humans). The subject may seek or require treatment, be undergoing treatment, will undergo treatment, or be a human or animal under the care of a trained professional for a particular disease or condition.

The terms "treat", "treated" or "treating" as used herein mean both therapeutic treatment and prophylactic (preventative) or preventative measures, wherein the object is to prevent or slow down (lessen) an undesired physiological condition, disorder or disease, or to obtain a beneficial or desired clinical result. Beneficial or desired clinical results include, but are not limited to, alleviation of symptoms; a reduced extent of the condition, disorder or disease; stable (i.e., no worsening) state of the condition, disorder or disease; delay in onset or slowing of progression of the condition, disorder or disease; amelioration or palliation (whether partial or total) of the condition, disorder or disease state; an improvement in at least one measurable physical parameter, not necessarily discernible by the patient; or enhancement or amelioration of a condition, disorder or disease. Treatment includes eliciting a clinically significant response without excessive levels of side effects. Treatment also includes prolonging survival compared to expected survival in the absence of treatment.

By "treatment regimen" is meant a dosing regimen that is administered in association with a desired or beneficial therapeutic outcome in the relevant population.

The term "therapeutically effective amount" means an amount sufficient to treat a disease, disorder, and/or condition when administered according to a therapeutic dosing regimen to a population suffering from or susceptible to such a disease, disorder, and/or condition. In some embodiments, a therapeutically effective amount is an amount that reduces the incidence and/or severity and/or delays the onset of one or more symptoms of a disease, disorder, and/or condition. One of ordinary skill in the art will appreciate that the term "therapeutically effective amount" does not actually require successful treatment in a particular individual. Of course, a therapeutically effective amount may be an amount that, when administered to a patient in need of such treatment, provides a particular desired pharmacological response in a substantial number of subjects. It is specifically understood that a particular subject may actually be "refractory" to a "therapeutically effective amount". To give just one example, refractory subjects may have low bioavailability such that clinical efficacy is not achieved. In some embodiments, reference to a therapeutically effective amount may refer to an amount as measured in one or more specific tissues (e.g., tissues affected by a disease, disorder, or condition) or fluids (e.g., blood, saliva, serum, sweat, tears, urine, etc.). One of ordinary skill in the art will appreciate that, in some embodiments, a therapeutically effective amount may be formulated and/or administered in a single dose. In some embodiments, a therapeutically effective amount may be formulated and/or administered in multiple doses (e.g., as part of a dosing regimen).

Detailed Description

The invention features compounds useful for treating neurological disorders, for example, by inhibiting alpha-synuclein toxicity in a cell (such as a nerve cell), or by inhibiting SCD5 and/or SCD1 in a cell (such as a nerve cell). Exemplary compounds described herein include compounds having the structure of formula I:

wherein

R1Is H, halo, CN, NO2Hydroxy, optionally substituted amino, optionally substituted C1-C6Alkyl, optionally substituted C1-C6Heteroalkyl, optionally substituted C1-C6Alkenyl, optionally substituted C1-C6Heteroalkenyl, optionally substituted C3-C10Carbocyclyl, optionally substituted C2-C9Heterocyclyl, optionally substituted C6-C10Aryl or optionally substituted C2-C9(ii) a heteroaryl group, wherein,

R2is H or optionally substituted C1-C6Alkyl, or

R1And R2Together with the atoms to which each is attached to form optionally substituted C3-C10Carbocyclylene, optionally substituted C2-C9Heterocyclylene radical, optionally substituted C6-C10Arylene or optionally substituted C2-C9A heteroarylene group;

L1is optionally substituted C1-C6Alkylene or optionally substituted C1-C6A heteroalkylene group; and is

R3Is optionally substituted C2-C9(ii) a heteroaryl group, wherein,

or a compound having the structure of formula II:

wherein

R1Is H, halo, CN, NO2Hydroxy, optionally substituted amino, optionally substituted C 1-C6Alkyl, optionally substituted C1-C6Heteroalkyl, optionally substituted C1-C6Alkenyl, optionally substituted C1-C6Heteroalkenyl, optionally substituted C3-C10Carbocyclyl, optionally substituted C2-C9Heterocyclyl, optionally substituted C6-C10Aryl or optionally substituted C2-C9A heteroaryl group; and is

R2Is H or optionallySubstituted C1-C6An alkyl group; or

R1And R2Together with the atoms to which each is attached to form optionally substituted C3-C10Carbocyclylene, optionally substituted C2-C9Heterocyclylene radical, optionally substituted C6-C10Arylene or optionally substituted C2-C9A heteroarylene group;

L1is optionally substituted C1-C6Alkylene or optionally substituted C1-C6A heteroalkylene group; and is

R3Is optionally substituted C2-C9A heterocyclic group,

or a pharmaceutically acceptable salt thereof.

In some embodiments, the compound has the structure of any one of compounds 1-464 of table 1.

Other embodiments and exemplary methods for synthesizing or producing these compounds are described herein.

Pharmaceutical use

The compounds described herein are useful in the methods of the invention, and while not being bound by theory, are believed to exert their desired effects through their ability to inhibit toxicity in cells caused by protein aggregation (e.g., alpha-synuclein aggregation).

Another aspect of the invention relates to a method of treating and/or preventing a neurological disorder (such as a neurodegenerative disease) in a subject in need thereof. The pathology of neurodegenerative diseases may be characterized by the presence of inclusion bodies in the brain tissue of the affected patient.

In certain embodiments, neurological disorders that can be treated and/or prevented by the methods of the invention include, but are not limited to, Alexander disease, Alper's disease, AD, amyotrophic lateral sclerosis, ataxia telangiectasia, Canavan disease, Cockayne syndrome, corticobasal degeneration, Creutzfeldt-Jakob disease, Huntington disease, Kennedy's disease, Krabbe disease, Lewy body dementia, Machado-Joseph disease, multiple sclerosis, PD, pernevus-merzbachia disease, sanhado-Joseph disease, peyer-merzbachia, primary peyer-merzbachia disease, primary pezier disease, pichoff disease, etc Sherder's disease, Steele-Richardson Olszewski disease, tuberculosis of the spinal cord and Guillain-Barre Syndrome.

The compounds described herein are useful as inhibitors of stearoyl-CoA desaturase (SCD), including SCD1 and/or SCD 5. It is known in the art that SCD inhibitors may be useful in methods of treating and/or preventing SCD-related disorders. SCD-related disorders are described, for example, in U.S. patent No. 8,148,378 and international patent application publication nos. WO 2011/047481, WO 2010/112520, WO 2010/045374, WO 2010/028761; WO 2009150196 and WO 2009/106991. Accordingly, another aspect of the present invention relates to a method of treating and/or preventing an SCD-related disorder in a subject in need thereof.

SCD-related disorders include metabolic disorders (e.g., insulin resistance, diabetes (e.g., type I diabetes, type II diabetes, non-insulin dependent diabetes, gestational diabetes and diabetic complications (e.g., diabetic peripheral neuropathy, diabetic nephropathy, diabetic retinopathy, diabetic macroangiopathy, diabetic angiopathy, and diabetic arteriosclerosis)), hyperglycemia, metabolic syndrome, hyperinsulinemia (hyperinsulinemia), glucose intolerance, body weight disorders (e.g., obesity (e.g., abdominal obesity), excess body weight, cachexia, body mass index, and anorexia), lipid disorders (e.g., abnormal lipid levels (e.g., elevated lipid levels, e.g., in plasma), dyslipidemia (e.g., diabetic dyslipidemia), mixed dyslipidemia, Hyperlipidemia, hypertriglyceridemia, hypoalphalipoproteinemia, hyperbetalipoproteinemia, atherosclerosis, hypercholesterolemia (e.g., familial hypercholesterolemia), low HDL, high LDL, diseases associated with lipid accumulation in the liver, familial histiocytosis, lipoprotein lipase deficiency, polyunsaturated fatty acid (PUFA) disorders, fatty acid desaturation index (e.g., 18:1/18:0 ratio of fatty acids, or other fatty acids) and abnormal lipid metabolism conditions), abnormal plasma lipoprotein disorders, pancreatic beta cell regeneration disorders, fatty liver, nonalcoholic steatohepatitis (NASH), nonalcoholic fatty liver disease (NAFLD), hypertension and microalbuminuria, leptin-related disorders, hyperleptin syndrome, appetite disorders, essential fatty acid deficiencies, and adverse weight gain associated with drug therapy.

Additional SCD-related disorders include cancer, including solid or hematological malignancies (e.g., esophageal, pancreatic, endometrial, kidney, hepatocellular, thyroid, gallbladder, prostate, leukemia (e.g., lymphomas and myelomas), ENT-related cancers, brain, colon, rectal, colorectal, ovarian, uterine, breast, skin, and prostate), neoplasias, malignancies, metastases, tumors (benign or malignant), carcinogenesis, and hepatocellular carcinoma.

In addition, SCD-related disorders include cardiovascular disease (e.g., heart disease, atherosclerosis, hypertension, lipidemia, dyslipidemia, elevated blood pressure, microalbuminuria, hyperuricemia, hypercholesterolemia, hyperlipidemia, hypertriglyceridemia, arteriosclerosis, coronary artery disease, myocardial infarction, diabetic vascular complications, and diabetic arteriosclerosis), inflammation, sinusitis, asthma, pancreatitis, osteoarthritis, rheumatoid arthritis, hepatitis (e.g., hepatitis sexualhepatis), meibomitis, cystic fibrosis, premenstrual syndrome, osteoporosis, thrombosis, cardiovascular risk, weight loss, angina, hypertension, ischemia, cardiac ischemia, reperfusion injury, angioplastic restenosis, infertility, liver disease (e.g., fatty liver, liver cirrhosis, non-alcoholic steatohepatitis), stroke, hepatic fibrosis and hepatitis c-associated steatosis), renal disease (e.g., tubulointerstitial fibrosis, renal lipid accumulation, glomerulosclerosis and proteinuria), osteoarthritis (e.g., knee osteoarthritis), gastroesophageal disease, sleep apnea, secondary hyperparathyroidism to renal osteodystrophy, peripheral vascular disease, cerebrovascular disease (e.g., stroke, ischemic stroke and transient ischemic attack (TlA) and ischemic retinopathy), hyperandrogenism, malignant syndrome, extrapyramidal symptoms, hyperuricemia, hypercoagulability, syndrome X, cataracts, polycystic ovary syndrome, dyspnea, sleep disordered breathing, lumbago, gout, cholelithiasis, myopathy, lipidosis (e.g., carnitine palmitoyl transferase deficiency (CPT I or CPT II)), autoimmune disease (e.g., lupus, host versus graft rejection, and organ transplant rejection), asthma, inflammatory bowel disease, nephropathy, retinopathy, erythropoietic protoporphyrinopathy, iron overload disorders, and hereditary hemochromatosis.

Still further SCD-related disorders include Central Nervous System (CNS) disorders, dementia, schizophrenia, mild cognitive impairment, alzheimer's disease, cerebral amyloid angiopathy, dementia associated with down's syndrome, other neurodegenerative diseases, neurological disorders, ocular diseases, immune disorders, multiple sclerosis, neuropathy, and depression.

Additional SCD-related disorders include skin disorders (e.g., acne vulgaris), psoriasis, hirsutism, rosacea, seborrheic skin, oily skin (seborrhea), seborrheic dermatitis, hyperseborrhea, eczema, keloids, skin aging, diseases associated with production or secretion from mucous membranes, wrinkles, lack of sufficient skin firmness, lack of sufficient skin hydration, insufficient sebum secretion, oily hair, oily shiny skin, greasy-looking hair, and other skin symptoms caused by lipid imbalance).

SCD-related disorders may also include diseases or conditions that are or are associated with viral diseases or infections.

In some embodiments, the SCD-related disorder is acne (e.g., acne vulgaris). In some embodiments, the SCD-related disorder is diabetes (e.g., type II diabetes, including diabetes with inadequate glycemic control). In some embodiments, the SCD-related disorder is non-alcoholic fatty liver disease (NAFLD). In some embodiments, the SCD-related disorder is non-alcoholic steatohepatitis (NASH). In some embodiments, the SCD-related disorder is cancer. In some embodiments, the SCD-related disorder is obesity. In some embodiments, the SCD-related disorder is metabolic syndrome (e.g., dyslipidemia, obesity, insulin resistance, hypertension, microalbuminuria, hyperuricemia, and hypercoagulability), syndrome X, diabetes, insulin resistance, decreased glucose tolerance, non-insulin dependent diabetes mellitus, type II diabetes, type I diabetes, diabetic complications, weight disorders (e.g., obesity, overweight, cachexia, and anorexia), weight loss, body mass index, leptin-related disorders, or skin disorders (e.g., eczema, acne, psoriasis, and keloids). In some embodiments, the SCD-related disorder is diabetes, metabolic syndrome, insulin resistance, obesity, a cardiovascular disorder, a CNS disorder, schizophrenia, or alzheimer's disease.

Combined preparation and application thereof

The compounds of the present invention may be combined with one or more therapeutic agents. In particular, the therapeutic agent can be a therapeutic agent that treats or prophylactically treats any of the neurological disorders described herein.

Combination therapy

The compounds of the present invention may be used alone or in combination with other agents that treat neurological disorders or symptoms associated with neurological disorders or in combination with other types of treatments to treat, prevent and/or reduce the risk of any neurological disorder. In combination therapy, the dosage of one or more therapeutic compounds may be reduced from the standard dosage when administered alone. For example, dosages can be determined empirically for drug combinations and permutations, or can be derived by isobologram analysis (e.g., Black et al, Neurology 65: S3-S6, 2005). In such cases, the dosages of the compounds in the combination should provide a therapeutic effect.

Pharmaceutical composition

Preferably, the compounds of the present invention are formulated into pharmaceutical compositions for administration to human subjects in a biocompatible form suitable for in vivo administration. Thus, in a further aspect, the present invention provides a pharmaceutical composition comprising a compound of the invention in admixture with a suitable diluent, carrier or excipient.

The compounds of the invention may be used in the form of the free base, as salts, solvates and in prodrug form. All forms are within the scope of the invention. As will be appreciated by those skilled in the art, the salts, solvates, or prodrugs thereof may be administered to a patient in a variety of forms depending on the route of administration selected, in accordance with the methods of the present invention. The compounds of the invention may be administered, for example, by oral, parenteral, buccal, sublingual, nasal, rectal, patch, pump or transdermal administration, and the pharmaceutical compositions formulated accordingly. Parenteral administration includes intravenous, intraperitoneal, subcutaneous, intramuscular, epithelial, nasal, intrapulmonary, intrathecal, rectal and topical modes of administration. Parenteral administration may be by continuous infusion over a selected period of time.

The compounds of the invention may be administered orally, for example, with an inert diluent or with an ingestible carrier, or they may be enclosed in hard or soft shell gelatin capsules, or they may be compressed into tablets, or they may be incorporated directly with the food of the diet. For oral therapeutic administration, the compounds of the present invention may be incorporated with excipients and used in the form of ingestible tablets, buccal tablets, troches, capsules, elixirs, suspensions, syrups and wafers.

The compounds of the invention may also be administered parenterally. Solutions of the compounds of the present invention may be prepared in water suitably mixed with a surfactant such as hydroxypropyl cellulose. Dispersions can also be prepared in glycerol, liquid polyethylene glycols, DMSO and mixtures thereof with or without alcohol, and in oils. Under normal conditions of storage and use, these formulations may contain preservatives to prevent microbial growth. Conventional procedures and ingredients for selecting and preparing suitable formulations are described, for example, in Remington's Pharmaceutical Sciences (2003, 20 th edition) and The United States Pharmacopeia, The National Formulary (USP 24 NF19), published 1999.

Pharmaceutical forms suitable for injectable use include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions. In all cases, the form must be sterile and must be fluid to the extent that it can be easily administered via a syringe.

Compositions for nasal administration may conveniently be formulated as aerosols, drops, gels and powders. Aerosol formulations typically comprise a solution or fine suspension of the active substance in a physiologically acceptable aqueous or non-aqueous solvent and are usually presented in sterile form in unit-dose or multi-dose form in a sealed container, which may take the form of a cartridge or refill for an atomising device. Alternatively, the sealed container may be an integral dispensing device, such as a single dose nasal inhaler or an aerosol dispenser equipped with a metering valve intended to be discarded after use. When the dosage form comprises an aerosol dispenser, it will contain a propellant which may be a compressed gas (such as compressed air) or an organic propellant (such as fluorochlorohydrocarbon). Aerosol dosage forms may also take the form of pump atomizers. Compositions suitable for buccal or sublingual administration include tablets, lozenges and pastilles, wherein the active ingredient is formulated with a carrier such as sugar, acacia, tragacanth, gelatin and glycerin. Compositions for rectal administration are conveniently in the form of suppositories with conventional suppository bases such as cocoa butter.

As described herein, the compounds of the present invention can be administered to an animal (e.g., a human) alone or in combination with a pharmaceutically acceptable carrier, in proportions determined by the solubility and chemical nature of the compound, the chosen route of administration, and standard pharmaceutical practice.

Dosage form

The dosage of the compounds of the invention and/or compositions comprising the compounds of the invention may vary depending on a number of factors, such as the pharmacodynamic properties of the compounds; the mode of administration; age, health, and weight of the recipient; the nature and extent of the symptoms; frequency of treatment and type of concurrent treatment (if any); and the clearance of the compound in the animal to be treated. One skilled in the art can determine the appropriate dosage based on the factors described above. The compounds of the invention may be administered initially in suitable dosages which may be adjusted as required in accordance with the clinical response. In general, satisfactory results are obtained when the compounds of the invention are administered to humans in daily doses (measured in solid form), for example, of from 0.05mg to 3000 mg. Dosage ranges include, for example, between 10-1000mg (e.g., 50-800 mg). In some embodiments, 50mg, 100mg, 150mg, 200mg, 250mg, 300mg, 350mg, 400mg, 450mg, 500mg, 550mg, 600mg, 650mg, 700mg, 750mg, 800mg, 850mg, 900mg, 950mg, or 1000mg of the compound is administered. Preferred dosage ranges include, for example, 0.05-15mg/kg or 0.5-15 mg/kg.

Alternatively, the weight of the patient may be used to calculate the dose amount. For example, the dose of the compound or pharmaceutical composition thereof administered to the patient can be in the range of 0.1-50mg/kg (e.g., 0.25-25 mg/kg). In exemplary non-limiting embodiments, the dose can be in the range of 0.5-5.0mg/kg (e.g., 0.5, 1.0, 1.5, 2.0, 2.5, 3.0, 3.5, 4.0, 4.5, or 5.0mg/kg) or 5.0-20mg/kg (e.g., 5.5, 6.0, 6.5, 7.0, 7.5, 8.0, 8.5, 9.0, 9.5, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 mg/kg).

Examples

General scheme 1

Benzylpyrazolidine I can be coupled with acid II under various peptide coupling conditions (e.g., HATU, HBTU, or T3P) to provide the desired amide III.

General scheme 2

4-Nitropyrazole I can be alkylated with an appropriately substituted benzyl or alkyl halide II (X is a halogen such as Cl or Br) in the presence of an inorganic base (e.g., cesium carbonate) to give pyrazole III. Pyrazole III is reduced to amine IV with iron. The amine can be coupled with acid V under various conditions to provide the desired amide VI.

General scheme 3

Mitsunobu reaction of an appropriately substituted benzyl alcohol I with 4-nitropyrazole II gives alkylated pyrazoles III. Pyrazole III is reduced to amine IV with iron or palladium on charcoal. The amine can be coupled with acid V under various peptide coupling conditions (e.g., HATU, HBTU, or T3P) to provide the desired amide VI.

General scheme 4

Halo-substituted isoxazolyl esters I (where X is typically bromo) can be reacted with the appropriately substituted boronic esters IIa or boronic acids IIb under metal-catalyzed conditions to give intermediates III. III is hydrolyzed under various conditions to give the acid IV which can be coupled with an appropriately substituted amine V under various peptide coupling conditions (e.g., HATU) to give the desired amide VI.

General scheme 5

Stannyl isoxazolyl esters I can be reacted with appropriately substituted aromatic halides II (where X is typically Br or Cl) under metal-catalyzed conditions to give intermediates III. Hydrolysis of ethyl ester III under various conditions affords isoxazolic acid IV which can be coupled with appropriately substituted pyrazolylamine Va or azetidinylamine Vb under various peptide coupling conditions (e.g., HATU) to afford the desired pyrazolamide VI or azetidinylamide VII.

General scheme 6

The appropriately substituted acid I can be coupled with pyrazolamine II under various peptide coupling conditions (e.g., HATU) to afford intermediate amide III. Alkylation of pyrazole amide III with an appropriately substituted benzyl halide IV (where X is typically Br or Cl) occurs in the presence of an inorganic base (e.g., potassium carbonate) to provide the desired amide V and a double alkylation by-product VI.

General scheme 7

The nitrile oxide generated in situ from the halogenated oxime I cycloaddition with 1, 2-dichloroethylene II under basic conditions (e.g., triethylamine) gives the isoxazole ester intermediate III. III is hydrolyzed under various conditions (e.g., lithium hydroxide) to provide acid IV, which can be coupled with an appropriately substituted amine V under various peptide coupling conditions (e.g., HATU) to provide intermediate amide VI. Replacement of the chloride ion with an appropriately selected amine VII in the presence of heat under basic conditions (e.g., potassium carbonate) affords amide VIII.

General scheme 8

The nitrile oxide generated in situ from the halogenated oxime I cycloadds with an appropriately substituted alkynyl heterocycle II under basic conditions (e.g., triethylamine) to give the isoxazole ester intermediate III. III is hydrolyzed under various conditions (e.g., lithium hydroxide) to give an acid IV which can be coupled with an appropriately substituted pyrazolylamine Va or azetidine Vb under various peptide coupling conditions (e.g., HATU) to give a pyrazolamide VI or azetidine amide VII.

General scheme 9

The nitrile oxide generated in situ from the halogenated oxime II cycloaddition with the alkynoic acid I under basic conditions (e.g., triethylamine) gives the isoxazolic acid intermediate III. Curtius rearrangement of III gives the protected amine IV. Deprotection of IV under various acidic conditions (e.g., trifluoroacetic acid) affords intermediate amines V which can be double alkylated with bis-bromoalkyl VI under basic conditions (e.g., potassium carbonate) in the presence of heat to provide esters VII. Hydrolysis of VII under various conditions (e.g., lithium hydroxide) affords acid VIII, which can be coupled with appropriately substituted pyrazolamine IX under various peptide coupling conditions (e.g., HATU) to afford pyrazolamide X.

General scheme 10

Appropriately substituted isoxazoloic acid I can be coupled with benzyl azetidine IIa or spirocyclic amine IIb, respectively, under various peptide coupling conditions (e.g., HATU, HBTU, or T3P) to afford the desired benzyl azetidine amide III or spirocyclic amide IV.

General scheme 11

The appropriately substituted vinylphenyl group I can be reacted with chlorosulfonyl isocyanate II to give the appropriately substituted phenylazetidin-2-one III. Reduction of III with lithium aluminum hydride affords 2-substituted azetidines IV, which can be coupled with appropriately substituted acids V under various conditions (e.g., HATU) to afford the desired amides VI.

General scheme 12

An appropriately substituted isoxazolic acid piperidine or piperazine I can be coupled with an appropriately substituted isoxazolic acid II under various peptide coupling conditions (e.g., HATU, HBTU, or T3P) to afford the desired amide III.

General scheme 13

An appropriately substituted aromatic halide I, wherein X is typically bromine, can be reacted with an appropriately substituted boronic acid II under metal-catalyzed conditions to provide an intermediate III. III is hydrogenated in the presence of palladium on charcoal and hydrogen atmosphere to obtain piperidine intermediate IV. Deprotection of IV under acidic conditions (e.g., hydrogen chloride) affords piperidine V, which can be coupled with an appropriately substituted acid VI under various peptide coupling conditions (e.g., HATU) to afford the desired amide VII.

Example 1 preparation of N- (3, 4-difluorobenzyl) -N- (1- (3, 4-difluorobenzyl) -1H-pyrazol-4-yl) -5- (pyridin-2-yl) isoxazole-3-carboxamide (62)

Step 1: preparation of N- (1H-pyrazol-4-yl) -5- (pyridin-2-yl) isoxazole-3-carboxamide

To a mixture of 5- (pyridin-2-yl) isoxazole-3-carboxylic acid (190mg, 1.0mmol), 1H-pyrazol-4-amine (91mg, 1.1mmol) and 2- (7-aza-1H-benzotriazol-1-yl) -1, 1, 3, 3-tetramethyluronium hexafluorophosphate (418mg, 1.1mmol) in N, N-dimethylformamide (5mL) was added N, N-diisopropylethylamine (387mg, 3.0 mmol). The mixture was stirred at 23 ℃ for 2 h. The reaction mixture was added dropwise to ice-cold 0.5 aqueous hydrochloric acid (50 mL). The solid precipitate was collected by filtration and dried in vacuo to give N- (1H-pyrazol-4-yl) -5- (pyridin-2-yl) isoxazole-3-carboxamide as a pale yellow solid (215mg, 0.84mmol, 84%). LCMS (ESI) M/z 256.1[ M + H ]]+

Step 2: preparation of N- (1- (3, 4-difluorobenzyl) -1H-pyrazol-4-yl) -5- (pyridin-2-yl) isoxazole-3-carboxamide and N- (3, 4-difluorobenzyl) -N- (1- (3, 4-difluorobenzyl) -1H-pyrazol-4-yl) -5- (pyridin-2-yl) isoxazole-3-carboxamide

To a suspension of N- (1H-pyrazol-4-yl) -5- (pyridin-2-yl) isoxazole-3-carboxamide (0.215g, 0.84mmol) and cesium carbonate (0.547g, 1.68mmol) in N, N-dimethylformamide (5mL) at 0 ℃ was added 4- (bromomethyl) -1, 2-difluorobenzene (0.173g, 0.84mmol) dropwise. The mixture was stirred at 23 ℃ for 18 h. After filtration, the residue was purified by preparative-HPLC (Sunfire preparative C1810 μm OBD 19 x 250 mm; mobile phase: [ water (0.05% trifluoroacetic acid-acetonitrile) ](ii) a B%: 60% -88%, 15min) to afford N- (1- (3, 4-difluorobenzyl) -1H-pyrazol-4-yl) -5- (pyridin-2-yl) isoxazole-3-carboxamide as a white solid (76.4mg, 0.20mmol, 24%).1H NMR (500MHz, dimethylsulfoxide-d)6)δ11.08(s,1H),8.77(d,J=4.5Hz,1H),8.24(s,1H),8.16-7.95(m,2H),7.70(s,1H),7.57(ddd,J=7.5,4.8,1.1Hz,1H),7.49(s,1H),7.43(dd,J=8.5,2.3Hz,1H),7.34(d,J=1.3Hz,1H),7.17-6.99(m,1H),5.34(s,2H)。LCMS(ESI)m/z:382.1[M+H]+And provided N- (3, 4-difluorobenzyl) -N- (1- (3, 4-difluorobenzyl) -1H-pyrazol-4-yl) -5- (pyridin-2-yl) isoxazole-3-carboxamide as a white solid (40.8mg, 0.08mmol, 10%).1H NMR (500MHz, dimethylsulfoxide-d)6)δ8.92-8.18(m,1H),8.01(ddd,J=9.4,8.8,2.7Hz,1H),7.89(d,J=7.9Hz,1H),7.73(d,J=57.1Hz,1H),7.60-7.47(m,1H),7.48-7.29(m,3H),7.17(s,1H),7.11-6.86(m,2H),6.71(s,1H),5.13(dd,J=94.7,71.8Hz,4H)。LCMS(ESI)m/z:508.0[M+H]+

Example 2 preparation of N- (1-benzylpyrazol-4-yl) -5- (2-fluorophenyl) isoxazole-3-carboxamide (38)

Step 1: preparation of ethyl 5- (2-fluorophenyl) isoxazole-3-carboxylate

A mixture of ethyl 5-tributylstannyl isoxazole-3-carboxylate (0.500g, 1.16mmol), 1-fluoro-2-iodo-benzene (0.272mL, 2.32mmol), bis (triphenylphosphine) palladium (II) dichloride (0.041g, 0.058mmol) in toluene (5mL) was purged with nitrogen (3 ×), and then the mixture was stirred at 110 ℃ under nitrogen for 12 h. The mixture was cooled to 15 ℃ and then poured into ice-water (5 mL). The aqueous phase was extracted with ethyl acetate (10 mL. times.3). The combined organic layers were washed with brine (5mL), dried over anhydrous sodium sulfate, filtered and concentrated in vacuo to give the crude product. The crude residue was purified by column chromatography (ISCO, 12g silica, 0-50% ethyl acetate in petroleum ether, 30min gradient) to give ethyl 5- (2-fluorophenyl) isoxazole-3-carboxylate (0.200g, crude) as a white solid. LCMS (ESI) M/z 236.1[ M + H ] ]+

Step 2: preparation of 5- (2-fluorophenyl) isoxazole-3-carboxylic acid

To a stirred solution of ethyl 5- (2-fluorophenyl) isoxazole-3-carboxylate (0.180g, 0.765mmol) in tetrahydrofuran (0.5mL) was added lithium hydroxide (2M, 0.77 mL). The mixture was stirred at 15 ℃ for 2 h. The mixture was diluted with water and adjusted to pH 5 by addition of aqueous hydrogen chloride (2M, 1 mL). The aqueous phase was extracted with ethyl acetate (5 mL. times.3). The combined organic phases were washed with brine (5mL), dried over anhydrous sodium sulfate, filtered and concentrated in vacuo to afford 5- (2-fluorophenyl) isoxazole-3-carboxylic acid (0.150g, crude) as a white solid. Used in the next step without further purification.

And step 3: preparation of ethyl N- (1-benzylpyrazol-4-yl) -5- (2-fluorophenyl) isoxazole-3-carboxamide

To a stirred solution of 5- (2-fluorophenyl) isoxazole-3-carboxylic acid (0.140g, 0.676mmol) in dichloromethane (2mL) was added propylphosphonic anhydride (0.482g, 0.811mmol, 50% wt), triethylamine (0.188mL, 1.35mmol) and 1-benzylpyrazol-4-amine (0.117g, 0.676 mmol). The mixture was stirred at 15 ℃ for 1 h. The crude residue was purified by preparative-HPLC (YMC-Actus Triart C18150X 305 um column; 30-70% acetonitrile in 10mM aqueous ammonium acetate, 10min gradient) to give N- (1-benzylpyrazol-4-yl) -5- (2-fluorophenyl) isoxazole-3-carboxamide (0.172g, 0.469mmol, 69%) as a white solid. 1H NMR (400MHz, dimethylsulfoxide-d 6), delta 11.05(s, 1H), 8.18(s, 1H), 8.08-7.97(m, 1H), 7.68(s, 1H), 7.66-7.59(m, 1H), 7.53-7.40(m, 2H), 7.38-7.21(m, 6H), 5.34(s, 2H); LCMS (ESI) M/z 363.0[ M + H ]]+

Example 3 preparation of N- (1-benzylpyrazol-4-yl) -5- (2-pyridinyl) isoxazole-3-carboxamide (8)

To a stirred solution of 1-benzylpyrazol-4-amine (0.120g, 0.693mmol) and 5- (pyridin-2-yl) isoxazole-3-carboxylic acid (0.157g, 0.831mmol) in N, N-dimethylformamide (1mL) was added N, N' -tetramethyl-O- (1H-benzotriazol-1-yl) uronium hexafluorophosphate (0.315g, 0.831mmol) and diisopropylethylamine (0.242mL, 1.39mmol) at 15 ℃ and then stirred for 3H at 15 ℃. The reaction mixture was directly purified by preparative-HPLC (Agela Venusil XBP C18150X 255 um column; 50% -75% acetonitrile in 10mM 0.04% ammonium hydroxide, 10min gradient) to give N- (1-benzylpyrazol-4-yl) -5- (2-pyridinyl) isoxazole-3-carboxamide (0.068g, 0.197mmol, 29%) as a pale yellow solid.1H NMR (400MHz, chloroform-d) δ 8.76(d, J ═ 4.5Hz, 1H), 8.55(br.s, 1H), 8.06(s, 1H), 7.97-7.92(m, 1H), 7.91-7.84(m, 1H), 7.64(s, 1H), 7.44-7.31(m, 5H), 7.30-7.26(m, 2H), 5.32(s, 2H); LCMS (ESI) M/z 346.0[ M + H ] ]+

EXAMPLE 4 preparation of 5- (1, 3-benzodioxol-5-yl) -N- (1-benzylpyrazol-4-yl) isoxazole-3-carboxamide (6)

Step 1: preparation of 5- (1, 3-benzodioxol-5-yl) -N- (1-benzylpyrazol-4-yl) isoxazole-3-carboxamide

To a stirred solution of 1-benzylpyrazol-4-amine (0.120g, 0.693mmol) and 5- (pyridin-2-yl) isoxazole-3-carboxylic acid (0.157g, 0.831mmol) in N, N-dimethylformamide (1mL) was added N, N' -tetramethyl-O- (1H-benzotriazol-1-yl) uronium hexafluorophosphate (0.315g, 0.831mmol) and diisopropylethylamine (0.242g, 1.39mmol) at 15 ℃. The reaction mixture was stirred at 15 ℃ for 3h and then purified directly by preparative-HPLC (Agela Venusil XBP C18150X 255 um column; acetonitrile in 10mM 0.04% ammonium hydroxide, 10min gradient; 50% -75%) to give 5- (1, 3-benzodioxol-5-yl) -N- (1-benzylpyrazol-4-yl) isoxazole-3-carboxamide (0.070g, 0.181mmol, 31%) as a white solid.1H NMR (400MHz, chloroform-d) δ 8.53(s, 1H), 8.04(s, 1H), 7.63(s, 1H), 7.41-7.30(m, 4H), 7.30-7.25(m, 3H), 6.93(d, J ═ 8.2Hz, 1H), 6.88(s, 1H), 6.07(s, 2H), 5.32(s, 2H); LCMS (ESI) M/z 389.1[ M + H ] ]+

Example 5 preparation of N- (1-benzylpyrazol-4-yl) -5-phenyl-isoxazole-3-carboxamide (7)

Step 1: preparation of 1-benzyl-4-nitro-pyrazoles

To a stirred solution of 4-nitro-1H-pyrazole (2.00g, 17.7mmol) in N, N-dimethylformamide (15mL) at 0 deg.C was added sodium hydride (0.778g, 19.0mmol, 60% purity in mineral oil). The reaction mixture was stirred at 15 ℃ for 1h, then cooled to 0 ℃ before benzyl bromide (2.10mL, 17.7mmol) was added. The reaction mixture was warmed to 15 ℃ and stirred for 15h, then quenched by addition of ice water (5mL) and extracted with ethyl acetate (15 mL. times.3). The combined organic layers were washed with water (10mL × 2) and brine (5mL), dried over sodium sulfate, filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (ISCO, 20g silica, 0-30% ethyl acetate in petroleum ether, 20min gradient) to give 1-benzyl-4-nitro-pyrazole as a white solid (2.80g, 13.8mmol, 78%).1H NMR (400MHz, chloroform-d) Δ 8.10(s, 1H), 8.04(s, 1H), 7.45-7.39(m, 3H), 7.32-7.28(m, 2H), 5.31(s, 2H); LCMS (ESI) M/z 204.1[ M + H ]]+

Step 2: preparation of 1-benzylpyrazole-4-amine

To a solution of 1-benzyl-4-nitro-pyrazole (1.50g, 7.38mmol) in methanol (10mL) under nitrogen was added palladium on activated carbon (0.500g, 10 wt%). The suspension was purged several times with hydrogen. The mixture was stirred under a hydrogen balloon at 15 ℃ for 16h, then purged with nitrogen and filtered. The filtrate was concentrated in vacuo to give 1-benzylpyrazol-4-amine (1.10g, 6.35mmol, 86%) as a pink solid. LCMS (ESI) M/z 174.1[ M + H ] ]+

And step 3: preparation of N- (1-benzylpyrazol-4-yl) -5-phenyl-isoxazole-3-carboxamide

To 1-benzylpyrazol-4-amine (0.120g, 0.693mmol) and 5-phenylisoxazole-3-carboxylic acid (0) was added at 15 ℃.157g, 0.831mmol) in N, N-dimethylformamide (1mL) were added N, N, N ', N' -tetramethyl-O- (1H-benzotriazol-1-yl) uronium hexafluorophosphate (0.315g, 0.831mmol) and diisopropylethylamine (0.242mL, 1.39mmol), followed by stirring at 15 ℃ for 3H. The reaction was purified by preparative-HPLC (Agela Venusil XBP C18150X 255 um column; 50% -75% acetonitrile in 10mM 0.04% ammonium hydroxide, 10min gradient) to give N- (1-benzylpyrazol-4-yl) -5-phenyl-isoxazole-3-carboxamide (0.087g, 0.253mmol, 36%) as a pale yellow solid.1H NMR (400MHz, chloroform-d) Δ 8.57(br.s, 1H), 8.05(s, 1H), 7.86-7.82(m, 2H), 7.65(s, 1H), 7.54-7.50(m, 3H), 7.41-7.33(m, 3H), 7.30-7.27(m, 2H), 7.03(s, 1H), 5.32(s, 2H); LCMS (ESI) M/z 345.1[ M + H ]]+

EXAMPLE 6 preparation of 5- (benzo [ d ] [1,3] dioxol-5-yl) -N- (1-benzyl-3, 5-dimethyl-1H-pyrazol-4-yl) isoxazole-3-carboxamide (6)

Diisopropylethylamine (121mL, 0.696mmol) was added to a solution of 1-benzyl-3, 5-dimethyl-1H-pyrazol-4-amine (0.700g, 0.348mmol), 5- (1, 3-benzodioxol-5-yl) isoxazole-3-carboxylic acid (0.0811g, 0.348mmol), N' -tetramethyl-O- (1H-benzotriazol-1-yl) uronium hexafluorophosphate (0.132g, 0.348mmol) in N, N-dimethylformamide (0.5mL) at 0 ℃. The reaction mixture was then stirred at 15 ℃ for 0.5 h. The mixture was passed through preparative-HPLC (column: Waters Xbridge 150X 255 u; mobile phase: [ water (10mM ammonium bicarbonate) -acetonitrile ](ii) a B%: 35% -65%, 12min gradient) to obtain 5- (benzo [ d ] as a white solid][1,3]Dioxol-5-yl) -N- (1-benzyl-3, 5-dimethyl-1H-pyrazol-4-yl) isoxazole-3-carboxamide (0.0892g, 0.208mmol, 60%).1H NMR (400MHz, dimethyl sulfoxide-d 6) δ 9.93(s, 1H), 7.52-7.45(m, 2H), 7.36-7.23(m, 4H), 7.15(d, J ═ 7.4Hz, 2H), 7.08(d, J ═ 7.8Hz, 1H), 6.12(s, 2H), 5.21(s, 2H), 2.06(s, 3H), 2.03(s, 3)H);LCMS(ESI)m/z:417.2[M+H]+

Example 7 preparation of N- (1-benzyl-1H-pyrazol-4-yl) -5- (furan-2-yl) isoxazole-3-carboxamide (4)

To a mixture of 5- (furan-2-yl) isoxazole-3-carboxylic acid (0.090g, 0.502mmol), N' -tetramethyl-O- (1H-benzotriazol-1-yl) uronium hexafluorophosphate (0.191g, 0.502mmol) and diisopropylethylamine (0.176mL, 1.00mmol) in N, N-dimethylformamide (1mL) was added 1-benzyl-1H-pyrazol-4-amine (0.087g, 0.502mmol) at 0 ℃. The reaction mixture was stirred at 15 ℃ for 12 h. The residue was purified by preparative-HPLC (column: Luna C8100X 305 u; mobile phase: [ water (10mM ammonium bicarbonate) -acetonitrile](ii) a B%: 36% -66%, 12min gradient) to afford N- (1-benzyl-1H-pyrazol-4-yl) -5- (furan-2-yl) isoxazole-3-carboxamide as a pink solid (0.055g, 0.162mmol, 32%). 1H NMR (400MHz, chloroform-d) δ 8.47(br.s., 1H), 8.02(s, 1H), 7.61(d, J ═ 6.6Hz, 2H), 7.40-7.31(m, 3H), 7.26(br.s., 1H), 6.99(d, J ═ 3.5Hz, 1H), 6.91(s, 1H), 6.65-6.53(m, 1H), 5.31(s, 2H); LCMS (ESI) M/z 335.1[ M + H ]]+

EXAMPLE 8 preparation of (R) -N- (1-benzyl-1H-pyrazol-4-yl) -5- (3-hydroxypyrrolidin-1-yl) isoxazole-3-carboxamide (186)

A mixture of N- (1-benzyl-1H-pyrazol-4-yl) -5-chloroisoxazole-3-carboxamide (130mg, 0.43mmol), (R) -pyrrolidin-3-ol hydrochloride (159mg, 1.30mmol) and potassium carbonate (90mg, 0.64mmol) in N, N-dimethylformamide (5mL) was heated in a sealed tube at 50 ℃ for 18H. After filtration, the filtrate was passed through preparative-HPLC (column: Sunfire preparative C1810 μm OBD 19 x 250 mm; mobile phase: [ water (0.05% trifluoroacetic acid-acetonitrile)](ii) a B%: 60% -88%, 15min) purification to giveWhite solid N- (1-benzyl-1H-pyrazol-4-yl) -5- (pyrrolidin-1-yl) isoxazole-3-carboxamide (70mg, 0.20mmol, 46%).1H NMR (500MHz, dimethylsulfoxide-d)6)δ10.62(s,1H),8.10(s,1H),7.64(s,1H),7.29(ddd,J=33.7,20.4,7.0Hz,5H),5.34(d,J=37.2Hz,3H),4.47-4.28(m,1H),3.67-3.29(m,6H),3.25(d,J=10.5Hz,1H),2.02(dd,J=8.7,4.4Hz,1H),1.95-1.75(m,1H);LCMS(ESI)m/z:354.1[M+H]+

Example 9 preparation of N- (1-benzyl-1H-pyrazol-4-yl) -5- (thiazol-2-yl) isoxazole-3-carboxamide (110)

To a solution of 1-benzyl-1H-pyrazol-4-amine (0.033g, 0.157mmol), 5- (thiazol-2-yl) isoxazole-3-carboxylic acid (30.8mg, 0.157mmol), and diisopropylethylamine (50.8mg, 0.393mmol) in N, N' -dimethylformamide (0.5mL) at 25 deg.C was added 1- [ bis (dimethylamino) methylene ] -4-amine ]-1H-1, 2, 3-triazolo [4, 5-b]Pyridinium 3-oxide hexafluorophosphate (0.123g, 0.157 mmol). The reaction mixture was stirred at 25 ℃ for 16 h. The reaction mixture was quenched with water (1mL) and the aqueous layer was extracted with ethyl acetate (5 mL. times.3). The combined organic layers were washed with brine, dried over magnesium sulfate, filtered and concentrated in vacuo. The crude residue was purified by column chromatography (ISCO, 12g silica, eluting with 40% ethyl acetate/hexanes for 20min) to give N- (1-benzyl-1H-pyrazol-4-yl) -5- (thiazol-2-yl) isoxazole-3-carboxamide (24.7mg, 0.0704mmol, 45%) as a yellow solid.1H NMR (300MHz, dimethylsulfoxide-d)6)δ11.10(s,1H),8.24-8.03(m,3H),7.67(d,J=0.7Hz,1H),7.49(s,1H),7.46-7.17(m,5H),5.34(s,2H);LCMS(ESI)m/z:352.2[M+H]+

Example 10 preparation of N- (1-benzyl-1H-pyrazol-4-yl) -5-isopropylisoxazole-3-carboxamide (117)

To 1-benzyl-1H-pyrazol-4-amine (0.050g, 0.289mmol), 5-isopropylisoxazole-3-carboxylic acid (44.7mg, 0.289mmol) and 1- [ bis (dimethylamino) methylene at 25 deg.C]-1H-1, 2, 3-triazolo [4, 5-b]Diisopropylethylamine (0.075mL, 0.433mmol) was added to a solution of pyridinium 3-oxide hexafluorophosphate (109mg, 0.289mmol) in N, N' -dimethylformamide (1 mL). The reaction mixture was stirred at 25 ℃ for 16 h. The reaction mixture was quenched with water (1 mL). The aqueous layer was extracted with ethyl acetate (5 mL. times.3). The combined organic layers were washed with brine, dried over magnesium sulfate, filtered and concentrated in vacuo. The crude residue was purified by column chromatography (ISCO, 12g silica, eluting with 40% ethyl acetate/hexanes for 20min) to give N- (1-benzyl-1H-pyrazol-4-yl) -5-isopropylisoxazole-3-carboxamide (63.3mg, 0.204mmol, 70%) as an off-white solid. 1H NMR (300MHz, dimethylsulfoxide-d)6)δ10.87(s,1H),8.13(d,J=1.6Hz,1H),7.64(d,J=1.7Hz,1H),7.40-7.05(m,5H),6.63(t,J=1.2Hz,1H),5.31(d,J=1.6Hz,2H),3.16(p,J=6.8Hz,1H),1.28(dd,J=7.0,1.7Hz,6H);LCMS(ESI)m/z:311.3[M+H]+

Example 11 preparation of N- (1-benzyl-1H-pyrazol-4-yl) -5- (tert-butyl) isoxazole-3-carboxamide (118)

To 1-benzyl-1H-pyrazol-4-amine (0.050g, 0.289mmol), 5- (tert-butyl) isoxazole-3-carboxylic acid (48.8mg, 0.289mmol) and 1- [ bis (dimethylamino) methylene at 25 deg.C]-1H-1, 2, 3-triazolo [4, 5-b]Diisopropylethylamine (0.075mL, 0.433mmol) was added to a solution of pyridinium 3-oxide hexafluorophosphate (109mg, 0.289mmol) in N, N' -dimethylformamide (1 mL). The reaction mixture was stirred at 25 ℃ for 16 h. The reaction mixture was quenched with water (1 mL). The aqueous layer was extracted with ethyl acetate (5 mL. times.3). The combined organic layers were washed with brine, dried over magnesium sulfate, filtered and concentrated in vacuo. Removing the crude residueThe material was purified by column chromatography (ISCO, 12g silica, eluting with 40% ethyl acetate/hexanes for 20min) to give N- (1-benzyl-1H-pyrazol-4-yl) -5- (tert-butyl) isoxazole-3-carboxamide as a yellow solid (65.3mg, 0.201mmol, 70%).1H NMR (300MHz, dimethylsulfoxide-d)6)δ10.86(s,1H),8.13(s,1H),7.63(s,1H),7.42-7.13(m,5H),6.62(d,J=1.2Hz,1H),5.31(s,2H),1.34(d,J=1.1Hz,10H)。M+H=325.3;LCMS(ESI)m/z:325.3[M+H]+

Example 12 preparation of N- (1-benzyl-1H-pyrazol-4-yl) -5- (pyrrolidin-1-yl) isoxazole-3-carboxamide (178)

Step 1: preparation of ethyl 5-chloroisoxazole-3-carboxylate

A solution of triethylamine (26.2mL, 182mmol) in 1, 1-dichloroethylene (150mL) was added dropwise over 2h to a solution of ethyl (Z) -2-chloro-2- (hydroxyimino) acetate (11.0g, 72.9mmol) in 1, 1-dichloroethylene (100 mL). The mixture was stirred at 23 ℃ for 18 h. After concentration, the residue was dissolved in ethyl acetate (300mL) and washed with brine (100mL), dried over sodium sulfate, filtered and concentrated in vacuo. The residue was purified by column chromatography (silica, petroleum ether/ethyl acetate-20/1) to give ethyl 5-chloroisoxazole-3-carboxylate (1.40g, 8.00mmol, 11%) as a pale yellow oil. LCMS (ESI) M/z 176.1[ M + H ]]+

Step 2: preparation of 5-chloroisoxazole-3-carboxylic acid

Ice-cold to ethyl 5-chloroisoxazole-3-carboxylate (2.80g, 16.0mmol) in 1, 4-dioxane (18mL)To the solution was added a solution of lithium hydroxide monohydrate (1.34g, 32.0mmol) in water (18 mL). The mixture was stirred at 0 ℃ for 1 h. After concentration, the residue was diluted with ethyl acetate/water (1/1, 250mL) and adjusted to pH 5 with 0.5N aqueous hydrochloric acid. The aqueous layer was extracted with ethyl acetate (2X 100 mL). The combined organic layers were washed with brine (100mL), dried over sodium sulfate, filtered and concentrated to afford 5-chloroisoxazole-3-carboxylic acid as a pale yellow solid (2.00g, 13.6mmol, 85%). LCMS (ESI) M/z 148.1[ M + H ] ]+. This material was used in the next step without further purification.

And step 3: preparation of N- (1-benzyl-1H-pyrazol-4-yl) -5-chloroisoxazole-3-carboxamide

To a solution of 5-chloroisoxazole-3-carboxylic acid (2.00g,13.6mmol), 1-benzyl-1H-pyrazol-4-amine hydrochloride (2.84g,13.6mmol) and 4- (4, 6-dimethoxy-1, 3, 5-triazin-2-yl) -4-methylmorpholinium chloride (4.01g,13.6mmol) in tetrahydrofuran (50mL) was added 4-methylmorpholine (4.12g,40.8 mmol). The mixture was stirred at 23 ℃ for 1 h. The reaction mixture was diluted with ethyl acetate (300mL) and washed with 0.5N aqueous hydrochloric acid (100mL), water (100mL) and brine (100 mL). The combined organic layers were dried over sodium sulfate, filtered and concentrated in vacuo. The residue was purified by column chromatography (silica, petroleum ether/ethyl acetate-2/1) to give N- (1-benzyl-1H-pyrazol-4-yl) -5-chloroisoxazole-3-carboxamide as a white yellow solid (2.38g,7.88mmol, 58%). LCMS (ESI) M/z 303.1[ M + H ]]+

And 4, step 4: preparation of N- (1-benzyl-1H-pyrazol-4-yl) -5- (pyrrolidin-1-yl) isoxazole-3-carboxamide

N- (1-benzyl-1H-pyrazol-4-yl) -5-chloroisoxazole-3-carboxamide (130mg, 0.43mmol), pyrrolidine (180mg, 1.30mmol) and potassium carbonate (90mg, 0.64mmol) in N, N-dimethylformamide (5mL) was stirred at 60 ℃ for 18 h. After filtration, the filtrate was passed through preparative-HPLC (column: Sunfire preparative C1810 μm OBD 19X 250 mm; mobile phase: [ water (0.05% trifluoroacetic acid-acetonitrile)](ii) a B%: 60% -88%, 15min) to afford N- (1-benzyl-1H-pyrazol-4-yl) -5- (pyrrolidin-1-yl) isoxazole-3-carboxamide as a white solid (60mg, 0.18mmol, 41%).1H NMR (500MHz, dimethylsulfoxide-d)6)δ10.62(s,1H),8.11(s,1H),7.64(s,1H),7.44-6.90(m,5H),5.34(d,J=34.0Hz,3H),3.37(t,J=6.6Hz,5H),2.00-1.74(m,4H);LCMS(ESI)m/z:338.1[M+H]+

Example 13 preparation of N- (1-benzyl-1H-pyrazol-4-yl) -5- (dimethylamino) isoxazole-3-carboxamide (203)

To a solution of N- (1-benzyl-1H-pyrazol-4-yl) -5-chloroisoxazole-3-carboxamide (0.160g, 0.53mmol) in N, N-dimethylformamide (15mL) was added dimethylamine hydrochloride (0.130g, 1.59mmol) and potassium carbonate (0.219g, 1.59 mmol).The reaction mixture was heated at 80 ℃ for 17 h. The reaction mixture was filtered and concentrated in vacuo. The crude sample was dissolved in a minimal amount of N, N-dimethylformamide and purified via preparative-HPLC (Boston C1821 × 250mm 10 μm column; acetonitrile/0.01% aqueous trifluoroacetic acid) to give N- (1-benzyl-1H-pyrazol-4-yl) -5- (dimethylamino) isoxazole-3-carboxamide (40.4mg, 0.13mmol, 25%) as a white solid. 1H NMR (400MHz, dimethylsulfoxide-d)6)δ10.61(s,1H),8.10(s,1H),7.74-7.53(m,1H),7.43-7.01(m,5H),5.50(s,1H),5.31(s,2H),2.98(s,6H);LCMS(ESI)m/z:312.1[M+H]+

Example 14 preparation of N- (1-benzyl-1H-pyrazol-4-yl) -5-cyclopropylisoxazole-3-carboxamide (119)

To 1-benzyl-1H-pyrazol-4-amine (0.050g, 0.289mmol), 5-cyclopropylisoxazole-3-carboxylic acid (44.1mg, 0.289mmol) and 1- [ bis (dimethylamino) methylene at 25 deg.C]-1H-1, 2, 3-triazolo [4, 5-b]Diisopropylethylamine (0.075mL, 0.433mmol) was added to a solution of pyridinium 3-oxide hexafluorophosphate (109mg, 0.289mmol) in N, N' -dimethylformamide (1 mL). The reaction mixture was stirred at 25 ℃ for 16 h. The reaction mixture was quenched with water (1 mL). The aqueous layer was extracted with ethyl acetate (5 mL. times.3). The combined organic layers were washed with brine, dried over magnesium sulfate, filtered and concentrated in vacuo. The crude residue was purified by column chromatography (ISCO, 12g silica, eluting with 40% ethyl acetate/hexanes for 20min) to give N- (1-benzyl-1H-pyrazol-4-yl) -5-cyclopropylisoxazole-3-carboxamide as an off-white solid (12.4mg, 0.041mmol, 14%).1H NMR (300MHz, chloroform-d) δ 8.42(s, 1H), 8.00(s, 1H), 7.59(s, 1H), 7.37-7.18(m, 5H), 6.39(s, 1H), 5.30(s, 2H), 2.11(tt, J ═ 8.5, 5.0Hz, 1H), 1.23-1.08(m, 2H), 1.08-0.92(m, 2H); LCMS (ESI) M/z 309.2[ M + H ] ]+

Example 15 preparation of N- (1-benzyl-1H-pyrazol-4-yl) -5- (pyridin-4-yl) isoxazole-3-carboxamide (124)

To 1-benzyl-1H-pyrazol-4-amine (0.050g, 0.289mmol), 5- (pyridin-4-yl) isoxazole-3-carboxylic acid (54.8mg, 0.289mmol) and 1- [ bis (dimethylamino) methylene ] at 25 deg.C]-1H-1, 2, 3-triazolo [4, 5-b]Diisopropylethylamine (0.075mL, 0.433mmol) was added to a solution of pyridinium 3-oxide hexafluorophosphate (109mg, 0.289mmol) in N, N' -dimethylformamide (1 mL). The reaction mixture was stirred at 25 ℃ for 16 h. The reaction mixture was quenched with water (1 mL). The aqueous layer was extracted with ethyl acetate (5 mL. times.3). The combined organic layers were washed with brine, dried over magnesium sulfate, filtered and concentrated in vacuo. The crude residue was purified by column chromatography (ISCO, 12g silica, 60% ethyl acetate/hexane)Alkane elution 20min) to afford N- (1-benzyl-1H-pyrazol-4-yl) -5- (pyridin-4-yl) isoxazole-3-carboxamide as a pale orange solid (26mg, 0.075mmol, 26%).1H NMR (300MHz, dimethylsulfoxide-d)6)δ11.10(s,1H),8.85-8.73(m,2H),8.18(s,1H),8.01-7.89(m,2H),7.75(s,1H),7.68(s,1H),7.40-7.19(m,5H),5.34(s,2H);LCMS(ESI)m/z:346.3[M+H]+

Example 16 preparation of N- (1-benzyl-1H-pyrazol-4-yl) -5- (2- (methoxymethyl) thiazol-5-yl) isoxazole-3-carboxamide (140)

Step 1: preparation of 2- (methoxymethyl) thiazole

To a solution of thiazol-2-ylmethanol (1.15g, 10mmol) in dry N, N-dimethylformamide (10mL) at 0 deg.C was added sodium hydride (60% in mineral oil, 0.52g, 13mmol) portionwise. After addition, the mixture was stirred at 0 ℃ for 20min, then iodomethane (1.49mL, 24mmol) was added dropwise. The reaction mixture was stirred at 0 ℃ for 2 h. The reaction was quenched with ethyl acetate/water (30mL/20mL) and extracted with ethyl acetate (20 mL. times.2). The combined organic phases were washed with brine (40mL), dried over sodium sulfate, filtered and concentrated in vacuo. The residue was purified by column chromatography (Biotage, 40g silica, eluting with 25% to 40% ethyl acetate in petroleum ether) to give 2- (methoxymethyl) thiazole (0.74g, 5.74mmol, 57%) as a colourless oil. LCMS (ESI) M/z 130.1[ M + H ]]+

Step 2: preparation of 2- (methoxymethyl) thiazole-5-carbaldehyde

To a solution of 2- (methoxymethyl) thiazole (0.73g, 5.66mmol) in anhydrous tetrahydrofuran (20mL) under nitrogen at-78 deg.C was added dropwise n-butyllithium (2.5M solution in hexane, 3.4mL, 8.48 mmol). After addition, the reaction was stirred at-78 ℃ for 1h, then N, N-dimethylformamide (0.87mL, 11.3mmol) was added dropwise. The reaction mixture was stirred at-78 ℃ for 2 h. The reaction was quenched with aqueous ammonium chloride (30mL) and extracted with ethyl acetate (30 mL. times.2). The combined organic phases were washed with brine (40mL), dried over sodium sulfate, filtered and concentrated in vacuo. The residue was purified by column chromatography (Biotage, 40g silica, eluting with 30% to 40% ethyl acetate in petroleum ether) to give 2- (methoxymethyl) thiazole-5-carbaldehyde as a yellow oil (0.48g, 3.06mmol, 53%). LCMS (ESI) M/z 158.1[ M + H ] ]+

And step 3: preparation of 5-ethynyl-2- (methoxymethyl) thiazole

To a solution of 2- (methoxymethyl) thiazole-5-carbaldehyde (0.46g, 2.93mmol) in methanol (30mL) at 25 ℃ were added dimethyl 1-diazo-2-oxopropylphosphonate (0.84g, 4.39mmol) and potassium carbonate (1.21g, 8.79 mmol). The reaction mixture was stirred at 25 ℃ for 3 h. Volatiles were removed in vacuo. The crude residue was taken up in ethyl acetate (40mL) and washed with water (30mL × 2) and brine (20 mL). The organic phase was dried over sodium sulfate, filtered and concentrated to give 5-ethynyl-2- (methoxymethyl) thiazole (0.46g, 2.93mmol, crude) as a yellow oil. LCMS (ESI) M/z 154.1[ M + H ]]+. This material was used directly in the next step without further purification.

And 4, step 4: preparation of 5-ethynyl-2- (methoxymethyl) thiazole

5-ethynyl-2- (methoxymethyl) silane at 25 ℃ under nitrogenYl) thiazole (0.41g, 2.68mmol) in N, N-dimethylformamide (10mL) ethyl 2-chloro-2- (hydroxyimino) acetate (0.27g, 1.78mmol) in N, N-dimethylformamide (3mL) was added dropwise. After addition, the reaction was stirred at 25 ℃ for 20min and then heated to 90 ℃. A solution of triethylamine (0.54g, 5.36mmol) in dimethylformamide (6mL) was slowly added dropwise. After addition, the reaction was heated at 90 ℃ for 18 h. The reaction mixture was cooled, diluted with ethyl acetate/water (40mL/40mL) and extracted twice with ethyl acetate (30 mL. times.2). The combined organic phases were washed with brine (40mL), dried over sodium sulfate, filtered and concentrated in vacuo. The residue was purified by column chromatography (Biotage, 40g silica, eluting with 20% to 30% ethyl acetate in petroleum ether) to give ethyl 5- (2- (methoxymethyl) thiazol-5-yl) isoxazole-3-carboxylate as a white solid (0.17g, 0.63mmol, 2 steps total 20.7%). LCMS (ESI) M/z 269.1[ M + H ] ]+

And 5: preparation of 5- (2- (methoxymethyl) thiazol-5-yl) isoxazole-3-carboxylic acid

To a solution of ethyl 5- (2- (methoxymethyl) thiazol-5-yl) isoxazole-3-carboxylate (0.15g, 0.56mmol) in tetrahydrofuran/water (20mL/5mL) at 0 ℃ was added lithium hydroxide monohydrate (35mg, 0.84mmol) in one portion. The reaction mixture was stirred for 1h, then the volatiles were removed. The crude residue was diluted with water (10mL) and the aqueous layer was adjusted to pH 3-4 with 1N aqueous hydrogen chloride solution. The white precipitate was collected and dried to give 5- (2- (methoxymethyl) thiazol-5-yl) isoxazole-3-carboxylic acid (0.14g, 0.58mmol, 93%) as a white solid. LCMS (ESI) M/z 241.1[ M + H ]]+

Step 6: preparation of (N- (1-benzyl-1H-pyrazol-4-yl) -5- (2- (methoxymethyl) thiazol-5-yl) isoxazole-3-carboxamide

To a mixture of 5- (2- (methoxymethyl) thiazol-5-yl) isoxazole-3-carboxylic acid (0.12g, 0.5mmol), 1-benzyl-1H-pyrazol-4-amine (0.125g, 0.6mmol), 2- (7-azabenzotriazol-1-yl) -N, N' -tetramethyluronium hexafluorophosphate (0.285, 0.75mmol) in N, N-dimethylformamide (15mL) was added N, N-diisopropylethylamine (0.19g, 1.5mmol) dropwise at 25 ℃. The reaction mixture was stirred at 25 ℃ for 18 h. The reaction mixture was diluted with ethyl acetate/water (20mL/20mL) and extracted with ethyl acetate (20 mL. times.2). The combined organic phases were washed with brine (20mL), dried over sodium sulfate, filtered and concentrated in vacuo. The residue was slurried in a mixture of ethyl acetate/petroleum ether (5mL/50mL), filtered and concentrated to give N- (1-benzyl-1H-pyrazol-4-yl) -5- (2- (methoxymethyl) thiazol-5-yl) isoxazole-3-carboxamide (130mg, 0.33mmol, 65.8%) as a brown solid. 1H NMR (500MHz, dimethylsulfoxide-d)6)δ11.06(s,1H),8.49(s,1H),8.17(s,1H),7.68(s,1H),7.40(s,1H),7.22-7.37(m,5H),5.34(s,2H),4.81(s,2H),3.46(s,3H);LCMS(ESI)m/z:396.1[M+H]+

Example 17 preparation of N- (1-benzyl-1H-pyrazol-4-yl) -5- (3-fluoropyridin-4-yl) isoxazole-3-carboxamide (147)

Step 1: preparation of 3-fluoro-4- ((trimethylsilyl) ethynyl) pyridine

A mixture of 4-bromo-3-fluoropyridine hydrochloride (3g, 14.2mmol), bis (triphenylphosphine) palladium (II) chloride (0.5g, 0.7mmol), cuprous iodide (0.27g, 1.41mmol) in triethylamine (20mL) was degassed and refilled with nitrogen three times. Ethynyltrimethylsilane (6mL, 42.5mmol) was then added dropwise via syringe. After addition, the reaction was stirred at 60 ℃ for 18 h. Mixing the reactionThe material was diluted with water (100mL) and extracted with ethyl acetate (100 mL. times.2). The combined organic phases were washed with brine (80mL), dried over sodium sulfate, filtered and concentrated in vacuo. The residue was purified by column chromatography (Biotage, 40g silica, eluting with 10% to 15% ethyl acetate in petroleum ether) to give 3-fluoro-4- ((trimethylsilyl) ethynyl) pyridine as a yellow oil (2g, 10.4mmol, 73%). LCMS (ESI) M/z 194.1[ M + H ]]+

Step 2: preparation of 4-ethynyl-3-fluoropyridine

To a solution of 3-fluoro-4- ((trimethylsilyl) ethynyl) pyridine (1.95g, 10.1mmol) in tetrahydrofuran (80mL) was added potassium hydroxide (1.13g, 20.2mmol) in methanol (16mL) dropwise at 0 ℃. After addition, the reaction was stirred at 25 ℃ for 1 h. Volatiles were removed in vacuo. The crude material was diluted with ethyl acetate/water (20mL/20mL) and extracted with ethyl acetate (20 mL. times.2). The combined organic phases were washed with brine (30mL), dried over sodium sulfate, filtered and concentrated to give 4-ethynyl-3-fluoropyridine (0.6g, 4.96mmol, 49%) as a pale yellow oil. This material was used directly in the next step without further purification.

And step 3: preparation of 5- (3-fluoropyridin-4-yl) isoxazole-3-carboxylic acid ethyl ester

To a solution of 4-ethynyl-3-fluoropyridine (0.6g, 4.96mmol) in N, N-dimethylformamide (6mL) was added dropwise a solution of ethyl 2-chloro-2- (hydroxyimino) acetate (0.5g, 3.30mmol) in N, N-dimethylformamide (3mL) under nitrogen at 25 ℃. After addition, the reaction was stirred at 25 ℃ for 20min, then heated to 90 ℃ and a solution of triethylamine (1.0g, 9.93mmol) in dimethylformamide (6mL) was added dropwise. The reaction mixture was heated at 90 ℃ for 18 h. Mixing the reaction mixtureCooled, diluted with water (20mL) and extracted with ethyl acetate (30 mL. times.2). The combined organic phases were washed with brine (30mL), dried over sodium sulfate, filtered and concentrated in vacuo. The residue was purified by column chromatography (Biotage, 40g silica, eluting with 20% to 30% ethyl acetate in petroleum ether) to give ethyl 5- (3-fluoropyridin-4-yl) isoxazole-3-carboxylate (0.2g, 0.85mmol, 25%) as a white solid. LCMS (ESI) M/z 237.1[ M + H ]]+

And 4, step 4: preparation of 5- (3-fluoropyridin-4-yl) isoxazole-3-carboxylic acid

To a solution of ethyl 5- (3-fluoropyridin-4-yl) isoxazole-3-carboxylate (0.18g, 0.76mmol) in tetrahydrofuran/water (20mL/5mL) was added lithium hydroxide monohydrate (48mg, 1.14mmol) at 0 ℃ in one portion. The reaction mixture was stirred at 0 ℃ for 2 h. Volatiles were removed in vacuo. The crude residue was diluted with water (20mL) and adjusted to pH 3-4 with 1N aqueous hydrogen chloride. The white precipitate was collected and dried to give 5- (3-fluoropyridin-4-yl) isoxazole-3-carboxylic acid (0.15g, 0.72mmol, 94%) as a white solid. LCMS (ESI) M/z 209.1[ M + H ] ]+. This material was used directly in the next step without further purification.

And 5: preparation of N- (1-benzyl-1H-pyrazol-4-yl) -5- (3-fluoropyridin-4-yl) isoxazole-3-carboxamide

To a solution of 5- (3-fluoropyridin-4-yl) isoxazole-3-carboxylic acid (80mg, 0.38mmol), 1-benzyl-1H-pyrazol-4-amine (80mg, 0.46mmol) and 2- (7-azabenzotriazol-1-yl) -N, N' -tetramethyluronium hexafluorophosphate (0.22g, 0.57mmol) in N, N-dimethylformamide (15mL) was slowly added N, N-diisopropylethylamine (0.15g, 1.15mmol) at 25 ℃. The resulting reaction mixture was stirred at 25 ℃ for 18 h. The reaction mixture was diluted with ethyl acetate/water (20mL/20mL) andextraction was performed with ethyl acetate (20 mL. times.2). The combined organic phases were washed with brine (20mL), dried over sodium sulfate, filtered and concentrated in vacuo. The residue was triturated in a mixture of ethyl acetate/petroleum ether (5mL/50mL), filtered and concentrated to give N- (1-benzyl-1H-pyrazol-4-yl) -5- (3-fluoropyridin-4-yl) isoxazole-3-carboxamide as a brown solid (125mg, 0.34mmol, 75%).1H NMR (500MHz, dimethylsulfoxide-d)6)δ11.12(s,1H),8.88(s,1H),8.64-8.68(m,1H),8.19(s,1H),8.02-8.07(m,1H),7.68(s,1H),7.52-7.54(m,1H),7.23-7.38(m,5H),5.34(s,2H);LCMS(ESI)m/z:364.1[M+H]+

Example 18 preparation of N- (1-benzyl-1H-pyrazol-4-yl) -5- (3-chloropyridin-4-yl) isoxazole-3-carboxamide (134)

Step 1: preparation of 3-chloro-4- ((trimethylsilyl) ethynyl) pyridine

A mixture of 4-bromo-3-chloropyridine (2g, 10.5mmol), bis (triphenylphosphine) palladium (II) chloride (0.37g, 0.52mmol), cuprous iodide (0.2g, 1.05mmol) in triethylamine (20mL) was degassed and refilled with nitrogen (2X). Ethynyltrimethylsilane (4.4mL, 31.4mmol) was then added dropwise via syringe. After addition, the reaction mixture was stirred at 60 ℃ for 18 h. The reaction mixture was diluted with water (100mL) and extracted with ethyl acetate (100 mL. times.2). The combined organic phases were washed with water (50mL) and brine (50mL), dried over sodium sulfate, filtered and concentrated in vacuo. The crude material was purified by column chromatography (Biotage, 40g silica, eluting with 10% to 15% ethyl acetate in petroleum ether) to give 3-chloro-4- ((trimethylsilyl) ethynyl) pyridine as a yellow oil (1.47g, 7.03mmol, 67%). LCMS (ESI) M/z 210.1[ M + H ]]+

Step 2: preparation of 3-chloro-4-ethynylpyridine

To a solution of 3-chloro-4- ((trimethylsilyl) ethynyl) pyridine (1.27g, 6.08mmol) in tetrahydrofuran (60mL) was added potassium hydroxide (0.68g, 12.2mmol) in methanol (12mL) dropwise at 0 ℃. After addition, the reaction was stirred at 25 ℃ for 1 h. Volatiles were removed in vacuo. The residue was diluted with ethyl acetate (100mL), washed with water (50mL × 2) and brine (50mL), dried over sodium sulfate, filtered and concentrated to give 3-chloro-4-ethynylpyridine (0.77g, 5.62mmol, 92%) as a dark brown solid. This material was used directly in the next step without further purification.

And step 3: preparation of ethyl 5- (3-chloropyridin-4-yl) isoxazole-3-carboxylate

To a solution of 3-chloro-4-ethynylpyridine (0.67g, 4.89mmol) in N, N-dimethylformamide (6mL) was added ethyl 2-chloro-2- (hydroxyimino) acetate (0.49g, 3.26mmol) in N, N-dimethylformamide (3mL) dropwise under nitrogen at 25 ℃. After addition, the reaction was stirred at 25 ℃ for 20min and heated to 90 ℃. A solution of triethylamine (0.98g, 9.73mmol) in N, N-dimethylformamide (6mL) was then added dropwise. The reaction mixture was heated at 90 ℃ for 18 h. The reaction mixture was cooled to room temperature, diluted with ethyl acetate/water (30mL/30mL) and extracted with ethyl acetate (50 mL. times.2). The combined organic phases were washed with water (40mL) and brine (50mL), dried over sodium sulfate, filtered and concentrated in vacuo. The residue was purified by column chromatography (Biotage, 40g silica, eluting with 30% to 40% ethyl acetate in petroleum ether) to give ethyl 5- (3-chloropyridin-4-yl) isoxazole-3-carboxylate (0.21g, 0.83mmol, 17%) as an off-white solid. LCMS (ESI) M/z 253.1[ M + H ]]+

And 4, step 4: preparation of 5- (3-chloropyridin-4-yl) isoxazole-3-carboxylic acid

To a solution of ethyl 5- (3-chloropyridin-4-yl) isoxazole-3-carboxylate (0.19g, 0.75mmol) in tetrahydrofuran/water (20mL/5mL) was added lithium hydroxide monohydrate (47.5mg, 1.13mmol) at 0 ℃ in one portion. The reaction mixture was stirred at 0 ℃ for 2 h. Volatiles were removed in vacuo. The residue was diluted with water (10mL) and the aqueous layer was adjusted to pH 3-4 with 1N aqueous hydrogen chloride solution. The white precipitate was collected and dried to give 5- (3-chloropyridin-4-yl) isoxazole-3-carboxylic acid (0.16g, 0.71mmol, 86%) as a white solid. LCMS (ESI) M/z 224.9[ M + H ] ]+. This material was used directly in the next step without further purification.

And 5: preparation of 5- (3-chloropyridin-4-yl) isoxazole-3-carbonyl chloride

To a suspension of 5- (3-chloropyridin-4-yl) isoxazole-3-carboxylic acid (50mg, 0.22mmol) in dichloromethane (20mL) at 25 ℃ was added oxalyl chloride (2mL) dropwise followed by two drops of N, N-dimethylformamide. The reaction mixture was then stirred at 25 ℃ for 30 min. The reaction was concentrated and dried to give crude 5- (3-chloropyridin-4-yl) isoxazol-3-carbonyl chloride (55mg, 0.22mmol) as a yellow solid. This material was used directly in the next step without further purification.

Step 6: preparation of N- (1-benzyl-1H-pyrazol-4-yl) -5- (3-chloropyridin-4-yl) isoxazole-3-carboxamide

To a suspension of 1-benzyl-1H-pyrazol-4-amine hydrochloride (52mg, 0.249mmol) in dichloromethane (20mL) at 0 deg.C was added triethylamine (68.6 m)g, 0.679mmol) followed by slow addition of a suspension of 5- (3-chloropyridin-4-yl) isoxazole-3-carbonyl chloride (55mg, 0.226mmol) in dichloromethane (10 mL). After addition, the reaction was stirred at 0 ℃ for 1 h. The reaction mixture was diluted with water (30mL) and extracted with dichloromethane (30 mL. times.2). The combined organic phases were washed with brine (30mL), dried over sodium sulfate, filtered and concentrated in vacuo. The residue was slurried with a mixture of ethyl acetate/petroleum ether (1mL/20mL, containing two drops of methanol) using a sonicator and filtered. The precipitate was filtered and concentrated to give N- (1-benzyl-1H-pyrazol-4-yl) -5- (3-chloro-pyridin-4-yl) isoxazole-3-carboxamide as a grey solid (60.4mg, 0.159mmol, 71% over 2 steps). 1H NMR (500MHz, dimethylsulfoxide-d)6)δ11.14(s,1H),8.91(s,1H),8.74(d,J=5.5Hz,1H),8.18(s,1H),8.02(d,J=5Hz,1H),7.69(d,J=2.5Hz,2H),7.23-7.38(m,5H),5.35(s,2H);LCMS(ESI)m/z:380.0[M+H]+

Example 19 preparation of N- (1-benzyl-1H-pyrazol-4-yl) -5- (1,3, 4-thiadiazol-2-yl) isoxazole-3-carboxamide (170)

Step 1: preparation of 5- (2-formylhydrazine-1-carbonyl) isoxazole-3-carboxylic acid ethyl ester

To a solution of 3- (ethoxycarbonyl) isoxazole-5-carboxylic acid (1.85g, 10.0mmol), formylhydrazine (1.2g, 20.0mmol) and 4-methylmorpholine (3.03g, 30mmol) in tetrahydrofuran (60mL) at 25 ℃ under nitrogen was added 4- (4, 6-dimethoxy-1, 3, 5-triazin-2-yl) -4-methylmorpholinium chloride (2.95g, 10.0 mmol). The mixture was stirred at 25 ℃ for 2 h. Volatiles were removed in vacuo. The crude residue was purified by column chromatography (silica, dichloromethane/methanol 30/1 to 20/1) to give ethyl 5- (2-formylhydrazine-1-carbonyl) isoxazole-3-carboxylate as a yellow oil(1.29g,5.68mmol,56%)。LCMS(ESI)m/z:227.9[M+H]+

Step 2: preparation of ethyl 5- (1,3, 4-thiadiazol-2-yl) isoxazole-3-carboxylate

To a solution of ethyl 5- (2-formylhydrazine-1-carbonyl) isoxazole-3-carboxylate (0.682g, 3.0mmol) in tetrahydrofuran (50mL) was added phosphorus pentasulfide (0.861g, 4.5mmol) at 25 ℃ under nitrogen. The mixture was stirred at 65 ℃ for 2 h. Volatiles were removed in vacuo. The crude residue was purified by column chromatography (silica, petroleum ether/ethyl acetate 4/1 to 3/1) to give ethyl 5- (1,3, 4-thiadiazol-2-yl) isoxazole-3-carboxylate (0.263g, 1.17mmol, 38%) as a pale yellow solid. LCMS (ESI) M/z 226.0[ M + H ] ]+

And step 3: preparation of 5- (1, 3, 4-thiadiazol-2-yl) isoxazole-3-carboxylic acid

To a solution of ethyl 5- (1, 3, 4-thiadiazol-2-yl) isoxazole-3-carboxylate (0.263g, 1.17mmol) in tetrahydrofuran (9mL) and water (3mL) was added lithium hydroxide hydrate (59mg, 1.4mmol) under nitrogen at 0 ℃. The mixture was stirred at 0 ℃ for 30 min. The aqueous phase was adjusted to pH 3 with 2N aqueous hydrochloric acid. Volatiles were removed in vacuo. The resulting precipitate was filtered and concentrated to give 5- (1, 3, 4-thiadiazol-2-yl) isoxazole-3-carboxylic acid (0.153g, 0.78mmol, 67%) as a yellow solid. LCMS (ESI) M/z 198.0[ M + H ]]+. This material was used directly in the next step without further purification.

