Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
THIAZOLE DERIVATIVES AS PROTEIN SECRETION INHIBITORS
Document Type and Number:
WIPO Patent Application WO/2020/176863
Kind Code:
A1
Abstract:
Provided herein are secretion inhibitors, such as inhibitors of Sec61, methods for their preparation, related pharmaceutical compositions, and methods for using the same, wherein the compound has a structure of Formula (I), (II), or (III).

Inventors:
MCMINN DUSTIN (US)
RAO MEERA (US)
Application Number:
PCT/US2020/020419
Publication Date:
September 03, 2020
Filing Date:
February 28, 2020
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
KEZAR LIFE SCIENCES (US)
International Classes:
A61P35/00; A61K31/427; C07D277/18; C07D401/06; C07D413/14; C07D417/12; C07D417/14
Domestic Patent References:
WO2005095386A12005-10-13
WO2003015778A12003-02-27
WO2010101949A12010-09-10
WO2014145642A22014-09-18
WO2010126914A12010-11-04
Foreign References:
US6468798B12002-10-22
US6472375B12002-10-29
US6471996B12002-10-29
US4522811A1985-06-11
Other References:
JUN ZUO ET AL: "Discovery of Structurally Diverse Small-Molecule Compounds with Broad Antiviral Activity against Enteroviruses", ANTIMICROBIAL AGENTS AND CHEMOTHERAPY, vol. 60, no. 3, 1 March 2016 (2016-03-01), US, pages 1615 - 1626, XP055694837, ISSN: 0066-4804, DOI: 10.1128/AAC.02646-15
KAI-UWE KALIES ET AL: "Inhibitors of Protein Translocation Across the ER Membrane : ER Protein Translocation Inhibitors", TRAFFIC, vol. 16, no. 10, 27 July 2015 (2015-07-27), DK, pages 1027 - 1038, XP055694929, ISSN: 1398-9219, DOI: 10.1111/tra.12308
DATABASE REGISTRY [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; 1 May 2011 (2011-05-01), XP002799007, Database accession no. 1287868-13-6
DATABASE REGISTRY [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; 27 May 2011 (2011-05-27), XP002799008, Database accession no. 1301352-46-4
DATABASE REGISTRY [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; 10 November 2010 (2010-11-10), XP002799009, Database accession no. 1252203-85-2
DATABASE REGISTRY [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; 12 April 2011 (2011-04-12), XP002799010, Database accession no. 1278765-37-9
DATABASE REGISTRY [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; 18 November 2018 (2018-11-18), XP002799011, Database accession no. 2249352-34-7
DATABASE REGISTRY [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; 24 May 2011 (2011-05-24), XP002799012, Database accession no. 1299339-46-0
KALIESROMISCH, TRAFFIC, vol. 16, no. 10, 2015, pages 1027 - 1038
SMITH, M. B.MARCH, J.: "March l:s Advanced Organic Chemistry: Reactions, Mechanisms, and Structure", 2001, JOHN WILEY & SONS
GREENE, T.W.WUTS, P.G. M.: "Protective Groups in Organic Synthesis", 1999, JOHN WILEY & SONS
BERGE ET AL.: "Pharmaceutical Salts", J. PHARM. SCI., vol. 66, 1977, pages 1 - 19, XP002675560, DOI: 10.1002/jps.2600660104
HAMAJIMA ET AL., CLIN. IMMUNOL. IMMUNOPATHOL., vol. 88, no. 2, 1998, pages 205 - 10
Attorney, Agent or Firm:
DANEK, Shelley, C. et al. (US)
Download PDF:
Claims:
What is Claimed is:

1. A compound having a structure of Formula (III):

wherein:

L1 is a bond, , wherein = indicates a double bond, a triple bond, or a fused cyclopropyl;

B is Co-3alkylene-X;

X is an aromatic or nonaromatic C4-iocarbocycle, or an aromatic or nonaromatic 4-10 membered heterocycle having 1-3 ring heteroatoms selected from N, O, and S;

R2 is H or Ci-3alkyl;

L2 is Co-3alkylene;

m is 0 to 2; and

each R4 independently is Ci-3alkyl, C^alkynyl, Ci-3haloalkyl, Ci-3alkoxy, halo, or NHCi-3alkylene-aryl;

or a pharmaceutically acceptable salt thereof.

2. The compound or salt of claim 1 , wherein L1 is a bond.

3. The compound or salt of claim 1 , wherein L1 is Ci-6alkylene.

4. The compound or salt of claim 3, wherein L1 is CH2, CH2CH2, C(CH3)2, C(CH3)2CH2, or C(CH3)2CH2CH2.

5. The compound or salt of claim 1 , wherein L1 is

6. The compound or salt of claim 5, wherein L1 is

7. The compound or salt of claim 5, wherein L1 is

8. The compound or salt of claim 5, wherein L1 is .

9. The compound or salt of any one of claims 5-8, wherein— indicates a double bond.

10. The compound or salt of claim 9, wherein the double bond is further substituted with

Ci-3alkyl.

1 1. The compound or salt of any one of claims 5-8, wherein = indicates a triple bond.

12. The compound or salt of any one of claims 5-8, wherein = indicates a fused cyclopropyl, e.g., .

13. The compound or salt of any one of claims 1-12, wherein B is Ci-3alkylene-X.

14. The compound or salt of any one of claims 1-12, wherein B is X.

15. The compound or salt of any one of claims 1-14, wherein X is pyrrolidinyl, cyclobutyl, cyclopentyl, cyclohexyl, cyclohexenyl, phenyl, piperidinyl, pyridinyl, piperazinyl,

tetrahydrofuranyl, furanyl, tetrahydropyranyl, quinolinyl, morpholinyl, pyrrolidonyl, pyrimidinyl, pyridazinyl, indenyl, dihydroindenyl, dihydrobenzofuranyl, chromanyl, isochromanyl, dihydroisoquinolinyl, or indolyl.

16. The compound or salt of any one of claims 1-15,

wherein

X is substituted with 1-3 G;

each G independently is selected from the group consisting of halo, OH, =0, CN, NO2, N(Rn)2, N(RN)C(0)Ci-3alkyl, Ci-3alkyl, Ci-3alkoxy, Ci-3 haloalkyl, C1 -3 haloalkoxy, C(0)Ci- salkyl, S(02)-Z, C(0)-Z, C(0)N(RN)2, silyl ether, and [O]0-i-C0-3alkylene-Z;

each RN independently is H or Ci-4alkyl;

Z is aromatic or nonaromatic C3-iocarbocycle, or aromatic or nonaromatic 4-10 membered heterocycle having 1-3 heteroatoms selected from the group consisting of N, O, and S;

Z is optionally substituted with 1-3 E; and,

each E independently is selected from Ci-3alkyl, Ci-3alkoxy, =0, Ci-3haloalkoxy, CN, and halo.

17. The compound or salt of claim 1 ,

wherein

X X

X

U-B is selected from the group consisting of , and ^ ;

X is aromatic or nonaromatic C4-7 carbocycle, or an aromatic or nonaromatic 4-9 membered heterocycle having 1 ring heteroatom;

X is optionally substituted with 1-3 G;

each G independently is selected from the group consisting of halo, OH, =0, CN, N02, N(Rn)2, N(RN)C(0)Ci.3alkyl, Ci-3alkyl, C^alkoxy, C1-3 haloalkyl, C1.3 haloalkoxy, C(0)C salkyl, S(02)-Z, C(0)-Z, C(0)N(RN)2, silyl ether, and [O]0-i-C0-3alkylene-Z;

each RN independently is H or Ci-4alkyl;

Z is aromatic or nonaromatic C3-iocarbocycle, or aromatic or nonaromatic 4-10 membered heterocycle having 1-3 heteroatoms selected from the group consisting of N, O, and S;

Z is optionally substituted with 1-3 E; and,

each E independently is selected from Ci-3alkyl, Ci-3alkoxy, =0, Ci-3haloalkoxy, CN, and halo.

18. The compound or salt of claim 17, wherein U-B is selected from the group consisting

19. The compound or salt of claim 17 or 18, wherein U-B is selected from the group

21. The compound or salt of any one of claims 1-20, wherein R2 is H.

22. The compound or salt of any one of claims 1-20, wherein R2 is Ci-3alkyl.

23. The compound or salt of claim 22, wherein R2 is methyl.

24. The compound or salt of any one of claims 1-23, wherein L2 is Coalkylene.

25. The compound or salt of any one of claims 1-23, wherein L2 is Cialkylene.

26. The compound or salt of any one of claims 1-23, wherein L2 is C2alkylene.

27. The compound or salt of any one of claims 1-23, wherein L2 is Csalkylene.

28. The compound or salt of any one of claims 1-27, wherein m is 0.

29. The compound or salt of any one of claims 1-27, wherein m is 1 or 2.

30. The compound or salt of claim 29, wherein R4 is Ci-3alkyl.

31. The compound or salt of claim 30, wherein R4 is methyl or ethyl.

32. The compound or salt of claim 29, wherein R4 is halo.

33. The compound or salt of claim 32, wherein R4 is F.

34. The compound or salt of claim 32, wherein R4 is Cl.

35. The compound or salt of claim 29, wherein R4 is C^alkynyl.

36. The compound or salt of claim 35, wherein R4 is C2alkynyl.

37. The compound or salt of claim 29, wherein R4 is Ci-3haloalkyl.

38. The compound or salt of claim 37, wherein R4 is CF3.

39. The compound or salt of claim 29, wherein R4 is Ci-3alkoxy.

40. The compound or salt of claim 39, wherein R4 is methoxy.

41. The compound or salt of claim 29, wherein R4 is NHCi-3alkylene-aryl.

42. The compound or salt of claim 41 , wherein R4 is NH-CFh-phenyl.

43. The compound or salt of claim 29, wherein m is 2, and one R4 is halo, and the other R4 is halo or methyl.

44. The compound or salt of any one of claims 1 to 23, having the structure of Formula

(IIIA):

45. The compound or salt of claim 44, wherein U-B is selected from the group consisting

wo 2020/176863

sa„ , claim 44, wherein L--B is selected rom isting 6 The compound

47. The compound or salt of claim 1 , wherein the compound or salt is selected from the

wherein:

ring A is an aromatic or nonaromatic C3-10 carbocycle, or an aromatic or nonaromatic 5-10 membered heterocycle having 1 or 2 ring heteroatoms selected from N, O, and S; one of Q and Q’ is U-B and the other is R2;

L1 is a bond, Ci-6alkylene, or , wherein = indicates a double bond, a triple bond, or a fused or spiro cyclopropyl;

B is Ci-3alkoxy,[0]o-i-Co-3alkylene-X, or NRNCi-3alkylene-X;

X is an aromatic or nonaromatic C3-10 carbocycle, or an aromatic or nonaromatic 5-10 membered heterocycle having 1-4 ring heteroatoms selected from N, O, and S;

L2 is Co-6alkylene , wherein = indicates a double bond, a triple bond, or a fused or spiro cyclopropyl;

W is a bond, O, or C(0)N(RN);

D is C6-ioaryl or an aromatic or nonaromatic 5-10-membered heterocycle having 1-4 ring heteroatoms selected from N, O, and S;

each RN independently is H or Ci-4alkyl;

R1 is H or Ci-3alkyl; and

R2 is H, Ci-3alkyl, or halo,

or a pharmaceutically acceptable salt thereof.

49. The compound or salt of claim 48, wherein Q is U-B and Q’ is R2.

50. The compound or salt of claim 48, wherein Q is R2 and Q’ is U-B.

51. The compound or salt of any one of claims 48 to 50, wherein ring A is a 5-6 membered heterocycle having 1 or 2 ring heteroatoms selected from N, O, and S.

52. The compound or salt of claim 48 or 49, having the structure of Formula (IA):

wherein ring A has 0 or 1 additional ring heteroatoms selected from N, O, and S, and R3 is H, Ci-3alkyl, Ci-3hydroxyalkyl, Ci-3haloalkyl, halo, or -C(0)N(RN)2.

53. The compound or salt of any one of claims 48 to 50, wherein ring A is an aromatic or nonaromatic C3-10 carbocycle.

54. The compound or salt of any one of claims 48 to 50, wherein ring A-L2 moiety is

55. The compound or salt of claim 54, wherein ring A-L2 moiety

56. The compound or salt of any one of claims 48-55, wherein R1 is H.

57. The compound or salt of any one of claims 48-55, wherein R1 is Ci-3alkyl.

58. The compound or salt of claim 57, wherein R1 is methyl or ethyl.

59. The compound or salt of any one of claims 48-58, wherein R2 is H.

60. The compound or salt of any one of claims 48-58, wherein R2 is Ci-3alkyl.

61. The compound or salt of claim 60, wherein R2 is methyl.

62. The compound or salt of claim 60, wherein R2 is ethyl.

63. The compound or salt of claim 60, wherein R2 is n-propyl or isopropyl.

64. The compound or salt of any one of claims 48-58, wherein R2 is halo.

65. The compound or salt of claim 64, wherein R2 is Br.

66. The compound or salt of claim 64, wherein R2 is F.

67. The compound or salt of claim 64, wherein R2 is Cl.

68. The compound or salt of any one of claims 48-67, wherein U is a bond.

69. The compound or salt of any one of claims 48-67, wherein U is a Ci-6alkylene.

70. The compound or salt of claim 69, wherein U is CH2, CH(CH3), CH2CH2, or

C(CH3) .

71. The compound or salt of any one of claims 48-67, wherein U is

72. The compound or salt of claim 71 , wherein = indicates a double bond.

73. The compound or salt of claim 72, wherein the double bond is tri- or tetra- substituted, and the 1 or 2 other substituents on the double bond are independently selected from Ci-3alkyl and halo.

74. The compound or salt of claim 71 , wherein— indicates a triple bond.

75. The compound or salt of claim 71 , wherein— indicates a fused cyclopropyl, .

76. The compound or salt of any one of claims 71-75, wherein Co-2alkylene is CH2, CH(CH3), or CH2CH2.

77. The compound or salt of any one of claims 48-76, wherein B is Ci-3alkoxy.

78. The compound or salt of any one of claims 48-76, wherein B is O-X.

79. The compound or salt of any one of claims 48-76, wherein B is 0-Ci-3alkylene-X.

80. The compound or salt of any one of claims 48-76, wherein B is Ci-3alkylene-X.

81. The compound or salt of any one of claims 48-76, wherein B is X.

82. The compound or salt of any one of claims 48-76, wherein B is NHCi-3alkylene-X.

83. The compound or salt of any one of claims 48-76, wherein B is N(CH3)CI-

3alkylene-X.

84. The compound or salt of any one of claims 48-83, wherein X is an aromatic C6- locarbocycle, or an aromatic or nonaromatic 5-10-membered heterocycle.

85. The compound or salt of any one of claims 48-84, wherein X is selected from phenyl, pyridyl, indolyl, tetrahydropyranyl, piperidinyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, or piperazinyl, and X is optionally substituted with 1-3 substituents independently selected from Ci-3alkyl, Ci-3alkoxy, halo, Ci-3 haloalkyl, Ci-3 haloalkoxy, C(0)Ci-3alkyl, and S02Ci-3alkyl.

86. The compound or salt of any one of claims 48-67, wherein U-B is selected from

the group consisting of:

87. The compound or salt of claim 86, wherein U-B is selected from the group

88. The compound or salt of any one of claims 48-87, wherein L2 is Co-6alkylene.

89. The compound or salt of any one of claims 48-87, wherein L2 is Ci-6alkylene

90. The compound or salt of any one of claims 48-87, wherein L2 is

91. The compound or salt of claim 90, wherein— indicates a double bond.

92. The compound or salt of claim 90, wherein— indicates a triple bond.

93. The compound or salt of claim 90, wherein— indicates a fused cyclopropyl, .

94. The compound or salt of any one of claims 48-91 , wherein W is a bond.

95. The compound or salt of any one of claims 48-91 , wherein W is O.

96. The compound or salt of any one of claims 48-91 , wherein W is C(0)N(RN).

97. The compound or salt of claim 96, wherein W is C(0)NH.

98. The compound or salt of claim 96, wherein W is C(0)N(CI -4 alkyl).

99. The compound or salt of claim 98, wherein W is C(0)N(Me).

100. The compound or salt of any one of claims 48-99, wherein D is C6-ioaryl.

101. The compound or salt of any one of claims 48-99, wherein D is an aromatic 5-10- membered heterocycle having 1-4 ring heteroatoms selected from N, O, and S.

102. The compound or salt of claim 101 , wherein D comprises pyridyl optionally substituted with 1-3 substituents independently selected from Ci-3alkyl, Ci-3alkoxy, halo, C1 -3 haloalkyl, Ci-3 haloalkoxy, N(RN)2, C3-6cycloalkyl, N02, N(RN)C(0)Ci-3alkyl, C(0)C1.3alkyl, NO2, CN, SC>2Ci-3alkyl, O , NHCi-3alkylene-aryl, OCi-3alkylene-aryl, Ci-3alkylene-aryl, and 5- 10 membered heterocycle having 1-3 ring heteroatoms selected from N, O, and S, and each RN is independently H or Ci-4alkyl.

103. The compound or salt of any one of claims 48-99, wherein D is a nonaromatic 5- 10 membered heterocycle having 1-4 ring heteroatoms selected from N, O, and S.

104. The compound or salt of any one of claims 48-87, wherein L2-W-D is selected

wherein:

one of Q and Q’ is U-B and the other is R2, or

Q and Q’ and the atoms to which they are attached join together to form an aromatic or nonaromatic 5 or 6 membered carbocycle or a 5 or 6 membered heterocycle having 1 or 2 ring heteroatoms selected from N, O, and S;

U is a bond, Ci-6alkylene, or wherein = indicates a double bond, a triple bond, or a fused or spiro cyclopropyl;

B is C1-6 alkyl, C1-3 haloalkyl, C1.3 hydroxyalkyl, C1.3 haloalkoxy, Ci-3alkoxy, [O]o-1-Co- 3alkylene-X or NRNCi-3alkylene-X, X is an aromatic or nonaromatic C3-10 carbocycle, or an aromatic or nonaromatic 5-10 membered heterocycle having 1-4 ring heteroatoms selected from N, O, and S; indicates a double bond, a triple bond, or a fused or spiro cyclopropyl;

W is a bond, O, or C(0)N(RN);

D comprises pyridyl or quinolinyl optionally substituted with 1-3 substituents independently selected from Ci-3alkyl, Ci-3alkoxy, halo, Ci-3 haloalkyl, Ci-3 haloalkoxy, N(Rn)2, Cs-ecycloalkyl, N02, C(0)N(RN)2, N(RN)C(0)C1.3alkyl, C(0)Ci^alkyl, N02, CN, S02Ci-3alkyl, O , NHCi-3alkylene-aryl, OCi-3alkylene-aryl, Ci-3alkylene-aryl, and 5-10 membered heterocycle having 1-3 ring heteroatoms selected from N, O, and S.

each RN is independently H or Ci-4alkyl;

R1 is H or Ci-3alkyl;

R2 is H, Ci-3alkyl, or halo; and

R3 is H, Ci-3alkyl, Ci-3hydroxyalkyl, Ci-3haloalkyl, halo, or -C(0)N(RN)2;

or a pharmaceutically acceptable salt thereof.