And 4, step 4: preparation of N- (1-benzyl-1H-pyrazol-4-yl) -5- (1, 3, 4-thiadiazol-2-yl) isoxazole-3-carboxamide

To a solution of 5- (1, 3, 4-thiadiazol-2-yl) isoxazole-3-carboxylic acid (75mg, 0.38mmol), 1-benzyl-1H-pyrazol-4-amine hydrochloride (80mg, 0.38mmol) and triethylamine (0.384g, 3.8mmol) in dichloromethane (30mL) at 0 ℃ under nitrogen was slowly added propylphosphonic anhydride (0.5M in ethyl acetate, 1.2g, 1.9 mmol). The mixture was stirred at room temperature for 20 h. The reaction mixture was quenched with water (30mL) and the aqueous layer was extracted with dichloromethane (50 mL. times.3). The organic layer was dried over sodium sulfate, filtered and concentrated in vacuo. The crude residue was purified by preparative-TLC (dichloromethane/ammonia in methanol (7N) ═ 25/1) to give N- (1-benzyl-1H-pyrazol-4-yl) -5- (1, 3, 4-thiadiazol-2-yl) isoxazole-3-carboxamide as a white solid (0.102g, 0.29mmol, 76%). 1H NMR (400MHz, dimethylsulfoxide-d)6)δ11.17(s,1H),9.90(s,1H),8.19(s,1H),7.77(s,1H),7.69(s,1H),7.27-7.38(m,3H),7.24-7.26(m,2H),5.35(s,2H);LCMS(ESI)m/z:353.1[M+H]+

Example 20 preparation of N- (1-benzyl-1H-pyrazol-4-yl) -5- (piperidin-1-yl) isoxazole-3-carboxamide (159)

Step 1: preparation of 3- (ethoxycarbonyl) isoxazole-5-carboxylic acid

To a solution of propiolic acid (8.0g, 114.2mmol) in N, N-dimethylformamide (60mL) was added dropwise ethyl (Z) -2-chloro-2- (hydroxyimino) acetate (6.9g, 45.7mmol) in N, N-dimethylformamide (20mL) under a nitrogen atmosphere over 40 min. After addition, the reaction mixture was heated to 90 ℃ and a solution of triethylamine (13.8g, 137mmol) in N, N-dimethylformamide (20mL) was added dropwise over 1 h. The reaction mixture was heated at 90 ℃ for 17h and then cooled to room temperature. The reaction mixture is evaporated to dryness and then dried overWater (30mL) was diluted and adjusted to pH 2 with 1N aqueous hydrogen chloride. The aqueous layer was extracted with ethyl acetate (30 mL. times.2). The combined organic layers were washed with brine (30mL × 2), dried over anhydrous sodium sulfate, filtered and concentrated in vacuo to afford 3- (ethoxycarbonyl) isoxazole-5-carboxylic acid as a yellow oil (3.2g, 17.3mmol, 38%). LCMS (ESI) M/z 186.1[ M + H ]]+

Step 2: preparation of ethyl 5- (tert-butoxycarbonylamino) isoxazole-3-carboxylate

To a solution of 3- (ethoxycarbonyl) isoxazole-5-carboxylic acid (2.7g, 14.6mmol) in toluene (40mL) was added triethylamine (1.8g, 17.5mmol), tert-butanol (2.7g, 36.5mmol) and diphenylphosphoryl azide (4.8g, 17.5 mmol). The reaction mixture was heated at 100 ℃ for 2h and concentrated to dryness. The residue was purified by column chromatography (silica, petroleum ether/ethyl acetate-8/1) to give ethyl 5- (tert-butoxycarbonylamino) isoxazole-3-carboxylate (1.8g, 7.0mmol, 49%) as a white solid. LCMS (ESI) M/z 257.2[ M + H ] ]+

And step 3: preparation of 5-aminoisoxazole-3-carboxylic acid ethyl ester

To a solution of ethyl 5- (tert-butoxycarbonylamino) isoxazole-3-carboxylate (1.8g, 7.0mmol) in dichloromethane (16.0mL) was added trifluoroacetic acid (8.0 mL). The reaction mixture was stirred at 25 ℃ for 3h, then concentrated in vacuo. The crude residue was purified by column chromatography (silica, petroleum ether/ethyl acetate 1/1) to give ethyl 5-aminoisoxazole-3-carboxylate (0.620g, 3.97mmol, 57%) as a yellow solid. LCMS (ESI) M/z 157.1[ M + H ]]+

And 4, step 4: preparation of 5- (piperidin-1-yl) isoxazole-3-carboxylic acid ethyl ester

To a solution of ethyl 5-aminoisoxazole-3-carboxylate (0.310g, 1.9mmol) in dimethylacetamide (20mL) was added cesium carbonate (1.8g, 5.7mmol) and 1, 5-dibromopentane (1.1g, 4.75 mmol). The reaction mixture was heated at 80 ℃ for 3h and then cooled to room temperature. Volatiles were removed in vacuo. The crude material was purified by column chromatography (silica, dichloromethane/methanol ═ 15/1) to give ethyl 5- (piperidin-1-yl) isoxazole-3-carboxylate (0.170g, 0.76mmol, 38%) as a white solid. LCMS (ESI) M/z 225.2[ M + H ]]+

And 5: preparation of 5- (piperidin-1-yl) isoxazole-3-carboxylic acids

To a solution of ethyl 5- (piperidin-1-yl) isoxazole-3-carboxylate (170mg, 0.76mmol) in tetrahydrofuran/water (v/v ═ 2/1, 9mL) was added lithium hydroxide/water (95.8mg, 2.28mmol) at-20 ℃. The reaction mixture was stirred at-20 ℃ for 10 min. The volatiles were removed in vacuo to give 5- (piperidin-1-yl) isoxazole-3-carboxylic acid (120mg, 0.61mmol, 81%) as a yellow oil. LCMS (ESI) M/z 197.1[ M + H ]]+. This material was used directly in the next step without further purification.

Step 6: preparation of N- (1-benzyl-1H-pyrazol-4-yl) -5- (piperidin-1-yl) isoxazole-3-carboxamide

To a solution of 5- (piperidin-1-yl) isoxazole-3-carboxylic acid (80mg, 0.4mmol) in N, N-dimethylformamide (15mL) was added 1-benzyl-1H-pyrazol-4-amine (69mg, 0.4mmol), 1- [ bis (dimethylamino) methylene ] methylene]-1H-1, 2, 3-triazolo [4, 5-b]Pyridinium 3-oxide hexafluorophosphate (0.456g, 1.2mmol) and diisopropylethylamine (0.155g, 1.2 mmol). Subjecting the mixture to 25 deg.CStirred for 17 h. The crude sample was dissolved in a minimal amount of N, N-dimethylformamide and purified by preparative-HPLC (Boston C1821 × 250mm 10 μm column; acetonitrile/0.01% aqueous trifluoroacetic acid) to give N- (1-benzyl-1H-pyrazol-4-yl) -5- (piperidin-1-yl) isoxazole-3-carboxamide (30.5mg, 0.08mmol, 23%) as a white solid. 1H NMR (500MHz, dimethylsulfoxide-d)6)δ10.64(s,1H),8.10(s,1H),7.63(s,1H),7.40-7.11(m,5H),5.62(s,1H),5.31(s,2H),3.33(s,4H),1.58(s,6H);LCMS(ESI)m/z:352.1[M+H]+

Example 21 preparation of N- (1-benzyl-1H-pyrazol-4-yl) -4,5,6, 7-tetrahydrobenzo [ d ] isoxazole-3-carboxamide (156)

To 1-benzyl-1H-pyrazol-4-amine (0.050g, 0.289mmol), 4,5,6, 7-tetrahydrobenzo [ d ] at 25 deg.C]Isoxazole-3-carboxylic acid (48.2mg, 0.289mmol) and 1- [ bis (dimethylamino) methylene]-1H-1, 2, 3-triazolo [4, 5-b]Diisopropylethylamine (0.075mL, 0.433mmol) was added to a solution of pyridinium 3-oxide hexafluorophosphate (109mg, 0.289mmol) in N, N' -dimethylformamide (1 mL). The reaction mixture was stirred at 25 ℃ for 16 h. The reaction mixture was quenched with water (1 mL). The aqueous layer was extracted with ethyl acetate (5 mL. times.3). The combined organic layers were washed with brine, dried over magnesium sulfate, filtered and concentrated in vacuo. The crude residue was purified by column chromatography (ISCO, 12g silica, eluting with 60% ethyl acetate/hexanes for 20min) to afford N- (1-benzyl-1H-pyrazol-4-yl) -4,5,6, 7-tetrahydrobenzo [ d ] as a pale yellow solid]Isoxazole-3-carboxamide (35.2mg, 0.109mmol, 38%).1H NMR (300MHz, dimethylsulfoxide-d)6)δ11.35(s,1H),8.55(s,1H),8.06(s,1H),7.82-7.59(m,5H),5.72(s,2H),3.16(q,J=5.9Hz,4H),2.13(dt,J=12.9,6.9Hz,4H);LCMS(ESI)m/z:323.3[M+H]+

Example 22 preparation of N- (1-benzyl-1H-pyrazol-4-yl) -5- (2-methyloxazol-5-yl) isoxazole-3-carboxamide (164)

Step 1: preparation of ethyl 5- (hydroxymethyl) isoxazole-3-carboxylate

To a solution of prop-2-yn-1-ol (8.4g, 149mmol) in N, N-dimethylformamide (25mL) was added dropwise a solution of ethyl (Z) -2-chloro-2- (hydroxyimino) acetate (7.5g, 49.7mmol) in N, N-dimethylformamide (50mL) under nitrogen over 40 min. After addition, the reaction mixture was heated to 90 ℃ and a solution of triethylamine (15.0g, 149mmol) in N, N-dimethylformamide (25mL) was added dropwise over 1 h. The reaction mixture was heated at 90 ℃ for 17h and then cooled to room temperature. The reaction mixture was diluted with ethyl acetate (50mL), washed with water (60mL × 2) and brine (60mL × 2), dried over anhydrous sodium sulfate, filtered and concentrated in vacuo. The residue was purified by column chromatography (silica, petroleum ether/ethyl acetate-6/1) to give ethyl 5- (hydroxymethyl) isoxazole-3-carboxylate (4.5g, 26.3mmol, 53%) as a yellow oil. LCMS (ESI) M/z 172.1[ M + H ]]+

Step 2: preparation of ethyl 5-formylisoxazole-3-carboxylate

To a solution of ethyl 5- (hydroxymethyl) isoxazole-3-carboxylate (4.5g, 26.3mmol) in ethyl acetate (80.0mL) was added 2-iodoxybenzoic acid (22.1g, 78.9 mmol). After addition, the reaction mixture was heated to 90 ℃ and stirred for 17h, then cooled to room temperature. The reaction mixture was filtered and concentrated in vacuo. The residue was purified by column chromatography (silica, petroleum ether/ethyl acetate-6/1) to give ethyl 5-formylisoxazole-3-carboxylate (3.5g, 20.7mmol, 80%) as a yellow oil.

And step 3: preparation of 5- (2-methyloxazol-5-yl) isoxazole-3-carboxylic acid

To a solution of 1- (1-isocyanoethylsulfonyl) -4-toluene (1.85g, 8.87mmol) in acetonitrile (30mL) was added potassium carbonate (2.4g, 17.7 mmol). The reaction mixture was stirred at 25 ℃ for 1 h. The reaction mixture was cooled to 0 ℃ and ethyl 5-formylisoxazole-3-carboxylate (1.5g, 8.87mmol) was added. The reaction mixture was then heated to 90 ℃ for 17 h. The reaction was cooled to room temperature, diluted with water (20mL) and adjusted to pH 3 with 1N aqueous hydrogen chloride. The aqueous layer was extracted with ethyl acetate (10 mL. times.3). The combined organic layers were washed with brine (10mL × 2), dried over anhydrous sodium sulfate, filtered and concentrated in vacuo to afford 5- (2-methyloxazol-5-yl) isoxazole-3-carboxylic acid (0.220g, 1.13mmol, 12%) as a white solid. LCMS (ESI) M/z 195.1[ M + H ]]+. This material was used directly in the next step without further purification.

And 4, step 4: preparation of N- (1-benzyl-1H-pyrazol-4-yl) -5- (2-methyloxazol-5-yl) isoxazole-3-carboxamide

To a solution of 5- (2-methyloxazol-5-yl) isoxazole-3-carboxylic acid (60mg, 0.3mmol) in N, N-dimethylformamide (15mL) was added 1-benzyl-1H-pyrazol-4-amine (52mg, 0.3mmol), 1- [ bis (dimethylamino) methylene ] amine ]-1H-1, 2, 3-triazolo [4, 5-b]Pyridinium 3-oxide hexafluorophosphate (0.352g, 0.9mmol) and diisopropylethylamine (0.116g, 0.9 mmol). The mixture was stirred at 25 ℃ for 17H and purified via prep-HPLC (Boston C1821 × 250mm 10 μm column; acetonitrile/0.01% aqueous trifluoroacetic acid) to give N- (1-benzyl-1H-pyrazol-4-yl) -5- (2-methyloxazol-5-yl) isoxazole-3-carboxamide (25.8mg, 0.07mmol, 25%) as a white solid.1H NMR (400MHz, dimethyl sulfoxide-d6)δ11.04(s,1H),8.60(s,1H),8.16(s,1H),7.67(s,1H),7.40-7.19(m,6H),5.34(s,2H),2.44(s,3H);LCMS(ESI)m/z:350.1[M+H]+

Example 23 preparation of N- (1- (3-fluorobenzyl) -1H-pyrazol-4-yl) -5- (prop-1-en-2-yl) isoxazole-3-carboxamide (58)

Step 1: preparation of 5- (prop-1-en-2-yl) isoxazole-3-carboxylic acid ethyl ester

To a solution of ethyl 5-bromoisoxazole-3-carboxylate (0.960g, 4.36mmol) in N, N-dimethylformamide (20mL) was added under nitrogen 4, 4, 5, 5-tetramethyl-2- (prop-1-en-2-yl) -1, 3, 2-dioxaborolan (0.879g, 5.23mmol), tetrakis (triphenylphosphine) palladium (0) (0.504g, 0.43mmol) and potassium phosphate (1.8g, 8.72 mmol). The mixture was heated at 90 ℃ for 5 h. The mixture was diluted with water (30mL) and extracted with ethyl acetate (20 mL. times.2). The combined organic layers were washed with water (20mL × 2) and brine (40mL × 2), then dried over anhydrous sodium sulfate, filtered and concentrated in vacuo. The residue was purified by column chromatography (silica, petroleum ether/ethyl acetate-20/1) to give ethyl 5- (prop-1-en-2-yl) isoxazole-3-carboxylate (0.660g, 3.64mmol, 84%) as a yellow oil. LCMS (ESI) M/z 182.2[ M + H ] ]+

Step 2: preparation of 5- (prop-1-en-2-yl) isoxazole-3-carboxylic acids

To a solution of ethyl 5- (prop-1-en-2-yl) isoxazole-3-carboxylate (0.660g, 3.64mmol) in tetrahydrofuran/water (v/v ═ 4/1, 20mL) was added sodium hydroxide (0.349g, 8.73mmol) at 15 ℃. Mixing the reactionThe mixture was stirred at room temperature for 17 h. The reaction mixture was evaporated to dryness, diluted with water (30mL) and adjusted to pH 2 with 1N aqueous hydrogen chloride. The reaction mixture was then extracted with ethyl acetate (20 mL. times.2). The combined organic layers were washed with brine (20mL × 2), dried over anhydrous sodium sulfate, filtered and concentrated in vacuo to afford 5- (prop-1-en-2-yl) isoxazole-3-carboxylic acid (0.350g, 2.28mmol, 63%) as a yellow solid. LCMS (ESI) M/z 154.1[ M + H ]]+. This material was used directly in the next step without further purification.

And step 3: preparation of N- (1- (3-fluorobenzyl) -1H-pyrazol-4-yl) -5- (prop-1-en-2-yl) isoxazole-3-carboxamide

To a solution of 5- (prop-1-en-2-yl) isoxazole-3-carboxylic acid (0.350g, 2.28mmol) in N, N-dimethylformamide (20mL) were added 1- (3-fluorobenzyl) -1H-pyrazol-4-amine (0.435g, 2.28mmol), triethylamine (0.691g, 6.84mmol), 1-hydroxybenzotriazole hydrate (0.462g, 3.42mmol) and N- (3-dimethylaminopropyl) -N' -ethylcarbodiimide hydrochloride (1.22g, 6.84 mmol). The mixture was stirred at room temperature for 17H, then purified directly by preparative-HPLC (Boston C1821X 250mm 10 μm column; acetonitrile/0.01% aqueous trifluoroacetic acid) to give N- (1- (3-fluorobenzyl) -1H-pyrazol-4-yl) -5- (prop-1-en-2-yl) isoxazole-3-carboxamide (0.433g, 1.32mmol, 58%) as a white solid. 1H NMR (400MHz, dimethylsulfoxide-d)6)δ10.97(s,1H),8.20(s,1H),7.68(s,1H),7.40(dt,J=14.1,7.1Hz,1H),7.21-7.10(m,1H),7.09-6.96(m,3H),5.86(s,1H),5.48(s,1H),5.36(s,2H),2.11(s,3H);LCMS(ESI)m/z:327.1[M+H]+

Example 24.preparation of 4-phenylpiperidin-1-yl) (5- (pyridin-4-yl) isoxazol-3-yl) methanone trifluoroacetate (313)

Step 1: preparation of 5- (pyridin-4-yl) isoxazole-3-carboxylic acid ethyl ester

Ethyl 5-bromoisoxazole-3-carboxylate (0.850g, 3.86mmol), pyridin-4-ylboronic acid (0.522g, 4.25mmol) and [1, 1' -bis (diphenylphosphino) ferrocene]A mixture of palladium (II) dichloride (0.285g, 0.39mmol) and potassium carbonate (1.07g, 7.72mmol) in acetonitrile (40mL) and water (10mL) was heated at 80 ℃ for 3h under nitrogen. The mixture was concentrated under reduced pressure and the residue was purified by column chromatography (silica, petroleum ether/ethyl acetate 4/1) to give ethyl 5- (pyridin-4-yl) isoxazole-3-carboxylate (0.280g, 1.28mmol, 33%) as a yellow solid. LCMS (ESI) M/z 219.1[ M + H ]]+

Step 2: preparation of 5- (pyridin-4-yl) isoxazole-3-carboxylic acids

To a stirred solution of ethyl 5- (pyridin-4-yl) isoxazole-3-carboxylate (0.280g, 1.28mmol) in tetrahydrofuran (10mL) and water (10mL) was added lithium hydroxide monohydrate (0.107g, 2.56mmol) at 0 ℃. The mixture was stirred at this temperature for 2 h. The volatiles were removed under reduced pressure and the aqueous layer was adjusted to pH 5 with 1N aqueous hydrogen chloride. The resulting precipitate was collected by filtration and dried in vacuo to give 5- (pyridin-4-yl) isoxazole-3-carboxylic acid (0.120g, 0.63mmol, 49%) as a white solid. LCMS (ESI) M/z 191.2[ M + H ] ]+

And step 3: preparation of (4-phenylpiperidin-1-yl) (5- (pyridin-4-yl) isoxazol-3-yl) methanone trifluoroacetate

To 5- (pyridin-4-yl) isoxazole-3-carboxylic acid (40.0mg, 0.21mmol),4-phenylpiperidine (40.0mg, 0.25mmol) and 1- [ bis (dimethylamino) methylene]-1H-1, 2, 3-triazolo [4, 5-b]To a stirred solution of pyridinium 3-oxide hexafluorophosphate (0.122g, 0.32mmol) in N, N' -dimethylformamide (3mL) was added diisopropylethylamine (54.0mg, 0.42 mmol). After addition, the reaction mixture was stirred at room temperature for 2 h. The mixture was passed through preparative-HPLC (column: Sunfire preparative C1810 μm OBD 19X 250 mm; mobile phase: [ water (0.05% trifluoroacetic acid) -acetonitrile](ii) a B%: 60% -88%, 15min gradient) to give (4-phenylpiperidin-1-yl) (5- (pyridin-4-yl) isoxazol-3-yl) methanone trifluoroacetate as a white solid (0.036g, 0.08mmol, 38%).1H NMR (500MHz, dimethylsulfoxide-d)6)δ8.88(d,J=6.0Hz,2H),8.07-8.06(m,2H),7.72(s,1H),7.33-7.19(m,5H),4.67-4.64(m,1H),4.11-4.08(m,1H),3.33-3.27(m,1H),2.99-2.86(m,2H),1.94-1.91(m,1H),1.85-1.82(m,1H),1.69-1.58(m,2H);LCMS(ESI)m/z:334.1[M+H]+

Example 25 preparation of N- (1- (3-fluorobenzyl) -1H-pyrazol-4-yl) -5-isopropylisoxazole-3-carboxamide (60)

Step 1: preparation of 5- (prop-1-en-2-yl) isoxazole-3-carboxylic acid ethyl ester

To a mixture of ethyl 5-bromoisoxazole-3-carboxylate (0.960g, 4.36mmol) in N, N-dimethylformamide (20mL) was added under nitrogen 4, 4, 5, 5-tetramethyl-2- (prop-1-en-2-yl) -1, 3, 2-dioxaborolan (0.879g, 5.23mmol), tetrakis (triphenylphosphine) palladium (0) (0.504g, 0.43mmol) and potassium phosphate (1.8g, 8.72 mmol). The mixture was heated at 90 ℃ for 5 h. The mixture was cooled, diluted with water (30mL), and extracted with ethyl acetate (20 mL. times.2). The combined organic layers were washed with water (20 mL. times.2), brine (40 mL. times.2), and dried over anhydrous water Dried over sodium sulfate, filtered and concentrated in vacuo. The residue was purified by column chromatography (silica, petroleum ether/ethyl acetate-20/1) to give ethyl 5- (prop-1-en-2-yl) isoxazole-3-carboxylate (0.660g, 3.64mmol, 84%) as a yellow oil. LCMS (ESI) M/z 182.2[ M + H ]]+

Step 2: preparation of 5- (prop-1-en-2-yl) isoxazole-3-carboxylic acids

To a solution of ethyl 5- (prop-1-en-2-yl) isoxazole-3-carboxylate (0.660g, 3.64mmol) in tetrahydrofuran/water (v/v ═ 4:1, 20mL) was added sodium hydroxide (0.349g, 8.73mmol) at 15 ℃. The reaction mixture was stirred at room temperature for 17 h. The reaction mixture was evaporated to dryness, diluted with water (30mL) and adjusted to pH 2 with 1N aqueous hydrogen chloride. The reaction mixture was then extracted with ethyl acetate (20 mL. times.2). The combined organic layers were washed with brine (20mL × 2), dried over anhydrous sodium sulfate, filtered and concentrated in vacuo to afford 5- (prop-1-en-2-yl) isoxazole-3-carboxylic acid (0.350g, 2.28mmol, 63%) as a yellow solid. LCMS (ESI) M/z 154.1[ M + H ]]+. This material was used directly in the next step without further purification.

And step 3: preparation of N- (1- (3-fluorobenzyl) -1H-pyrazol-4-yl) -5- (prop-1-en-2-yl) isoxazole-3-carboxamide

To a solution of 5- (prop-1-en-2-yl) isoxazole-3-carboxylic acid (0.350g, 2.28mmol) in N, N-dimethylformamide (20mL) were added 1- (3-fluorobenzyl) -1H-pyrazol-4-amine (0.435g, 2.28mmol), triethylamine (0.691g, 6.84mmol), 1-hydroxybenzotriazole hydrate (0.462g, 3.42mmol) and N- (3-dimethylaminopropyl) -N' -ethylcarbodiimide hydrochloride (1.22g, 6.84 mmol). The mixture was stirred at room temperature for 17h and passed directly through preparative-HPLC (Boston C1821X 250mm 10 μm column; acetonitrile/0.01% trifluoroacetic acidAqueous solution)) to yield N- (1- (3-fluorobenzyl) -1H-pyrazol-4-yl) -5- (prop-1-en-2-yl) isoxazole-3-carboxamide as a white solid (0.433g, 1.32mmol, 58%).1H NMR (400MHz, dimethylsulfoxide-d)6)δ10.97(s,1H),8.20(s,1H),7.68(s,1H),7.40(dt,J=14.1,7.1Hz,1H),7.21-7.10(m,1H),7.09-6.96(m,3H),5.86(s,1H),5.48(s,1H),5.36(s,2H),2.11(s,3H);LCMS(ESI)m/z:327.1[M+H]+

And 4, step 4: preparation of N- (1- (3-fluorobenzyl) -1H-pyrazol-4-yl) -5-isopropylisoxazole-3-carboxamide

To a stirred solution of N- (1- (3-fluorobenzyl) -1H-pyrazol-4-yl) -5- (prop-1-en-2-yl) isoxazole-3-carboxamide (0.380g, 1.16mmol) in ethanol (20mL) was added palladium on activated carbon (0.200g, 10 wt% Pd) at room temperature. The mixture was stirred at room temperature under hydrogen for 17 h. Passing the mixture throughFiltered and then concentrated in vacuo. The crude product was dissolved in a minimal amount of N, N-dimethylformamide and purified by preparative-HPLC (Boston C1821 × 250mm 10 μm column; acetonitrile/0.01% aqueous trifluoroacetic acid) to give N- (1- (3-fluorobenzyl) -1H-pyrazol-4-yl) -5-isopropylisoxazole-3-carboxamide (0.105g, 0.32mmol, 27%) as a white solid. 1H NMR (400MHz, dimethylsulfoxide-d 6) δ 10.90(s, 1H), 8.18(s, 1H), 7.66(s, 1H), 7.40(dt, J ═ 14.0, 7.2Hz, 1H), 7.20-6.93(m, 3H), 6.64(d, J ═ 0.6Hz, 1H), 5.35(s, 2H), 3.17(hept, J ═ 6.9Hz, 1H), 1.29(d, J ═ 6.9Hz, 6H); LCMS (ESI) M/z 329.1[ M + H ]]+

Example 26 preparation of N- (1- (3-fluorobenzyl) -1H-pyrazol-4-yl) -5- (2-methyloxazol-5-yl) isoxazole-3-carboxamide (67)

Step 1: preparation of 5- (1-hydroxyethyl) isoxazole-3-carboxylic acid ethyl ester

To a solution of but-3-yn-2-ol (5.0g, 71.4mmol) in N, N-dimethylformamide (50mL) was added dropwise a solution of ethyl (Z) -2-chloro-2- (hydroxyimino) acetate (4.33g, 28.6mmol) in N, N-dimethylformamide (10mL) under nitrogen over 40 min. After addition, the reaction mixture was heated to 90 ℃ and a solution of triethylamine (8.7g, 85.7mmol) in N, N-dimethylformamide (10mL) was added dropwise over 1 h. The reaction mixture was heated at 90 ℃ for 17h and then cooled to room temperature. The reaction mixture was diluted with ethyl acetate (150mL), washed with water (100mL × 2) and brine (100mL × 2), dried over anhydrous sodium sulfate, filtered and concentrated in vacuo. The residue was purified by column chromatography (silica, petroleum ether/ethyl acetate 4/1) to give ethyl 5- (1-hydroxyethyl) isoxazole-3-carboxylate (1.86g, 10.1mmol, 35%) as a yellow oil. LCMS (ESI) M/z 186.1[ M + H ] ]+

Step 2: preparation of ethyl 5-acetylisoxazole-3-carboxylate

To a solution of ethyl 5- (1-hydroxyethyl) isoxazole-3-carboxylate (5.0g, 27.0mmol) in ethyl acetate (50mL) was added 2-iodoxybenzoic acid (22.7g, 81.1 mmol). After addition, the reaction mixture was heated to 80 ℃ and stirred for 17h, then cooled to room temperature. The reaction mixture was filtered and concentrated in vacuo. The residue was purified by column chromatography (silica, petroleum ether/ethyl acetate-6/1) to give ethyl 5-acetylisoxazole-3-carboxylate (4.4g, 24.0mmol, 89%) as a yellow oil. LCMS (ESI) M/z 184.1[ M + H ]]+

And step 3: preparation of ethyl 5- (2-methyloxazol-5-yl) isoxazole-3-carboxylate

To a solution of iodobenzene diacetate (2.35g, 5.46mmol) in acetonitrile (20mL) was added dropwise trifluoromethanesulfonic acid (1.64g, 10.9mmol) under nitrogen at room temperature. After 30min, a solution of ethyl 5-acetylisoxazole-3-carboxylate (1.0g, 5.46mmol) in acetonitrile (10mL) was added dropwise over 30 min. After addition, the reaction mixture was heated to 80 ℃ and stirred for 6h, then cooled to room temperature. The reaction mixture was concentrated in vacuo. The residue was diluted with saturated aqueous sodium bicarbonate (30mL) and extracted with ethyl acetate (50 mL. times.2). The combined organic layers were washed with brine (50mL × 2), dried over anhydrous sodium sulfate, filtered and concentrated in vacuo. The residue was purified by column chromatography (silica, petroleum ether/ethyl acetate-4/1) to give ethyl 5- (2-methyloxazol-5-yl) isoxazole-3-carboxylate (0.31g, 1.40mmol, 26%) as a yellow oil. LCMS (ESI) M/z 223.1[ M + H ]+

And 4, step 4: preparation of 5- (2-methyloxazol-5-yl) isoxazole-3-carboxylic acid

To a solution of ethyl 5- (2-methyloxazol-5-yl) isoxazole-3-carboxylate (0.32g, 1.44mmol) in tetrahydrofuran/water (v/v ═ 4/1, 10mL) was added lithium hydroxide (0.0908g, 2.16mmol) at 0 ℃. The reaction mixture was warmed to room temperature and stirred for 3 h. The reaction was concentrated to dryness, diluted with water (15mL) and adjusted to pH 2 with 1N aqueous hydrogen chloride. The aqueous layer was extracted with dichloromethane (20 mL. times.2). The combined organic layers were washed with saturated sodium bicarbonate solution (20mL × 2), brine (20mL × 2), dried over anhydrous sodium sulfate, filtered and concentrated in vacuo to afford 5- (2-methyloxazol-5-yl) isoxazole-3-carboxylic acid (0.20g, 1.03mmol, 72%) as a white solid. LCMS (ESI) M/z 195.1[ M + H ]]+. The material was used without further purificationIn the next step.

And 5: preparation of N- (1- (3-fluorobenzyl) -1H-pyrazol-4-yl) -5- (2-methyloxazol-5-yl) isoxazole-3-carboxamide

To a mixture of 5- (2-methyloxazol-5-yl) isoxazole-3-carboxylic acid (0.20g, 1.03mmol), 1- (3-fluorobenzyl) -1H-pyrazol-4-amine (0.20g, 1.24mmol) and 1- [ bis (dimethylamino) methylene ] -amine]-1H-1, 2, 3-triazolo [4, 5-b]To a solution of pyridinium 3-oxide hexafluorophosphate (0.47g, 1.24mmol) in N, N-dimethylformamide (4mL) was added triethylamine (0.31g, 3.09 mmol). The mixture was stirred at room temperature for 2H then purified directly by preparative-HPLC (Boston C1821 × 250mm 10 μm column; acetonitrile/0.01% aqueous trifluoroacetic acid)) to give N- (1- (3-fluorobenzyl) -1H-pyrazol-4-yl) -5- (2-methyloxazol-5-yl) isoxazole-3-carboxamide (0.0516g, 0.14mmol, 14%) as a white solid. 1H NMR (500MHz, dimethylsulfoxide-d)6)δ11.08(s,1H),8.21(s,1H),7.86(s,1H),7.69(s,1H),7.42-7.37(m,1H),7.24(s,1H),7.17-7.00(m,3H),5.34(s,2H),2.53(s,3H);LCMS(ESI)m/z:368.0[M+H]+

Example 27 preparation of N- (1- (3-fluorobenzyl) -1H-pyrazol-4-yl) -5-vinylisoxazole-3-carboxamide (106)

Step 1: preparation of ethyl 5-vinylisoxazole-3-carboxylate

To a mixture of ethyl 5-bromoisoxazole-3-carboxylate (0.400g, 1.83mmol) and potassium vinyltrifluoroborate (0.295g, 2.20mmol) in 1, 4-dioxane (8mL) and water (2mL) was added [1, 1' -bis (diphenylphosphino) ferrocene]Palladium (II) dichloride (0.067g, 0.092mmol) and cesium carbonate (1.19g, 3.66 mmol). The reaction was heated at 90 ℃ for 2h under nitrogen. The reaction mixture was cooled, then diluted with water (50mL) and passed throughFiltered and washed with ethyl acetate (20 mL). The filtrate was extracted with ethyl acetate (20 mL. times.3). The combined organic layers were washed with brine (30mL), dried over sodium sulfate, filtered and concentrated in vacuo to give ethyl 5-vinylisoxazole-3-carboxylate (0.380g, crude) as a brown oil which was used in the next step without further purification. LCMS (ESI) M/z 168.1[ M + H ]]+

Step 2: preparation of 5-vinyl isoxazole-3-carboxylic acid

A mixture of ethyl 5-vinylisoxazole-3-carboxylate (0.300g, 1.8mmol) in tetrahydrofuran (8mL) and water (2mL) was cooled to 0 deg.C and lithium hydroxide monohydrate (0.151g, 3.6mmol) was added. The mixture was stirred at 0 ℃ for 0.5 h. The reaction was diluted with water (50mL) and extracted with ethyl acetate (20 mL. times.2). The aqueous layer was separated and acidified with 1N aqueous hydrogen chloride solution to pH 2-3 and extracted with ethyl acetate (20mL × 3). The combined extracts were dried over sodium sulfate, filtered and concentrated in vacuo to give 5-vinylisoxazole-3-carboxylic acid (0.380g, crude) which was used directly in the next step. LCMS (ESI) M/z 140.1[ M + H ] ]+

And step 3: preparation of N- (1- (3-fluorobenzyl) -1H-pyrazol-4-yl) -5-vinyl isoxazole-3-carboxamide

To a solution of 5-vinylisoxazole-3-carboxylic acid (0.100g, 0.719mmol) in dichloromethane (2mL) at 0 ℃ was added N, N-dimethylformamide (1 drop) and oxalyl chloride (0.183g, 1.44 mmol). Will reactThe mixture was stirred at 0 ℃ for 0.5h, then concentrated in vacuo. The crude oil was dissolved in dichloromethane (2mL) and added to a mixture of 1- (3-fluorobenzyl) -1H-pyrazol-4-amine hydrochloride (0.163g, 0.719mmol) and pyridine (0.228g, 2.88mmol) in dichloromethane (2mL) at 0 ℃. The reaction mixture was stirred at 0 ℃ for 20min, then concentrated in vacuo. The crude product was dissolved in a minimal amount of N, N-dimethylformamide and purified by preparative-HPLC (Boston C1821 × 250mm 10 μm column; acetonitrile/0.01% aqueous trifluoroacetic acid) to give N- (1- (3-fluorobenzyl) -1H-pyrazol-4-yl) -5-vinylisoxazole-3-carboxamide (0.106g, 0.34mmol, 47%) as a white solid.1H NMR (400MHz, dimethylsulfoxide-d)6)δ10.98(s,1H),8.19(s,1H),7.67(s,1H),7.45-7.32(m,1H),7.11(tt,J=12.0,6.0Hz,1H),7.07-6.95(m,3H),6.84(dd,J=18.0,11.6Hz,1H),6.14(d,J=18.0Hz,1H),5.74(d,J=11.6Hz,1H),5.36(s,2H);LCMS(ESI)m/z:313.1[M+H]+

EXAMPLE 28 preparation of 5-Ethyl-N- (1- (3-fluorobenzyl) -1H-pyrazol-4-yl) isoxazole-3-carboxamide (104)

Step 1: preparation of 5-ethyl-N- (1- (3-fluorobenzyl) -1H-pyrazol-4-yl) isoxazole-3-carboxamide

A mixture of N- (1- (3-fluorobenzyl) -1H-pyrazol-4-yl) -5-vinylisoxazole-3-carboxamide (0.320g, 1.03mmol) and 10% palladium on activated carbon (0.030g, 10 wt% Pd) in methanol (5mL) was stirred at room temperature under hydrogen (balloon) for 2H. Passing the reaction mixture throughFiltered and washed with methanol (20 mL). The filtrate was concentrated in vacuo and dissolved in a minimum amountWas added to N, N-dimethylformamide and purified by preparative-HPLC (Boston C1821 × 250mm 10 μm column; acetonitrile/0.01% aqueous trifluoroacetic acid) to give 5-ethyl-N- (1- (3-fluorobenzyl) -1H-pyrazol-4-yl) isoxazole-3-carboxamide (0.167g, 0.53mmol, 52%) as a pink solid.1H NMR (400MHz, dimethylsulfoxide-d)6)δ10.90(s,1H),8.17(s,1H),7.66(s,1H),7.40(dt,J=14.0,7.2Hz,1H),7.18-7.09(m,1H),7.05(t,J=8.4Hz,2H),6.64(s,1H),5.34(s,2H),2.84(q,J=7.6Hz,2H),1.26(t,J=7.6Hz,3H);LCMS(ESI)m/z:315.1[M+H]+

EXAMPLE 29 preparation of 5-chloro-N- (1- (3-fluorobenzyl) -1H-pyrazol-4-yl) isoxazole-3-carboxamide (105)

Step 1: preparation of ethyl 5-chloroisoxazole-3-carboxylate

To a solution of (Z) -2-chloro-2- (hydroxyimino) acetic acid ethyl ester (10.0g, 66.2mmol) in 1, 1-dichloroethylene (100mL) was added a solution of triethylamine (16.8g, 166mmol) in 1, 1-dichloroethylene (100mL) over a period of 2 h. The reaction mixture was stirred at room temperature for 16 h. The reaction was poured into water (500mL) and extracted with dichloromethane (200 mL. times.3). The combined organic layers were washed with brine (500mL), dried over sodium sulfate, filtered and concentrated in vacuo. The crude product was purified by column chromatography (silica, ethyl acetate/petroleum ether ═ 1/20) to give ethyl 5-chloroisoxazole-3-carboxylate (1.67g, mmol, 14%) as a yellow oil. LCMS (ESI) M/z 376.1[ M + H ]+

Step 2: preparation of 5-chloroisoxazole-3-carboxylic acid

A mixture of ethyl 5-chloroisoxazole-3-carboxylate (0.200g, 1.14mmol) in tetrahydrofuran (1.6mL) and water (0.4mL) was cooled to 0 ℃ and lithium hydroxide hydrate (0.096g, 2.28mmol) was added. The mixture was stirred at 0 ℃ for 0.5h, then diluted with water (10mL) and extracted with ethyl acetate (10 mL. times.2). The aqueous phase was separated and acidified to pH 2-3 with 1N aqueous hydrochloric acid solution, followed by extraction with ethyl acetate (10mL × 3). The combined extracts were dried over sodium sulfate, filtered and concentrated in vacuo to give 5-chloroisoxazole-3-carboxylic acid (140mg, crude) as a white solid, which was used directly in the next step. LCMS (ESI) M/z 148.1[ M + H ]]+

And step 3: preparation of 5-chloro-N- (1- (3-fluorobenzyl) -1H-pyrazol-4-yl) isoxazole-3-carboxamide

To a solution of 5-chloroisoxazole-3-carboxylic acid (0.120g, 0.816mmol) in dichloromethane (2mL) was added N, N-dimethylformamide (1 drop) and oxalyl chloride (0.207g, 1.63mmol) at 0 ℃. The reaction mixture was stirred at 0 ℃ for 0.5h, then concentrated in vacuo. The crude yellow oil was dissolved in dichloromethane (2mL) and added to a mixture of 1- (3-fluorobenzyl) -1H-pyrazol-4-amine hydrochloride (0.185g, 0.816mmol) and pyridine (0.258g, 3.26mmol) in dichloromethane (2mL) at 0 ℃. The reaction mixture was stirred at 0 ℃ for 20 min. The reaction mixture was concentrated in vacuo, then dissolved in a minimal amount of N, N-dimethylformamide and purified by preparative-HPLC (Boston C1821 × 250mm 10 μm column; acetonitrile/0.01% aqueous trifluoroacetic acid) to give 5-chloro-N- (1- (3-fluorobenzyl) -1H-pyrazol-4-yl) isoxazole-3-carboxamide (0.111g, 0.348mmol, 36%) as a white solid. 1H NMR (500MHz, dimethylsulfoxide-d)6)δ11.07(s,1H),8.19(s,1H),7.67(s,1H),7.44-7.36(m,1H),7.17-7.09(m,2H),7.05(t,J=9.5Hz,2H),5.36(s,2H);LCMS(ESI)m/z:321.1[M+H]+

Example 30 preparation of N- (1- (3-fluorobenzyl) -1H-pyrazol-4-yl) -5-morpholinoisoxazole-3-carboxamide (101)

Step 1: preparation of 3- (ethoxycarbonyl) isoxazole-5-carboxylic acid

To a solution of propiolic acid (23.0g, 328.5mmol) in N, N-dimethylformamide (200mL) was added dropwise a solution of (Z) -2-chloro-2- (hydroxyimino) acetic acid ethyl ester (20.0g, 132mmol) in N, N-dimethylformamide (100mL) at room temperature under nitrogen over 40 min. After addition, the reaction mixture was heated to 90 ℃ and a solution of triethylamine (40.1g, 397mmol) in N, N-dimethylformamide (100mL) was added dropwise over 1 h. The reaction mixture was heated at 90 ℃ for 17h and then cooled to room temperature. The reaction mixture was concentrated to dryness, diluted with water (200mL) and adjusted to pH 2 with 1N aqueous hydrogen chloride. The reaction mixture was then extracted with ethyl acetate (100mL × 2) and the combined organic layers were washed with brine (10mL × 2), dried over anhydrous sodium sulfate, filtered and concentrated in vacuo. Crude 3- (ethoxycarbonyl) isoxazole-5-carboxylic acid (10.3g, 55.6mmol, 16.9%) was obtained as a yellow oil and used directly in the next step without further purification. LCMS (ESI) M/z 186.0[ M + H ] ]+

Step 2: preparation of ethyl 5- (tert-butoxycarbonylamino) isoxazole-3-carboxylate

To a solution of 3- (ethoxycarbonyl) isoxazole-5-carboxylic acid (10.3g, 55.6mmol) in toluene (30mL) was added triethylamine (6.73g, 66.7mmol), tert-butanol (10.3g, 139.0mmol) and diphenylphosphoryl azide (18.3g, 66.7 mmol). The reaction mixture was heated at 100 ℃ for 2h, then cooled and concentrated under reduced pressure. The residue was purified by column chromatography (silica, petroleum ether/ethyl acetate-8/1) to give ethyl 5- (tert-butoxycarbonylamino) isoxazole-3-carboxylate (3.1g, 12.1mmol, 22%) as a white solid.

And step 3: preparation of 5-aminoisoxazole-3-carboxylic acid ethyl ester

To a solution of ethyl 5- (tert-butoxycarbonylamino) isoxazole-3-carboxylate (3.1g, 12.1mmol) in dichloromethane (30mL) was added trifluoroacetic acid (15 mL). The mixture was stirred at room temperature for 3h, then concentrated in vacuo. The residue was purified by column chromatography (silica, petroleum ether/ethyl acetate 1/1) to give ethyl 5-aminoisoxazole-3-carboxylate (0.740g, 4.74mmol, 41%) as a white solid. LCMS (ESI) M/z 157.1[ M + H ]]+

And 4, step 4: preparation of 5-morpholinoisoxazole-3-carboxylic acid ethyl ester

To a solution of 5-aminoisoxazole-3-carboxylic acid ethyl ester (0.320g, 2.05mmol) in dimethylacetamide (20mL) was added cesium carbonate (1.99g, 6.15mmol) and 1-bromo-2- (2-bromoethoxy) ethane (1.18g, 5.12 mmol). The reaction mixture was stirred at 80 ℃ for 3h and then cooled to room temperature. The mixture was diluted with water (30mL) and then extracted with ethyl acetate (10 mL. times.2). The combined organic layers were washed with brine (10mL × 2), dried over anhydrous sodium sulfate, filtered and concentrated in vacuo. The residue was purified by column chromatography (silica, petroleum ether/ethyl acetate ═ 3:1) to give ethyl 5-morpholinoisoxazole-3-carboxylate (0.260g, 1.15mmol, 56%) as a white solid. LCMS (ESI) M/z 227.2[ M + H ]]+

And 5: preparation of 5-morpholinoisoxazole-3-carboxylic acid

To a solution of ethyl 5-morpholinoisoxazole-3-carboxylate (0.260g, 1.15mmol) in tetrahydrofuran/water (v/v ═ 4:1, 20mL) was added lithium hydroxide hydrate (0.145g, 3.45mmol) at-20 ℃. The reaction mixture was stirred at 0 ℃ for 1h, then concentrated in vacuo, diluted with water (15mL) and adjusted to pH 2 with 1N aqueous hydrogen chloride. The reaction mixture was then extracted with dichloromethane (10 mL. times.2). The combined organic layers were washed with brine (10mL × 2), dried over anhydrous sodium sulfate, filtered and concentrated in vacuo to give 5-morpholinoisoxazole-3-carboxylic acid (0.200g, 1.01mmol, 88%) as a white solid, which was used without further purification. LCMS (ESI) M/z 199.1[ M + H ] ]+

Step 6: preparation of N- (1- (3-fluorobenzyl) -1H-pyrazol-4-yl) -5-morpholinoisoxazole-3-carboxamide

To a solution of 5-morpholinoisoxazole-3-carboxylic acid (0.200g, 1.01mmol) in dichloromethane (15mL) at 0 ℃ were added oxalyl chloride (0.256g, 2.02mmol) and N, N-dimethylformamide (0.01 mL). The reaction mixture was warmed to room temperature for 1h and then concentrated in vacuo. The residue was diluted with dichloromethane (8mL) and added to a solution of 1- (3-fluorobenzyl) -1H-pyrazol-4-amine (0.193g, 1.01mmol) and pyridine (0.359g, 4.54mmol) in dichloromethane (10mL) at 0 ℃. The reaction mixture was stirred at room temperature for 2 h. The reaction mixture was evaporated to dryness. The crude product was dissolved in a minimal amount of N, N-dimethylformamide and purified by preparative-HPLC (Boston C1821 × 250mm 10 μm column; acetonitrile/0.01% aqueous trifluoroacetic acid) to give N- (1- (3-fluorobenzyl) -1H-pyrazol-4-yl) -5-morpholinoisoxazole-3-carboxamide (0.0812g, 0.21mmol, 22%) as a white solid.1H NMR (400MHz, dimethylsulfoxide-d)6)δ10.72(s,1H),8.16(s,1H),7.66(s,1H),7.39(dt,J=14.0,7.1Hz,1H),7.18-6.95(m,3H),5.72(s,1H),5.34(s,2H),3.76-3.61(m,4H),3.36-3.30(m,4H);LCMS(ESI)m/z:372.1[M+H]+

EXAMPLE 31 preparation of 5-cyclohexenyl-N- (1- (3-fluorobenzyl) -1H-pyrazol-4-yl) isoxazole-3-carboxamide (85)

Step 1: preparation of 5-cyclohexenylisoxazole-3-carboxylic acid ethyl ester

To a mixture of ethyl 5-bromoisoxazole-3-carboxylate (1.5g, 6.81mmol) in 1, 4-dioxane (30mL) and water (10mL) was added cyclohexenylboronic acid (1.02g, 8.17mmol), [1, 1' -bis (diphenylphosphino) ferrocene under nitrogen]Palladium (II) dichloride (0.497g, 0.68mmol) and cesium carbonate (6.63g, 20.4 mmol). The mixture was heated at 90 ℃ for 3h and then cooled to room temperature. The reaction mixture was diluted with water (20mL) and extracted with ethyl acetate (20 mL. times.2). The combined organic layers were washed with water (20mL × 2) and brine (20mL × 2), dried over anhydrous sodium sulfate, filtered and concentrated in vacuo. The residue was purified by column chromatography (silica, petroleum ether/ethyl acetate-20/1) to give ethyl 5-cyclohexenylisoxazole-3-carboxylate (1.1g, 4.97mmol, 73%) as a yellow oil. LCMS (ESI) M/z 222.1[ M + H ]]+

Step 2: preparation of 5-cyclohexenylisoxazole-3-carboxylic acid

To a solution of ethyl 5-cyclohexenylisoxazole-3-carboxylate (1.1g, 4.97mmol) in tetrahydrofuran/water (v/v ═ 4:1, 20mL) was added sodium hydroxide (0.476g, 11.9mmol) at room temperature. The reaction mixture was stirred at room temperature for 17 h. The reaction mixture was evaporated to dryness, diluted with water (20mL) and washed with 1N aqueous hydrogen chloride The solution was adjusted to pH 2. The aqueous layer was extracted with ethyl acetate (10mL × 2) and the combined organic layers were washed with brine (10mL × 2), dried over anhydrous sodium sulfate, filtered and concentrated in vacuo to give 5-cyclohexenylisoxazole-3-carboxylic acid as a white solid (0.570g, 2.95mmol, 59%). LCMS (ESI) M/z 194.2[ M + H ]]+. This material was used directly in the next step without further purification.

And step 3: preparation of 5-cyclohexenyl-N- (1- (3-fluorobenzyl) -1H-pyrazol-4-yl) isoxazole-3-carboxamide

To 5-cyclohexenylisoxazole-3-carboxylic acid (0.410g, 2.12mmol), 1- (3-fluorobenzyl) -1H-pyrazol-4-amine (0.405g, 2.12mmol) and 1- [ bis (dimethylamino) methylene]-1H-1, 2, 3-triazolo [4, 5-b]To a solution of pyridinium 3-oxide hexafluorophosphate (2.41g, 6.36mmol) in N, N-dimethylformamide (15mL) was added diisopropylethylamine (0.820g, 6.36 mmol). The mixture was stirred at room temperature for 17h, then concentrated in vacuo. The crude product was dissolved in a minimal amount of N, N-dimethylformamide and purified by preparative-HPLC (Boston C1821 × 250mm 10 μm column; acetonitrile/0.01% aqueous trifluoroacetic acid) to give 5-cyclohexenyl-N- (1- (3-fluorobenzyl) -1H-pyrazol-4-yl) isoxazole-3-carboxamide (0.300g, 0.81mmol, 27%) as a white solid. 1H NMR (500MHz, dimethylsulfoxide-d)6)δ10.92(s,1H),8.18(s,1H),7.67(s,1H),7.39(td,J=7.9,6.3Hz,1H),7.13(td,J=8.6,2.3Hz,1H),7.05(t,J=9.8Hz,2H),6.83(s,1H),6.68(d,J=4.0Hz,1H),5.35(s,2H),2.29(dd,J=60.1,2.9Hz,4H),1.77-1.54(m,4H);LCMS(ESI)m/z:367.1[M+H]+

Example 32.preparation of 5-cyclohexyl-N- (1- (3-fluorobenzyl) -1H-pyrazol-4-yl) isoxazole-3-carboxamide (83)

Step 1: preparation of 5-cyclohexenylisoxazole-3-carboxylic acid ethyl ester

To a solution of ethyl 5-bromoisoxazole-3-carboxylate (1.5g, 6.81mmol) in 1, 4-dioxane (30mL) and water (10mL) was added cyclohexenylboronic acid (1.02g, 8.17mmol) and [1, 1' -bis (diphenylphosphino) ferrocene under nitrogen]Palladium (II) dichloride (0.497g, 0.68mmol) and cesium carbonate (6.63g, 20.4 mmol). The mixture was heated at 90 ℃ for 3h, then cooled, diluted with water (20mL) and extracted with ethyl acetate (20 mL. times.2). The combined organic layers were washed with water (20mL × 2) and brine (20mL × 2), dried over anhydrous sodium sulfate, filtered and concentrated in vacuo. The residue was purified by column chromatography (silica, petroleum ether/ethyl acetate-20/1) to give ethyl 5-cyclohexenylisoxazole-3-carboxylate (1.1g, 4.97mmol, 73%) as a yellow oil. LCMS (ESI) M/z 222.1[ M + H ]]+

Step 2: preparation of 5-cyclohexenylisoxazole-3-carboxylic acid

To a solution of ethyl 5-cyclohexenylisoxazole-3-carboxylate (1.1g, 4.97mmol) in tetrahydrofuran/water (v/v ═ 4:1, 20mL) was added sodium hydroxide (0.476g, 11.9mmol) at room temperature. The reaction mixture was stirred at room temperature for 17 h. The reaction mixture was evaporated to dryness, diluted with water (20mL) and adjusted to pH 2 with 1N aqueous hydrogen chloride. The aqueous layer was then extracted with ethyl acetate (10mL × 2) and the combined organic layers were washed with brine (10mL × 2), dried over anhydrous sodium sulfate, filtered and concentrated in vacuo to give 5-cyclohexenylisoxazole-3-carboxylic acid as a white solid (0.570g, 2.95mmol, 59%). LCMS (ESI) M/z 194.2[ M + H ] ]+. This material was used directly in the next step without further purification.

And step 3: preparation of 5-cyclohexenyl-N- (1- (3-fluorobenzyl) -1H-pyrazol-4-yl) isoxazole-3-carboxamide

To 5-cyclohexenylisoxazole-3-carboxylic acid (0.410g, 2.12mmol), 1- (3-fluorobenzyl) -1H-pyrazol-4-amine (0.405g, 2.12mmol) and 1- [ bis (dimethylamino) methylene]-1H-1, 2, 3-triazolo [4, 5-b]To a solution of pyridinium 3-oxide hexafluorophosphate (2.41g, 6.36mmol) in N, N-dimethylformamide (15mL) was added diisopropylethylamine (0.820g, 6.36 mmol). The mixture was stirred at room temperature for 17h, then concentrated under reduced pressure. The crude product was dissolved in a minimal amount of N, N-dimethylformamide and purified by preparative-HPLC (Boston C1821 × 250mm 10 μm column; acetonitrile/0.01% aqueous trifluoroacetic acid) to give 5-cyclohexenyl-N- (1- (3-fluorobenzyl) -1H-pyrazol-4-yl) isoxazole-3-carboxamide (0.300g, 0.81mmol, 27%) as a white solid.1H NMR (500MHz, dimethylsulfoxide-d)6)δ10.92(s,1H),8.18(s,1H),7.67(s,1H),7.39(td,J=7.9,6.3Hz,1H),7.13(td,J=8.6,2.3Hz,1H),7.05(t,J=9.8Hz,2H),6.83(s,1H),6.68(d,J=4.0Hz,1H),5.35(s,2H),2.29(dd,J=60.1,2.9Hz,4H),1.77-1.54(m,4H);LCMS(ESI)m/z:367.1[M+H]+

And 4, step 4: preparation of 5-cyclohexyl-N- (1- (3-fluorobenzyl) -1H-pyrazol-4-yl) isoxazole-3-carboxamide

To a stirred solution of 5-cyclohexenyl-N- (1- (3-fluorobenzyl) -1H-pyrazol-4-yl) isoxazole-3-carboxamide (0.240g, 0.65mmol) in ethanol (10mL) was added palladium on activated carbon (0.100g, 10 wt% Pd) at room temperature. The mixture was stirred under hydrogen atmosphere (balloon) for 0.5 h. Passing the mixture through Filtering and filteringThe solution was concentrated in vacuo. The crude product was dissolved in a minimal amount of N, N-dimethylformamide and purified by preparative-HPLC (Boston C1821 × 250mm 10 μm column; acetonitrile/0.01% aqueous trifluoroacetic acid) to give 5-cyclohexyl-N- (1- (3-fluorobenzyl) -1H-pyrazol-4-yl) isoxazole-3-carboxamide (0.139g, 0.37mmol, 58%) as a white solid.1H NMR (400MHz, dimethylsulfoxide-d)6)δ10.90(s,1H),8.18(s,1H),7.66(s,1H),7.39(dt,J=14.0,7.2Hz,1H),7.18-6.92(m,3H),6.62(s,1H),5.35(s,2H),2.89(td,J=10.9,3.4Hz,1H),2.00(d,J=11.4Hz,2H),1.82-1.59(m,3H),1.42(tt,J=24.3,7.3Hz,4H),1.31-1.17(m,1H);LCMS(ESI)m/z:369.2[M+H]+

Example 33 preparation of N- (1- (3-fluorobenzyl) -1H-pyrazol-4-yl) -5- (piperidin-1-yl) isoxazole-3-carboxamide (102)

Step 1: preparation of 3- (ethoxycarbonyl) isoxazole-5-carboxylic acid

To a solution of propiolic acid (23.0g, 3295mmol) in N, N-dimethylformamide (200mL) was added dropwise a solution of (Z) -2-chloro-2- (hydroxyimino) acetic acid ethyl ester (20.0g, 132mmol) in N, N-dimethylformamide (100mL) at room temperature under nitrogen over 40 min. After the addition, the reaction mixture was heated to 90 ℃, and then a solution of triethylamine (40.1g, 397mmol) in N, N-dimethylformamide (100mL) was added dropwise over 1 h. The reaction mixture was heated at 90 ℃ for 17h, then cooled to room temperature and evaporated to dryness. The residue was diluted with water (200mL) and adjusted to pH 2 with 1N aqueous hydrogen chloride. The aqueous layer was extracted with ethyl acetate (100 mL. times.2) and the combined organic layers were washed with brine (10 mL. times.2), dried over anhydrous sodium sulfate, filtered and concentrated in vacuo to give 3- (ethoxycarbonyl) isoxazole- 5-Carboxylic acid (10.3g, 55.6mmol, 17%). LCMS (ESI) M/z 186.1[ M + H ]]+. This material was used directly in the next step without further purification.

Step 2: preparation of ethyl 5- (tert-butoxycarbonylamino) isoxazole-3-carboxylate

To a solution of 3- (ethoxycarbonyl) isoxazole-5-carboxylic acid (10.3g, 55.6mmol) in toluene (30mL) was added triethylamine (6.73g, 66.7mmol), tert-butanol (10.3g, 139.0mmol) and diphenylphosphoryl azide (18.3g, 66.7 mmol). The reaction mixture was heated at 100 ℃ for 2h, then cooled and concentrated to dryness. The residue was purified by column chromatography (silica, petroleum ether/ethyl acetate-8/1) to give ethyl 5- (tert-butoxycarbonylamino) isoxazole-3-carboxylate (3.1g, 12.1mmol, 22%) as a white solid. LCMS (ESI) M/z 257.2[ M + H ]]+

And step 3: preparation of 5-aminoisoxazole-3-carboxylic acid ethyl ester

To a solution of ethyl 5- (tert-butoxycarbonylamino) isoxazole-3-carboxylate (3.1g, 12.1mmol) in dichloromethane (30mL) was added trifluoroacetic acid (15 mL). After addition, the mixture was stirred at room temperature for 3h, then concentrated in vacuo. The residue was purified by column chromatography (silica, petroleum ether/ethyl acetate 1/1) to give ethyl 5-aminoisoxazole-3-carboxylate (0.740g, 4.74mmol, 41%) as a white solid. (LCMS (ESI) M/z:157.1[ M + H ] ]+

And 4, step 4: preparation of 5- (piperidin-1-yl) isoxazole-3-carboxylic acid ethyl ester

To 5-aminoiso-amino at room temperatureTo a solution of oxazole-3-carboxylic acid ethyl ester (0.280g, 1.79mmol) in N, N-dimethylacetamide (20mL) were added cesium carbonate (1.74g, 5.37mmol) and 1, 5-dibromopentane (1.02g, 4.47 mmol). The reaction mixture was heated at 80 ℃ for 3h, then cooled to room temperature and diluted with water (30 mL). The aqueous layer was extracted with ethyl acetate (10mL × 2) and the combined organic layers were washed with brine (10mL × 2), dried over anhydrous sodium sulfate, filtered and concentrated in vacuo. The residue was purified by column chromatography (silica, petroleum ether/ethyl acetate 3/1) to give ethyl 5- (piperidin-1-yl) isoxazole-3-carboxylate (0.300g, 1.33mmol, 75%) as a white solid. LCMS (ESI) M/z 225.3[ M + H ]]+

And 5: preparation of 5- (piperidin-1-yl) isoxazole-3-carboxylic acids

To a solution of ethyl 5- (piperidin-1-yl) isoxazole-3-carboxylate (0.300mg, 1.33mmol) in tetrahydrofuran/water (v/v ═ 4:1, 20mL) was added lithium hydroxide hydrate (0.894g, 3.99mmol) at-20 ℃. The reaction mixture was stirred at 20 ℃ for 10min, then evaporated to dryness to give 5- (piperidin-1-yl) isoxazole-3-carboxylic acid (0.220g, 1.12mmol, 84%) as a white solid. (LCMS (ESI) M/z 197.1[ M + H ] ]+. This material was used directly in the next step without further purification.

Step 6: preparation of N- (1- (3-fluorobenzyl) -1H-pyrazol-4-yl) -5- (piperidin-1-yl) isoxazole-3-carboxamide

To a solution of 5- (piperidin-1-yl) isoxazole-3-carboxylic acid (0.220g, 1.12mmol) in N, N-dimethylformamide (15mL) was added 1- (3-fluorobenzyl) -1H-pyrazol-4-amine (0.214g, 1.12mmol), 1- [ bis (dimethylamino) methylene ] -methylene]-1H-1, 2, 3-triazolo [4, 5-b]Pyridinium 3-oxide hexafluorophosphate (1.27g, 3.36mmol) and diisopropylethylamine (0.433g, 3.36 mmol). The mixture was cooled to room temperatureStirred for 17h then concentrated in vacuo. The crude product was dissolved in a minimal amount of N, N-dimethylformamide and purified by preparative-HPLC (Boston C1821 × 250mm 10 μm column; acetonitrile/0.01% aqueous trifluoroacetic acid) to give N- (1- (3-fluorobenzyl) -1H-pyrazol-4-yl) -5- (piperidin-1-yl) isoxazole-3-carboxamide (0.0317g, 0.08mmol, 8%) as a white solid.1H NMR (500MHz, dimethylsulfoxide-d)6)δ10.66(s,1H),8.15(s,1H),7.65(s,1H),7.39(td,J=8.0,6.3Hz,1H),7.18-6.89(m,3H),5.62(s,1H),5.34(s,2H),3.34(s,4H),1.58(s,6H);LCMS(ESI)m/z:370.1[M+H]+

Example 34 preparation of N- (1- (3-fluorobenzyl) -1H-pyrazol-4-yl) -5- (oxazol-5-yl) isoxazole-3-carboxamide (86)

Step 1: preparation of ethyl 5- (hydroxymethyl) isoxazole-3-carboxylate

To a solution of prop-2-yn-1-ol (3.3g, 59.4mmol) in N, N-dimethylformamide (40mL) was added dropwise a solution of ethyl (Z) -2-chloro-2- (hydroxyimino) acetate (3.0g, 19.8mmol) in N, N-dimethylformamide (10mL) under nitrogen over 40 min. After the addition, the reaction mixture was heated to 90 ℃, and then a solution of triethylamine (5.9g, 59.4mmol) in N, N-dimethylformamide (10mL) was added dropwise over 1 h. The reaction mixture was heated at 90 ℃ for 17h and then cooled to room temperature. The reaction mixture was diluted with ethyl acetate (50mL), washed with water (60mL × 2) and brine (60mL × 2), dried over anhydrous sodium sulfate, filtered and concentrated in vacuo. The residue was purified by column chromatography (silica, petroleum ether/ethyl acetate-2/1) to give ethyl 5- (hydroxymethyl) isoxazole-3-carboxylate (1.6g, 9.3mmol, 49%) as a yellow oil. LCMS (ESI) M/z 172.1[ M + H ] ]+

Step 2: preparation of ethyl 5-formylisoxazole-3-carboxylate

To a solution of ethyl 5- (hydroxymethyl) isoxazole-3-carboxylate (1.2g, 7.0mmol) in ethyl acetate (20mL) was added 2-iodoxybenzoic acid (5.9g, 21.0 mmol). After addition, the reaction mixture was heated to 90 ℃ and stirred for 17h, then cooled to room temperature. The reaction mixture was filtered and concentrated in vacuo. The residue was purified by column chromatography (silica, petroleum ether/ethyl acetate 5/1) to give ethyl 5-formylisoxazole-3-carboxylate (0.540g, 3.2mmol, 46%) as a yellow oil.

And step 3: preparation of 5- (oxazol-5-yl) isoxazole-3-carboxylic acids

To a solution of 1- (isocyanomethylsulfonyl) -4-toluene (0.622g, 3.19mmol) in acetonitrile (20mL) was added potassium carbonate (0.527g, 3.82 mmol). The reaction mixture was stirred at room temperature for 1 h. The reaction mixture was cooled to 0 ℃ and 5- (oxazol-5-yl) isoxazole-3-carboxylic acid (0.540g, 3.19mmol) was added. The solution was heated to 90 ℃ and stirred for 17 h. The reaction mixture was cooled to room temperature, diluted with water (15mL) and adjusted to pH 2 with 1N aqueous hydrogen chloride. The reaction mixture was then extracted with ethyl acetate (10 mL. times.2). The combined organic layers were washed with brine (10mL × 2), dried over anhydrous sodium sulfate, filtered and concentrated in vacuo to afford 5- (oxazol-5-yl) isoxazole-3-carboxylic acid (0.140mg, 0.77mmol, 24%) as a white solid. LCMS (ESI) M/z 181.1[ M + H ] ]+

And 4, step 4: preparation of N- (1- (3-fluorobenzyl) -1H-pyrazol-4-yl) -5- (oxazol-5-yl) isoxazole-3-carboxamide

To a mixture of 5- (oxazol-5-yl) isoxazole-3-carboxylic acid (0.140g, 0.77mmol), 1- (3-fluorobenzyl) -1H-pyrazol-4-amine (0.147g, 0.77mmol) and 1- [ bis (dimethylamino) methylene]-1H-1, 2, 3-triazolo [4, 5-b]To a solution of pyridinium 3-oxide hexafluorophosphate (0.877g, 2.31mmol) in N, N-dimethylformamide (6mL) was added diisopropylethylamine (0.298g, 2.31 mmol). The mixture was stirred at room temperature for 17 h. The crude reaction mixture was purified by preparative-HPLC (Boston C1821X 250mm 10 μm column; acetonitrile/0.01% aqueous trifluoroacetic acid) to give N- (1- (3-fluorobenzyl) -1H-pyrazol-4-yl) -5- (oxazol-5-yl) isoxazole-3-carboxamide (0.070g, 0.19mmol, 26%) as a white solid.1H NMR (500MHz, dimethylsulfoxide-d)6)δ11.11(s,1H),8.74(s,1H),8.20(d,J=18.7Hz,1H),8.02(s,1H),7.69(s,1H),7.45-7.28(m,2H),7.20-6.95(m,3H),5.37(s,2H);LCMS(ESI)m/z:354.1[M+H]+

Example 35 preparation of N- (1- (3-fluorobenzyl) -1H-pyrazol-4-yl) -5- (isothiazol-5-yl) isoxazole-3-carboxamide (81)

Step 1: preparation of 5-ethynylisothiazole

To a stirred mixture of dimethyl 1-diazo-2-oxopropylphosphonate (2.12g, 11.0mmol), potassium carbonate (2.76g, 20.0mmol) in methanol (100mL) at 0 ℃ was added dropwise a solution of isothiazole-5-carbaldehyde (1.13g, 10.0mmol) in methanol (5 mL). The reaction mixture was stirred at room temperature for 16h, then poured into water (50mL) and extracted with ethyl acetate (100mL × 2). The combined organic phases were dried over sodium sulfate, filtered and concentrated to give 5-ethynylisothiazole as a red-brown oil (0.250g, 2.29mmol, 23%). 1H NMR (500MHz, chloroform-d) δ 8.44(d, J ═ 1.5Hz, 1H), 7.36 (d),J=1.5Hz,1H),3.80(s,1H)。

Step 2: preparation of 5- (isothiazol-5-yl) isoxazole-3-carboxylic acid ethyl ester

To a stirred solution of 5-ethynylisothiazole (0.250g, 2.3mmol) in N, N-dimethylformamide (2mL) was added dropwise a solution of ethyl 2-chloro-2- (hydroxyimino) acetate (0.347g, 2.3mmol) in N, N-dimethylformamide (2 mL). After the addition was complete, the mixture was stirred at room temperature for 1h, then heated to 90 ℃. Triethylamine (0.465g, 4.6mmol) was added dropwise and the reaction mixture was heated at 90 ℃ for 16 h. The reaction mixture was cooled to room temperature and quenched with water (20mL), and extracted with ethyl acetate (40mL × 2). The combined organic phases were dried over sodium sulfate, filtered and concentrated in vacuo. The residue was purified by column chromatography (silica, petroleum ether/ethyl acetate-10/1) to give ethyl 5- (isothiazol-5-yl) isoxazole-3-carboxylate (0.080g, 0.357mmol, 16%) as an off-white solid. LCMS (ESI) M/z 225.1[ M + H ]]+

And step 3: preparation of 5- (isothiazol-5-yl) isoxazole-3-carboxylic acid

The synthesis of 5- (isothiazol-5-yl) isoxazole-3-carboxylic acid was carried out following the same procedure as in example 24. Compound 5- (isothiazol-5-yl) isoxazole-3-carboxylic acid (40mg, 0.94mmol, 58%) was obtained as an off-white solid. LCMS (ESI) M/z 197.1[ M + H ] ]+

And 4, step 4: preparation of N- (1- (3-fluorobenzyl) -1H-pyrazol-4-yl) -5- (isothiazol-5-yl) isoxazole-3-carboxamide

Following the same procedure as in example 24The same procedure was used for the synthesis of N- (1- (3-fluorobenzyl) -1H-pyrazol-4-yl) -5- (isothiazol-5-yl) isoxazole-3-carboxamide. The compound N- (1- (3-fluorobenzyl) -1H-pyrazol-4-yl) -5- (isothiazol-5-yl) isoxazole-3-carboxamide (0.0329g, 0.089mmol, 45%) was obtained as a white solid.1H NMR (500MHz, dimethylsulfoxide-d)6)δ.11.11(s,1H),8.76(d,J=2.0Hz,1H),8.22(s,1H),8.10(d,J=1.5Hz,1H),7.69(s,1H),7.61(s,1H),7.40-7.37(m,1H),7.13(td,J=8.5,2.0Hz,1H),7.07-7.03(m,2H),5.36(s,2H);LCMS(ESI)m/z:370.0[M+H]+

Example 36 preparation of N- (1- (3-fluorobenzyl) -1H-pyrazol-4-yl) -5- (pyridin-4-yl) isoxazole-3-carboxamide trifluoroacetate (56)

The synthesis of N- (1- (3-fluorobenzyl) -1H-pyrazol-4-yl) -5- (pyridin-4-yl) isoxazole-3-carboxamide trifluoroacetate was performed following the same procedure as in example 24. N- (1- (3-fluorobenzyl) -1H-pyrazol-4-yl) -5- (pyridin-4-yl) isoxazole-3-carboxamide trifluoroacetate was obtained as a white solid (0.0509g, 0.11mmol, 52%).1H NMR (500MHz, dimethylsulfoxide-d)6)δ11.15(s,1H),8.88-8.87(m,2H),8.24(s,1H),8.10-8.08(m,2H),7.85(s,1H),7.72(s,1H),7.43-7.38(m,3H),5.38(s,2H);LCMS(ESI)m/z:364.1.[M+H]+

Example 37 preparation of N- (1- (3-fluorobenzyl) -1H-pyrazol-4-yl) -5- (thiazol-5-yl) isoxazole-3-carboxamide (69)

Step 1: preparation of 5- ((trimethylsilyl) ethynyl) thiazole

5-bromothiazole (4.0g, 24.5mmol), ethynyltrimethylsilane (4.8g, 48.96mmol) and [1, 1' -bis (diphenylphosphino) ferrocene ]A mixture of palladium (II) dichloride (1.96g, 2.4mmol) and cuprous iodide (475mg, 2.4mmol) in triethylamine (30mL) was heated at 80 ℃ for 16 h. The mixture was concentrated and purified by column chromatography (silica, petroleum ether/ethyl acetate 10/1) to give 5- ((trimethylsilyl) ethynyl) thiazole (3.2g, 17.7mmol, 72%) as a pale yellow oil. LCMS (ESI) M/z 182.1[ M + H ]]+

Step 2: preparation of 5-ethynylthiazole

A mixture of 5- ((trimethylsilyl) ethynyl) thiazole (4.0g, 22.0mmol), KOH (1.24g, 22.0mmol) in methanol (30mL) was stirred at room temperature for 1 h. The mixture was quenched with saturated aqueous ammonium chloride solution and extracted with ethyl acetate (150 mL. times.2). The combined organic phases were dried over sodium sulfate, filtered and concentrated to give 5-ethynylthiazole (1.5g, 13.8mmol, 63%) as a brown oil.1H NMR (500MHz, chloroform-d) delta 8.72(s, 1H), 8.04(s, 1H), 3.46(s, 1H).

And step 3: preparation of 5- (thiazol-5-yl) isoxazole-3-carboxylic acid ethyl ester

The synthesis of ethyl 5- (thiazol-5-yl) isoxazole-3-carboxylate was achieved using the same procedure as example 35 to give ethyl 5- (thiazol-5-yl) isoxazole-3-carboxylate as a pale yellow solid (0.800g, 3.57mmol, 26%). LCMS (ESI) M/z 225.1[ M + H ] ]+

And 4, step 4: preparation of 5- (thiazol-5-yl) isoxazole-3-carboxylic acids

The synthesis of 5- (thiazol-5-yl) isoxazole-3-carboxylic acid was performed following the same procedure as example 24. Compound 5- (thiazol-5-yl) isoxazole-3-carboxylic acid (0.280g, 1.42mmol, 80%) was obtained as a pale yellow solid. LCMS (ESI) M/z 197.1[ M + H ]]+

And 5: preparation of N- (1- (3-fluorobenzyl) -1H-pyrazol-4-yl) -5- (thiazol-5-yl) isoxazole-3-carboxamide

The synthesis of N- (1- (3-fluorobenzyl) -1H-pyrazol-4-yl) -5- (thiazol-5-yl) isoxazole-3-carboxamide was performed following the same procedure as example 24. The compound N- (1- (3-fluorobenzyl) -1H-pyrazol-4-yl) -5- (thiazol-5-yl) isoxazole-3-carboxamide (0.0581g, 0.157mmol, 39%) was obtained as a white solid.1H NMR (500MHz, dimethylsulfoxide-d)6)δ11.08(s,1H),9.36(s,1H),8.64(s,1H),8.22(s,1H),7.69(s,1H),7.44(s,1H),7.42-7.37(m,1H),7.13(td,J=8.5,2.0Hz,1H),7.07-7.04(m,2H),5.36(s,2H);LCMS(ESI)m/z:370.0[M+H]+

Example 38 preparation of N- (1- (3-fluorobenzyl) -1H-pyrazol-4-yl) -5- (pyrimidin-4-yl) isoxazole-3-carboxamide (90) and 5-acetyl-N- (1- (3-fluorobenzyl) -1H-pyrazol-4-yl) isoxazole-3-carboxamide (91)

Step 1: preparation of 5-acetyl isoxazole-3-carboxylic acid

The synthesis of 5-acetylisoxazole-3-carboxylic acid was carried out following the same procedure as in example 24 to give 5-acetylisoxazole-3-carboxylic acid (0.300g, crude) as a yellow solid. L is CMS(ESI)m/z:156.1[M+H]+

Step 2: preparation of 5-acetyl-N- (1- (3-fluorobenzyl) -1H-pyrazol-4-yl) isoxazole-3-carboxamide

The synthesis of 5-acetyl-N- (1- (3-fluorobenzyl) -1H-pyrazol-4-yl) isoxazole-3-carboxamide was performed following the same procedure as in example 24 to give 5-acetyl-N- (1- (3-fluorobenzyl) -1H-pyrazol-4-yl) isoxazole-3-carboxamide (0.370g, 1.13mmol, 58%) as a white solid.1H NMR (500MHz, dimethylsulfoxide-d)6)δ11.17(s,1H),8.22(s,1H),7.70(d,J=10.4Hz,2H),7.40(d,J=6.4Hz,1H),7.08(dd,J=27.9,20.3Hz,3H),5.37(s,2H),2.62(s,3H);LCMS(ESI)m/z:329.0[M+H]+

And step 3: preparation of (Z) -5- (3- (dimethylamino) acryloyl) -N- (1- (3-fluorobenzyl) -1H-pyrazol-4-yl) isoxazole-3-carboxamide

A mixture of 5-acetyl-N- (1- (3-fluorobenzyl) -1H-pyrazol-4-yl) isoxazole-3-carboxamide (0.342g, 0.891mmol) and N, N-dimethylformamide dimethyl acetal (0.21g, 1.78mmol) in toluene (2.6mL) was stirred at 110 ℃ for 18H. The volatiles were removed and the residue was recrystallized from ether/petroleum ether to give (Z) -5- (3- (dimethylamino) acryloyl) -N- (1- (3-fluorobenzyl) -1H-pyrazol-4-yl) isoxazole-3-carboxamide (0.290g, 0.76mmol, 83%) as a yellow solid. LCMS (ESI) M/z 384.1[ M + H ]]+

And 4, step 4: n- (1- (3-fluorobenzyl) -1H-pyrazol-4-yl) -5- (pyrimidin-4-yl) isoxazole-3-carboxamides

A mixture of (Z) -5- (3- (dimethylamino) acryloyl) -N- (1- (3-fluorobenzyl) -1H-pyrazol-4-yl) isoxazole-3-carboxamide (0.240g, 0.62mmol), formamidine acetate (0.196g, 1.88mmol), and potassium carbonate (0.259g, 1.88mmol) in N, N-dimethylformamide (6mL) was heated at 100 ℃ for 2H in a sealed tube. The reaction mixture was filtered and the filtrate was purified by preparative-HPLC (Boston C1821 × 250mm 10 μm column; acetonitrile/0.01% aqueous trifluoroacetic acid) to give N- (1- (3-fluorobenzyl) -1H-pyrazol-4-yl) -5- (pyrimidin-4-yl) isoxazole-3-carboxamide (0.060g, 0.16mmol, 26%) as a white solid. 1H NMR (500MHz, dimethylsulfoxide-d)6)δ11.16(s,1H),9.39(d,J=1.1Hz,1H),9.08(d,J=5.2Hz,1H),8.38-8.09(m,2H),7.75(d,J=35.1Hz,2H),7.54-7.26(m,1H),7.11(dt,J=18.8,9.1Hz,3H),5.38(s,2H);LCMS(ESI)m/z:365.0[M+H]+

Example 39 preparation of N- (1- (3-fluorobenzyl) -1H-pyrazol-4-yl) -5- (1-methoxycyclobutyl) isoxazole-3-carboxamide (115)

Step 1: preparation of 1-ethynyl cyclobutanol

To a stirred solution of cyclobutanone (1.4g, 20.0mmol) in tetrahydrofuran (20mL) was added ethynylmagnesium bromide (0.5M in tetrahydrofuran, 40mL) dropwise at 0 ℃. After the addition was complete, the reaction mixture was stirred at 0 ℃ for an additional 2h, then quenched with saturated aqueous ammonium chloride (40mL) and extracted with ethyl acetate (100 mL. times.2). The combined organic phases were dried over sodium sulfate, filtered and concentrated to provide 1-ethynyl cyclobutanol as a red oil (1.4g, 14.6mmol, 73%).1H NMR (500MHz, chloroform-d) delta 2.54(s, 1H), 2.46-2.42(m, 2H), 2.29-2.23(m, 2H), 1.88-1.80(m, 2H).