106. The compound or salt of claim 105, wherein Q is U-B and Q’ is R2.

107. The compound or salt of claim 105, wherein Q is R2 and Q’ is U-B.

108. The compound or salt of claim 105, wherein Q and Q’ and the atoms to which they are attached join together to form an aromatic or nonaromatic 5 or 6 membered carbocycle or a 5 or 6 membered heterocycle having 1 or 2 ring heteroatoms selected from N, O, and S.

109. The compound or salt of any one of claims 105-108, wherein R3 is H.

110. The compound or salt of any one of claims 105-108, wherein R3 is Ci-3alkyl.

111. The compound or salt of claim 110, wherein R3 is methyl.

112. The compound or salt of any one of claims 105-108, wherein R3 is halo.

113. The compound or salt of claim 112, wherein R3 is Cl.

114. The compound or salt of any one of claims 105-108, wherein R3 is Ci-

3hydroxyalkyl.

115. The compound or salt of claim 114, wherein R3 is -CH2OH.

116. The compound or salt of any one of claims 105-108, wherein R3 is -C(0)N(RN)2.

117. The compound or salt of claim 116, wherein R3 is -C(0)NH2.

118. The compound or salt of claim 116, wherein R3 is -C(0)N(Me)2.

119. The compound or salt of any one of claims 105-108, wherein R3 is Ci-3haloalkyl.

120. The compound or salt of any one of claims 105-108, wherein the pyrrole ring-L2 moiety is selected from the group consisting of:

122. The compound or salt of any one of claims 105-121 , wherein R1 is H.

123. The compound or salt of any one of claims 105-121 , wherein R1 is Ci-3alkyl.

124. The compound or salt of claim 123, wherein R1 is methyl or ethyl.

125. The compound or salt of any one of claims 105-124, wherein R2 is H.

126. The compound or salt of any one of claims 105-124, wherein R2 is Ci-3alkyl.

127. The compound or salt of claim 126, wherein R2 is methyl.

128. The compound or salt of claim 126, wherein R2 is ethyl.

129. The compound or salt of claim 126, wherein R2 is n-propyl or isopropyl.

130. The compound or salt of any one of claims 105-124, wherein R2 is halo.

131. The compound or salt of claim 130, wherein R2 is Br.

132. The compound or salt of claim 130, wherein R2 is F.

133. The compound or salt of claim 130, wherein R2 is Cl.

134. The compound or salt of any one of claims 105-133, wherein U is a bond.

135. The compound or salt of any one of claims 105-133, wherein U is a Ci-6alkylene.

136. The compound or salt of claim 135, wherein U is CH2, CH(CH3), CH2CH2, or C(CH3)2.

137. The compound or salt of any one of claims 105-133, wherein U is

138. The compound or salt of claim 137, wherein— indicates a double bond.

139. The compound or salt of claim 138, wherein the double bond is tri- or tetra- substituted, and the 1 or 2 other substituents on the double bond are independently selected from Ci-3alkyl and halo.

140. The compound or salt of claim 137, wherein— indicates a triple bond.

141. The compound or salt of claim 137, wherein = indicates a fused cyclopropyl, .

142. The compound or salt of any one of claims 105-141 , wherein B is Ci-e alkyl.

143. The compound or salt of claim 142, wherein B is methyl, ethyl, n-propyl, isopropyl, n-butyl, sec-butyl, tert-butyl, n-pentyl, or n-hexyl.

144. The compound or salt of any one of claims 105-141 , wherein B is C1-3 haloalkyl.

145. The compound or salt of claim 144, wherein B is -CF3 or -CF2CH3.

146. The compound or salt of any one of claims 105-141 , wherein B is C1-3

hydroxyalkyl.

147. The compound or salt of claim 146, wherein B is -CH2CH2OH.

148. The compound or salt of any one of claims 105-141 , wherein B is C1-3 haloalkoxy.

149. The compound or salt of claim 148, wherein B is -OCH2CF3.

150. The compound or salt of any one of claims 105-141 , wherein B is Ci-3alkoxy.

151. The compound or salt of any one of claims 105-141 , wherein B is O-X.

152. The compound or salt of any one of claims 105-141 , wherein B is 0-Ci-3alkylene-

X.

153. The compound or salt of any one of claims 105-141 , wherein B is Ci-3alkylene-X.

154. The compound or salt of any one of claims 105-141 , wherein B is X.

155. The compound or salt of any one of claims 105-141 , wherein B is NHCi-

3alkylene-X.

156. The compound or salt of any one of claims 105-141 , wherein B is N(CH3)CI- 3alkylene-X.

157. The compound or salt of any one of claims 105-156, wherein X is an aromatic Ce- locarbocycle, or an aromatic or nonaromatic 5-10-membered heterocycle.

158. The compound or salt of any one of claims 105-157, wherein X is selected from phenyl, pyridyl, indolyl, tetrahydropyranyl, piperidinyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, or piperazinyl, and X is optionally substituted with 1-3 substituents independently selected from Ci-3alkyl, Ci-3alkoxy, halo, C1 -3 haloalkyl, C1 -3 haloalkoxy, C(0)Ci-3alkyl, and SC>2Ci-3alkyl.

159. The compound or salt of any one of claims 105-133, wherein U-B is selected

from the group consisting of:

161. The compound or salt of any one of claims 105-160, wherein L2 is Ci-6alkylene.

162. The compound or salt of any one of claims 105-160, wherein L2 is

j— C0-2alkylene - C0-2alkylene— j

163. The compound or salt of claim 162, wherein L2 is

164. The compound or salt of claim 162, wherein L2 is

165. The compound or salt of claim 162, wherein L2 is

166. The compound or salt of any one of claims 162-165, wherein = indicates a double bond.

167. The compound or salt of any one of claims 162-165, wherein = indicates a triple bond.

168. The compound or salt of any one of claim 162-165, wherein = indicates a fused cyclopropyl, e.g., , or a spiro cyclopropyl, e.g., .

169. The compound or salt of any one of claims 105-168, wherein W is a bond.

170. The compound or salt of any one of claims 105-168, wherein W is O.

171. The compound or salt of any one of claims 105-168, wherein W is C(0)N(RN).

172. The compound or salt of claim 171 , wherein W is C(0)NH2

173. The compound or salt of claim 171 , wherein W is C(0)N(CI -4 alkyl)2.

174. The compound or salt of claim 173, wherein W is C(0)N(Me)2.

175. The compound or salt of any one of claims 105-160, wherein L2-W-D is selected

176. The compound or salt of claim 175, wherein L2-W-D is

177. A compound listed in Table A, or a pharmaceutically salt thereof.

178. The compound or salt of claim 177, wherein the compound or salt is selected

180. A compound selected from a compound listed in Table B, or a pharmaceutically acceptable salt thereof.

181. A pharmaceutical composition comprising the compound or salt of any one of claims 1-180 and a pharmaceutically acceptable carrier.

182. A method of inhibiting protein secretion in a cell comprising contacting the cell with the compound or salt of any one of claims 1-180 or the composition of claim 181 in an amount effective to inhibit secretion.

183. The method of claim 182, wherein the protein is a checkpoint protein.

184. The method of claim 182, wherein the protein is a cell-surface protein, endoplasmic reticulum associated protein, or secreted protein involved in regulation of anti tumor immune response.

185. The method of claim 182, wherein the protein is at least one of PD-1 , PD-L1 , TIM-1 , LAG-3, CTLA4, BTLA, OX-40, B7H1 , B7H4, CD137, CD47, CD96, CD73, CD40, VISTA, TIGIT, LAIR1 , CD160, 2B4, TGFF^ and combinations thereof.

186. The method of claim 182, wherein the protein is selected from the group consisting of HER3, TNFa, IL2, and PD1.

187. The method of any one of claims 182-186, wherein the contacting comprising administering the compound or the composition to a subject in need thereof.

188. A method for treating inflammation in a subject comprising administering to the subject a therapeutically effective amount of the compound or salt of any one of claims 1- 180 or the pharmaceutical composition of claim 181.

189. A method for treating cancer in a subject comprising administering to the subject a therapeutically effective amount of the compound or salt of any one of claims 1-180 or the pharmaceutical composition of claim 181.

190. The method of claim 189, wherein the cancer is melanoma, multiple myeloma, prostate cancer, lung cancer, pancreatic cancer, squamous cell carcinoma, leukemia, lymphoma, a neuroendocrine tumor, bladder cancer, or colorectal cancer.

191. The method of claim 189, wherein the cancer is selected from the group consisting of prostate, lung, bladder, colorectal, and multiple myeloma.

192. The method of claim 189, wherein the cancer is non-small cell lung carcinoma, squamous cell carcinoma, leukemia, acute myelogenous leukemia, chronic myelogenous leukemia, lymphoma, NPM/ALK-transformed anaplastic large cell lymphoma, diffuse large B cell lymphoma, neuroendocrine tumors, breast cancer, mantle cell lymphoma, renal cell carcinoma, rhabdomyosarcoma, ovarian cancer, endometrial cancer, small cell carcinoma, adenocarcinoma, gastric carcinoma, hepatocellular carcinoma, pancreatic cancer, thyroid carcinoma, anaplastic large cell lymphoma, hemangioma, or head and neck cancer.

193. The method of claim 189, wherein the cancer is a solid tumor.

194. The method of claim 189, wherein the cancer is head and neck cancer, squamous cell carcinoma, gastric carcinoma, or pancreatic cancer.

195. A method for treating an autoimmune disease in a subject comprising

administering to the subject a therapeutically effective amount of the compound or salt of any one of claims 1-180 or the pharmaceutical composition of claim 181.

196. The method of claim 195, wherein the autoimmune disease is psoriasis, dermatitis, systemic scleroderma, sclerosis, Crohn’s disease, ulcerative colitis; respiratory distress syndrome, meningitis; encephalitis; uveitis; colitis; glomerulonephritis; eczema, asthma, chronic inflammation; atherosclerosis; leukocyte adhesion deficiency; rheumatoid arthritis; systemic lupus erythematosus (SLE); diabetes mellitus; multiple sclerosis;

Reynaud’s syndrome; autoimmune thyroiditis; allergic encephalomyelitis; Sjorgen’s syndrome; juvenile onset diabetes; tuberculosis, sarcoidosis, polymyositis, granulomatosis and vasculitis; pernicious anemia (Addison’s disease); diseases involving leukocyte diapedesis; central nervous system (CNS) inflammatory disorder; multiple organ injury syndrome; hemolytic anemia; myasthenia gravis; antigen-antibody complex mediated diseases; anti-glomerular basement membrane disease; antiphospholipid syndrome; allergic neuritis; Graves’ disease; Lambert-Eaton myasthenic syndrome; pemphigoid bullous;

pemphigus; autoimmune polyendocrinopathies; Reiter’s disease; stiff-man syndrome;

Behcet disease; giant cell arteritis; immune complex nephritis; IgA nephropathy; IgM polyneuropathies; immune thrombocytopenic purpura (ITP) or autoimmune

thrombocytopenia.

197. A method for the treatment of an immune-related disease in a subject comprising administering to the subject a therapeutically effective amount of the compound or salt of any one of claims 1-180 or the pharmaceutical composition of claim 181.

198. The method of claim 197, wherein the immune-related disease is rheumatoid arthritis, lupus, inflammatory bowel disease, multiple sclerosis, or Crohn’s disease.

199. A method for treating neurodegenerative disease in a subject comprising administering to the subject a therapeutically effective amount of the compound or salt of any one of claims 1-180 or the pharmaceutical composition of claim 181.

200. The method of claim 199, wherein the neurodegenerative disease is multiple sclerosis.

201. A method for treating an inflammatory disease in a subject comprising administering to the subject a therapeutically effective amount of the compound or salt of any one of claims 1-180 or the pharmaceutical composition of claim 181.

202. The method of claim 201 , wherein the inflammatory disease is bronchitis, conjunctivitis, myocarditis, pancreatitis, chronic cholecstitis, bronchiectasis, aortic valve stenosis, restenosis, psoriasis or arthritis.

Description:
THIAZOLE DERIVATIVES AS PROTEIN SECRETION INHIBITORS

BACKGROUND

Field of the Invention

[0001] The present disclosure relates to protein secretion inhibitors, including methods of making and using the same.

Incorporation by Reference of Material Submitted Electronically

[0002] This application contains, as a separate part of the disclosure, a sequence listing in computer-readable form (filename: 40061_Seqlisting.txt; 912 bytes; created: February 28, 2020) which is incorporated by reference in its entirety.

Description of Related Technology

[0003] Protein translocation into the endoplasmic reticulum (“ER”) constitutes the first step of protein secretion. ER protein import is essential in all eukaryotic cells and is particularly important in fast-growing tumor cells. Thus, the process of protein secretion can serve as a target both for potential cancer drugs and for bacterial virulence factors. See Kalies and Romisch, Traffic, 16(10):1027-1038 (2015).

[0004] Protein transport to the ER is initiated in the cytosol when N-terminal hydrophobic signal peptides protrude from the ribosome. Binding of signal recognition particle (“SRP”) to the signal sequence allows targeting of the ribosome-nascent chain-SRP complex to the ER membrane where contact of SRP with its receptor triggers handing over of the signal peptide to Sec61. Sec61 is an ER membrane protein translocator (aka translocon) that is doughnut-shaped with 3 major subunits (heterotrimeric). It includes a“plug,” which blocks transport into or out of the ER. The plug is displaced when the hydrophobic region of a nascent polypeptide interacts with the“seam” region of Sec61 , allowing translocation of the polypeptide into the ER lumen. In mammals, only short proteins (<160 amino acids) can enter the ER posttranslationally, and proteins smaller than 120 amino acids are obliged to use this pathway. Some of the translocation competence is maintained by the binding of calmodulin to the signal sequence. Upon arrival at the Sec61 channel, the signal peptide or signal anchor intercalates between transmembrane domains (“TMDs”) 2 and 7 of Sec61a, which form the lateral portion of the gate, allowing the channel to open for soluble secretory proteins. As the Sec61 channel consists of 10 TMDs (Sec61a) surrounded by a

hydrophobic clamp formed by Sec61y, channel opening is dependent on conformational changes that involve practically all TMDs.

[0005] Inhibition of protein transport across the ER membrane has the potential to treat or prevent diseases, such as the growth of cancer cells and inflammation. Known secretion inhibitors, which range from broad-spectrum to highly substrate-specific, can interfere with virtually any stage of this multistep process, and even with transport of endocytosed antigens into the cytosol for cross-presentation. These inhibitors interact with the signal peptide, chaperones, or the Sec61 channel to block substrate binding or to prevent the conformational changes needed for protein import into the ER. Examples of protein secretion inhibitors include, calmodulin inhibitors (e.g., E6 Berbamine and Ophiobolin A), Lanthanum, sterols, cyclodepsipeptides (e.g., HUN-7293, CAM741 , NFI028, Cotrainsin, Apratoxin A, Decatransin, Valinomycin), CADA, Mycolactone, Eeyarestatin I (“ESI”), and Exotoxin A. However, the above secretion inhibitors suffer from one or more of the following: lack selectivity for the Sec61 channel, challenging manufacture due to structural complexity, and molecular weight limited administration, bio-availability and distribution.

[0006] Thus, a need exits for new inhibitors of protein secretion.

SUMMARY

[0007] Provided herein are compounds having a structure of Formula (I), or a

pharmaceutically acceptable salt thereof:

wherein ring A is an aromatic or nonaromatic C3-10 carbocycle, or an aromatic or

nonaromatic 5-10 membered heterocycle having 1 or 2 ring heteroatoms selected from N, O, and S; one of Q and Q’ is U-B and the other is R 2 ; U is a bond, Ci- 6 alkylene, or w (-, erejn ^ indicates a double bond, a triple bond, or a fused or spiro cyclopropyl; B is Ci- 3 alkoxy,[0]o-i-Co- 3 alkylene-X, or NR N Ci-3alkylene-X; X is an aromatic or nonaromatic C 3-10 carbocycle, or an aromatic or nonaromatic 5-10 membered heterocycle having 1-4 ring heteroatoms selected from N, O, and S; L 2 is Co- 6 alkylene (e.g.,

C 1-6 alkylene) wherein— indicates a double bond, a triple bond, or a fused or spiro cyclopropyl; W is a bond, O, or C(0)N(R N ); D is C 6 -ioaryl or an aromatic or nonaromatic 5-10-membered heterocycle having 1-4 ring heteroatoms selected from N, O, and S; each R N independently is H or Ci-4alkyl; R 1 is H or Ci-3alkyl; and R 2 is H, Ci-3alkyl, or halo.

[0008] In various embodiments, R 1 is H. In various embodiments, R 1 is Ci-3alkyl. In some cases, R 1 is methyl or ethyl. [0009] In some embodiments, R 2 is H. In some embodiments, R 2 is Ci-3alkyl. In some embodiments, R 2 is halo. In some cases, R 2 is methyl. In some cases, R 2 is ethyl. In some cases, R 2 is n-propyl or isopropyl. In some cases, R 2 is Br. In some cases, R 2 is F. In some cases, R 2 is Cl.

[0010] In some embodiments, Q is U-B and Q’ is R 2 . In some embodiments, Q is R 2 and Q’ is U-B.

[0011] In various embodiments, U is a bond. In various embodiments, U is a Ci- 6 alkylene. In some cases, U is CH2, CH(CH 3 ), CH2CH2, or C(CH3)2. In various embodiments, cases, Co-2alkylene is CH2, CH(CH3), or

CH2CH2. In some cases, = indicates a double bond. In various cases, the double bond is tri- or tetra-substituted, and the 1 or 2 other substituents on the double bond are

independently selected from Ci-3alkyl and halo. In some cases,— indicates a triple bond.

In some cases,— indicates a fused cyclopropyl, e.g., , or a spiro cyclopropyl,

[0012] In some embodiments, B is Ci-3alkoxy. In some embodiments, B is O-X. In some embodiments, B is 0-Ci- 3 alkylene-X. In some embodiments, B is Ci-3alkylene-X. In some embodiments, B is X. In some embodiments, B is NHCi-3alkylene-X. In some

embodiments, B is N(CH3)Ci-3alkylene-X. In various embodiments, X is an aromatic C 6 - l ocarbocycle, or an aromatic or nonaromatic 5-10-membered heterocycle. In some cases, X is selected from phenyl, pyridyl, indolyl, tetrahydropyranyl, piperidinyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, or piperazinyl, and X is optionally substituted with 1-3 substituents independently selected from Ci-3alkyl, Ci-3alkoxy, halo, Ci-3 haloalkyl, C1 -3 haloalkoxy, C(0)Ci- 3 alkyl, and SC>2Ci-3alkyl.