Step 2: preparation of ethyl 5- (1-hydroxycyclobutyl) isoxazole-3-carboxylate

The synthesis of the compound ethyl 5- (1-hydroxycyclobutyl) isoxazole-3-carboxylate was carried out following the same procedure as in example 35 to give ethyl 5- (1-hydroxycyclobutyl) isoxazole-3-carboxylate (3.0g, 0.014mmol, 24%) as a pale yellow oil. LCMS (ESI) M/z 212.1[ M + H ]]+

And step 3: 5- (1-methoxycyclobutyl) isoxazole-3-carboxylic acid

To a stirred solution of sodium hydride (0.208g, 60% in petroleum ether, 5.2mmol) in N, N-dimethylformamide (6mL) was added dropwise a solution of ethyl 5- (1-hydroxycyclobutyl) isoxazole-3-carboxylate (1.0g, 4.74mmol) in N, N-dimethylformamide (2mL) at room temperature. After addition, the mixture was stirred for a further 1h, then iodomethane (0.673g, 4.74mmol) was added dropwise. The reaction mixture was stirred at room temperature for an additional 2h, quenched with water (20mL), and extracted with ethyl acetate (50 mL. times.2). The combined organic phases were dried over sodium sulfate, filtered and concentrated in vacuo. The crude residue was dissolved in a minimal amount of N, N-dimethylformamide and purified by preparative-HPLC (Boston C1821 × 250mm 10 μm column. mobile phase acetonitrile/0.01% aqueous trifluoroacetic acid) to give 5- (1-methoxycyclobutyl) isoxazole-3-carboxylic acid as a pale yellow solid (0.200g, 1.01mmol, 21%). LCMS (ESI) M/z 198.1[ M + H ]]+

And 4, step 4: n- (1- (3-fluorobenzyl) -1H-pyrazol-4-yl) -5- (1-methoxycyclobutyl) isoxazole-3-carboxamide

N- (1- (3) was carried out following the same procedure as in example 24-fluorobenzyl) -1H-pyrazol-4-yl) -5- (1-methoxycyclobutyl) isoxazole-3-formamide synthesis. The crude sample was dissolved in a minimal amount of N, N-dimethylformamide and purified by preparative-HPLC (Boston C1821 × 250mM 10 μm column. mobile phase acetonitrile/10 mM aqueous ammonium acetate) to give N- (1- (3-fluorobenzyl) -1H-pyrazol-4-yl) -5- (1-methoxycyclobutyl) isoxazole-3-carboxamide as a colorless oil (72.7mg, 0.196mmol, 65%). 1H NMR (500MHz, dimethylsulfoxide-d)6)δ.10.96(s,1H),8.18(s,1H),7.67(s,1H),7.39(dd,J=14.0,7.5Hz,1H),7.14-7.10(m,1H),7.07-7.03(m,3H),5.35(s,2H),3.03(s,3H),2.45-2.34(m,4H),1.86-1.84(m,1H),1.70-1.65(m,1H);LCMS(ESI)m/z:371.1[M+H]+

EXAMPLE 40 preparation of N- [1- [ (3-fluorophenyl) methyl ] pyrazol-4-yl ] -5- (2-furyl) isoxazole-3-carboxamide (30)

Step 1: preparation of 1- [ (3-fluorophenyl) methyl ] -4-nitro-pyrazole

To a solution of 4-nitro-1H-pyrazole (0.100g, 0.884mmol) in N, N-dimethylformamide (3mL) was added cesium carbonate (0.864g, 2.65mmol) and 1- (bromomethyl) -3-fluoro-benzene (0.109mL, 0.884 mmol). The mixture was stirred at 20 ℃ for 3 h. The reaction mixture was diluted with water (20mL) and extracted with ethyl acetate (10 mL. times.3). The combined organic layers were washed with brine (20mL), dried over sodium sulfate, filtered and concentrated under reduced pressure to give a colorless residue. The crude product 1- [ (3-fluorophenyl) methyl group]-4-nitro-pyrazole (0.196g, 884mmol) was used in the next step without further purification. LCMS (ESI) M/z 222.1[ M + H ]]+

Step 2: preparation of 1- [ (3-fluorophenyl) methyl ] pyrazol-4-amine

To 1- [ (3-fluorophenyl) methyl group under nitrogen]To a solution of-4-nitro-pyrazole (0.196g, 0.884mmol) in ethanol (10mL) and water (2mL) were added ammonium chloride (155mL, 4.42mmol) and iron powder (0.247g, 4.42 mmol). The mixture was heated at 50 ℃ for 4 h. The reaction mixture was concentrated under reduced pressure to remove ethanol (2 mL). The residue was diluted with water (20mL) and extracted with ethyl acetate (10 mL. times.3). The combined organic layers were washed with brine (20mL), dried over sodium sulfate, filtered and concentrated under reduced pressure to give a residue as a brown oil. The crude product 1- [ (3-fluorophenyl) methyl group ]Pyrazol-4-amine (0.169g, 0.884mmol) was used in the next step without further purification. LCMS (ESI) M/z 192.1[ M + H ]]+

And step 3: preparation of N- [1- [ (3-fluorophenyl) methyl ] pyrazol-4-yl ] -5- (2-furyl) isoxazole-3-carboxamide

To a solution of 5- (2-furyl) isoxazole-3-carboxylic acid (0.100g, 0.558mmol) in N, N-dimethylformamide (3mL) at 15 deg.C were added N, N, N ', N' -tetramethyl-O- (1H-benzotriazol-1-yl) uronium hexafluorophosphate (0.254g, 0.670mmol), 1- [ (3-fluorophenyl) methyl]Pyrazol-4-amine (0.117g, 0.614mmol) and diisopropylethylamine (292mL, 1.67 mmol). The mixture was stirred at 15 ℃ for 2 h. The residue was purified by preparative-HPLC (YMC-Actus Triart C18100X 30mM X5 μm; mobile phase: [ water (10mM ammonium bicarbonate) -acetonitrile](ii) a B%: 40-60 percent, 12min) to obtain the N- [1- [ (3-fluorophenyl) methyl ] which is light yellow solid]Pyrazol-4-yl]-5- (2-furyl) isoxazole-3-carboxamide (0.126g, 0.36mmol, 64%).1H NMR (400MHz, chloroform-d) δ 8.60(br.s, 1H), 8.08(s, 1H), 7.61(br.d, J ═ 11.0Hz, 2H), 7.38-7.28(m, 1H), 7.12-6.81(m, 5H), 6.58(br.s, 1H), 5.30(s, 2H); LCMS (ESI) M/z 353.1[ M + H ]]+

Example 41 preparation of N- (1- (3-fluorobenzyl) -1H-pyrazol-4-yl) -5- (3-fluoropyridin-2-yl) isoxazole-3-carboxamide (108)

Step 1: preparation of 3-fluoro-2- ((trimethylsilyl) ethynyl) pyridine

The synthesis of the compound 3-fluoro-2- ((trimethylsilyl) ethynyl) pyridine was performed following the same procedure as example 37. Compound 3-fluoro-2- ((trimethylsilyl) ethynyl) pyridine (5.0g, 25.9mmol, 94%) was obtained as a red-brown oil. LCMS (ESI) M/z 194.1[ M + H ]]+

Step 2: preparation of 2-ethynyl-3-fluoropyridine

The synthesis of compound 2-ethynyl-3-fluoropyridine was performed following the same procedure as example 37. The compound 2-ethynyl-3-fluoropyridine (2.0g, 16.5mmol, 64%) was obtained as a dark brown oil.

And step 3: preparation of 5- (3-fluoropyridin-2-yl) isoxazole-3-carboxylic acid ethyl ester

The synthesis of the compound ethyl 5- (3-fluoropyridin-2-yl) isoxazole-3-carboxylate was performed following the same procedure as example 35. Compound ethyl 5- (3-fluoropyridin-2-yl) isoxazole-3-carboxylate (1.2g, 3.57mmol, 39%) was obtained as a yellow-green solid. LCMS (ESI) M/z 237.1[ M + H ]]+

And 4, step 4: preparation of 5- (3-fluoropyridin-2-yl) isoxazole-3-carboxylic acid

The synthesis of 5- (3-fluoropyridin-2-yl) isoxazole-3-carboxylic acid was carried out following the same procedure as example 24. Compound 5- (3-fluoropyridin-2-yl) isoxazole-3-carboxylic acid (0.400g, 1.92mmol, 76%) was obtained as a grey solid. LCMS (ESI) M/z 209.1[ M + H ] ]+

And 5: preparation of N- (1- (3-fluorobenzyl) -1H-pyrazol-4-yl) -5- (3-fluoropyridin-2-yl) isoxazole-3-carboxamide

The synthesis of N- (1- (3-fluorobenzyl) -1H-pyrazol-4-yl) -5- (3-fluoropyridin-2-yl) isoxazole-3-carboxamide was performed following the same procedure as example 24. The compound N- (1- (3-fluorobenzyl) -1H-pyrazol-4-yl) -5- (3-fluoropyridin-2-yl) isoxazole-3-carboxamide (0.151g, 0.395mmol, 82%) was obtained as a white solid.1H NMR (500MHz, dimethylsulfoxide-d)6)δ.11.12(s,1H),8.65(d,J=4.5Hz,1H),8.23(s,1H),8.03(t,J=10.0Hz,1H),7.73-7.69(m,2H),7.40-7.38(m,2H),7.13(td,J=8.5,2.0Hz,1H),7.08-7.04(m,2H),5.37(s,2H);LCMS(ESI)m/z:382.0[M+H]+

Example 42 preparation of N- (1- (3-fluorobenzyl) -1H-pyrazol-4-yl) -5- (oxetan-3-yl) isoxazole-3-carboxamide (139)

Step 1: preparation of 2, 2-dimethyl-1, 3-dioxane-5-carbaldehyde

To a solution of (2, 2-dimethyl-1, 3-dioxan-5-yl) methanol (11g, 75.3mmol) in ethyl acetate (150mL) under nitrogen was added 2-iodoxybenzoic acid (25.3g, 90.3 mmol). The reaction mixture was heated to 95 ℃ and stirred for 17h, then cooled to room temperature. The reaction mixture was filtered and concentrated in vacuo. The residue was purified by column chromatography (silica, petroleum ether/ethyl acetate ═ 1/1) to give 2, 2-dimethyl-1, 3-dioxane-5-carbaldehyde (2.8g, 19.4mmol, 26%) as a colorless oil. Three identical batches (each from 2.8g of starting material) were prepared and combined to give a total of 8.4g of 2, 2-dimethyl-1, 3-dioxane-5-carbaldehyde.

Step 2: preparation of 5-ethynyl-2, 2-dimethyl-1, 3-dioxane

To a solution of 2, 2-dimethyl-1, 3-dioxane-5-carbaldehyde (6.1g, 42.4mmol) in methanol (60mL) and diethyl ether (30mL) was added dimethyl (1-diazo-2-oxopropyl) phosphonate (16.2g, 84.7mmol) and potassium carbonate (23.4g, 169 mmol). The reaction mixture was stirred at 25 ℃ for 17 h. The reaction mixture was diluted with water (30mL) and extracted with petroleum ether (20 mL. times.3). The combined organic layers were washed with brine (10mL × 2), dried over anhydrous sodium sulfate, filtered and concentrated in vacuo to give crude 5-ethynyl-2, 2-dimethyl-1, 3-dioxane as a colorless oil (4.0g, 28.6mmol, 68%). LCMS (ESI) M/z 141.1[ M + H ]]+

And step 3: preparation of ethyl 5- (2, 2-dimethyl-1, 3-dioxan-5-yl) isoxazole-3-carboxylate

To a solution of 5-ethynyl-2, 2-dimethyl-1, 3-dioxane (4.0g, 28.5mmol) in N, N-dimethylformamide (10mL) was added dropwise a solution of (Z) -2-chloro-2- (hydroxyimino) acetic acid ethyl ester (4.3g, 28.5mmol) in N, N-dimethylformamide (40mL) under nitrogen over 40 min. After the addition, the reaction was mixedThe mixture was heated to 90 ℃ and a solution of triethylamine (8.6g, 85.5mmol) in N, N-dimethylformamide (10mL) was added dropwise over 1 h. The reaction mixture was heated at 90 ℃ for 17h and cooled to room temperature. The reaction mixture was diluted with ethyl acetate (20mL), washed with water (10mL × 2) and brine (10mL × 2), dried over anhydrous sodium sulfate, filtered and concentrated in vacuo. The residue was purified by column chromatography (silica, petroleum ether/ethyl acetate-10/1) to give ethyl 5- (2, 2-dimethyl-1, 3-dioxan-5-yl) isoxazole-3-carboxylate (1.6g, 6.27mmol, 22%) as a yellow solid. LCMS (ESI) M/z 256.1[ M + H ] ]+

And 4, step 4: preparation of ethyl 5- (1, 3-dihydroxypropan-2-yl) isoxazole-3-carboxylate

To a solution of ethyl 5- (2, 2-dimethyl-1, 3-dioxan-5-yl) isoxazole-3-carboxylate (1.5g, 5.88mmol) in methanol (20mL) was added 1N hydrochloric acid (10 mL). The reaction mixture was stirred at 25 ℃ for 1 h. The pH of the reaction mixture was adjusted to pH 8 with solid sodium bicarbonate and the aqueous layer was extracted with ethyl acetate (10mL × 2). The combined organic layers were washed with water (10mL) and brine (10mL), dried over anhydrous sodium sulfate, filtered and concentrated in vacuo to afford ethyl 5- (1, 3-dihydroxypropan-2-yl) isoxazole-3-carboxylate (0.94g, 4.37mmol, 78%) as a yellow oil. LCMS (ESI) M/z 216.1[ M + H ]]+

And 5: preparation of ethyl 5- (oxetan-3-yl) isoxazole-3-carboxylate

To a solution of ethyl 5- (1, 3-dihydroxypropan-2-yl) isoxazole-3-carboxylate (0.6g, 2.79mmol) in toluene (20mL) was added cyanomethylenetributylphosphine (1.0g, 4.18 mmol). The reaction mixture was stirred at 120 ℃ for 17 h. The reaction mixture was cooled to room temperature, filtered and concentrated in vacuo. Preparing the residue-TLC (silica, petroleum ether/ethyl acetate 1/1) to give ethyl 5- (oxetan-3-yl) isoxazole-3-carboxylate (0.120g, 0.61mmol, 22%) as a yellow oil. LCMS (ESI) M/z 198.1[ M + H ] ]+

Step 6: preparation of 5- (oxetan-3-yl) isoxazole-3-carboxylic acids

To a solution of ethyl 5- (oxetan-3-yl) isoxazole-3-carboxylate (0.120g, 0.61mmol) in tetrahydrofuran/water (v/v ═ 2/1, 12mL) was added lithium hydroxide hydrate (77mg, 1.83mmol) at 0 ℃. The reaction mixture was stirred at 25 ℃ for 2 h. The reaction mixture was quenched with ice-water and then adjusted to pH 3 with 1N aqueous hydrogen chloride. The aqueous layer was extracted with ethyl acetate (10 mL. times.2). The combined organic layers were washed with brine (10mL), dried over anhydrous sodium sulfate, filtered and concentrated in vacuo to afford 5- (oxetan-3-yl) isoxazole-3-carboxylic acid (0.085g, 0.5mmol, 83%) as a white solid. LCMS (ESI) M/z 170.1[ M + H ]]+

And 7: preparation of N- (1- (3-fluorobenzyl) -1H-pyrazol-4-yl) -5- (oxetan-3-yl) isoxazole-3-carboxamide

To a solution of 5- (oxetan-3-yl) isoxazole-3-carboxylic acid (80mg, 0.47mmol) in N, N-dimethylformamide (15mL) was added 1- (3-fluorobenzyl) -1H-pyrazol-4-amine (90mg, 0.47mmol), 1- [ bis (dimethylamino) methylene ] -methylene]-1H-1, 2, 3-triazolo [4, 5-b]Pyridinium 3-oxide hexafluorophosphate (536mg, 1.41mmol) and diisopropylethylamine (0.182g, 1.41 mmol). The mixture was stirred at 25 ℃ for 3 h. The crude sample was dissolved in a minimal amount of N, N-dimethylformamide and purified by preparative-HPLC (Boston C1821X 250mm 10 μm column; acetonitrile/0.01% aqueous trifluoroacetic acid) to give N- (1- (3-fluorobenzyl) -1H-pyrazol-4-yl as a white solid ) -5- (oxetan-3-yl) isoxazole-3-carboxamide (57.8mg, 0.17mmol, 36%).1H NMR (500MHz, dimethylsulfoxide-d)6)δ10.98(s,1H),8.20(s,1H),7.68(s,1H),7.39(dd,J=14.0,7.9Hz,1H),7.21-6.93(m,4H),5.95(s,1H),5.67(s,1H),5.36(s,2H),4.31(s,2H);LCMS(ESI)m/z:343.1[M+H]+

Example 43 preparation of N- {1- [ (3-fluorophenyl) methyl ] -1H-pyrazol-4-yl } -4,5,6, 7-tetrahydro-1, 2-benzoxazole-3-carboxamide (193)

Step 1: preparation of 1- [ (3-fluorophenyl) methyl ] -4-nitro-1H-pyrazole

To a solution of 4-nitro-1H-pyrazole (0.500g, 4.42mmol) and 1- (bromomethyl) -3-fluorobenzene (918mg, 4.86mmol) in N, N' -dimethylformamide (11mL) at 25 ℃ was added cesium carbonate (2.16g, 6.63mmol) and the reaction was stirred at 25 ℃ for 24H, then quenched by addition of water (10mL) and extracted with ethyl acetate (20mL × 3). The combined organic layers were dried over magnesium sulfate, filtered and concentrated in vacuo. The crude residue was purified by column chromatography (ISCO, 40g silica, 0-30% ethyl acetate in hexanes, 20min gradient) to afford 1- [ (3-fluorophenyl) methyl group as a yellow oil]-4-nitro-1H-pyrazole (0.98g, 4.40mmol, 99%).1H NMR (300MHz, chloroform-d) δ 8.12(d, J ═ 3.8Hz, 2H), 7.49-7.32(m, 1H), 7.17-6.89(m, 3H), 5.33(s, 2H); LCMS (ESI) M/z 222.1[ M + H ]]+

Step 2: preparation of 1- [ (3-fluorophenyl) methyl ] -1H-pyrazol-4-amine

To 1- [ (3-fluorophenyl) methyl group at 70 DEG C]To a solution of-4-nitro-1H-pyrazole (1.90g, 8.58mmol) and ammonium chloride (1.83g, 34.3mmol) in methanol (40mL) and water (8.2mL) was added iron (1.91g, 34.3mmol) in one portion. The reaction mixture was stirred at 70 ℃ for 16h, after which the reaction was cooled to room temperature and diluted with saturated aqueous sodium bicarbonate (40 mL). The reaction mixture was diluted with ethyl acetate (50mL) and passed throughThe pad is filtered. The filter pad was washed with ethyl acetate (20 mL. times.2). The filtrate was concentrated and the aqueous layer was extracted with ethyl acetate (50 mL. times.2). The combined organic layers were dried over magnesium sulfate, filtered and concentrated to give 1- [ (3-fluorophenyl) methyl group as a crude red solid]-1H-pyrazol-4-amine (1.40g, 7.32mmol, 85%). The crude material was used without further purification.1H NMR (300MHz, dimethylsulfoxide-d)6)δ7.42-7.22(m,1H),7.18-6.87(m,5H),5.15(s,2H);LCMS(ESI)m/z:192.2[M+H]+

And step 3: preparation of N- {1- [ (3-fluorophenyl) methyl ] -1H-pyrazol-4-yl } -4, 5, 6, 7-tetrahydro-1, 2-benzoxazole-3-carboxamide

To 1- [ (3-fluorophenyl) methyl group at 25 DEG C]-1H-pyrazol-4-amine (57.1mg, 0.299mmol), 4, 5, 6, 7-tetrahydro-1, 2-benzoxazole-3-carboxylic acid (50.0mg, 0.299mmol) and 1- [ bis (dimethylamino) methylene]-1H-1, 2, 3-triazolo [4, 5-b ]To a solution of pyridinium 3-oxide hexafluorophosphate (113mg, 0.299mmol) in N, N' -dimethylformamide (1mL) was added diisopropylethylamine (130. mu.L, 0.748 mmol). The reaction mixture was stirred at 25 ℃ for 16h, then quenched with water (1 mL). The aqueous layer was extracted with ethyl acetate (5 mL. times.3). The combined organic layers were washed with brine, dried over magnesium sulfate, filtered and concentrated in vacuo. The crude residue was purified by column chromatography (ISCO, 12g silica, eluting with 60% ethyl acetate/hexanes for 20min) to afford N- {1- [ (3-fluorophenyl) methyl ] as a yellow solid]-1H-pyrazoles-4-yl } -4, 5, 6, 7-tetrahydro-1, 2-benzoxazole-3-carboxamide (60.3mg, 0.177mmol, 60%).1H NMR (300MHz, dimethylsulfoxide-d)6)δ10.95(s,1H),8.18(d,J=0.7Hz,1H),7.66(d,J=0.7Hz,1H),7.39(td,J=8.0,6.1Hz,1H),7.21-6.94(m,3H),5.33(s,2H),2.74(q,J=5.9Hz,4H),1.81-1.56(m,4H);LCMS(ESI)m/z:341.2[M+H]+

EXAMPLE 44.preparation of 5-ethynyl-N- (1- (3-fluorobenzyl) -1H-pyrazol-4-yl) isoxazole-3-carboxamide (154)

Step 1: preparation of 5-ethynylisoxazole-3-carboxylic acid potassium

To an ice-cooled solution of ethyl 5-formylisoxazole-3-carboxylate (2.0g, 11.8mmol) in methanol (40mL) were added potassium carbonate (3.27g, 23.7mmol) and dimethyl 1-diazo-2-oxopropylphosphonate (2.5g, 13.0 mmol). The mixture was warmed to 23 ℃ and stirred for 2 h. Water (4mL) was added to the mixture and stirred for 2 h. The reaction mixture was concentrated and dried in vacuo to give potassium 5-ethynylisoxazole-3-carboxylate (4.0g, crude) as a yellow solid. LCMS (ESI) M/z 138.1[ M + H ] ]+. This material was used in the next step without further purification.

Step 2: preparation of 5-ethynyl-N- (1- (3-fluorobenzyl) -1H-pyrazol-4-yl) isoxazole-3-carboxamide

To a mixture of 5-ethynylisoxazole-3-carboxylic acid potassium ester (4.00g, 11.8mmol), 1- (3-fluorobenzyl) -1H-pyrazol-4-amine hydrochloride (2.69g, 11.8mmol) and 2- (7-aza-1H-benzotriazol-1-yl) -1, 1, 3, 3-tetramethyluronium hexafluorophosphate (4.5g,11.8mmol) in N, N-dimethylformamide (15mL) was added N, N-diisopropylethylamine (6.3mL, 35.5 mmol). The mixture was stirred at 23 ℃ for 2 h. The reaction mixture was diluted with ethyl acetate (300mL) and washed with hydrochloric acid (0.5N, 100mL), water (100mL) and brine (100 mL). The organic layer was dried over sodium sulfate, filtered and concentrated in vacuo. The residue was purified by column chromatography (silica, petroleum ether/ethyl acetate ═ 2/1) to give 5-ethynyl-N- (1- (3-fluorobenzyl) -1H-pyrazol-4-yl) isoxazole-3-carboxamide as a white yellow solid (313mg, 1.01mmol, 9%).1H NMR (500MHz, dimethylsulfoxide-d)6)δ11.08(s,1H),8.19(s,1H),7.68(s,1H),7.39(d,J=6.2Hz,1H),7.33(s,1H),7.13(d,J=1.9Hz,1H),7.05(t,J=9.4Hz,2H),5.37(d,J=14.7Hz,3H);LCMS(ESI)m/z:311.0[M+H]+

EXAMPLE 45. preparation of N- (1- (2-methoxybenzyl) -1H-pyrazol-4-yl) -5-phenylisoxazole-3-carboxamide (125)

Step 1: preparation of (2-methoxyphenyl) methanol

To a cooled solution of 2-methoxybenzaldehyde (1.55g, 11.3mmol) in methanol (8mL) at 0 deg.C was added dropwise a solution of sodium borohydride (0.854g, 22.6mmol) in water. The reaction mixture was stirred in the cooling bath for a further 1h, after which the cooling bath was removed. The reaction mixture was stirred at room temperature for 18 h. The reaction mixture was cooled to 0 ℃ and carefully adjusted to pH 3 with 1M aqueous hydrogen chloride. The mixture was stirred in the cooling bath for a further 30 min. The acidic mixture was neutralized to pH 6-7 with 1M aqueous sodium hydroxide solution and stirred for 1 h. The organics were concentrated and the residual aqueous layer was extracted with ethyl acetate (25 mL. times.5). The combined organics were dried over sodium sulfate, filtered and concentrated in vacuo. Subjecting the crude material to column chromatography(ISCO, 12g silica, eluting with 0% to 25% ethyl acetate in hexanes) to give (2-methoxyphenyl) methanol as a colorless oil (1.16g, 8.25mmol, 73%).1H NMR (300MHz, dimethylsulfoxide-d)6)δ7.36(ddt,J=7.5,1.8,0.9Hz,1H),7.27-7.15(m,1H),6.98-6.87(m,2H),4.96(t,J=5.7Hz,1H),4.48(d,J=5.6Hz,2H),3.76(s,3H)。

Step 2: preparation of 1- (chloromethyl) -2-methoxybenzene

To a solution of (2-methoxyphenyl) methanol (1.15g, 8.32mmol) and triethylamine (1.24g, 12.3mmol, 1.7mL) in dichloromethane (16mL) at 0 deg.C was added chlorotrimethylsilane (0.939g, 8.65mmol) dropwise. The reaction mixture was stirred in the cooling bath for a further 30min, after which the cooling bath was removed and stirred at room temperature for 18 h. The reaction mixture was washed with water (10mL × 2), dried over sodium sulfate, filtered and concentrated to give crude 1- (chloromethyl) -2-methoxybenzene (1.36g, crude) as an orange oil. This material was used directly in the next step without further purification.

And step 3: preparation of 1- (2-methoxybenzyl) -4-nitro-1H-pyrazole

To a suspension of 4-nitro-1H-pyrazole (1.19g, 10.6mmol) and freshly ground anhydrous potassium carbonate (4.40g, 31.9mmol) in anhydrous acetone (13mL) was added crude 1- (chloromethyl) -2-methoxybenzene (1.84g, 11.7 mmol). The reaction mixture was stirred at room temperature for 18h, after which it was heated at 50 ℃ for 30 min. The reaction mixture was filtered and the filtrate was concentrated in vacuo. The crude product was purified by column chromatography (ISCO, 24g silica, 0-25% ethyl acetate in hexanes) to give 1- (2-methoxybenzyl) -4-nitro-1H-pyrazole (0.97g, 4.15mmol, 39%).1H NMR (300MHz, dimethylsulfoxide-d)6)δ8.86(d,J=0.7Hz,1H),8.25(d,J=0.8Hz,1H),7.34(td,J=7.9,1.8Hz,1H),7.10(dd,J=7.5,1.8Hz,1H),7.04(d,J=8.0Hz,1H),6.93(td,J=7.4,1.1Hz,1H),5.35(s,2H),3.81(s,3H)。

And 4, step 4: preparation of 1- (2-methoxybenzyl) -1H-pyrazol-4-amine

To a solution of 1- (2-methoxybenzyl) -4-nitro-1H-pyrazole (0.97g, 4.15mmol) and ammonium chloride (1.10g, 20.7mmol) in methanol (8mL) and water (2mL) at 70 deg.C was added iron (1.15g, 20.7mmol) in one portion. The reaction mixture was heated to 70 ℃ for 16 h. The reaction mixture was cooled to room temperature and diluted with saturated aqueous sodium bicarbonate (40mL) and ethyl acetate (50 mL). Passing the mixture throughThe pad was filtered and the filter cake was washed with ethyl acetate (20 mL. times.3). The aqueous layer was extracted with ethyl acetate (50 mL. times.2). The combined organic layers were dried over magnesium sulfate, filtered and concentrated to give 1- (2-methoxybenzyl) -1H-pyrazol-4-amine (0.595g, 2.92mmol, 70%) as a crude red oil. This material was used directly in the next step without further purification. 1H NMR (300MHz, dimethylsulfoxide-d)6)δ7.35-7.17(m,1H),7.01(td,J=4.0,1.0Hz,2H),6.92(d,J=0.9Hz,1H),6.87(td,J=7.4,1.1Hz,1H),6.77(dd,J=7.4,1.8Hz,1H),5.08(s,2H),3.81(s,3H)。

And 5: preparation of N- (1- (2-methoxybenzyl) -1H-pyrazol-4-yl) -5-phenylisoxazole-3-carboxamide

To 5-phenylisoxazole-3-carboxylic acid (0.075, 0.396mmol), 1- (2-methoxybenzyl) -1H-pyrazol-4-amine (0.088g, 0.435mmol) and 1- [ bis (dimethyl)Amino) methylene]-1H-1, 2, 3-triazolo [4, 5-b]To a solution of pyridinium 3-oxide hexafluorophosphate (0.190g, 0.593mmol) in N, N-dimethylformamide (1mL) was added diisopropylethylamine (0.152g, 1.18 mmol). The reaction mixture was stirred at room temperature for 12h, then concentrated in vacuo. The residue was dissolved in ethyl acetate (5mL), washed with saturated sodium bicarbonate (2mL × 2), and concentrated in vacuo. The crude product was purified by column chromatography (ISCO, 24g silica, 0-40% ethyl acetate in hexanes) to give N- (1- (2-methoxybenzyl) -1H-pyrazol-4-yl) -5-phenylisoxazole-3-carboxamide as an off-white solid (29.8mg, 0.0792mmol, 20%).1H NMR (300MHz, dimethylsulfoxide-d)6)δ11.00(s,1H),8.06(d,J=0.7Hz,1H),8.01-7.92(m,2H),7.67(d,J=0.7Hz,1H),7.62-7.53(m,3H),7.45(s,1H),7.31(ddd,J=8.2,5.1,4.0Hz,1H),7.05(d,J=8.2Hz,1H),6.91(dd,J=4.3,0.8Hz,2H),5.27(s,2H),3.84(s,3H);LCMS(ESI)m/z:375.3[M+H]+

Example 46 preparation of N- (1-benzylpyrazol-4-yl) -5-thiazol-5-yl-isoxazole-3-carboxamide (37)

Step 1: preparation of 5-thiazol-5-ylisoxazole-3-carboxylic acid ethyl ester

A mixture of ethyl 5-tributylstannyl isoxazole-3-carboxylate (1.00g, 2.32mmol), 5-bromothiazole (0.763g, 4.65mmol) and bis (tri-tert-butylphosphine) palladium (0) (0.059g, 0.116mmol) in toluene (10mL) was purged with nitrogen (3X) and the mixture was then heated at 100 ℃ under nitrogen for 12 h. The mixture was cooled to 15 ℃ and then poured into ice-water (10 mL). The aqueous phase was extracted with ethyl acetate (15 mL. times.3). The combined organic phases were washed with brine (10mL), dried over anhydrous sodium sulfate, filtered and concentrated in vacuo to give the crude product And (5) preparing a product. The crude product was purified by column chromatography (silica, petroleum ether/ethyl acetate 100/1 to 5/1) to give ethyl 5-thiazol-5-ylisoxazole-3-carboxylate (0.060mg, 0.268mmol, 12%) as a yellow solid. LCMS (ESI) M/z 225.0[ M + H ]]+

Step 2: preparation of 5-thiazol-5-yl isoxazole-3-carboxylic acids

To a stirred solution of ethyl 5-thiazol-5-ylisoxazole-3-carboxylate (0.06g, 0.268mmol) in tetrahydrofuran (1mL) was added lithium hydroxide (2M, 0.268 mL). The mixture was stirred at 15 ℃ for 1 h. The mixture was diluted with water (1mL) and adjusted to pH 5 with hydrogen chloride solution (2M, 1 mL). The aqueous phase was extracted with ethyl acetate (5 mL. times.3). The combined organic phases were washed with brine (5mL), dried over anhydrous sodium sulfate, filtered and concentrated in vacuo to give crude 5-thiazol-5-ylisoxazole-3-carboxylic acid (0.05g, 0.255mmol) as a yellow solid which was used without further purification.

And step 3: preparation of N- (1-benzylpyrazol-4-yl) -5-thiazol-5-yl-isoxazole-3-carboxamide

To a stirred solution of 5-thiazol-5-ylisoxazole-3-carboxylic acid (0.05g, 0.255mmol) in dichloromethane (0.5mL) was added propylphosphonic anhydride (0.227mL, 0.382mmol, 50% wt), triethylamine (0.071mL, 0.510mmol) and 1-benzylpyrazol-4-amine (0.044g, 0.255 mmol). The mixture was stirred at 15 ℃ for 1 h. The reaction mixture was concentrated under reduced pressure to give a residue. The crude residue was purified by preparative-HPLC (YMC-Actus Triart C18150X 305 um column; 24-54% acetonitrile in 10mM aqueous ammonium acetate, 10min gradient) to give N- (1-benzylpyrazol-4-yl) -5-thiazol-5-yl-isoxazole-3-carboxamide (0.024g, 0.068mmol, 27%) as a pale yellow solid. 1H NMR (400MHz, dimethylsulfoxide-d 6) delta 11.05(s, 1H), 9.36(s,1H),8.64(s,1H),8.17(s,1H),7.67(s,1H),7.43(s,1H),7.38-7.21(m,5H),5.33(s,2H);LCMS(ESI)m/z:352.1[M+H]+

example 47.5- (2-fluorophenyl) -N- (1- ((4- (trifluoromethyl) pyridin-3-yl) methyl) -1H-pyrazol-4-yl) isoxazole-3-carboxamide (114) preparation

Step 1: preparation of 4- (trifluoromethyl) nicotinic acid

To a solution of 4- (trifluoromethyl) nicotinonitrile (5.0g, 29.0mmol) in water (30mL) was slowly added concentrated sulfuric acid (30 mL). The reaction mixture was heated to 110 ℃ and stirred for 12 h. The mixture was cooled and then an aqueous sodium carbonate solution was added to adjust the pH to 3-5. The aqueous phase was extracted with dichloromethane (200 mL. times.2). The combined organic layers were dried over sodium sulfate, filtered and concentrated to give 4- (trifluoromethyl) nicotinic acid as a white solid (2.0g, crude, 36%). The material was used in the procedure without additional purification.

Step 2: preparation of (4- (trifluoromethyl) pyridin-3-yl) methanol

To a solution of 4- (trifluoromethyl) nicotinic acid (2.0g, 10.5mmol) in tetrahydrofuran (20mL) was added borane-tetrahydrofuran (31mL, 31.4mmol, 1M). The reaction mixture was stirred at 20 ℃ for 12h and then quenched with 1N aqueous hydrogen chloride solution. The crude mixture was heated to 90 ℃ and stirred for 1 h. The solid is filtered and an aqueous sodium hydroxide solution is added to adjust the pH to 8-10. The aqueous phase was extracted with dichloromethane (100mL × 2), dried over magnesium sulfate, filtered and concentrated under reduced pressure to give 4- (trifluoromethyl) pyridin-3-yl) methanol as a yellow solid (0.87g, 47%). L is CMS(ESI)m/z:178.1[M+H]+

And step 3: preparation of 3- ((4-nitro-1H-pyrazol-1-yl) methyl) -4- (trifluoromethyl) pyridine

A mixture of (4- (trifluoromethyl) pyridin-3-yl) methanol (0.72g, 4.07mmol), 4-nitro-1H-pyrazole (0.46g, 4.07mmol), triphenylphosphine (4.3g, 16.3mmol) and tetrahydrofuran (30mL) was stirred at 20 deg.C for 20 min. Dipropyl azodicarboxylate (3.3g, 16.3mmol) was slowly added to the reaction, which was then stirred at 20 ℃ for 2 h. The reaction mixture was diluted with ethyl acetate (100mL) and washed with water (100mL), brine (100mL), dried over sodium sulfate, filtered and concentrated in vacuo. The crude residue was purified by column chromatography (silica, petroleum ether/ethyl acetate 5/1 to 2/1) to afford 3- ((4-nitro-1H-pyrazol-1-yl) methyl) -4- (trifluoromethyl) pyridine (0.700g, 2.6mmol, 64%) as a yellow oil. LCMS (ESI) M/z 273.1[ M + H ]]+

And 4, step 4: preparation of 1- ((4- (trifluoromethyl) pyridin-3-yl) methyl) -1H-pyrazol-4-amine

To a solution of 3- ((4-nitro-1H-pyrazol-1-yl) methyl) -4- (trifluoromethyl) pyridine (0.300g, 1.10mmol) in methanol (10mL) under nitrogen was added palladium on charcoal (10 wt%, 0.030 g). The reaction mixture was purged with hydrogen and stirred at 20 ℃ for 30 min. The reaction mixture was filtered and the filtrate was concentrated in vacuo to give 1- ((4- (trifluoromethyl) pyridin-3-yl) methyl) -1H-pyrazol-4-amine (0.228g, 9.35mmol, 85%) as a red oil (which was used in the next step without further purification); LCMS (ESI) M/z 243.2[ M + H ] ]+

And 5: preparation of ethyl 5- (2-fluorophenyl) isoxazole-3-carboxylate

A mixture of ethyl (Z) -2-chloro-2- (hydroxyimino) acetate (2.0g, 13.2mmol) in N, N-dimethylformamide (5mL) was slowly added under nitrogen to a solution of 1-ethynyl-2-fluorobenzene (3.97g, 33.1mmol) in N, N-dimethylformamide (10 mL). The mixture was heated to 90 ℃ and then a solution of triethylamine (4.02g, 39.7mmol) in N, N-dimethylformamide (5mL) was added slowly. The reaction mixture was heated at 90 ℃ for 16h and then concentrated under reduced pressure. The residue was purified by column chromatography (silica, petroleum ether/ethyl acetate-2/1) to give ethyl 5- (2-fluorophenyl) isoxazole-3-carboxylate (2.0g, 8.45mmol, 64%) as a red solid. (LCMS (ESI) M/z:236.1[ M + H ]]+

Step 6: preparation of 5- (2-fluorophenyl) isoxazole-3-carboxylic acid

To a solution of ethyl 5- (2-fluorophenyl) isoxazole-3-carboxylate (0.500g, 2.13mmol) in tetrahydrofuran (5mL) was slowly added lithium hydroxide hydrate (0.0894g, 2.13mmol) in water (4mL) at 0 ℃. The reaction was then stirred at 10 ℃ for 20min, then 1N aqueous hydrogen chloride was added to adjust the pH to 2-3. The volatiles were removed under reduced pressure to give crude 5- (2-fluorophenyl) isoxazole-3-carboxylic acid (0.280g, 1.36mmol, 64%) as a yellow solid (which was used in the next step without further purification); LCMS (ESI) M/z 208.1[ M + H ] ]+

And 7: preparation of 5- (2-fluorophenyl) -N- (1- ((4- (trifluoromethyl) pyridin-3-yl) methyl) -1H-pyrazol-4-yl) isoxazole-3-carboxamide

To a solution of 5- (2-fluorophenyl) isoxazole-3-carboxylic acid (0.150g, 0.724mmol) in dichloromethane (2mL) at 20 deg.CTo which oxalyl chloride (2mL) was added. The mixture was stirred at room temperature for 0.5h, then the solvent was removed under reduced pressure. The resulting solid was dissolved in dichloromethane (2mL) and added dropwise to a mixture of 1- ((4- (trifluoromethyl) pyridin-3-yl) methyl) -1H-pyrazol-4-amine (0.228g, 0.942mmol) and triethylamine (220mg, 2.172mmol) in dichloromethane (5 mL). The reaction mixture was stirred for a further 0.5H and then purified by preparative-TLC (silica, petroleum ether/ethyl acetate ═ 1/1) to give 5- (2-fluorophenyl) -N- (1- ((4- (trifluoromethyl) pyridin-3-yl) methyl) -1H-pyrazol-4-yl) isoxazole-3-carboxamide (0.0630g, 0.145mmol, 20%) as a white solid. (1H NMR (400MHz, dimethylsulfoxide-d)6)δ11.10(s,1H),8.81(s,1H),8.29-8.35(m,2H),8.02-8.04(m,1H),7.75-7.80(m,2H),7.64-7.65(m,1H),7.42-7.52(m,2H),7.27(s,1H),5.61(s,2H);LCMS(ESI)m/z:432.0[M+H]+

Example 48 preparation of N- (1- { [4- (trifluoromethyl) pyridin-3-yl ] methyl } -1H-pyrazol-4-yl) -4,5,6, 7-tetrahydro-1, 2-benzoxazole-3-carboxamide (194)

Step 1: preparation of 3- ((4-nitro-1H-pyrazol-1-yl) methyl) -4- (trifluoromethyl) pyridine

To [4- (trifluoromethyl) pyridin-3-yl ] at 0 deg.C]To a solution of methanol (0.870g, 4.91mmol), 4-nitro-1H-pyrazole (0.555g, 4.91mmol) and triphenylphosphine (1.93g, 7.36mmol) in tetrahydrofuran (12.2mL) was added dipropyl azodicarboxylate (1.4mL, 7.36 mmol). The reaction mixture was warmed to room temperature over 16 h. The volatiles were concentrated under reduced pressure. The crude residue was purified by column chromatography (ISCO, 24g silica, eluting with 70% ethyl acetate/hexanes for 20min) to give 3- ((4-nitro-1H-pyrazol-1-yl) methyl) -4- (trifluoromethyl) pyridine (0.90g, 3.3mmol, 67%) as a yellow solid.1H NMR (300MHz, chloroform-d) δ 8.86(d, J ═ 5.1Hz, 1H), 8.71(s, 1H), 8.23-8.11(m, 2H), 7.64(d, J ═ 5.1Hz, 1H), 5.56(s, 2H), 4.98(H, J ═ 6.3Hz, 1H), 1.28(d, J ═ 6.2Hz, 4H); LCMS (ESI) M/z 273.1[ M + H ]]+

Step 2: preparation of 1- { [4- (trifluoromethyl) pyridin-3-yl ] methyl } -1H-pyrazol-4-amine

To 3- [ (4-nitro-1H-pyrazol-1-yl) methyl at 70 ℃]To a solution of-4- (trifluoromethyl) pyridine (1.80g, 6.61mmol) and ammonium chloride (1.41g, 26.4mmol) in methanol (26mL) and water (6.6mL) was added iron (1.47g, 26.4mmol) in one portion. The reaction mixture was heated to 70 ℃ for 16 h. The reaction mixture was cooled to room temperature and diluted with saturated aqueous sodium bicarbonate (40mL) and ethyl acetate (50 mL). Passing the mixture through The pad was filtered and the filter cake was washed with ethyl acetate (20 mL. times.3). The aqueous layer was extracted with ethyl acetate (50 mL. times.2). The combined organic layers were dried over magnesium sulfate, filtered and concentrated to give 1- { [4- (trifluoromethyl) pyridin-3-yl as a crude red solid]Methyl } -1H-pyrazol-4-amine (1.50g, 6.19mmol, 94%). This material was used directly in the next step without further purification.1H NMR (300MHz, dimethylsulfoxide-d)6)δ8.91(s,1H),8.78(d,J=5.0Hz,1H),8.16(s,1H),7.78(d,J=5.1Hz,1H),7.20(s,1H),7.03(d,J=3.7Hz,1H),5.41(s,2H),4.03(d,J=19.7Hz,2H)。

And step 3: preparation of N- (1- { [4- (trifluoromethyl) pyridin-3-yl ] methyl } -1H-pyrazol-4-yl) -4, 5, 6, 7-tetrahydro-1, 2-benzoxazole-3-carboxamide

To 1- { [4- (trifluoromethyl) at 25 deg.C) Pyridin-3-yl]Methyl } -1H-pyrazol-4-amine (72.4mg, 0.299mmol), 4, 5, 6, 7-tetrahydro-1, 2-benzoxazole-3-carboxylic acid (50.0mg, 0.299mmol) and 1- [ bis (dimethylamino) methylene]-1H-1, 2, 3-triazolo [4, 5-b]To a solution of pyridinium 3-oxide hexafluorophosphate (113mg, 0.299mmol) in N, N' -dimethylformamide (1mL) was added diisopropylethylamine (130. mu.L, 0.748 mmol). The reaction mixture was stirred at 25 ℃ for 16 h. The reaction mixture was quenched with water (1 mL). The aqueous layer was extracted with ethyl acetate (5 mL. times.3). The combined organic layers were washed with brine, dried over magnesium sulfate, filtered and concentrated in vacuo. The crude residue was purified by column chromatography (ISCO, 12g silica, eluting with 60% ethyl acetate/hexanes for 20min) to give N- (1- { [4- (trifluoromethyl) pyridin-3-yl) as a yellow solid ]Methyl } -1H-pyrazol-4-yl) -4, 5, 6, 7-tetrahydro-1, 2-benzoxazole-3-carboxamide (55.4mg, 0.142mmol, 47%).1H NMR (300MHz, dimethylsulfoxide-d)6)δ11.00(s,1H),8.84-8.73(m,1H),8.34(s,1H),8.24(s,1H),7.79(d,J=5.1Hz,1H),7.71(d,J=0.7Hz,1H),5.58(s,2H),2.75(q,J=6.1Hz,4H),1.72(dd,J=12.7,7.6Hz,4H);LCMS(ESI)m/z:392.3[M+H]+

EXAMPLE 49 preparation of 5- (pyridin-2-yl) -N- (1- { [4- (trifluoromethyl) pyridin-3-yl ] methyl } -1H-pyrazol-4-yl) -1, 2-oxazole-3-carboxamide (197)

Step 1: preparation of 3- ((4-nitro-1H-pyrazol-1-yl) methyl) -4- (trifluoromethyl) pyridine

To [4- (trifluoromethyl) pyridin-3-yl ] at 0 deg.C]To a solution of methanol (0.870g, 4.91mmol), 4-nitro-1H-pyrazole (0.555g, 4.91mmol) and triphenylphosphine (1.93g, 7.36mmol) in tetrahydrofuran (12.2mL) was added dipropyl azodicarboxylate (1.4mL, 7.36 mmol). After 16h, the mixture is cooledThe reaction mixture was warmed to room temperature. The volatiles were concentrated under reduced pressure. The crude residue was purified by column chromatography (ISCO, 24g silica, eluting with 70% ethyl acetate/hexanes for 20min) to give 3- ((4-nitro-1H-pyrazol-1-yl) methyl) -4- (trifluoromethyl) pyridine (0.90g, 3.3mmol, 67%) as a yellow solid.1H NMR (300MHz, chloroform-d) δ 8.86(d, J ═ 5.1Hz, 1H), 8.71(s, 1H), 8.23-8.11(m, 2H), 7.64(d, J ═ 5.1Hz, 1H), 5.56(s, 2H), 4.98(H, J ═ 6.3Hz, 1H), 1.28(d, J ═ 6.2Hz, 4H); LCMS (ESI) M/z 273.1[ M + H ] ]+

Step 2: preparation of 1- { [4- (trifluoromethyl) pyridin-3-yl ] methyl } -1H-pyrazol-4-amine

To 3- [ (4-nitro-1H-pyrazol-1-yl) methyl at 70 ℃]To a solution of-4- (trifluoromethyl) pyridine (1.80g, 6.61mmol) and ammonium chloride (1.41g, 26.4mmol) in methanol (26mL) and water (6.6mL) was added iron (1.47g, 26.4mmol) in one portion. The reaction mixture was heated to 70 ℃ for 16 h. The reaction mixture was cooled to room temperature and diluted with saturated aqueous sodium bicarbonate (40mL) and ethyl acetate (50 mL). Passing the mixture throughThe pad was filtered and the filter cake was washed with ethyl acetate (20 mL. times.3). The aqueous layer was extracted with ethyl acetate (50 mL. times.2). The combined organic layers were dried over magnesium sulfate, filtered and concentrated to give 1- { [4- (trifluoromethyl) pyridin-3-yl as a crude red solid]Methyl } -1H-pyrazol-4-amine (1.50g, 6.19mmol, 94%). This material was used directly in the next step without further purification.1H NMR (300MHz, dimethylsulfoxide-d)6)δ8.91(s,1H),8.78(d,J=5.0Hz,1H),8.16(s,1H),7.78(d,J=5.1Hz,1H),7.20(s,1H),7.03(d,J=3.7Hz,1H),5.41(s,2H),4.03(d,J=19.7Hz,2H)。

And step 3: preparation of 5- (pyridin-2-yl) -N- (1- { [4- (trifluoromethyl) pyridin-3-yl ] methyl } -1H-pyrazol-4-yl) -1, 2-oxazole-3-carboxamide

To 5- (pyridin-2-yl) -1, 2-oxazole-3-carboxylic acid (50mg, 0.263mmol), 1- { [4- (trifluoromethyl) pyridin-3-yl ]Methyl } -1H-pyrazol-4-amine (63.6mg, 0.263mmol) and 1- [ bis (dimethylamino) methylene]-1H-1, 2, 3-triazolo [4, 5-b]To a solution of pyridinium 3-oxide hexafluorophosphate (99.9mg, 0.263mmol) in N, N' -dimethylformamide (1.1mL) was added diisopropylethylamine (91.5. mu.L, 0.526mmol) dropwise. The reaction mixture was stirred at 25 ℃ for 16 h. The reaction mixture was quenched with water (1 mL). The aqueous layer was extracted with ethyl acetate (5 mL. times.3). The combined organic layers were washed with brine, dried over magnesium sulfate, filtered and concentrated in vacuo. The crude residue was purified by column chromatography (ISCO, 12g silica, eluting with 0-80% ethyl acetate/hexanes for 20min) to give 5- (pyridin-2-yl) -N- (1- { [4- (trifluoromethyl) pyridin-3-yl) as a pale yellow solid]Methyl } -1H-pyrazol-4-yl) -1, 2-oxazole-3-carboxamide (53.0mg, 0.128mmol, 49%).1H NMR (300MHz, chloroform-d) δ 8.85-8.73(m, 2H), 8.60(s, 1H), 8.40(s, 1H), 8.18(s, 1H), 7.91(dtd, J ═ 17.1, 8.0, 1.5Hz, 2H), 7.69(s, 1H), 7.58(d, J ═ 5.1Hz, 1H), 7.50-7.36(m, 2H), 5.57(s, 2H); LCMS (ESI) M/z 415.3[ M + H ]]+

EXAMPLE 50.preparation of 5- (furan-2-yl) -N- (1- { [4- (trifluoromethyl) pyridin-3-yl ] methyl } -1H-pyrazol-4-yl) -1, 2-oxazole-3-carboxamide (198)

Step 1: preparation of 3- ((4-nitro-1H-pyrazol-1-yl) methyl) -4- (trifluoromethyl) pyridine

To [4- (trifluoromethyl) pyridin-3-yl ] at 0 deg.C]To a solution of methanol (0.870g, 4.91mmol), 4-nitro-1H-pyrazole (0.555g, 4.91mmol) and triphenylphosphine (1.93g, 7.36mmol) in tetrahydrofuran (12.2mL) was added dipropyl azodicarboxylate (1.4mL, 7.36 mmol). The reaction mixture was warmed to room temperature over 16 h. The volatiles were concentrated under reduced pressure. The crude residue was purified by column chromatography (ISCO, 24g silica, eluting with 70% ethyl acetate/hexanes for 20min) to give 3- ((4-nitro-1H-pyrazol-1-yl) methyl) -4- (trifluoromethyl) pyridine (0.90g, 3.3mmol, 67%) as a yellow solid.1H NMR (300MHz, chloroform-d) δ 8.86(d, J ═ 5.1Hz, 1H), 8.71(s, 1H), 8.23-8.11(m, 2H), 7.64(d, J ═ 5.1Hz, 1H), 5.56(s, 2H), 4.98(H, J ═ 6.3Hz, 1H), 1.28(d, J ═ 6.2Hz, 4H); LCMS (ESI) M/z 273.1[ M + H ]]+

Step 2: preparation of 1- { [4- (trifluoromethyl) pyridin-3-yl ] methyl } -1H-pyrazol-4-amine

To 3- [ (4-nitro-1H-pyrazol-1-yl) methyl at 70 ℃]To a solution of-4- (trifluoromethyl) pyridine (1.80g, 6.61mmol) and ammonium chloride (1.41g, 26.4mmol) in methanol (26.0mL) and water (6.61mL) was added iron (1.47g, 26.4mmol) in one portion. The reaction mixture was heated to 70 ℃ for 16 h. The reaction mixture was cooled to room temperature and diluted with saturated aqueous sodium bicarbonate (40mL) and ethyl acetate (50 mL). Passing the mixture through The pad was filtered and the filter cake was washed with ethyl acetate (20 mL. times.3). The aqueous layer was extracted with ethyl acetate (50 mL. times.2). The combined organic layers were dried over magnesium sulfate, filtered and concentrated to give 1- { [4- (trifluoromethyl) pyridin-3-yl as a red crude solid]Methyl } -1H-pyrazol-4-amine (1.50g, 6.19mmol, 94%). This material was used directly in the next step without further purification.1H NMR (300MHz, dimethylsulfoxide-d)6)δ8.91(s,1H),8.78(d,J=5.0Hz,1H),8.16(s,1H),7.78(d,J=5.1Hz,1H),7.20(s,1H),7.03(d,J=3.7Hz,1H),5.41(s,2H),4.03(d,J=19.7Hz,2H)。

And step 3: preparation of 5- (furan-2-yl) -N- (1- { [4- (trifluoromethyl) pyridin-3-yl ] methyl } -1H-pyrazol-4-yl) -1, 2-oxazole-3-carboxamide

To 5- (furan-2-yl) -1, 2-oxazole-3-carboxylic acid (0.05g, 0.279mmol), 1- { [4- (trifluoromethyl) pyridin-3-yl at 25 deg.C]Methyl } -1H-pyrazol-4-amine (67.5mg, 0.279mmol) and 1- [ bis (dimethylamino) methylene]-1H-1, 2, 3-triazolo [4, 5-b]To a solution of pyridinium 3-oxide hexafluorophosphate (106mg, 0.279mmol) in N, N' -dimethylformamide (1.1mL) was added diisopropylethylamine (97.1. mu.L, 0.558mmol) dropwise. The reaction mixture was stirred at 25 ℃ for 16 h. The reaction mixture was quenched with water (1 mL). The aqueous layer was extracted with ethyl acetate (5 mL. times.3). The combined organic layers were washed with brine, dried over magnesium sulfate, filtered and concentrated in vacuo. The crude residue was purified by column chromatography (ISCO, 12g silica, eluting with 0-80% ethyl acetate/hexanes for 20min) to give 5- (furan-2-yl) -N- (1- { [4- (trifluoromethyl) pyridin-3-yl) as a yellow solid ]Methyl } -1H-pyrazol-4-yl) -1, 2-oxazole-3-carboxamide (48.5mg, 0.120mmol, 50%).1H NMR (300MHz, chloroform-d) δ 8.77(d, J ═ 5.1Hz, 1H), 8.55(s, 1H), 8.40(s, 1H), 8.15(s, 1H), 7.68(d, J ═ 0.7Hz, 1H), 7.63-7.46(m, 2H), 7.01(dd, J ═ 3.5, 0.7Hz, 1H), 6.93(s, 1H), 6.59(dd, J ═ 3.5, 1.8Hz, 1H), 5.56(s, 2H); LCMS (ESI) M/z 404.2[ M + H ]]+

EXAMPLE 51.preparation of 5- (2H-1, 3-benzodioxol-5-yl) -N- (1- { [4- (trifluoromethyl) pyridin-3-yl ] methyl } -1H-pyrazol-4-yl) -1, 2-oxazole-3-carboxamide (199)

Step 1: preparation of 3- ((4-nitro-1H-pyrazol-1-yl) methyl) -4- (trifluoromethyl) pyridine

To [4- (trifluoromethyl) pyridin-3-yl ] at 0 deg.C]To a solution of methanol (0.870g, 4.91mmol), 4-nitro-1H-pyrazole (0.555g, 4.91mmol) and triphenylphosphine (1.93g, 7.36mmol) in tetrahydrofuran (12.2mL) was added dipropyl azodicarboxylate (1.4mL, 7.36 mmol). The reaction mixture was warmed to room temperature over 16 h. The volatiles were concentrated under reduced pressure. The crude residue was purified by column chromatography (ISCO, 24g silica, eluting with 70% ethyl acetate/hexanes for 20min) to give 3- ((4-nitro-1H-pyrazol-1-yl) methyl) -4- (trifluoromethyl) pyridine (0.90g, 3.3mmol, 67%) as a yellow solid. 1H NMR (300MHz, chloroform-d) δ 8.86(d, J ═ 5.1Hz, 1H), 8.71(s, 1H), 8.23-8.11(m, 2H), 7.64(d, J ═ 5.1Hz, 1H), 5.56(s, 2H), 4.98(H, J ═ 6.3Hz, 1H), 1.28(d, J ═ 6.2Hz, 4H); LCMS (ESI) M/z 273.1[ M + H ]]+

Step 2: preparation of 1- { [4- (trifluoromethyl) pyridin-3-yl ] methyl } -1H-pyrazol-4-amine

To 3- [ (4-nitro-1H-pyrazol-1-yl) methyl at 70 ℃]To a hot solution of-4- (trifluoromethyl) pyridine (1.80g, 6.61mmol) and ammonium chloride (1.41g, 26.4mmol) in methanol (26.0mL) and water (6.61mL) was added iron (1.47g, 26.4mmol) in one portion. The reaction mixture was heated to 70 ℃ for 16 h. The reaction mixture was cooled to room temperature and diluted with saturated aqueous sodium bicarbonate (40mL) and ethyl acetate (50 mL). Passing the mixture throughThe pad was filtered and the filter cake was washed with ethyl acetate (20 mL. times.3). The aqueous layer was extracted with ethyl acetate (50 mL. times.2). The combined organic layers were dried over magnesium sulfate, filtered and concentrated,1- { [4- (trifluoromethyl) pyridin-3-yl ] obtained as a red crude solid]Methyl } -1H-pyrazol-4-amine (1.50g, 6.19mmol, 94%). This material was used directly in the next step without further purification.1H NMR (300MHz, dimethylsulfoxide-d)6)δ8.91(s,1H),8.78(d,J=5.0Hz,1H),8.16(s,1H),7.78(d,J=5.1Hz,1H),7.20(s,1H),7.03(d,J=3.7Hz,1H),5.41(s,2H),4.03(d,J=19.7Hz,2H)。

And step 3: preparation of 5- (2H-1, 3-benzodioxol-5-yl) -N- (1- { [4- (trifluoromethyl) pyridin-3-yl ] methyl } -1H-pyrazol-4-yl) -1, 2-oxazole-3-carboxamide

To 5- (2H-1, 3-benzodioxol-5-yl) -1, 2-oxazole-3-carboxylic acid (0.05g, 0.214mmol), 1- { [4- (trifluoromethyl) pyridin-3-yl at 25 deg.C]Methyl } -1H-pyrazol-4-amine (51.9mg, 0.214mmol) and 1- [ bis (dimethylamino) methylene]-1H-1, 2, 3-triazolo [4, 5-b]To a solution of pyridinium 3-oxide hexafluorophosphate (81.5mg, 0.214mmol) in N, N' -dimethylformamide (0.9mL) was added diisopropylethylamine (74.6. mu.L, 0.429mmol) dropwise. The reaction mixture was stirred at 25 ℃ for 16 h. The reaction mixture was quenched with water (1 mL). The aqueous layer was extracted with ethyl acetate (5 mL. times.3). The combined organic layers were washed with brine, dried over magnesium sulfate, filtered and concentrated in vacuo. The crude residue was purified by column chromatography (ISCO, 12g silica, eluting with 0-80% ethyl acetate/hexanes for 20min) to give 5- (2H-1, 3-benzodioxol-5-yl) -N- (1- { [4- (trifluoromethyl) pyridin-3-yl) as a white solid]Methyl } -1H-pyrazol-4-yl) -1, 2-oxazole-3-carboxamide (48.5mg, 0.106mmol, 55%).1H NMR (300MHz, chloroform-d) δ 8.77(d, J ═ 5.1Hz, 1H), 8.56(s, 1H), 8.40(s, 1H), 8.15(s, 1H), 7.68(d, J ═ 0.7Hz, 1H), 7.58(d, J ═ 5.1Hz, 1H), 7.37(dd, J ═ 8.1, 1.7Hz, 1H), 7.28(s, 1H), 6.99 to 6.85(m, 2H), 6.08(s, 2H), 5.56(s, 2H). LCMS (ESI) M/z 458.3[ M + H ] ]+

Example 52.preparation of 5- (pyrimidin-4-yl) -N- (1- ((4- (trifluoromethyl) pyridin-3-yl) methyl) -1H-pyrazol-4-yl) isoxazole-3-carboxamide (200)

Step 1: preparation of (4- (trifluoromethyl) pyridin-3-yl) methanol

To a solution of 3-bromo-4- (trifluoromethyl) pyridine (2.26g, 10.0mmol) in toluene (50mL) under nitrogen at-78 deg.C was added n-butyllithium (4.8mL, 12 mmol). The mixture was stirred at-78 ℃ for 30min, then N, N-dimethylformamide (1.1g, 15mmol) was added. The reaction mixture was stirred at-78 ℃ for 10min, then sodium borohydride (0.756g, 20mmol) and methanol (5mL) were added sequentially at-78 ℃ and the reaction was warmed to 25 ℃ and stirred for 1 h. Upon completion, aqueous ammonium chloride solution was added to quench the reaction and the mixture was diluted with ethyl acetate (80mL × 3). The organic layer was dried over sodium sulfate, filtered and concentrated in vacuo. The crude residue was purified by column chromatography (silica, petroleum ether/ethyl acetate 2/1 to 1/1) to give (4- (trifluoromethyl) pyridin-3-yl) methanol (1.23g, 6.95mmol, 69%) as a colourless oil. LCMS (ESI) M/z 178.1[ M + H ]]+

Step 2: preparation of ethyl (4- (trifluoromethyl) pyridin-3-yl) methanesulfonate

To a solution of ethyl (4- (trifluoromethyl) pyridin-3-yl) methanol (1.23g, 6.9mmol) and triethylamine (2.1g, 20.8mmol) in tetrahydrofuran (50mL) at 0 deg.C under nitrogen was added methanesulfonyl chloride (1.59g, 13.9 mmol). The mixture was stirred at 0 ℃ for 30 min. The reaction mixture was used directly in the next step. LCMS (ESI) M/z 256.0[ M + H ] ]+

And step 3: preparation of 3- ((4-nitro-1H-pyrazol-1-yl) methyl) -4- (trifluoromethyl) pyridine

To a solution of 4-nitro-1H-pyrazole (1.57g, 13.9mmol) and cesium carbonate (6.78g, 20.8mmol) in acetonitrile (50mL) was added ethyl (4- (trifluoromethyl) pyridin-3-yl) methyl methanesulfonate (crude solution in tetrahydrofuran, 6.9mmol) under nitrogen at 25 ℃. The mixture was heated at 80 ℃ for 1 h. Upon completion, ethyl acetate (100mL) was added to dilute the mixture, and then the mixture was washed with water (50mL × 2). The organic layer was dried over sodium sulfate, filtered and concentrated in vacuo. The crude residue was purified by column chromatography (silica, petroleum ether/ethyl acetate 3/1) to give 3- ((4-nitro-1H-pyrazol-1-yl) methyl) -4- (trifluoromethyl) pyridine (0.870g, 3.2mmol, 46%) as a white solid. LCMS (ESI) M/z 273.1[ M + H ]]+

And 4, step 4: preparation of 1- ((4- (trifluoromethyl) pyridin-3-yl) methyl) -1H-pyrazol-4-amine

To a solution of 3- ((4-nitro-1H-pyrazol-1-yl) methyl) -4- (trifluoromethyl) pyridine (436mg, 1.6mmol) in methanol (50mL) under a hydrogen balloon at 25 ℃ was added 10% palladium on charcoal (0.170g, 1.6 mmol). The mixture was stirred at 25 ℃ for 1 h. After completion, the reaction mixture is passed through Filtered and washed with methanol. The filtrate was concentrated to give 1- ((4- (trifluoromethyl) pyridin-3-yl) methyl) -1H-pyrazol-4-amine (384mg, 1.6mmol, 100%) as a red oil. LCMS (ESI) M/z 243.1[ M + H ]]+

And 5: preparation of N- (1-benzyl-1H-pyrazol-4-yl) -5- (1, 3, 4-thiadiazol-2-yl) isoxazole-3-carboxamide

To a solution of 1- ((4- (trifluoromethyl) pyridin-3-yl) methyl) -1H-pyrazol-4-amine (384mg, 1.6mmol), 5-acetylisoxazole-3-carboxylic acid (272mg, 1.75mmol), and triethylamine (1.62g, 16mmol) in dichloromethane (50mL) at 0 ℃ under nitrogen was slowly added propylphosphonic anhydride (0.5M in ethyl acetate, 5.1g, 8.0 mmol). The mixture was stirred at room temperature for 16 h. Upon completion, water (30mL) was added and the mixture was extracted with dichloromethane (100mL × 3). The organic layer was dried over sodium sulfate, filtered and concentrated in vacuo. The crude residue was purified by preparative-TLC (dichloromethane/ammonia in methanol (7N) ═ 40/1) to give N- (1-benzyl-1H-pyrazol-4-yl) -5- (1, 3, 4-thiadiazol-2-yl) isoxazole-3-carboxamide as a white solid (180mg, 0.47mmol, 30%). LCMS (ESI) M/z 380.1[ M + H ]]+

Step 6: (E) preparation of (E) -5- (3- (dimethylamino) acryloyl) -N- (1- ((4- (trifluoromethyl) pyridin-3-yl) methyl) -1H-pyrazol-4-yl) isoxazole-3-carboxamide

To a solution of N- (1-benzyl-1H-pyrazol-4-yl) -5- (1, 3, 4-thiadiazol-2-yl) isoxazole-3-carboxamide (0.180g, 0.47mmol) in toluene (15mL) under nitrogen was added N, N-dimethylformamide dimethyl acetal (0.283g, 2.37 mmol). The mixture was stirred at 110 ℃ for 1h, then cooled and concentrated in vacuo. The crude residue was triturated with diethyl ether (10mL × 2) and dried in vacuo to give (E) -5- (3- (dimethylamino) acryloyl) -N- (1- ((4- (trifluoromethyl) pyridin-3-yl) methyl) -1H-pyrazol-4-yl) isoxazole-3-carboxamide (0.170g, 0.39mmol, 83%) as an orange solid. LCMS (ESI) M/z 435.1[ M + H ]]+

And 7: preparation of 5- (pyrimidin-4-yl) -N- (1- ((4- (trifluoromethyl) pyridin-3-yl) methyl) -1H-pyrazol-4-yl) isoxazole-3-carboxamide

A mixture of (E) -5- (3- (dimethylamino) acryloyl) -N- (1- ((4- (trifluoromethyl) pyridin-3-yl) methyl) -1H-pyrazol-4-yl) isoxazole-3-carboxamide (136mg, 0.31mmol), formamide acetate (65mg, 0.62mmol), and potassium carbonate (87mg, 0.62mmol) in N, N-dimethylformamide (6mL) was heated at 100 ℃ for 2H in a sealed tube. The mixture was cooled, then diluted with ethyl acetate (50mL) and washed with brine (30mL × 3). The organic layer was dried over sodium sulfate, filtered and concentrated in vacuo. The crude residue was purified by prep-HPLC (dissolved in a minimal amount of N, N-dimethylformamide and loaded on a Boston C1821 × 250mM 10 μm column with acetonitrile/10 mM aqueous ammonium acetate as mobile phase) to give 5- (pyrimidin-4-yl) -N- (1- ((4- (trifluoromethyl) pyridin-3-yl) methyl) -1H-pyrazol-4-yl) isoxazole-3-carboxamide as a white solid (49mg, 0.12mmol, 38%). 1H NMR (400MHz, dimethylsulfoxide-d)6)δ11.18(s,1H),9.38(d,J=1.2Hz,1H),9.07(d,J=5.2Hz,1H),8.80(d,J=4.8Hz,1H),8.34(s,1H),8.28(s,1H),8.16(dd,J1=1.2Hz,J2=4.8Hz,1H),7.77-7.79(m,2H),7.74(s,1H),5.60(s,2H);LCMS(ESI)m/z:379.0[M+H]+

Example 53.preparation of 5- (piperidin-1-yl) -N- (1- ((4- (trifluoromethyl) pyridin-3-yl) methyl) -1H-pyrazol-4-yl) isoxazole-3-carboxamide (182)

Step 1: preparation of 3- (ethoxycarbonyl) isoxazole-5-carboxylic acid

Under nitrogenTo a solution of propiolic acid (8.0g, 114.2mmol) in N, N-dimethylformamide (60mL) was added dropwise a solution of ethyl (Z) -2-chloro-2- (hydroxyimino) acetate (6.9g, 45.7mmol) in N, N-dimethylformamide (20mL) under an atmosphere over 40 min. After addition, the reaction mixture was heated to 90 ℃ and a solution of triethylamine (13.8g, 137mmol) in N, N-dimethylformamide (20mL) was added dropwise over 1 h. The reaction mixture was heated at 90 ℃ for 17h and then cooled to room temperature. The reaction mixture was evaporated to dryness, diluted with water (30mL) and adjusted to pH 2 with 1N aqueous hydrogen chloride. The aqueous layer was extracted with ethyl acetate (30 mL. times.2). The combined organic layers were washed with brine (30mL × 2), dried over anhydrous sodium sulfate, filtered and concentrated in vacuo to afford 3- (ethoxycarbonyl) isoxazole-5-carboxylic acid as a yellow oil (3.2g, 17.3mmol, 38%). LCMS (ESI) M/z 186.1[ M + H ]]+

Step 2: preparation of ethyl 5- (tert-butoxycarbonylamino) isoxazole-3-carboxylate

To a solution of 3- (ethoxycarbonyl) isoxazole-5-carboxylic acid (2.7g, 14.6mmol) in toluene (40mL) was added triethylamine (1.8g, 17.5mmol), tert-butanol (2.7g, 36.5mmol) and diphenylphosphoryl azide (4.8g, 17.5 mmol). The reaction mixture was heated at 100 ℃ for 2h and concentrated to dryness. The residue was purified by column chromatography (silica, petroleum ether/ethyl acetate-8/1) to give ethyl 5- (tert-butoxycarbonylamino) isoxazole-3-carboxylate (1.8g, 7.0mmol, 49%) as a white solid. LCMS (ESI) M/z 257.2[ M + H ]]+

And step 3: preparation of 5-aminoisoxazole-3-carboxylic acid ethyl ester

To a solution of ethyl 5- (tert-butoxycarbonylamino) isoxazole-3-carboxylate (0.5g, 1.95mmol) in dichloromethane (12mL) was added trifluoroacetic acid (6 mL). Will be reversedThe mixture was stirred at 25 ℃ for 3h, then concentrated in vacuo. The residue was purified by column chromatography (silica, petroleum ether/ethyl acetate 1/1) to give ethyl 5-aminoisoxazole-3-carboxylate (0.170g, 1.08mmol, 57%) as a yellow solid. LCMS (ESI) M/z 157.1[ M + H ]]+

And 4, step 4: preparation of 5- (piperidin-1-yl) isoxazole-3-carboxylic acid ethyl ester

To a solution of 5-aminoisoxazole-3-carboxylic acid ethyl ester (0.170g, 1.1mmol) in dimethylacetamide (20mL) was added cesium carbonate (1.07g, 3.3mmol) and 1, 5-dibromopentane (0.630g, 2.75 mmol). The reaction mixture was heated at 80 ℃ for 3h, then cooled to room temperature and concentrated in vacuo. The residue was purified by column chromatography (silica, dichloromethane/methanol ═ 15/1) to give ethyl 5- (piperidin-1-yl) isoxazole-3-carboxylate (0.100g, 0.44mmol, 38%) as a white solid. LCMS (ESI) M/z 225.2[ M + H ] ]+

And 5: preparation of 5- (piperidin-1-yl) isoxazole-3-carboxylic acids

To a solution of ethyl 5- (piperidin-1-yl) isoxazole-3-carboxylate (0.100g, 0.44mmol) in tetrahydrofuran/water (v/v ═ 2/1, 9mL) was added lithium hydroxide hydrate (56mg, 1.32mmol) at-20 ℃. The reaction mixture was stirred at-20 ℃ for 10min, then evaporated to dryness to give 5- (piperidin-1-yl) isoxazole-3-carboxylic acid (75mg, 0.38mmol, 86%) as a yellow solid. LCMS (ESI) M/z 197.1[ M + H ]]+

Step 6: preparation of 5- (piperidin-1-yl) -N- (1- ((4- (trifluoromethyl) pyridin-3-yl) methyl) -1H-pyrazol-4-yl) isoxazole-3-carboxamide

To a solution of 5- (piperidin-1-yl) isoxazole-3-carboxylic acid (55mg, 0.28mmol) in dichloromethane (15mL) at 0 ℃ was added 1- ((4- (trifluoromethyl) pyridin-3-yl) methyl) -1H-pyrazol-4-amine (68mg, 0.28mmol) and triethylamine (0.283g, 2.8mmol) and propylphosphonic anhydride (0.445g, 1.4 mmol). The mixture was stirred at 25 ℃ for 17 h. The crude sample was dissolved in a minimal amount of N, N-dimethylformamide and purified via preparative-HPLC (Boston C1821 × 250mm 10 μm column; acetonitrile/0.01% aqueous trifluoroacetic acid) to give (piperidin-1-yl) -N- (1- ((4- (trifluoromethyl) pyridin-3-yl) methyl) -1H-pyrazol-4-yl) isoxazole-3-carboxamide (7.5mg, 0.02mmol, 6%) as a white solid. 1H NMR (500MHz, dimethylsulfoxide-d)6)δ10.72(s,1H),8.80(d,J=5.0Hz,1H),8.32(d,J=12.9Hz,1H),8.22(s,1H),7.79(d,J=5.1Hz,1H),7.70(d,J=4.6Hz,1H),5.63(d,J=6.5Hz,1H),5.58(s,2H),3.35-3.31(m,4H),1.58(s,6H);LCMS(ESI)m/z:421.0[M+H]+

Example 54.preparation of 5- (prop-1-en-2-yl) -N- (1- ((4- (trifluoromethyl) pyridin-3-yl) methyl) -1H-pyrazol-4-yl) isoxazole-3-carboxamide (166)

Step 1: preparation of 4- (trifluoromethyl) nicotinic acid

To a mixture of 4- (trifluoromethyl) nicotinonitrile (5.0g, 29.1mmol) in water (15mL) was slowly added concentrated sulfuric acid (15 mL). The reaction mixture was heated at 110 ℃ for 12h, then cooled and extracted with tetrahydrofuran (50 mL. times.2). The organic layer was dried over sodium sulfate, filtered and concentrated under reduced pressure to give 4- (trifluoromethyl) nicotinic acid (5.2g, crude) as a white solid. LCMS (ESI) M/z 192.1[ M + H ]]+. This material was used in the next step without further purification.