[0013] In some embodiments, U-B is selected from the group consisting of:

[0014] In some embodiments, ring A is a 5-6 membered heterocycle having 1 or 2 ring heteroatoms selected from N, O, and S. In some cases, the compound has a structure of Formula (IA):

wherein ring A has 0 or 1 additional ring heteroatoms selected from N, O, and S, and R 3 is H, Ci-3alkyl, Ci-3hydroxyalkyl, Ci-3haloalkyl, halo, or -C(0)N(R N ) 2 . In some cases, ring A is an aromatic or nonaromatic C3-10 carbocycle. In some cases, the ring A-L 2 moiety is

[0015] In some embodiments, L 2 is Co- 6 alkylene. In some embodiments, L 2 is Ci-

6 alkylene. In some embodiments, L 2 is In some cases,— indicates a double bond. In some cases, = indicates a triple bond. In some cases,— indicates a fused cyclopropyl, e.g., , or a spiro cyclopropyl, e.g., .

[0016] In some embodiments, W is a bond. In some embodiments, W is O. In some embodiments, W is C(0)N(R N ). In various cases, W is C(0)NH. In various cases, W is C(0)N(Ci- 4 alkyl). In various cases, W is C(0)N(Me).

[0017] In some embodiments, D is Ce-ioaryl. In some embodiments, D is a nonaromatic 5- 10 membered heterocycle having 1-4 ring heteroatoms selected from N, O, and S. In some embodiments, D is an aromatic 5-10-membered heterocycle having 1-4 ring heteroatoms selected from N, O, and S. In some cases, D comprises pyridyl optionally substituted with 1- 3 substituents independently selected from Ci-3alkyl, Ci-3alkoxy, halo, Ci-3 haloalkyl, C1 -3 haloalkoxy, N(R N ) 2 , Cs-ecycloalkyl, N0 2 , N(R N )C(0)Ci. 3 alkyl, C(0)Ci- 3 alkyl, N0 2 , CN, SOiC^ 3alkyl, O , NHCi-3alkylene-aryl, OCi-3alkylene-aryl, Ci-3alkylene-aryl, and 5-10 membered heterocycle having 1-3 ring heteroatoms selected from N, O, and S, and each R N is independently H or Ci-4alkyl.

[0018] In some embodiments, L 2 -W-D is selected from the group consisting of:

[0019] Also provided herein are compounds having a structure of Formula (II), or a pharmaceutically acceptable salt thereof:

wherein one of Q and Q’ is the atoms to which they are attached join together to form an aromatic or nonaromatic 5 or 6 membered carbocycle or a 5 or 6 membered heterocycle having 1 or 2 ring heteroatoms selected from N, O, and S;

U is a bond, Ci- 6 alkylene, or wherein = indicates a double bond, a triple bond, or a fused or spiro cyclopropyl; B is Ci-e alkyl, C1 -3 haloalkyl, C1 -3 hydroxyalkyl, C1 -3 haloalkoxy, Ci-3alkoxy, [0]o-i-Co- 3 alkylene-X or NR N Ci-3alkylene-X, X is an aromatic or nonaromatic C3-10 carbocycle, or an aromatic or nonaromatic 5-10 membered heterocycle having 1-4 ring heteroatoms selected from N, O, and S; L 2 is Ci- 6 alkylene or \ w|ierejn ___ indicates a double bond, a triple bond, or a fused or spiro cyclopropyl; W is a bond, O, or C(0)N(R N ); D comprises pyridyl or quinolinyl optionally substituted with 1-3 substituents independently selected from Ci-3alkyl, Ci-3alkoxy, halo, Ci-3 haloalkyl, C^ haloalkoxy, N(R N ) 2 , Cs-ecycloalkyl, N0 2 , C(0)N(R N ) 2 , IM(R N )C(0)Ci- 3 alkyl, C(0)Ci- 3 alkyl, N0 2 , CN, S0 2 Ci- 3 alkyl, O , NHCi-3alkylene-aryl, OCi-3alkylene-aryl, Ci- 3alkylene-aryl, and 5-10 membered heterocycle having 1-3 ring heteroatoms selected from N, O, and S; each R N is independently H or Ci-4alkyl; R 1 is H or Ci-3alkyl; R 2 is H, Ci-3alkyl, or halo; and R 3 is H, Ci-3alkyl, Ci-3hydroxyalkyl, Ci-3haloalkyl, halo, or -C(0)N(R N ) 2 .

[0020] In some embodiments, R 1 is H. In some embodiments, R 1 is Ci-3alkyl. In various cases, R 1 is methyl or ethyl.

[0021] In some embodiments, R 2 is H. In some embodiments, R 2 is Ci-3alkyl. In some embodiments, R 2 is halo. In some cases, R 2 is methyl. In some cases, R 2 is ethyl. In some cases, R 2 is n-propyl or isopropyl. In some cases, R 2 is Br. In some cases, R 2 is F. In some cases, R 2 is Cl.

[0022] In some embodiments, R 3 is H. In some embodiments, R 3 is Ci-3alkyl. In some embodiments, R 3 is halo. In some embodiments, R 3 is Ci-3hydroxyalkyl. In some

embodiments, -C(0)N(R N ) 2 . In some embodiments, R 3 is Ci-3haloalkyl. In various cases, R 3 is methyl. In various cases, R 3 is Cl. In various cases, R 3 is -CH2OH. In various cases, R 3 is -C(0)NH 2 . In various cases, R 3 is -C(0)N(Me) 2 .

[0023] In some embodiments, Q is U-B and Q’ is R 2 . In some embodiments, Q is R 2 and Q’ is U-B. In some embodiments, Q and Q’ and the atoms to which they are attached join together to form an aromatic or nonaromatic 5 or 6 membered carbocycle or a 5 or 6 membered heterocycle having 1 or 2 ring heteroatoms selected from N, O, and S.

[0024] In various embodiments, U is a bond. In various embodiments, U is a Ci- 6 alkylene. In some cases, U is CH2, CH(CH 3 ), CH2CH2, or C(CH3)2. In various

embodiments, U is | n varjOUS cases ^ indicates a double bond. In some cases, the double bond is tri- or tetra-substituted, and the 1 or 2 other substituents on the double bond are independently selected from Ci-3alkyl and halo. In various cases,— indicates a triple bond. In various cases,— indicates a fused cyclopropyl, e.g., or spiro cyclopropyl, e.g.,

[0025] In some embodiments, B is Ci- 6 alkyl. In some embodiments, B is C1-3 haloalkyl.

In some embodiments, B is C1-3 hydroxyalkyl. In some embodiments, B is C1-3 haloalkoxy.

In some embodiments, B is Ci-3alkoxy. In some cases, B is methyl, ethyl, n-propyl, isopropyl, n-butyl, sec-butyl, tert-butyl, n-pentyl, or n-hexyl. In some cases, B is -CF3 or - CF2CH3. In some cases, B is -CH2CH2OH. In some cases, B is -OCH2CF3. In some embodiments, B is O-X. In some embodiments, B is 0-Ci- 3 alkylene-X. In some

embodiments, B is Ci-3alkylene-X. In some embodiments, B is X. In some embodiments, B is NHCi-3alkylene-X. In some embodiments, B is N(CH3)Ci-3alkylene-X. In various embodiments, X is an aromatic C 6 -iocarbocycle, or an aromatic or nonaromatic 5-10- membered heterocycle. In some cases, X is selected from phenyl, pyridyl, indolyl, tetrahydropyranyl, piperidinyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, or piperazinyl, and X is optionally substituted with 1-3 substituents independently selected from Ci-3alkyl, Ci-3alkoxy, halo, C1 -3 haloalkyl, C1 -3 haloalkoxy, C(0)Ci- 3 alkyl, and SC>2Ci-3alkyl.

, . In some cases, U-B is selected from the group [0027] In some embodiments, the pyrrole ring-L 2 moiety is selected from the group

[0028] In some embodiments, L 2 is Ci- 6 alkylene. In some embodiments, L 2 is some cases, L 2 is . in some cases, L 2 is I G In various cases,— indicates a double bond. In various cases, = indicates a triple bond. In various cases,— indicates a fused cyclopropyl, e.g., , or a spiro cyclopropyl, e.g., .

[0029] In some embodiments, W is a bond. In some embodiments, W is O. In some embodiments, W is C(0)N(R N ). In various cases, W is C(0)NH 2. In various cases, W is C(0)N(Ci-4alkyl)2. In some cases, W is C(0)N(Me) 2 .

[0030] In some embodiments, L 2 -W-D is selected from the group consisting of:

,

[0031] Also provided herein are compounds having a structure of Formula (III), or a pharmaceutically acceptable salt thereof:

wherein L 1 is a bond,

indicates a double bond, a triple bond, or a fused cyclopropyl; B is Co-3alkylene-X; X is an aromatic or nonaromatic C4-iocarbocycle, or an aromatic or nonaromatic 4-10 membered heterocycle having 1-3 ring heteroatoms selected from N, O, and S; R 2 is H or Ci-3alkyl; L 2 is Co-3alkylene; m is 0 to 2; and each R 4 independently is Ci-3alkyl, C2-3alkynyl, Ci-3haloalkyl, Ci-3alkoxy, halo, or NHCi-3alkylene-aryl; or a pharmaceutically acceptable salt thereof.

[0032] In various embodiments, L 1 is a bond. In various embodiments, L 1 is Ci- 6 alkylene. In some cases, L 1 is CH 2 , CH 2 CH 2 , C(CH 3 ) 2 , C(CH 3 ) 2 CH 2 , or C(CH 3 ) 2 CH 2 CH 2 . In various embodiments, U is I - c o- 3alk y |ene c 0-2 aikyiene— j | n some cases, L 1 is | n some cases, L 1 is I ~ Co^alkylene j | n some cases |_1 js embodiments,— indicates a double bond. In some cases, the double bond is further substituted with Ci-3alkyl. In various embodiments,— indicates a triple bond. In various embodiments, = indicates a fused cyclopropyl, e.g.,

[0033] In various embodiments, B is Ci-3alkylene-X. In various embodiments, B is X. In various embodiments, X is pyrrolidinyl, cyclobutyl, cyclopentyl, cyclohexyl, cyclohexenyl, phenyl, piperidinyl, pyridinyl, piperazinyl, tetrahydrofuranyl, furanyl, tetrahydropyranyl, quinolinyl, morpholinyl, pyrrolidonyl, pyrimidinyl, pyridazinyl, indenyl, dihydroindenyl, dihydrobenzofuranyl, chromanyl, isochromanyl, dihydroisoquinolinyl, or indolyl.

[0034] In various embodiments, X is substituted with 1-3 G; each G independently is selected from the group consisting of halo, OH, =0, CN, N0 2 , N(R N ) 2 , N(R N )C(0)Ci- 3 alkyl, Ci-3alkyl, Ci-3alkoxy, C1 -3 haloalkyl, C1 -3 haloalkoxy, C(0)Ci- 3 alkyl, S(0 2 )-Z, C(0)-Z,

C(0)N(R n ) 2 , silyl ether, and [0]o-i-Co- 3 alkylene-Z; each R N independently is H or Ci-4alkyl; Z is aromatic or nonaromatic C3-iocarbocycle, or aromatic or nonaromatic 4-10 membered heterocycle having 1-3 heteroatoms selected from the group consisting of N, O, and S; Z is optionally substituted with 1-3 E; and, each E independently is selected from Ci-3alkyl, Ci- 3alkoxy, =0, Ci-3haloalkoxy, CN, and halo.

X

[0035] In various embodiments, U-B is selected from the group consisting of ; X is aromatic or nonaromatic C 4-7 carbocycle, or an aromatic or nonaromatic 4-9 membered heterocycle having 1 ring heteroatom; X is optionally substituted with 1-3 G; each G independently is selected from the group consisting of halo, OH, =0, CN, NO2, N(R n ) 2 , N(R N )C(0)Ci- 3 alkyl, Ci-3alkyl, Ci-3alkoxy, Ci-3 haloalkyl, C1 -3 haloalkoxy, C(0)Ci- salkyl, S(0 2 )-Z, C(0)-Z, C(0)N(R N ) 2 , silyl ether, and [O] 0 -i-C 0 -3alkylene-Z; each R N independently is H or Ci-4alkyl; Z is aromatic or nonaromatic C3-iocarbocycle, or aromatic or nonaromatic 4-10 membered heterocycle having 1-3 heteroatoms selected from the group consisting of N, O, and S; Z is optionally substituted with 1-3 E; and, each E independently is selected from Ci-3alkyl, Ci-3alkoxy, =0, Ci-3haloalkoxy, CN, and halo. [0036] In some cases, U-B is selected from the group consisting

[0037] In some cases, L 1 -B is selected from the group consisting

[0038] In various embodiments, U-B is selected from the group consisting of

[0039] In various embodiments, R 2 is H. In various embodiments, R 2 is Ci-3alkyl. In some cases, R 2 is methyl.

[0040] In various embodiments, L 2 is Coalkylene. In various embodiments, L 2 is

Cialkylene. In various embodiments, L 2 is C2alkylene. In various embodiments, L 2 is C3alkylene.

[0041] In various embodiments, m is 0. In various embodiments, m is 1 or 2.

[0042] In various embodiments, R 4 is Ci-3alkyl. In some cases, R 4 is methyl or ethyl. In various embodiments, R 4 is halo. In some cases, R 4 is F. In some cases, R 4 is Cl. In various embodiments, R 4 is C2-3alkynyl. In some cases, R 4 is C2alkynyl. In various embodiments, R 4 is Ci-3haloalkyl. In some cases R 4 is CF3. In various embodiments, R 4 is Ci-3alkoxy. In some cases, R 4 is methoxy. In various embodiments, R 4 is NHCi-3alkylene- aryl. In some cases, R 4 is NH-CFh-phenyl.

[0043] In various embodiments, m is 2, and one R 4 is halo, and the other R 4 is halo or methyl. [0044] In various embodiments, the compound or salt has a structure of Formula (IIIA):

pharmaceutically salt thereof. In some embodiments, the compound or salt is selected from A1-A210. In some embodiments, the compound or salt is selected from A211-A403.

Further provided are the compounds listed in Table B, or a pharmaceutically acceptable salt thereof. In some embodiments, the compound or salt is selected from B1-B29. In some cases, the compound or salt is selected from the group consisting of

the group consisting

[0049] Also provided are pharmaceutical compositions comprising the compound or salt described herein and a pharmaceutically acceptable carrier.

[0050] Further provided are methods of inhibiting protein secretion in a cell comprising contacting the cell with the compound, salt, or pharmaceutical composition described herein in an amount effective to inhibit secretion. In some embodiments, the protein is a checkpoint protein. In some embodiments, the protein is a cell-surface protein, endoplasmic reticulum associated protein, or secreted protein involved in regulation of anti-tumor immune response. In various cases, the protein is at least one of PD-1 , PD-L1 , TIM-1 , LAG-3, CTLA4, BTLA, OX-40, B7H1 , B7H4, CD137, CD47, CD96, CD73, CD40, VISTA, TIGIT, LAIR1 , CD160, 2B4, TGFF^ and combinations thereof. In some cases, the protein is selected from the group consisting of HER3, TNFa, IL2, and PD1. In some embodiments, the contacting comprises administering the compound or the composition to a subject in need thereof. [0051] The disclosure also provides methods for treating inflammation in a subject comprising administering to the subject a therapeutically effective amount of the compound, salt, or pharmaceutical composition described herein.

[0052] The disclosure further provides methods for treating cancer in a subject comprising administering to the subject a therapeutically effective amount of the compound, salt, or pharmaceutical composition described herein. In some embodiments, the cancer is melanoma, multiple myeloma, prostate cancer, lung cancer, pancreatic cancer, squamous cell carcinoma, leukemia, lymphoma, a neuroendocrine tumor, bladder cancer, or colorectal cancer. In some cases, the cancer is selected from the group consisting of prostate, lung, bladder, colorectal, and multiple myeloma. In some cases, the cancer is non-small cell lung carcinoma, squamous cell carcinoma, leukemia, acute myelogenous leukemia, chronic myelogenous leukemia, lymphoma, NPM/ALK-transformed anaplastic large cell lymphoma, diffuse large B cell lymphoma, neuroendocrine tumors, breast cancer, mantle cell lymphoma, renal cell carcinoma, rhabdomyosarcoma, ovarian cancer, endometrial cancer, small cell carcinoma, adenocarcinoma, gastric carcinoma, hepatocellular carcinoma, pancreatic cancer, thyroid carcinoma, anaplastic large cell lymphoma, hemangioma, or head and neck cancer. In various cases, the cancer is a solid tumor. In various cases, the cancer is head and neck cancer, squamous cell carcinoma, gastric carcinoma, or pancreatic cancer.

[0053] Further provided are methods for treating an autoimmune disease in a subject comprising administering to the subject a therapeutically effective amount of the compound, salt, or pharmaceutical composition described herein. In some embodiments, the

autoimmune disease is psoriasis, dermatitis, systemic scleroderma, sclerosis, Crohn’s disease, ulcerative colitis; respiratory distress syndrome, meningitis; encephalitis; uveitis; colitis; glomerulonephritis; eczema, asthma, chronic inflammation; atherosclerosis; leukocyte adhesion deficiency; rheumatoid arthritis; systemic lupus erythematosus (SLE); diabetes mellitus; multiple sclerosis; Reynaud’s syndrome; autoimmune thyroiditis; allergic

encephalomyelitis; Sjorgen’s syndrome; juvenile onset diabetes; tuberculosis, sarcoidosis, polymyositis, granulomatosis and vasculitis; pernicious anemia (Addison’s disease);

diseases involving leukocyte diapedesis; central nervous system (CNS) inflammatory disorder; multiple organ injury syndrome; hemolytic anemia; myasthenia gravis; antigen- antibody complex mediated diseases; anti-glomerular basement membrane disease;

antiphospholipid syndrome; allergic neuritis; Graves’ disease; Lambert-Eaton myasthenic syndrome; pemphigoid bullous; pemphigus; autoimmune polyendocrinopathies; Reiter’s disease; stiff-man syndrome; Behcet disease; giant cell arteritis; immune complex nephritis; IgA nephropathy; IgM polyneuropathies; immune thrombocytopenic purpura (ITP) or autoimmune thrombocytopenia. [0054] The disclosure also provides methods for the treatment of an immune-related disease in a subject comprising administering to the subject a therapeutically effective amount of the compound, salt, or pharmaceutical composition described herein. In some embodiments, the immune-related disease is rheumatoid arthritis, lupus, inflammatory bowel disease, multiple sclerosis, or Crohn’s disease.