Step 2: preparation of (4- (trifluoromethyl) pyridin-3-yl) methanol

To a solution of 4- (trifluoromethyl) nicotinic acid (5.2g, 27.2mmol) in tetrahydrofuran (150mL) was added borane tetrahydrofuran solution (82mL, 1M, 82 mmol). The reaction mixture was stirred for 16h, then 3N sodium hydroxide was added. The mixture was heated to 60 ℃ and stirred for 1 h. The biphasic mixture was separated and the organic layer was concentrated to give (4- (trifluoromethyl) pyridin-3-yl) methanol (3.3g, crude) as a yellow crude oil. LCMS (ESI) M/z 178.1[ M + H ] ]+. This material was used in the next step without further purification.

And step 3: preparation of 3- ((4-nitro-1H-pyrazol-1-yl) methyl) -4- (trifluoromethyl) pyridine

A mixture of (4- (trifluoromethyl) pyridin-3-yl) methanol (3.2g, 18.1mmol), 4-nitro-1H-pyrazole (2.05g, 18.1mmol) and triphenylphosphine (10.4g, 39.8mmol) in tetrahydrofuran (20mL) was stirred at 20 ℃ for 0.5H. Dipropyl azodicarboxylate (8.04g, 39.8mmol) was added slowly and the reaction mixture was stirred at 20 ℃ for 4 h. The volatiles were removed under reduced pressure and the crude material was purified by column chromatography (silica, petroleum ether/ethyl acetate ═ 1/1) to give 3- ((4-nitro-1H-pyrazol-1-yl) methyl) -4- (trifluoromethyl) pyridine (1.24g, 4.56mmol, 25%) as a yellow solid. LCMS (ESI) M/z 273.1[ M + H ]]+

And 4, step 4: preparation of 1- ((4- (trifluoromethyl) pyridin-3-yl) methyl) -1H-pyrazol-4-amine

To 3- ((4-nitro-1H-pyrazol-1-yl) methyl) -4- (tris under nitrogenTo a solution of fluoromethyl) pyridine (1.24g, 4.56mmol) in methanol (15mL) was added palladium on charcoal (0.248g, 10 wt% Pd). The reaction mixture was stirred under a hydrogen balloon at 20 ℃ for 0.5 h. The mixture was filtered and the filtrate was concentrated in vacuo to give 1- ((4- (trifluoromethyl) pyridin-3-yl) methyl) -1H-pyrazol-4-amine (1.1g, crude) as a red solid. LCMS (ESI) M/z 243.1[ M + H ] ]+

And 5: preparation of 5- (prop-1-en-2-yl) -N- (1- ((4- (trifluoromethyl) pyridin-3-yl) methyl) -1H-pyrazol-4-yl) isoxazole-3-carboxamide

To a solution of 5- (prop-1-en-2-yl) isoxazole-3-carboxylic acid (100mg, 0.653mmol) in dichloromethane (2mL) at 20 ℃ was added oxalyl chloride (1 mL). The reaction mixture was stirred at room temperature for 0.5 h. The volatiles were removed in vacuo and the residue was dissolved in dichloromethane (2mL) and added dropwise to a mixture of 1- ((4- (trifluoromethyl) pyridin-3-yl) methyl) -1H-pyrazol-4-amine (158mg, 0.653mmol) and triethylamine (198mg, 1.96mmol) in dichloromethane (5 mL). The reaction mixture was stirred for 0.5H and dissolved in a minimal amount of N, N-dimethylformamide and purified by preparative-HPLC (Boston C1821 × 250mm 10 μm column. mobile phase acetonitrile/0.01% aqueous trifluoroacetic acid) to afford 5- (prop-1-en-2-yl) -N- (1- ((4- (trifluoromethyl) pyridin-3-yl) methyl) -1H-pyrazol-4-yl) isoxazole-3-carboxamide as a white solid (8.8mg, 0.023mmol, 4%).1H NMR (500MHz, dimethylsulfoxide-d)6)δ11.02(s,1H),8.81(d,J=5.5Hz,1H),8.33(s,1H),8.27(s,1H),7.79(d,J=5.0Hz,1H),7.72(s,1H),7.02(s,1H),5.87(s,1H),5.60(s,2H),5.48(s,1H),2.12(s,3H);LCMS(ESI)m/z:378.1[M+H]+

Example 55.preparation of 5-phenyl-N- (1- ((4- (trifluoromethyl) pyridin-3-yl) methyl) -1H-pyrazol-4-yl) isoxazole-3-carboxamide (160)

Step 1: preparation of 1- ((4- (trifluoromethyl) pyridin-3-yl) methyl) -1H-pyrazol-4-amine

To a solution of 3- ((4-nitro-1H-pyrazol-1-yl) methyl) -4- (trifluoromethyl) pyridine (300mg, 1.03mmol) in methanol (10mL) under nitrogen was added palladium on charcoal (90mg, 10 wt% Pd). The reaction mixture was stirred under a hydrogen balloon at 20 ℃ for 0.5 h. The mixture was filtered and the filtrate was concentrated in vacuo to give 1- ((4- (trifluoromethyl) pyridin-3-yl) methyl) -1H-pyrazol-4-amine (250mg, crude) as a red oil. LCMS (ESI) M/z 243.1[ M + H ]]+

Step 2: preparation of 5-phenyl-N- (1- ((4- (trifluoromethyl) pyridin-3-yl) methyl) -1H-pyrazol-4-yl) isoxazole-3-carboxamide

To a solution of 5-phenylisoxazole-3-carboxylic acid (117mg, 0.620mmol) in dichloromethane (2mL) at 20 ℃ was added oxalyl chloride (1 mL). The reaction mixture was stirred at room temperature for 0.5 h. Volatiles were removed in vacuo. The crude residue was dissolved in dichloromethane (2mL) and added dropwise to a mixture of 1- ((4- (trifluoromethyl) pyridin-3-yl) methyl) -1H-pyrazol-4-amine (0.150g, 0.620mmol) and triethylamine (0.188g, 1.86mmol) in dichloromethane (5 mL). The reaction mixture was stirred for 0.5H and purified by preparative-HPLC (crude sample dissolved in N, N-dimethylformamide and loaded onto a Boston C1821 × 250mm 10 μm column mobile phase acetonitrile/0.01% aqueous trifluoroacetic acid) to afford 5-phenyl-N- (1- ((4- (trifluoromethyl) pyridin-3-yl) methyl) -1H-pyrazol-4-yl) isoxazole-3-carboxamide as a white solid (65.5mg, 0.158mmol, 25%). 1H NMR (500MHz, dimethylsulfoxide-d)6)δ11.09(s,1H),8.81(d,J=5.0Hz,1H),8.34(s,1H),8.29(s,1H),7.98(dd,J=7.6,1.6Hz,2H),7.80(d,J=5.0Hz,1H),7.75(s,1H),7.59–7.56(m,3H),7.47(s,1H),5.61(s,2H);LCMS(ESI)m/z:414.1[M+H]+

Example 56.5 preparation of isopropyl-N- (1- ((4- (trifluoromethyl) pyridin-3-yl) methyl) -1H-pyrazol-4-yl) isoxazole-3-carboxamide (161)

Step 1: preparation of 5- (prop-1-en-2-yl) isoxazole-3-carboxylic acid ethyl ester

To a solution of ethyl 5-bromoisoxazole-3-carboxylate (0.5g, 2.28mmol), 4, 5, 5-tetramethyl-2- (prop-1-en-2-yl) -1, 3, 2-dioxaborolane (461mg, 2.74mmol) and potassium phosphate (968mg, 4.568mmol) in N, N-dimethylformamide (5mL) was added palladium tetraphosphate (257mg, 0.223mmol) under nitrogen. The reaction mixture was heated to 90 ℃ and stirred for 2 h. The organic layer was diluted with ethyl acetate (50mL) and washed with water (100 mL). The organic layer was concentrated in vacuo. The crude material was purified by preparative-TLC (silica, petroleum ether/ethyl acetate-4/1) to give ethyl 5- (prop-1-en-2-yl) isoxazole-3-carboxylate (350mg, 1.93mmol, 85%) as a yellow oil. LCMS (ESI) M/z 182.1[ M + H ]]+

Step 2: preparation of 5- (prop-1-en-2-yl) isoxazole-3-carboxylic acids

To a solution of ethyl 5- (prop-1-en-2-yl) isoxazole-3-carboxylate (0.350g, 1.93mmol) in tetrahydrofuran (4.0mL) was added lithium hydroxide hydrate (81mg, 1.93mmol) in water (2mL) at 0 ℃. The residue was stirred at 0 ℃ for 15 min. Dissolving in 1N hydrochloric acid The aqueous layer was adjusted to pH 3-5. The volatiles were removed in vacuo to give 5- (prop-1-en-2-yl) isoxazole-3-carboxylic acid (0.240g, crude) as a yellow solid. LCMS (ESI) M/z 154.1[ M + H ]]+

And step 3: preparation of 5- (prop-1-en-2-yl) -N- (1- ((4- (trifluoromethyl) pyridin-3-yl) methyl) -1H-pyrazol-4-yl) isoxazole-3-carboxamide

To a solution of 5- (prop-1-en-2-yl) isoxazole-3-carboxylic acid (76mg, 0.496mmol) in dichloromethane (2mL) at 20 ℃ was added oxalyl chloride (1 mL). The reaction mixture was stirred at room temperature for 0.5 h. Volatiles were removed in vacuo. The crude solid was dissolved in dichloromethane (2mL) and added dropwise to a mixture of 1- ((4- (trifluoromethyl) pyridin-3-yl) methyl) -1H-pyrazol-4-amine (120mg, 0.496mmol) and triethylamine (150mg, 1.488mmol) in dichloromethane (3.0 mL). The reaction mixture was stirred for 0.5 h. The mixture was directly purified by column chromatography (silica, petroleum ether/ethyl acetate ═ 2/1) to give 5- (prop-1-en-2-yl) -N- (1- ((4- (trifluoromethyl) pyridin-3-yl) methyl) -1H-pyrazol-4-yl) isoxazole-3-carboxamide as a white solid (110mg, 0.292mmol, 59%). LCMS (ESI) M/z 378.1[ M + H ]]+

And 4, step 4: preparation of 5-isopropyl-N- (1- ((4- (trifluoromethyl) pyridin-3-yl) methyl) -1H-pyrazol-4-yl) isoxazole-3-carboxamide

To a solution of 5- (prop-1-en-2-yl) -N- (1- ((4- (trifluoromethyl) pyridin-3-yl) methyl) -1H-pyrazol-4-yl) isoxazole-3-carboxamide (110mg, 0.292mmol) in methanol (5mL) under nitrogen was added 10% palladium on charcoal (33mg, 10%). The reaction mixture was stirred under a hydrogen balloon at 20 ℃ for 25 min. The volatiles were removed in vacuo and the residue was purified by preparative-HPLC (crude sample was dissolved in N, N-dimethylformamide and loaded onto BostonC1821X 250mm 10 μm column. Mobile phase acetonitrile/0.01% aqueous trifluoroacetic acid) to give 5-isopropyl-N- (1- ((4- (trifluoromethyl) pyridin-3-yl) methyl) -1H-pyrazol-4-yl) isoxazole-3-carboxamide as a white solid (51.2mg, 0.135mmol, 46%).1H NMR (500MHz, dimethylsulfoxide-d)6)δ10.95(s,1H),8.81(d,J=5.0Hz,1H),8.33(s,1H),8.25(s,1H),7.79(d,J=5.0Hz,1H),7.71(s,1H),6.65(d,J=1.0Hz,1H),5.60(s,2H),3.20-3.15(m,1H),1.29(d,J=7.0Hz,6H);LCMS(ESI)m/z:380.1[M+H]+

Example 57 preparation of N- (1- (3, 4-dichlorobenzyl) -3-methyl-1H-pyrazol-4-yl) -5- (furan-2-yl) isoxazole-3-carboxamide (17)

Step 1: preparation of 1- [ (3, 4-dichlorophenyl) methyl ] -5-methyl-4-nitro-pyrazole and 1- [ (3, 4-dichlorophenyl) methyl ] -3-methyl-4-nitro-pyrazole

A mixture of 5-methyl-4-nitro-1H-pyrazole (1.00g, 7.87mmol), 1, 2-dichloro-4- (chloromethyl) benzene (1.6mL, 11.8mmol) and cesium carbonate (2.56g, 7.87mmol) in N, N-dimethylformamide (10mL) was purged with nitrogen (3X) and the mixture was stirred at 18 ℃ under nitrogen for 16H. The reaction mixture was washed with water (20mL), and then extracted with ethyl acetate (10 mL. times.2). The combined organic layers were dried over sodium sulfate, filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (silica, petroleum ether/ethyl acetate 40/1) to give 1- [ (3, 4-dichlorophenyl) methyl group as a pale yellow solid ]-5-methyl-4-nitro-pyrazole (1.6g, crude) and 1- [ (3, 4-dichlorophenyl) methyl]-3-methyl-4-nitro-pyrazole (0.8g, crude). LCMS (ESI) M/z 285.9[ M + H ]]+

Step 2: preparation of 1- [ (3, 4-dichlorophenyl) methyl ] -3-methyl-pyrazol-4-amine and 1- [ (3, 4-dichlorophenyl) methyl ] -5-methyl-pyrazol-4-amine

1- [ (3, 4-dichlorophenyl) methyl]-5-methyl-4-nitro-pyrazole (0.800g, 2.80mmol), 1- [ (3, 4-dichlorophenyl) methyl]A mixture of-3-methyl-4-nitro-pyrazole (0.400g, 1.40mmol), iron powder (0.781g, 14.0mmol) and ammonium chloride (0.489mL, 14.0mmol) in ethanol (8mL) and water (2mL) was purged with nitrogen (3X) and the mixture was then heated at 50 ℃ under nitrogen for 2 h. The reaction mixture was filtered and the filtrate was extracted with dichloromethane (10mL × 2). The combined organic layers were washed with brine (5 mL. times.2), dried over sodium sulfate, filtered and concentrated under reduced pressure to give 1- [ (3, 4-dichlorophenyl) methyl group as a yellow oil]-3-methyl-pyrazol-4-amine (0.5g, crude) and 1- [ (3, 4-dichlorophenyl) methyl]-5-methyl-pyrazol-4-amine (1g, crude). LCMS (ESI) M/z 256.0[ M + H ]]+

And step 3: preparation of N- (1- (3, 4-dichlorobenzyl) -5-methyl-1H-pyrazol-4-yl) -5- (furan-2-yl) isoxazole-3-carboxamide and N- (1- (3, 4-dichlorobenzyl) -3-methyl-1H-pyrazol-4-yl) -5- (furan-2-yl) isoxazole-3-carboxamide

1- [ (3, 4-dichlorophenyl) methyl]-3-methyl-pyrazol-4-amine (0.107g, 0.419mmol), 1- [ (3, 4-dichlorophenyl) methyl]A mixture of-5-methyl-pyrazol-4-amine (0.214g, 0.837mmol), 5- (2-furyl) isoxazole-3-carboxylic acid (0.150g, 838mmol), diisopropylethylamine (438mL, 2.51mmol) and N, N' -tetramethyl-O- (1H-benzotriazol-1-yl) uronium hexafluorophosphate (0.381g, 1.00mmol) in N, N-dimethylformamide (3mL) was purged with nitrogen (3 ×), and then the mixture was stirred at 18 ℃ under nitrogen for 2 hours. The reaction mixture was washed with water (10mL), and then extracted with ethyl acetate (5 mL. times.2). The combined organic layers were washed with brine(5 mL. times.2) dried over sodium sulfate, filtered and concentrated under reduced pressure to give a residue. The residue is purified by column chromatography (silica, petroleum ether/ethyl acetate 7/3, then by preparative-HPLC (column: Agela Durashell C18150X 255 u; mobile phase: [ water (0.04% ammonia) -acetonitrile](ii) a B%: 35% -85%, 12min) to yield first N- [1- [ (3, 4-dichlorophenyl) methyl group as a brown solid]-3-methyl-pyrazol-4-yl]-5- (2-furyl) isoxazole-3-carboxamide (0.177g, 0.409mmol, 49%).1H NMR (400MHz, chloroform-d) δ 8.22(s, 1H), 8.07(s, 1H), 7.61(d, J1.2 Hz, 1H), 7.42(d, J8.4 Hz, 1H), 7.33(d, J2.0 Hz, 1H), 7.07(dd, J2.0, 8.4Hz, 1H), 7.00(d, J3.6 Hz, 1H), 6.92(s, 1H), 6.59(dd, J1.6, 3.5Hz, 1H), 5.19(s, 2H), 2.33(s, 3H); LCMS (ESI) M/z 417.2[ M + H ] ]+And secondly to yield N- [1- [ (3, 4-dichlorophenyl) methyl group as a white solid]-5-methyl-pyrazol-4-yl]-5- (2-furyl) isoxazole-3-carboxamide (0.040g, 0.0957mmol, 11%).1H NMR (400MHz, chloroform-d) δ 8.07(s, 1H), 7.83(s, 1H), 7.61(d, J1.6 Hz, 1H), 7.41(d, J8.4 Hz, 1H), 7.24(d, J2.0 Hz, 1H), 7.00(d, J3.2 Hz, 1H), 6.96(dd, J1.6, 8.3Hz, 1H), 6.94(s, 1H), 6.59(dd, J2.0, 3.3Hz, 1H), 5.26(s, 2H), 2.21(s, 3H); LCMS (ESI) M/z 417.0[ M + H ]]+

EXAMPLE 58. preparation of N- [1- [ [ 5-fluoro-2- (trifluoromethyl) phenyl ] methyl ] pyrazol-4-yl ] -5- (2-furyl) isoxazole-3-carboxamide (36)

Step 1: preparation of 1- [ [ 5-fluoro-2- (trifluoromethyl) phenyl ] methyl ] -4-nitro-pyrazole

To a solution of 4-nitro-1H-pyrazole (0.150g, 1.33mmol) in N, N-dimethylformamide (4mL) was addedCesium carbonate (0.864g, 2.65mmol) and 2- (bromomethyl) -4-fluoro-1- (trifluoromethyl) benzene (0.358g, 1.39 mmol). The mixture was stirred at 20 ℃ for 3 h. The reaction mixture was diluted with water (20mL) and extracted with ethyl acetate (10 mL. times.3). The combined organic layers were washed with brine (20mL), dried over sodium sulfate, filtered and concentrated under reduced pressure to give 1- [ [ 5-fluoro-2- (trifluoromethyl) phenyl ] as a red solid ]Methyl radical]-4-nitro-pyrazole (0.380g, crude) which was used in the next step without further purification. LCMS (ESI) mz 290.0[ M + H ]]+

Step 2: preparation of 1- [ [ 5-fluoro-2- (trifluoromethyl) phenyl ] methyl ] pyrazol-4-amine

1- [ [ 5-fluoro-2- (trifluoromethyl) phenyl group]Methyl radical]A mixture of-4-nitro-pyrazole (0.380g, 1.31mmol), iron powder (0.367g, 6.57mmol), ammonium chloride (0.230mL, 6.57mmol) in ethanol (20mL) and water (5mL) was purged with nitrogen (3X). The reaction mixture was warmed at 50 ℃ under nitrogen for 4h, then concentrated under reduced pressure and diluted with water (20 mL). The aqueous layer was extracted with ethyl acetate (10 mL. times.3). The combined organic layers were washed with brine (20mL), dried over sodium sulfate, filtered and concentrated under reduced pressure to give 1- [ [ 5-fluoro-2- (trifluoromethyl) phenyl ] residue as a red residue]Methyl radical]Pyrazol-4-amine (0.300g, 1.16mmol, 88%) was used in the next step without further purification. LCMS (ESI) M/z 260.1[ M + H ]]+

And step 3: preparation of N- [1- [ [ 5-fluoro-2- (trifluoromethyl) phenyl ] methyl ] pyrazol-4-yl ] -5- (2-furyl) isoxazole-3-carboxamide

To a solution of 5- (2-furyl) isoxazole-3-carboxylic acid (0.150g, 0.837mmol) in N, N-dimethylformamide (3mL) at 15 deg.C was added N, N, N ', N' -tetramethyl-O- (1H-benzotriazol-1-yl) uronium hexafluorophosphate (0.381g, 1.00mmol), diisopropylethyl acetate Amine (0.438mL, 2.51mmol) and 1- [ [ 5-fluoro-2- (trifluoromethyl) phenyl ]]Methyl radical]Pyrazol-4-amine (0.239g, 0.921 mmol). The mixture was stirred at 15 ℃ for 2 h. The residue was purified by preparative-HPLC (YMC-Actus Triart C18150X 305 u; mobile phase: [ water (10mM ammonium bicarbonate) -acetonitrile](ii) a B%: 50-70 percent, 10min) to obtain the N- [1- [ [ 5-fluorine-2- (trifluoromethyl) phenyl ] which is light yellow solid]Methyl radical]Pyrazol-4-yl]-5- (2-furyl) isoxazole-3-carboxamide (0.083g, 0.20mmol, 24%).1H NMR (400MHz, chloroform-d) δ 8.61(s, 1H), 8.12(s, 1H), 7.74-7.67(m, 2H), 7.60(d, J ═ 1.1Hz, 1H), 7.11-7.04(m, 1H), 6.99(d, J ═ 3.5Hz, 1H), 6.93(s, 1H), 6.65(dd, J ═ 1.9, 9.4Hz, 1H), 6.58(dd, J ═ 1.8, 3.5Hz, 1H), 5.53(s, 2H); LCMS (ESI) M/z 421.1[ M + H ]]+

Example 59 preparation of N- (1- (2-chloro-5-fluorobenzyl) -1H-pyrazol-4-yl) -5- (furan-2-yl) isoxazole-3-carboxamide (28)

Step 1: preparation of 1- [ (2-chloro-5-fluoro-phenyl) methyl ] -4-nitro-pyrazole

A mixture of 4-nitro-1H-pyrazole (0.2g, 1.77mmol), 2- (bromomethyl) -1-chloro-4-fluoro-benzene (0.395g, 1.77mmol) and cesium carbonate (1.73g, 5.31mmol) in N, N-dimethylformamide (2mL) was purged with nitrogen (3 ×), and the mixture was then stirred at 18 ℃ under nitrogen for 16H. The reaction mixture was diluted with water (5mL) and extracted with ethyl acetate (5 mL. times.2). The combined organic layers were washed with brine (5mL × 2), dried over sodium sulfate, filtered and concentrated under reduced pressure to give 1- [ (2-chloro-5-fluoro-phenyl) methyl ] as a yellow solid ]-4-nitro-pyrazole (0.407g, 1.59mmol, 90%). The material was used directly in the next step without further purification.1H NMR (400MHz, chloroform-d) delta 8.20(s, 1H), 8.142(s, 1H), 7.44-7.41(m, 1H), 7.09-7.07(m,1H),6.98-6.95(m,1H),5.42(s,2H);LCMS(ESI)m/z:256.0[M+H]+

Step 2: preparation of 1- [ (2-chloro-5-fluoro-phenyl) methyl ] pyrazol-4-amine

Reacting 1- [ (2-chloro-5-fluoro-phenyl) methyl]A mixture of-4-nitro-pyrazole (0.407g, 1.59mmol), iron powder (0.444g, 7.96mmol) and ammonium chloride (0.278mL, 7.96mmol) in ethanol (4mL) and water (1mL) was purged with nitrogen (3X) and the mixture was then heated at 50 ℃ under nitrogen for 2 h. The reaction mixture was filtered and the filtrate was extracted with dichloromethane (10mL × 2). The combined organic layers were washed with brine (5mL × 2), dried over sodium sulfate, filtered and concentrated under reduced pressure to give 1- [ (2-chloro-5-fluoro-phenyl) methyl as a dark brown oil]Pyrazol-4-amine (0.279g, 1.24mmol, 78%).1H NMR (400MHz, chloroform-d) Δ 7.35-7.31(m, 1H), 7.25(s, 1H), 7.09(s, 1H), 6.95-6.93(m, 1H), 6.58-6.55(m, 1H), 5.27(s, 2H); LCMS (ESI) M/z 226.4[ M + H ]]+

And step 3: preparation of N- [1- [ (2-chloro-5-fluoro-phenyl) methyl ] pyrazol-4-yl ] -5- (2-furyl) isoxazole-3-carboxamide

Reacting 1- [ (2-chloro-5-fluoro-phenyl) methyl ]A mixture of pyrazol-4-amine (0.101g, 0.447mmol), 5- (2-furyl) isoxazole-3-carboxylic acid (0.080g, 447mmol), N' -tetramethyl-O- (1H-benzotriazol-1-yl) uronium hexafluorophosphate (0.203g, 0.536mmol) and diisopropylethylamine (0.233mL, 1.34mmol) in N, N-dimethylformamide (2mL) was purged with nitrogen (3 ×), and the mixture was then stirred at 18 ℃ under nitrogen for 2H. The residue was purified by preparative-HPLC (Waters Xbridge C18150X 25mM X5 um column; acetonitrile in 10mM aqueous ammonium acetate, gradient of 11min 37-67%) to give N- [1- [ (2-chloro-5-fluoro-phenyl) methyl ] as a pale yellow solid]Pyrazol-4-yl]-5- (2-furyl) isoxazole-3-carboxamide (0.075g, 0.192mmol, 43%).1H NMR (400MHz, chloroform-d) δ 8.58(s, 1H), 8.14(s, 1H), 7.67(s, 1H), 7.60(s, 1H), 7.37(dd, J ═ 5.2, 8.8Hz, 1H), 7.05-6.95(m, 2H), 6.93(s, 1H), 6.69(dd, J ═ 3.2, 8.9Hz, 1H), 6.58(dd, J ═ 1.6, 3.2Hz, 1H), 5.41(s, 2H); LCMS (ESI) M/z 387.0[ M + H ]]+

EXAMPLE 60 preparation of 5- (2-furyl) -N- [1- [ [3- (trifluoromethyl) phenyl ] methyl ] pyrazol-4-yl ] isoxazole-3-carboxamide (20)

Step 1: preparation of 4-nitro-1- [ [3- (trifluoromethyl) phenyl ] methyl ] pyrazole

To a stirred solution of 4-nitro-1H-pyrazole (0.3g, 2.65mmol), 1- (bromomethyl) -3- (trifluoromethyl) benzene (0.404mL, 2.65mmol) in N, N-dimethylformamide (3mL) was added cesium carbonate (2.59g, 7.96mmol), and the mixture was stirred at 20 ℃ for 12H. The mixture was poured into water (20mL), extracted with ethyl acetate (20mL × 3) and the combined organic layers were concentrated under reduced pressure to give 4-nitro-1- [ [3- (trifluoromethyl) phenyl ] as a yellow oil]Methyl radical]Pyrazole (0.5g, 1.84mmol, 69%). LCMS (ESI) M/z 272.3[ M + H ]]+. This material was used in the next step without further purification.

Step 2: preparation of 1- [ [3- (trifluoromethyl) phenyl ] methyl ] pyrazol-4-amine

To 4-nitro-1- [ [3- (trifluoromethyl) phenyl]Methyl radical]To a stirred solution of pyrazole (0.5g, 1.84mmol) in ethanol (20mL) and water (5mL) were added iron powder (0.515g, 9.22mmol) and ammonium chloride (0.129mL, 3.69mmol) and the mixture is heated at 50 ℃ for 2 h. The mixture was filtered and the filtrate was extracted with dichloromethane (20mL × 3) and the combined organic layers were concentrated under reduced pressure to give 1- [ [3- (trifluoromethyl) phenyl ] as a yellow oil]Methyl radical]Pyrazol-4-amine (0.3g, crude). LCMS (ESI) M/z 242.4[ M + H ]]+. This material was used in the next step without further purification.

And step 3: preparation of 5- (2-furyl) -N- [1- [ [3- (trifluoromethyl) phenyl ] methyl ] pyrazol-4-yl ] isoxazole-3-carboxamide

To a solution of 5- (2-furyl) isoxazole-3-carboxylic acid (0.090g, 0.502mol) in N, N-dimethylformamide (2mL) were added N, N, N ', N' -tetramethyl-O- (1H-benzotriazol-1-yl) uronium hexafluorophosphate (0.286g, 0.754mmol), diisopropylethylamine (0.263mL, 1.51mmol) and 1- [ [3- (trifluoromethyl) phenyl ] ethyl phosphate]Methyl radical]Pyrazol-4-amine (0.133g, 0.553 mmol). The reaction mixture was stirred at 15 ℃ for 2 h. The crude residue was purified by preparative-HPLC (YMC-Actus Triart C18100X 30mM X5 um; acetonitrile in 10mM aqueous ammonium acetate, 12min gradient, 40-60%) to give 5- (2-furyl) -N- [1- [ [3- (trifluoromethyl) phenyl ] as a pale yellow solid]Methyl radical]Pyrazol-4-yl]Isoxazole-3-carboxamide (0.030g, 0.073mmol, 14%).1H NMR (400MHz, chloroform-d) δ 8.50(br.s, 1H), 8.07(s, 1H), 7.62(s, 1H), 7.59-7.55(m, 2H), 7.52-7.43(m, 2H), 7.42-7.38(m, 1H), 6.98(d, J ═ 3.3Hz, 1H), 6.90(s, 1H), 6.57(dd, J ═ 1.5, 3.3Hz, 1H), 5.35(s, 2H); LCMS (ESI) M/z 403.0[ M + H ]]+

Example 61.preparation of 5- (furan-2-yl) -N- (1- (2- (trifluoromethyl) benzyl) -1H-pyrazol-4-yl) isoxazole-3-carboxamide (14)

Step 1: preparation of 4-nitro-1- [ [2- (trifluoromethyl) phenyl ] methyl ] pyrazole

A mixture of 4-nitro-1H-pyrazole (0.3g, 2.65mmol), 1- (bromomethyl) -2- (trifluoromethyl) benzene (0.403mL, 2.65mmol) and cesium carbonate (2.59g, 7.95mmol) in N, N-dimethylformamide (8mL) was purged with nitrogen (3X) and the mixture was stirred at 18 ℃ under nitrogen for 16H. The reaction mixture was diluted with water (5mL) and extracted with ethyl acetate (5 mL. times.2). The combined organic layers were washed with brine (5mL × 2), dried over sodium sulfate, filtered and concentrated under reduced pressure to give 4-nitro-1- [ [2- (trifluoromethyl) phenyl ] as a yellow solid]Methyl radical]Pyrazole (0.748g, 2.43mmol, 92%). This material was used in the next step without further purification.1H NMR (400MHz, chloroform-d) δ 8.13(s, 1H), 8.09(s, 1H), 7.77-7.75(d, J ═ 8.0Hz, 1H), 7.62-7.58(t, J ═ 7.6, 1H), 7.54-7.50(t, J ═ 7.6, 1H), 7.31-7.29(t, J ═ 7.6, 1H), 5.54(s, 2H); LCMS (ESI) M/z 272.1[ M + H]+

Step 2: preparation of 1- [ [2- (trifluoromethyl) phenyl ] methyl ] pyrazol-4-amine

4-nitro-1- [ [2- (trifluoromethyl) phenyl group]Methyl radical]A mixture of pyrazole (0.374g, 1.38mmol), iron powder (0.385g, 6.90mmol) and ammonium chloride (241mL, 6.90mmol) in ethanol (4mL) and water (1mL) was purged with nitrogen (3X) and the mixture was heated at 50 ℃ under nitrogen for 2 h. The reaction mixture was filtered and the filtrate was extracted with dichloromethane (10mL × 2). The combined organic layers were washed with brine (5mL × 2), dried over sodium sulfate, filtered and concentrated under reduced pressure to give 1- [ [2- (trifluoromethyl) phenyl ] as a yellow oil ]Methyl radical]Pyrazol-4-amine (0.192g, 0.798mmol, 56%). This material was used in the next step without further purification.1H NMR (400MHz, chloroform-d) δ 7.67-7.65(t, J ═ 7.6, 1H), 7.48-7.44(t, J ═ 7.6, 1H),7.39-7.35(t,J=7.6,1H),7.28(s,1H),7.03(s,1H),6.95-6.93(d,J=8.0Hz,1H),5.54(s,2H);LCMS(ESI)m/z:242.3[M+H]+

and step 3: preparation of 5- (2-furyl) -N- [1- [ [2- (trifluoromethyl) phenyl ] methyl ] pyrazol-4-yl ] isoxazole-3-carboxamide

1- [ [2- (trifluoromethyl) phenyl group]Methyl radical]A mixture of pyrazol-4-amine (0.108g, 0.447mmol), 5- (2-furyl) isoxazole-3-carboxylic acid (0.080g, 447mmol), N, N, N ', N' -tetramethyl-O- (1H-benzotriazol-1-yl) uronium hexafluorophosphate (0.203g, 0.536mmol) and diisopropylethylamine (0.233mL, 1.34mmol) in N, N-dimethylformamide (2mL) was purged with nitrogen (3X). The reaction mixture was stirred at 18 ℃ for 1h under nitrogen. The residue was purified by preparative-HPLC (Agela Durashell C18150X 255 um column; 45-95% acetonitrile in 0.04% ammonium hydroxide, 12min gradient) to give 5- (2-furyl) -N- [1- [ [2- (trifluoromethyl) phenyl ] as a pale yellow solid]Methyl radical]Pyrazol-4-yl]Isoxazole-3-carboxamide (0.088g, 0.217mmol, 49%).1H NMR (400MHz, chloroform-d) δ 8.55(br.s, 1H), 8.07(s, 1H), 7.72-7.66(m, 2H), 7.60(s, 1H), 7.53-7.45(m, 1H), 7.44-7.36(m, 1H), 7.05-6.96(m, 2H), 6.92(s, 1H), 6.58(dd, J ═ 2.0, 3.4Hz, 1H), 5.54(s, 2H); LCMS (ESI) M/z 403.1[ M + H ] ]+

Example 62 preparation of N- [1- [ (4-fluorophenyl) methyl ] pyrazol-4-yl ] -5- (2-furyl) isoxazole-3-carboxamide (18)

Step 1: preparation of 1- [ (4-fluorophenyl) methyl ] -4-nitro-pyrazole

To a solution of 4-nitro-1H-pyrazole (0.300g, 2.65mmol) in N, N-dimethylformamide (5mL) were added cesium carbonate (2.59g, 7.95mmol) and 1- (chloromethyl) -4-fluoro-benzene (317mL, 2.65 mmol). The mixture was stirred at 15 ℃ for 15 h. The reaction mixture was partitioned between ethyl acetate (40mL) and water (40 mL). The organic phase was separated, washed with brine (20mL), dried over sodium sulfate, filtered and concentrated under reduced pressure to give 1- [ (4-fluorophenyl) methyl group as a brown oil]-4-nitro-pyrazole (0.586g, 2.65mmol), which was used directly in the next step. LCMS (ESI) M/z 222.0[ M + H ]]+

Step 2: preparation of 1- [ (4-fluorophenyl) methyl ] pyrazol-4-amine

To 1- [ (4-fluorophenyl) methyl group under nitrogen]To a solution of-4-nitro-pyrazole (0.586g, 2.65mmol) in ethanol (20mL) and water (5mL) were added ammonium chloride (463mg, 13.3mmol) and iron powder (0.740g, 13.3 mmol). The mixture was heated at 50 ℃ for 4 h. The reaction mixture was concentrated under reduced pressure to remove ethanol. The residue was diluted with water (20mL) and extracted with ethyl acetate (10 mL. times.3). The combined organic layers were washed with brine (20mL), dried over sodium sulfate, filtered and concentrated under reduced pressure to give 1- [ (4-fluorophenyl) methyl group as a red oil ]Pyrazol-4-amine (0.390g, crude) was used in the next step without further purification. LCMS (ESI) M/z 192.3[ M + H ]]+

And step 3: preparation of N- [1- [ (4-fluorophenyl) methyl ] pyrazol-4-yl ] -5- (2-furyl) isoxazole-3-carboxamide

To a solution of 5- (2-furyl) isoxazole-3-carboxylic acid (0.090g, 0.50mmol) in N, N-dimethylformamide (3mL) at 15 ℃ were added N, N, N ', N' -tetramethyl-O- (1H-benzotriazol-1-yl) uronium hexafluorophosphate (0.229g, 0.60mmol), 1- [ (4)-fluorophenyl) methyl]Pyrazol-4-amine (0.106g, 0.55mmol), diisopropylethylamine (263mL, 1.51 mmol). The mixture was stirred at 15 ℃ for 2 h. The mixture was purified by preparative-HPLC (Waters X bridge 150X 255 μm column; 30-65% acetonitrile in 10mM aqueous ammonium acetate, 10min gradient) to give N- [1- [ (4-fluorophenyl) methyl group as a pale yellow solid]Pyrazol-4-yl]-5- (2-furyl) isoxazole-3-carboxamide (0.077g, 0.21mmol, 43%).1H NMR (400MHz, chloroform-d) δ 8.51(s, 1H), 8.04(s, 1H), 7.60(s, 2H), 7.27-7.23(m, 2H), 7.04(t, J ═ 8.6Hz, 2H), 6.99(d, J ═ 3.5Hz, 1H), 6.91(s, 1H), 6.58(dd, J ═ 1.8, 3.5Hz, 1H), 5.27(s, 2H); LCMS (ESI) M/z 353.1[ M + H ] ]+

EXAMPLE 63. preparation of N- [1- [ (2-fluorophenyl) methyl ] pyrazol-4-yl ] -5- (2-furyl) isoxazole-3-carboxamide (19)

Step 1: preparation of 1- [ (2-fluorophenyl) methyl ] -4-nitro-pyrazole

To a solution of 4-nitro-1H-pyrazole (0.30g, 2.65mmol) in N, N-dimethylformamide (5mL) was added cesium carbonate (2.59g, 7.96mmol) and 1- (bromomethyl) -2-fluoro-benzene (0.319mL, 2.65 mmol). The mixture was stirred at 15 ℃ for 15 h. The reaction mixture was diluted with water (20mL) and extracted with ethyl acetate (10 mL. times.3). The combined organic layers were washed with brine (20mL), dried over sodium sulfate, filtered and concentrated under reduced pressure to give 1- [ (2-fluorophenyl) methyl group as a brown oil]-4-nitro-pyrazole (0.586g, 2.65 mmol). LCMS (ESI) M/z 222.0[ M + H ]]+. This material was used in the next step without further purification.

Step 2: preparation of 1- [ (2-fluorophenyl) methyl ] pyrazol-4-amine

To 1- [ (2-fluorophenyl) methyl group under nitrogen]To a solution of-4-nitro-pyrazole (0.586g, 2.65mmol) in water (5mL) and ethanol (20mL) were added ammonium chloride (0.463mL, 13.3mmol) and iron powder (0.740g, 13.25 mmol). The mixture was heated at 50 ℃ for 4 h. The reaction mixture was concentrated under reduced pressure to remove ethanol. The residue was diluted with water (20mL) and extracted with ethyl acetate (10 mL. times.3). The combined organic layers were washed with 20mL brine, dried over sodium sulfate, filtered and concentrated under reduced pressure to give 1- [ (2-fluorophenyl) methyl group as a brown oil ]Pyrazol-4-amine (0.400g, crude). LCMS (ESI) M/z 192.4[ M + H ]]+. This material was used in the next step without further purification.

And step 3: preparation of N- [1- [ (2-fluorophenyl) methyl ] pyrazol-4-yl ] -5- (2-furyl) isoxazole-3-carboxamide

To a solution of 5- (2-furyl) isoxazole-3-carboxylic acid (0.090g, 0.50mmol) in N, N-dimethylformamide (3mL) at 15 ℃ were added N, N, N ', N' -tetramethyl-O- (1H-benzotriazol-1-yl) uronium hexafluorophosphate (0.229g, 0.60mmol), 1- [ (2-fluorophenyl) methyl ] phosphonium hexafluorophosphate (2.229 g, 0.60mmol)]Pyrazol-4-amine (0.106g, 0.56mmol) and diisopropylethylamine (0.263mL, 1.51 mmol). The mixture was stirred at 15 ℃ for 2 h. The mixture was purified by preparative-HPLC (Waters X bridge 150X 255 μm column; 35-65% acetonitrile in 10mM aqueous ammonium acetate, 10min gradient) to give N- [1- [ (2-fluorophenyl) methyl group as a pale yellow solid]Pyrazol-4-yl]-5- (2-furyl) isoxazole-3-carboxamide (0.075g, 0.21mmol, 43%).1H NMR (400MHz, chloroform-d) δ 8.51(s, 1H), 8.08(s, 1H), 7.64-7.58(m, 2H), 7.36-7.28(m, 1H), 7.23-7.17(m, 1H), 7.15-7.07(m, 2H), 6.99(d, J ═ 3.5Hz, 1H), 6.92(s, 1H), 6.58(dd, J ═ 1.8, 3.3Hz, 1H), 5.37(s, 2H); LCMS (ESI) M/z 353.1M + H ]+

Example 64 preparation of N- [1- [ (3-chlorophenyl) methyl ] pyrazol-4-yl ] -5- (2-furyl) isoxazole-3-carboxamide (22)

Step 1: preparation of 1- [ (3-chlorophenyl) methyl ] -4-nitro-pyrazole

To a stirred solution of 4-nitro-1H-pyrazole (0.300g, 2.65mmol) in N, N-dimethylformamide (4mL) were added cesium carbonate (2.59g, 7.96mmol) and 1- (bromomethyl) -3-chloro-benzene (0.347mL, 2.65 mmol). The mixture was then stirred at 20 ℃ for 16h, then added to water (5mL) and extracted with ethyl acetate (10 mL. times.3). The combined organic layers were washed with brine (10 mL. times.2), dried over sodium sulfate, filtered and concentrated under reduced pressure to give 1- [ (3-chlorophenyl) methyl group as a yellow oil]-4-nitro-pyrazole (0.700g, 2.94mmol, crude). This material was used in the next step without further purification.1H NMR (400MHz, chloroform-d) Δ 8.10(s, 1H), 8.01(s, 1H), 7.37-7.16(m, 4H), 5.28(s, 2H).

Step 2: preparation of 1- [ (3-chlorophenyl) methyl ] pyrazol-4-amine

To 1- [ (3-chlorophenyl) methyl group]To a stirred solution of-4-nitro-pyrazole (0.350g, 1.47mmol) in ethanol (4mL) and water (1mL) were added iron powder (0.411g, 7.36mmol) and ammonium chloride (0.26mL, 7.36 mmol). The reaction mixture was stirred at 60 ℃ for 3h, then filtered and extracted with ethyl acetate (10 mL. times.3). The combined organic layers were washed with brine (10 mL. times.2), dried over sodium sulfate, filtered and concentrated under reduced pressure to give 1- [ (3-chlorophenyl) methyl group as a red oil ]Pyrazol-4-amine (0.300g, crude). LCMS (ESI) M/z 208.1[ M + H ]]+

And step 3: preparation of N- [1- [ (3-chlorophenyl) methyl ] pyrazol-4-yl ] -5- (2-furyl) isoxazole-3-carboxamide

To 1- [ (3-chlorophenyl) methyl group]To a solution of pyrazol-4-amine (0.115g, 0.554mmol) in N, N-dimethylformamide (3mL) were added diisopropylethylamine (0.276mL, 1.58mmol) and N, N' -tetramethyl-O- (1H-benzotriazol-1-yl) uronium hexafluorophosphate (0.200g, 0.527mmol) and 5- (2-furyl) isoxazole-3-carboxylic acid (0.094g, 0.527 mmol). The mixture was stirred at 20 ℃ for 16 h. The reaction mixture was filtered and purified by preparative-HPLC (column: Agela Durashell C18150X 255 μm; mobile phase: [ water (10mM ammonium bicarbonate) -acetonitrile](ii) a B%: 40% -75%, 12min gradient) to obtain N- [1- [ (3-chlorophenyl) methyl group as a white solid]Pyrazol-4-yl]-5- (2-furyl) isoxazole-3-carboxamide (0.0450g, 0.122mmol, 23%).1H NMR (400MHz, chloroform-d) δ 8.55(s, 1H), 8.06(s, 1H), 7.65-7.55(m, 2H), 7.31-7.26(m, 2H), 7.22(s, 1H), 7.16-7.08(m, 1H), 6.98(d, J ═ 3.5Hz, 1H), 6.91(s, 1H), 6.57(m, 1H), 5.27(s, 2H); LCMS (ESI) M/z 369.1[ M + H ]]+

EXAMPLE 65. preparation of N- [1- [ (2-chlorophenyl) methyl ] pyrazol-4-yl ] -5- (2-furyl) isoxazole-3-carboxamide (23)

Step 1: preparation of 1- [ (2-chlorophenyl) methyl ] -4-nitro-pyrazole

To a stirred solution of 4-nitro-1H-pyrazole (0.300g, 2.65mmol) in N, N-dimethylformamide (4mL) were added cesium carbonate (2.59g, 7.95mmol) and 1-chloro-2- (chloromethyl) benzene (0.336mL, 2.65 mmol). The mixture is then brought to 20 DEG CStirred for 16h, then added to water (5mL) and extracted with ethyl acetate (10 mL. times.3). The combined organic layers were washed with brine (10 mL. times.2), dried over sodium sulfate, filtered and concentrated under reduced pressure to give 1- [ (2-chlorophenyl) methyl group as a yellow oil]-4-nitro-pyrazole (0.700g, 2.94 mmol).1H NMR (400MHz, chloroform-d) Δ 8.15(s, 1H), 8.11(s, 1H), 7.49-7.28(m, 4H), 5.46(s, 2H). This material was used in the next step without further purification.

Step 2: preparation of 1- [ (2-chlorophenyl) methyl ] pyrazol-4-amine

To 1- [ (2-chlorophenyl) methyl group]To a stirred solution of-4-nitro-pyrazole (0.700g, 2.94mmol) in ethanol (4mL) and water (1mL) were added iron powder (0.411g, 7.36mmol) and ammonium chloride (0.257mL, 7.36 mmol). The reaction mixture was stirred at 60 ℃ for 3h, then filtered and extracted with ethyl acetate (10 mL. times.3). The combined organic layers were washed with brine (10 mL. times.2), dried over sodium sulfate, filtered and concentrated under reduced pressure to give 1- [ (2-chlorophenyl) methyl group as a red oil ]Pyrazol-4-amine (0.270g, crude). LCMS (ESI) M/z 208.1[ M + H ]]+. The material was used in the next step without further purification.

And step 3: preparation of [1- [ (2-chlorophenyl) methyl ] pyrazol-4-yl ] -5- (2-furyl) isoxazole-3-carboxamide

To 1- [ (2-chlorophenyl) methyl group]To a solution of pyrazol-4-amine (0.150g, 0.722mmol) in N, N-dimethylformamide (3mL) were added diisopropylethylamine (359mL, 2.06mmol), N' -tetramethyl-O- (1H-benzotriazol-1-yl) uronium hexafluorophosphate (0.261g, 0.688mmol) and 5- (2-furyl) isoxazole-3-carboxylic acid (0.123g, 0.688 mmol). The mixture was stirred at 20 ℃ for 16 h. The reaction mixture was filtered. The crude residue was passed through preparative-HPLC (Agela Durashell 150X 255 μm column(ii) a 40-70% acetonitrile in 10mM aqueous ammonium bicarbonate, 12min gradient) to yield N- [1- [ (2-chlorophenyl) methyl ] methyl as a white solid]Pyrazol-4-yl]-5- (2-furyl) isoxazole-3-carboxamide (0.066mg, 0.180mmol, 26%).1H NMR (400MHz, chloroform-d) δ 8.53(s, 1H), 8.08(s, 1H), 7.64(s, 1H), 7.59(d, J ═ 1.1Hz, 1H), 7.40(dd, J ═ 1.7, 7.6Hz, 1H), 7.28-7.22(m, 2H), 7.06(dd, J ═ 1.8, 7.3Hz, 1H), 6.98(d, J ═ 3.5Hz, 1H), 6.91(s, 1H), 6.58(m, 1H), 5.43(s, 2H); LCMS (ESI) M/z 369.0[ M + H ] ]+

Example 66.5- (2-furyl) -N- [1- (4,4, 4-trifluorobutyl) pyrazol-4-yl ] isoxazole-3-carboxylic acid amide (34)

Step 1: preparation of 4-nitro-1- (4,4, 4-trifluorobutyl) pyrazole

To a stirred solution of 4-nitro-1H-pyrazole (0.100g, 0.884mmol) and 1, 1, 1-trifluoro-4-iodo-butane (0.232g, 0.973mol) in N, N-dimethylformamide (2mL) was added potassium carbonate (0.134g, 0.973mmol) at 15 ℃, followed by stirring at 15 ℃ for 20H. The reaction mixture was quenched with ice water (2mL) and extracted with ethyl acetate (10 mL. times.3). The combined organic layers were washed with water (3mL × 3) and brine (3mL), dried over sodium sulfate, filtered and concentrated under reduced pressure to give a crude residue. The residue was purified by column chromatography (ISCO, 10g silica, 0-25% ethyl acetate in petroleum ether, 20min gradient) to give 4-nitro-1- (4,4, 4-trifluorobutyl) pyrazole as a colorless oil (0.150g, 0.672mmol, 76%). LCMS (ESI) M/z 224.0[ M + H ]]+

Step 2: preparation of 1- (4,4, 4-trifluorobutyl) pyrazol-4-amine

To a solution of 4-nitro-1- (4,4, 4-trifluorobutyl) pyrazole (0.130g, 0.583mmol) in methanol (5mL) was added palladium on activated carbon (0.020g, 0.058mmol, 10 wt% Pd) under nitrogen. The suspension was purged with hydrogen (3 ×). The mixture was stirred at 15 ℃ for 4h under hydrogen (15 psi). The mixture was filtered and the filtrate was concentrated in vacuo to give 1- (4,4, 4-trifluorobutyl) pyrazol-4-amine (0.100g, 0.466mmol, 80%) as a pink solid. This material was used in the next step without further purification. LCMS (ESI) M/z 194.1[ M + H ] ]+

And step 3: preparation of 5- (2-furyl) -N- [1- (4, 4, 4-trifluorobutyl) pyrazol-4-yl ] isoxazole-3-carboxamide

To a stirred solution of 1- (4, 4, 4-trifluorobutyl) pyrazol-4-amine (0.090g, 0.466mmol) and 5- (2-furyl) isoxazole-3-carboxylic acid (0.092g, 0.513mmol) in N, N-dimethylformamide (1mL) was added N, N' -tetramethyl-O- (1H-benzotriazol-1-yl) uronium hexafluorophosphate (0.194g, 0.513mmol) and diisopropylethylamine (0.162mL, 0.932mmol) at 15 ℃. The reaction mixture was stirred at 15 ℃ for 3h and then purified by preparative-HPLC (Agela Durashell C18150X 255 μm column; 20% -70% acetonitrile in 0.04% ammonium hydroxide, 12min gradient). Then purified by preparative-HPLC (YMC-Actus ODS-AQ 100X 305 um column; acetonitrile in 0.225% aqueous formic acid 38% -68%, 12min gradient) to give 5- (2-furyl) -N- [1- (4, 4, 4-trifluorobutyl) pyrazol-4-yl as a white solid]Isoxazole-3-carboxamide (0.035g, 0.099mmol, 21%).1H NMR (400MHz, chloroform-d) δ 8.55(s, 1H), 8.06(s, 1H), 7.62-7.56(m, 2H), 6.99(d, J ═ 3.4Hz, 1H), 6.93(s, 1H), 6.58(dd, J ═ 1.8, 3.5Hz, 1H), 4.20(t, J ═ 6.4Hz, 2H), 2.22-2.03(m, 4H); LCMS (ESI) M/z 355.1[ M + H ] ]+

Example 67: preparation of N- [1- (cyclohexylmethyl) pyrazol-4-yl ] -5- (2-furyl) isoxazole-3-carboxamide (26)

Step 1: preparation of 1- (cyclohexylmethyl) -4-nitro-pyrazoles

To a stirred solution of 4-nitro-1H-pyrazole (0.300g, 2.65mmol) and bromomethylcyclohexane (0.564g, 3.18mmol, 0.444mL) in N, N-dimethylformamide (5mL) was added potassium carbonate (0.440g, 3.18mmol) at 15 deg.C, followed by stirring for 16H at 15 deg.C. The reaction mixture was added to ice water (2mL) and extracted with ethyl acetate (10 mL. times.3). The combined organic layers were washed with water (3mL × 3) and brine (3mL), dried over sodium sulfate, filtered and concentrated under reduced pressure to give a residue. The residue was purified by column chromatography (ISCO 10g silica, 0-25% ethyl acetate in petroleum ether, 20min gradient) to give 1- (cyclohexylmethyl) -4-nitro-pyrazole as a white solid (420mg, 1.97mmol, 74%).1H NMR (400MHz, chloroform-d) δ 8.07(d, J ═ 6.1Hz, 2H), 3.96(d, J ═ 7.5Hz, 2H), 1.97 to 1.84(m, 1H), 1.79 to 1.57(m, 5H), 1.31 to 1.10(m, 3H), 1.04 to 0.91(m, 2H); LCMS (ESI) M/z 210.1[ M + H ]]+

Step 2: preparation of 1- (cyclohexylmethyl) pyrazol-4-amine

To a solution of 1- (cyclohexylmethyl) -4-nitro-pyrazole (0.200g, 0.956mmol) in methanol (5mL) under nitrogen was added palladium on charcoal (0.020g, 0.096mmol, 10 wt% Pd). The suspension was purged with hydrogen (3 ×). The mixture was stirred at 15 ℃ for 4h under hydrogen (15 psi). The reaction mixture was filtered and the filtrate was concentrated in vacuo to give 1- (cyclohexane) as a pink solid Ylmethyl) pyrazol-4-amine (0.160g, 0.803mmol, 84%). LCMS (ESI) M/z 180.2[ M + H ]]+. This material was used in the next step without further purification.

And step 3: preparation of N- [1- (cyclohexylmethyl) pyrazol-4-yl ] -5- (2-furyl) isoxazole-3-carboxamide

To a stirred solution of 1- (cyclohexylmethyl) pyrazol-4-amine (0.090g, 0.502mmol) and 5- (2-furyl) isoxazole-3-carboxylic acid (0.090g, 0.502mmol) in N, N-dimethylformamide (1mL) was added N, N' -tetramethyl-O- (1H-benzotriazol-1-yl) uronium hexafluorophosphate (0.190g, 0.502mmol) and diisopropylethylamine (0.175mL, 1.00mmol) at 15 ℃, followed by stirring for 3H at 15 ℃. The reaction mixture was directly purified by preparative-HPLC (Agela Durashell C18150X 255 μm column; 40% -90% acetonitrile in 0.04% ammonium hydroxide, 12min gradient) to give N- [1- (cyclohexylmethyl) pyrazol-4-yl as a white solid]-5- (2-furyl) isoxazole-3-carboxamide (0.055g, 0.160mmol, 32%).1H NMR (400MHz, chloroform-d) δ 8.50(br.s, 1H), 7.99(s, 1H), 7.62-7.53(m, 2H), 6.99(d, J ═ 3.4Hz, 1H), 6.92(s, 1H), 6.57(dd, J ═ 1.7, 3.4Hz, 1H), 3.93(d, J ═ 7.2Hz, 2H), 1.89(ttd, J ═ 3.8, 7.4, 14.8Hz, 1H), 1.76-1.61(m, 5H), 1.30-1.10(m, 3H), 1.05-0.91(m, 2H); LCMS (ESI) M/z 341.2[ M + H ] ]+

EXAMPLE 68. preparation of N- (1- (3, 4-dichlorobenzyl) -3, 5-dimethyl-1H-pyrazol-4-yl) -5- (furan-2-yl) isoxazole-3-carboxamide (3)

Step 1: preparation of N- (1- (3, 4-dichlorobenzyl) -3, 5-dimethyl-1H-pyrazol-4-yl) -5- (furan-2-yl) isoxazole-3-carboxamide

To a mixture of 5- (furan-2-yl) isoxazole-3-carboxylic acid (0.120g, 0.669mmol), N' -tetramethyl-O- (1H-benzotriazol-1-yl) uronium hexafluorophosphate (0.254g, 0.670mmol) and diisopropylethylamine (0.234g, 1.34mmol) in N, N-dimethylformamide (1mL) was added 1- (3, 4-dichlorobenzyl) -3, 5-dimethyl-1H-pyrazol-4-amine (0.199g, 0.737mmol) at 0 ℃. The reaction mixture was stirred at 15 ℃ for 12 h. The residue was purified by preparative-HPLC (column: Luna C8100X 305. mu.; mobile phase: [ water (10mM ammonium bicarbonate) -acetonitrile](ii) a B%: 40% -65%, 12min gradient) to yield N- (1- (3, 4-dichlorobenzyl) -3, 5-dimethyl-1H-pyrazol-4-yl) -5- (furan-2-yl) isoxazole-3-carboxamide as a yellow solid (0.0963g, 0.222mmol, 33%).1H NMR (400MHz, chloroform-d) δ 7.90(s, 1H), 7.61(s, 1H), 7.41(d, J ═ 7.9Hz, 1H), 7.24(s, 1H), 7.00(d, J ═ 3.5Hz, 1H), 6.96(d, J ═ 8.4Hz, 1H), 6.93(s, 1H), 6.59(dd, J ═ 1.8, 3.5Hz, 1H), 5.18(s, 2H), 2.24(s, 3H), 2.15(s, 3H); LCMS (ESI) M/z 431.1[ M + H ] ]+

Example 69.preparation of 5- (oxazol-5-yl) -N- (1- ((4- (trifluoromethyl) pyridin-3-yl) methyl) -1H-pyrazol-4-yl) isoxazole-3-carboxamide (171)

Step 1: preparation of ethyl 5- (hydroxymethyl) isoxazole-3-carboxylate

To a solution of prop-2-yn-1-ol (8.4g, 149.1mmol) in N, N-dimethylformamide (50mL) was added dropwise a solution of ethyl (Z) -2-chloro-2- (hydroxyimino) acetate (7.5g, 49.7mmol) in N, N-dimethylformamide (50mL) under nitrogen over 40 min. After the addition, the reaction mixture was heated to 90 ℃ and added dropwise over 1hA solution of triethylamine (15.0g, 149.1mmol) in N, N-dimethylformamide (50mL) was added. The reaction mixture was heated at 90 ℃ for 17h and cooled to room temperature. The reaction mixture was diluted with ethyl acetate (50mL), washed with water (60mL × 2) and brine (60mL × 2), dried over anhydrous sodium sulfate, filtered and concentrated in vacuo. The residue was purified by column chromatography (silica, petroleum ether/ethyl acetate-6/1) to give ethyl 5- (hydroxymethyl) isoxazole-3-carboxylate (3.35g, 19.6mmol, 39%) as a yellow oil. LCMS (ESI) M/z 172.1[ M + H ]]+

Step 2: preparation of ethyl 5-formylisoxazole-3-carboxylate

To a solution of ethyl 5- (hydroxymethyl) isoxazole-3-carboxylate (1.2g, 7.0mmol) in ethyl acetate (20mL) was added 2-iodoxybenzoic acid (5.9g, 21.0 mmol). The reaction mixture was heated at 90 ℃ for 17h and then cooled to room temperature. The reaction mixture was filtered and the filtrate was concentrated in vacuo. The residue was purified by column chromatography (silica, petroleum ether/ethyl acetate-5/1) to give ethyl 5-formylisoxazole-3-carboxylate (780mg, 4.6mmol, 71%) as a yellow oil.

And step 3: preparation of 5- (oxazol-5-yl) isoxazole-3-carboxylic acids

To a solution of 1- (isocyanomethylsulfonyl) -4-toluene (0.460g, 2.36mmol) in acetonitrile (15mL) was added potassium carbonate (0.391g, 2.83 mmol). The reaction mixture was stirred at 25 ℃ for 1h, then ethyl 5-formylisoxazole-3-carboxylate (0.400g, 2.36mmol) was added at 0 ℃. The reaction mixture was heated at 90 ℃ for 17h and then cooled to room temperature. The reaction mixture was diluted with water (15mL) and the aqueous layer was adjusted to pH 2 with 1N aqueous hydrogen chloride. The aqueous layer was extracted with ethyl acetate (10 mL. times.2). The combined organic layers were washed with brine (10mL × 2), dried over anhydrous sodium sulfate, filtered and concentrated in vacuo to afford 5- (oxazol-5-yl) isoxazole-3-carboxylic acid (140mg, 0.77mmol, 33%) as a white solid. This material was used in the next step without further purification.

And 4, step 4: preparation of 5- (oxazol-5-yl) -N- (1- ((4- (trifluoromethyl) pyridin-3-yl) methyl) -1H-pyrazol-4-yl) isoxazole-3-carboxamide

To a solution of 5- (oxazol-5-yl) isoxazole-3-carboxylic acid (0.100mg, 0.55mmol), 1- ((4- (trifluoromethyl) pyridin-3-yl) methyl) -1H-pyrazol-4-amine (133mg, 0.55mmol) and 4- (4, 6-dimethoxy-1, 3, 5-triazin-2-yl) -4-methylmorpholinium chloride (0.182g, 0.66mmol) in tetrahydrofuran (15mL) was added N-methylmorpholine (167mg, 1.65 mmol). The mixture was stirred at 25 ℃ for 4 h. The reaction mixture was diluted with water (15mL) and extracted with ethyl acetate (10 mL. times.2). The combined organic layers were washed with brine (10mL), dried over sodium sulfate, filtered and concentrated in vacuo. The crude sample was dissolved in a minimal amount of N, N-dimethylformamide and purified via preparative-HPLC (Boston C1821 × 250mm 10 μm column; acetonitrile/0.01% aqueous trifluoroacetic acid) to give 5- (oxazol-5-yl) -N- (1- ((4- (trifluoromethyl) pyridin-3-yl) methyl) -1H-pyrazol-4-yl) isoxazole-3-carboxamide (35.3mg, 0.08mmol, 16%) as a white solid. 1H NMR (400MHz, dimethylsulfoxide-d)6)δ11.19(d,J=31.8Hz,1H),8.81(d,J=5.0Hz,1H),8.73(s,1H),8.31(d,J=23.9Hz,2H),8.02(s,1H),7.80(d,J=5.1Hz,1H),7.74(s,1H),7.35(s,1H),5.61(s,2H);LCMS(ESI)m/z:405.1[M+H]+

Example 70 preparation of N- (1-benzyl-1H-pyrazol-4-yl) -5- (3-methylpyridin-4-yl) isoxazole-3-carboxamide (201)

At 23 deg.CTo a solution of 1-benzyl-1H-pyrazol-4-amine hydrochloride (257mg, 1.23mmol) in 1, 4-dioxane (6mL) was slowly added triethylaluminum (2.0M in toluene, 0.60mL, 1.20mmol) under argon. The mixture was stirred at 23 ℃ for 30min, then ethyl 5- (3-methylpyridin-4-yl) isoxazole-3-carboxylate (65mg, 0.30mmol) in 1, 4-dioxane (2mL) was added. The resulting reaction mixture was heated to 100 ℃ and stirred for 3 h. After cooling to room temperature, the mixture was quenched with 1N aqueous hydrochloric acid (25mL) and extracted with ethyl acetate (50 mL). The organic layer was washed with 0.5N aqueous hydrochloric acid (25mL × 2) and brine (25mL), dried over sodium sulfate, filtered and concentrated in vacuo. The crude material was purified by preparative-HPLC (column: Sunfire preparative C1810 μm OBD 19 x 250 mm; mobile phase: [ water (0.05% trifluoroacetic acid) -acetonitrile](ii) a B%: 60% -88%, 15min) to afford N- (1-benzyl-1H-pyrazol-4-yl) -5- (3-methylpyridin-4-yl) isoxazole-3-carboxamide as a white solid (12mg, 0.03mmol, 11%). 1H NMR (500MHz, methanol-d)4)δ8.94(d,1H),8.55(dd,11.2Hz,3H),7.98(d,1H),7.63(d,1H),7.33-7.04(m,5H),5.21(d,2H),2.62(s,2H),2.30(s,1H);LCMS(ESI)m/z:360.1[M+H]+

EXAMPLE 71 preparation of (S) -N- (1-benzyl-1H-pyrazol-4-yl) -5- (3-methoxypyrrolidin-1-yl) isoxazole-3-carboxamide (189)

To a solution of N- (1-benzyl-1H-pyrazol-4-yl) -5-chloroisoxazole-3-carboxamide (0.1g, 0.331mmol), (R) -3-methoxypyrrolidine hydrochloride (0.136g, 0.993mmol) in N, N-dimethylformamide (3.0mL) was added potassium carbonate (0.068g, 0.497 mmol). The reaction mixture was heated at 90 ℃ for 1h, cooled to room temperature and concentrated in vacuo. The crude sample was dissolved in a minimal amount of N, N-dimethylformamide and purified by preparative-HPLC (Boston C1821X 250mM 10 μm column. mobile phase acetonitrile/10 mM ammonium acetate in water) to give (R) -N- (1-benzyl-1H-pyrazol-4-yl) -5- (3-methoxypyrrolidin-1-yl) isoxazole-3-carboxylic acid as a pale yellow solidAmine (19.0mg, 0.052mmol, 15%).1H NMR (500MHz, dimethylsulfoxide-d)6)δ10.64(s,1H),8.11(s,1H),7.63(s,1H),7.35-7.22(m,5H),5.41(s,1H),5.31(s,2H),4.09-4.08(m,1H),3.52–3.36(m,4H),3.26(s,3H),2.09-2.03(m,2H);LCMS(ESI)m/z:368.2[M+H]+

Example 72.preparation of tert-butyl 4- (3- (1-benzyl-1H-pyrazol-4-ylcarbamoyl) isoxazol-5-yl) piperazine-1-carboxylate (185)

Step 1: preparation of N- (1-benzyl-1H-pyrazol-4-yl) -5- (piperazin-1-yl) isoxazole-3-carboxamide

To a solution of N- (1-benzyl-1H-pyrazol-4-yl) -5-chloroisoxazole-3-carboxamide (0.3g, 0.993mmol) and piperazine (256mg, 2.98mmol) in N, N-dimethylformamide (5mL) was added potassium carbonate (0.205g, 1.44 mmol). The reaction mixture was heated to 90 ℃ and stirred for 1 h. The crude sample was dissolved in a minimal amount of N, N-dimethylformamide and purified by preparative-HPLC (Boston C1821 × 250mM 10 μm column. mobile phase acetonitrile/10 mM aqueous ammonium acetate) to give N- (1-benzyl-1H-pyrazol-4-yl) -5- (piperazin-1-yl) isoxazole-3-carboxamide (130mg, 0.369mmol, 37%) as a white solid. LCMS (ESI) M/z 353.1[ M + H ] ]+

Step 2: preparation of 4- (3- (1-benzyl-1H-pyrazol-4-ylcarbamoyl) isoxazol-5-yl) piperazine-1-carboxylic acid tert-butyl ester

To N- (1-benzyl-1H-pyrazol-4-yl) -5- (piperazin-1-yl) isoxazole-3-carboxamide (60mg, 0.17mmol) and triethylamine (34mg, 0.34mmol) in tetrahydrofuran (5mL)To the solution of (3), di-tert-butyl dicarbonate (37mg, 0.17mmol) was added. The reaction mixture was heated to 60 ℃ and stirred for 1 h. Volatiles were removed in vacuo. The crude sample was dissolved in a minimal amount of N, N-dimethylformamide and purified by preparative-HPLC (Boston C1821 × 250mM 10 μm column. mobile phase acetonitrile/10 mM aqueous ammonium acetate) to give tert-butyl 4- (3- (1-benzyl-1H-pyrazol-4-ylcarbamoyl) isoxazol-5-yl) piperazine-1-carboxylate (51.5mg, 0.114mmol, 66%) as a white solid.1H NMR (500MHz, dimethylsulfoxide-d)6)δ10.69(s,1H),8.10(s,1H),7.63(s,1H),7.35–7.22(m,5H),5.71(s,1H),5.31(s,2H),3.44(d,J=5.5Hz,4H),3.35(d,J=5.5Hz,4H),1.42(s,9H);LCMS(ESI)m/z:453.3[M+H]+

EXAMPLE 73 preparation of (R) -N- (1-benzyl-1H-pyrazol-4-yl) -5- (3-methoxypyrrolidin-1-yl) isoxazole-3-carboxamide (188)

To a solution of N- (1-benzyl-1H-pyrazol-4-yl) -5-chloroisoxazole-3-carboxamide (0.1g, 0.331mmol) and (R) -3-methoxypyrrolidine hydrochloride (113mg, 0.828mmol) in N, N-dimethylformamide (3.0mL) was added potassium carbonate (68mg, 0.497 mmol). The reaction mixture was heated to 90 ℃ and stirred for 1 h. The crude sample was dissolved in a minimal amount of N, N-dimethylformamide and purified by preparative-HPLC (Boston C1821 × 250mM 10 μm column. mobile phase is acetonitrile/10 mM aqueous ammonium acetate) to give (R) -N- (1-benzyl-1H-pyrazol-4-yl) -5- (3-methoxypyrrolidin-1-yl) isoxazole-3-carboxamide (21.1mg, 0.057mmol, 17%) as a pale yellow solid. 1H NMR (500MHz, dimethylsulfoxide-d)6)δ10.64(s,1H),8.11(s,1H),7.63(s,1H),7.35-7.22(m,5H),5.41(s,1H),5.31(s,2H),4.09-4.08(m,1H),3.52–3.36(m,4H),3.26(s,3H),2.08-2.03(m,2H);LCMS(ESI)m/z:368.0[M+H]+

EXAMPLE 74.1 preparation of benzyl-N- [5- (2-furyl) isoxazol-3-yl ] pyrazole-4-carboxamide (49)

Step 1: preparation of 3- (2-furyl) -3-oxo-propionitrile

To a stirred solution of acetonitrile (7.51mL, 1423mmol) and sodium hydride (8.56g, 214mmol, 60% in mineral oil) in tetrahydrofuran (100mL) at 20 ℃ was added dropwise ethyl furan-2-carboxylate (10.0g, 71.4 mmol). The reaction mixture was heated to 80 ℃ for 1 h. The mixture was cooled to 0 ℃ and water (40mL) was added until no air bubbles were generated, then neutralized with 4M aqueous hydrogen chloride to pH 7. The aqueous layer was extracted with ethyl acetate (100 mL. times.3). The combined organic layers were washed with brine (20mL), dried over sodium sulfate, filtered and concentrated in vacuo. The residue was purified by column chromatography (ISCO, 40g silica, 0-30% ethyl acetate in petroleum ether, 20min gradient) to give 3- (2-furyl) -3-oxo-propionitrile (7.5g, 55.5mmol, 78%) as a yellow solid.1H NMR (400MHz, chloroform-d) δ 7.68(d, J ═ 1.1Hz, 1H), 7.40(d, J ═ 3.5Hz, 1H), 6.66(dd, J ═ 1.7, 3.7Hz, 1H), 3.99(s, 2H).

Step 2: preparation of 5- (2-furyl) isoxazol-3-amine

To a solution of 3- (2-furyl) -3-oxo-propionitrile (4.00g, 29.6mmol) in ethanol (60mL) and water (40mL) was added ammonia hydroxide (2.26g, 32.6mmol) and sodium hydroxide (1.42g, 35.5 mmol). The reaction mixture was heated at 90 ℃ for 16 h. The mixture was cooled to 20 ℃ and concentrated hydrogen chloride (4mL) was added to the above solution, and the mixture was heated at 90 ℃ for 2 h. The mixture was basified to pH 8 with 1M lithium hydroxide hydrate and extracted with dichloromethane (20mL × 4). Mixing the organic phaseWashed with brine (10mL × 2), dried over sodium sulfate, filtered and concentrated in vacuo. The crude residue was purified by column chromatography (ISCO, 40g silica, 40-55% ethyl acetate in petroleum ether, 20min gradient) to give 5- (2-furyl) isoxazol-3-amine (1g, 6.10mmol, 21%) as a yellow solid.1H NMR (400MHz, acetone-d6)δ7.75-7.67(m,1H),6.87(d,J=3.4Hz,1H),6.61(dd,J=1.8,3.4Hz,1H),6.13(s,1H),5.32-4.90(m,2H)。

Step 3.1 preparation of benzyl pyrazole-4-carboxylic acid methyl ester

A mixture of methyl 1H-pyrazole-4-carboxylate (1.10g, 8.72mmol), benzyl bromide (1.55mL, 13.1mmol) and cesium carbonate (8.53g, 26.2mmol) in N, N-dimethylformamide (20mL) was stirred at 20 ℃ for 16H. The reaction mixture was diluted with water (30mL) and extracted with dichloromethane (15 mL. times.2). The combined organic layers were washed with brine (20mL × 2), dried over sodium sulfate, filtered and concentrated under reduced pressure. The crude residue was purified by column chromatography (ISCO, 12g silica, 0-30% ethyl acetate in petroleum ether, 15min gradient) to give methyl 1-benzylpyrazole-4-carboxylate (1.0g, 4.62mmol, 53%) as a white solid. 1H NMR (400MHz, acetone-d6)δ7.86(s,1H),7.77(s,1H),7.31-7.25(m,2H),7.18-7.16(m,2H),5.22(s,2H),3.72(ds,1H),6.13(s,3H)。

Step 4. preparation of methyl 1-benzylpyrazole-4-carboxylic acid

To a solution of methyl 1-benzylpyrazole-4-carboxylate (1.00g, 4.62mmol) in tetrahydrofuran (20mL) was added sodium hydroxide (2M, 4.6 mL). The reaction mixture was stirred at 20 ℃ for 16 h. The reaction mixture was adjusted to pH 3 with 2M aqueous hydrogen chloride solution and the aqueous phase was extracted with ethyl acetate (10mL × 2). The combined organic layers were washed with brine (10mL × 2), dried over sodium sulfate, filtered and concentrated under reduced pressure to give 1-benzylpyrazole-4-carboxylic acid (0.600g, 2.97mmol, 64%) as a white solid. This material was used in the next step without further purification.

And 5: preparation of 1-benzyl-N- [5- (2-furyl) isoxazol-3-yl ] pyrazole-4-carboxamide

To a solution of 1-benzylpyrazole-4-carboxylic acid (0.242g, 1.20mmol) in dichloromethane (3mL) at 0 ℃ was added oxalyl chloride (0.26mL, 3.00mmol) dropwise, followed by 1 drop of anhydrous N, N-dimethylformamide. The reaction mixture was stirred at 20 ℃ for 30 min. The reaction mixture was concentrated in vacuo, dissolved in dichloromethane (1mL) and added slowly to a solution of 5- (2-furyl) isoxazol-3-amine (150mg, 0.999mmol) and pyridine (158mg, 2.00mmol) in dichloromethane (3 mL). The reaction mixture was stirred at 15 ℃ for 5h, then filtered and purified by column chromatography (ISCO, 12g silica, 40-60% ethyl acetate in petroleum ether, 20min gradient) to give 1-benzyl-N- [5- (2-furyl) isoxazol-3-yl as a white solid ]Pyrazole-4-carboxamide (0.100g, 0.298mmol, 30%).1H NMR (400MHz, dimethylsulfoxide-d)6)δ11.22(s,1H),8.56(s,1H),8.16(s,1H),7.96(d,J=1.2Hz,1H),7.45-7.26(m,5H),7.23-7.15(m,2H),6.74(dd,J=1.8,3.5Hz,1H),5.40(s,2H);LCMS(ESI)m/z:335.1[M+H]+

Example 75 preparation of N- (1-benzyl-1H-pyrazol-4-yl) -3-isopropylisoxazole-5-carboxamide (158)

Step 1: preparation of 3- (ethoxycarbonyl) isoxazole-5-carboxylic acid

A solution of ethyl (Z) -2-chloro-2- (hydroxyimino) acetate (3.36g, 22.3mmol) in N, N-dimethylformamide (10mL) was slowly added to a solution of propiolic acid (3.9g, 55.7mmol) in N, N-dimethylformamide (10mL) under nitrogen. The reaction mixture was heated to 90 ℃ and triethylamine (6.76g, 66.9mmol) was slowly added to the mixture. The solution was then heated at 90 ℃ for 12 h. The volatiles were removed under reduced pressure and the crude residue was extracted with ethyl acetate (40 mL). The aqueous layer was adjusted to pH 3-5 with 1N aqueous hydrochloric acid, and then extracted with dichloromethane (50mL × 2). The dichloromethane layer was dried over sodium sulfate, filtered and concentrated in vacuo to give 3- (ethoxycarbonyl) isoxazole-5-carboxylic acid (4.5g, crude) as a brown oil. LCMS (ESI) M/z 186.1[ M + H ]]+. This material was used in the next step without further purification.

Step 2: preparation of ethyl 5- (1-benzyl-1H-pyrazol-4-ylcarbamoyl) isoxazole-3-carboxylate

To a solution of 3- (ethoxycarbonyl) isoxazole-5-carboxylic acid (0.6g, 3.24mmol) in diisopropylethylamine (1.26g, 9.72mmol) in tetrahydrofuran (20mL) at 20 deg.C was added 1- [ bis (dimethylamino) methylene chloride]-1H-1, 2, 3-triazolo [4, 5-b]Pyridinium 3-oxide hexafluorophosphate (1.86g, 4.91 mmol). The reaction mixture was stirred at 20 ℃ for 30min, then 1-benzyl-1H-pyrazol-4-amine (0.561g, 3.24mmol) was added. The reaction mixture was stirred at 20 ℃ for 12 h. Volatiles were removed in vacuo. The crude product was purified by column chromatography (silica, petroleum ether/ethyl acetate 1/1) to give ethyl 5- (1-benzyl-1H-pyrazol-4-ylcarbamoyl) isoxazole-3-carboxylate (0.300g, 0.882mmol, 27%) as a white solid. LCMS (ESI) M/z 341.1[ M + H ]]+

And step 3: preparation of ethyl 5- (1-benzyl-1H-pyrazol-4-ylcarbamoyl) isoxazole-3-carboxylate

To a solution of ethyl 5- (1-benzyl-1H-pyrazol-4-ylcarbamoyl) isoxazole-3-carboxylate (0.200g, 0.588mmol) in tetrahydrofuran (10mL) under nitrogen at 0 ℃ was added methylmagnesium bromide (1mL, 2.94 mmol). The reaction mixture was quenched with aqueous ammonium chloride (10mL) and extracted with dichloromethane (100 mL). The organic layer was dried over sodium sulfate, filtered and concentrated in vacuo. The crude sample was dissolved in a minimal amount of N, N-dimethylformamide and purified via preparative-HPLC (Boston C1821 × 250mm 10 μm column; acetonitrile/0.01% aqueous trifluoroacetic acid) to give ethyl 5- (1-benzyl-1H-pyrazol-4-ylcarbamoyl) isoxazole-3-carboxylate (72.7mg, 0.223mmol, 25%) as a white solid. LCMS (ESI) M/z 327.2[ M + H ]+

And 4, step 4: preparation of N- (1-benzyl-1H-pyrazol-4-yl) -3- (prop-1-en-2-yl) isoxazole-5-carboxamide

To a solution of ethyl (Z) -2-chloro-2- (hydroxyimino) acetate (40mg, 0.123mmol) in dichloromethane (10mL) was added methanesulfonic acid (0.5 mL). The reaction mixture was refluxed for 2 h. Volatiles were removed in vacuo. The crude sample was dissolved in a minimal amount of N, N-dimethylformamide and purified via preparative-HPLC (Boston C1821 × 250mm 10 μm column; acetonitrile/0.01% aqueous trifluoroacetic acid) to give N- (1-benzyl-1H-pyrazol-4-yl) -3- (prop-1-en-2-yl) isoxazole-5-carboxamide (22.2mg, 0.072mmol, 58%) as a white solid.1H NMR (500MHz, dimethylsulfoxide-d)6)δ11.07(s,1H),8.16(s,1H),7.65(s,1H),7.50(s,1H),7.36-7.23(m,5H),5.88(s,1H),5.52(s,1H),5.33(s,2H),2.11(s,3H);LCMS(ESI)m/z:309.1[M+H]+

And 5: preparation of N- (1-benzyl-1H-pyrazol-4-yl) -3- (prop-1-en-2-yl) isoxazole-5-carboxamide

To a solution of N- (1-benzyl-1H-pyrazol-4-yl) -3- (prop-1-en-2-yl) isoxazole-5-carboxamide (80mg, 0.26mmol) in methanol (10mL) under nitrogen was added palladium on charcoal (8mg, 10 wt% Pd). The reaction mixture was stirred under a hydrogen balloon at 20C for 1 h. Volatiles were removed in vacuo. The crude sample was dissolved in a minimal amount of N, N-dimethylformamide and purified via preparative-HPLC (Boston C1821 × 250mm 10 μm column; acetonitrile/0.01% aqueous trifluoroacetic acid) to give N- (1-benzyl-1H-pyrazol-4-yl) -3- (prop-1-en-2-yl) isoxazole-5-carboxamide (14.5mg, 0.047mmol, 18%) as a white solid. 1H NMR (500MHz, dimethylsulfoxide-d)6)δ11.00(s,1H),8.15(s,1H),7.64(s,1H),7.36-7.23(m,5H),7.14(s,1H),5.33(s,2H),3.11-3.06(m,1H),1.26(d,J=7.0Hz,6H);LCMS(ESI)m/z:311.2[M+H]+

Example 76: preparation of N- (1-benzylpyrazol-4-yl) -3- (2-furyl) isoxazole-5-carboxamide (11)

Step 1: preparation of 1-benzyl-4-nitro-pyrazoles

To a stirred solution of 4-nitro-1H-pyrazole (2.00g, 17.7mmol) in N, N-dimethylformamide (15mL) at 0 deg.C was added sodium hydride (0.778g, 19.0mmol, 60% purity by weight in mineral oil). The reaction mixture was stirred at 15 ℃ for 1h, then cooled to 0 ℃ and benzyl bromide (3.03g, 17.7mmol, 2.1mL) was added. The reaction mixture was warmed to 15 ℃ and stirred for 15 h. The reaction mixture was added to ice water (5mL) and extracted with ethyl acetate (15 mL. times.3). The combined organic layers were washed with water (10 mL. times.2), thenThen washed with brine (5mL), dried over sodium sulfate, filtered and concentrated under reduced pressure. The crude residue was purified by column chromatography (ISCO, 20g silica, 0-30% ethyl acetate in petroleum ether, 20min gradient) to give 1-benzyl-4-nitro-pyrazole as a white solid (2.80g, 13.8mmol, 78%).1H NMR (400MHz, chloroform-d) Δ 8.10(s, 1H), 8.04(s, 1H), 7.45-7.39(M, 3H), 7.32-7.28(M, 2H), 5.31(s, 2H), LCMS (ESI) M/z 204.1[ M + H ] ]+

Step 2: preparation of 1-benzylpyrazole-4-amine

To a solution of 1-benzyl-4-nitro-pyrazole (1.50g, 7.38mmol) in methanol (10mL) under nitrogen was added palladium on activated carbon (10 wt% Pd, 0.500 g). The suspension was evacuated and purged several times with hydrogen. The mixture was stirred under hydrogen at 15 ℃ for 16 h. The reaction mixture was filtered and the filtrate was concentrated in vacuo to give 1-benzylpyrazol-4-amine (1.10g, 6.35mmol, 86%) as a pink solid. LCMS (ESI) M/z 174.1[ M + H ]]+. This material was used in the next step without further purification.

And step 3: preparation of N- (1-benzylpyrazol-4-yl) -3- (2-furyl) isoxazole-5-carboxamide

To a stirred solution of 1-benzylpyrazol-4-amine (0.120g, 0.693mmol) and 3- (furan-2-yl) isoxazole-5-carboxylic acid (0.157g, 0.831mmol) in N, N-dimethylformamide (1mL) was added N, N' -tetramethyl-O- (1H-benzotriazol-1-yl) uronium hexafluorophosphate (0.315g, 0.831mmol) and diisopropylethylamine (0.179mg, 1.39mmol) at 15 ℃. The reaction mixture was stirred at 15 ℃ for 3 h. After completion of the reaction, the reaction mixture was passed through preparative-HPLC (Agela Venusil XBP C18150X 255 um column; 50-75% acetonitrile in 10mM 0.04% ammonium hydroxide, 10min gradient) Purification gave N- (1-benzylpyrazol-4-yl) -3- (2-furyl) isoxazole-5-carboxamide (0.070g, 0.209mmol, 36%) as a white solid.1H NMR (400MHz, chloroform-d) δ 8.55(br.s, 1H), 8.04(s, 1H), 7.65-7.59(m, 2H), 7.40-7.32(m, 3H), 7.30-7.26(m, 2H), 7.00(d, J ═ 3.4Hz, 1H), 6.92(s, 1H), 6.59(dd, J ═ 1.8, 3.5Hz, 1H), 5.32(s, 2H); LCMS (ESI) M/z 335.1[ M + H ]]+

EXAMPLE 77 preparation of (5- (5-fluoropyridin-2-yl) isoxazol-3-yl) (3-phenylazetidin-1-yl) methanone (243)

Step 1: preparation of 5-fluoro-2- ((trimethylsilyl) ethynyl) pyridine

2-bromo-5-fluoropyridine (4.9g, 27.8mmol), ethynyltrimethylsilane (3.27g, 33.4mmol) and [1, 1' -bis (diphenylphosphino) ferrocene]A mixture of palladium (II) dichloride (2.03g, 2.78mmol) and cuprous iodide (1.06g, 5.57mmol) in triethylamine (100mL) was heated at 90 ℃ for 2 h. The mixture was diluted with brine (100mL), extracted with ethyl acetate (100mL × 2) and purified by column chromatography (silica, petroleum ether/ethyl acetate ═ 20/1) to give 5-fluoro-2- ((trimethylsilyl) ethynyl) pyridine as a yellow oil (1.5g, 7.7mmol, 28%). LCMS (ESI) M/z 194.2[ M + Na ] ]+

Step 2: preparation of 2-ethynyl-5-fluoropyridine

A mixture of 5-fluoro-2- ((trimethylsilyl) ethynyl) pyridine (3.5g, 18.1mmol) and potassium hydroxide (1.02g, 18.1mmol) in methanol (100mL) was stirred at 20 ℃ for 5 h. The mixture is mixed with brineDiluted (100mL), extracted with ethyl acetate (100mL × 2) and purified by column chromatography (silica, petroleum ether/ethyl acetate 20/1) to give 2-ethynyl-5-fluoropyridine (1.75g, 14.5mmol, 80%) as a yellow oil. LCMS (ESI) M/z 122.0[ M + H ]]+

And step 3: preparation of 5- (5-fluoropyridin-2-yl) isoxazole-3-carboxylic acid ethyl ester

To a solution of 2-ethynyl-5-fluoropyridine (1.5g, 12.4mmol) in N, N-dimethylformamide (20mL) was added ethyl (Z) -2-chloro-2- (hydroxyimino) acetate (2.81g, 18.6mmol) at 23 ℃. The reaction mixture was stirred for 1h, then triethylamine (1.88g, 18.6mmol) was added. The mixture was heated at 90 ℃ for 16 h. The mixture was diluted with brine (100mL), extracted with ethyl acetate (100mL × 2) and purified by column chromatography (silica, petroleum ether/ethyl acetate ═ 20/1) to give ethyl 5- (5-fluoropyridin-2-yl) isoxazole-3-carboxylate (300mg, 1.27mmol, 10%) as a grey oil. LCMS (ESI) M/z 237.1[ M + H ] ]+

And 4, step 4: preparation of 5- (5-fluoropyridin-2-yl) isoxazole-3-carboxylic acid

A mixture of ethyl 5- (5-fluoropyridin-2-yl) isoxazole-3-carboxylate (0.2g, 0.85mmol) and lithium hydroxide (1.7mL, 3.39mmol, aq. 2M) in ethanol (1.7mL) was stirred at 20 ℃ for 2 h. The mixture was extracted with ethyl acetate and concentrated in vacuo to give crude 5- (5-fluoropyridin-2-yl) isoxazole-3-carboxylic acid (1.3g, 0.63mmol, 73%) as a grey solid. LCMS (ESI) M/z 209.1[ M + H ]]+. This material was used in the next step without further purification.

And 5: preparation of (5- (5-fluoropyridin-2-yl) isoxazol-3-yl) (3-phenylazetidin-1-yl) methanone

To a solution of 5- (5-fluoropyridin-2-yl) isoxazole-3-carboxylic acid (0.1g, 0.48mmol) and N, N-dimethylformamide (0.001g, 0.01mmol) in dichloromethane (5mL) at 0 ℃ was added oxalyl chloride (0.106g, 1.44 mmol). The reaction mixture was stirred at 23 ℃ for 1 h. The reaction mixture was concentrated in vacuo. The crude residue was diluted with dichloromethane (5mL) and pyridine (0.19g, 2.4mmol) before 3-phenylazetidine (0.122g, 0.72mmol) was added. The reaction mixture was stirred for 2 h. The reaction mixture was extracted with ethyl acetate (20 mL. times.2) and washed with water (30mL) and brine (30 mL). The combined organic phases were dried over anhydrous sodium sulfate, filtered and concentrated in vacuo. The crude sample was dissolved in a minimal amount of N, N-dimethylformamide and purified via preparative-HPLC (Boston C1821 × 250mm 10 μm column; acetonitrile/0.01% aqueous trifluoroacetic acid) to give (5- (5-fluoropyridin-2-yl) isoxazol-3-yl) (3-phenylazetidin-1-yl) methanone (0.033g, 0.1mmol, 21%) as a white solid. 1H NMR (500MHz, dimethylsulfoxide-d)6)δ8.77(d,J=2.8Hz,1H),8.17(dd,J=8.8,4.4Hz,1H),7.97(td,J=8.7,2.9Hz,1H),7.43(m,2H),7.41-7.35(m,3H),7.28(t,J=7.2Hz,1H),4.92(t,J=9.2Hz,1H),4.57-4.46(m,2H),4.10(dd,J=10.0,6.5Hz,1H),4.06-3.98(m,1H)。LCMS(ESI)m/z:324.1[M+H]+

EXAMPLE 78 preparation of (5- (6-methoxypyridazin-3-yl) isoxazol-3-yl) (3-phenylazetidin-1-yl) methanone (255)

Step 1: preparation of ethyl 5- (6-methoxypyridazin-3-yl) isoxazole-3-carboxylate

The reaction vial was charged with 3-bromo-6-methoxypyridazine (58.7mg, 0.311mmol), cesium fluoride (85.9mg, 0.566mmol), Pd-PEPSI-IPent catalyst (17.8mg, 0.023mmol), crushed 3A molecular sieve (100mg) and ethyl 5- (tributylstannyl) -1, 2-oxazole-3-carboxylate (0.122g, 0.283 mmol). The vial was sealed, purged, and placed under an argon atmosphere, then 1, 4-dioxane (1.5mL) was added. The reaction mixture was heated to 80 ℃ and stirred for 5 h. The reaction mixture was cooled to room temperature, filtered, and concentrated in vacuo. The crude material was purified by column chromatography (ISCO, 4g silica, eluting with 0% to 30% ethyl acetate in hexanes with 2% triethylamine) to give ethyl 5- (6-methoxypyridazin-3-yl) isoxazole-3-carboxylate (46.9mg, 0.190mmol, 67%) as a yellow solid.1H NMR (300MHz, dimethylsulfoxide-d)6)δ8.29(d,J=9.3Hz,1H),7.63(s,1H),7.47(d,J=9.2Hz,1H),4.42(q,J=7.1Hz,2H),4.13(s,3H),1.35(t,J=7.1Hz,3H)。

Step 2: preparation of 5- (6-methoxypyridazin-3-yl) isoxazole-3-carboxylic acid

To a solution of ethyl 5- (6-methoxypyridazin-3-yl) -1, 2-oxazole-3-carboxylate (66.5mg, 0.266mmol) in ethanol/THF/water (1:4:8, 3.25mL) was added lithium hydroxide monohydrate (55.8mg, 1.33mmol) at 25 ℃ in one portion. The reaction mixture was stirred at 25 ℃ for 18 h. The reaction was cooled in an ice bath and quenched with 1N aqueous hydrogen chloride (1.5 mL). The volume of the reaction mixture was doubled with water and the volatiles were concentrated in vacuo. The residual aqueous layer was extracted with ethyl acetate (20 mL. times.3). The combined organics were washed with brine (5mL) and concentrated in vacuo. Crude 5- (6-methoxypyridazin-3-yl) isoxazole-3-carboxylic acid (53.0mg, 0.239mmol, 90%) was obtained as a pale yellow solid. This material was used in the next step without further purification. 1H NMR (300MHz, dimethylsulfoxide-d)6)δ8.26(d,J=9.2Hz,1H),7.54(s,1H),7.46(d,J=9.3Hz,1H),4.12(s,3H);LCMS(ESI)m/z:222.1[M+H]+

And step 3: preparation of (5- (6-methoxypyridazin-3-yl) isoxazol-3-yl) (3-phenylazetidin-1-yl) methanone

To a suspension of 5- (6-methoxypyridazin-3-yl) -1, 2-oxazole-3-carboxylic acid (25mg, 0.113mmol), N' -tetramethyl-O- (1H-benzotriazol-1-yl) uronium hexafluorophosphate (0.192g, 0.508mmol) and diisopropylethylamine (58.4mg, 0.452mmol) in N, N-dimethylformamide/dichloromethane (1/5, 1.2mL) was added 3-phenylazetidine hydrochloride (28.6mg, 0.169mmol) at 25 ℃. The reaction mixture was stirred at room temperature for 24 h. The reaction was poured into ethyl acetate (20mL) and water (2 mL). The aqueous phase was removed and the organic layer was concentrated in vacuo. The crude oil was purified by column chromatography (ISCO, 4g silica, eluting with 0% to 50% ethyl acetate in hexanes) to give (5- (6-methoxypyridazin-3-yl) isoxazol-3-yl) (3-phenylazetidin-1-yl) methanone as a white solid (12.1mg, 0.036mmol, 32%).1H NMR (300MHz, dimethylsulfoxide-d)6)δ8.27(dd,J=9.3,1.0Hz,1H),7.53(d,J=0.9Hz,1H),7.51-7.35(m,5H),7.35-7.21(m,1H),4.93(t,J=9.2Hz,1H),4.52(td,J=11.2,9.9,7.5Hz,2H),4.12(s,3H),4.10-3.93(m,3H);LCMS(ESI)m/z:337.2[M+H]+

EXAMPLE 79 preparation of (3-Phenylazetidin-1-yl) (5- (pyrazin-2-yl) isoxazol-3-yl) methanone (259)

Step 1: preparation of 5- (pyrazin-2-yl) isoxazole-3-carboxylic acid ethyl ester

The reaction vial was charged with 2-bromopyrazine (50mg,0.314mmol), lithium chloride (13mg,0.314mmol), Pd-PEPSI-IPent catalyst (19.8mg,0.025mmol), and toluene (2 mL). The vial was sealed, purged, and placed under argon and ethyl 5- (tributylstannyl) -1, 2-oxazole-3-carboxylate (148mg,0.345mmol) was added. The reaction mixture was heated in a microwave reactor at 130 ℃ for 2 h. The reaction mixture was cooled to room temperature, filtered, and concentrated in vacuo. The crude material was purified by column chromatography (ISCO, 4g silica, eluting with 0% to 40% ethyl acetate in hexanes) to give 5- (pyrazin-2-yl) isoxazole-3-carboxylic acid ethyl ester (0.035g,0.126mmol, 40%).

Step 2: preparation of 5- (pyrazin-2-yl) isoxazole-3-carboxylic acids

To a solution of ethyl 5- (pyrazin-2-yl) -1, 2-oxazole-3-carboxylate (35.9mg,0.126mmol) in ethanol (0.050mL) and THF (0.50mL) was added lithium hydroxide monohydrate (34.1mg,0.815mmol) in one portion followed by water (1 mL). The reaction mixture was stirred at room temperature for 18 h. The reaction was cooled in an ice bath, diluted with water (2mL), and quenched with 1M aqueous hydrogen chloride (0.90 mL). All volatiles were removed in vacuo, the residual aqueous layer was extracted with ethyl acetate 5 × (2mL), and the combined organics were concentrated in vacuo to give 5- (pyrazin-2-yl) -1, 2-oxazole-3-carboxylic acid (26.1mg,0.130mmol, 84%) as a yellow solid. This material was used in the next step without further purification. 1H NMR (300MHz, dimethylsulfoxide-d)6)δ9.33(d,J=1.5Hz,1H),8.83(dd,J=2.5,1.5Hz,1H),8.80(d,J=2.5Hz,1H),7.62(s,1H);LCMS(ESI)m/z:192.1[M+H]+

And step 3: preparation of (3-phenylazetidin-1-yl) (5- (pyrazin-2-yl) isoxazol-3-yl) methanones

To a suspension of 5- (pyrazin-2-yl) -1, 2-oxazole-3-carboxylic acid (25.0mg, 0.130mmol), N' -tetramethyl-O- (1H-benzotriazol-1-yl) uronium hexafluorophosphate (73.5mg, 0.194mmol) and diisopropylethylamine (50.2mg, 0.389mmol) in N, N-dimethylformamide/dichloromethane (1/5, 1.2mL) was added 3-phenylazetidine hydrochloride (24.2mg, 0.143 mmol). The reaction mixture was stirred at room temperature for 8 h. The reaction was poured into ethyl acetate (20mL), washed with water (2mL), 1M aqueous sodium hydroxide (2mL × 2), brine (2mL) and concentrated in vacuo. The crude product was purified by column chromatography (ISCO, 4g silica, eluting with 0% to 50% ethyl acetate in hexanes) to afford (3-phenylazetidin-1-yl) (5- (pyrazin-2-yl) isoxazol-3-yl) methanone as an off-white solid (9.0mg, 0.0299mmol, 23%).1H NMR (300MHz, dimethylsulfoxide-d)6)δ9.34(d,J=1.5Hz,1H),8.84(dd,J=2.5,1.5Hz,1H),8.80(d,J=2.5Hz,1H),7.61(s,1H),7.49-7.24(m,5H),4.93(t,J=9.2Hz,1H),4.60-4.46(m,2H),4.14-3.95(m,2H);LCMS(ESI)m/z:307.1[M+H]+

EXAMPLE 80 preparation of (3-Phenylazetidin-1-yl) (5- (pyrimidin-4-yl) isoxazol-3-yl) methanone (257)

Step 1: preparation of 1- (3- (3-phenylazetidin-1-carbonyl) isoxazol-5-yl) ethan-1-one

To a suspension of 5-acetyl-1, 2-oxazole-3-carboxylic acid (50mg, 0.322mmol), N' -tetramethyl-O- (1H-benzotriazol-1-yl) uronium hexafluorophosphate (170mg, 0.448mmol) and diisopropylethylamine (124mg, 0.965mmol) in N, N-dimethylformamide/dichloromethane (1/5, 1.2mL) was added 3-phenylazetidine hydrochloride (65.4mg, 0.386mmol) in one portion. The reaction mixture was stirred at room temperatureStirring for 24 h. The reaction was poured into ethyl acetate (20mL) and water (2 mL). The aqueous phase was removed and the organic layer was concentrated in vacuo. The crude material was purified by column chromatography (ISCO, 4g silica, eluting with 0% to 50% ethyl acetate in hexanes) to give 1- (3- (3-phenylazetidin-1-carbonyl) isoxazol-5-yl) ethan-1-one (20mg, 0.0739mmol, 23%).1H NMR (300MHz, dimethylsulfoxide-d)6)δ7.65(s,1H),7.45-7.34(m,4H),7.32-7.25(m,1H),4.95-4.82(m,1H),4.56-4.43(m,2H),4.17-3.93(m,2H),2.60(s,3H);LCMS(ESI)m/z:271.0[M+H]+

Step 2: (E) preparation of (E) -3- (dimethylamino) -1- (3- (3-phenylazetidin-1-carbonyl) isoxazol-5-yl) prop-2-en-1-one

A solution of 1- (3- (3-phenylazetidin-1-carbonyl) isoxazol-5-yl) ethan-1-one (0.026g, 0.0961mmol), N-dimethoxymethyl-N, N-dimethylamine (0.028mL, 0.192 mmol) in toluene (1mL) was heated at 110 ℃ for 24 h. 1H NMR (300MHz, dimethylsulfoxide-d)6)δ7.85(d,J=12.1Hz,1H),7.45-7.36(m,4H),7.35(s,2H),7.31-7.22(m,2H),5.80(d,J=12.1Hz,1H),4.88(t,J=9.2Hz,1H),4.55-4.41(m,2H),4.12-3.94(m,2H),3.20(s,3H),2.96(s,3H);LCMS(ESI)m/z:326.2[M+H]+

And step 3: preparation of (3-phenylazetidin-1-yl) (5- (pyrimidin-4-yl) isoxazol-3-yl) methanones

Reacting (E) -3- (dimethyl)A mixture of amino) -1- (3- (3-phenylazetidin-1-carbonyl) isoxazol-5-yl) prop-2-en-1-one (30.5mg, 0.0952mmol), formamidine acetate (30mg, 0.285mmol), potassium carbonate (39mg, 0.285mmol) in N, N-dimethylformamide (0.5mL) was heated at 100 ℃ for 8 h. The reaction mixture was cooled to room temperature, diluted with ethyl acetate (10mL), filtered and concentrated in vacuo. The crude residue was suspended in hot heptane/ethyl acetate (1/1, 2-3mL) and allowed to stand at room temperature for 24 h. The yellow crystals were filtered to give (3-phenylazetidin-1-yl) (5- (pyrimidin-4-yl) isoxazol-3-yl) methanone (10mg, 0.031mmol, 33%).1H NMR (300MHz, dimethylsulfoxide-d)6)δ9.38(d,J=1.4Hz,1H),9.07(d,J=5.3Hz,1H),8.16(dd,J=5.2,1.5Hz,1H),7.69(s,1H),7.50-7.32(m,4H),7.32-7.21(m,1H),4.92(t,J=9.2Hz,1H),4.59-4.46(m,2H),4.15-3.93(m,2H);LCMS(ESI)m/z:307.2[M+H]+

EXAMPLE 81 preparation of (3-Benzylethetidin-1-yl) (5- (pyrimidin-4-yl) isoxazol-3-yl) methanone (258)

Step 1: preparation of 1- [3- (3-benzylazetidine-1-carbonyl) -1, 2-oxazol-5-yl ] ethan-1-one

To a suspension of 5-acetyl-1, 2-oxazole-3-carboxylic acid (0.100g, 0.644mmol), 3-benzyl azetidine hydrochloride (0.141g, 0.772mmol), 1H-1, 2, 3-benzotriazol-1-ol (0.182g, 1.34mmol) and (3N- (3-dimethylaminopropyl) -N' -ethylcarbodiimide hydrochloride (0.184g, 0.965mmol) in N, N-dimethylformamide (1mL) was added diisopropylethylamine (0.124g, 0.965mmol), the reaction mixture was stirred for 48H, the reaction mixture was diluted with ethyl acetate (20mL), washed with water (2mL) and concentrated in vacuo, the crude product was passed through column chromatography (ISCO, 4g silica, with silica in vacuo 0% to 50% ethyl acetate in hexane) to yield 1- [3- (3-benzylazetidine-1-carbonyl) -1, 2-oxazol-5-yl ] -1- [3- (3-benzylazetidine-1-carbonyl) -1, 2-oxazol-5-yl ester]Ethan-1-one (36.1mg, 0.126mmol, 20%)1H NMR (300MHz, dimethylsulfoxide-d)6)δ7.59(s,1H),7.32-7.20(m,5H),4.49(t,J=8.6Hz,1H),4.20-4.08(m,2H),3.81(dd,J=10.0,5.0Hz,1H),3.01-2.91(m,1H),2.95(s,2H),2.59(s,3H);LCMS(ESI)m/z:285.2[M+H]+

Step 2: (E) preparation of (E) -1- (3- (3-benzylazetidine-1-carbonyl) isoxazol-5-yl) -3- (dimethylamino) prop-2-en-1-one

A solution of 1- [3- (3-benzylazetidine-1-carbonyl) -1, 2-oxazol-5-yl ] ethan-1-one (0.032g, 0.112mmol), N-dimethoxymethyl-N, N-dimethylamine (0.030mL, 0.224mmol) in toluene (1mL) was heated at 110 ℃ for 24 h. The reaction mixture was cooled to room temperature and concentrated in vacuo. Crude (E) -1- (3- (3-benzylazetidine-1-carbonyl) isoxazol-5-yl) -3- (dimethylamino) prop-2-en-1-one (30mg, 0.0886mmol) was obtained as a red oil. This material was used in the next step without further purification.

And step 3: preparation of (3-benzyl azetidin-1-yl) (5- (pyrimidin-4-yl) isoxazol-3-yl) methanones

A mixture of (E) -1- (3- (3-benzylazetidine-1-carbonyl) isoxazol-5-yl) -3- (dimethylamino) prop-2-en-1-one (29mg, 0.0854mmol), formamidine acetate (26.6mg, 0.256mmol), potassium carbonate (35.3mg, 0.256mmol) in N, N-dimethylformamide (0.5mL) was heated at 100 ℃ for 8 h. The reaction mixture was cooled to room temperature, diluted with ethyl acetate (10mL), filtered and concentrated in vacuo. The crude residue was purified by column chromatography (ISCO, 4g silica, using 0% to 50% acetic acid in hexanes) Ethyl ester elution) to afford (3-benzyl azetidin-1-yl) (5- (pyrimidin-4-yl) isoxazol-3-yl) methanone as an off-white solid (15mg, 0.0468mmol, 55%).1H NMR (300MHz, dimethylsulfoxide-d)6)δ9.37(d,J=1.4Hz,1H),9.06(d,J=5.2Hz,1H),8.14(dd,J=5.2,1.5Hz,1H),7.64(s,1H),7.39-7.15(m,5H),4.52(t,J=8.6Hz,1H),4.27-4.09(m,2H),3.83(dd,J=10.2,4.9Hz,1H),3.03-2.93(m,2H),2.96(s,2H);LCMS(ESI)m/z:321.1[M+H]+

EXAMPLE 82. preparation of N-methyl-4- (3- (3-phenylazetidin-1-carbonyl) isoxazol-5-yl) benzamide (231)

The synthesis of N-methyl-4- (3- (3-phenylazetidin-1-carbonyl) isoxazol-5-yl) benzamide was carried out following the same procedure as in example 24. The compound N-methyl-4- (3- (3-phenylazetidin-1-carbonyl) isoxazol-5-yl) benzamide (25.9mg, 0.07mmol, 22%) was obtained as a white solid.1H NMR (500MHz, dimethylsulfoxide-d 6) δ 8.61(d, J ═ 9.0Hz, 4.0Hz, 1H), 8.05(d, J ═ 8.5Hz, 2H), 7.99(d, J ═ 8.5Hz, 2H), 7.52(s, 1H), 7.44-7.27(m, 5H), 4.92(t, J ═ 9.0Hz, 1H), 4.55-4.48(m, 2H), 4.12-4.08(m, 1H), 4.04-3.98(m, 1H), 2.81(d, J ═ 4.5Hz, 3H); LCMS (ESI) M/z 362.2[ M + H ]]+

EXAMPLE 83.5- (3, 4-Dimethoxyphenyl) isoxazol-3-yl) (3- (3-fluorophenyl) azetidin-1-yl) methanone (236) preparation

Step 1: preparation of ethyl 5- (3, 4-dimethoxyphenyl) isoxazole-3-carboxylate

The synthesis of ethyl 5- (3, 4-dimethoxyphenyl) isoxazole-3-carboxylate was carried out following the same procedure as in example 24. The compound ethyl 5- (3, 4-dimethoxyphenyl) isoxazole-3-carboxylate (230mg, 0.83mmol, 46%) was obtained as a yellow solid. LCMS (ESI) M/z 277/1[ M + H ]]+

Step 2: preparation of 5- (3, 4-dimethoxyphenyl) isoxazole-3-carboxylic acid

The synthesis of 5- (3, 4-dimethoxyphenyl) isoxazole-3-carboxylic acid was carried out following the same procedure as in example 24. The compound 5- (3, 4-dimethoxyphenyl) isoxazole-3-carboxylic acid (90mg, 0.36mmol, 43%) was obtained as a white solid. LCMS (ESI) M/z 250.2[ M + H ]]+

And step 3: preparation of (5- (3, 4-dimethoxyphenyl) isoxazol-3-yl) (3- (3-fluorophenyl) azetidin-1-yl) methanone

The synthesis of (5- (3, 4-dimethoxyphenyl) isoxazol-3-yl) (3- (3-fluorophenyl) azetidin-1-yl) methanone was carried out following the same procedure as example 24. The compound (5- (3, 4-dimethoxyphenyl) isoxazol-3-yl) (3- (3-fluorophenyl) azetidin-1-yl) methanone (60.6mg, 0.16mmol, 44%) was obtained as a white solid.1H NMR (500MHz, dimethylsulfoxide-d)6)δ7.53-7.50(m,2H),7.44-7.40(m,1H),7.34-7.26(m,3H),7.13-7.09(m,2H),4.90(t,J=9.0Hz,1H),4.52-4.47(m,2H),4.12-4.03(m,2H),3.85(s,3H),3.83(s,3H);LCMS(ESI)m/z:383.0[M+H]+

EXAMPLE 84 preparation of (5- (2-methyl-2H-indazol-6-yl) isoxazol-3-yl) (3-phenylazetidin-1-yl) methanone (235)

Step 1: preparation of 6-bromo-2-methyl-2H-indazole

To a solution of 6-bromo-1H-indazole (6.48g, 32.9mmol) in anhydrous tetrahydrofuran (80mL) at 0 ℃ was added sodium hydride (60% in mineral oil, 1.39g, 34.5 mmol). The reaction mixture was warmed to room temperature for 2h, then iodomethane (18.68g, 131.6mmol) was added. The reaction mixture was stirred at room temperature for 3 h. The reaction mixture was diluted with water (300mL) and extracted with ethyl acetate (100 mL. times.3). The combined organic layers were washed with brine, dried over sodium sulfate, filtered, and concentrated in vacuo. The crude residue was purified by column chromatography (silica, petroleum ether/ethyl acetate-4/1) to give 6-bromo-2-methyl-2H-indazole (3.4g, 16.11mmol, 49%) as a yellow oil. LCMS (ESI) M/z 211.1[ M + H ]]+

Step 2: preparation of ethyl 5- (2-methyl-2H-indazol-6-yl) isoxazole-3-carboxylate

To a solution of 6-bromo-2-methyl-2H-indazole (0.600g, 2.84mmol) in 1, 4-dioxane (10mL) was added ethyl 5- (tributylstannyl) isoxazole-3-carboxylate (1.35g, 3.13mmol) and bis (triphenylphosphine) palladium (II) dichloride (0.199g, 0.29mmol) under nitrogen at room temperature. The reaction mixture was heated at 90 ℃ for 16 h. The reaction mixture was cooled to room temperature and diluted with water (100mL), and extracted with ethyl acetate (50mL × 3). The combined organic layers were washed with brine, dried over sodium sulfate, filtered, and concentrated in vacuo. The crude residue was purified by column chromatography (silica, petroleum ether/ethyl acetate 5/1) to give ethyl 5- (2-methyl-2H-indazol-6-yl) isoxazole-3-carboxylate (620mg, 2.29) as a yellow solid mmol,81%)。LCMS(ESI)m/z:272.1[M+H]+

And step 3: preparation of ethyl 5- (2-methyl-2H-indazol-6-yl) isoxazole-3-carboxylic acid

To a solution of 5- (2-methyl-2H-indazol-6-yl) isoxazole-3-carboxylic acid ester (0.570g, 2.10mmol) in tetrahydrofuran (5mL) was added lithium hydroxide hydrate (0.441g, 10.5mmol) in water (5mL) at room temperature. The reaction mixture was stirred at room temperature for 1h, and then the aqueous layer was adjusted to pH 3-4 with 2N aqueous hydrogen chloride solution. The aqueous layer was extracted with ethyl acetate (50 mL. times.3). The combined organic layers were washed with brine, dried over sodium sulfate, filtered and concentrated to provide 5- (2-methyl-2H-indazol-6-yl) isoxazole-3-carboxylic acid as a yellow solid (0.220g, 0.91mmol, 43%). LCMS (ESI) M/z 244.1[ M + H ]]+

And 4, step 4: preparation of 5(5- (2-methyl-2H-indazol-6-yl) isoxazol-3-yl) (3-phenylazetidin-1-yl) methanone

To a solution of 5- (2-methyl-2H-indazol-6-yl) isoxazole-3-carboxylic acid (0.120g, 0.90mmol) in anhydrous dichloromethane (15mL) at 0 ℃ was added N, N' -dimethylformamide (7mg, 0.09mmol) and oxalyl chloride (0.229g, 1.80 mmol). The mixture was stirred at room temperature for 1h, then the volatiles were removed in vacuo. The residue was dissolved in anhydrous tetrahydrofuran (6mL) and added to a solution of 3-phenylazetidine hydrochloride (263mg, 1.08mmol) in anhydrous tetrahydrofuran (6 mL). The reaction mixture was stirred at room temperature for 3 h. The reaction mixture was diluted with water (100mL) and extracted with dichloromethane (50 mL. times.3). The combined organic layers were washed with brine, dried over sodium sulfate, filtered, and concentrated in vacuo. The crude sample was dissolved in a minimum amount of N, N-dimethylformamide and subjected to preparative-HPLC (Boston C1821X 250mm 10 μm column; acetonitrile/0.0 1% aqueous trifluoroacetic acid) to yield 5(5- (2-methyl-2H-indazol-6-yl) isoxazol-3-yl) (3-phenylazetidin-1-yl) methanone as a yellow solid (60mg, 0.17mmol, 19%).1H NMR (500MHz, dimethylsulfoxide-d)6)δ8.34(s,1H),8.15(s,1H),7.92(d,J=8.4Hz,1H),7.70(d,J=8.4Hz,1H),7.51(s,1H),7.44(d,J=7.4Hz,2H),7.39(t,J=7.6Hz,2H),7.29(t,J=7.2Hz,1H),4.94(t,J=9.2Hz,1H),4.52(dt,J=9.6,7.9Hz,2H),4.16-4.09(m,4H),4.07-3.97(m,1H);LCMS(ESI)m/z:359.2[M+H]+

EXAMPLE 85 (5- (1-methyl-1H-indazol-6-yl) isoxazol-3-yl) (3-phenylazetidin-1-yl) methanone (232)

Step 1: preparation of 6-bromo-1-methyl-1H-indazole

To a solution of 6-bromo-1H-indazole (6.48g, 32.9mmol) in anhydrous tetrahydrofuran (80mL) at 0 ℃ was added sodium hydride (60% in mineral oil, 1.39g, 34.5 mmol). The reaction mixture was warmed to room temperature for 2h, then iodomethane (18.68g, 132mmol) was added and the reaction mixture was stirred at room temperature for 3 h. The reaction mixture was diluted with water (300mL) and extracted with ethyl acetate (100 mL. times.3). The combined organic layers were washed with brine, dried over sodium sulfate, filtered and concentrated in vacuo. The crude residue was purified by column chromatography (silica, petroleum ether/ethyl acetate-4/1) to give 6-bromo-1-methyl-1H-indazole (3.2g, 15.2mmol, 46%) as a yellow oil. LCMS (ESI) M/z 211.1[ M + H ]]+

Step 2: preparation of ethyl 5- (1-methyl-1H-indazol-6-yl) isoxazole-3-carboxylate

The synthesis of ethyl 5- (1-methyl-1H-indazol-6-yl) isoxazole-3-carboxylate was performed following the same procedure as example 84. Compound 5- (1-methyl-1H-indazol-6-yl) isoxazole-3-carboxylic acid ester (0.680g, 2.51mmol, 88% yield) was obtained as a product. LCMS (ESI) M/z 272.1[ M + H]+

And step 3: preparation of 5- (1-methyl-1H-indazol-6-yl) isoxazole-3-carboxylic acid

The synthesis of 5- (1-methyl-1H-indazol-6-yl) isoxazole-3-carboxylic acid was performed following the same procedure as example 84. Compound 5- (1-methyl-1H-indazol-6-yl) isoxazole-3-carboxylic acid (0.300g, 1.23mmol, 49%) was obtained as a yellow solid. LCMS (ESI) M/z 244.1[ M + H ]]+

And 4, step 4: preparation of ethyl (5- (1-methyl-1H-indazol-6-yl) isoxazol-3-yl) (3-phenylazetidin-1-yl) methanone

The synthesis of ethyl (5- (1-methyl-1H-indazol-6-yl) isoxazol-3-yl) (3-phenylazetidin-1-yl) methanone was performed following the same procedure as example 84. Ethyl (5- (1-methyl-1H-indazol-6-yl) isoxazol-3-yl) (3-phenylazetidin-1-yl) methanone (0.100g, 0.28mmol, 45%) was obtained as a yellow solid.1H NMR (500MHz, dimethylsulfoxide-d)6)δ8.44(s,1H),8.24(s,1H),7.87(d,J=8.7Hz,1H),7.73-7.47(m,1H),7.44(d,J=7.1Hz,3H),7.39(t,J=7.6Hz,2H),7.29(t,J=7.2Hz,1H),4.93(t,J=9.2Hz,1H),4.52(dt,J=9.7,7.8Hz,2H),4.22(s,3H),4.10(dd,J=9.9,6.4Hz,1H),4.05-3.97(m,1H);LCMS(ESI)m/z:359.2[M+H]+

EXAMPLE 86 preparation of (3-Phenylazetidin-1-yl) (5- (pyridin-2-yl) isoxazol-3-yl) methanone (233)

To a solution of 5- (pyridin-2-yl) isoxazole-3-carboxylic acid (0.150g, 0.79mmol) in dichloromethane (2mL) at 20 ℃ was added oxalyl chloride (2 mL). The reaction mixture was stirred at room temperature for 0.5h, then the volatiles were removed in vacuo. The residue was dissolved in dichloromethane (2mL) and added dropwise to a mixture of 3-phenylazetidine hydrochloride (0.137g, 1.03mmol) and triethylamine (0.239g, 2.37mmol) in dichloromethane (5 mL). The reaction mixture was stirred for a further 0.5h and concentrated in vacuo. The crude sample was dissolved in a minimal amount of N, N-dimethylformamide and purified via preparative-HPLC (Boston C1821 × 250mm 10 μm column; acetonitrile/0.01% aqueous trifluoroacetic acid) to give (3-phenylazetidin-1-yl) (5- (pyridin-2-yl) isoxazol-3-yl) methanone as a white solid (72.8mg, 0.237mmol, 30%).1H NMR (400MHz, dimethylsulfoxide-d)6)δ8.76(s,1H),8.03-8.08(m,2H),7.55-7.57(m,1H),7.37-7.44(m,5H),7.28-7.30(m,1H),4.90-4.92(m,1H),4.48-4.54(m,2H),4.09-4.12(m,1H),4.02(m,1H);LCMS(ESI)m/z:306.1[M+H]+

Example 87.preparation of 3-Phenylazetidin-1-yl) (5- (pyridin-3-yl) isoxazol-3-yl) methanone trifluoroacetate (234)

The synthesis of (3-phenylazetidin-1-yl) (5- (pyridin-3-yl) isoxazol-3-yl) methanone trifluoroacetate was performed following the same procedure as example 24. The compound (3-phenylazetidin-1-yl) (5- (pyridin-3-yl) isoxazol-3-yl) methanone trifluoroacetate was obtained as a white solid (29.4mg, 0.07mmol, 33%). 1H NMR (500MHz, dimethylsulfoxide-d)6)δ9.20(d,J=2.0Hz,1H),8.75-8.73(m,1H),8.39(d,J=8.5Hz,1H),7.66-7.63(m,1H),7.60(s,1H),7.44-7.40(m,2H),7.38-7.37(m,2H),7.30-7.27(m,1H),4.92(d,J=9.0Hz,1H),4.55-4.48(m,2H),4.12-4.03(m,1H),4.02-3.99(m,1H);LCMS(ESI)m/z:306.1[M+H]+

EXAMPLE 88 preparation of (3-Phenylazetidin-1-yl) (5- (pyridin-4-yl) isoxazol-3-yl) methanone trifluoroacetate (229)

The synthesis of (3-phenylazetidin-1-yl) (5- (pyridin-4-yl) isoxazol-3-yl) methanone trifluoroacetate was performed following the same procedure as example 24. The compound (3-phenylazetidin-1-yl) (5- (pyridin-4-yl) isoxazol-3-yl) methanone trifluoroacetate was obtained as a white solid (8.0mg, 0.02mmol, 10%).1H NMR (500MHz, dimethylsulfoxide-d)6)δ8.82-8.80(m,2H),7.98-7.97(m,2H),7.75(s,1H),7.44-7.37(m,4H),7.28(t,J=7.5Hz,1H),4.92(t,J=9.0Hz,1H),4.56-4.48(m,2H),4.12-4.05(m,1H),4.04-4.00(m,1H);LCMS(ESI)m/z:306.1[M+H]+

EXAMPLE 89 preparation of (5- (5-Chloropyridin-2-yl) isoxazol-3-yl) (3-phenylazetidin-1-yl) methanone (241)

Step 1: preparation of 5-chloro-2- ((trimethylsilyl) ethynyl) pyridine

2-bromo-5-chloropyridine (3.5g, 18.2mmol), ethynyltrimethylsilane (2.14g, 21.9mmol) and [1, 1' -bis (diphenylphosphino) ferrocene]Palladium (II) dichloride (1.33g, 1.82mmol) and copper (I) iodide (0.68g, 3.65mmol) in triethylamine (50mL)The mixture of (a) was heated at 90 ℃ for 2 h. The mixture was diluted with brine (100mL) and extracted with ethyl acetate (100 mL. times.2). The combined organic layers were dried over sodium sulfate, filtered and concentrated in vacuo. The crude residue was purified by column chromatography (silica, petroleum ether/ethyl acetate ═ 20/1) to give 5-chloro-2- ((trimethylsilyl) ethynyl) pyridine as a yellow oil (1.4g, 6.7mmol, 37%). LCMS (ESI) M/z 210.1[ M + H ] ]+

Step 2: preparation of 5-chloro-2-ethynylpyridine

A mixture of 5-chloro-2- ((trimethylsilyl) ethynyl) pyridine (1.4g, 6.7mmol) and potassium hydroxide (0.38g, 6.7mmol) in methanol (50mL) was stirred at 20 ℃ for 5 h. The reaction mixture was diluted with brine (100mL) and extracted with ethyl acetate (100 mL. times.2). The combined organic layers were dried over sodium sulfate, filtered and concentrated in vacuo. The crude residue was purified by column chromatography (silica, petroleum ether/ethyl acetate 20/1) to give 5-chloro-2-ethynylpyridine as a yellow oil (0.75g, 5.5mmol, 82%). LCMS (ESI) M/z 138.0[ M + H ]]+

And step 3: preparation of 5- (5-chloropyridin-2-yl) isoxazole-3-carboxylic acid ethyl ester

The synthesis of ethyl 5- (5-chloropyridin-2-yl) isoxazole-3-carboxylate was performed following the same procedure as example 89. Ethyl 5- (5-chloropyridin-2-yl) isoxazole-3-carboxylate (290mg, 1.15mmol, 21%) was obtained as a grey oil. LCMS (ESI) M/z 253.1[ M + H ]]+

And 4, step 4: preparation of 5- (5-chloropyridin-2-yl) isoxazole-3-carboxylic acid

A mixture of ethyl 5- (5-chloropyridin-2-yl) isoxazole-3-carboxylate (0.29g, 1.15mmol) and aqueous lithium hydroxide (2.3mL, 4.6mmol, 2M) in ethanol (2.3mL) was stirred at 20 ℃ for 2 h. The reaction mixture was extracted with ethyl acetate (20mL) and concentrated in vacuo to give 1- (5-fluoro-2- (trifluoromethyl) benzyl) -1H-pyrazole-4-carboxylic acid (0.200g, 0.89mmol, 78%) as a grey solid. LCMS (ESI) M/z 225.1[ M + H ] ]+

And 5: preparation of (5- (5-chloropyridin-2-yl) isoxazol-3-yl) (3-phenylazetidin-1-yl) methanone

3-Phenylazetidine hydrochloride (143mg, 1.07mmol), 5- (5-chloropyridin-2-yl) isoxazole-3-carboxylic acid (200mg, 0.89mmol), 1- [ bis (dimethylamino) methylene]-1H-1, 2, 3-triazolo [4, 5-b]A mixture of pyridinium 3-oxide hexafluorophosphate (407mg, 1.07mmol) and triethylamine (270mg, 2.67mmol) in dry N, N' -dimethylformamide (5.0mL) was stirred at 20 ℃ for 16 h. The reaction mixture was extracted with ethyl acetate (20 mL. times.2) and washed with water (30mL) and brine (30 mL). The combined organic phases were dried over anhydrous sodium sulfate, filtered, and concentrated in vacuo. The crude sample was dissolved in a minimal amount of N, N-dimethylformamide and purified via preparative-HPLC (Boston C1821 × 250mm 10 μm column; acetonitrile/0.01% aqueous trifluoroacetic acid) to give (5- (5-chloropyridin-2-yl) isoxazol-3-yl) (3-phenylazetidin-1-yl) methanone (28mg, 0.08mmol, 9%) as a grey solid.1H NMR (500MHz, dimethylsulfoxide-d)6)δ8.82(d,J=2.4Hz,1H),8.19(dd,J=8.5,2.4Hz,1H),8.12(d,J=8.5Hz,1H),7.44(d,J=8.2Hz,3H),7.38(t,J=7.6Hz,2H),7.28(t,J=7.2Hz,1H),4.91(t,J=9.1Hz,1H),4.51(dt,J=9.6,7.8Hz,2H),4.10(dd,J=9.9,6.5Hz,1H),4.01(dt,J=15.2,7.6Hz,1H);LCMS(ESI)m/z:340.0[M+H]+

EXAMPLE 90 preparation of (5- (5-methoxypyridin-2-yl) isoxazol-3-yl) (3-phenylazetidin-1-yl) methanone (242)

Step 1: preparation of 5-methoxy-2- ((trimethylsilyl) ethynyl) pyridine

The synthesis of 5-methoxy-2- ((trimethylsilyl) ethynyl) pyridine was performed following the same procedure as example 89. Compound 5-methoxy-2- ((trimethylsilyl) ethynyl) pyridine (1.6g, 7.8mmol, 30%) was obtained as a colorless oil. LCMS (ESI) M/z 206.2[ M + H ]]+

Step 2: preparation of 2-ethynyl-5-methoxypyridine

The synthesis of 2-ethynyl-5-methoxypyridine was performed following the same procedure as example 89. Compound 2-ethynyl-5-methoxypyridine (0.8g, 6.0mmol, 77%) was obtained as a colorless oil. LCMS (ESI) M/z 134.2[ M + H ]]+

And step 3: preparation of 5- (5-methoxypyridin-2-yl) isoxazole-3-carboxylic acid ethyl ester

The synthesis of ethyl 5- (5-methoxypyridin-2-yl) isoxazole-3-carboxylate was performed following the same procedure as example 89. The compound ethyl 5- (5-methoxypyridin-2-yl) isoxazole-3-carboxylate (0.330g, 1.33mmol, 22%) was obtained as a grey solid. LCMS (ESI) M/z 249.1[ M + H ]]+

And 4, step 4: preparation of 5- (5-methoxypyridin-2-yl) isoxazole-3-carboxylic acid

The synthesis of 5- (5-methoxypyridin-2-yl) isoxazole-3-carboxylic acid was performed following the same procedure as example 89. The compound (5- (5-methoxypyridin-2-yl) isoxazol-3-yl) (3-phenylazetidin-1-yl) methanone (0.200g, 0.90mmol, 68%) was obtained as a grey solid. LCMS (ESI) M/z 221.1[ M + H ] ]+

And 5: preparation of ((5- (5-methoxypyridin-2-yl) isoxazol-3-yl) (3-phenylazetidin-1-yl) methanone

The synthesis of ((5- (5-methoxypyridin-2-yl) isoxazol-3-yl) (3-phenylazetidin-1-yl) methanone was carried out following the same procedure as example 89 the compound (5- (5-methoxypyridin-2-yl) isoxazol-3-yl) (3-phenylazetidin-1-yl) methanone was obtained as a grey solid (32.8mg, 0.1mmol, 8%).1H NMR (500MHz, dimethylsulfoxide-d)6)δ8.46(d,J=2.9Hz,1H),8.04(d,J=8.8Hz,1H),7.59(dd,J=8.8,2.9Hz,1H),7.47-7.35(m,4H),7.28(t,J=7.3Hz,1H),7.23(s,1H),4.91(t,J=9.2Hz,1H),4.59-4.44(m,2H),4.17-4.05(m,1H),4.00(td,J=8.7,4.3Hz,1H),3.91(d,J=11.6Hz,3H);LCMS(ESI)m/z:336.1[M+H]+

EXAMPLE 91 preparation of (5- (benzo [ d ] [1,3] dioxol-5-yl) isoxazol-3-yl) (3- (3-fluorophenyl) azetidin-1-yl) methanone (239)

Step 1: preparation of 5-ethynylbenzo [ d ] [1,3] dioxoles

To a solution of (benzo [ d ] [1,3] dioxol-5-ylethynyl) trimethylsilane (10.0g, 45.8mmol) in methanol (80mL) and tetrahydrofuran (80mL) was added potassium carbonate (12.6g, 91.6 mmol). The mixture was stirred at room temperature for 2 h. The mixture was diluted with water (100mL) and extracted with ethyl acetate (80 mL. times.2). The combined organic layers were washed with brine (60 mL. times.2), dried over anhydrous sodium sulfate, filtered and concentrated in vacuo to afford 5-ethynylbenzo [ d ] [1,3] dioxole as a yellow oil (5.8g, 39.7mmol, 88%). This material was used in the next step without further purification.

Step 2: preparation of ethyl 5- (benzo [ d ] [1, 3] dioxol-5-yl) isoxazole-3-carboxylate

To 5-ethynylbenzo [ d ] over 40min under nitrogen][1,3]To a solution of dioxole (5.8g, 39.7mmol) in N, N '-dimethylformamide (60.0mL) was added dropwise a solution of ethyl (Z) -2-chloro-2- (hydroxyimino) acetate (5.9g, 39.7mmol) in N, N' -dimethylformamide (20 mL). After addition, the reaction mixture was heated to 90 ℃ and a solution of triethylamine (12.0g, 119mmol) in N, N' -dimethylformamide (20mL) was added dropwise over 1 h. The reaction mixture was stirred at this temperature for 17h and cooled to room temperature. The reaction mixture was diluted with ethyl acetate (50mL), washed with water (60mL × 2) and brine (60mL × 2), dried over anhydrous sodium sulfate, filtered and concentrated in vacuo. The residue was purified by column chromatography (silica, petroleum ether/ethyl acetate 20/1) to give 5- (benzo [ d ] as a yellow solid][1,3]Dioxol-5-yl) isoxazole-3-carboxylic acid ethyl ester (3.7g, 14.1mmol, 36%). LCMS (ESI) M/z 262.1[ M + H ]]+

And step 3: preparation of 5- (benzo [ d ] [1, 3] dioxol-5-yl) isoxazole-3-carboxylic acid

To 5- (benzo [ d ] at room temperature ][1,3]To a solution of dioxol-5-yl) isoxazole-3-carboxylic acid ethyl ester (220.0mg, 0.84mmol) in tetrahydrofuran/water (v/v ═ 4/1, 20mL) was added sodium hydroxide (80.6mg, 2.01 mmol). The reaction mixture was stirred at room temperature for 17h, then the volatiles were removed in vacuo. The residue was diluted with water (10mL) and adjusted to pH 2 with 1N aqueous hydrogen chloride. The aqueous layer was extracted with ethyl acetate (10 mL. times.2). The combined organic layers were washed with brine (10 mL. times.2), dried over anhydrous sodium sulfate and concentrated in vacuo to give 5- (benzo [ d ] a yellow solid][1,3]Dioxol-5-yl) isoxazole-3-carboxylic acid (120mg, 0.51mmol, 67%). LCMS (ESI) M/z 234.1[ M + H ]]+. This material was used in the next step without further purification.

And 4, step 4: preparation of (5- (benzo [ d ] [1, 3] dioxol-5-yl) isoxazol-3-yl) (3- (3-fluorophenyl) azetidin-1-yl) methanone

At 0 ℃ to 5- (benzo [ d ]][1,3]To a solution of dioxol-5-yl) isoxazole-3-carboxylic acid (0.120g, 0.51mmol) in dichloromethane (6mL) was added oxalyl chloride (0.129g, 1.02mmol) and N, N' -dimethylformamide (0.01 mL). The reaction mixture was stirred at room temperature for 1h and then evaporated to dryness. The residue was dissolved in dichloromethane (8mL) and added to 3- (3-fluorophenyl) azetidine (92.1mg, 0.61mmol) and pyridine (0.181g, 2.29mmol) in dichloromethane (10mL) at 0 ℃. The reaction mixture was stirred at room temperature for 2 h. The reaction mixture was concentrated in vacuo. The crude sample was dissolved in a minimal amount of N, N-dimethylformamide and purified by preparative-HPLC (Boston C1821X 250mm 10 μm column; acetonitrile/0.01% aqueous trifluoroacetic acid) to give (5- (benzo [ d ] as a white solid ][1,3]Intermediate twoOxopenen-5-yl) isoxazol-3-yl) (3- (3-fluorophenyl) azetidin-1-yl) methanone (33.8mg, 0.09mmol, 18%).1H NMR (500MHz, dimethylsulfoxide-d)6)δ7.60-7.46(m,2H),7.45-7.37(m,1H),7.35-7.24(m,3H),7.15-7.04(m,2H),6.13(s,2H),4.94-4.80(m,1H),4.49(dd,J=17.3,9.1Hz,2H),4.14-3.98(m,2H);LCMS(ESI)m/z:367.0[M+H]+

EXAMPLE 92 preparation of (5- (benzo [ d ] [1,3] dioxol-5-yl) isoxazol-3-yl) (2-phenylazetidin-1-yl) methanone (204)

Step 1: preparation of 4-phenyl azetidin-2-ones

To a solution of 3-amino-3-phenyl-propionic acid (2.00g, 12.1mmol) in acetonitrile (20mL) was added 2-chloro-1-methyl-pyridin-1-ium iodide (3.40g, 13.3mmol) and triethylamine (2.70g, 26.6 mmol). The reaction mixture was heated to 80 ℃ and stirred for 18 h. The mixture was cooled to 25 ℃ and filtered. The filtrate was concentrated in vacuo. The crude product was purified by column chromatography (silica, petroleum ether/ethyl acetate 10/1 to 1/2) to afford 4-phenylazetidin-2-one as a white solid (0.900g, 6.12mmol, 50%).1H NMR (400MHz, chloroform-d) δ 7.47-7.33(m, 5H), 6.12(br.s, 1H), 4.76(dd, J ═ 2.6, 5.3Hz, 1H), 3.48(ddd, J ═ 2.4, 5.3, 14.8Hz, 1H), 2.92(dd, J ═ 1.6, 14.9Hz, 1H)

Step 2: preparation of 2-phenyl azetidines

To a solution of lithium aluminum hydride (0.361g, 9.52mmol) in tetrahydrofuran (10mL) was added 4-phenylazetidin-2-one (0.400g, 2.72 mmol). The reaction mixture was heated to 80 ℃ and stirred for 2 h. The reaction mixture was cooled to 0 ℃ and quenched with saturated aqueous ammonium chloride (1 mL). The mixture was then filtered. The filter cake was washed with dichloromethane/methanol (═ 10/1, 10mL) and the filtrate was concentrated in vacuo. A mixture of the compound 2-phenylazetidine (190mg, crude) and 3-amino-3-phenyl-propan-1-ol (190mg, 1.26mmol, 46%) was obtained as a colorless oil.

And step 3: preparation of (5- (benzo [ d ] [1, 3] dioxol-5-yl) isoxazol-3-yl) (2-phenylazetidin-1-yl) methanone 5- (benzo [ d ] [1, 3] dioxol-5-yl) -N- (3-hydroxy-1-phenylpropyl) isoxazole-3-carboxamide

To 5- (benzo [ d ]][1,3]To a solution of dioxol-5-yl) isoxazole-3-carboxylic acid (0.231g, 0.992mmol) in N, N '-dimethylformamide (3mL) were added N, N' -tetramethyl-O- (1H-benzotriazol-1-yl) uronium hexafluorophosphate (0.376g, 0.992mmol), diisopropylethylamine (0.256g, 1.98mmol) and 3-amino-3-phenyl-propan-1-ol (0.150g, 0.992 mmol). The reaction mixture was stirred at 25 ℃ for 0.5 h. The reaction mixture was directly purified by preparative-HPLC (Waters Xbridge 150X 255 μm column; 37-67% acetonitrile in 10mM aqueous ammonium bicarbonate, 12min gradient) to give (5- (benzo [ d ] b) as a yellow solid][1,3]Dioxol-5-yl) isoxazol-3-yl) (2-phenylazetidin-1-yl) methanone (53mg, 0.141mmol, 14%).1H NMR (400MHz, chloroform-d) δ 7.31-7.34(m, 3H), 7.24-7.27(m, 2.6vH), 7.16-7.24(m, 2H), 6.82-6.84(m, 1H), 6.71(s 1H), 6.5(s, 1H), 5.96(d, J ═ 1.2, 2H), 5.86-5.88(m, 1H), 5.3-5.5(m, 1H), 4.59-4.68(m, 1H), 4.25-4.32(m, 1H), 2.78-2.85(m, 1H), 2.10-2.28(m, 1H); LCMS (ESI M/z:349.1[ M + H ] ]+

EXAMPLE 93 preparation of [5- (1, 3-benzodioxol-5-yl) isoxazol-3-yl ] - [2- (3-fluorophenyl) azetidin-1-yl ] methanone (220)

Step 1: preparation of 4- (3-fluorophenyl) azetidin-2-one

To a stirred solution of N- (chloromethylene) sulfamoyl chloride (1.16g, 8.19mmol) in toluene (5mL) at 0 deg.C was added dropwise 1-fluoro-3-vinylbenzene (1.00g, 8.19mmol) in toluene (5 mL). The mixture was then stirred at 30 ℃ for 16 h. The reaction mixture was poured into a solution of sodium sulfate (0.4g) and sodium carbonate (1.6g) in water (15mL), then stirred at 15 ℃ for 30 min. The aqueous layer was extracted with ethyl acetate (20 mL. times.3). The combined organic layers were washed with brine (5mL), dried over sodium sulfate, filtered and concentrated in vacuo. The crude product was purified by column (ISCO, 20g silica, 30-60% ethyl acetate in petroleum ether, 20min gradient) to give 4- (3-fluorophenyl) azetidin-2-one as a pale yellow solid (0.300g, 1.82mmol, 22%).

Step 2: preparation of 2- (3-fluorophenyl) azetidines

To a solution of lithium aluminium hydride (0.207g, 5.45mmol) in tetrahydrofuran (5mL) was added 4- (3-fluorophenyl) azetidin-2-one (0.300g, 1.82mmol) portionwise at 0 ℃. The mixture was then heated at 80 ℃ for 1 h. The reaction mixture was quenched sequentially with water, 10% sodium hydroxide and water (1:2:1, 0.4mL total). The reaction mixture was dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated in vacuo to give 2- (2-fluorophenyl) azetidine (0.070g, 0.463mmol, 25%) as a colorless oil.

And step 3: preparation of [5- (1, 3-benzodioxol-5-yl) isoxazol-3-yl ] - [2- (3-fluorophenyl) azetidin-1-yl ] methanone

To a stirred solution of 5- (1, 3-benzodioxol-5-yl) isoxazole-3-carboxylic acid (0.090g, 0.386mmol) in N, N ' -dimethylformamide (0.5mL) were added N, ' N ' -tetramethyl-O- (1H-benzotriazol-1-yl) uronium hexafluorophosphate (0.176g, 0.463mmol), diisopropylethylamine (150mg, 1.16mmol) and 2- (3-fluorophenyl) azetidine (0.070g, 0.463 mmol). The mixture was then stirred at 15 ℃ for 1h and purified directly by preparative-HPLC (Waters X bridge 150X 255 μm column; 35-60% acetonitrile in 10mM aqueous ammonium bicarbonate, 11min gradient) to give [5- (1, 3-benzodioxol-5-yl) isoxazol-3-yl as a brown solid]- [2- (3-fluorophenyl) azetidin-1-yl]Methanone (49mg, 0.133mmol, 31%).1H NMR (400MHz, chloroform-d) δ 7.44-7.30(m, 1.8H), 7.26-6.95(m, 4.2H), 6.95-6.85(m, 1H), 6.83-6.67(m, 1H), 6.12-6.02(m, 2H), 5.98(dd, J ═ 5.6, 9.2Hz, 0.4H), 5.59(dd, J ═ 6.0, 8.8Hz, 0.6H), 4.81-4.64(m, 1.4H), 4.44-4.29(m, 0.6H), 3.03-2.84(m, 1H), 2.37-2.15(m, 1H); LCMS (ESI) M/z 367.0[ M + H ] ]+

EXAMPLE 94 preparation of [5- (1, 3-benzodioxol-5-yl) isoxazol-3-yl ] - [2- (4-fluorophenyl) azetidin-1-yl ] methanone (223)

[5- (1, 3-Benzodioxolen-5-yl) isoxazol-3-yl ] was carried out following the procedure used in example 93]- [2- (4-fluorophenyl) azetidin-1-yl]And (5) synthesizing the ketone. The compound [5- (1, 3-benzodioxol-5-yl) isoxazol-3-yl ] is obtained as a white solid]- [2- (4-fluorophenyl) azetidin-1-yl]Methanone (39mg, 0.104mmol, 24%).1H NMR (400MHz, chloroform)-d)7.41(dd,J=5.6,8.4Hz,1H),7.37-7.30(m,1H),7.26-7.20(m,1H),7.07(t,J=8.4Hz,1H),7.00(t,J=8.8Hz,1H),6.90(t,J=1H),6.85(d,J=8.0Hz,1H),6.78(s,1H),6.61(s,1H),6.05(s,1H),6.02(s,1H),5.94(dd,J=4.8,8.8Hz,1H),5.56(dd,J=6.0,8.8Hz,1),4.84-4.55(m,1H),4.45-4.26(m,1H),3.00-2.77(m,1H),2.38-2.14(m,1H);LCMS(ESI)m/z:367.1[M+H]+

Example 95.5- (benzo [ d ] [1,3] dioxol-5-yl) isoxazol-3-yl) (4-phenylpiperazin-1-yl) methanone (265) preparation

At 25 ℃ to 5- (benzo [ d ]][1,3]Dioxol-5-yl) isoxazole-3-carboxylic acid (0.050g, 0.214mmol) and 1-phenylpiperazine (42mg, 0.257mmol) in N, N '-dimethylformamide (1mL) was added to a stirred mixture in one portion of N, N' -tetramethyl-O- (1H-benzotriazol-1-yl) uronium hexafluorophosphate (81mg, 0.214mmol) and diisopropylethylamine (0.055g, 0.429 mmol). The mixture was stirred at 25 ℃ for 15 h. The solvent was removed under vacuum. The residue was purified by preparative-HPLC (Luna C18100X 305. mu.m column; mobile phase: 45-65% acetonitrile in 0.1% aqueous trifluoroacetic acid, 12min gradient) to give (5- (benzo [ d ] b) as a white solid ][1,3]Dioxol-5-yl) isoxazol-3-yl) (4-phenylpiperazin-1-yl) methanone (64mg, 0.262mmol, 79%).1H NMR (400MHz, acetonitrile-d)3)δ7.43(dd,J=1.7,8.0Hz,1H),7.35(d,J=1.5Hz,1H),7.29-7.24(m,2H),7.00-6.95(m,3H),6.86(t,J=7.3Hz,1H),6.78(s,1H),6.06(s,2H),3.88-3.83(m,4H),3.28-3.17(m,4H);LCMS(ESI)m/z:378.2[M+H]+

EXAMPLE 96 preparation of (3-Phenylazetidin-1-yl) - (5-phenylisoxazol-3-yl) methanone (212)

The synthesis of (3-phenylazetidin-1-yl) - (5-phenylisoxazol-3-yl) methanone was performed following the procedure used in example 95. The compound (3-phenylazetidin-1-yl) - (5-phenylisoxazol-3-yl) methanone (51mg, 0.168mmol, 40%) was obtained as a pale yellow solid.1H NMR (400MHz, chloroform-d) δ 7.82(dd, J ═ 1.8, 7.5Hz, 2H), 7.55 to 7.45(m, 3H), 7.43 to 7.35(m, 4H), 7.33 to 7.28(m, 1H), 6.99(s, 1H), 5.04(t, J ═ 9.9Hz, 1H), 4.77 to 4.58(m, 2H), 4.30(dd, J ═ 6.1, 10.1Hz, 1H), 4.06 to 3.86(m, 1H); LCMS (ESI) M/z 305.1[ M + H ]]+

EXAMPLE 97 preparation of [5- (1, 3-benzodioxol-5-yl) isoxazol-3-yl ] - (3-phenylazetidin-1-yl) methanone (217)

Step 1: preparation of [5- (1, 3-benzodioxol-5-yl) isoxazol-3-yl ] - (3-phenylazetidin-1-yl) methanone

To a stirred solution of 5- (1, 3-benzodioxol-5-yl) isoxazole-3-carboxylic acid (0.100g, 0.429mmol) and 3-phenylazetidine hydrochloride (87mg, 0.515mmol) in dichloromethane (2mL) was added propylphosphonic anhydride (0.409g, 0.643mmol, 50% by weight in ethyl acetate) and triethylamine (0.130g, 1.29 mmol). The mixture was stirred at 20 ℃ for 15 h. The reaction mixture was quenched with water (5mL) and extracted with dichloromethane (5 mL. times.3). The combined organic layers were washed with brine (10mL), dried over sodium sulfate, filtered and concentrated under reduced pressure. The residue was purified by preparative-HPLC (YMC-actual Triart C18100X 30mM X5 μm column; acetonitrile 60-80% in 10mM aqueous ammonium bicarbonate, 12min gradient) to give [5- (1, 3-benzodioxol-5-yl) isoxaprop-e as a white solid Oxazol-3-yl]- (3-phenylazetidin-1-yl) methanone (19mg, 0.053mmol, 12%).1H NMR (400MHz, dimethylsulfoxide-d)6)δ7.53(d,J=1.3Hz,1H),7.49(dd,J=1.5,8.1Hz,1H),7.45-7.35(m,4H),7.31-7.25(m,2H),7.09(d,J=8.1Hz,1H),6.13(s,2H),4.89(t,J=9.2Hz,1H),4.55-4.43(m,2H),4.12-3.94(m,2H);LCMS(ESI)m/z:349.1[M+H]+

EXAMPLE 98 preparation of [5- (2-furyl) isoxazol-3-yl ] - (3-phenylazetidin-1-yl) methanone (207)

To a solution of 5- (2-furyl) isoxazole-3-carboxylic acid (140mg, 0.78mmol) and N, N '-tetramethyl-O- (1H-benzotriazol-1-yl) uronium hexafluorophosphate (356mg, 0.94mmol) in N, N' -dimethylformamide (3mL) was added diisopropylethylamine (303mg, 2.34mmol) and 3-phenylazetidine hydrochloride (146mg, 0.86 mmol). The mixture was stirred at 15 ℃ for 2 h. The mixture was purified by preparative-HPLC (Waters X bridge 150X 255. mu.M column; 35-70% acetonitrile in 10mM aqueous ammonium acetate, 10min gradient) to give [5- (2-furyl) isoxazol-3-yl as a brown solid]- (3-phenylazetidin-1-yl) methanone (0.086g, 0.29mmol, 37%).1H NMR (400MHz, chloroform-d) δ 7.59(s, 1H), 7.41-7.35(m, 4H), 7.31(br.d, J ═ 7.0Hz, 1H), 6.95(d, J ═ 3.5Hz, 1H), 6.89(s, 1H), 6.56(dd, J ═ 1.8, 3.5Hz, 1H), 5.02(t, J ═ 9.6Hz, 1H), 4.68-4.61(m, 2H), 4.32-4.26(m, 1H), 4.01-3.92(m, 1H); LCMS (ESI) M/z 295.0[ M + H ] ]+

EXAMPLE 99.3- (3-Phenylazetidine-1-carbonyl) -4,5,6, 7-tetrahydro-1, 2-benzoxazole (252)

To 3-phenylazetidine hydrochloride (50.7mg, 0.299) at 25 deg.Cmmol), 4,5,6, 7-tetrahydro-1, 2-benzoxazole-3-carboxylic acid (50.0mg, 0.299mmol) and 1- [ bis (dimethylamino) methylene]-1H-1, 2, 3-triazolo [4, 5-b]To a solution of pyridinium 3-oxide hexafluorophosphate (0.113g, 0.299mmol) in N, N' -dimethylformamide (1mL) was added diisopropylethylamine (130. mu.L, 0.748 mmol). The reaction mixture was stirred at 25 ℃ for 16 h. The reaction mixture was quenched with water (1 mL). The aqueous layer was extracted with ethyl acetate (5 mL. times.3). The combined organic layers were washed with brine, dried over magnesium sulfate, filtered and concentrated in vacuo. The crude residue was purified by column chromatography (ISCO, 12g silica, eluting with 60% ethyl acetate/hexanes for 20min) to give 3- (3-phenylazetidine-1-carbonyl) -4,5,6, 7-tetrahydro-1, 2-benzoxazole (34.3mg, 0.121mmol, 40%) as a clear oil.1H NMR (300MHz, dimethylsulfoxide-d)6)δ7.45-7.20(m,5H),4.91(t,J=8.7Hz,1H),4.53-4.41(m,2H),4.07-3.91(m,2H),2.71(t,J=6.1Hz,4H),1.71(tt,J=12.7,5.5Hz,4H);LCMS(ESI)m/z:283.3[M+H]+

Example 100.3- (3-Benzylethazetidine-1-carbonyl) -4,5,6, 7-tetrahydro-1, 2-benzoxazole (253) preparation

To 3-benzylazetidine hydrochloride (54.9mg, 0.299mmol), 4, 5, 6, 7-tetrahydro-1, 2-benzoxazole-3-carboxylic acid (50.0mg, 0.299mmol) and 1- [ bis (dimethylamino) methylene ] at 25 deg.C]-1H-1, 2, 3-triazolo [4, 5-b]To a solution of pyridinium 3-oxide hexafluorophosphate (113mg, 0.299mmol) in N, N' -dimethylformamide (1mL) was added diisopropylethylamine (130. mu.L, 0.748 mmol). The reaction mixture was stirred at 25 ℃ for 16 h. The reaction mixture was quenched with water (1 mL). The aqueous layer was extracted with ethyl acetate (5 mL. times.3). The combined organic layers were washed with brine, dried over magnesium sulfate, filtered and concentrated in vacuo. The crude residue was purified by column chromatography (ISCO, 12g silica, eluting with 60% ethyl acetate/hexanes for 20min) to afford 3- (3-benzylnitrogen as a white solidAzetidine-1-carbonyl) -4, 5, 6, 7-tetrahydro-1, 2-benzoxazole (33.3mg, 0.112mmol, 37%).1H NMR (300MHz, dimethylsulfoxide-d)6)δ7.36-7.15(m,5H),4.51(t,J=8.4Hz,1H),4.23-4.02(m,2H),3.75(dd,J=9.9,4.9Hz,1H),3.05-2.87(m,3H),2.68(q,J=6.3Hz,4H),1.69(dt,J=14.1,4.8Hz,4H)。LCMS(ESI)m/z:297.2[M+H]+

Example 101.preparation of 5- (1, 3-benzodioxol-5-yl) isoxazol-3-yl ] - (3-benzylazetidin-1-yl) methanone (216)

5- (1, 3-Benzodioxolen-5-yl) isoxazol-3-yl was carried out following the procedure reported for example 95 ]Synthesis of (3-benzyl azetidin-1-yl) methanone. The compound [5- (1, 3-benzodioxol-5-yl) isoxazol-3-yl ] is obtained as a yellow solid]- (3-benzyl azetidin-1-yl) methanone (32mg, 0.085mmol, 20%).1H NMR (400MHz, chloroform-d) δ 7.36-7.29(m, 3H), 7.26-7.21(m, 2H), 7.18(d, J ═ 7.1Hz, 2H), 6.91(d, J ═ 7.9Hz, 1H), 6.81(s, 1H), 6.05(s, 2H), 4.71-4.61(m, 1H), 4.39-4.23(m, 2H), 3.96(dd, J ═ 4.4, 10.8Hz, 1H), 3.11-3.07(m, 1H), 3.05-2.92(m, 2H); LCMS (ESI) M/z 363.1[ M + H ]]+

Example 102.3 preparation of benzyl azetidin-1-yl) - (5-phenylisoxazol-3-yl) methanone (211)

The synthesis of 3-benzylazetidin-1-yl) - (5-phenylisoxazol-3-yl) methanone was performed following the procedure reported for example 95. The compound (3-benzylazetidin-1-yl) - (5-phenylisoxazol-3-yl) methanone (77mg, 0.241mmol, 46%) was obtained as a pale yellow solid.1H NMR (400MHz, chloroform-d) δ 7.79(dd, J ═ 2.0, 7.7Hz,2H),7.52-7.42(m,3H),7.36-7.28(m,2H),7.27-7.15(m,3H),6.95(s,1H),4.73-4.62(m,1H),4.38-4.23(m,2H),3.96(dd,J=4.8,10.5Hz,1H),3.11-2.92(m,3H);LCMS(ESI)m/z:319.1[M+H]+

EXAMPLE 103 preparation of (3-Benzylethazetidin-1-yl) - [5- (2-furyl) isoxazol-3-yl ] methanone (205)

To a solution of 5- (2-furyl) isoxazole-3-carboxylic acid (0.100g, 0.56mmol) and N, N '-tetramethyl-O- (1H-benzotriazol-1-yl) uronium hexafluorophosphate (0.254g, 0.67mmol) in N, N' -dimethylformamide (3mL) was added diisopropylethylamine (0.216g, 1.67mmol) and 3-benzyl azetidine hydrochloride (0.113g, 0.61 mmol). The mixture was stirred at 15 ℃ for 1h and then purified directly by preparative-HPLC (Waters X bridge 150X 255. mu.M column; 35-75% acetonitrile in 10mM aqueous ammonium acetate, 10min gradient) to give (3-benzylazetidin-1-yl) - [5- (2-furyl) isoxazol-3-yl ] -as a yellow solid ]Methanone (0.062g, 0.20mmol, 36%).1H NMR (400MHz, chloroform-d) δ 7.57(s, 1H), 7.31(d, J ═ 7.6Hz, 2H), 7.26-7.22(m, 1H), 7.18(d, J ═ 7.2Hz, 2H), 6.94(d, J ═ 3.4Hz, 1H), 6.85(s, 1H), 6.56(dd, J ═ 1.7, 3.3Hz, 1H), 4.68-4.62(m, 1H), 4.33-4.26(m, 2H), 3.95(dd, J ═ 4.7, 10.8Hz, 1H), 3.05-2.97(m, 3H); LCMS (ESI) M/z 309.0[ M + H ]]+

EXAMPLE 104 preparation of [5- (2-furyl) isoxazol-3-yl ] - (3-phenoxyazetidin-1-yl) methanone (206)

To 5- (2-furyl) isoxazole-3-carboxylic acid (0.120g, 0.67mmol) and N, N, N, ' N ' -tetramethyl-O- (1H-benzotriazol-1-yl) uronium hexafluorophosphate (0.305g, 0.80mmol) in N, N ' -dimethylformamide (3mL)To the solution were added diisopropylethylamine (260mg, 2.01mmol) and 3-phenoxyazetidine hydrochloride (137mg, 0.74 mmol). The mixture was stirred at 15 ℃ for 2 h. The mixture was purified by preparative-HPLC (Waters X bridge 150X 255 um column; 35-65% acetonitrile in 10mM aqueous ammonium acetate, 11min gradient) to give [5- (2-furyl) isoxazol-3-yl as a pale yellow solid]- (3-phenoxy azetidin-1-yl) methanone (52mg, 0.17mmol, 25%). 1H NMR (400MHz, chloroform-d) δ 7.58(s, 1H), 7.32(t, J ═ 7.9Hz, 2H), 7.05-7.00(m, 1H), 6.95(d, J ═ 3.5Hz, 1H), 6.87(s, 1H), 6.79(d, J ═ 7.9Hz, 2H), 6.56(dd, J ═ 1.8, 3.5Hz, 1H), 5.08-4.97(m, 2H), 4.66-4.58(m, 2H), 4.30(dd, J ═ 4.2, 11.2Hz, 1H); LCMS (ESI) M/z 311.1[ M + H ]]+

EXAMPLE 105.6-hydroxy-6-phenyl-2-azaspiro [3.3] hept-2-yl) - (5-phenylisoxazol-3-yl) methanone (226) and [5- (1, 3-benzodioxol-5-yl) isoxazol-3-yl ] - (6-hydroxy-6-phenyl-2-azaspiro [3.3] hept-2-yl) methanone (YUMAX-3015) and preparation of (6-phenyl-2-azaspiro [3.3] hept-6-en-2-yl) - (5-phenylisoxazol-3-yl) methanone (225)

Step 1: preparation of tert-butyl 6-hydroxy-6-phenyl-2-azaspiro [3.3] heptane-2-carboxylate

6-oxo-2-azaspiro [3.3] at 0 ℃ under nitrogen]To a stirred solution of tert-butyl heptane-2-carboxylate (0.43g, 2.04mmol) in tetrahydrofuran (20mL) was added dropwise phenylmagnesium bromide (3M, 0.746 mL). The mixture was stirred at 0 ℃ for 2 h. The reaction mixture was quenched by addition of water (30mL) and extracted with ethyl acetate (50 mL. times.2). The combined organic layers were dried over sodium sulfate, filtered and concentrated under reduced pressure to give crude 6-hydroxy-6-phenyl-2-azaspiro [3.3] as a white solid ]Heptane-2-carboxylic acid tert-butyl esterEster (0.500g, 1.73mmol, 85%). This material was used directly in the next step.1H NMR (400MHz, chloroform-d) Δ 7.45-7.33(m, 4H), 7.32-7.24(m, 1H), 4.07(s, 2H), 3.80(s, 2H), 2.79-2.72(m, 2H), 2.55(m, 2H), 2.18-2.00(m, 1H), 1.42(s, 9H); LCMS (ESI) M/z 234.1[ M-56+ H]+

Step 2: preparation of 6-phenyl-2-azaspiro [3.3] hept-6-ol and 6-phenyl-2-azaspiro [3.3] hept-6-ene

6-hydroxy-6-phenyl-2-azaspiro [3.3] was treated dropwise with trifluoroacetic acid (6.15g, 53.9mmol) at 0 deg.C]A solution of tert-butyl heptane-2-carboxylate (0.400g, 1.38mmol) in dichloromethane (8 mL). The reaction mixture was then stirred at 25 ℃ for 1 h. The reaction mixture was concentrated under reduced pressure and the crude residue was purified directly by preparative-HPLC (Phenomenex Gemini C18250X 5010 μm column; 12-40% acetonitrile in 10mM aqueous ammonium bicarbonate, 20min gradient) to give (6-phenyl-2-azaspiro [ 3.3.3 ] as a white solid]Hept-6-ol (0.120g, crude). This material was used directly in the next step.1H NMR (400MHz, chloroform-d) δ 7.41-7.31(m, 4H), 7.28(m, 1H), 5.47(br.s, 1H), 3.84(br.s, 2H), 3.56(br.s, 2H), 3.35(br.s, 1H), 2.73(br.d, J ═ 12.8Hz, 2H), 2.59-2.48(br.d, J ═ 12.8Hz, 2H); LCMS (ESI) M/z 190.1[ M + H ] ]+And 6-phenyl-2-azaspiro [3.3] is obtained as a white solid]Hept-6-ene (0.200g, crude):1h NMR (400MHz, chloroform-d) δ 7.43-7.20(m, 5H), 6.41(s, 1H), 5.35-5.07(m, 1H), 4.14(br.s, 1H), 3.94(br.d, J ═ 16.5Hz, 3H), 2.98(br.s, 2H); LCMS (ESI) M/z 172.1[ M + H ]]+

And step 3: preparation of (6-hydroxy-6-phenyl-2-azaspiro [3.3] hept-2-yl) - (5-phenylisoxazol-3-yl) methanones

To 5-phenylisoxazole-3-carboxylic acid (60mg, 0.317mmol), 6-phenyl-2-azaspiro [3.3] at 0 deg.C]A stirred solution of hept-6-ol (60mg, 0.317mmol) and triethylamine (96mg, 0.951mmol) in dichloromethane (3mL) was added dropwise to propylphosphonic anhydride (0.303g, 0.476mmol, 50% in ethyl acetate) in ethyl acetate. The reaction mixture was stirred at 20 ℃ for 15 h. The reaction mixture was quenched with water (10mL) and then extracted with dichloromethane (10 mL. times.3). The combined organic layers were dried over sodium sulfate, filtered and concentrated in vacuo. The crude product was purified by preparative-HPLC (Waters Xbridge 150X 255 μm column; 30-70% acetonitrile in 10mM aqueous ammonium bicarbonate, 11min gradient) to give (6-hydroxy-6-phenyl-2-azaspiro [3.3] as a pale yellow solid ]Hept-2-yl) - (5-phenylisoxazol-3-yl) methanone (29mg, 0.080mmol, 25%).1H NMR (400MHz, chloroform-d) δ 7.79(dt, J ═ 2.1, 8.0Hz, 2H), 7.54-7.38(m, 7H), 7.36-7.29(m, 1H), 6.94(d, J ═ 1.1Hz, 1H), 4.78(s, 1H), 4.50(s, 1H), 4.39(s, 1H), 4.11(s, 1H), 2.94-2.82(m, 2H), 2.74-2.60(m, 2H), 2.00(d, J ═ 4.0Hz, 1H); LCMS (ESI) M/z 361.1[ M + H ]]+

And 4, step 4: preparation of (6-phenyl-2-azaspiro [3.3] hept-6-en-2-yl) - (5-phenylisoxazol-3-yl) methanones

To 5-phenylisoxazole-3-carboxylic acid (0.062g, 0.330mmol), 6-phenyl-2-azaspiro [3.3] at 0 deg.C]Hept-6-ol trifluoroacetic acid (20mg, 0.066mmol), 6-phenyl-2-azaspiro [ 3.3%]To a stirred solution of hept-6-ene trifluoroacetic acid (80mg, 0.280mmol) and triethylamine (0.100g, 0.989mmol) in dichloromethane (3mL) was added propylphosphonic anhydride (0.504g, 0.791mmol, 50% in ethyl acetate) dropwise. The reaction mixture was stirred at 15 ℃ for 15 h. The reaction mixture was quenched with water (10mL) and then extracted with dichloromethane (10 mL. times.3). The combined organic layers were dried over sodium sulfate and concentrated in vacuo. The crude product was passed through preparative-HPLC (YMC-actual Triart C18150X 305. mu.m column; 65-90% in 10mM ammonium bicarbonate Acetonitrile in aqueous solution, 10min gradient) to give (6-phenyl-2-azaspiro [3.3] as a brown solid]Hept-6-en-2-yl) - (5-phenylisoxazol-3-yl) methanone (10mg, 0.026mmol, 40%).1H NMR (400MHz, chloroform-d) Δ 7.82-7.77(m, 2H), 7.51-7.44(m, 3H), 7.39-7.26(m, 5H), 6.96(s, 1H), 6.42(s, 1H), 4.86-4.71(m, 2H), 4.49-4.35(m, 2H), 3.04(s, 2H); LCMS (ESI) M/z 343.1[ M + H ]]+

EXAMPLE 106 preparation of (6-phenyl-2-azaspiro [3.3] hept-2-yl) - (5-phenylisoxazol-3-yl) methanone (227)

Step 1: preparation of 6-phenyl-2-azaspiro [3.3] heptane

6-phenyl-2-azaspiro [3.3] was reacted under hydrogen (15psi) at 30 deg.C]A solution of hept-6-ene (540mg, 1.68mmol) and palladium on charcoal (50mg, 10 wt% Pd) in methanol (20mL) was stirred for 12 h. The reaction mixture was filtered and concentrated under reduced pressure to give 6-phenyl-2-azaspiro [3.3] as a yellow solid]Heptane (0.520g, 1.61mmol, 96%). LCMS (ESI) M/z 174.3[ M + H ]]+. This material was used directly in the next step without further purification.