[0055] Further provided are methods for treating neurodegenerative disease in a subject comprising administering to the subject a therapeutically effective amount of the compound, salt, or pharmaceutical composition described herein. In some cases, the

neurodegenerative disease is multiple sclerosis.

[0056] Also provided are methods for treating an inflammatory disease in a subject comprising administering to the subject a therapeutically effective amount of the compound, salt, or pharmaceutical composition described herein. In some embodiments, the inflammatory disease is bronchitis, conjunctivitis, myocarditis, pancreatitis, chronic cholecstitis, bronchiectasis, aortic valve stenosis, restenosis, psoriasis or arthritis.

[0057] Further aspects and advantages will be apparent to those of ordinary skill in the art from a review of the following detailed description. The description hereafter includes specific embodiments with the understanding that the disclosure is illustrative, and is not intended to limit the disclosure to the specific embodiments described herein.

DETAILED DESCRIPTION

[0058] Provided herein are compounds that inhibit protein secretion. The compounds described herein can be used to treat or prevent diseases associated with excessive protein secretion, such as inflammation and cancer, improving the quality of life for afflicted individuals.

[0059] Compounds disclosed herein can have a structure of Formula (I), (II), or (III):

[0060] In some cases, the compound has a structure of Formula (IA):

[0061] In some cases, the compound has a structure of Formula (IIIA):

[0062] Without being bound by any particular theory, the compounds described herein inhibit protein secretion by binding to and disabling components of the translocon, including but not limited to Sec61 , and in some cases, disrupting in a sequence specific fashion interactions between the nascent signaling sequence of translated proteins with components of the translocon including but not limited to Sec61.

[0063] The compounds described herein can advantageously inhibit the secretion of a protein of interest with an IC50 of up to 5 mM, or up to 3mM, or up to 1 mM. In various cases, the compounds disclosed herein can inhibit the secretion of TNFa with an IC50 of up to 5 mM, or up to 3mM, or up to 1 mM. In various cases, the compounds disclosed herein can inhibit the secretion of Her3 with an IC50 of up to 5 mM, or up to 3mM, or up to 1 mM. In some cases, the compounds disclosed herein can inhibit the secretion of IL2 with an IC50 of up to 5 mM, or up to 3mM, or up to 1 mM. In various cases, the compounds disclosed herein can inhibit the secretion of PD-1 with an IC50 of up to 5 mM, or up to 3mM, or up to 1 mM.

Chemical Definitions

[0064] As used herein, the term“alkyl” refers to straight chained and branched saturated hydrocarbon groups containing one to thirty carbon atoms, for example, one to twenty carbon atoms, or one to ten carbon atoms. The term C n means the alkyl group has“n” carbon atoms. For example, C4alkyl refers to an alkyl group that has 4 carbon atoms. Ci- 6 alkyl refers to an alkyl group having a number of carbon atoms encompassing the entire range (i.e., 1 to 6 carbon atoms), as well as all subgroups (e.g., 1-5, 2-5, 1-4, 2-5, 1 , 2, 3, 4, 5, and 6 carbon atoms). Nonlimiting examples of alkyl groups include, methyl, ethyl, n- propyl, isopropyl, n-butyl, sec-butyl (2-methylpropyl), and t-butyl (1 ,1-dimethylethyl). Unless otherwise indicated, an alkyl group can be an unsubstituted alkyl group or a substituted alkyl group.

[0065] As used herein, the term "alkylene" refers to a bivalent saturated aliphatic radical. The term C n means the alkylene group has "n" carbon atoms. For example, Ci- 6 alkylene refers to an alkylene group having a number of carbon atoms encompassing the entire range, as well as all subgroups, as previously described for "alkyl" groups.

[0066] As used herein, the term“alkene” or“alkenyl” is defined identically as“alkyl” except for containing at least one carbon-carbon double bond, and having two to thirty carbon atoms, for example, two to twenty carbon atoms, or two to ten carbon atoms. The term C n means the alkenyl group has“n” carbon atoms. For example, C4alkenyl refers to an alkenyl group that has 4 carbon atoms. C^alkenyl refers to an alkenyl group having a number of carbon atoms encompassing the entire range (i.e., 2 to 7 carbon atoms), as well as all subgroups (e.g., 2-6, 2-5, 3-6, 2, 3, 4, 5, 6, and 7 carbon atoms). Specifically contemplated alkenyl groups include ethenyl, 1-propenyl, 2-propenyl, and butenyl. Unless otherwise indicated, an alkenyl group can be an unsubstituted alkenyl group or a substituted alkenyl group. Unless otherwise indicated, an alkenyl group can be a cis-alkenyl or trans-alkenyl.

[0067] As used herein, the term“alkyne” or“alkynyl” is defined identically as“alkyl” except for containing at least one carbon-carbon triple bond, and having two to thirty carbon atoms, for example, two to twenty carbon atoms, or two to ten carbon atoms. The term C n means the alkynyl group has“n” carbon atoms. For example, C4alkynyl refers to an alkynyl group that has 4 carbon atoms. C^alkynyl refers to an alkynyl group having a number of carbon atoms encompassing the entire range (i.e., 2 to 7 carbon atoms), as well as all subgroups (e.g., 2-6, 2-5, 3-6, 2, 3, 4, 5, 6, and 7 carbon atoms). Specifically contemplated alkynyl groups include ethynyl, 1-propynyl, 2-propynyl, and butynyl. Unless otherwise indicated, an alkynyl group can be an unsubstituted alkynyl group or a substituted alkynyl group.

[0068] As used herein, the term“carbocycle” refers to an aromatic or nonaromatic ring in which each atom of the ring is carbon. A carbocycle can include, for example, from three to ten carbon atoms, four to eight carbon atoms, or five to six carbon atoms. As used herein, the term“carbocycle” also includes polycyclic ring systems having two or more cyclic rings in which two or more carbons are common to two adjoining rings wherein at least one of the rings is carbocyclic, e.g., the other cyclic rings can be cycloalkyls, cycloalkenyls, aryls, heteroaryls, and/or heterocycles.

[0069] As used herein, the term“cycloalkyl” specifically refers to a non-aromatic carbocycle. The term C n means the cycloalkyl group has“n” carbon atoms. For example,

C5 cycloalkyl refers to a cycloalkyl group that has 5 carbon atoms in the ring. C5-8 cycloalkyl refers to cycloalkyl groups having a number of carbon atoms encompassing the entire range (i.e., 5 to 10 carbon atoms), as well as all subgroups (e.g., 5-10, 5-9, 5-8, 5-6, 6-8, 7-8, 5-7,

5, 6, 7, 8, 9 and 10 carbon atoms). Nonlimiting examples of cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl. Unless otherwise indicated, a cycloalkyl group can be an unsubstituted cycloalkyl group or a substituted cycloalkyl group.

[0070] As used herein, the term“aryl” refers to an aromatic carbocycle, and can be monocyclic or polycyclic (e.g., fused bicyclic and fused tricyclic) carbocyclic aromatic ring systems. Examples of aryl groups include, but are not limited to, phenyl, naphthyl, tetrahydronaphthyl, phenanthrenyl, biphenylenyl, indanyl, indenyl, anthracenyl, fluorenyl, tetralinyl. Unless otherwise indicated, an aryl group can be an unsubstituted aryl group or a substituted aryl group.

[0071] As used herein, the term“heterocycle” is defined similarly as carbocycle, except the ring contains one to four heteroatoms independently selected from oxygen, nitrogen, and sulfur. For example, a heterocycle can be a 5-10 membered ring having 1 or 2 heteroatoms selected from N, O, and S. As another example, a heterocycle can be a 5-6 membered ring having 1 or 2 ring heteroatoms selected from N, O, and S. Nonlimiting examples of heterocycle groups include piperdine, tetrahydrofuran, tetrahydropyran, dihydrofuran, morpholine, oxazepaneyl, thiazole, pyrrole, and pyridine. Carbocyclic and heterocyclic groups can be saturated or partially unsaturated ring systems optionally substituted with, for example, one to three groups, independently selected alkyl, alkoxy, alkyleneOH, C(0)NH 2 , NH2, oxo (=0), aryl, haloalkyl, haloalkoxy, C(0)-alkyl, SC>2alkyl, halo, OH, NHCi-3alkylene- aryl, OCi-3alkylene-aryl, Ci-3alkylene-aryl, and C3-6heterocycloalkyl having 1-3 heteroatoms selected from N, O, and S. Heterocyclic groups optionally can be further N-substituted as described herein.

[0072] As used herein, the term“heteroaryl” refers to an aromatic heterocycle, and can be monocyclic or polycyclic (e.g., fused bicyclic and fused tricyclic) aromatic ring systems, wherein one to four-ring atoms are selected from oxygen, nitrogen, or sulfur, and the remaining ring atoms are carbon, said ring system being joined to the remainder of the molecule by any of the ring atoms. Nonlimiting examples of heteroaryl groups include, but are not limited to, pyridyl, pyridazinyl, pyrazinyl, pyrimidinyl, pyrrolyl, pyrazolyl, imidazolyl, thiazolyl, tetrazolyl, oxazolyl, isooxazolyl, thiadiazolyl, oxadiazolyl, furanyl, thienyl, quinolinyl, isoquinolinyl, benzoxazolyl, benzimidazolyl, benzofuranyl, benzothiazolyl, triazinyl, triazolyl, purinyl, pyrazinyl, purinyl, indolinyl, phthalzinyl, indazolyl, quinolinyl, isoquinolinyl, cinnolinyl, quinazolinyl, naphthyridinyl, pyridopyridinyl, indolyl, 3H-indolyl, pteridinyl, and quinooxalinyl. Unless otherwise indicated, a heteroaryl group can be an unsubstituted heteroaryl group or a substituted heteroaryl group.