Step 2: preparation of (6-phenyl-2-azaspiro [3.3] hept-2-yl) - (5-phenylisoxazol-3-yl) methanones

(6-phenyl-2-azaspiro [3.3] was performed following the procedure reported for the preparation of example 105 ]Synthesis of hept-2-yl) - (5-phenylisoxazol-3-yl) methanone. The compound (6-phenyl-2-azaspiro [3.3 ] is obtained as a brown solid]Hept-2-yl) - (5-phenylisoxazol-3-yl) methanone (10mg, 0.028mmol, 4%).1H NMR(400MHz, chloroform-d) δ 7.77-7.64(m, 2H), 7.47-7.32(m, 3H), 7.24(m, 1H), 7.19(s, 1H), 7.17-7.09(m, 3H), 6.88(d, J ═ 3.8Hz, 1H), 4.68(s, 1H), 4.46(s, 1H), 4.29(s, 1H), 4.07(s, 1H), 3.51-3.29(m, 1H), 2.70-2.50(m, 2H), 2.40-2.14(m, 2H); LCMS (ESI) M/z 345.0[ M + H ]]+

Example 107.preparation of 4-phenylpiperidin-1-yl) (5- (pyridin-2-yl) isoxazol-3-yl) methanone (318)

To a solution of 5- (pyridin-2-yl) isoxazole-3-carboxylic acid (150mg, 0.79mmol) in dichloromethane (2mL) at 20 ℃ was added oxalyl chloride (2 mL). The mixture was stirred at room temperature for 0.5 h. The solvent was removed in vacuo and the solid was dissolved in dichloromethane (2mL) and added dropwise to a mixture of 4-phenylpiperidine (166mg, 1.03mmol), triethylamine (239mg, 2.37mmol) in dichloromethane (5 mL). The reaction mixture was stirred for 0.5 h. The mixture was purified by preparative-HPLC to afford 4-phenylpiperidin-1-yl) (5- (pyridin-2-yl) isoxazol-3-yl) methanone as a white solid (0.104g, 0.316mmol, 40%). 1H NMR (400MHz, dimethylsulfoxide-d)6)δ8.76(s,1H),8.02-8.04(m,2H),7.55-7.57(m,1H),7.20-7.36(m,6H),4.65-4.67(m,1H),4.04-4.06(m,1H),3.26-3.31(m,1H),2.85-2.97(m,2H),1.90-1.92(m,1H),1.81-1.83(m,1H),1.62-1.68(m,2H);LCMS(ESI)m/z:334.1[M+H]+

EXAMPLE 108 preparation of (4-phenylpiperidin-1-yl) (5- (pyridin-3-yl) isoxazol-3-yl) methanone trifluoroacetate (315)

Step 1: preparation of 5- (pyridin-3-yl) isoxazole-3-carboxylic acid ethyl ester

The synthesis of ethyl 5- (pyridin-3-yl) isoxazole-3-carboxylate was carried out following the same procedure as example 24. The compound ethyl 5- (pyridin-3-yl) isoxazole-3-carboxylate (0.360g, 1.65mmol, 43%) was obtained as a yellow solid. LCMS (ESI) M/z 219.1[ M + H ]]+

Step 2: preparation of 5- (pyridin-3-yl) isoxazole-3-carboxylic acids

The synthesis of 5- (pyridin-3-yl) isoxazole-3-carboxylic acid was carried out following the same procedure as example 24. Compound 5- (pyridin-3-yl) isoxazole-3-carboxylic acid (0.180g, 0.94mmol, 57%) was obtained as a white solid. LCMS (ESI) M/z 191.2[ M + H ]]+

And step 3: preparation of (4-phenylpiperidin-1-yl) (5- (pyridin-3-yl) isoxazol-3-yl) methanone trifluoroacetate

The synthesis of (4-phenylpiperidin-1-yl) (5- (pyridin-3-yl) isoxazol-3-yl) methanone trifluoroacetate was performed following the same procedure as example 24. The compound (4-phenylpiperidin-1-yl) (5- (pyridin-3-yl) isoxazol-3-yl) methanone trifluoroacetate (0.080g, 0.18mmol, 58%) was obtained as a white solid. 1H NMR (500MHz, dimethylsulfoxide-d)6)δ9.19(d,J=2.0Hz,1H),8.76-8.74(m,1H),8.39-8.37(m,1H),7.67-7.65(m,1H),7.50(s,1H),7.33-7.20(m,5H),4.68-4.65(m,1H),4.12-4.10(m,1H),3.32-3.27(m,1H),2.98-2.85(m,2H),1.93-1.82(m,2H),1.69-1.59(m,2H);LCMS(ESI)m/z:334.1[M+H]+

EXAMPLE 109 preparation of 4- (3- (4-phenylpiperidine-1-carbonyl) isoxazol-5-yl) benzonitrile (316)

Step 1: preparation of ethyl 5- (4-cyanophenyl) isoxazole-3-carboxylate

Ethyl 5-bromoisoxazole-3-carboxylate (0.300g, 1.37mmol), 4-cyanophenylboronic acid (201mg, 1.37mmol), 1' -bis (diphenylphosphino) ferrocene]A mixture of palladium (II) dichloride (224mg, 0.27mmol) and potassium phosphate (580mg, 2.74mmol) in 1, 4-dioxane (15mL) and water (4mL) was heated to 80 ℃ under nitrogen for 16 h. The mixture was concentrated under reduced pressure and the residue was purified by column chromatography (silica, petroleum ether/ethyl acetate 4/1) to give ethyl 5- (4-cyanophenyl) isoxazole-3-carboxylate (240mg, 0.99mmol, 67%) as a grey solid. LCMS (ESI) M/z 243.1[ M + H ]]+

Step 2: preparation of 5- (4-cyanophenyl) isoxazole-3-carboxylic acids

The synthesis of 5- (4-cyanophenyl) isoxazole-3-carboxylic acid was carried out following the same procedure as example 24. Compound 5- (4-cyanophenyl) isoxazole-3-carboxylic acid (0.120g, 0.56mmol, 56%) was obtained as a grey solid. LCMS (ESI) M/z 215.1[ M + H ]]+

And step 3: preparation of 4- (3- (4-phenylpiperidine-1-carbonyl) isoxazol-5-yl) benzonitrile

The synthesis of 4- (3- (4-phenylpiperidine-1-carbonyl) isoxazol-5-yl) benzonitrile was performed following the same procedure as in example 24. The compound 4- (3- (4-phenylpiperidine-1-carbonyl) isoxazol-5-yl) benzonitrile (0.051g, 0.14mmol, 51%) was obtained as a white solid.1H NMR (500MHz, dimethylsulfoxide-d)6)δ8.13(d,J=8.0Hz,2H),8.05(d,J=8.0Hz,2H),7.55(s,1H),7.32-7.28(m,4H),7.22-7.19(m,1H),4.65(d,J=13.0Hz,1H),4.08(d,J=13.0Hz,1H),3.32-3.26(m,1H),2.97-2.88(m,2H),1.92(d,J=8.0Hz,1H),1.83(d,J=8.0Hz,1H),1.65-1.60(m,2H);LCMS(ESI)m/z:358.2[M+H]+

EXAMPLE 110 preparation of 2- (3- (4-phenylpiperidine-1-carbonyl) isoxazol-5-yl) benzonitrile (326)

Step 1: preparation of ethyl 5- (2-cyanophenyl) isoxazole-3-carboxylate

A mixture of 2-iodobenzonitrile (0.600g, 2.62mmol), ethyl 5- (tributylstannyl) isoxazole-3-carboxylate (1.69g, 3.93mmol) and bis (triphenylphosphine) palladium (II) dichloride (0.184g, 0.26mmol) in 1, 4-dioxane (40mL) was heated to 90 ℃ under nitrogen for 3 h. The mixture was concentrated under reduced pressure and the residue was purified by column chromatography (silica, petroleum ether/ethyl acetate 5/1) to give ethyl 5- (2-cyanophenyl) isoxazole-3-carboxylate (0.191g, 0.79mmol, 30%) as a yellow oil. LCMS (ESI) M/z 243.1[ M + H ]]+

Step 2: preparation of 5- (2-cyanophenyl) isoxazole-3-carboxylic acid)

The synthesis of 5- (2-cyanophenyl) isoxazole-3-carboxylic acid was carried out following the same procedure as example 24. Compound 5- (2-cyanophenyl) isoxazole-3-carboxylic acid (0.123g, 0.57mmol, 72%) was obtained as a yellow solid. LCMS (ESI) M/z 215.1[ M + H ] ]+

And step 3: preparation of 2- (3- (4-phenylpiperidine-1-carbonyl) isoxazol-5-yl) benzonitrile

The synthesis of 2- (3- (4-phenylpiperidine-1-carbonyl) isoxazol-5-yl) benzonitrile was performed following the same procedure as in example 24. Compound 2- (3- (4-phenylpiperidine-1-carbonyl) isoxazol-5-yl) benzonitrile (0.019g, 0.05mmol, 18%) was obtained as a white solid.1H NMR (500MHz, dimethylsulfoxide-d)6)δ8.14-8.09(m,2H),7.92(t,J=7.5Hz,1H),7.76(t,J=7.5Hz,1H),7.44(s,1H),7.33-7.27(m,4H),7.22-7.19(m,1H),4.68-4.65(m,1H),4.14-4.11(m,1H),3.33-3.28(m,1H),2.99-2.86(m,2H),1.93-1.82(m,2H),1.71-1.62(m,2H);LCMS(ESI)m/z:358.1[M+H]+

Example 111.preparation of 5-Phenylisoxazol-3-yl) (4-phenylpiperidin-1-yl) methanone (314)

The synthesis of (5-phenylisoxazol-3-yl) (4-phenylpiperidin-1-yl) methanone was carried out following the same procedure as example 24. The compound (5-phenylisoxazol-3-yl) (4-phenylpiperidin-1-yl) methanone (67.5mg, 0.20mmol, 26%) was obtained as a white solid.1H NMR (500MHz, dimethylsulfoxide-d)6)δ7.94(dd,J=8.0,1.5Hz,2H),7.63-7.44(m,3H),7.36-7.25(m,5H),7.21(t,J=7.0Hz,1H),4.66(d,J=13.0Hz,1H),4.10(d,J=13.5Hz,1H),3.27(dd,J=18.5,7.5Hz,1H),3.00-2.79(m,2H),1.91(d,J=13.0Hz,1H),1.83(d,J=12.5Hz,1H),1.73-1.50(m,2H);LCMS(ESI)m/z:333.1[M+H]+

EXAMPLE 112.3 preparation of 3-fluoro-4- (3- (4-phenylpiperidine-1-carbonyl) isoxazol-5-yl) benzonitrile (325)

Step 1: preparation of 3-fluoro-4- ((trimethylsilyl) ethynyl) benzonitrile

To a solution of 4-bromo-3-fluorobenzonitrile (4.0g, 20.1mmol) and triethylamine (5mL) in tetrahydrofuran (5mL) was added ethynyltrimethylsilane (2.17g, 22.1mmol), tetrakis (triphenylphosphine) palladium (0) (0.460g, 0.40mmol), and copper (I) iodide (84mg, 0.44mmol) under nitrogen. The mixture was then stirred at 20 ℃ for 3 h. The solvent was removed under reduced pressure. The residue was dissolved in dichloromethane (100mL), then washed with water (100mL) and brine (100mL), dried over sodium sulfate, filtered and concentrated in vacuo. The crude product was purified by column chromatography (silica, petroleum ether/ethyl acetate ═ 6/1) to give 3-fluoro-4- ((trimethylsilyl) ethynyl) benzonitrile as a white solid (3.0g, 13.9mmol, 69%); LCMS (ESI) M/z 218.1[ M + H ] ]+

Step 2: preparation of 4-ethynyl-3-fluorobenzonitrile

To a solution of 3-fluoro-4- ((trimethylsilyl) ethynyl) benzonitrile (3.0g, 13.8mmol) in tetrahydrofuran (25mL) was added tetrabutylammonium fluoride in tetrahydrofuran (27mL, 1M) at 20 ℃. The solution mixture was stirred at 20 ℃ for 0.5 h. Volatiles were removed under reduced pressure. The residue was dissolved in dichloromethane (100mL), then washed with water (100mL) and brine (100mL), dried over sodium sulfate, filtered and concentrated in vacuo. The crude product was purified by column chromatography (petroleum ether/ethyl acetate ═ 6/1) to give 4-ethynyl-3-fluorobenzonitrile (0.88g, 6.07mmol, 44%) as a yellow solid.

And step 3: preparation of ethyl 5- (4-cyano-2-fluorophenyl) isoxazole-3-carboxylate

To a solution of (Z) -2-chloro-2- (hydroxyimino) acetic acid ethyl ester (0.366g, 2.43mmol) in N, N '-dimethylformamide (2mL) under nitrogen at 20 ℃ was slowly added a solution of 4-ethynyl-3-fluorobenzonitrile (0.88g, 6.07mmol) in N, N' -dimethylformamide (10 mL). The solution mixture was heated to 90 ℃ and then triethylamine (0.737g, 7.29mmol) in N, N' -dimethylformamide (2mL) was slowly added. The reaction mixture was heated at 90 ℃ for 2 h. The reaction was diluted with dichloromethane (100mL), then washed with water (100mL) and brine (100mL), dried over sodium sulfate, filtered and concentrated in vacuo. The crude product was purified by column chromatography (silica, petroleum ether/ethyl acetate-4/1) to give ethyl 5- (4-cyano-2-fluorophenyl) isoxazole-3-carboxylate (0.36g, 1.39mmol, 57%) as a yellow oil. LCMS (ESI) M/z 261.1[ M + H ] ]+

And 4, step 4: preparation of 5- (4-cyano-2-fluorophenyl) isoxazole-3-carboxylic acid

To a solution of ethyl 5- (4-cyano-2-fluorophenyl) isoxazole-3-carboxylate (0.2g, 0.769mmol) in 1/1 mixture of tetrahydrofuran/water (3mL) was added lithium hydroxide hydrate (32mg, 0.769 mmol). The reaction mixture was stirred at 10 ℃ for 0.5 h. The volatiles were removed in vacuo and the aqueous layer was adjusted to pH 3' 5 with 1N aqueous hydrogen chloride. Water was then removed to give 5- (4-cyano-2-fluorophenyl) isoxazole-3-carboxylic acid (0.130g, 0.561mmol, 73%) as a white solid. This material was used in the next step without further purification. LCMS (ESI) M/z 233.1[ M + H ]]+

And 5: preparation of 3-fluoro-4- (3- (4-phenylpiperidine-1-carbonyl) isoxazol-5-yl) benzonitrile

To a solution of 5- (4-cyano-2-fluorophenyl) isoxazole-3-carboxylic acid (0.120g, 0.577mmol) in dichloromethane (3mL) at 20 ℃ was added oxalyl chloride (1 mL). The reaction mixture was stirred at room temperature for 0.5 h. The solvent was removed in vacuo and the resulting solid was dissolved in dichloromethane (5mL) and added dropwise to a mixture of 4-phenylpiperidine (0.108g, 0.672mmol) and triethylamine (0.157g, 1.55mmol) in dichloromethane (3 mL). The mixture was then stirred for a further 0.5 h. The reaction was purified by column chromatography (silica, petroleum ether/ethyl acetate-1/1) to afford 3-fluoro-4- (3- (4-phenylpiperidine-1-carbonyl) isoxazol-5-yl) benzonitrile (0.128g, 0.340mmol, 59%) as a pale yellow solid. 1H NMR (400MHz, dimethylsulfoxide-d)6)δ8.16-8.21(m,2H),7.91-7.93(m,1H),7.27-7.35(m,5H),7.19-7.22(m,1H),4.64-4.67(m,1H),4.02-4.05(m,1H),3.27-3.29(m,1H),2.87-2.97(m,2H),1.90-1.92(m,1H),1.80-1.83(m,1H),1.62-1.68(m,2H);LCMS(ESI)m/z:376.1[M+H]+

Example 113 preparation of 5- (1, 3-benzodioxol-5-yl) isoxazol-3-yl ] - (4-phenyl-1-piperidinyl) methanone (275)

To a stirred solution of 5- (1, 3-benzodioxol-5-yl) isoxazole-3-carboxylic acid (0.080g, 0.343mmol) and N, N '-tetramethyl-O- (1H-benzotriazol-1-yl) uronium hexafluorophosphate (0.130g, 0.343mmol) in N, N' -dimethylformamide (2mL) were added diisopropylethylamine (0.089g, 0.686mmol) and 4-phenylpiperidine (0.055g, 0.343 mmol). The mixture was stirred at 25 ℃ for 2h and purified directly by preparative-HPLC (Luna C18150X 255 μm; 47-77% acetonitrile in 10mM aqueous ammonium bicarbonate, 12min gradient) to give [5- (1, 3-benzodioxol-5-yl) isoxazol-3-yl as a yellow solid]- (4-phenyl-1-piperidinyl) methanone (64mg, 0.171mmol, 50%).1H NMR (400MHz, chloroform-d) δ 7.35(br.t, J ═ t8.3Hz,3H),7.29(s,1H),7.27-7.22(m,3H),6.93(d,J=8.2Hz,1H),6.71(s,1H),6.08(s,2H),4.93(br.d,J=13.2Hz,1H),4.66(br.d,J=13.7Hz,1H),3.35-3.22(m,1H),2.98-2.81(m,2H),2.09-1.93(m,2H),1.89-1.75(m,2H);LCMS(ESI)m/z:377.1[M+H]+

EXAMPLE 114.preparation of 5-phenylisoxazol-3-yl) - [4- [2- (trifluoromethyl) phenyl ] -1-piperidinyl ] methanone (289)

To a stirred solution of 5-phenylisoxazole-3-carboxylic acid (0.100g, 0.529mmol) in dichloromethane (3mL) was added 4- [2- (trifluoromethyl) phenyl ]Piperidine (0.155g, 0.582mmol), triethylamine (0.160g, 1.59mmol) and propylphosphonic anhydride (0.505g, 0.793mmol, 50% in ethyl acetate). The mixture was stirred at 25 ℃ for 15 h. The reaction mixture was quenched with water (10mL) and extracted with dichloromethane (10 mL. times.3). The combined organic layers were washed with brine (30mL), dried over sodium sulfate, filtered and concentrated under reduced pressure. The residue was purified by preparative-HPLC (YMC-Actus Triart C18100X 30mM X5 μm column; acetonitrile in 10mM aqueous ammonium bicarbonate solution 60-90%, 12min gradient) to give (5-phenylisoxazol-3-yl) - [4- [2- (trifluoromethyl) phenyl ] isoxazol-3-yl) - [ as a pale yellow solid]-1-piperidinyl group]Methanone (0.135g, 0.336mmol, 64%).1H NMR (400MHz, chloroform-d) δ 7.82(dd, J ═ 1.8, 7.5Hz, 2H), 7.66(d, J ═ 7.9Hz, 1H), 7.57-7.43(m, 5H), 7.33(t, J ═ 7.7Hz, 1H), 6.85(s, 1H), 4.95(br.d, J ═ 13.6Hz, 1H), 4.69(br.d, J ═ 13.6Hz, 1H), 3.35-3.21(m, 2H), 2.92(dt, J ═ 2.9, 13.0Hz, 1H), 2.02-1.75(m, 4H); LCMS (ESI) M/z 401.1[ M + H ]]+

EXAMPLE 115 preparation of [5- (1, 3-benzodioxol-5-yl) isoxazol-3-yl ] - [4- [2- (trifluoromethyl) phenyl ] -1-piperidinyl ] methanone (302)

5- (1, 3-Benzodioxolen-5-yl) isoxazol-3-yl is carried out following the same procedure as in example 114]- [4- [2- (trifluoromethyl) phenyl group]-1-piperidinyl group]And (5) synthesizing the ketone. The compound [5- (1, 3-benzodioxol-5-yl) isoxazol-3-yl ] is obtained as a white solid]- [4- [2- (trifluoromethyl) phenyl group]-1-piperidinyl group]Methanone (0.072g, 0.162mmol, 38%).1H NMR (400MHz, dimethylsulfoxide-d)6)δ7.72-7.60(m,3H),7.51-7.45(m,2H),7.45-7.39(m,1H),7.18(s,1H),7.10(d,J=8.1Hz,1H),6.14(s,2H),4.67(br.d,J=13.1Hz,1H),4.16(br.d,J=13.6Hz,1H),3.32-3.24(m,1H),3.17(br.t,J=10.9Hz,1H),2.94(dt,J=2.9,12.6Hz,1H),1.89-1.66(m,4H);LCMS(ESI)m/z:445.1[M+H]+

Example 116.preparation of 5- (1, 3-benzodioxol-5-yl) isoxazol-3-yl ] - [4- (2-chloro-5-fluoro-phenyl) -1-piperidinyl ] methanone (308)

Step 1: preparation of 4- (2-chloro-5-fluoro-phenyl) -3, 6-dihydro-2H-pyridine-1-carboxylic acid tert-butyl ester

To a sealed tube were added 1-chloro-4-fluoro-2-iodo-benzene (1g, 3.90mmol), 4- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -3, 6-dihydro-2H-pyridine-1-carboxylic acid tert-butyl ester (1.21g, 3.90mmol), potassium carbonate (1.62g, 11.7mmol), [1, 1' -bis (diphenylphosphino) ferrocene]Palladium (II) dichloride (0.318g, 0.390mmol) and 1, 4-dioxane (10mL) and water (1 mL). The mixture was then degassed with nitrogen for 30 seconds and heated to 100 ℃ for 16 h. The reaction mixture was cooled to room temperature and purified by column chromatography (ISCO, 20g silica, 0-5% ethyl acetate in petroleum ether, 20min gradient) to give 4- (2) as a colorless gum -chloro-5-fluoro-phenyl) -3, 6-dihydro-2H-pyridine-1-carboxylic acid tert-butyl ester (0.860g, 2.76mmol, 71%). LCMS (ESI) M/z 256.3[ M + H-56 ]]+

Step 2: preparation of tert-butyl 4- (2-chloro-5-fluoro-phenyl) piperidine-1-carboxylate and tert-butyl 4- (3-fluorophenyl) piperidine-1-carboxylate

To a solution of tert-butyl 4- (2-chloro-5-fluoro-phenyl) -3, 6-dihydro-2H-pyridine-1-carboxylate (0.700g, 2.25mmol) in methanol (5mL) under nitrogen was added palladium on charcoal (10 wt% Pd, 0.05 g). The suspension was degassed under vacuum and purged with hydrogen (3 ×). The mixture was then stirred under hydrogen (15psi) at 15 ℃ for 40 h. The mixture was filtered and the filtrate was concentrated in vacuo. The crude product was purified by preparative-HPLC (Waters X bridge 150X 255 μm column; acetonitrile in 10mM aqueous ammonium bicarbonate solution 51-71%, 11min gradient) to afford first tert-butyl 4- (2-chloro-5-fluoro-phenyl) piperidine-1-carboxylate (0.050g, 0.159mmol, 7%) as a white solid. LCMS (ESI) M/z 257.9[ M + H-56 ]]+And secondly tert-butyl 4- (3-fluorophenyl) piperidine-1-carboxylate (0.160g, 0.573mmol, 25%) as a white solid.

And step 3: preparation of 4- (2-chloro-5-fluoro-phenyl) piperidine

To a stirred solution of tert-butyl 4- (2-chloro-5-fluoro-phenyl) piperidine-1-carboxylate (0.044g, 0.140mmol) in dichloromethane (3mL) was added trifluoroacetic acid (1 mL). The reaction mixture was stirred at 20 ℃ for 1h, then concentrated to give 4- (2-chloro-5-fluoro-phenyl) piperidine (0.050g, crude, trifluoroacetate salt) as a yellow gum. LCMS (ESI) M/z 214.4[ M + H ] ]+. This material was used in the next step without further purification.

And 4, step 4: preparation of [5- (1, 3-benzodioxol-5-yl) isoxazol-3-yl ] - [4- (2-chloro-5-fluoro-phenyl) -1-piperidinyl ] methanone

To a stirred solution of 5- (1, 3-benzodioxol-5-yl) isoxazole-3-carboxylic acid (32mg, 0.137mmol) in N, N ' -dimethylformamide (2mL) were added N, ' N ' -tetramethyl-O- (1H-benzotriazol-1-yl) uronium hexafluorophosphate (62mg, 0.165mmol), diisopropylethylamine (89mg, 0.687mmol) and 4- (2-chloro-5-fluoro-phenyl) piperidine trifluoroacetate (45mg, 0.137 mmol). The mixture was then stirred at 20 ℃ for 1 h. The mixture was directly purified by preparative-HPLC (Waters X bridge 150X 255 μm column; acetonitrile in 10mM aqueous ammonium bicarbonate solution 46-66%, 11min gradient) to give [5- (1, 3-benzodioxol-5-yl) isoxazol-3-yl as a yellow solid]- [4- (2-chloro-5-fluoro-phenyl) -1-piperidinyl]Methanone (31mg, 0.071mmol, 52%).1H NMR (400MHz, chloroform-d) δ 7.37-7.30(m, 2H), 7.25(s, 1H), 6.97(dd, J ═ 2.0, 9.6Hz, 1H), 6.94-6.85(m, 2H), 6.70(s, 1H), 6.06(s, 2H), 4.94(br.d, J ═ 13.2Hz, 1H), 4.70(d, J ═ 13.4Hz, 1H), 3.41-3.22(m, 2H), 2.94(t, J ═ 11.2Hz, 1H), 2.11-1.91(m, 2H), 1.78-1.59(m, 2H); LCMS (ESI M/z:429.0[ M + H ] ]+

Example 117.preparation of 4- (2-chloro-5-fluoro-phenyl) -1-piperidinyl ] - (5-phenylisoxazol-3-yl) methanone (310)

Step 1: preparation of 4- (2-chloro-5-fluoro-phenyl) -3, 6-dihydro-2H-pyridine-1-carboxylic acid tert-butyl ester

To a sealed tube were added 1-chloro-4-fluoro-2-iodo-benzene (1.0g, 3.90mmol), 4- (4, 4, 5, 5-tetramethyl-1, 3, 2-dioxaborolan in that orderCyclopentan-2-yl) -3, 6-dihydro-2H-pyridine-1-carboxylic acid tert-butyl ester (1.21g, 3.90mmol), potassium carbonate (1.62g, 11.7mmol), [1, 1' -bis (diphenylphosphino) ferrocene]Palladium (II) dichloride (0.318g, 0.390mmol) and 1, 4-dioxane (10mL) and water (1 mL). The mixture was then degassed with nitrogen for 30 seconds and heated to 100 ℃ for 16 h. The reaction mixture was concentrated in vacuo. The crude product was purified by column chromatography (ISCO, 20g silica, 0-5% ethyl acetate in petroleum ether, 20min gradient) to give tert-butyl 4- (2-chloro-5-fluoro-phenyl) -3, 6-dihydro-2H-pyridine-1-carboxylate (0.860g, 2.76mmol, 71%) as a colourless gum. LCMS (ESI) M/z 256.3[ M + H-56 ]]+

Step 2: preparation of tert-butyl 4- (2-chloro-5-fluoro-phenyl) piperidine-1-carboxylate and tert-butyl 4- (3-fluorophenyl) piperidine-1-carboxylate

To a solution of tert-butyl 4- (2-chloro-5-fluoro-phenyl) -3, 6-dihydro-2H-pyridine-1-carboxylate (0.300g, 0.962mmol) in methanol (5mL) under nitrogen was added palladium on charcoal (0.05g, 0.962mmol, 10 wt% Pd). The suspension was degassed under vacuum and purged with hydrogen (3 ×). The reaction mixture was stirred under hydrogen (15psi) at 15 ℃ for 16 h. The mixture was filtered and the filtrate was concentrated in vacuo. A mixture of tert-butyl 4- (2-chloro-5-fluoro-phenyl) piperidine-1-carboxylate and tert-butyl 4- (3-fluorophenyl) piperidine-1-carboxylate (0.280g, crude, 1:1) was obtained as a yellow gum. This material was used in the next step without further purification.

And step 3: preparation of 4- (2-chloro-5-fluoro-phenyl) piperidine and 4- (3-fluorophenyl) piperidine

To a mixture of tert-butyl 4- (2-chloro-5-fluoro-phenyl) piperidine-1-carboxylate and tert-butyl 4- (3-fluorophenyl) piperidine-1-carboxylate (0.280g, 0.892mmol) in dichloromethane (4mL) was added trifluoroacetic acid (1 mL). The mixture was then stirred at 20 ℃ for 2 h. The mixture was concentrated in vacuo to give a mixture of 4- (2-chloro-5-fluoro-phenyl) piperidine (trifluoroacetic acid) and 4- (3-fluorophenyl) piperidine (300 mg total, crude, trifluoroacetic acid, 1:1) as a yellow gum. This material was used in the next step without further purification.

And 4, step 4: preparation of [5- (1, 3-benzodioxol-5-yl) isoxazol-3-yl ] - [4- (2-chloro-5-fluoro-phenyl) -1-piperidinyl ] methanone

To a stirred solution of 5-phenylisoxazole-3-carboxylic acid (0.120g, 0.634mmol) in N, N' -dimethylformamide (4mL) was added [4- (2-chloro-5-fluoro-phenyl) piperidine and 4- (3-fluorophenyl) piperidine (0.250mg, 0.763mmol, trifluoroacetate, mixture, 1:1)]Then N, N, N, 'N' -tetramethyl-O- (1H-benzotriazol-1-yl) uronium hexafluorophosphate (0.289g, 0.761mmol) and diisopropylethylamine (0.410g, 3.17mmol) were added. The mixture was stirred at 20 ℃ for 2h and purified directly by preparative-HPLC (Waters X bridge 150X 255 μm column; acetonitrile in 10mM aqueous ammonium bicarbonate solution 46-70%, 11min gradient) to give [4- (2-chloro-5-fluoro-phenyl) -1-piperidinyl group as a brown solid ]- (5-phenylisoxazol-3-yl) methanone (49mg, 0.125mmol, 20%).1H NMR (400MHz, chloroform-d) δ 7.88-7.76(m, 2H), 7.55-7.44(m, 3H), 7.39-7.31(m, 1H), 6.98(dd, J ═ 2.4, 9.6Hz, 1H), 6.93-6.86(m, 1H), 6.85(d, J ═ 1.2Hz, 1H), 4.95(br.d, J ═ 13.2Hz, 1H), 6.70(d, J ═ 13.6Hz, 1H), 3.42-3.22(m, 2H), 2.94(td, J ═ 2.4, 13.2Hz, 1H), 2.10-1.93(m, 2H), 1.81-1.63(m, 2H); LCMS (ESI) M/z 385.0[ M + H ]]+

Example 118 preparation of N-methyl-2- [3- (4-phenylpiperidine-1-carbonyl) isoxazol-5-yl ] benzamide (301)

Step 1: preparation of 2-bromo-N-methyl-benzamide

To a stirred solution of 2-bromobenzoic acid (2.00g, 9.95mmol) and methylamine (2M, 25mL) in dichloromethane (50mL) was added propylphosphonic anhydride (9.50g, 14.9mmol, 50% in ethyl acetate) and triethylamine (3.02g, 29.9 mmol). The reaction mixture was stirred at 20 ℃ for 15 h. The reaction mixture was quenched with water (80mL) and extracted with dichloromethane (80 mL. times.3). The combined organic layers were washed with brine (150mL), dried over sodium sulfate, filtered and concentrated under reduced pressure. Crude 2-bromo-N-methyl-benzamide (1.20g, 5.61mmol, 56%) was obtained as a brown solid. 1H NMR (400MHz, chloroform-d) δ 7.50(d, J ═ 7.9Hz, 1H), 7.43(dd, J ═ 1.3, 7.6Hz, 1H), 7.26(t, J ═ 7.5Hz, 1H), 7.21-7.15(m, 1H), 6.07(br.s, 1H), 2.93(d, J ═ 4.9Hz, 3H); LCMS (ESI) M/z 214.0[ M + H ]]+. The material was used directly in the next step without further purification.

Step 2: preparation of ethyl 5- [2- (methylcarbamoyl) phenyl ] isoxazole-3-carboxylate

A solution of ethyl 5-tributylstannyl isoxazole-3-carboxylate (0.60g, 1.39mmol), 2-bromo-N-methyl-benzamide (0.358g, 1.67mmol) and bis (tri-tert-butylphosphine) palladium (0) (0.036g, 0.070mmol) in toluene (5mL) was degassed and then heated to 100 ℃ under nitrogen for 15 h. The reaction mixture was concentrated in vacuo. The crude product was purified by column chromatography (silica, petroleum ether/ethyl acetate 10/1-1/1) to give 5- [2- (methylcarbamoyl) phenyl ] as a dark brown oil]Isoxazole-3-carboxylic acid ethyl ester (0.090g, 0.328mmol, 24%).1H NMR (400MHz, chloroform-d) δ 7.81(d, J ═ 7.3Hz, 1H), 7.65-7.41(m, 3H), 6.94(s, 1H), 5.94-5.74(m, 1H), 4.46(q, J ═ 7.2Hz, 2H), 2.99(d, J ═ 4.9Hz, 3H), 1.43(t,J=7.1Hz,3H);LCMS(ESI)m/z:275.0[M+H]+

And step 3: preparation of 5- [2- (methylcarbamoyl) phenyl ] isoxazole-3-carboxylic acid

To the 5- [2- (methylcarbamoyl) phenyl group]To a stirred solution of isoxazole-3-carboxylic acid ethyl ester (90mg, 0.328mmol) in tetrahydrofuran (2mL) and water (2mL) was added lithium hydroxide (24mg, 0.984 mmol). The reaction mixture was stirred at 20 ℃ for 2 h. The reaction mixture was concentrated and the residue was dissolved in water (3 mL). The aqueous phase was extracted with methyl tert-butyl ether (3mL × 4) and the aqueous phase was adjusted to pH 3 with 1N aqueous hydrogen chloride solution. The aqueous phase was extracted with dichloromethane (3 mL. times.4). The combined organic phases were dried over anhydrous sodium sulfate, filtered and concentrated in vacuo. The compound 5- [2- (methylcarbamoyl) phenyl ] was obtained as a yellow oil]Isoxazole-3-carboxylic acid (0.080g, 0.325mmol, 99%). The material was used directly in the procedure. LCMS (ESI) M/z 246.9[ M + H ]]+

And 4, step 4: preparation of N-methyl-2- [3- (4-phenylpiperidine-1-carbonyl) isoxazol-5-yl ] benzamide

To the 5- [2- (methylcarbamoyl) phenyl group]To a stirred solution of isoxazole-3-carboxylic acid (40mg, 0.163mmol) and 4-phenylpiperidine (26mg, 0.163mmol) in dichloromethane (1mL) were added triethylamine (49mg, 0.487mmol) and propylphosphonic anhydride (124mg, 0.195mmol, 50% in ethyl acetate). The reaction mixture was stirred at 20 ℃ for 2 h. The reaction mixture was concentrated and the residue was purified by preparative-HPLC (Waters Xbridge 150X 255 μm column; 25-60% acetonitrile in 10mM aqueous ammonium bicarbonate, 11min gradient) to give N-methyl-2- [3- (4-phenylpiperidine-1-carbonyl) isoxazol-5-yl as a brown solid ]Benzamide (46mg, 0.118mmol, 73%).1H NMR (400MHz, chloroform-d) δ 7.84(d, J ═ d7.5Hz,1H),7.60-7.48(m,3H),7.37-7.29(m,2H),7.26-7.19(m,3H),6.82(s,1H),5.89(br.d,J=4.4Hz,1H),4.89(td,J=1.8,13.1Hz,1H),4.57-4.49(m,1H),3.30-3.20(m,1H),3.00(d,J=4.9Hz,3H),2.95-2.79(m,2H),2.05-1.90(m,2H),1.86-1.72(m,2H);LCMS(ESI)m/z:390.2[M+H]+

Example 119 preparation of N-methyl-4- [3- (4-phenylpiperidine-1-carbonyl) isoxazol-5-yl ] benzamide (304)

N-methyl-4- [3- (4-phenylpiperidine-1-carbonyl) isoxazol-5-yl was carried out following the same procedure as in example 97]And (3) synthesizing benzamide. The compound N-methyl-4- [3- (4-phenylpiperidine-1-carbonyl) isoxazol-5-yl is obtained as a white solid]Benzamide (0.075g, 0.193mmol, 53%).1H NMR (400MHz, chloroform-d) δ 7.97-7.83(m, 4H), 7.38-7.32(m, 2H), 7.28-7.22(m, 3H), 6.90(s, 1H), 6.36(br.d, J ═ 4.5Hz, 1H), 4.93(td, J ═ 1.9, 13.3Hz, 1H), 4.72-4.58(m, 1H), 3.37-3.24(m, 1H), 3.07(d, J ═ 4.9Hz, 3H), 2.99-2.90(m, 1H), 2.90-2.82(m, 1H), 2.15-1.94(m, 2H), 1.88-1.78(m, 2H); LCMS (ESI) M/z 390.2[ M + H ]]+

Example 120.5- (1, 3-benzodioxol-5-yl) isoxazol-3-yl ] - [4- (3-fluorophenyl) -1-piperidinyl ] methanone (309)

Step 1: preparation of tert-butyl 4- (2-chloro-5-fluoro-phenyl) piperidine-1-carboxylate and tert-butyl 4- (3-fluorophenyl) piperidine-1-carboxylate

To 4- (3-fluorophenyl) piperidine-1-carboxylic acid tert-butyl ester (b 0.160g, 0.573mmol) in dichloromethane (4mL) was added trifluoroacetic acid (1 mL). The mixture was stirred at 15 ℃ for 2 h. The reaction mixture was concentrated to give 4- (3-fluorophenyl) piperidine trifluoroacetate salt (0.240g, crude) as a yellow gum. LCMS (ESI) M/z 179.9[ M + H ]]+

Step 2: preparation of [5- (1, 3-benzodioxol-5-yl) isoxazol-3-yl ] - [4- (2-chloro-5-fluoro-phenyl) -1-piperidinyl ] methanone

To a solution of 5- (1, 3-benzodioxol-5-yl) isoxazole-3-carboxylic acid (0.065g, 0.279mmol) in N, N '-dimethylformamide (3mL) were added N, N' -tetramethyl-O- (1H-benzotriazol-1-yl) uronium hexafluorophosphate (0.127g, 0.334mmol), diisopropylethylamine (0.180g, 1.39mmol) and 4- (3-fluorophenyl) piperidine (0.050g, 0.279 mmol). The mixture was stirred at 15 ℃ for 1h and then purified directly by preparative-HPLC (Waters X bridge 150X 255. mu.M column; acetonitrile in 10mM aqueous ammonium bicarbonate solution 41-76%, 11min gradient) to give [5- (1, 3-benzodioxol-5-yl) isoxazol-3-yl as a pale yellow solid]- [4- (3-fluorophenyl) -1-piperidinyl group]Methanone (0.075g, 0.191mmol, 69%).1H NMR (400MHz, chloroform-d) δ 7.35(dd, J ═ 1.6, 8.0Hz, 1H), 7.32 to 7.28(m, 1H), 7.25(d, J ═ 2.0Hz, 1H), 7.01(d, J ═ 7.6Hz, 1H), 6.97 to 6.88(m, 3H), 6.69(s, 1H), 6.06(s, 2H), 4.97 to 4.86(m, 1H), 4.70 to 4.61(m, 1H), 3.26(td, J ═ 2.4, 13.2Hz, 1H), 2.97 to 2.80(m, 2H), 2.06 to 1.90(m, 2H), 1.84 to 1.67(m, 2H); LCMS (ESI) M/z 395.1[ M + H ] ]+

EXAMPLE 121 preparation of [4- (3-fluorophenyl) -1-piperidinyl ] - (5-phenylisoxazol-3-yl) methanone (307)

Prepared according to the procedure described for example 120Reported synthetic procedure for the synthesis of [4- (3-fluorophenyl) -1-piperidinyl]- (5-phenylisoxazol-3-yl) methanones. The compound [4- (3-fluorophenyl) -1-piperidinyl group ] is obtained as a yellow solid]- (5-phenylisoxazol-3-yl) methanone (66mg, 0.186mmol, 64%).1H NMR (400MHz, chloroform-d) δ 7.85-7.65(m, 2H), 7.52-7.36(m, 3H), 7.32-7.18(m, 1H), 6.97(br.d, J ═ 7.6Hz, 1H), 6.92-6.82(m, 2H), 6.80(s, 1H), 4.87(br.d, J ═ 13.2Hz, 1H), 4.63(br.d, J ═ 13.6Hz, 1H), 3.22(td, J ═ 2.4, 13.2Hz, 1H), 2.94-2.68(m, 2H), 2.06-1.84(m, 2H), 1.82-1.64(m, 2H); LCMS (ESI) M/z 351.2[ M + H ]]+

EXAMPLE 122.preparation of 4- {1- [5- (2H-1, 3-benzodioxol-5-yl) -1, 3-oxazole-2-carbonyl ] piperidin-4-yl } benzonitrile (291)

To 4- (piperidin-4-yl) benzonitrile (0.06g, 0.4mmol), 5- (benzo [ d ] at 23 deg.C][1,3]Dioxol-5-yl) isoxazole-3-carboxylic acid (0.1g, 0.4mmol) in N, N-dimethylformamide (2mL) was added to a stirred solution of N, N' -tetramethyl-O- (1H-benzotriazol-1-yl) uronium hexafluorophosphate (0.06g, 0.4mmol) and diisopropylethylamine (0.05g, 0.5 mmol). The reaction mixture was stirred at 23 ℃ for 12H and then purified by column chromatography (ISCO, silica, 0-100% ethyl acetate) to give 4- {1- [5- (2H-1, 3-benzodioxol-5-yl) -1, 3-oxazole-2-carbonyl as an orange solid ]Piperidin-4-yl } benzonitrile (0.096g, 0.24mmol, 60%).1H NMR (300MHz, chloroform-d) δ 7.35(dd, J ═ 8.2, 1.7Hz, 1H), 7.31-7.14(m, 4H), 7.08-6.88(m, 3H), 6.70(s, 1H), 6.38(s, 0H), 6.07(s, 2H), 4.92(d, J ═ 13.1Hz, 1H), 4.65(d, J ═ 13.6Hz, 1H), 3.26(t, J ═ 11.7Hz, 1H), 2.97-2.77(m, 1H), 1.95(d, J ═ 16.4Hz, 2H), 1.78(t, J ═ 12.6Hz, 1H); LCMS (ESI) M/z 402.1[ M + H ]]+

EXAMPLE 123.1 preparation of- [5- (2H-1, 3-benzodioxol-5-yl) -1, 3-oxazole-2-carbonyl ] -4- (3, 4-difluorophenyl) piperidine (292)

To 4- (3, 4-difluorophenyl) piperidine hydrochloride (0.02g, 0.09mmol), 5- (benzo [ d ] at 23 deg.C][1,3]To a stirred solution of dioxol-5-yl) isoxazole-3-carboxylic acid (0.02g, 0.09mmol) in N, N-dimethylformamide (3mL) were added N, N' -tetramethyl-O- (1H-benzotriazol-1-yl) uronium hexafluorophosphate (0.1g, 0.09mmol) and diisopropylethylamine (0.1g, 0.09 mmol). The reaction mixture was stirred at 23 ℃ for 12H and then purified by column chromatography (ISCO, 4g silica, 0-100% ethyl acetate) to give 1- [5- (2H-1, 3-benzodioxol-5-yl) -1, 3-oxazole-2-carbonyl as a white solid ]-4- (3, 4-difluorophenyl) piperidine (0.02g, 0.mmol, 53%).1H NMR (300MHz, chloroform-d) δ 8.57(d, J ═ 5.1Hz, 2H), 7.40-7.22(m, 2H), 7.18(d, J ═ 5.7Hz, 2H), 6.93(d, J ═ 8.2Hz, 1H), 6.71(s, 1H), 6.07(d, J ═ 0.7Hz, 2H), 5.32(s, 0H), 4.94(d, J ═ 13.5Hz, 1H), 4.72(d, J ═ 14.0Hz, 1H), 4.14(d, J ═ 7.1Hz, 1H), 3.28(t, J ═ 12.0Hz, 1H), 2.90(q, J ═ 13.0Hz, 2H), 2.09-1.92(m, 2H), 1.81(t, J ═ 12.0Hz, 1H), t, 1H), 2.9, t ═ 13.5H, 1H); LCMS (ESI) M/z 413.4[ M + H ]]+

EXAMPLE 124.4 preparation of- [5- (1, 3-benzodioxol-5-yl) isoxazole-3-carbonyl ] -1-phenyl-piperazin-2-one (280)

To a stirred solution of 5- (1, 3-benzodioxol-5-yl) isoxazole-3-carboxylic acid (40mg, 0.172mmol) in N, N '-dimethylformamide (2mL) was added N, N' -tetramethyl-O- (1H-benzotriazol-1-yl) uronium hexafluorophosphate (0.078g, 0.206mmol), diisopropylethylamine (0.067g, 0.515mmol) and 1-phenylpiperazin-2-one (0.033g, 0.189mmol) at 30 ℃. The mixture was then stirred at 30 ℃ for 1 h. Mixing the mixturePurification by preparative-HPLC (Waters X bridge 150X 255 μm column; acetonitrile 25-55% in 10mM aqueous ammonium bicarbonate, 12min gradient) gave 4- [5- (1, 3-benzodioxol-5-yl) isoxazole-3-carbonyl as a brown solid ]-1-phenyl-piperazin-2-one (26mg, 0.067mmol, 39%).1H NMR (400MHz, chloroform-d) δ 7.44(t, J ═ 7.2Hz, 2H), 7.38 to 7.28(m, 4H), 7.26(t, J ═ 1.6Hz, 1H), 6.93(d, J ═ 8.0Hz, 1H), 6.79(d, J ═ 18.0Hz, 1H), 6.07(s, 2H), 4.85(s, 1H), 4.61(s, 1H), 4.43(t, J ═ 4.2Hz, 1H), 4.17(t, J ═ 4.2Hz, 1H), 3.91 to 3.66(m, 2H); LCMS (ESI) M/z 392.1[ M + H ]]+

EXAMPLE 125.1 preparation of- [5- (2H-1, 3-benzodioxol-5-yl) -1, 2-oxazole-3-carbonyl ] -4-methyl-4-phenylpiperidine (290)

4-methyl-4-phenylpiperidine hydrochloride (0.25g, 1.2mmol), 5- (benzo [ d ] at 23 deg.C][1,3]To a stirred solution of dioxol-5-yl) isoxazole-3-carboxylic acid (0.17g, 1.1mmol) in N, N-dimethylformamide (2mL) were added N, N' -tetramethyl-O- (1H-benzotriazol-1-yl) uronium hexafluorophosphate (0.37g, 0.98mmol) and diisopropylethylamine (0.29g, 2.26 mmol). The reaction mixture was stirred at 23 ℃ for 12H and purified directly by column chromatography (ISCO, silica 12g, 0-100% ethyl acetate) to give 1- [5- (2H-1, 3-benzodioxol-5-yl) -1, 2-oxazole-3-carbonyl as a yellow solid]-4-methyl-4-phenylpiperidine (0.047g, 0.12mmol, 11%). 1H NMR (300MHz, chloroform-d) δ 7.42-7.21(m, 6H), 6.91(d, J ═ 8.1Hz, 1H), 6.65(d, J ═ 1.9Hz, 1H), 6.06(s, 1H), 6.06(d, J ═ 3.1Hz, 0H), 3.97(d, J ═ 13.3Hz, 2H), 3.73-3.59(m, 1H), 2.25(s, 2H), 1.84(s, 3H), 1.33(s, 2H); LCMS (ESI) M/z 391.2[ M + H ]]+

Example 126.preparation of 5- (1, 3-benzodioxol-5-yl) isoxazol-3-yl ] - [4- (2-chloro-5-fluoro-phenyl) -3, 6-dihydro-2H-pyridin-1-yl ] methanone (306)

[5- (1, 3-Benzodioxolen-5-yl) isoxazol-3-yl ] was carried out following the same procedure as in example 120]- [4- (2-chloro-5-fluoro-phenyl) -3, 6-dihydro-2H-pyridin-1-yl]And (5) synthesizing the ketone. The compound [5- (1, 3-benzodioxol-5-yl) isoxazol-3-yl ] is obtained as a yellow solid]- [4- (2-chloro-5-fluoro-phenyl) -3, 6-dihydro-2H-pyridin-1-yl]Methanone (0.095g, 0.222mmol, 57%).1H NMR (400MHz, chloroform-d) δ 7.42-7.31(m, 2H), 7.28-7.26(m, 1H), 7.07-6.85(m, 3H), 6.74(d, J ═ 12.8Hz, 1H), 6.06(s, 2H), 5.87-5.65(m, 1H), 4.70-4.35(m, 2H), 4.10(t, J ═ 5.4Hz, 1H), 4.03(t, J ═ 5.2Hz, 1H), 2.73-2.47(m, 2H); LCMS (ESI) M/z 427.1[ M + H ] ]+

Example 127.4- (2-chloro-5-fluoro-phenyl) -3, 6-dihydro-2H-pyridin-1-yl ] - (5-phenylisoxazol-3-yl) methanone (305)

4- (2-chloro-5-fluoro-phenyl) -3, 6-dihydro-2H-pyridin-1-yl was performed using the same procedure as in example 120]Synthesis of- (5-phenylisoxazol-3-yl) methanone. The compound 4- (2-chloro-5-fluoro-phenyl) -3, 6-dihydro-2H-pyridin-1-yl is obtained as a yellow solid]- (5-phenylisoxazol-3-yl) methanone (0.084g, 0.219mmol, 41%).1H NMR (400MHz, chloroform-d) δ 7.98-7.70(m, 2H), 7.62-7.38(m, 3H), 7.36-7.30(m, 1H), 7.05-6.80(m, 3H), 5.95-5.60(m, 1H), 4.60(d, J ═ 2.8Hz, 1H), 4.43(d, J ═ 2.8Hz, 1H), 4.11(t, J ═ 5.6Hz, 1H), 4.04(t, J ═ 5.6Hz, 1H), 2.75-2.46(m, 2H); LCMS (ESI) M/z 383.1[ M + H ]]+

EXAMPLE 128 preparation of [5- (1, 3-benzodioxol-5-yl) isoxazol-3-yl ] - (4-phenoxy-1-piperidinyl) methanone (298)

Synthesis of [5- (1, 3-benzodioxol-5-yl) isoxazol-3-yl ] according to the synthetic procedure reported for the preparation of example 95]- (4-phenoxy-1-piperidinyl) methanone. The compound [5- (1, 3-benzodioxol-5-yl) isoxazol-3-yl ] is obtained as a brown solid ]- (4-phenoxy-1-piperidinyl) methanone (0.059g, 0.150mmol, 35%).1H NMR (400MHz, methanol-d)4)δ7.42(dd,J=1.7,8.0Hz,1H),7.33(d,J=1.5Hz,1H),7.26(t,J=8.0Hz,2H),7.01-6.90(m,4H),6.85(s,1H),6.04(s,2H),4.75-4.65(m,1H),4.02-3.89(m,2H),3.86-3.77(m,1H),3.71(ddd,J=3.9,7.2,13.7Hz,1H),2.05(tdd,J=4.4,8.5,12.9Hz,2H),1.92-1.78(m,2H);LCMS(ESI)m/z:393.1[M+H]+

EXAMPLE 129 preparation of (4-phenoxy-1-piperidinyl) - (5-phenylisoxazol-3-yl) methanone (288)

The synthesis of (4-phenoxy-1-piperidinyl) - (5-phenylisoxazol-3-yl) methanone was performed following the same procedure as example 95. The compound (4-phenoxy-1-piperidinyl) - (5-phenylisoxazol-3-yl) methanone (0.100g, 0.287mmol, 54%) was obtained as a pale yellow solid.1H NMR (400MHz, chloroform-d) Δ 7.88-7.75(m, 2H), 7.54-7.45(m, 3H), 7.35-7.28(m, 2H), 7.02-6.92(m, 3H), 6.83(s, 1H), 4.65(br.s, 1H), 4.10-3.81(m, 4H), 2.11-1.89(m, 4H); LCMS (ESI) M/z 349.1[ M + H ]]+

EXAMPLE 130 preparation of (5- (benzo [ d ] [1,3] dioxol-5-yl) isoxazol-3-yl) (4- (3-methoxyphenyl) piperazin-1-yl) methanone (271)

To 1- (3-methoxyphenyl) piperazine (0.20g, 0.75mmol), 5- (benzo [ d ] at 23 deg.C][1,3]To a stirred solution of dioxol-5-yl) isoxazole-3-carboxylic acid (0.157g, 0.831mmol) in N, N-dimethylformamide (2.7mL) were added N, N' -tetramethyl-O- (1H-benzotriazol-1-yl) uronium hexafluorophosphate (0.37g, 0.98mmol) and diisopropylethylamine (0.29g, 2.26 mmol). The reaction mixture was stirred at 23 ℃ for 12h and purified by column chromatography (ISCO, 12g silica, 0-100% ethyl acetate) to give (5- (benzo [ d ] b) as a white solid ][1,3]Dioxol-5-yl) isoxazol-3-yl) (4- (3-methoxyphenyl) piperazin-1-yl) methanone (0.268g, 0.658mmol, 87%).1H NMR (300MHz, dimethylsulfoxide-d)6)δ7.54-7.42(m,2H),7.23-7.05(m,3H),6.61-6.36(m,3H),6.14(s,2H),3.85-3.68(m,7H),3.34-3.13(m,4H)。3.30(s,3H);LCMS(ESI)m/z:408.4[M+H]+

EXAMPLE 131 preparation of (4- (2-chloro-5-fluorophenyl) piperazin-1-yl) (5-phenylisoxazol-3-yl) methanone (297)

To a solution of (5-phenylisoxazol-3-yl) (piperazin-1-yl) methanone (0.25g, 0.851mmol) and 1-chloro-4-fluoro-2-iodobenzene (0.327g, 1.28mmol) in toluene (5mL) was added cesium carbonate (0.832g, 2.55mmol), tris (dibenzylideneacetone) dipalladium (0) (0.078g, 0.085mmol) and BINAP (0.053g, 0.085 mmol). The reaction mixture was purged with nitrogen for 3min and stirred in a sealed tube at 115 ℃ for 15 h. The mixture was quenched with water (10mL) and extracted with ethyl acetate (15 mL. times.3). The combined organic layers were washed with brine (5mL), dried over sodium sulfate, filtered and concentrated in vacuo. The crude product was purified by column chromatography (ISCO, 10g silica, 8-10% ethyl acetate in petroleum ether, 15min gradient) followed by preparative-HPLC (YMC-Actus ODS-AQ 100X 305 μm column; 45-85% acetonitrile in 0.05% aqueous hydrochloric acid, 12min gradient) to give (4- (2-chloro-5-fluorophenyl) piperazin-1-yl) (5-phenylisoxazol-3-yl) methanone hydrochloride as a white solid (0.172g of 0.172g ,0.404mmol,47%)。1H NMR (400MHz, chloroform-d) Δ 7.89-7.80(m, 2H), 7.57-7.46(m, 3H), 7.40-7.31(m, 1H), 6.89(s, 1H), 6.82-6.71(m, 2H), 4.20-4.10(m, 2H), 4.08-3.97(m, 2H), 3.18-3.13(m, 4H); LCMS (ESI) M/z 386.0[ M + H ]]+

Example 132.preparation of 5- (1, 3-benzodioxol-5-yl) isoxazol-3-yl ] - [4- (2-chloro-5-fluoro-phenyl) piperazin-1-yl ] methanone (299)

[5- (1, 3-Benzodioxolen-5-yl) isoxazol-3-yl ] was carried out following the same procedure as in example 131]- [4- (2-chloro-5-fluoro-phenyl) piperazin-1-yl]And (5) synthesizing the ketone. The compound [5- (1, 3-benzodioxol-5-yl) isoxazol-3-yl ] is obtained as a pale yellow solid]- [4- (2-chloro-5-fluoro-phenyl) piperazin-1-yl]Methanone (0.065g, 0.147mmol, 33%).1H NMR (400MHz, chloroform-d) δ 7.39-7.30(m, 2H), 7.25(d, J ═ 1.5Hz, 1H), 6.92(d, J ═ 8.2Hz, 1H), 6.81-6.68(m, 3H), 6.06(s, 2H), 4.18-4.06(m, 2H), 4.05-3.97(m, 2H), 3.14(td, J ═ 5.0, 10.4Hz, 4H); LCMS (ESI) M/z 430.0[ M + H ]]+

Example 133.5 preparation of phenyl isoxazol-3-yl) (4-phenylpiperazin-1-yl) methanone (266)

To a stirred solution of 5-phenylisoxazole-3-carboxylic acid (0.150g, 0.793mmol) and N, N '-tetramethyl-O- (1H-benzotriazol-1-yl) uronium hexafluorophosphate (0.301g, 0.793mmol) in N, N' -dimethylformamide (2mL) was added diisopropylethylamine (0.205g, 1.59mmol) and 1-phenylpiperazine (0.129g, 0.793 mmol). The mixture was stirred at 25 ℃ for 2h and then purified directly by preparative-HPLC (Luna C18100X 305 μm column; 53-83% acetonitrile in 10mM aqueous ammonium bicarbonate, 10min gradient) to give ((5-benzene) as a white solid Methylisoxazol-3-yl) (4-phenylpiperazin-1-yl) methanone (0.198g, 0.589mmol, 74%).1H NMR (400MHz, dimethylsulfoxide-d)6)δ7.94(dd,J=1.5,7.7Hz,2H),7.60-7.52(m,3H),7.33(s,1H),7.27-7.20(m,2H),6.97(d,J=7.9Hz,2H),6.82(t,J=7.2Hz,1H),3.81(td,J=4.9,15.1Hz,4H),3.26-3.15(m,4H);LCMS(ESI)m/z:334.2[M+H]+

EXAMPLE 134 preparation of (5- (benzo [ d ] [1,3] dioxol-5-yl) isoxazol-3-yl) (2-methyl-4-phenylpiperazin-1-yl) methanone (270)

To 3-methyl-1-phenylpiperazine (0.050g, 0.280mmol), 5- (benzo [ d ] at 15 deg.C][1,3]To a stirred solution of dioxol-5-yl) isoxazole-3-carboxylic acid (0.157g, 0.831mmol) in N, N-dimethylformamide (1mL) were added N, N' -tetramethyl-O- (1H-benzotriazol-1-yl) uronium hexafluorophosphate (0.315g, 0.831mmol) and diisopropylethylamine (0.242g, 1.39 mmol). The reaction mixture was stirred at 15 ℃ for 12h and purified by column chromatography (ISCO, 12g silica, 0-100% ethyl acetate) to give (5- (benzo [ d ] b) as a yellow oil][1,3]Dioxol-5-yl) isoxazol-3-yl) (2-methyl-4-phenylpiperazin-1-yl) methanone (0.105g, 0.268mmol, 95%).1H NMR (300MHz, dimethylsulfoxide-d)6)δ7.55-7.43(m,2H),7.29-7.05(m,2H),6.97-6.88(m,2H),6.78(t,J=6.7Hz,1H),6.14(d,J=1.4Hz,2H),5.76(s,1H),4.41(d,J=12.7Hz,1H),4.25(d,J=12.7Hz,1H),4.18-3.87(m,2H),3.42(d,J=14.7Hz,1H),3.04(d,J=11.6Hz,1H),2.40(d,J=18.9Hz,1H),0.91(dd,J=10.9,6.4Hz,2H);LCMS(ESI)m/z:392.4[M+H]+

Example 135 preparation of N- (1-benzyl-1H-pyrazol-4-yl) -5- (oxazol-5-yl) isoxazole-3-carboxamide (149)

Step 1: preparation of ethyl 5- (hydroxymethyl) isoxazole-3-carboxylate

To a solution of prop-2-yn-1-ol (3.3g, 59.4mmol) in N, N-dimethylformamide (20mL) was added dropwise ethyl (Z) -2-chloro-2- (hydroxyimino) acetate (3.0g, 19.8mmol) in N, N-dimethylformamide (20mL) under nitrogen over 40 min. After addition, the reaction mixture was heated to 90 ℃ and a solution of triethylamine (6.0g, 59.4mmol) in N, N-dimethylformamide (20mL) was added dropwise over 1 h. The reaction mixture was stirred at this temperature for 17h and then cooled to room temperature. The reaction mixture was diluted with ethyl acetate (30mL), washed with water (20mL × 3) and brine (20mL), dried over anhydrous sodium sulfate, filtered and concentrated in vacuo. The residue was purified by column chromatography (silica, petroleum ether/ethyl acetate-6/1) to give ethyl 5- (hydroxymethyl) isoxazole-3-carboxylate (1.4g, 8.19mmol, 41%) as a yellow oil. LCMS (ESI) M/z 172.1[ M + H ] ]+

Step 2: preparation of ethyl 5-formylisoxazole-3-carboxylate

To a solution of ethyl 5- (hydroxymethyl) isoxazole-3-carboxylate (1.4g, 8.18mmol) in ethyl acetate (30mL) was added 2-iodoxybenzoic acid (6.9g, 24.5 mmol). The reaction mixture was heated at 90 ℃ for 17h and then cooled to room temperature. The reaction mixture was filtered and the filtrate was concentrated in vacuo. The residue was purified by column chromatography (silica, petroleum ether/ethyl acetate-6/1) to give ethyl 5-formylisoxazole-3-carboxylate (1.1g, 6.5mmol, 80%) as a yellow oil.

And step 3: preparation of 5- (oxazol-5-yl) isoxazole-3-carboxylic acids

To a solution of 1- (isocyanomethylsulfonyl) -4-toluene (0.460g, 2.36mmol) in acetonitrile (15mL) was added potassium carbonate (0.391g, 2.83 mmol). The reaction mixture was stirred at 25 ℃ for 1h, then ethyl 5-formylisoxazole-3-carboxylate (0.400g, 2.36mmol) was added at 0 ℃. The reaction mixture was heated to 90 ℃ and stirred for 17 h. The reaction mixture was cooled to room temperature, diluted with water (20mL) and the aqueous layer was adjusted to pH 3 with 1N aqueous hydrogen chloride. The reaction mixture was then extracted with ethyl acetate (10 mL. times.3). The combined organic layers were washed with brine (10mL × 2), dried over anhydrous sodium sulfate, filtered and concentrated in vacuo. Crude 5- (oxazol-5-yl) isoxazole-3-carboxylic acid (0.160g, 0.89mmol, 38%) was obtained as a white solid. This material was used directly in the next step without further purification.

And 4, step 4: preparation of N- (1-benzyl-1H-pyrazol-4-yl) -5- (oxazol-5-yl) isoxazole-3-carboxamide

To a solution of 5- (oxazol-5-yl) isoxazole-3-carboxylic acid (0.140g, 0.77mmol) in N, N-dimethylformamide (16mL) was added 1-benzyl-1H-pyrazol-4-amine (0.133g, 0.77mmol), 1- [ bis (dimethylamino) methylene ] -amine]-1H-1, 2, 3-triazolo [4, 5-b]Pyridinium 3-oxide hexafluorophosphate (0.878g, 2.31mmol) and diisopropylethylamine (0.298g, 2.31 mmol). The mixture was stirred at 25 ℃ for 17 h. The crude sample was dissolved in a minimal amount of N, N-dimethylformamide and purified via preparative-HPLC (Boston C1821 × 250mm 10 μm column; acetonitrile/0.01% aqueous trifluoroacetic acid) to give N- (1-benzyl-1H-pyrazol-4-yl) -5- (oxazol-5-yl) isoxazole-3-carboxamide (20.4mg, 0.06mmol, 8%) as a white solid.1H NMR (500MHz, dimethylsulfoxide-d)6)δ11.09(s,1H),8.73(s,1H),8.17(s,1H),8.02(s,1H),7.67(s,1H),7.30(ddd,J=29.6,18.3,7.0Hz,6H),5.34(s,2H);LCMS(ESI)m/z:336.1[M+H]+

EXAMPLE 136 characterization data for Compounds of the invention

The following compounds were synthesized by a method similar to the above method.

Compound 1:1H NMR(400MHz,CDCl3)δ7.87(br.s.,1H),7.61(s,1H),7.00(d,J=3.1Hz,1H),6.94(s,1H),6.63-6.55(m,1H),3.75(s,3H),2.20(d,J=5.3Hz,6H);LCMS(ESI)m/z:287.1[M+H]+

compound 2:1H NMR(400MHz,CDCl3)δ7.61(d,J=1.1Hz,1H),7.37-7.29(m,3H),7.13(d,J=6.9Hz,2H),7.00(d,J=3.5Hz,1H),6.93(s,1H),6.59(dd,J=1.8,3.5Hz,1H),5.25(s,2H),2.24(s,3H),2.14(s,3H);LCMS(ESI)m/z:363.2[M+H]+

compound 9:1H NMR(400MHz,DMSO-d6)δ7.98-7.88(m,2H),7.63-7.51(m,3H),7.48-7.33(m,2H),7.23-7.08(m,3H),4.77(s,1H),4.74(s,1H),3.11(s,1.5H),2.96(s,1.5H);LCMS(ESI)m/z:311.0[M+H]+

compound 10:1h NMR (400MHz, chloroform-d) δ 8.54(br s, 1H), 8.02(s, 1H), 7.63-7.54(m, 2H), 7.00(d, J ═ 3.5Hz, 1H), 6.94(s, 1H), 6.59(dd, J ═ 1.7, 3.3Hz, 1H), 3.93(s, 3H); LCMS (ESI) M/z 259.0[ M + H ] ]+

Compound 12:1h NMR (400MHz, chloroform-d) δ 8.69-8.53(m, 2H), 7.87(s, 1H), 7.60(s, 1H), 6.99(d, J ═ 3.4Hz, 1H), 6.93(s, 1H), 6.63-6.53(m, 1H), 1.66(s, 9H); LCMS (ESI) M/z 345.1[ M + H ]]+

Compound 13:1H NMR(400MHz,DMSO-d6)δ12.72(br s,1H),10.96(s,1H),7.99(d,J=1.1Hz,2H),7.70(br s,1H),7.27(d,J=3.5Hz,1H),7.14(s,1H),6.76(dd,J=1.8,3.5Hz,1H);LCMS(ESI)m/z:245.0[M+H]+

compound 15:1h NMR (400MHz, chloroform-d) δ 8.53(br.s, 1H), 8.05(s, 1H), 7.70-7.53(m, 2H), 7.33(d, J ═ 8.4Hz, 2H), 7.19(d, J ═ 8.4Hz, 2H), and,J=8.4Hz,2H),6.99(d,J=3.6Hz,1H),6.91(s,1H),6.64-6.60(m,1H),5.27(s,2H);LCMS(ESI)m/z:369.0[M+H]+

compound 16:1h NMR (400MHz, chloroform-d) δ 8.55(br s, 1H), 8.10(s, 1H), 7.62(br d, J9.2 Hz, 3H), 7.35(br d, J8.0 Hz, 2H), 7.00(d, J3.2 Hz, 1H), 6.92(s, 1H), 6.58(br d, J1.2 Hz, 1H), 5.37(s, 2H); LCMS (ESI) M/z 403.1[ M + H ]]+

Compound 21:1h NMR (400MHz, chloroform-d) δ 8.07(s, 1H), 7.83(s, 1H), 7.61(d, J1.6 Hz, 1H), 7.41(d, J8.4 Hz, 1H), 7.24(d, J2.0 Hz, 1H), 7.00(d, J3.2 Hz, 1H), 6.96(dd, J1.6, 8.3Hz, 1H), 6.94(s, 1H), 6.59(dd, J2.0, 3.3Hz, 1H), 5.26(s, 2H), 2.21(s, 3H); LCMS (ESI) M/z 417.0[ M + H ]]+

Compound 24:1h NMR (400MHz, chloroform-d) δ 8.48(s, 1H), 7.86(s, 1H), 7.61-7.57(m, 2H), 7.32-7.20(m, 3H), 7.13(d, J ═ 7.1Hz, 2H), 6.98(d, J ═ 3.5Hz, 1H), 6.91(s, 1H), 6.57(dd, J ═ 1.9, 3.4Hz, 1H), 4.33(t, J ═ 7.5Hz, 2H), 3.18(t, J ═ 7.5Hz, 2H); LCMS (ESI) M/z 349.1[ M + H ] ]+

Compound 25:1h NMR (400MHz, chloroform-d) Δ 8.47(s, 2H), 7.95-7.87(m, 2H), 7.59-7.49(m, 3H), 7.23(s, 1H); LCMS (ESI) M/z 255.1[ M + H ]]+

Compound 27:1H NMR(400MHz,DMSO-d6)δ10.82(s,1H),8.01(s,1H),7.60-7.50(m,2H),7.33-7.24(m,2H),7.20(s,1H),6.77(br d,J=1.3Hz,1H),6.13(s,1H),3.86(s,2H),3.66(s,3H);LCMS(ESI)m/z:417.0[M+H]+

compound 29:1H NMR(400MHz,DMSO-d6)δ11.03(s,1H),8.23(s,1H),8.00(d,J=1.2Hz,1H),7.69(s,1H),7.62(d,J=8.3Hz,1H),7.51(d,J=1.8Hz,1H),7.28(d,J=3.5Hz,1H),7.21(dd,J=2.0,8.3Hz,1H),7.15(s,1H),6.77(dd,J=1.7,3.4Hz,1H),5.36(s,2H);LCMS(ESI)m/z:403.0[M+H]+

compound 31:1h NMR (400MHz, chloroform-d) δ 8.57(s, 1H), 8.04(s, 1H), 7.63-7.55(m, 2H), 6.99(d, J ═ 3.3Hz, 1H), 6.93(s, 1H), 6.58(dd, J ═ 1.7, 3.4Hz, 1H), 4.01-3.95(m, 4H), 3.37(dt, J ═ 1.8, 11.7Hz, 2H), 2.18(ttt, J ═ 3.8, 7.5, 11.4Hz, 1H), 1.55-1.49(m, 2H), 1.44-1.33(m, 2H); LCMS (ESI) M/z 343.1[ M + H ]]+

Compound 32:1H NMR(400MHz,DMSO-d6)δ8.24(s,0.1H),7.97(s,0.9H),7.72(s,0.75H),7.62(s,0.25H),7.46-7.25(m,5.4H),7.24-7.04(m,3.6H),6.85-6.70(m,3H),5.32-5.13(m,2H),5.12-4.90(m,2H);LCMS(ESI)m/z:493.1[M+H]+

compound 33:1h NMR (400MHz, chloroform-d) δ 8.65(s, 1H), 8.59(s, 1H), 7.80(s, 1H), 7.71(d, J7.9 Hz, 2H), 7.60(s, 1H), 7.46(t, J7.9 Hz, 2H), 7.32-7.27(m, 1H), 6.99(d, J3.5 Hz, 1H), 6.95(s, 1H), 6.58(dd, J1.7, 3.4Hz, 1H); LCMS (ESI) M/z 321.0[ M + H ]]+

Compound 35:1h NMR (400MHz, chloroform-d) Δ 8.63(s, 1H), 8.61(s, 1H), 7.89(s, 1H), 7.86-7.80(m, 2H), 7.56-7.47(m, 3H), 7.05(s, 1H), 1.68(s, 9H); LCMS (ESI) M/z 255.1[ M + H ]]+

Compound 39: 1H NMR (400MHz, DMSO-d6) δ 11.10(s, 1H), 8.73(s, 2H), 8.33(s, 1H), 8.05(d, J ═ 8.0Hz, 1H), 7.99-7.96(m, 2H), 7.77-7.73(m, 2H), 7.61-7.54(m, 3H), 7.47(s, 1H), 5.52(s, 2H); LCMS (ESI) M/z 346.1[ M + H ] ]+

Compound 40: 1H NMR (400MHz, DMSO-d6) δ 11.08(s, 1H), 8.58(s, 1H), 8.25(s, 1H), 7.99-7.97(m, 2H), 7.85(t, J ═ 7.6Hz, 1H), 7.72(s, 1H), 7.58-7.57(m, 3H), 7.48-7.37(m, 2H), 7.10(d, J ═ 8.4Hz, 1H), 7.47(s, 1H), 5.48(s, 2H); LCMS (ESI) M/z 346.1[ M + H ]]+

Compound 41: 1H NMR (400MHz, DMSO — d6) δ 10.99(s, 1H), 8.12(s, 1H), 7.98(dd, J ═ 7.6, 2.0Hz, 2H), 7.65(s, 1H), 7.61-7.52(m, 3H), 7.46(s, 1H), 3.97(d,J=7.0Hz,2H),1.28-1.16(m,1H),0.57-0.44(m,2H),0.40-0.29(m,2H);LCMS(ESI)m/z:309.1[M+H]+

Compound 42: 1H NMR (500MHz, DMSO-d6) δ 11.07(s, 1H), 8.26(s, 1H), 7.97(dd, J ═ 7.5Hz, J ═ 2.0Hz, 2H), 7.83(d, J ═ 8.5Hz, 2H), 7.72(s, 1H), 7.61-7.52(m, 3H), 7.46(s, 1H), 7.36(d, J ═ 8.5Hz, 2H), 5.47(s, 2H); LCMS (ESI) M/z 370.1[ M + H ]]+

Compound 43: 1H NMR (500MHz, DMSO-d6) δ 11.08(s, 1H), 8.27(s, 1H), 7.97(d, J ═ 6.0Hz, 2H), 7.86(d, J ═ 3.0Hz, 1H), 7.73(s, 1H), 7.58-7.57(m, 3H), 7.47(s, 1H), 7.25(d, J ═ 3.5Hz, 1H), 5.66(s, 2H); LCMS (ESI) M/z 376.1[ M + H]+

Compound 44: LCMS (ESI) M/z 346.1[ M + H ]]+

Compound 45: 1H NMR (500MHz, DMSO-d6) δ 11.13(s, 1H), 9.35(d, J ═ 1.5Hz, 1H), 8.84(dd, J ═ 2.5, 1.5Hz, 1H), 8.81(d, J ═ 1.5Hz, 1H), 8.23(s, 1H), 7.70(s, 1H), 7.69(s, 1H), 7.41-7.37(m, 1H), 7.13(td, J ═ 8.5, 2.0Hz, 1H), 7.08-7.04(m, 2H), 5.37(s, 2H); LCMS (ESI) M/z 365.1[ M + H ] ]+

Compound 46: 1H NMR (500MHz, DMSO-d6) δ 11.13(s, 1H), 9.04(d, J ═ 4.0Hz, 2H), 8.23(s, 1H), 7.70(s, 1H), 7.68-7.65(m, 1H), 7.55(s, 1H), 7.40-7.38(m, 1H), 7.15-7.12(m, 1H), 7.07-7.04(m, 2H), 5.37(s, 2H); LCMS (ESI) M/z 365.1[ M + H ]]+

Compound 47: LCMS (ESI) M/z 485.1[ M + H ]]+

Compound 48:1H NMR(400MHz,DMSO-d6)δ11.07(br s,1H),8.45(s,1H),8.15(s,1H),8.08(s,1H),7.66(s,1H),7.41(s,1H),7.38-7.26(m,3H),7.23(d,J=7.1Hz,2H),5.33(s,2H);LCMS(ESI)m/z:312.0[M+H]+

compound 50: 1H NMR (500MHz, DMSO-d6) Δ 11.08(s, 1H), 8.12(s, 1H), 7.99-7.97(m, 2H), 7.66(s, 1H), 7.60-7.56(m, 3H), 7.47(s, 1H), 4.14-4.07(m, 2H), 3.78-3.74(m, 1H), 3.66-3.61(m,2H),3.47-3.44(m,1H),2.74-2.67(m,1H),1.94-1.87(m,1H),1.63-1.58(m,1H);LCMS(ESI)m/z:339.1[M+H]+

Compound 51: 1H NMR (500MHz, DMSO-d6) δ 11.01(s, 1H), 8.14(s, 1H), 7.98-7.96(m, 2H), 7.71(s, 1H), 7.60-7.53(m, 3H), 7.45(s, 1H), 7.36-7.26(m, 5H), 5.64(q, J ═ 7.1Hz, 1H), 1.81(d, J ═ 7.1Hz, 3H); LCMS (ESI) M/z 359.2[ M + H ]]+

Compound 52: LCMS (ESI) M/z 346.1[ M + H ]]+

Compound 53: 1H NMR (500MHz, DMSO-d6) δ 11.05(s, 1H), 8.23(s, 1H), 7.97(dd, J ═ 7.5, 2.0Hz, 2H), 7.71(s, 1H), 7.62-7.52(m, 3H), 7.46(s, 1H), 7.43-7.34(m, 1H), 7.20-7.09(m, 1H), 7.06(t, J ═ 9.0Hz, 2H), 5.37(s, 2H); LCMS (ESI) M/z 363.0[ M + H ]]+

Compound 54: 1H NMR (400MHz, DMSO-d6) Δ 11.10(s, 1H), 8.77(s, 1H), 8.23(s, 1H), 8.02-8.10(m, 2H), 7.72(s, 1H), 7.56-7.58(m, 1H), 7.51(s, 1H), 7.38-7.41(m, 1H), 7.05-7.14(m, 3H), 5.37(s, 2H); LCMS (ESI) M/z 364.1[ M + H ] ]+

Compound 55: 1H NMR (500MHz, DMSO-d6) Δ 11.15(s, 1H), 9.21(s, 1H), 8.75-8.74(m, 1H), 8.41-8.39(m, 1H), 8.23(s, 1H), 7.71(s, 1H), 7.66-7.63(m, 2H), 7.42-7.38(m, 1H), 7.16-7.04(m, 3H), 5.37(s, 2H); LCMS (ESI) M/z 364.1[ M + H ]]+

Compound 57: 1H NMR (400MHz, DMSO-d6) Δ 11.09(s, 1H), 8.77(s, 1H), 8.25(s, 1H), 8.04-8.10(m, 2H), 7.72(s, 1H), 7.56-7.57(m, 1H), 7.50(s, 1H), 7.38-7.40(m, 2H), 7.31(s, 1H), 7.21-7.22(m, 1H), 5.37(s, 2H); LCMS (ESI) M/z 364.1[ M + H ]]+

Compound 59:1H NMR(400MHz,DMSO-d6)δ11.25(s,1H),8.59(s,1H),8.18(s,1H),7.96(d,J=1.3Hz,1H),7.66(d,J=8.3Hz,1H),7.61(d,J=1.8Hz,1H),7.29(dd,J=2.0,8.3Hz,1H),7.22-7.18(m,2H),6.74(dd,J=1.7,3.4Hz,1H),5.43(s,2H);LCMS(ESI)m/z:403.0[M+H]+

compound 61: 1H NMR (500MHz, DMSO-d6) δ 11.08(s, 1H), 8.77(d, J ═ 4.5Hz, 1H), 8.24(s, 1H), 8.16-7.95(m, 2H), 7.70(s, 1H), 7.57(ddd, J ═ 7.5, 4.8, 1.1Hz, 1H), 7.49(s, 1H), 7.43(dd, J ═ 8.5, 2.3Hz, 1H), 7.34(d, J ═ 1.3Hz, 1H), 7.17-6.99(m, 1H), 5.34(s, 2H); LCMS (ESI) M/z 382.1[ M + H ]]+

Compound 63: 1H NMR (500MHz, DMSO-d6) δ 11.08(s, 1H), 9.00(d, J ═ 1.5Hz, 1H), 8.34-8.18(m, 2H), 7.97(dd, J ═ 7.7, 1.7Hz, 2H), 7.74(s, 1H), 7.58(q, J ═ 5.3Hz, 3H), 7.47(s, 1H), 7.17(d, J ═ 8.2Hz, 1H), 5.58(s, 2H); LCMS (ESI) M/z 371.1[ M + H ] ]+

Compound 64: 1H NMR (500MHz, DMSO-d6) δ 11.01(s, 1H), 8.07(s, 1H), 7.98-7.95(m, 2H), 7.75(s, 1H), 7.60-7.55(m, 3H), 7.43(s, 1H), 7.33-7.23(m, 3H), 7.02(d, J ═ 7.6Hz, 2H), 1.92(s, 6H); LCMS (ESI) M/z 373.1[ M + H ]]+

Compound 65: 1H NMR (500MHz, CDCl3) Δ 8.96(s, 1H), 8.07(s, 1H), 7.78-7.77(m, 3H), 7.47-7.46(m, 3H), 7.31-7.26(m, 3H), 7.22-7.21(m, 2H), 7.04(s, 1H), 5.62(s, 1H), 5.54(s, 1H), 3.25(s, 1H), 2.99(s, 2H), 2.91(s, 6H), 2.60(s, 1H); LCMS (ESI) M/z 416.1[ M + H ]]+

Compound 66: 1H NMR (500MHz, DMSO-d6) Δ 10.21(s, 1H), 8.05(s, 1H), 7.97-7.95(m, 2H), 7.58-7.55(m, 3H), 7.44(s, 1H), 7.37-7.26(m, 5H), 5.24(s, 2H), 2.16(s, 3H); LCMS (ESI) M/z 359.2[ M + H ]]+

Compound 68: 1H NMR (500MHz, DMSO-d6) δ 11.19(s, 1H), 8.40(s, 1H), 7.98(dd, J ═ 7.6, 1.9Hz, 2H), 7.63-7.55(m, 3H), 7.52(s, 1H), 7.42-7.32(m, 5H), 5.47(s, 2H); LCMS (ESI) M/z 370.1[ M + H ]]+

Compound 70: 1H NMR (500MHz, DMSO-d6) delta.11.02 (s, 1H), 8.19(s, 1H), 7.97-7.97(m, 2H), 7.73(s, 1H), 7.59-7.55(m, 3H), 7.44(s, 1H), 7.38-7.33(m, 4H), 7.30-7.27(m, 1H), 5.59-5.57(m, 1H), 2.67-2.65(m, 1H), 2.36-2.29(m, 3H) ,2.05(s,3H);LCMS(ESI)m/z:419.1[M+H]+

Compound 71: 1H NMR (500MHz, DMSO-d6) δ.11.03(s, 1H), 8.20(d, J ═ 1.5Hz, 1H), 7.97-7.95(m, 2H), 7.74(s, 1H), 7.59-7.55(m, 3H), 7.44(s, 1H), 7.39-7.34(m, 4H), 7.32-7.29(m, 1H), 5.63-5.60(m, 1H), 7.75-2.43(m, 7H); LCMS (ESI) M/z 435.1[ M + H ]]+

Compound 72: 1H NMR (400MHz, DMSO-d6) δ 11.16(s, 1H), 8.90(d, J ═ 2.0Hz, 1H), 8.23(s, 1H), 7.66(d, J ═ 34.9Hz, 2H), 7.38(dd, J ═ 12.2, 4.1Hz, 2H), 7.22-6.86(m, 3H), 5.37(s, 2H); LCMS (ESI) M/z 354.1[ M + H ]]+

Compound 73: 1H NMR (500MHz, CDCl3) δ 8.55(s, 1H), 8.13(s, 1H), 7.82(dd, J ═ 7.4, 2.2Hz, 2H), 7.68(s, 1H), 7.54-7.46(m, 3H), 7.36(d, J ═ 4.4Hz, 4H), 7.33(dd, J ═ 8.5, 4.3Hz, 1H), 7.01(s, 1H), 5.41(dd, J ═ 9.0, 5.9Hz, 1H), 3.60(dd, J ═ 13.9, 9.0Hz, 1H), 3.24(dd, J ═ 13.9, 5.9Hz, 1H), 2.03(s, 3H); LCMS (ESI) M/z 405.0[ M + H ]]+

Compound 74: LCMS (ESI) M/z 431.0[ M + H ]]+

Compound 75: 1H NMR (500MHz, DMSO-d6) δ 11.06(s, 1H), 8.20(s, 1H), 7.97(dd, J ═ 2.0Hz, 2H), 7.77(s, 1H), 7.59-7.55(m, 3H), 7.45(s, 1H), 7.38-7.37(m, 4H), 7.34-7.31(m, 1H), 5.64-5.61(m, 1H), 3.13-3.07(m, 1H), 3.02(s, 3H), 2.91-2.78(m, 2H), 2.55-2.52(m, 1H); LCMS (ESI) M/z 451.1[ M + H ] +.

Compound 76: 1H NMR (400MHz, DMSO-d6) delta 8.45(s, 1H), 7.10-7.99(m, 9H), 6.43-6.73(m, 1H), 5.47-5.60(m, 2H), 3.34-3.50(m, 3H); LCMS (ESI) M/z 445.0[ M + H ] +.

Compound 77: 1H NMR (400MHz, DMSO-d6) Δ 11.17(s, 1H), 8.78(s, 1H), 8.29(s, 1H), 8.03-8.09(m, 2H), 7.79-7.89(m, 2H), 7.38-7.56(m, 3H), 6.65(s, 1H), 5.58(s, 2H); LCMS (ESI) M/z 432.1[ M + H ] +.

Compound 78: 1H NMR (500MHz, DMSO-d6) δ 11.02(d, J ═ 9.1Hz, 1H), 8.29(d, J ═ 25.1Hz, 1H), 8.01-7.93(m, 2H), 7.76(d, J ═ 11.6Hz, 1H), 7.56(m, 3H), 7.50-7.30(m, 6H), 6.01-5.84(m, 1H), 4.13-3.77(m, H), 3.54(m, 1H), 2.61(d, J ═ 21.4Hz, 3H); LCMS (ESI) M/z 421.1[ M + H ] +.

Compound 79: 1H NMR (500MHz, CDCl3) δ 9.13(s, 1H), 7.82(dd, J ═ 6.5, 3.0Hz, 2H), 7.57-7.44(m, 3H), 7.42-7.28(m, 5H), 7.09(s, 1H), 4.24(s, 2H); LCMS (ESI) M/z 347.1[ M + H ] +.

Compound 80: 1H NMR (500MHz, DMSO-d6) δ 11.03(s, 1H), 8.35(s, 1H), 7.96(dd, J ═ 7.7, 1.8Hz, 2H), 7.76(s, 1H), 7.60-7.54(m, 3H), 7.47-7.43(m, 3H), 7.34(dt, J ═ 24.8, 7.1Hz, 3H), 6.10(dd, J ═ 9.8, 3.8Hz, 1H), 4.53(dd, J ═ 14.8, 9.9Hz, 1H), 3.94(dd, J ═ 14.9, 3.7Hz, 1H), 2.73(s, 3H); LCMS (ESI) M/z 437.0[ M + H ] +.

Compound 82: 1H NMR (500MHz, DMSO-d6) Δ 10.33(s, 1H), 8.32(s, 1H), 8.02-7.90(m, 2H), 7.61-7.53(m, 3H), 7.47(s, 1H), 7.43-7.30(m, 5H), 5.44(s, 2H); LCMS (ESI) M/z 413.0[ M + H ] +.

Compound 84: 1H NMR (400MHz, DMSO-d6) Δ 11.05(s, 1H), 8.27(s, 1H), 7.86-7.89(m, 1H), 7.76(s, 1H), 7.37-7.41(m, 1H), 7.03(s, 1H), 6.62-6.64(m, 1H), 5.87(s, 1H), 5.57(s, 2H), 5.48(s, 1H), 2.12(s, 3H); LCMS (ESI) M/z 395.0[ M + H ] +.

Compound 87: 1H NMR (400MHz, DMSO-d6) δ 11.08(s, 1H), 9.04(d, J ═ 4.6Hz, 1H), 8.22(s, 1H), 7.69(s, 1H), 7.47-7.30(m, 2H), 7.19-6.93(m, 3H), 5.37(s, 2H), 2.81(d, J ═ 4.6Hz, 3H); LCMS (ESI) M/z 344.1[ M + H ] +

Compound 88: 1H NMR (500MHz, DMSO-d6) Δ 11.09(s, 1H), 8.32(s, 1H), 8.03-7.97(m, 3H), 7.75(s, 1H), 7.60-7.54(m, 3H), 7.47(s, 1H), 7.45-7.41(m, 1H), 7.11-7.08(m, 1H), 5.57(s, 2H); LCMS (ESI) M/z 388.1[ M + H ] +.

Compound 89: 1H NMR (500MHz, DMSO-d6) δ 10.99(s, 1H), 8.22-8.19(m, 2H), 8.02(dd, J ═ 6.6, 1.9Hz, 1H), 7.69(s, 1H), 7.47(s, 1H), 7.42-7.37(m, 1H), 7.17-6.97(m, 3H), 6.48(t, J ═ 6.9Hz, 1H), 5.36(s, 2H), 3.59(s, 3H); LCMS (ESI) M/z 394.1[ M + H ] +.

Compound 92: 1H NMR (500MHz, DMSO-d6) δ 11.14(s, 1H), 8.80(s, 1H), 8.20(s, 1H), 8.04-7.95(m, 2H), 7.75(d, J ═ 14.5Hz, 2H), 7.51(dd, J ═ 7.6, 1.6Hz, 1H), 7.36(pd, J ═ 7.4, 1.7Hz, 2H), 7.28(dd, J ═ 8.3, 7.2Hz, 1H), 7.03(dd, J ═ 7.3, 2.0Hz, 1H), 6.07-5.96(m, 1H), 5.46(s, 2H), 5.11(dt, J ═ 14.5, 6.9Hz, 4H); LCMS (ESI) M/z 475.1[ M + H ] +.

Compound 93: 1H NMR (500MHz, DMSO-d6) δ 11.18(s, 1H), 8.46(s, 1H), 8.22(s, 1H), 8.08(dd, J ═ 8.0, 0.8Hz, 1H), 7.76(s, 1H), 7.67(dd, J ═ 7.2, 0.8Hz, 1H), 7.51(dd, J ═ 7.6, 1.6Hz, 1H), 7.41-7.28(m, 4H), 7.02(dd, J ═ 7.3, 1.9Hz, 1H), 5.47(s, 2H), 5.45-5.37(m, 1H), 5.05(t, J ═ 6.3Hz, 2H), 4.78(t, J ═ 7.0Hz, 2H); LCMS (ESI) M/z 475.1[ M + H ] +.

Compound 94: 1H NMR (500MHz, DMSO-d6) δ 11.13(s, 1H), 8.70(s, 1H), 8.21(s, 1H), 7.95(d, J ═ 8.5Hz, 1H), 7.87(d, J ═ 7.0Hz, 1H), 7.74(s, 1H), 7.67(s, 1H), 7.61-7.59(m, 1H), 7.52-7.50(m, 1H), 7.38-7.32(m, 2H), 7.02-7.00(m, 1H), 6.20-6.17(m, 1H), 5.46(s, 2H), 5.09-5.03(m, 4H); LCMS (ESI) M/< 475.0[ M + H ] +.

Compound 95: 1H NMR (500MHz, DMSO-d6) δ 11.10(s, 1H), 8.80(s, 1H), 8.18(s, 1H), 8.00(t, J ═ 7.8Hz, 2H), 7.72(d, J ═ 2.0Hz, 2H), 7.36(t, J ═ 7.3Hz, 2H), 7.32-7.19(m, 4H), 6.05-5.97(m, 1H), 5.35(s, 2H), 5.11(dt, J ═ 14.6, 7.0Hz, 4H); LCMS (ESI) M/z 441.1[ M + H ] +.

Compound 96: 1H NMR (500MHz, DMSO-d6) δ 11.15(s, 1H), 8.46(s, 1H), 8.21(s, 1H), 8.08(dd, J ═ 8.0, 0.8Hz, 1H), 7.72(s, 1H), 7.67(dd, J ═ 7.1, 0.8Hz, 1H), 7.39-7.28(m, 5H), 7.26(d, J ═ 7.0Hz, 2H), 5.45-5.38(m, 1H), 5.36(s, 2H), 5.05(t, J ═ 6.3Hz, 2H), 4.78(t, J ═ 7.1Hz, 2H); LCMS (ESI) M/z 441.1[ M + H ] +.

Compound 97: 1H NMR (500MHz, DMSO-d6) δ 11.09(s, 1H), 8.69(s, 1H), 8.19(s, 1H), 7.94(d, J ═ 8.5Hz, 1H), 7.87(d, J ═ 7.5Hz, 1H), 7.70-7.59(m, 3H), 7.38-7.25(m, 5H), 6.21-6.15(m, 1H), 5.35(s, 2H), 5.09-5.03(m, 4H); LCMS (ESI) M/z 441.1[ M + H ] +.

Compound 98: 1H NMR (500MHz, DMSO-d6) δ 11.07(s, 1H), 8.30(s, 1H), 7.98(dd, J ═ 8.52.0 Hz, 2H), 7.84-7.82(m, 1H), 7.73(s, 1H), 7.60-7.55(m, 4H), 7.47-7.44(m, 2H), 5.44(s, 2H); LCMS (ESI) M/z 388.1[ M + H ] +.

Compound 99:1H NMR (400MHz, DMSO-d6) δ 11.21(s, 1H), 9.21(s, 1H), 8.56-8.58(d, J ═ 8Hz, 1H), 8.30-8.31(d, J ═ 4Hz, 2H), 7.87-7.90(m, 1H), 7.72-7.79(m, 2H), 7.39-7.42(m, 1H), 6.64-6.66(d, J ═ 8Hz, 1H), 5.58(s, 2H); LCMS (ESI) M/z 457.0[ M + H ] +.

Compound 100:1H NMR(500MHz,DMSO-d6)δ.10.93(s,1H),8.19(s,1H),7.67(s,1H),7.39(dd,J=14.0,3.5Hz,1H),7.14-7.11(m,1H),7.07-7.03(m,2H),6.78(s,1H),6.25(s,1H),5.35(s,2H),2.50-2.45(m,2H),2.35-2.29(m,2H),1.88-1.85(m,1H),1.74-1.70(m,1H;LCMS(ESI)m/z:357.1[M+H]+

compound 103: 1H NMR (500MHz, DMSO-d6) Δ 11.16(s, 1H), 8.23(s, 1H), 8.17-8.10(m, 2H), 7.95-7.93(m, 1H), 7.92-7.55(m, 4H), 7.43-7.38(m, 1H), 7.16-7.05(m, 3H), 5.38(s, 2H); LCMS (ESI) M/z 388.0[ M + H ] +.

Compound 107: 1H NMR (500MHz, DMSO-d6) δ.11.18(s, 1H), 8.66-8.65(m, 1H), 8.30(s, 1H), 8.04(td, J ═ 10.0, 1.0Hz, 1H), 7.88(dd, J ═ 8.5, 5.5Hz, 1H), 7.78(s, 1H), 7.73-7.70(m, 1H), 7.42(d, J ═ 2.5Hz, 1H), 7.38(dd, J ═ 8.5, 2.0Hz, 1H), 6.66(dd, J ═ 10.0, 2.0Hz, 1H), 5.58(s, 2H);

compound 109:1H NMR(300MHz,DMF-d7)δ11.02(s,1H),8.18(s,1H),8.00-7.93(m,2H),7.67(d,J=0.7Hz,1H),7.61-7.54(m,3H),7.45(s,1H),7.38-7.25(m,2H),7.23-7.13(m,2H),5.32(s,2H);LCMS(ESI)m/z:375.3[M+H]+

compound 111:1H NMR(300MHz,DMSO-d6)δ11.03(s,1H),8.17(s,1H),8.00-7.93(m,2H),7.68(d,J=0.7Hz,1H),7.57(dd,J=5.2,2.0Hz,3H),7.45(s,1H),7.43-7.32(m,1H),7.27-7.15(m,3H),5.40(s,2H);LCMS(ESI)m/z:363.3[M+H]+

compound 112:1H NMR(300MHz,DMF-d7)δ11.02(s,1H),8.18(s,1H),8.00-7.93(m,2H),7.67(d,J=0.7Hz,1H),7.61-7.54(m,3H),7.45(s,1H),7.38-7.25(m,2H),7.23-7.13(m,2H),5.32(s,2H);LCMS(ESI)m/z:363.2[M+H]+

compound 116: 1H NMR (500MHz, DMSO-d6) δ.10.96(s, 1H), 8.20(s, 1H), 7.67(s, 1H), 7.39(dd, J ═ 14.0, 7.5Hz, 1H), 7.13(td, J ═ 8.5, 2.0Hz, 1H), 7.07-7.04(m, 2H), 6.91(s, 1H), 5.35(s, 2H), 4.84(d, J ═ 6.0Hz, 2H), 4.55(d, J ═ 6.0Hz, 2H), 1.72(s, 3H); LCMS (ESI) M/z 357.2[ M + H ] +.

Compound 120:1H NMR(500MHz,DMSO-d6)δ.10.96(s,1H),8.14(s,1H),7.65(s,1H),7.36-7.23(m,5H),7.03(s,1H),5.32(s,2H),3.02(s,3H),2.45-2.35(m,4H),1.88-1.82(m,1H),1.70-1.64(m,1H);LCMS(ESI)m/z:353.2[M+H]+

compound 121:1H NMR(500MHz,DMSO-d6)δ.10.09(s,1H),8.14(s,1H),7.64(s,1H),7.36-7.23(m,5H),6.90(s,1H),5.32(s,2H),4.83(d,J=6.0Hz,2H),4.54(d,J=6.0Hz,2H),1.72(s,3H);LCMS(ESI)m/z:339.1[M+H]+

compound 122:1H NMR(500MHz,DMSO-d6)δ.11.09(s,1H),8.65(d,J=4.5Hz,1H),8.18(s,1H),8.03(td,J=10.0,1.0Hz,1H),7.72-7.69(m,1H),7.68(s,1H),7.39(d,J=2.5Hz,1H),7.36-7.24(m,5H),5.34(s,2H);LCMS(ESI)m/z:364.1[M+H]+

compound 123: 1H NMR (400MHz, DMSO-d6) Δ 11.16(s, 1H), 8.20(s, 1H), 7.96-7.97(m, 1H), 7.69-7.73(m, 2H), 7.55-7.56(m, 3H), 7.25-7.37(m, 5H), 5.35(s, 2H); LCMS (ESI) M/z 345.1[ M + H ] +.

Compound 126: 1H NMR (500MHz, DMSO-d6) δ.11.10(s, 1H), 8.76(d, J ═ 1.5Hz, 1H), 8.17(s, 1H), 8.09(d, J ═ 1.5Hz, 1H), 7.67(s, 1H), 7.61(s, 1H), 7.36-7.23(m, 5H), 5.33(s, 2H); LCMS (ESI) M/z 352.0[ M + H ] +.

Compound 128: 1H NMR (500MHz, DMSO-d6) δ 11.07(s, 1H), 8.17-8.16(m, 3H), 8.05(d, J ═ 8.5Hz, 2H), 7.68(d, J ═ 1.0Hz, 2H), 7.36-7.23(m, 5H), 5.33(s, 2H); LCMS (ESI) M/z 370.1[ M + H ] +.

Compound 129: 1H NMR (400MHz, DMSO-d6) Δ 11.15(s, 1H), 8.76-8.77(m, 1H), 8.20(s, 1H), 8.10-8.12(m, 1H), 7.99-8.03(m, 1H), 7.68-7.69(m, 2H), 7.56-7.59(m, 1H), 7.25-7.37(m, 5H), 5.35(s, 2H); LCMS (ESI) M/z 346.1[ M + H ] +.

Compound 130: 1H NMR (400MHz, DMSO-d6) Δ 11.16(s, 1H), 8.19(s, 1H), 7.98-7.99(m, 1H), 7.58-7.68(m, 3H), 7.46-7.48(m, 2H), 7.25-7.42(m, 5H), 5.35(s, 2H); LCMS (ESI) M/z 363.1[ M + H ] +.

Compound 131: 1H NMR (400MHz, DMSO-d6) Δ 11.22(s, 1H), 9.32(s, 1H), 8.67(s, 1H), 8.19(s, 1H), 7.80(s, 1H), 7.68(s, 1H), 7.24-7.37(m, 5H), 5.34(s, 2H); LCMS (ESI) M/z 352.0[ M + H ] +.

Compound 132: 1H NMR (300MHz, DMSO-d6) δ 11.05(s, 1H), 8.26(s, 1H), 8.06-7.90(m, 2H), 7.79(dt, J ═ 6.5, 2.1Hz, 1H), 7.71(s, 2H), 7.61-7.52(m, 5H), 7.46(s, 1H), 5.42(s, 2H); LCMS (ESI) M/z 370.3[ M + H ] +.

Compound 144: 1H NMR (300MHz, DMSO-d6) δ 11.05(s, 1H), 8.17(d, J ═ 0.7Hz, 1H), 7.92-7.84(m, 1H), 7.84-7.79(m, 1H), 7.68(d, J ═ 0.7Hz, 1H), 7.63(td, J ═ 8.0, 5.9Hz, 1H), 7.56(s, 1H), 7.45-7.21(m, 6H), 5.34(s, 2H); LCMS (ESI) M/z 363.2[ M + H ] +.

Compound 145: 1H NMR (400MHz, CDCl3) δ 8.23(dd, J ═ 2.2, 15.8Hz, 1H), 7.32-7.23(m, 1H), 7.19(s, 1H), 7.17-7.09(m, 1H), 7.06-6.97(m, 1H), 6.92-6.82(m, 1H), 6.75(s, 0.6H), 6.53(s, 0.3H), 6.05-5.97(m, 2H), 5.77(m, 0.4H), 4.25-4.07(m, 0.6H), 3.98-3.86(m, 1H), 3.83-3.74(m, 3H), 2.44-2.30(m, 1H), 2.24-2.13(m, 2H), 2.06-1.89(m, 1H); LCMS (ESI) M/z [ M + H ] +:394.1

Compound 148:1H NMR(500MHz,DMSO-d6)δ11.13(s,1H),9.39(d,J=1.0Hz,1H),9.08(d,J=5.0Hz,1H),8.18-8.16(m,2H),7.77(s,1H),7.69(s,1H),7.37-7.34(m,2H),7.31-7.28(m,1H),7.25(d,J=7.0Hz,2H),5.34(s,2H);LCMS(ESI)m/z:347.1[M+H]+

compound 153:1H NMR(500MHz,DMSO-d6)δ11.07(s,1H),8.16(s,1H),7.65(s,1H),7.50(s,1H),7.36-7.23(m,5H),5.88(s,1H),5.52(s,1H),5.33(s,2H),2.11(s,3H);LCMS(ESI)m/z:309.1[M+H]+

compound 167:1H NMR(300MHz,DMSO-d6)δ10.82(s,1H),7.91(s,1H),7.57(s,1H),6.63(d,J=0.9Hz,1H),3.77(s,2H),3.17(p,J=7.1Hz,1H),1.57-1.41(m,6H),1.30(dd,J=10.9,7.2Hz,13H);LCMS(ESI)m/z:343.3[M+H]+

compound 176:1H NMR(300MHz,DMSO-d6)δ11.35(s,1H),8.64-8.49(m,1H),7.64(dt,J=8.9,1.3Hz,1H),7.22(ddd,J=8.9,6.7,1.1Hz,1H),6.96(d,J=0.9Hz,1H),6.91-6.68(m,2H),3.26-3.06(m,1H),1.29(dd,J=6.9,1.3Hz,6H);LCMS(ESI)m/z:271.1[M+H]+

compound 202:1h NMR (300MHz, chloroform-d) δ 8.86-8.70(m, 3H), 8.56(s, 1H), 8.41(s, 1H), 8.16(s, 1H), 7.77-7.65(m, 3H), 7.59(d, J ═ 5.0Hz, 1H), 7.23(s, 1H), 5.57(s, 2H); LCMS (ESI) M/z 415.5[ M + H ]]+

Compound 208:1h NMR (400MHz, chloroform-d) δ 7.39-7.30(m, 3H), 7.26(s, 1H), 7.04(t, J ═ 7.3Hz, 1H), 6.93(d, J ═ 8.2Hz, 1H), 6.86-6.77(m, 3H), 6.07(s, 2H), 5.12-4.97(m, 2H), 4.70-4.58(m, 2H), 4.31(dd, J ═ 3.7, 11.2Hz, 1H); LCMS (ESI) M/z 365.1[ M + H ]]+

Compound 209:1h NMR (400MHz, chloroform)-d)δ7.85(dd,J=2.0,7.5Hz,2H),7.60-7.47(m,3H),7.32-7.28(m,2H),7.00(s,1H),6.85(t,J=7.3Hz,1H),6.54(d,J=7.8Hz,2H),4.83(s,2H),4.44(s,2H),4.19-4.04(m,4H);LCMS(ESI)m/z:346.1[M+H]+

Compound 210:1h NMR (400MHz, chloroform-d) δ 7.57(d, J ═ 0.9Hz, 1H), 7.20(m, 2H), 6.94(d, J ═ 3.5Hz, 1H), 6.87-6.77(m, 2H), 6.60-6.51(m, 3H), 5.01-4.90(m, 1H), 4.60(m, 1H), 4.45-4.33(m, 2H), 4.09(br s, 1H), 4.05-3.96(m, 1H); LCMS (ESI) M/z 310.1[ M + H ]]+

Compound 213:1H NMR(400MHz,CDCl3)δ7.81(dd,J=2.0,7.7Hz,2H),7.54-7.44(m,3H),7.32(t,J=8.1Hz,2H),7.03(t,J=7.5Hz,1H),6.97(s,1H),6.80(d,J=7.9Hz,2H),5.12-4.99(m,2H),4.70-4.58(m,2H),4.31(dd,J=3.7,11.6Hz,1H);LCMS(ESI)m/z:321.1[M+H]+

compound 214:1H NMR(400MHz,CDCl3)δ7.80(dd,J=2.1,7.4Hz,2H),7.57-7.46(m,3H),7.26-7.20(m,2H),6.97(s,1H),6.82(t,J=7.4Hz,1H),6.58(d,J=7.7Hz,2H),5.06-4.94(m,1H),4.62(dd,J=6.3,10.7Hz,1H),4.47-4.37(m,2H),4.12(br s,1H),4.03(dd,J=4.6,11.7Hz,1H);LCMS(ESI)m/z:320.2[M+H]+

compound 215:1H NMR(400MHz,DMSO-d6)δ7.95(dd,J=2.0,7.3Hz,2H),7.61-7.48(m,3H),7.38(s,1H),7.09(t,J=7.8Hz,2H),6.67(d,J=7.9Hz,2H),6.55(t,J=7.3Hz,1H),5.80(br t,J=6.2Hz,1H),4.41-4.24(m,2H),4.05(s,2H),3.99-3.85(m,2H),3.40(br d,J=6.2Hz,2H);LCMS(ESI)m/z:382.1[M+H]+

compound 218:1H NMR(400MHz,DMSO-d6)δ7.53(d,J=1.6Hz,0.7H),7.52-7.46(m,1.2H),7.46-7.40(m,0.8H),7.39-7.33(m,1H),7.31-7.24(m,0.3H),7.26-7.24(m,1H)7.24-7.19(m,1H),7.19-7.11(m,1H),7.09(d,J=8.4Hz,0.7H),7.02(d,J=8.0Hz,0.3H),6.13-6.10(m,2H),6.05(dd,J=5.2,9.2Hz,0.3H),5.66(dd,J=6.0,8.8Hz,0.7H),4.64-4.50(m,1.5H),4.30-4.14(m,0.5H),3.00-2.92(m,0.3H),2.90-2.78(m,0.7H),2.23-2.05(m,1H);LCMS(ESI)m/z:367.1[M+H]+

compound 219:1h NMR (400MHz, chloroform-d) δ 7.46-7.28(m, 4H), 7.26-7.16(m, 2H), 6.92(d, J ═ 8.0Hz, 0.7H), 6.87-6.87(m, 1H), 6.75(s, 0.2H), 6.28(dd, J ═ 4.8, 8.8Hz, 0.2H), 6.06(s, 1.4H), 6.01(s, 0.6H), 5.85(dd, J ═ 6.0, 8.8Hz, 0.7H), 4.67(t, J ═ 7.6Hz, 1.4H), 4.42-4.20(m, 0.6H), 3.11-2.90(m, 1H), 2.27-2.07(m, 1H); LCMS (ESI) M/z 383.0[ M + H ] ]+

Compound 221:1H NMR(400MHz,CDCl3)δ7.33(dd,J=1.5,8.2Hz,1H),7.26-7.18(m,3H),6.91(d,J=8.2Hz,1H),6.85-6.78(m,2H),6.57(d,J=7.7Hz,2H),6.05(s,2H),5.04-4.94(m,1H),4.61(dd,J=6.4,10.8Hz,1H),4.47-4.36(m,2H),4.02(dd,J=4.4,12.1Hz,2H);LCMS(ESI)m/z:364.1[M+H]+

compound 222:1H NMR(400MHz,DMSO-d6)δ7.52(d,J=1.8Hz,1H),7.48(dd,J=1.8,8.2Hz,1H),7.25(s,1H),7.17(t,J=7.9Hz,2H),7.08(d,J=8.2Hz,1H),6.69(t,J=7.4Hz,1H),6.44(d,J=7.7Hz,2H),6.13(s,2H),4.65(s,2H),4.29(s,2H),3.98(s,4H);LCMS(ESI)m/z:390.2[M+H]+

compound 224:1H NMR(400MHz,CDCl3)δ7.47-7.36(m,4H),7.34-7.28(m,2H),7.22(dd,J=1.5,7.3Hz,1H),6.90(dd,J=3.5,8.2Hz,1H),6.78(d,J=1.1Hz,1H),6.05(d,J=2.6Hz,2H),4.76(s,1H),4.48(s,1H),4.37(s,1H),4.09(s,1H),2.92-2.81(m,2H),2.75-2.61(m,2H),2.19(br s,1H);LCMS(ESI)m/z:405.1[M+H]+

compound 228:1H NMR(400MHz,MeOD)δ7.47-7.40(m,1H),7.34(br d,J=3.4Hz,1H),7.32-7.26(m,2H),7.25-7.20(m,2H),7.19-7.13(m,1H),6.96(dd,J=3.2,8.1Hz,1H),6.92(d,J=3.5Hz,1H),6.05(d,J=2.0Hz,2H),4.74(s,1H),4.50(s,1H),4.34(s,1H),4.11(s,1H),3.52-3.42(m,1H),2.67(br t,J=8.9Hz,2H),2.49-2.28(m,1H),2.49-2.28(m,1H);LCMS(ESI)m/z:389.0[M+H]+。

compound 230: LCMS (ESI) M/z 376.1[ M + H ] +.

Compound 237: LCMS (ESI) M/z 348.1[ M + H ] +.

Compound 238: LCMS (ESI) M/z 362.0[ M + H ] +.

Compound 240: LCMS (ESI) M/z 348.1[ M + H ] +.

Compound 244: 1H NMR (300MHz, DMSO-d6) δ 7.99(d, J ═ 1.8Hz, 1H), 7.26(d, J ═ 3.5Hz, 1H), 7.08(s, 1H), 6.76(dd, J ═ 3.5, 1.8Hz, 1H), 5.57(tt, J ═ 6.2, 3.2Hz, 1H), 5.38(tt, J ═ 6.1, 3.1Hz, 1H), 4.79(dd, J ═ 21.5, 11.5, 5.9, 1.9Hz, 1H), 4.61-4.34(m, 2H), 4.23-4.02(m, 1H); LCMS (ESI) M/z 237.2[ M + H ] +.

Compound 245: 1H NMR (300MHz, DMSO-d6) δ 8.03-7.95(m, 1H), 7.24(d, J ═ 3.5Hz, 1H), 7.04(s, 1H), 6.78-6.70(m, 1H), 4.28(s, 2H), 3.92(s, 2H), 1.87-1.69(m, 5H), 1.69-1.48(m, 5H); LCMS (ESI) M/z 273.2[ M + H ] +.

Compound 246:1H NMR(300MHz,DMSO-d6)δ7.98(d,J=1.8Hz,1H),7.24(d,J=3.5Hz,1H),7.02(d,J=0.9Hz,1H),6.81-6.66(m,1H),4.41(s,2H),4.04(s,2H),2.17(t,J=7.6Hz,5H),1.78(p,J=7.7Hz,2H);LCMS(ESI)m/z:259.2[M+H]+。

compound 247: 1H NMR (300MHz, DMSO-d6) δ 7.99(d, J ═ 1.8Hz, 1H), 7.25(d, J ═ 3.5Hz, 1H), 7.03(s, 1H), 6.76(dd, J ═ 3.6, 1.8Hz, 1H), 4.70(d, J ═ 1.1Hz, 4H), 4.62(s, 2H), 4.25(s, 2H); LCMS (ESI) M/z 261.2[ M + H ] +.

Compound 248:1h NMR (300MHz, DMSO-d6) δ 7.43-7.20(m, 5H), 5.66(s, 1H), 4.80(dd, J ═ 9.8, 8.2Hz, 1H), 4.45(t, J ═ 8.9Hz, 1H), 4.35(dd, J ═ 9.7, 6.1Hz, 1H), 4.07-3.87(m, 2H), 3.69(dd, J ═ 5.9, 3.8Hz, 4H), 3.29-3.35(4H, under water peak); LCMS (ESI) M/z 336.2[ M + Na ]]+。

Compound 249:1H NMR(300MHz,DMSO-d6)δ7.47-7.33(m,5H),7.32-7.23(m,1H),5.61(s,1H),4.79(dd,J=9.9,8.1Hz,1H),4.44(t,J=8.8Hz,1H),4.35(dd,J=9.4,6.2Hz,1H),4.07-3.88(m,2H),3.60-3.48(m,2H),3.40(ddd,J=11.5,7.8,3.3Hz,9H),3.16(ddd,J=12.9,8.9,3.5Hz,2H),1.97-1.78(m,2H),1.49(dtd,J=12.9,8.7,4.0Hz,2H);LCMS(ESI)m/z:364.3[M+Na]+。

compound 250: 1H NMR (300MHz, DMSO-d6) δ 7.42-7.32(m, 4H), 7.32-7.23(m, 1H), 6.56(d, J ═ 0.9Hz, 1H), 4.89-4.76(m, 1H), 4.53-4.33(m, 2H), 4.08-3.89(m, 2H), 3.22-3.06(m, 1H), 1.27(d, J ═ 6.9Hz, 6H); LCMS (ESI) M/z 271.2[ M + H ] +.

Compound 251:1H NMR(300MHz,DMSO-d6)δ7.37-7.10(m,5H),6.51(d,J=0.9Hz,1H),4.51-4.36(m,1H),4.16-4.01(m,2H),3.76(dd,J=10.1,4.8Hz,1H),3.12(pd,J=7.0,0.9Hz,1H),2.93(s,3H),1.25(d,J=6.9Hz,6H);LCMS(ESI)m/z:285.2[M+H]+。

compound 254:1H NMR(300MHz,DMSO-d6)δ8.81(d,J=2.3Hz,1H),8.24-8.01(m,2H),7.39(d,J=1.2Hz,1H),7.37-7.11(m,6H),4.51(t,J=8.4Hz,1H),4.25-4.06(m,2H),3.81(dd,J=9.9,4.9Hz,1H),2.95(d,J=6.2Hz,3H);LCMS(ESI)m/z:354.3[M+H]+。

compound 256:1H NMR(300MHz,DMSO-d6)δ8.25(d,J=9.3Hz,1H),7.56-7.39(m,2H),7.37-7.11(m,5H),4.53(t,J=8.6Hz,1H),4.12(s,5H),3.83(dd,J=9.8,5.0Hz,1H),2.95(d,J=6.1Hz,3H);LCMS(ESI)m/z:351.2[M+H]+。

compound 260:1H NMR(400MHz,DMSO-d6)δ7.48(s,1H),7.48-7.46(d,J=8.8Hz,1H),7.18-7.15(m,4H),7.12(s,1H),7.08-7.06(d,J=8.8Hz,1H),6.11(s,2H),4.80-4.79(m,2H),3.87-3.80(m,2H),2.89-2.83(m,2H);LCMS(ESI)m/z:349.1[M+H]+。

compound 261: h NMR (400MHz, CDCl3) δ 7.50-7.15(m, 2H), 7.115-7.06(m, 2H), 7.05-6.90(m, 2H), 6.82-6.80(d, J ═ 8.0Hz, 1H), 6.60-6.20(m, 1H), 5.96(s, 2H), 3.96-3.93(t, J ═ 6.4Hz, 2H), 2.81-2.78(t, J ═ 6.4Hz, 2H), 2.04-1.93(m, 2H); LCMS (ESI) M/z 349.1[ M + H ] +.

Compound 262: 1HNMR (400MHz, DMSO-d6) δ 8.04(brs, 1H), 7.50-7.46(m, 2H), 7.24(s, 1H), 7.15-7.04(m, 2H), 6.95(dd, J1 ═ 8.4Hz, J2 ═ 2.0Hz, 1H), 6.89(brs, 1H), 6.14(s, 2H), 4.40-4.25(m, 2H), 4.03(t, J ═ 5.2Hz, 2H); LCMS (ESI) M/z 351.1[ M + H ] +.

Compound 263:1H NMR(400MHz,DMSO-d6)δ7.49-7.40(m,2H),7.14(m,3H),7.08(d,J=7.9Hz,1H),7.02-6.95(m,1H),6.12(s,2H),3.87-3.80(m,2H),2.79(t,J=6.6Hz,2H),2.02-1.94(m,2H);LCMS(ESI)m/z:367.1[M+H]+。

compound 264:1H NMR(400MHz,DMSO-d6)δ7.41-7.39(m,2H),7.18-7.14(m,1H),7.09-7.07(m,1H),7.04-7.01(m,2H),6.98-6.93(m,1H),6.09(s,2H),3.90-3.65(m,2H),2.84-2.80(t,J=6.8Hz,2H),2.00-1.97(m,2H);LCMS(ESI)m/z:367.0[M+H]+。

compound 267:1H NMR(300MHz,DMSO-d6)δ7.55-7.43(m,2H),7.29-7.05(m,2H),6.97-6.88(m,2H),6.78(t,J=6.7Hz,1H),6.14(d,J=1.4Hz,2H),5.76(s,1H),4.41(d,J=12.7Hz,0H),4.25(d,J=12.7Hz,1H),4.18-3.87(m,2H),3.42(d,J=14.7Hz,1H),3.04(d,J=11.6Hz,1H),2.40(d,J=18.9Hz,1H),0.91(dd,J=10.9,6.4Hz,2H);LCMS(ESI)m/z:402.4[M+H]+。

compound 268:1H NMR(400MHz,CDCl3-d)δ7.82(br d,J=3.5Hz,2H),7.51(br s,3H),7.08(d,J=14.8Hz,1H),6.97-6.85(m,2H),5.29(s,1H),5.05(s,1H),4.44(br t,J=5.2Hz,1H),4.29-4.11(m,3H);LCMS(ESI)m/z:295.1[M+H]+。

compound 269:1H NMR(400MHz,CDCl3-d)δ7.34(br d,J=8.4Hz,1H),7.25(s,1H),7.07(d,J=13.9Hz,1H),6.95-6.88(m,2H),6.79(s,0.5H),6.71(s,0.5H),6.06(d,J=2.4Hz,2H),5.26(s,1H),5.04(s,1H),4.42(t,J=5.2Hz,1H),4.28-4.10(m,3H);LCMS(ESI)m/z:339.1[M+H]+。

compound 272: 1H NMR (400MHz, CDCl3) δ 8.41-8.29(m, 1H), 7.37-7.26(m, 2H), 7.22-7.15(m, 1H), 7.12-7.07(m, 1H), 6.90-6.79(m, 0.6H), 6.75-6.70(m, 0.6H), 6.61-6.53(m, 0.3H), 6.05-5.97(m, 2H), 5.84(br d, J7.3, 0.3H), 5.37(dd, J ═ 3.3, 8.4Hz, 0.5H), 4.27-4.10(m, 1H), 3.90(br dd, J ═ 6.7, 15.8Hz, 1H), 2.44-2.30(m, 1H), 2.88-1H (m, 1H); LCMS (ESI) M/z [ M + H ] +: 382.1.

Compound 273: 1H NMR (400MHz, CDCl3-d) δ 7.78(s, 1H), 7.72(d, J ═ 8.4Hz, 1H), 7.51-7.39(m, 4H), 7.36(br d, J ═ 6.6Hz, 2H), 6.65(s, 1H), 4.87-4.74(m, 2H), 4.67(t, J ═ 8.9Hz, 1H), 4.23(t, J ═ 8.0Hz, 1H), 4.07(s, 3H), 3.99(d, J ═ 15.4Hz, 1H), 2.60(s, 3H); lcms (esi) M/z [ M + H ] + ═ 389.1.

Compound 274:1H NMR(400MHz,DMSO-d6)δ7.54-7.44(m,2H),7.28-7.20(m,3H),7.13(d,J=8.4Hz,1H),6.98(d,J=8.2Hz,2H),6.83(t,J=7.3Hz,1H),3.87-3.77(m,10H),3.22(br d,J=19.8Hz,4H);LCMS(ESI)m/z:394.2[M+H]+。

compound 276:1H NMR(400MHz,CDCl3-d)δ8.75(br s,2H),7.33(br d,J=4.0Hz,3H),7.24(br s,1H),6.92(br d,J=7.7Hz,1H),6.73(s,1H),6.06(s,2H),4.16-3.73(m,6H),3.51(br s,2H);LCMS(ESI)m/z:407.0[M+H]+。

compound 277:1H NMR(400MHz,DMSO-d6)δ7.50(d,J=1.76Hz,1H)7.47(dd,J=8.16,1.76Hz,1H)7.18(s,1H)7.04-7.16(m,4H)6.97-7.03(m,1H)6.13(s,2H)3.81(dt,J=17.03,4.82Hz,4H)3.07(dt,J=18.41,5.02Hz,4H);LCMS(ESI)m/z:396.2[M+H]+。

compound 278:1H NMR(400MHz,CDCl3)δ7.84(s,1H),7.76-7.74(d,J=8.4Hz,1H),7.52-7.50(d,J=8.4Hz,1H),7.33-7.27(m,2H),6.99-6.94(m,4H),4.16-4.13(m,2H),4.05(s,3H),4..02-3.99(m,2H),3.33-3.27(m,4H),2.61(s,3H);LCMS(ESI)m/z:402.1[M+H]+。

compound 279:1H NMR(400MHz,CDCl3)δ7.37(dd,J=1.6,8.0Hz,1H),7.28(d,J=6.4Hz,1H),7.06-6.98(m,2H),6.97-6.89(m,3H),6.74(s,1H),6.08(s,2H),4.12(t,J=4.8Hz,2H),4.00(t,J=4.2Hz,2H),3.21(td,J=5.2,14.8Hz,4H);LCMS(ESI)m/z:396.2[M+H]+。

compound 281:1H NMR(400MHz,CDCl3)δ7.53-7.42(m,5H),7.36-7.30(m,1H),7.24(s,1H),6.91(d,J=8.0Hz,1H),6.72(s,1H),6.06(s,2H),4.04-3.51(m,8H);LCMS(ESI)m/z:406.2[M+H]+。

compound 282: 1HNMR (400MHz, CDCl3) δ 7.35(d, J ═ 8.0Hz, 1H), 7.29-7.19(m, 2H), 6.92(d, J ═ 8.0Hz, 1H), 6.76-6.67(m, 2H), 6.66-6.55(m, 2H), 6.06(s, 2H), 4.11(t, J ═ 5.2Hz, 2H), 3.97(t, J ═ 5.2Hz, 2H), 3.29(td, J ═ 5.2, 15.2Hz, 4H); LCMS (ESI) M/z 396.1[ M + H ] +.

Compound 283:1H NMR(400MHz,CDCl3)δ7.89-7.77(m,2H),7.56-7.46(m,3H),7.04-6.97(m,2H),6.96-6.90(m,2H),6.87(s,1H),4.20-4.07(m,2H),4.04-3.94(m,2H),3.20(td,J=5.1,13.3Hz,4H);LCMS(ESI)m/z:352.1[M+H]+。

compound 284:1H NMR(400MHz,CDCl3)δ7.86-7.79(m,2H),7.55-7.45(m,3H),7.12-7.04(m,2H),7.03-6.94(m,2H),6.87(s,1H),4.19-4.09(m,2H),4.06-3.97(m,2H),3.19(td,J=5.2,10.6Hz,4H);LCMS(ESI)m/z:352.1[M+H]+。

compound 285:1h NMR (400MHz, chloroform-d) δ 7.58(d, J0.8 Hz, 1H), 7.38-7.29(m, 4H), 7.29-7.25(m, 1H), 6.96(d, J3.2 Hz, 1H), 6.86(s, 1H), 6.71(br d, J7.6 Hz, 1H), 6.61-6.50(m, 1H), 4.06-3.93(m, 1H), 3.53(s, 2H), 2.86(br d, J12 Hz, 2H), 2.19(br t, J11.2 Hz, 2H), 2.05-1.97(m, 2H), 1.68-1.56(m, 2H); LCMS (ESI) M/z 352.1[ M + H ]]+。

Compound 286:1h NMR (400MHz, chloroform-d) δ 7.56(d, J0.8 Hz, 1H), 7.26-7.20(m, 2H), 6.97-6.91(m,3H),6.88-6.81(m,1H),6.73(br d,J=8.0Hz,1H),6.54(dd,J=2.0,2.9Hz,1H),4.21-4.03(m,1H),3.66(br d,J=12.8Hz,2H),3.00-2.82(m,2H),2.13(br d,J=10.0Hz,2H),1.71(dq,J=4.0,11.6Hz,2H);LCMS(ESI)m/z:338.1[M+H]+。

Compound 287:1h NMR (400MHz, chloroform-d) δ 7.59(d, J1.1 Hz, 1H), 6.97(d, J3.3 Hz, 1H), 6.87(s, 1H), 6.71(br d, J7.5 Hz, 1H), 6.57(dd, J1.8, 3.5Hz, 1H), 4.23-3.97(m, 4H), 2.92(br t, J11.8 Hz, 2H), 2.03(br d, J12.8 Hz, 3H), 1.48(s, 9H); LCMS (ESI) M/z 306.1[ M-55 ]]+。

Compound 293: 1H NMR (300MHz, chloroform-d) δ 8.57(s, 2H), 7.35(dd, J ═ 8.1, 1.7Hz, 1H), 7.31-7.16(m, 3H), 6.92(d, J ═ 8.2Hz, 1H), 6.71(s, 1H), 6.07(s, 2H), 4.94(d, J ═ 13.3Hz, 1H), 4.72(d, J ═ 13.3Hz, 1H), 3.36-3.20(m, 1H), 3.01-2.81(m, 2H), 2.01(s, 2H), 1.91-1.68(m, 2H); LCMS (ESI) M/z 378.1[ M + H ] +.

Compound 294:1H NMR(400MHz,CDCl3)δ7.86-7.79(m,2H),7.55-7.46(m,3H),7.27-7.18(m,1H),6.88(s,1H),6.71(dd,J=2.0,8.2Hz,1H),6.67-6.56(m,2H),4.24-4.07(m,2H),4.07-3.85(m,2H),3.31(td,J=5.2,14.2Hz,4H);LCMS(ESI)m/z:352.1[M+H]+。

compound 295:1H NMR(400MHz,CDCl3)δ7.85-7.76(m,2H),7.54-7.42(m,3H),6.97(s,1H),4.19-4.01(m,1H),3.98-3.87(m,1H),3.86-3.75(m,1H),3.74-3.59(m,1H),3.55-3.26(m,4H),2.10-1.83(m,4H),1.47(d,J=6.2Hz,9H);LCMS(ESI)m/z:342.1[M-55]+。

compound 296:1H NMR(400MHz,CDCl3)δ7.82(dd,J=2.2,7.5Hz,2H),7.56-7.46(m,5H),7.40(t,J=7.7Hz,2H),7.34-7.28(m,1H),6.84(s,1H),4.79-4.68(m,1H),4.48(br d,J=13.6Hz,1H),3.72(dt,J=2.6,13.2Hz,1H),3.47-3.32(m,1H),2.25-2.12(m,2H),1.98-1.84(m,2H),1.67(br s,1H);LCMS(ESI)m/z:347.1[M-H]-。

compound 300:1h NMR (400MHz, chloroform-d) δ 7.51(d, J ═ 7.3Hz, 2H), 7.40(t, J ═ 7.5Hz, 2H), 7.36-7.28(m, 2H), 7.25(d, J ═ 1.5Hz, 1H), 6.92(d, J ═ 8.2Hz, 1H), 6.69(s, 1H), 6.06(s, 2H), 4.72(td, J ═ 2.1, 13.1Hz, 1H), 4.46(td, J ═ 2.0, 13.6Hz, 1H), 3.71(dt, J ═ 2.5, 13.1Hz, 1H), 3.38(dt, J ═ 2.8, 13.0Hz, 1H), 2.24-2.10(m, 2H), 1.84(m, 97.84H), 1H); LCMS (ESI) M/z 391.0[ M-H ]]-。

Compound 303:1H NMR(400MHz,CDCl3)δ8.19(s,1H),7.90(br d,J=7.7Hz,1H),7.86(d,J=7.7Hz,1H),7.55(t,J=7.7Hz,1H),7.38-7.29(m,2H),7.27-7.20(m,3H),6.86(s,1H),6.52(br s,1H),4.91(br d,J=13.2Hz,1H),4.58(br d,J=13.7Hz,1H),3.34-3.21(m,1H),3.06(d,J=4.9Hz,3H),2.97-2.80(m,2H),2.08-1.92(m,2H),1.87-1.74(m,2H);LCMS(ESI)m/z:390.2[M+H]+。

compound 311: 1H NMR (500MHz, DMSO-d6) δ 11.61(s, 1H), 9.38(d, J ═ 2.0Hz, 1H), 8.00(dd, J ═ 7.5, 2.0Hz, 2H), 7.84(d, J ═ 2.0Hz, 1H), 7.66-7.53(m, 4H), 7.52-7.45(m, 2H), 7.35-7.21(m, 3H); LCMS (ESI) M/z 359.0[ M + H ] +.

Compound 312: 1H NMR (500MHz, DMSO-d6) δ 11.28(s, 1H), 8.11(d, J ═ 5.6Hz, 1H), 7.99-7.97(m, 2H), 7.63-7.53(m, 5H), 7.50(d, J ═ 1.5Hz, 1H), 7.45-7.42(m, 2H), 7.23(t, J ═ 7.4Hz, 1H), 7.18-7.12(m, 2H); LCMS (ESI) M/z 358.1[ M + H ] +.

Compound 317: 1H NMR (500MHz, DMSO-d6) δ.8.46(s, 1H), 8.25(d, J ═ 8.5Hz, 1H), 8.01(d, J ═ 7.5Hz, 1H), 7.78(t, J ═ 8.0Hz, 1H), 7.48(s, 1H), 7.33-7.27(m, 4H), 7.22-7.19(m, 1H), 4.65(d, J ═ 13.0Hz, 1H), 4.09(d, J ═ 14.0Hz, 1H), 3.32-3.28(m, 1H), 2.95-2.88(m, 2H), 1.92(d, J ═ 13.5Hz, 1H), 1.83(d, J ═ 12.5, 1H), 1.65-1.61(m, 2H); LCMS (ESI) M/z 358.2[ M + H ] +.

Compound 319: 1H NMR (500MHz, DMSO-d6) δ 8.61(t, J ═ 4.0Hz, 1H), 8.05-7.99(m, 4H), 7.48-7.43(m, 3H), 7.38-7.35(m, 2H), 7.30-7.27(m, 1H), 6.26-6.16(m, 1H), 4.35-4.34(m, 2H), 3.93-3.82(m, 2H), 2.82-2.81(m, 2H), 2.62(m, 2H); LCMS (ESI) M/z 388.1[ M + H ] +.

Compound 320: 1H NMR (500MHz, DMSO-d6) δ 7.91(dd, J ═ 7.5Hz, 1.5Hz, 1H), 7.56-7.52(m, 1H), 7.33-7.12(m, 7H), 7.07(s, 1H), 4.67-4.64(m, 1H), 4.11-4.08(m, 1H), 3.97(s, 3H), 3.34-3.25(m, 1H), 2.96-2.84(m, 2H), 1.92-1.81(m, 2H), 1.68-1.58(m, 2H); LCMS (ESI) M/z 363.1[ M + H ] +.

Compound 322: 1H NMR (500MHz, DMSO-d6) δ 8.82(s, 1H), 8.73(s, 1H), 8.33(s, 1H), 7.95(dd, J ═ 8.0, 1.5Hz, 2H), 7.89-7.80(m, 1H), 7.62-7.52(m, 3H), 7.32(s, 1H), 4.70(d, J ═ 13.0Hz, 1H), 4.19(d, J ═ 13.5Hz, 1H), 3.38-3.25(m, 1H), 3.14(t, J ═ 12.0Hz, 1H), 2.97(td, J ═ 13.0, 2.5Hz, 1H), 1.95 (J, J ═ 39.0, 12.5Hz, 2H), 1.75 (t, J ═ 12.0, 2H), 1.75(q ═ 0, 2H); LCMS (ESI) M/z 334.1[ M + H ] +.

Compound 323:1H NMR(400MHz,DMSO-d6)δ7.92-7.94(m,2H),7.56-7.57(m,3H),7.40-7.42(m,4H),7.32(s,2H),4.43-4.46(m,1H),3.90-3.93(m,1H),3.48-3.51(m,1H),3.14-3.20(m,1H),2.93(s,3H),1.86-2.15(m,4H);LCMS(ESI)m/z:385.2[M+Na]+。

compound 324:1H NMR(400MHz,DMSO-d6)δ7.11-7.13(m,1H),6.86(s,1H),6.68-6.71(m,1H),5.82(s,1H),5.45(s,1H),4.23-4.26(m,1H),3.79-3.82(m,1H),3.47-3.48(m,1H),3.24-3.25(m,1H),2.67-2.70(m,2H),2.09(s,3H),1.76-1.85(m,4H),1.66-1.70(m,2H);LCMS(ESI)m/z:357.1[M+H]+。

compound 327, compound (xxvi): 1H NMR(400MHz,DMSO-d6)δ7.45(br.s.,1H),7.39-7.19(m,4H),7.18-6.68(m,4H),6.16-6.01(m,2H),5.55-5.25(m,1H),4.09-3.72(m,2H),2.43-2.30(m,1H),2.07-1.70(m,3H);LCMS(ESI)m/z:363.2[M+H]+。

Compound 328: 1HNMR (400MHz, DMSO-d6) δ 7.54(d, J ═ 1.6Hz, 1H), 7.50(dd, J1 ═ 8.0Hz, J2 ═ 1.6Hz, 1H), 7.42-7.37(m, 2H), 7.33-7.30(m, 3H), 7.10(d, J ═ 8.2Hz, 1H), 8.14(s, 2H), 5.15(s, 2H), 4.90(s, 2H); LCMS (ESI) M/z 335.1[ M + H ] +.

Compound 329:1H NMR(400MHz,DMSO-d6)δ7.54-7.45(m,2H),7.36-7.31(m,4H),7.28-7.22(m,2H),7.09(dd,J=6.0,8.2Hz,1H),6.13(d,J=4.0Hz,2H),4.22(m,0.5H),4.07-3.96(m,1H),3.84-3.74(m,1H),3.69-3.60(m,0.5H),3.60-3.42(m,2H),2.37-2.25(m,1H),2.13-1.99(m,1H);LCMS(ESI)m/z:363.2[M+H]+。

compound 330:1H NMR(400MHz,DMSO-d6)δ7.94(ddd,J=2.0,7.6,9.3Hz,2H),7.61-7.49(m,3H),7.38-7.30(m,5H),7.28-7.21(m,1H),4.24(dd,J=7.5,10.5Hz,0.5H),4.11-3.98(m,1H),3.86-3.75(m,1H),3.69-3.54(m,1H),3.53-3.41(m,1.5H),2.38-2.25(m,1H),2.13-1.99(m,1H);LCMS(ESI)m/z:319.2[M+H]+。

compound 331: 1HNMR (400MHz, CDCl3) δ 7.57(d, J7.6 Hz, 0.5H), 7.51(d, J7.6 Hz, 0.5H), 7.44-7.20(m, 4H), 7.15-7.12(m, 1H), 7.10-7.02(m, 1H), 6.96-6.91(m, 0.3H), 6.90(d, J8.0 Hz, 0.4H), 6.82(d, J8.0 Hz, 0.4H), 6.72(s, 0.5H), 6.46(s, 0.5H), 6.03(s, 1H), 6.00(s, 1H), 5.94-5.92(m, 0.5H), 5.48-5.45(m, 0.5H), 7.26 (t, 2.2H), 2.3H, 2H, 3H, 2H; LCMS (ESI) M/z 413.0[ M + H ] +.

Compound 332:1H NMR(400MHz,DMSO-d6)δ9.08(d,J=6.8Hz,1H),7.99(d,J=1.1Hz,1H),7.25(d,J=3.5Hz,1H),7.09(s,1H),6.76(dd,J=1.8,3.5Hz,1H),4.49-4.34(m,1H),3.59-3.47(m,1H),3.45-3.35(m,1H),3.32-3.16(m,2H),2.17-2.02(m,1H),1.96-1.85(m,1H),1.40(s,9H);LCMS(ESI)m/z:292.1[M+H]+。

compound 333:1H NMR(400MHz,DMSO-d6)δ7.96(ddd,J=2.0,7.4,12.0Hz,1H),7.74-7.67(m,1H),7.62-7.52(m,2H),7.35(dd,J=4.2,7.3Hz,4H),7.31(d,J=3.1Hz,1H),7.28-7.22(m,1H),4.24(dd,J=7.7,10.6Hz,0.5H),4.11-3.99(m,1H),3.86-3.78(m,1H),3.72-3.63(m,0.5H),3.62-3.41(m,2H),2.38-2.27(m,1H),2.14-2.01(m,1H);LCMS(ESI)m/z:353.1[M+H]+。

compound 334:1H NMR(400MHz,DMSO-d6)δ7.99-7.88(m,2H),7.60-7.47(m,3H),7.44-7.30(m,3H),7.16(dt,J=6.8,8.8Hz,2H),4.23(dd,J=7.5,10.6Hz,0.5H),4.11-3.94(m,1H),3.87-3.72(m,1H),3.66-3.59(m,0.5H),3.59-3.48(m,1H),3.48-3.39(m,1H),2.37-2.21(m,1H),2.13-1.94(m,1H);LCMS(ESI)m/z:337.1[M+H]+。

compound 335:1H NMR(400MHz,CDCl3)δ7.81(dd,J=2.0,7.7Hz,1H),7.71-7.64(m,1H),7.53-7.39(m,3H),7.27-7.27(m,2H),7.26-7.14(m,1H),7.12-6.98(m,1H),6.93(s,0.5H),6.70(s,0.5H),6.06(br d,J=7.0Hz,0.5H),5.59(dd,J=4.2,8.1Hz,0.5H),4.34-4.09(m,1H),4.05-3.82(m,1H),2.53-2.35(m,1H),2.14-1.89(m,3H);LCMS(ESI)m/z:337.1[M+H]+。

compound 336:1H NMR(400MHz,DMSO-d6)δ9.22(br d,J=6.8Hz,3H),8.00(d,J=1.1Hz,1H),7.26(d,J=3.3Hz,1H),7.16(s,1H),6.76(dd,J=1.8,3.5Hz,1H),4.65-4.50(m,1H),3.47-3.35(m,2H),3.27-3.15(m,2H),2.24-2.13(m,1H),2.07-1.93(m,1H);LCMS(ESI)m/z:248.0[M+H]+。

compound 337:1h NMR (400MHz, chloroform-d) δ 7.57(s, 1H), 7.33(d, J ═ 3.9Hz, 4H), 7.26(br s, 1H), 7.12(br d, J ═ 7.0Hz, 1H), 6.95(d, J ═ 3.1Hz, 1H), 6.84(s, 1H), 6.55(br d, J ═ 1.8Hz, 1H), 4.68-4.59(m, 1H), 3.69-3.59(m, 2H), 2.91-2.84(m, 1H), 2.68(d, J ═ 4.4Hz, 2H), 2.40-2.31(m, 2H), 1.81-1.72(m, 1H); LCMS (ESI) M/z 338.1[ M + H ] ]+。

Compound 338:1h NMR (400MHz, chloroform-d) δ 7.55(br d, J ═ 3.5Hz, 1H), 7.36-7.28(m, 2H), 7.24(br d, J ═ 6.1Hz, 3H), 6.92(dd, J ═ 3.1, 8.3Hz, 1H), 6.84(d, J ═ 5.7Hz, 1H), 6.53(br s, 1H), 4.41(dd, J ═ 7.9, 11.4Hz, 0.5H), 4.19(br dd, J ═ 7.7, 12.1Hz, 1H), 4.01-3.91(m, 1H), 3.82(t, J ═ 10.7Hz, 0.5H), 3.76-3.68(m,0.5H),3.67-3.61(m,0.5H),3.52-3.40(m,1H),2.37(br d,J=17.5Hz,1H),2.17-2.04(m,1H);LCMS(ESI)m/z:309.1[M+H]+。

compound 339:1H NMR(400MHz,CDCl3)δ7.82(dt,J=1.8,7.2Hz,2H),7.54-7.46(m,3H),7.37-7.29(m,1H),7.07(br t,J=6.7Hz,1H),7.03-6.91(m,3H),4.50(dd,J=7.5,11.5Hz,0.5H),4.33-4.18(m,1H),4.08-3.97(m,1H),3.88(br t,J=10.6Hz,0.5H),3.81-3.61(m,1H),3.56-3.42(m,1H),2.52-2.34(m,1H),2.23-2.03(m,1H);LCMS(ESI)m/z:337.1[M+H]+。

compound 340:1H NMR(400MHz,MeOD)δ=7.92-7.85(m,2H),7.59-7.48(m,3H),7.09(d,J=4.0Hz,1H),4.06(t,J=6.9Hz,1H),4.00(s,1H),3.79(t,J=7.2Hz,1H),3.72(s,1H),3.51-3.33(m,4H),2.18-2.07(m,4H);LCMS(ESI)m/z:298.1[M+H]+。

compound 341:1H NMR(400MHz,MeOD)δ=7.92-7.85(m,2H),7.59-7.48(m,3H),7.09(d,J=4.0Hz,1H),4.06(t,J=6.9Hz,1H),4.00(s,1H),3.79(t,J=7.2Hz,1H),3.72(s,1H),3.51-3.33(m,4H),2.18-2.07(m,4H);LCMS(ESI)m/z:374.2[M+H]+。

compound 342:1H NMR(400MHz,CDCl3)δ7.85-7.75(m,2H),7.52-7.43(m,3H),6.94(d,J=8.2Hz,1H),4.06-3.96(m,1H),3.95-3.81(m,1H),3.79-3.71(m,1H),3.70-3.58(m,1H),2.75-2.56(m,2H),2.56-2.46(m,2H),2.38-2.32(m,3H),2.13-1.76(m,4H);LCMS(ESI)m/z:312.2[M+H]+。

compound 343:1H NMR(400MHz,CDCl-d)δ7.59(s,1H),7.29(br s,1H),7.25(br s,1H),7.06-6.95(m,2H),6.88(s,1H),6.75(br t,J=7.3Hz,1H),6.65-6.52(m,3H),4.83(br s,1H),3.68(dd,J=6.1,9.8Hz,1H),3.59-3.49(m,1H),3.46-3.29(m,2H),2.48-2.36(m,1H),2.14(br d,J=5.1Hz,1H);LCMS(ESI)m/z:324.1[M+H]+。

compound 344:1h NMR (400MHz, chloroform-d) δ 7.80(dd, J ═ 2.1, 7.5Hz, 2H), 7.56-7.45(m, 3H), 6.95(s),1H),4.73(s,2H),4.34(s,2H),4.13(s,4H),1.46(s,9H);LCMS(ESI)m/z:370.1[M+H]+。

Compound 345:1h NMR (400MHz, chloroform-d) Δ 7.86-7.76(m, 2H), 7.56-7.42(m, 3H), 6.94(s, 1H), 4.71(s, 2H), 4.33(s, 2H), 3.85(s, 4H); LCMS (ESI) M/z 270.1[ M + H ]]+。

Compound 346:1H NMR(400MHz,MeOD)δ7.92(br t,J=6.6Hz,1H),7.86-7.69(m,3H),7.40-7.29(m,4H),7.28-7.20(m,1H),6.98(d,J=2.2Hz,1H),4.39(dd,J=7.6,11.4Hz,0.5H),4.25-4.10(m,1H),4.03-3.89(m,1H),3.83(t,J=10.6Hz,0.5H),3.76-3.66(m,0.5H),3.63-3.46(m,1.5H),2.47-2.33(m,1H),2.24-2.05(m,1H);LCMS(ESI)m/z:387.1[M+H]+。

compound 347:1h NMR (400MHz, chloroform-d) Δ 8.95-8.99(m, 1H)7.38-7.31(m, 4H), 7.30-7.26(m, 2H)4.17-4.16(m, 0.5H), 4.04-4.03(m, 0.5H), 3.96-3.77(m, 0.5H), 3.61-3.59(m, 1H), 3.58-3.47(m, 1H), 3.47-3.43(m, 1.5H), 2.80-2.78(m, 3H), 2.32-2.31(m, 1H), 2.07-1.99(m, 1H); LCMS (ESI) M/z 300.1[ M + H ]]+。

Compound 348: 1H NMR(400MHz,DMSO-d6)δ8.40(br d,J=9.7Hz,1H),8.04(br d,J=7.7Hz,1H),7.36-7.31(m,5H),7.25(td,J=4.4,8.7Hz,1H),4.18(dd,J=7.6,10.5Hz,0.5H),4.08-3.92(m,1H),3.85-3.71(m,1H),3.67-3.53(m,1H),3.51-3.41(m,1.5H),2.36-2.25(m,1H),2.13-1.99(m,1H);LCMS(ESI)m/z:286.0[M+H]+。

Compound 349:1H NMR(300MHz,DMSO-d6)δ7.37-7.13(m,5H),6.53(dd,J=1.7,0.9Hz,1H),4.15(dd,J=10.6,7.2Hz,1H),4.07-3.84(m,1H),3.84-3.65(m,1H),3.65-3.37(m,3H),3.20-3.01(m,1H),2.27(s,1H),2.04(t,J=10.7Hz,1H),1.27(dd,J=6.9,6.1Hz,6H);LCMS(ESI)m/z:285.2[M+H]+。

compound 350:1H NMR(300MHz,DMSO-d6)δ7.45-7.28(m,1H),7.18(q,J=7.4,6.9Hz,2H),7.08(ddd,J=9.1,7.6,2.5Hz,1H),6.53(t,J=1.2Hz,1H),4.01-3.89(m,1H),3.82-3.66(m,1H),3.65-3.38(m,2H),3.13(dq,J=13.6,6.8Hz,1H),2.38-2.21(m,1H),2.12-1.96(m,1H),1.27(dd,J=6.9,5.4Hz,6H);LCMS(ESI)m/z:303.2[M+H]+。

compound 351:1H NMR(300MHz,DMSO-d6)δ8.75(tt,J=4.7,1.5Hz,1H),8.12-7.96(m,2H),7.58-7.48(m,1H),7.44-7.16(m,6H),4.22(dd,J=10.6,7.4Hz,1H),4.11-3.89(m,1H),3.81(td,J=10.1,6.7Hz,1H),3.73-3.39(m,3H),2.18-2.00(m,1H);LCMS(ESI)m/z:320.3[M+H]+。

compound 352:1H NMR(300MHz,DMSO-d6)δ8.75(tt,J=4.7,1.4Hz,1H),8.09-7.96(m,2H),7.55(dddd,J=6.8,5.0,3.5,1.9Hz,1H),7.46-7.29(m,2H),7.21(ddt,J=10.9,8.7,4.8Hz,2H),7.08(s,1H),4.23(dd,J=10.5,7.4Hz,1H),4.14-3.93(m,1H),3.80(td,J=10.2,6.8Hz,1H),3.72-3.39(m,3H),2.33(s,1H),2.10(s,1H);LCMS(ESI)m/z:338.3[M+H]+。

compound 353:1H NMR(400MHz,DMSO-d6)δ7.51-7.32(m,2H),7.17-6.94(m,2H),6.24-6.00(m,3H),5.80-5.24(m,1H),4.01-3.60(m,2H),2.31(br.s.,1H),2.21-2.10(m,3H),2.02(d,J=8.4Hz,3H);LCMS(ESI)m/z:368.2[M+H]+。

compound 354:1H NMR(400MHz,DMSO-d6)δ7.51-7.41(m,1H),7.38-7.19(m,4H),7.18-7.03(m,2H),7.02-6.68(m,1H),6.21-6.01(m,3H),5.58-5.19(m,1H),4.10-3.74(m,2H),2.46-2.29(m,1H),2.03-1.76(m,3H);LCMS(ESI)m/z:363.1[M+H]+。

compound 355:1H NMR(400MHz,DMSO-d6)δ7.50-7.25(m,2H),7.24-6.98(m,4H),6.97-6.67(m,2H),6.20-6.02(m,2H),5.55-5.07(m,1H),4.11-3.67(m,2H),2.45-2.16(m,3H),2.01-1.76(m,3H);LCMS(ESI)m/z:377.2[M+H]+。

compound 356:1H NMR(400MHz,DMSO-d6)δ7.50-7.25(m,2H),7.22-6.85(m,5H),6.83-6.64(m,1H),6.16-6.04(m,2H),5.50-5.16(m,1H),4.06-3.78(m,2H),3.77-3.59(m,3H),2.34(d,J=12.3Hz,1H),1.93(d,J=6.2Hz,3H);LCMS(ESI)m/z:393.2[M+H]+。

compound 357:1H NMR(400MHz,DMSO-d6)δ7.93(br.s.,1H),7.25-7.11(m,1H),7.02-6.82(m,1H),6.73(br.s.,1H),6.26-6.03(m,1H),5.75-5.32(m,1H),3.99-3.74(m,2H),2.33(br.s.,1H),2.24-2.11(m,3H),2.04(d,J=6.6Hz,3H);LCMS(ESI)m/z:314.1[M+H]+。

compound 358:1H NMR(400MHz,DMSO-d6)δ7.49-7.41(m,1H),7.37-7.19(m,5H),7.18-6.67(m,3H),6.18-5.99(m,2H),5.59-5.18(m,1H),4.05-3.71(m,2H),2.46-2.29(m,1H),2.06-1.73(m,3H);LCMS(ESI)m/z:363.2[M+H]+。

compound 359:1H NMR(400MHz,DMSO-d6)δ7.43(br.s.,1H),7.31-7.12(m,2H),7.09-6.94(m,2H),6.92-6.63(m,3H),6.08(d,J=7.9Hz,2H),5.45-5.19(m,1H),4.01-3.57(m,5H),2.31(br.s.,1H),2.01-1.75(m,3H);LCMS(ESI)m/z:393.2[M+H]+。

compound 360:1H NMR(400MHz,DMSO-d6)δ7.44(br.s.,1H),7.28(br.s.,1H),7.19(d,J=7.5Hz,1H),7.11-6.96(m,2H),6.94-6.63(m,3H),6.10(d,J=8.4Hz,2H),5.49-5.18(m,1H),4.03-3.59(m,5H),2.35(d,J=12.3Hz,1H),1.95(br.s.,3H);LCMS(ESI)m/z:393.2[M+H]+。

compound 361:1H NMR(400MHz,MeOD-d4)δ8.49(d,J=6.2Hz,1H),8.45-8.39(m,1H),7.42(s,1H),7.40-7.32(m,2H),7.24(d,J=5.7Hz,1H),7.19(d,J=1.8Hz,0.2H),6.96(d,J=7.9Hz,0.6H),6.91(s,0.9H),6.72(s,0.3H),6.09-5.95(m,2H),5.80-5.72(m,0.4H),5.33-5.25(m,0.6H),4.25-4.05(m,1H),4.00-3.84(m,1H),2.51(dd,J=7.9,12.3Hz,1H),2.12-1.83(m,3H);LCMS(ESI)m/z:364.1[M+H]+。

compound 362:1H NMR(400MHz,DMSO-d6)δ8.53(d,J=2.1Hz,1H),8.45(dd,J=1.6,4.7Hz,1H),8.36(dt,J=2.0,4.6Hz,1H),7.70-7.64(m,0.6H),7.55(d,J=7.9Hz,0.4H),7.52-7.44(m,1H),7.40-7.32(m,1H),7.31-7.25(m,0.4H),7.21(s,0.6H),7.09(d,J=8.2Hz,0.6H),7.03(d,J=8.2Hz,0.2H),6.96(s,0.2H),6.15-6.07(m,2H),5.60(s,1H),5.24(dd,J=4.4,7.9Hz,1H),4.05(s,1H),3.94(s,1H),2.40(dd,J=7.5,12.3Hz,1H),1.95(t,J=6.7Hz,2H);LCMS(ESI)m/z:364.2[M+H]+。

compound 363:1H NMR(400MHz,MeOD-d4)δ8.49(d,J=6.2Hz,1H),8.45-8.39(m,1H),7.42(s,1H),7.40-7.32(m,2H),7.24(d,J=5.7Hz,1H),7.19(d,J=1.8Hz,0.2H),6.96(d,J=7.9Hz,0.6H),6.91(s,0.9H),6.72(s,0.3H),6.09-5.95(m,2H),5.80-5.72(m,0.4H),5.33-5.25(m,0.6H),4.25-4.05(m,1H),4.00-3.84(m,1H),2.51(dd,J=7.9,12.3Hz,1H),2.12-1.83(m,3H);LCMS(ESI)m/z:364.2[M+H]+。

compound 364:1H NMR(400MHz,MeOD-d4)δ7.43(dd,J=1.6,8.0Hz,0.5H),7.34(d,J=1.5Hz,0.5H),7.26-7.19(m,1H),7.18-7.10(m,1H),7.06(d,J=7.5Hz,0.5H),7.01-6.79(m,4H),6.42(s,0.5H),6.03(d,J=14.8Hz,2H),5.77(dd,J=2.7,7.6Hz,0.5H),5.56(dd,J=3.5,8.1Hz,0.5H),4.23-4.14(m,0.5H),4.05-3.76(m,4.5H),2.43-2.28(m,1H),2.07-1.80(m,3H);LCMS(ESI)m/z:393.2[M+H]+。

compound 365:1h NMR (400MHz, methanol-d 4) δ 7.43(dd, J ═ 1.5, 8.2Hz, 0.6H), 7.34(d, J ═ 1.3Hz, 0.5H), 7.27-7.21(m, 1H), 7.18-7.11(m, 1H), 6.96(d, J ═ 7.9Hz, 0.6H), 6.91-6.87(m, 1H), 6.86-6.77(m, 1.7H), 6.71-6.64(m, 1H), 6.59(s, 0.5H), 6.44(s, 0.5H), 6.03(d, J ═ 13.2Hz, 2H), 5.57-5.52(m, 0.5H), 5.28(dd, J ═ 4.2, 7.68, 4.68, 3.78H), 3.49-3.3H (m, 1H), 3.3.9H, 1.5H), 3.49-6.6H, 1H, 6.6H, 6.6.6H, 6.6.6.6H, 6.6.6.6.6.2 Hz, 13 (m, 1H); LCMS (ESI) M/z 393.1[ M + H ]]+。

Compound 366:1H NMR(400MHz,DMSO-d6)δ7.52-7.46(m,1.3H),7.39-7.32(m,1H),7.31-7.26(m,0.7H),7.24-7.18(m,2H),7.18-7.12(m,1H),7.11-7.05(m,1.5H),7.04-6.98(m,0.8H),6.15-6.06(m,2H),5.78(d,J=6.2Hz,0.4H),5.37(dd,J=4.2,8.2Hz,0.7H),4.09-3.70(m,2H),2.43-2.32(m,1H),1.94(m,3H);LCMS(ESI)m/z:381.1[M+H]+。

compound 367:1H NMR(400MHz,MeOD-d4)δ7.43(d,J=8.4Hz,0.7H),7.37-7.31(m,1H),7.27(d,J=7.1Hz,1H),7.19-7.17(m,0.4H),7.10(d,J=7.5Hz,0.7H),6.91(s,3.6H),6.57(s,0.4H),6.04(d,J=12.8Hz,2H),5.66(d,J=7.5Hz,0.5H),5.30(dd,J=4.4,7.5Hz,0.6H),4.21-3.85(m,2H),2.51-2.41(m,1H),2.06-1.90(m,3H);LCMS(ESI)m/z:381.1[M+H]+。

compound 368:1H NMR(400MHz,MeOD-d4)δ7.43(dd,J=1.5,8.2Hz,0.6H),7.36-7.23(m,2H),7.17(s,0.4H),7.14-7.02(m,2H),7.00-6.88(m,2H),6.52(s,0.6H),6.04(d,J=12.3Hz,2H),5.61(d,J=4.9Hz,0.6H),5.29(dd,J=4.4,7.9Hz,0.7H),4.19-4.15(m,0.5H),4.08-4.01(m,0.7H),3.95-3.87(m,1H),2.45(d,J=7.1Hz,1H),2.05-1.88(m,3H);LCMS(ESI)m/z:381.0[M+H]+。

compound 369:1H NMR(400MHz,MeOD-d4)δ7.45-7.39(m,1H),7.36-7.31(m,1H),6.96(d,J=8.4Hz,1H),6.88(s,1H),6.05(s,2H),4.70-4.62(m,0.4H),4.38-4.29(m,0.7H),3.91-3.63(m,2H),2.18-1.67(m,4H),1.33(d,J=6.2Hz,2H),1.20(d,J=6.6Hz,1H);LCMS(ESI)m/z:301.1[M+H]+。

compound 370:1H NMR(400MHz,MeOD-d4)δ7.45-7.39(m,1H),7.36-7.31(m,1H),6.96(d,J=8.4Hz,1H),6.88(s,1H),6.05(s,2H),4.70-4.62(m,0.4H),4.38-4.29(m,0.7H),3.91-3.63(m,2H),2.18-1.67(m,4H),1.33-1.20(m,3H);LCMS(ESI)m/z:301.1[M+H]+。

compound 371:1H NMR(400MHz,MeOD-d4)δ7.41(dd,J=1.3,7.9Hz,1H),7.32(d,J=1.3Hz,1H),6.95(d,J=7.9Hz,1H),6.79(s,1H),6.05(s,2H),3.89(t,J=6.8Hz,2H),2.09-1.96(m,6H),0.63(d,J=1.8Hz,2H);LCMS(ESI)m/z:313.1[M+H]+。

compound 372:1H NMR(400MHz,MeOD-d4)δ7.44(dd,J=1.8,8.4Hz,1H),7.35(d,J=1.3Hz,1H),7.00-6.94(m,2H),6.05(s,2H),5.73-5.64(m,0.4H),5.10-4.99(m,0.8H),4.02-3.60(m,2H),2.39-2.00(m,4H);LCMS(ESI)m/z:355.1[M+H]+。

compound 373:1H NMR(400MHz,MeOD-d4)δ7.44(dd,J=1.5,8.2Hz,1H),7.35(d,J=1.3Hz,1H),7.01-6.91(m,2H),6.06(s,2H),5.73-5.64(m,0.4H),5.11-5.02(m,0.8H),3.96-3.63(m,2H),2.40-2.04(m,4H);LCMS(ESI)m/z:355.1[M+H]+。

compound 374:1H NMR(400MHz,DMSO-d6)δ7.54-7.45(m,1H),7.39-7.29(m,1H),7.24(br.s.,0.5H),7.20-7.09(m,1.5H),7.08-6.96(m,1.2H),6.94-6.86(m,1.4H),6.79(d,J=7.5Hz,0.7H),5.53(d,J=6.6Hz,0.4H),5.20(br.s.,0.5H),4.00(br.s.,0.7H),3.92-3.79(m,6H),3.73(br.s.,2.3H),3.65(br.s.,1H),2.41-2.25(m,1H),2.11-1.71(m,3H);LCMS(ESI)m/z:409.2[M+H]+。

compound 375:1H NMR(400MHz,DMSO-d6)δ7.47(d,J=11.7Hz,2H),7.35-7.27(m,0.7H),7.26-7.16(m,1.5H),7.14-6.96(m,1.7H),6.94-6.74(m,2.2H),5.45(d,J=4.7Hz,0.4H),5.23(br.s.,0.5H),4.04-3.88(m,1H),3.88-3.73(m,5H),3.66(br.s.,1H),2.42-2.28(m,1H),2.02-1.76(m,3H);LCMS(ESI)m/z:379.2[M+H]+。

compound 376:1H NMR(400MHz,DMSO-d6)δ7.85(d,J=8.2Hz,1H),7.68(d,J=7.8Hz,1H),7.19(d,J=7.8Hz,1H),7.15-6.96(m,3H),6.93-6.84(m,1H),6.83-6.74(m,1H),6.66(br.s.,1H),5.52-5.39(m,1H),5.22(br.s.,1H),4.04-3.87(m,1H),3.87-3.71(m,5H),3.66(br.s.,1H),2.42-2.29(m,1H),2.01-1.76(m,3H);LCMS(ESI)m/z:379.2[M+H]+。

compound 377: delta1H NMR(400MHz,DMSO-d6)7.91(d,J=5.5Hz,1H),7.75(br.s.,1H),7.52(d,J=20.0Hz,3H),7.26-7.14(m,2H),7.01(d,J=7.4Hz,1H),6.89(d,J=7.8Hz,1H),6.84-6.73(m,1H),5.44(br.s.,0.5H),5.23(br.s.,0.5H),4.05-3.85(m,2H),3.84-3.72(m,2H),3.65(br.s.,1H),2.35(d,J=16.4Hz,1H),1.95(d,J=5.9Hz,3H);LCMS(ESI)m/z:349.2[M+H]+。

Compound 378, i.e.:1H NMR(400MHz,DMSO-d6)δ8.09(t,J=9.0Hz,1H),7.99-7.79(m,1H),7.71-7.54(m,1H),7.40(br.s.,1H),7.17(d,J=7.9Hz,1H),7.08-6.95(m,1H),6.88(d,J=7.5Hz,1H),6.77(d,J=7.9Hz,1H),5.47(d,J=6.6Hz,0.4H),5.20(br.s.,0.6H),4.00(br.s.,1H),3.73(br.s.,3H),3.63(br.s.,1H),2.32(dd,J=8.6,18.7Hz,1H),1.99-1.71(m,3H);LCMS(ESI)m/z:385.1[M+H]+。

compound 379:1H NMR(400MHz,DMSO-d6)δ7.91(d,J=7.5Hz,0.5H),7.78(d,J=7.5Hz,0.5H),7.57-7.45(m,1H),7.29-6.98(m,4H),6.89(d,J=7.5Hz,1H),6.80(d,J=7.9Hz,1H),6.65(s,1H),5.44(d,J=6.6Hz,0.5H),5.19(br.s.,0.5H),4.11-3.54(m,8H),2.41-2.26(m,1H),1.99-1.71(m,3H);LCMS(ESI)m/z:397.2[M+H]+。

compound 380:1H NMR(400MHz,DMSO-d6)δ8.06(d,J=7.9Hz,1H),7.89(d,J=7.5Hz,1H),7.59-7.42(m,2H),7.28(s,1H),7.20(d,J=7.9Hz,1H),7.00(d,J=7.1Hz,1H),6.89(d,J=8.8Hz,1H),6.78(d,J=7.5Hz,1H),5.42(br.s.,0.5H),5.23(br.s.,0.5H),4.07-3.58(m,5H),2.33(br.s.,1H),1.95(d,J=5.7Hz,3H);LCMS(ESI)m/z:433.2[M+H]+。

compound 381:1H NMR(400MHz,DMSO-d6)δ8.15(d,J=7.5Hz,1H),8.04-7.81(m,3H),7.42(s,1H),7.20(d,J=7.9Hz,1H),7.00(br.s.,1H),6.90(d,J=7.5Hz,1H),6.77(d,J=7.1Hz,1H),5.42(br.s.,0.5H),5.24(br.s.,0.5H),4.07-3.56(m,5H),2.35(d,J=19.0Hz,1H),2.02-1.78(m,3H);LCMS(ESI)m/z:417.2[M+H]+。

compound 382:1H NMR(400MHz,DMSO-d6)δ7.82-7.67(m,1H),7.63-7.52(m,1H),7.39-7.25(m,1H),7.23-7.11(m,2H),6.99(d,J=7.1Hz,1H),6.89(d,J=7.5Hz,1H),6.78(d,J=9.3Hz,1H),5.43(br.s.,0.5H),5.22(br.s.,0.5H),4.02-3.59(m,8H),2.41-2.27(m,1H),1.95(br.s.,3H);LCMS(ESI)m/z:397.2[M+H]+。

compound 383:1H NMR(400MHz,DMSO-d6)δ7.68(d,J=7.5Hz,1H),7.50(br.s.,1H),7.42-7.12(m,3H),7.01(d,J=7.9Hz,1H),6.94-6.72(m,2H),5.46(br.s.,0.5H),5.23(br.s.,0.5H),4.05-3.57(m,8H),2.33(br.s.,1H),2.04-1.75(m,3H);LCMS(ESI)m/z:397.2[M+H]+。

compound 384: 1H NMR(400MHz,MeOD)δ7.24-7.18(m,1H),7.16-7.01(m,3H),6.94(d,J=7.5Hz,1H),6.41-6.33(s,0.6H),5.89(s,0.4H),5.45-5.41(m,0.5H),5.22-5.13(m,0.5H),4.06-3.97(m,0.7H),3.92-3.85(m,0.7H),3.78-3.72(m,0.8H),2.62(d,J=7.1Hz,1H),2.43-2.38(m,1H),2.37-2.26(m,1.3H),1.95-1.75(m,3.5H),1.02-0.94(m,0.6H),0.78-0.70(m.,0.6H),0.49(d,J=6.6Hz,1H),0.41-0.35(m,1H),0.16(d,J=5.3Hz,1H),0.01(d,J=4.9Hz,1H);LCMS(ESI)m/z:297.2[M+H]+。

Compound 385:1H NMR(400MHz,DMSO-d6)δ7.94(dd,J=1.8,7.7Hz,1H),7.77(dd,J=2.9,6.7Hz,1H),7.60-7.52(m,2H),7.49(dd,J=1.8,5.0Hz,1H),7.37-7.29(m,2H),7.28-7.20(m,2H),7.17-7.05(m,1H),6.98(s,1H),5.56(d,J=5.5Hz,0.4H),5.24(dd,J=4.0,7.9Hz,0.6H),4.09-3.99(m,1H),3.96-3.72(m,1H),2.45-2.30(m,1H),1.98-1.73(m,3H);LCMS(ESI)m/z:319.2[M+H]+。

compound 386:1H NMR(400MHz,MeOD-d4)δ7.88(dd,J=2.0,7.7Hz,1H),7.71-7.65(m,1H),7.57-7.43(m,3H),7.37-7.21(m,3H),7.16-7.06(m,1H),7.03(s,1H),6.58(s,1H),5.68-5.58(m,0.5H),5.33(dd,J=4.3,7.8Hz,0.5H),4.27-4.13(m,0.5H),4.06(td,J=7.2,11.4Hz,0.5H),3.98-3.82(m,1H),2.57-2.35(m,1H),2.16-1.85(m,3H);LCMS(ESI)m/z:319.2[M+H]+。

compound 387:1H NMR(400MHz,DMSO-d6)δ7.95(dd,J=1.8,7.5Hz,1H),7.80-7.75(m,1H),7.55(s,2H),7.53-7.47(m,1H),7.36-7.31(m,2H),7.29-7.19(m,2H),7.16-7.07(m,1H),7.00(s,1H),5.59-5.54(m,0.4H),5.25(dd,J=4.0,7.9Hz,0.6H),4.11-3.98(m,0.6H),3.96-3.81(m,1H),3.81-3.72(m,0.4H),2.44-2.29(m,1H),1.97-1.74(m,3H);LCMS(ESI)m/z:319.2[M+H]+。

compound 388:1H NMR(400MHz,DMSO-d6)δ7.56-7.19(m,6H),7.09(d,J=7.9Hz,1H),6.95(s,1H),6.17-6.06(m,2H),5.60-5.50(m,0.3H),5.25-5.15(m,0.7H),4.12-3.99(m,1H),3.97-3.70(m,1H),2.37(dd,J=7.7,12.1Hz,1H),1.91(d,J=6.6Hz,3H);LCMS(ESI)m/z:441.1[M+H]+。

compound 389:1H NMR(400MHz,DMSO-d6)δ7.56-7.40(m,3H),7.38(d,J=1.3Hz,1H),7.27-7.18(m,2H),7.12-6.91(m,2H),6.18-6.05(m,2H),5.55(s,0.4H),5.18(d,J=3.1Hz,0.6H),4.01(s,1H),3.90(d,J=11.0Hz,1H),2.44-2.28(m,1H),1.91(br.s.,3H);LCMS(ESI)m/z:441.1[M+H]+。

compound 390:1H NMR(400MHz,DMSO-d6)δ7.52(br.s.,2H),7.40-6.90(m,6H),6.22-6.02(m,2H),5.90-5.73(m,0.4H),5.40(d,J=4.0Hz,0.6H),4.15(d,J=4.0Hz,1H),3.95(d,J=7.5Hz,1H),2.44-2.33(m,1H),2.03-1.62(m,3H);LCMS(ESI)m/z:441.1[M+H]+。

compound 391:1H NMR(400MHz,MeOD-d4)δ9.15-9.02(m,1H),7.84-7.75(m,1.3H),7.70(d,J=3.3Hz,0.6H),7.40(dd,J=1.7,8.0Hz,0.7H),7.33-7.24(m,1H),7.18(d,J=1.8Hz,0.3H),6.94(d,J=8.2Hz,0.6H),6.90-6.86(m,1H),6.74(s,0.3H),6.05-5.99(m,2H),5.94(dd,J=2.5,8.0Hz,0.3H),5.47(dd,J=4.9,8.4Hz,0.6H),4.26-4.12(m,1.3H),4.03-3.86(m,0.7H),2.65-2.49(m,1H),2.22-1.93(m,3H);LCMS(ESI)m/z:365.2[M+H]+。

compound 392:1H NMR(400MHz,MeOD-d4)δ8.10(d,J=8.8Hz,1H),7.91(d,J=8.8Hz,1H),7.84-7.73(m,2H),7.22-7.16(m,1.5H),6.97(d,J=8.6Hz,1H),6.87(d,J=8.6Hz,1H),6.75-6.69(m,1.5H),5.46(m,0.5H),5.30-5.24(m,0.5H),4.21-4.12(m,0.5H),4.07-3.99(m,0.5H),3.88(t,J=7.1Hz,1H),3.75(s,1.5H),3.59(s,1.5H),2.48-2.35(m,1H),2.08-1.84(m,3H);LCMS(ESI)m/z:467.2[M+H]+。

compound 393:1H NMR(400MHz,CDCl3)δ7.60-7.51(m,1H),7.46-7.37(m,1H),7.24-7.18(m,1.4H),7.14(d,J=8.4Hz,0.4H),7.03(d,J=8.7Hz,1H),6.92-6.87(m,1.59H),6.79(d,J=8.8Hz,1H),6.53(s,0.5H),5.77-5.70(m,0.5H),5.36(dd,J=4.1,7.5Hz,0.5H),4.30-4.12(m,1H),3.94(br d,J=8.2Hz,1H),3.84-3.70(m,3H),2.49-2.32(m,1H),2.17-1.93(m,3H);LCMS(ESI)m/z:429.2[M+H]+。

compound 394:1H NMR(400MHz,DMSO-d6)δ8.20(s,0.4H),8.22-8.18(m,0.3H),8.05(s,1H),7.84(d,J=8.4Hz,1H),7.61(dd,J=1.3,8.4Hz,0.5H),7.45(dd,J=1.3,8.4Hz,0.5H),7.38(s,0.5H),7.16(d,J=8.6Hz,1H),7.03-6.96(m,1H),6.86(d,J=8.6Hz,1H),6.75(d,J=8.8Hz,1H),5.52-5.48(m,0.5H),5.21-5.16(m,0.5H),3.99(d,J=15.2Hz,3H),3.70(s,2H),3.81-3.70(s,3H),3.3(s,2H),2.45(m,1H),2.39-2.23(m,1H),1.89-1.80(m,3H);LCMS(ESI)m/z:417.3[M+H]+。

compound 395:1H NMR(400MHz,DMSO-d6)δ7.80-7.64(m,2H),7.50-7.42(m,3H),7.38-7.24(m,1H),7.12-6.97(m,1.6H),6.82(s,0.4H),6.12-6.07(m,2H),5.60(br d,J=6.2Hz,0.3H),5.33-5.24(m,0.7H),4.08-3.78(m,2H),2.43(br dd,J=7.4,12.5Hz,1H),2.01-1.77(m,3H);LCMS(ESI)m/z:388.2[M+H]+。

compound 396:1H NMR(400MHz,DMSO-d6)δ7.75-7.42(m,5H),7.30(br s,1H),7.15-6.98(m,1.5H),6.82(br s,0.5H),6.15-6.06(m,2H),5.57(br s,0.3H),5.27(br s,0.7H),4.13-3.77(m,2H),2.47-2.37(m,1H),2.03-1.79(m,3H);LCMS(ESI)m/z:388.1[M+H]+。

compound 397:1H NMR(400MHz,CDCl3-d)δ=7.63(br d,J=6.8Hz,1H),7.51-7.28(m,5H),7.24(s,1H),6.92(br d,J=7.9Hz,1H),6.76(d,J=1.5Hz,1H),6.06(d,J=1.5Hz,2H),5.84(br s,1H),4.63-4.33(m,2H),4.00(br d,J=7.5Hz,2H),3.74(br t,J=11.8Hz,1H),3.47(br s,1H),3.15(br s,0.6H),1.58(d,J=1.5Hz,0.4H);LCMS(ESI)m/z:379.2[M+H]+。

compound 398:1H NMR(400MHz,DMSO-d6)δ7.78(br d,J=7.7Hz,1H),7.72-7.54(m,1H),7.47-7.41(m,2H),7.38-7.26(m,1H),7.10-6.95(m,2H),6.83(s,1H),6.12-6.05(m,2H),5.78(br d,J=6.0Hz,0.3H),5.38(br t,J=6.7Hz,0.7H),4.14-3.79(m,2H),2.57-2.50(m,1H),2.07-1.77(m,3H);LCMS(ESI)m/z:388.2[M+H]+。

compound 399:1H NMR(400MHz,CDCl3)δ7.61(d,J=1.4Hz,0.5H),7.54(dd,J=1.6,8.2Hz,0.5H),7.48(d,J=1.4Hz,0.5H),7.40(dd,J=1.6,8.2Hz,0.5H),7.34-7.23(m,1H),7.13(d,J=8.7Hz,1H),6.95(d,J=8.5Hz,1H),6.79(d,J=8.7Hz,1H),6.75(s,1H),6.69(d,J=8.8Hz,1H),6.38(s,0.5H),5.69-5.63(m,0.5H),5.27(dd,J=4.0,7.7Hz,0.5H),4.09(s,1H),3.85(br d,J=7.3Hz,1H),3.74-3.59(m,3H),3.17(d,J=6.5Hz,6H),2.38-2.23(m,1H),2.08-1.83(m,3H);LCMS(ESI)m/z:433.2[M+H]+。

compound 400:1H NMR(400MHz,DMSO-d6)δ8.89(d,J=10.9Hz,1H),8.43(d,J=1.4Hz,0.5H),8.27(s,0.3H),8.09-8.03(m,0.5H),8.00-7.96(m,0.5H),7.95-7.87(m,1H),7.43(s,0.5H),7.20(d,J=8.7Hz,1H),7.07(s,0.3H),7.02(d,J=8.7Hz,0.7H),6.90(d,J=8.8Hz,1H),6.79(d,J=8.8Hz,1H),5.51(dd,J=2.4,7.2Hz,0.4H),5.22(dd,J=4.0,7.8Hz,0.5H),4.09-4.01(m,0.5H),3.96-3.88(m,0.5H),3.86-3.79(m,0.5H),3.76-3.61(m,3H),2.33(br dd,J=7.8,12.3Hz,1H),1.98-1.76(m,3H);LCMS(ESI)m/z:390.2[M+H]+。

compound 401:1H NMR(400MHz,CDCl3)δ8.11(d,J=11.3Hz,1H),7.98(s,0.5H),7.83(m,1H),7.78-7.72(m,1H),7.60(m,0.5H),7.13(br d,J=8.4Hz,1H),6.98-6.89(m,1.4H),6.80(br d,J=8.5Hz,1H),6.69(br d,J=8.5Hz,1H),6.54(s,0.4H),5.66(m,0.5H),5.28(m,0.5H),4.22-4.07(m,1H),3.93-3.82(m,1H),3.76-3.58(m,3H),2.42-2.22(m,1H),2.09-1.83(m,3H);LCMS(ESI)m/z:390.0[M+H]+。

compound 402:1H NMR(400MHz,DMSO-d6)δ8.16-7.80(m,3H),7.79-7.64(m,1H),7.43-7.07(m,5H),6.99(s,1H),5.50(br d,J=6.0Hz,0.4H),5.29(br dd,J=4.4,7.5Hz,0.6H),4.13-3.80(m,2H),2.46-2.34(m,1H),2.03-1.81(m,3H);LCMS(ESI)m/z:344.2[M+H]+。

compound 403:1H NMR(400MHz,DMSO-d6)δ8.39(s,1H),8.24(br d,J=8.2Hz,1H),8.06(br d,J=7.5Hz,1H),8.00-7.88(m,1H),7.82-7.66(m,1H),7.42(s,1H),7.38-7.20(m,3H),7.18-7.02(m,1H),5.53(br d,J=7.1Hz,0.4H),5.28(br dd,J=3.7,7.7Hz,0.6H),4.07-3.88(m,1H),3.84(br s,1H),2.47-2.34(m,1H),2.02-1.79(m,3H);LCMS(ESI)m/z:344.2[M+H]+。

compound 404:1H NMR(400MHz,DMSO-d6)δ8.11(br d,J=7.9Hz,1H),8.04-7.87(m,3H),7.45(s,1H),7.36-7.19(m,3H),7.16-7.01(m,2H),5.50(br d,J=7.5Hz,0.4H),5.27(br s,0.6H),4.06-3.77(m,2H),2.46-2.34(m,1H),2.04-1.80(m,3H)。LCMS(ESI)m/z:344.2[M+H]+。

compound 405:1H NMR(400MHz,DMSO-d6)δ=9.13(br s,0.5H),8.95(br s,0.5H),8.76-8.63(m,1H),8.29(br d,J=7.5Hz,1H),8.12(br d,J=7.1Hz,1H),7.63-7.46(m,1H),7.41-7.20(m,4H),7.16-6.98(m,1H),5.52(br d,J=6.0Hz,0.4H),5.28(br s,0.6H),4.08-3.90(m,1H),3.84(br s,1H),2.47-2.33(m,1H),2.01-1.77(m,3H)。LCMS(ESI)m/z:320.2[M+H]+。

compound 406:1H NMR(400MHz,CDCl3)δ7.45-7.37(m,3H),7.33(br d,J=1.6Hz,2H),7.27-7.21(m,2H),7.16-7.11(m,0.3H),6.96-6.85(m,1H),6.81(s,0.6H),6.57(s,0.3H),6.10-6.00(m,2H),5.88-5.83(m,0.3H),5.65(d,J=4.4Hz,1.3H),5.58-5.53(m,0.3H),5.41-5.34(m,1H),4.42(dd,J=6.7,8.8Hz,1H),4.16-4.11(m,0.3H),4.06(m,0.7H);LCMS(ESI)m/z:365.1[M+H]+。

compound 407: 1HNMR (400MHz, DMSO-d6) δ 7.50(t, J ═ 1.6Hz, 1H), 7.46(td, J1 ═ 8.0Hz, J2 ═ 2.0Hz, 1H), 7.17-7.16(m, 1H), 7.09-7.07(m, 1H), 6.13(s, 2H), 4.04(t, J ═ 8.0Hz, 0.35H), 3.90-3.86(m, 0.65H), 3.78-3.72(m, 0.65H), 3.68-3.56(m, 1.35H), 2.05-1.97(m, 1.4H), 1.93-1.79(m, 2H), 1.74-1.68(m, 0.7H), 1.08-1.02(m, 0.65H), 0.94 (m, 0.65H), 0.90-0.35H), 0.58-0.35H, 0.65 (m), 0.58-0.35H, 0.65 (m, 0.65H); LCMS (ESI) M/z 327.2[ M + H ] +.

Compound 408:1H NMR(400MHz,DMSO-d6)δ=8.79-8.63(m,1H),8.07-7.83(m,2H),7.59-7.43(m,1H),7.38-7.19(m,4H),7.18-7.05(m,1H),6.89(s,1H),5.52(br d,J=6.4Hz,0.4H),5.28(br dd,J=4.1,7.2Hz,0.6H),4.09-3.78(m,2H),2.47-2.33(m,1H),2.04-1.78(m,3H)。LCMS(ESI)m/z:320.2[M+H]+。

compound 409:1H NMR(400MHz,DMSO-d6)δ8.81-8.67(m,2H),7.86(br d,J=5.5Hz,1H),7.69(br d,J=4.9Hz,1H),7.51(s,1H),7.39-7.19(m,4H),7.16-7.05(m,1H),5.50(br d,J=7.5Hz,0.4H),5.27(br dd,J=3.7,7.5Hz,0.6H),4.08-3.76(m,2H),2.47-2.36(m,1H),2.01-1.77(m,3H)。LCMS(ESI)m/z:320.2[M+H]+。

compound 410:1H NMR(400MHz,DMSO-d6)δ8.75(s,0.5H),8.57(br s,0.5H),8.19(br d,J=7.1Hz,0.6H),8.02(br d,J=7.7Hz,0.4H),7.39-7.07(m,5H),7.01-6.88(m,1H),6.83(s,1H),5.53(br d,J=6.4Hz,0.4H),5.27(br dd,J=4.1,7.4Hz,0.6H),4.07-3.89(m,4H),3.82(br d,J=6.2Hz,1H),2.46-2.34(m,1H),2.01-1.79(m,3H);LCMS(ESI)m/z:350.2[M+H]+。

compound 411:1H NMR(400MHz,CDCl3)δ7.28-7.20(m,1H),7.17-7.12(m,1H),6.86-6.79(m,1H),6.72(m,0.6H),6.67(m,0.4H),6.00-5.96(m,2H),5.91(m,0.3H),5.76(m,0.6H),5.56(s,0.3H),5.33(m,0.6H),4.16-4.08(m,1H),3.98(br s,1H),3.89-3.82(m,3H),3.81-3.66(m,1H),2.27-2.15(m,2H),2.07-1.95(m,2H);LCMS(ESI)m/z:384.1[M+H]+。

compound 412:1H NMR(400MHz,CDCl3)δ7.29-7.20(m,1H),7.17-7.11(m,1H),6.86-6.79(m,1H),6.74-6.68(m,1H),6.23(m,0.6H),6.07-6.02(m,0.6H),5.99-5.96(m,2H),5.43(dd,J=2.9,7.6Hz,0.6H),4.20-3.68(m,2H),2.30-1.98(m,4H);LCMS(ESI)m/z:431.9,433.9[M+H]+。

compound 413:1H NMR(400MHz,CDCl3)δ7.40-7.32(m,1H),7.28-7.24(m,1H),6.96-6.90(m,1H),6.82-6.76(m,1H),6.10-6.04(m,2H),4.96-4.78(m,0.2H),4.35(m,0.7H),4.34-3.78(m,3H),3.78-3.80(1H),3.43-3.38(m,1H),2.33-2.34(m,1H),2.09-1.91(m,4.6H),1.58-1.33(m,5H);LCMS(ESI)m/z:371.2[M+H]+。

compound 414:1H NMR(400MHz,DMSO-d6)δ7.68-7.52(m,0.5H),7.49-7.39(m,1H),7.36-6.95(m,7H),6.63-6.54(m,0.5H),6.16-6.04(m,2H),5.52-5.37(m,0.3H),5.32-5.15(m,0.7H),5.03-4.83(m,1H),4.38(br s,1H),4.20-3.97(m,0.8H),3.92-3.79(m,0.8H),3.76-3.68(m,0.2H),3.59(br s,0.2H),2.66(br d,J=1.8Hz,0.3H),2.43-2.31(m,0.7H),2.09-1.75(m,1H);LCMS(ESI)m/z:379.2[M+H]+。

compound 415:1H NMR(400MHz,DMSO-d6)δ8.48-8.34(m,1H),7.97(d,J=8.6Hz,0.6H),7.83(br d,J=8.8Hz,0.4H),7.61-7.45(m,1H),7.35-7.01(m,4H),6.81(s,1H),5.73(br d,J=7.9Hz,0.4H),5.41(br dd,J=5.0,7.6Hz,0.6H),4.09-3.78(m,5H),2.46-2.36(m,1H),2.06-1.75(m,3H);LCMS(ESI)m/z:368.0[M+H]+;LCMS(ESI)m/z:368.0[M+H]+。

compound 416:1H NMR(400MHz,DMSO-d6)δ=8.47-8.34(m,1H),7.97(br d,J=8.6Hz,1H),7.80(br d,J=8.8Hz,1H),7.60-7.46(m,1H),7.39-7.19(m,3H),7.16-7.04(m,1H),6.73(s,1H),5.52(br d,J=7.1Hz,0.5H),5.30-5.21(m,0.5H),4.06-3.97(m,1H),3.95-3.87(m,3H),3.82(br s,1H),2.46-2.34(m,1H),2.00-1.79(m,3H);LCMS(ESI)m/z:350.0[M+H]+。

compound 417: 1HNMR (400MHz, DMSO-d6) δ 7.50(d, J ═ 2.0Hz, 0.6H), 7.47(dd, J1 ═ 8.0Hz, J2 ═ 2.4Hz, 0.6H), 7.42(d, J ═ 2.0Hz, 0.3H), 7.39(dd, J1 ═ 8.4Hz, J2 ═ 2.0Hz, 0.3H), 7.28(d, J ═ 2.0Hz, 0.5H), 7.20(s, 0.6H), 7.19(d, J ═ 2.0Hz, 0.3H), 7.08(d, J ═ 8.4Hz, 0.6H), 7.04(d, J ═ 8.0, 0.3H), 7.00(s, 0.92, 6H), 7.70 (d, J ═ 3.5H), 3.5H, 3.70 (d, 3.5H), 3.5H, 3.70H, 3.5H, 3.70 (d, 3.5H), 3.5H, 3.70H, 3.5H, 3H, 3.5H, 3H, 3.5H, 1H) 2.05-1.82(m, 3H); LCMS (ESI) M/z 367.0[ M + H ] +.

Compound 418: 1HNMR (400MHz, DMSO-d6) δ 7.50(d, J ═ 2.0Hz, 0.6H), 7.47(dd, J1 ═ 8.4Hz, J2 ═ 2.0Hz, 0.6H), 7.39(d, J ═ 1.6Hz, 0.3H), 7.35(dd, J1 ═ 8.0Hz, J2 ═ 1.6Hz, 0.3H), 7.19(s, 0.6H), 7.08(d, J ═ 8.0Hz, 0.7H), 7.02(d, J ═ 8.0Hz, 0.3H), 6.99(s, 0.3H), 6.18-6.08(m, 2H), 5.65(dd, J1 ═ 8.0, J ═ 63, J ═ 2.63, J673H), 6.3H, 5.65 (dd0.3H, 3H), 7.3H, 3H, 7.3H, 3H, 3.3H, 3H, 3.8.3H, 3H, 3.7.7.06 (ddh, 3H, 7.8.; LCMS (ESI) M/z 365.0[ M + H ] +.

Compound 419:1H NMR(400MHz,CDCl3)δ7.74(s,0.5H),7.68-7.57(m,1.4H),7.40(br d,J=8.4Hz,0.8H),7.36-7.27(m,3H),7.26-7.20(m,1.5H),7.15(m,0.5H),6.94(s,0.5H),6.71(s,0.2H),5.78(m,0.3H),5.59(m,1.3H),5.49(m,0.2H),5.36-5.27(m,1H),4.38-4.31(m,1H),4.09-4.03(m,0.4H),4.01-3.92(m,3.5H),2.59-2.44(m,3H);LCMS(ESI)m/z:389.2[M+H]+。

compound 420:1H NMR,(400MHz,DMSO-d6)δ=7.49-6.95(m,8.7H),6.56(br s,0.3H),6.15-6.05(m,2H),5.48(br s,1H),5.41-5.26(m,1H),4.37-3.90(m,2H),2.76(br s,1H),2.28-2.04(m,1H);LCMS(ESI)m/z:381.2[M+H]+。

compound 421:1H NMR(400MHz,DMSO-d6)7.65-7.44(m,2H),7.33-7.23(m,3H),7.21-6.98(m,3.8H),6.70(s,0.2H),6.14-6.06(m,2H),5.43(dd,J=5.3,8.4Hz,0.3H),5.18-5.05(m,1.7H),4.43-4.28(m,1H),4.16-3.99(m,1H),3.73(dd,J=5.7,11.2Hz,0.7H),3.56(dd,J=4.7,12.5Hz,0.3H),2.67-2.54(m,1H),1.92-1.69(m,1H);LCMS(ESI)m/z:379.1[M+H]+。

compound 422:1H NMR(400MHz,DMSO-d6)δ7.53-7.44(m,1H),7.35-7.12(m,5H),7.11-6.99(m,2H),6.65(s,1H),6.16-6.06(m,2H),5.44(t,J=7.5Hz,1H),5.23-5.09(m,1H),4.41-4.30(m,1H),4.02(dd,J=3.6,11.8Hz,1H),3.89-3.75(m,1H),2.40-2.30(m,1H),2.02-1.81(m,1H);LCMS(ESI)m/z:379.1[M+H]+。

compound 423: 1HNMR (400MHz, DMSO-d6) δ ═ 8.03(d, J ═ 0.8Hz, 0.5H), 7.99(d, J ═ 0.8Hz, 0.3H), 7.52(d, J ═ 1.6Hz, 0.5H), 7.49(d, J ═ 1.6Hz, 0.3H), 7.46(dd, J1 ═ 5.6Hz, J2 ═ 1.6Hz, 0.6H), 7.41(dd, J1 ═ 8.0Hz, J2 ═ 2.0Hz, 0.3H), 7.22(s, 0.6H), 7.15(d, J ═ 0.8Hz, 0.5H), 7.12(s, 0.4H), 7.10(s, 0.3H), 7.3H (s, 0.3.3H), 7.6H), 7.15(d, J ═ 0.8Hz, 0.5H), 7.12(s, 0.4H), 7.3H, 3H, 3.3H, 0.3H, 3.3H, 7.3H, 3.3H, 3.3.3H, 3.3.3.3.3H, 7.3H, 3.3H, 7.3.3.3.3H, 3.3.3H, 3H, 3.3.6H, 7.3H, 7.7.7.3.3.7.7.3.7.7.6H, 3H, 3.3H, 3.3.3, 2.12-1.96(m, 2.7H), 1.90-1.77(m, 0.4H). LCMS (ESI) M/z 354.1[ M + H ] +.

Compound 424: 1HNMR (400MHz, DMSO-d6) δ 7.48-7.45(m, 1H), 7.46(dd, J1 ═ 8.0Hz, J2 ═ 1.8Hz, 1H), 7.19-7.17(m, 1H), 7.09-7.07(m, 1H), 6.13(s, 2H), 4.41-4.14(m, 1H), 3.74-3.53(m, 2H), 2.80-2.66(m, 2H), 2.15-2.02(3H), 1.90-1.59(m, 7H), 1.51-1.44(m, 2H), 1.36-1.24(m, 2H); LCMS (ESI) M/z 384.2[ M + H ] +.

Compound 425:1H NMR(400MHz,DMSO-d6)δ8.04(s,0.4H),7.96(s,0.3H),7.62(dd,J=1.0,7.8Hz,0.5H),7.54(d,J=7.3Hz,0.3H),7.51-7.45(m,1H),7.31(d,J=1.8Hz,0.4H),7.26(dd,J=1.8,8.2Hz,0.4H),7.21(s,0.4H),7.10-6.99(m,2H),6.98-6.92(m,0.8H),6.56(d,J=7.5Hz,0.4H),6.15-6.05(m,2H),6.04(d,J=5.7Hz,0.4H),4.28(d,J=17.2Hz,3H),4.18(dt,J=4.0,7.4Hz,0.5H),4.02-3.90(m,1H),3.81-3.73(m,0.5H),2.55(br d,J=11.5Hz,1H),2.06-1.79(m,3H);LCMS(ESI)m/z:417.2[M+H]+。

compound 426:1H NMR(400MHz,CDCl3)δ8.41-8.26(m,1H),7.37-7.17(m,2H),6.97-6.86(m,1H),6.75(s,0.3H),6.42(d,J=1.5Hz,0.6H),6.24(d,J=1.5Hz,0.3H),6.09-6.03(m,2.3H),5.56(dd,J=4.0,6.7Hz,1H),4.23-4.03(m,2H),3.92-3.82(m,1H),2.41-2.26(m,2H),2.16-2.03(m,1H);LCMS(ESI)m/z:354.2[M+H]+。

compound 427:1H NMR(400MHz,DMSO-d6)δ7.52-6.96(m,8H),6.82(br s,1H),6.11(br s,2H),5.76(br s,0.3H),5.56-5.34(m,1.7H),4.36-4.00(m,2H),2.87-2.59(m,1H),2.38-2.18(m,1H);LCMS(ESI)m/z:381.2[M+H]+。

compound 428:1H NMR(400MHz,DMSO-d6)δ=7.89-7.72(m,1H),7.43(s,1H),7.38-7.21(m,3H),7.18-7.01(m,1H),6.92(s,1H),6.76-6.65(m,1H),6.49(br d,J=6.8Hz,1H),5.48(br d,J=7.3Hz,0.4H),5.28(br dd,J=4.0,7.5Hz,0.6H),4.06-3.80(m,2H),3.53-3.42(m,3H),2.46-2.35(m,1H),2.02-1.79(m,3H);LCMS(ESI)m/z:350.0[M+H]+。

compound 429:1H NMR(400MHz,DMSO-d6)δ8.44(br s,0.6H),8.29(br s,0.4H),7.86(br d,J=9.3Hz,0.6H),7.69(br d,J=8.2Hz,0.4H),7.36-7.19(m,4H),7.18-7.05(m,1H),6.96(s,0.6H),6.63(s,0.4H),6.56-6.44(m,1H),5.54(br d,J=6.4Hz,0.4H),5.26(br dd,J=4.2,7.3Hz,0.6H),4.04-3.76(m,2H),3.56-3.46(m,3H),2.47-2.31(m,1H),2.01-1.78(m,3H);LCMS(ESI)m/z:350.2[M+H]+。

compound 430: 1HNMR (400MHz, DMSO-d6) δ ═ 7.51(d, J ═ 2.0Hz, 0.6H)7.48, (dd, J1 ═ 8.0Hz, J2 ═ 2.0Hz, 0.6H), 7.37(d, J ═ 1.6Hz, 0.4H), 7.33(dd, J1 ═ 8.0Hz, J2 ═ 1.6Hz, 0.4H), 7.20(s, 0.6H), 7.09(d, J ═ 8.0Hz, 0.6H), 7.05-6.98(m, 1H), 6.96-6.87(m, 1.4H), 6.82(dd, J1 ═ 8.4Hz, J2 ═ 2.4, 0.3H, 6.70H), 6.96-6.87(m, 1.4H), 6.82(dd, J1 ═ 8.4Hz, J3.4, J3.8.4, J3, J5 ═ 8.7.7.7.7.7.7.7.7H, J-6H, J5 ═ 2.7.7.7.7.7.7.7.7.8, 7.8, 7.8.8, 7.8, 7.7.8, 7.7.7.7.8, 7.8, 7, 7.8, 7., 1.96-1.66(m, 3H); LCMS (ESI) M/z 411.2[ M + H ] +.

Compound 431:1H NMR(400MHz,DMSO-d6)δ8.74(d,J=5.1Hz,1H),8.68(d,J=4.9Hz,1H),7.52-7.43(m,1H),7.39-7.25(m,2H),7.15(m,0.5H),7.06-7.08(d,J=8.2Hz,0.5H),7.02-6.95(m,0.5H),6.97(s,0.4H),6.09(d,J=11.9Hz,2H),5.62(dd,J=2.4,8.2Hz,0.4H),5.22(dd,J=4.4,8.2Hz,0.5H),4.04-3.92(m,1H),3.83-3.66(m,1H),2.45-2.32(m,1H),2.13-1.69(m,3H);LCMS(ESI)m/z:365.2[M+H]+。

compound 432:1h NMR (400MHz, chloroform-d) δ 8.73-8.61(m, 1H), 8.43(d, J ═ 13.7Hz, 1H), 8.24(s, 0.5H), 8.07(s, 0.5H), 7.43-7.32(m, 1H), 7.28(br s, 1H), 7.27-7.23(m, 2H), 7.19-7.09(m, 2H), 6.76(s, 1H), 5.82-5.76(m, 0.5H), 5.41(dd, J ═ 4.0, 7.6Hz, 0.5H), 4.33-4.15(m, 1H), 4.04-3.89(m, 1H), 2.56-2.34(m, 1H), 2.18-1.95(m, 3H); LCMS (ESI) M/z 360.2[ M + H ]]+。

Compound 433:1H NMR(400MHz,DMSO-d6)δ9.41-9.27(m,1H),8.34(dd,J=1.5,8.5Hz,0.6H),8.18(dd,J=1.5,8.5Hz,0.4H),7.99-7.83(m,1H),7.62(s,1H),7.39-7.19(m,4H),7.16-7.06(m,1H),5.54(dd,J=2.4,7.5Hz,0.4H),5.26(dd,J=4.2,7.8Hz,0.6H),4.11-4.03(m,0.5H),3.93(td,J=7.2,10.9Hz,0.5H),3.87-3.70(m,1H),2.45-2.34(m,1H),2.03-1.71(m,3H);LCMS(ESI)m/z:321.2[M+H]+。

compound 434:1H NMR(400MHz,DMSO-d6)δ7.54-7.45(m,2H),7.36-7.29(m,1H),7.28-7.20(m,3H),7.19-7.06(m,3H),6.13(s,2H),4.66(br s,0.4H),4.35(br s,0.6H),3.72-3.53(m,2H),3.15(dd,J=3.0,12.9Hz,0.6H),2.88(dd,J=4.3,13.1Hz,0.4H),2.76-2.58(m,1H),2.05-1.61(m,4H);LCMS(ESI)m/z:377.1[M+H]+。

compound 435:1H NMR(400MHz,DMSO-d6)δ9.23-9.08(m,1H),8.60-8.45(m,1H),8.26(d,J=8.4Hz,0.6H),8.09(d,J=8.2Hz,0.4H),7.57(s,1H),7.37-7.30(m,1H),7.29-7.17(m,3H),7.14-7.07(m,1H),5.50(dd,J=2.6,7.5Hz,0.4H),5.24(dd,J=4.3,7.8Hz,0.6H),4.04(td,J=6.4,10.9Hz,1H),3.95-3.74(m,1H),2.47-2.32(m,1H),1.99-1.75(m,3H);LCMS(ESI)m/z:345.1[M+H]+。

compound 436:1H NMR(400MHz,DMSO-d6)δ8.00-7.90(m,2H),7.62-7.50(m,3H),7.37-7.29(m,2H),7.28-7.21(m,2H),7.19-7.09(m,2H),4.68(br s,0.4H),4.37(br s,0.6H),3.74-3.55(m,2H),3.16(dd,J=3.2,12.9Hz,0.6H),2.89(dd,J=4.6,12.8Hz,0.4H),2.77-2.60(m,1H),2.08-1.67(m,4H);LCMS(ESI)m/z:333.1[M+H]+。

compound 437:1H NMR(400MHz,CDCl3)δ8.10-7.98(m,1H),7.32-7.26(m,0.5H),7.20(d,J=1.3Hz,0.5H),7.16-7.09(m,1.4H),7.07-6.95(m,1H),6.89-6.78(m,1H),6.72(s,0.5H),6.38(s,0.3H),6.00(d,J=10.8Hz,2H),5.96(d,J=2.6Hz,0.2H),5.68(dd,J=4.2,7.9Hz,0.5H),4.27-4.15(m,1H),4.07-3.99(m,0.4H),3.90-3.77(m,3.3H),2.42-2.29(m,1H),2.21-2.12(m,1H),2.03-1.90(m,2H);LCMS(ESI)m/z:394.0[M+H]+。

compound 438: 1HNMR (400MHz, DMSO-d6) δ 7.88(d, J ═ 8.8Hz, 0.6H), 7.82(d, J ═ 8.8Hz, 0.4H), 7.77(d, J ═ 9.2Hz, 0.6H), 7.58(d, J ═ 9.2Hz, 0.4H), 7.50-7.46(m, 0.6H), 7.41-7.37(m, 0.4H), 7.34-7.27(m, 2.5H), 7.26-7.08(m, 3.5H), 5.73(d, J ═ 8.0Hz, 0.4H), 5.30(dd, J1 ═ 8.0Hz, J2 ═ 4.8Hz, 0.6H), 4.33-4.27(m, 0.6H), 4.6H, 4.3.3-3.79 (m, 3.3H), 3.3.3-7H, 3.3.3H), 3.3.3.3H, 3.3.3H, 3H, 3.3H, 3H; LCMS (ESI) M/z 393.0[ M + H ] +.

Compound 439: 1HNMR (400MHz, DMSO-d6) δ — 8.62-8.18(m, 1H), 7.54-7.37(m, 2H), 7.25-7.17(m, 0.7H), 7.13-6.97(m, 1.3H), 6.17-6.07(m, 2H), 5.74-5.66(m, 0.3H), 5.37(dd, J ═ 4.8, 8.0Hz, 0.6H), 4.03-3.86(m, 1.3H), 3.81-3.68(m, 0.6H), 2.43-2.26(m, 1H), 2.20-1.89(m, 5.2H), 1.79(s, 0.8H); LCMS (ESI) M/z 368.0[ M + H ] +.

Compound 440:1H NMR(400MHz,CDCl3)δ9.16-9.09(m,1H),8.68-8.58(m,1H),7.33-7.26(m,1H),7.23-7.10(m,2H),6.92-6.80(m,1H),6.76-6.66(m,1H),6.06-5.94(m,2H),5.84(br d,J=8.6Hz,0.3H),5.30(dd,J=3.3,8.4Hz,0.6H),4.29-4.12(m,1H),3.96-3.86(m,1H),2.53-2.37(m,1H),2.22-1.93(m,3H);LCMS(ESI)m/z:365.2[M+H]+。

compound 441:1H NMR(400MHz,DMSO-d6)δ9.71(s,0.5H),9.56(s,0.5H),8.69-8.58(m,1H),8.18(dd,J=1.4,9.4Hz,1H),8.08-7.94(m,1H),7.54(s,1H),7.37-7.30(m,1H),7.29-7.21(m,3H),7.17-7.08(m,1H),5.59(br d,J=6.8Hz,0.4H),5.26(dd,J=4.2,7.7Hz,0.6H),4.11-4.03(m,1H),3.98-3.71(m,1H),2.46-2.31(m,1H),1.97-1.75(m,3H);LCMS(ESI)m/z:360.0[M+H]+。

compound 442:1H NMR(400MHz,CDCl3-d)δ7.35-7.29(m,1H),7.25-7.19(m,1H),6.93-6.86(m,1H),6.80(s,1H),6.74(s,1H),6.07-5.96(m,2H),5.83(s,0.4H),5.45(br d,J=6.4Hz,0.6H),4.21-4.00(m,1H),3.91-3.77(m,1H),2.43-2.33(m,3H),2.32-2.16(m,3H),2.14-1.95(m,1H);LCMS(ESI)m/z:368.1[M+H]+。

compound 443:1H NMR(400MHz,DMSO-d6)δ7.83(dd,J=1.8,12.1Hz,0.6H),7.75-7.64(m,1H),7.57(br d,J=9.0Hz,0.4H),7.37-7.20(m,5H),7.16-7.05(m,1H),6.95(s,1H),5.57(br d,J=6.2Hz,0.4H),5.24(dd,J=4.0,7.9Hz,0.6H),4.17(qd,J=7.0,14.1Hz,2H),4.07-3.69(m,2H),2.45-2.30(m,1H),1.97-1.73(m,3H),1.42-1.31(m,3H);LCMS(ESI)m/z:381.0[M+H]+。

compound 444:1H NMR(400MHz,CDCl3-d)δ7.58(d,J=8.2Hz,1H),7.44-7.39(m,1H),7.37-7.31(m,1H),7.30-7.27(m,1H),7.27-7.21(m,2H),7.20-7.10(m,1.5H),6.92(s,0.5H),6.58(s,1H),5.81(br d,J=6.6Hz,0.5H),5.45-5.37(m,0.5H),4.33-4.11(m,1H),4.04-3.87(m,1H),3.46(d,J=19.2Hz,3H),2.56-2.34(m,1H),2.14-1.95(m,3H);LCMS(ESI)m/z:390.2[M+H]+。

compound 445: 1H NMR (300MHz, chloroform-d) δ 9.23(s, 1H), 7.76(d, J ═ 8.8Hz, 2H), 7.37-7.24(m, 2H), 7.03-6.85(m, 2H), 6.79(s, 1H), 6.70(d, J ═ 9Hz, 2H), 4.19(dd, J ═ 2Hz, 1H), 3.86(s, 3H), 3.81(dd, J ═ 9Hz, 1H), 3.35-3.23(m, 1H), 2.43-2.33(m, 2H), 2.19-1.98(m, 2H); LCMS (ESI) M/z 364.0[ M + H ] +.

Compound 446:1H NMR(400MHz,CDCl3)δ7.29(dd,J=8.4,2.1Hz,0.7H),7.22(dd,J=8.0,2.0Hz,0.3H),7.20(d,J=1.6Hz,0.7H),7.13(d,J=1.2Hz,0.3H),6.97(d,J=1.2Hz,0.7H),6.92-6.85(m,1.3H),6.78(d,J=1.2Hz,0.7H),6.72(s,0.7H),6.65(d,J=1.2Hz,0.3H),6.54(s,0.3H),6.04(s,1.7H),6.03(s,0.3H),5.77(dd,J=8.0,3.6Hz,0.3H),5.31(dd,J=7.6,5.2Hz,0.7H),2.62-2.51(m,0.8H),2.36-2.25(m,2H),2.12-2.00(m,1.2H);LCMS(ESI)m/z:367.0[M+H]+。

compound 447:1H NMR(400MHz,CDCl3)δ8.44-8.33(m,1H),7.84-7.71(m,1H),7.28(m,0.5H),7.21-7.15(m,0.4H),7.09(d,J=1.3Hz,1H),7.04-6.92(m,1H),6.90-6.79(m,1H),6.73(s,0.5H),6.53(s,0.4H),6.14-6.07(m,0.3H),6.00(d,J=11.9Hz,2H),5.72(dd,J=4.4,7.7Hz,0.56H),4.32-4.19(m,1H),4.12-4.04(m,0.4H),3.90-3.82(m,0.4H),2.55-2.39(m,1H),2.18(m,0.6H),2.08-1.92(m,2.6H);LCMS(ESI)m/z:441.9[M+H]+。

compound 448:1H NMR(400MHz,CDCl3)δ8.69(dd,J=1.5,4.9Hz,0.6H),8.61(br d,J=3.5Hz,0.4H),7.95-7.88(m,0.85H),7.30-7.25(m,1H),7.22-7.17(m,1H),7.11(s,0.23H),6.90-6.78(m,1H),6.74-6.62(m,1H),6.17(br d,J=7.5Hz,0.3H),6.05-5.96(m,2H),5.64-5.57(m,0.7H),4.31-4.22(m,1.4H),4.09-4.03(m,0.3H),3.94-3.85(m,0.3H),2.63-2.46(m,1H),2.32-2.20(m,1H),2.12-1.96(m,2H);LCMS(ESI)m/z:389.0[M+H]+。

compound 449:1H NMR(400MHz,MeOD)δ8.01(s,0.4H)7.89(d,J=8.60Hz,0.4H)7.79(s,0.4H)7.58(d,J=8.38Hz,0.9H)7.20-7.44(m,2.9H)6.93-7.11(m,2.4H)5.59(dd,J=7.39,4.52Hz,0.5H)5.39(dd,J=7.83,4.30Hz,0.5H)4.20-4.29(m,0.5H)4.06-4.15(m,0.5H)3.98-4.10(m,1H)2.40-2.58(m,1H)1.90-2.13(m,3H);LCMS(ESI)m/z:371.0[M+H]+。

compound 450:1H NMR(400MHz,DMSO-d6)δ9.07-8.99(m,2H),8.68(d,J=1.8Hz,1H),8.53-8.39(m,1H),8.31-8.17(m,1H),7.69(s,1H),7.39-7.20(m,4H),7.12(br d,J=7.9Hz,1H),5.58(br d,J=6.6Hz,0.4H),5.27(dd,J=3.9,7.9Hz,0.6H),4.13-4.03(m,1H),4.00-3.73(m,1H),2.47-2.34(m,1H),2.01-1.74(m,3H);LCMS(ESI)m/z:371.0[M+H]+。

compound 451:1H NMR(400MHz,DMSO-d6)δ8.10(d,J=2.0Hz,0.6H),7.97-7.89(m,1H),7.77(dd,J=2.5,9.6Hz,0.4H),7.36-7.29(m,1H),7.28-7.20(m,3H),7.18-7.06(m,1.7H),6.80(s,0.3H),6.52-6.39(m,1H),5.57(br d,J=6.2Hz,0.4H),5.23(dd,J=4.1,7.8Hz,0.6H),4.05-3.95(m,1H),3.92-3.69(m,1H),2.44-2.31(m,1H),1.97-1.72(m,3H);LCMS(ESI)m/z:336.1[M+H]+。

compound 452:1H NMR(400MHz,CDCl3)δ7.30(dd,J=1.5,8.2Hz,1H),7.22-7.17(m,1H),7.11(s,0.3H),7.04(t,J=8.5Hz,0.6H),6.91-6.80(m,1.4H),6.75(s,0.6H),6.64-6.48(m,2.3H),6.01(d,J=11.7Hz,2H),5.90(m,0.4H),5.46(m,0.6H),4.23-4.05(m,1H),3.97-3.84(m,1H),3.72(d,J=19.8Hz,3H),2.40-2.28(m,1H),2.08-1.87(m,3H);LCMS(ESI)m/z:410.9[M+H]+。

compound 453:1H NMR(400MHz,CDCl3)δ7.27(dd,J=1.5,8.2Hz,0.6H),7.18(d,J=1.3Hz,0.6H),7.16-7.07(m,1H),7.04-6.93(m,1H),6.88-6.79(m,1H),6.72-6.60(m,2H),6.52-6.42(m,1H),6.06(s,0.3H),6.02-5.98(m,2H),5.73(t,J=7.2Hz,0.3H),5.59(t,J=7.7Hz,0.6H),4.27-4.19(m,0.7H),4.06-3.92(m,1H),3.85(s,2H),3.79-3.71(m,1.6H),2.41-2.27(m,1H),2.16-1.89(m,3H);LCMS(ESI)m/z:411.2[M+H]+。

compound 454:1H NMR(400MHz,MeOD)δ8.09(d,J=8.38Hz,0.5H)7.90(s,0.5H)7.78(d,J=8.16Hz,0.5H)7.67-7.69(m,0.5H)7.50-7.59(m,1.6H)7.29-7.44(m,2.5H)7.21-7.29(m,0.5H)7.02-7.12(m,2H)6.96(s,0.5H)6.52(d,J=1.98Hz,0.5H)6.45(d,J=1.76Hz,0.5H)5.64(dd,J=7.61,4.74Hz,0.5H)5.39-5.45(m,0.5H)4.23-4.32(m,0.5H)4.09-4.19(m,0.5H)3.91-4.04(m,2.5H)3.87(s,1.5H)2.40-2.62(m,1H)1.89-2.15(m,3H);LCMS(ESI)m/z:373.0[M+H]+。

compound 455: 1HNMR (400MHz, CDCl3) δ 9.31(s, 0.8H), 8.19(s, 0.2H), 7.55(d, J ═ 7.6Hz, 0.8H), 7.51(d, J ═ 8.4Hz, 0.2H), 7.36-7.25(m, 4H), 7.09(t, J ═ 7.6Hz, 1H), 6.92(d, J ═ 8.4Hz, 0.8H), 6.88(d, J ═ 8.0Hz, 0.2H), 6.06(s, 1.6H), 6.04(s, 0.4H), 6.21(br d, J ═ 8.4Hz, 0.2H), 5.00(br d, J ═ 6.0, 0.8H), 4.12-3.21 (br d, J ═ 8.4Hz, 0.2H), 2.00 (br d, J ═ 6.0, 0.8H), 4.12-3.85 (m), 2H), 1.01-2H, 1.2 m (m-2H), 1.2H, 1.2 m-2H, 1.2H, 2H, 1.2H, 2H; LCMS (ESI) M/z 406.2[ M + H ] +.

Compound 456:1H NMR(400MHz,CDCl3)δ8.31(m,0.5H),8.01(m,0.5H),7.95(s,0.4H),7.76(s,0.4H),7.60(m,0.52H),7.49(m,0.4H),7.35-7.29(m,1H),7.27(m,1.2H),7.23(m,0.2H),7.14-7.08(m,1H),7.05-6.97(m,1.32H),5.80-5.69(m,0.5H),5.42(dd,J=4.2,7.5Hz,0.5H),4.32(m,0.6H),4.23-4.13(m,0.6H),4.05-3.94(m,1H),2.51-2.36(m,1H),2.16-1.91(m,3H);LCMS(ESI)m/z:318.2[M+H]+。

compound 457: 1H NMR(400MHz,CDCl3)δ8.34-8.23(m,1H),7.36(m,0.5H),7.31-7.27(m,1H),7.22-7.15(m,2H),7.12-7.07(m,1H),6.90-6.81(m,1H),6.74(s,0.5H),6.56(s,0.3H),6.08(m,0.4H),6.04-5.96(m,2H),5.61(m,.0.6H),4.24(t,J=6.6Hz,1.4H),4.05(m,0.3H),3.93-3.83(m,0.3H),2.54-2.36(m,1H),2.29-2.18(m,1H),2.11-1.96(m,3H);LCMS(ESI)m/z:381.9[M+H]+。

Compound 458:1H NMR(400MHz,CDCl3)δ9.17-9.04(m,2H),7.80(s,0.6H),7.74-7.64(m,1.3H),7.46(d,J=8.4Hz,0.6H),7.38-7.30(m,1H),6.99-6.90(m,1H),5.97(m,0.3H),5.32(m,0.6H),4.32-4.17(m,1.3H),4.07-3.99(m,3H),3.98-3.90(m,0.4H),2.60-2.52(m,3H),2.53-2.43(m,1H),2.14-2.01(m,2H),1.99-1.82(m,1H);LCMS(ESI)m/z:389.2[M+H]+。

compound 459:1H NMR(400MHz,CDCl3)δ8.13-8.03(m,1H),7.34-7.19(m,2H),7.08-6.93(m,1H),6.91-6.81(m,1H),6.77-6.69(m,1H),6.09(m,0.3H),6.06-5.98(m,2H),5.49(dd,J=5.1,8.2Hz,0.7H),4.28-4.14(m,1H),3.99-3.85(m,1H),2.56-2.39(m,1H),2.11-1.83(m,3H);LCMS(ESI)m/z:460.1[M+H]+。

compound 460:1H NMR(400MHz,CDCl3)δ8.43-8.38(m,1H),8.35-8.25(m,1H),7.31(dd,J=1.5,8.2Hz,1H),7.23-7.17(m,1H),7.13-6.96(m,1H),6.91-6.81(m,1H),6.78-6.65(m,1H),6.10(m,0.3H),6.04-5.94(m,2H),5.52(m,0.6H),4.28-4.11(m,1H),3.99-3.85(m,1H),2.46(m,1H),2.10-1.89(m,3H);LCMS(ESI)m/z:381.9[M+H]+。

compound 461:1H NMR(400MHz,CDCl3-d)δ7.74-7.64(m,1H),7.57-7.45(m,1H),7.36-7.28(m,3H),7.25-7.20(m,1H),7.13(d,J=1.6Hz,0.3H),6.90(d,J=8.1Hz,0.7H),6.86-6.75(m,1H),6.14(dd,J=1.9,7.5Hz,0.4H),6.08-5.98(m,2H),5.57(dd,J=3.2,7.9Hz,0.6H),4.42-4.31(m,0.7H),4.26-4.05(m,1H),3.87(td,J=8.4,12.1Hz,0.3H),2.54-2.26(m,3H),2.22-2.03(m,1H);LCMS(ESI)m/z:404.4[M+H]+。

compound 462:1H NMR(400MHz,CDCl3)δ.05-8.96(m,1H),7.74(s,1H),7.67-7.58(m,1H),7.46(br d,J=8.6Hz,1H),7.42-7.36(m,1H),7.33-7.28(m,1H),6.91-6.80(m,1H),6.03(m,0.3H),5.49(m,0.6H),4.29-4.11(m,1H),4.03-3.91(m,4H),2.54-2.48(m,3H),2.47-2.36(m,1H),2.35-2.18(m,2H),2.11-1.99(m,1H);LCMS(ESI)m/z:389.4[M+H]+。

compound 463:1H NMR(400MHz,CDCl3-d)δ7.37-7.29(m,2H),7.27-7.18(m,2H),7.11(d,J=7.7Hz,1H),5.74(br d,J=6.4Hz,0.5H),5.46(s,0.5H),5.37(dd,J=3.5,8.1Hz,0.5H),5.09(s,0.5H),4.20-4.03(m,1H),3.95-3.86(m,1H),3.86-3.80(m,2H),3.74(t,J=4.7Hz,2H),3.40-3.33(m,2H),3.24-3.10(m,2H),2.48-2.28(m,1H),2.11-1.88(m,3H);LCMS(ESI)m/z:328.1[M+H]+。

compound 464:1H NMR(400MHz,CDCl3-d)δ8.53(s,1H),7.66(dd,J=0.8,8.3Hz,1H),7.42(s,1H),7.40-7.34(m,3H),7.27-7.23(m,2H),7.02(dd,J=1.3,8.2Hz,1H),5.03(d,J=16.3Hz,1H),4.96(dd,J=6.1,8.9Hz,1H),4.54(t,J=8.8Hz,1H),4.17(dd,J=6.0,8.8Hz,1H),4.13(s,0.5H),4.09(s,0.5H),4.04(s,3H),2.58(s,3H);LCMS(ESI)m/z:389.2[M+H]+。

example 137 stearoyl-CoA desaturase (SCD) is a target for the Compounds of the invention

A. Materials and methods: compound spectrum analysis method

Strains expressing SCD1 or SCD5 as the only desaturase, human SCD1 and SCD5 genes were used to evaluate the inhibition of SCD1/SCD5 using reduced growth as a surrogate for SCD inhibition. These yeast strains express human SCD1 or SCD5 from plasmids carried in strains lacking the yeast OLE1 gene.

All compound profiling experiments were performed using the same basic protocol. Use of standard techniques in the absence ofYeast were grown in complete synthetic medium supplemented with 2% (w/v) glucose (SD-Ura) depleted of uracil and containing yeast nitrogen base. The starter culture was inoculated in 3mL SD-Ura medium containing 0.01% tween and 0.2mM palmitoleic and oleic acids. The cultures were incubated overnight at 30 ℃ in a shaker incubator (225 rpm). Saturated morning cultures were centrifuged, washed in SD-Ura medium lacking TWEEN-20 and fatty acids, and then diluted 1:20 in fresh SD-Ura medium also lacking TWEEN-20 and fatty acids. Cells were grown at 30 ℃ for 6h with shaking to an OD of about 0.4-0.8 600(optical density).

Compound stocks (10mM in 100% DMSO) were arrayed into 384 round-hole v-bottom polypropylene plates and diluted according to the dilution factors indicated. Compound administration was performed in two separate steps. First, using MULTIDROPTMThe Combi reagent dispenser dispenses 15 μ L SD-Ura into 384 well clear assay plates. An automated workstation equipped with a 384 pin tool (Perkin Elmer JANUS) was then usedTM) The diluted compound stock plate was applied to an assay plate, the tool containing a slotted pin that delivered 100nL of compound. The above cultures were centrifuged and washed with medium lacking TWEEN-20 or oleic acid and palmitoleic acid. The culture was then concentrated to OD600 (final OD) at 2-fold of 0.026000.0.01) was resuspended in SD-Ura. Then 15. mu.L of the diluted culture was dispensed into a marketed assay plate to achieve 30. mu.L of 1 XOD600Culture (0.01) and maximum drug concentration of 33.3. mu.M.

After yeast delivery, assay plates were incubated for 40h at 30 ℃ under humidified conditions. By using microplate reader (Perkin Elmer EnVision)TM) Read OD of each well600Yeast growth was monitored. The data were analyzed as follows. The values were converted to the percentage of untreated conditions for this strain [ (EXP-0.035)/(DMSO-0.035). times.100%) ]To process the raw data.

B. Results

Using the methods described above, compounds of the invention were tested for inhibition of SCD1 and SCD 5. The results are shown in Table 2.

TABLE 2 inhibition of SCD1 and SCD5 by compounds of the present invention

Other embodiments

While the present invention has been described with reference to what are presently considered to be the preferred embodiments, it is to be understood that the invention is not limited to the disclosed embodiments. On the contrary, the invention is intended to cover various modifications and equivalent arrangements included within the spirit and scope of the appended claims.

All publications, patents, and patent applications are herein incorporated by reference in their entirety to the same extent as if each individual publication, patent, or patent application was specifically and individually indicated to be incorporated by reference in its entirety. Where a term in the present application is found to be defined differently in a document incorporated by reference herein, the definition provided herein will be used as a definition of the term.

Other embodiments are within the scope of the following claims.

296页详细技术资料下载
上一篇:一种医用注射器针头装配设备
下一篇:GLP-1受体激动剂及其用途

网友询问留言

已有0条留言

还没有人留言评论。精彩留言会获得点赞!

精彩留言,会给你点赞!