[0073] As used herein, the term“hydroxy” or“hydroxyl” as used herein refers to an“-OH” group. Accordingly, a“hydroxyalkyl” refers to an alkyl group substituted with one or more - OH groups.

[0074] As used herein, the term“alkoxy” or“alkoxyl” refers to a“— O-alkyl” group.

[0075] As used herein, the term "halo" is defined as fluoro, chloro, bromo, and iodo.

Accordingly, a“haloalkyl” refers to an alkyl group substituted with one or more halo atoms.

A“haloalkoxy” refers to an alkoxy group that is substituted with one or more halo atoms.

[0076] A“substituted” functional group (e.g., a substituted alkyl, cycloalkyl, aryl, or heteroaryl) is a functional group having at least one hydrogen radical that is substituted with a non-hydrogen radical (i.e. , a substituent). Examples of non-hydrogen radicals (or substituents) include, but are not limited to, alkyl, cycloalkyl, alkenyl, cycloalkenyl, alkynyl, ether, aryl, O-alkylene aryl, N-alkylene aryl, alkylene aryl, heteroaryl, heterocycloalkyl, hydroxy, hydroxyalkyl, haloalkoxy, amido, oxy (or oxo), alkoxy, ester, thioester, acyl, carboxyl, cyano, nitro, amino, sulfhydryl, and halo. When a substituted alkyl group includes more than one non-hydrogen radical, the substituents can be bound to the same carbon or two or more different carbon atoms.

Protein Secretion Inhibitors - Formula (I)

[0077] Provided herein are compounds that have a structure of Formula (I), or a pharmaceutically acceptable salt thereof:

wherein ring A is an aromatic or nonaromatic C3-10 carbocycle, or an aromatic or

nonaromatic 5-10 membered heterocycle having 1 or 2 ring heteroatoms selected from N, O, and S; one of Q and Q’ is U-B and the other is R 2 ; L 1 is a bond, Ci- 6 alkylene, or , wherein = indicates a double bond, a triple bond, or a fused or spiro cyclopropyl; B is Ci-3alkoxy, [0]o-i-Co- 3 alkylene-X, or NR N Ci-3alkylene-X; X is an aromatic or nonaromatic C 3-10 carbocycle, or an aromatic or nonaromatic 5-10 membered heterocycle, having 1-4 ring heteroatoms selected from N, O, and S; L 2 is Co- 6 alkylene (e.g.,

Ci- 6 alkylene) or , wherein = indicates a double bond, a triple bond, or a fused or spiro cyclopropyl; W is a bond, O, or C(0)N(R N ); D is C 6 -ioaryl or an aromatic or nonaromatic 5-10-membered heterocycle having 1-4 ring heteroatoms selected from N, O, and S; each R N independently is H or Ci-3alkyl; R 1 is H or Ci-3alkyl; and R 2 is H, Ci-3alkyl, or halo.

[0078] In some embodiments, Q is U-B and Q’ is R 2 . In some embodiments, Q is R 2 and Q’ is U-B.

[0079] Ring A is an aromatic or nonaromatic C3-10 carbocycle, or an aromatic or nonaromatic 5-10 membered heterocycle having 1 or 2 ring heteroatoms selected from N, O, and S. Ring A can be an aromatic carbocycle, such as a phenyl or naphthyl. Ring A can be a non-aromatic carbocycle, such as a cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl.

Ring A can be substituted with 1 or 2 R 3 groups. Each R 3 can independently be H, Ci-3alkyl, Ci-3hydroxyalkyl, Ci-3haloalkyl, halo, oxo (=0) or -C(0)N(R N ) 2 .

[0080] In some embodiments, ring A is an aromatic or nonaromatic 5-10 membered heterocycle having 1 or 2 ring heteroatoms selected from N, O, and S. For example, ring A can be a 5-6 membered heterocycle having 1 or 2 ring heteroatoms selected from N, O, and S. Some examples of ring A include, but are not limited to, pyrrolidinyl, pyrrolyl, indolyl, imidozolyl, and pyrazolyl. In various embodiments, ring A includes a ring nitrogen. In various cases, the ring nitrogen is bonded to L 2 . In some cases, the ring nitrogen is not bonded to L 2 and is unsubstituted. In some cases, the ring nitrogen is not bonded to L 2 and is substituted, e.g., with an R 3 group.

[0081] In some embodiments, the compound has a structure of Formula (IA):

wherein ring A has 0 or 1 additional ring heteroatoms selected from N, O, and S. In some embodiments, ring A is an aromatic or nonaromatic C3-10 carbocycle. For example, in some cases, ring A is cyclopentyl, cyclohexyl, or phenyl, and can be substituted with 1 or 2 R 3 groups.

[0082] Specifically contemplated ring A-L 2 moieties include

[0083] As disclosed herein, R 1 is H or Ci-3alkyl. In some embodiments, R 1 is H. In some embodiments, R 1 is Ci-3alkyl. Examples of contemplated R 1 groups include, but are not limited to, H, methyl, ethyl, n-propyl, and isopropyl. In various cases, R 1 is H or methyl.

[0084] As disclosed herein, R 2 is H, Ci-3alkyl, or halo. In some embodiments, R 2 is H. In some embodiments, R 2 is Ci-3alkyl. Examples of contemplated R 2 groups include, but are not limited to, H, methyl, ethyl, n-propyl, isopropyl, Br, Cl, and F. In various cases, R 2 is methyl. In various cases, R 2 is ethyl. In various cases, R 2 is n-propyl or isopropyl. In some embodiments, R 2 is halo. In some cases, R 2 is Br. In some cases, R 2 is F. In some cases, R 2 is Cl.

[0085] As disclosed herein, U is a bond, Ci- 6 alkylene, wherein

— indicates a double bond, a triple bond, or a fused ( ' * ) or spiro ( ) cyclopropyl.

[0086] In some embodiments, U is a bond. In some embodiments, U is a Ci- 6 alkylene. In various cases, U is CH 2 , CH(CH 3 ), CH 2 CH 2 , or C(CH 3 ).

[0087] In some embodiments, U is | n varjOUS case s when U is — indicates a double bond. The double bond can be 1 ,1- . . ), or 1 ,2-substituted (e.g. ). In some cases, the double bond is tri- or tetra-substituted. The double bond can be substituted with substitutions independently selected from Ci- 3 alkyl and halo. For example, the double bond can be substituted with one or two groups independently selected from methyl, ethyl, n- propyl, isopropyl, F, Br, and Cl. The double bond orientation can be cis or trans, or when tri or tetra-substituted, E- or Z-. In some cases, the double bond is cis.

[0088] In various cases when U is = indicates a triple bond.

[0089] In various cases when U is ,— indicates a fused cyclopropyl, e.g., . In some cases,— indicates a spiro cyclopropyl, e.g., . The fused or spiro cyclopropyl can be further substituted with one to four substituents. Examples of suitable substituents include, but are not limited to Ci- 3 alkyl (e.g., methyl, ethyl, n-propyl, isopropyl), N(R N )C(0)Ci- 3 alkyl, and N(R N ) 2 , where each R N is independently H or Ci-4alkyl.

[0090] In some cases when

CH 2 CH 2 . In some cases, Co-2alkylene is null (Co).

[0091] As disclosed herein, B is Ci- 3 alkoxy, [0]o-rCo- 3 alkylene-X, or NR N Ci- 3 alkylene-X. X is an aromatic or nonaromatic C 3 -io carbocycle, or an aromatic or nonaromatic 5-10 membered heterocycle, having 1-4 ring heteroatoms selected from N, O, and S. R N is H or Ci-4alkyl, and in some cases, is H or methyl.

[0092] In various embodiments, B is Ci-3alkoxy. Specifically contemplated B include methoxy, ethoxy, propoxy, and isopropoxy.

[0093] In various embodiments, B is [0]o-i-Co- 3 alkylene-X. For example, in some cases, B is O-X. In some cases, B is 0-Ci- 3 alkylene-X. In some cases, B is Ci-3alkylene-X. In some cases, B is X. In some cases, B is NR N Ci-3alkylene-X, such as NHCi-3alkylene-X or

N(CH 3 )Ci- 3 alkylene-X.

[0094] X is an aromatic or nonaromatic C3-10 carbocycle, or an aromatic or nonaromatic 5- 10 membered heterocycle, having 1-4 ring heteroatoms selected from N, O, and S. X can be optionally substituted with 1-3 substituents. Contemplated substituents of X include Ci- 3alkyl, Ci-3alkoxy, halo, C1 -3 haloalkyl, Ci-3 haloalkoxy, C(0)Ci- 3 alkyl, and SC>2Ci-3alkyl.

[0095] In embodiments, X is an aromatic Ce-io carbocycle, such as phenyl or naphthyl. In embodiments, X is an aromatic or nonaromatic 5-10 membered heterocycle. In some cases, X is a 5-10 membered nonaromatic heterocycle, such as morpholinyl, piperidinyl,

tetrahydropyranyl, or piperazinyl. In some cases, X is a 5-10 membered heteroaryl, such as indolyl, or pyridyl. Specifically contemplated X include, but are not limited to, phenyl, pyridyl, indolyl, tetrahydropyranyl, piperidinyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, and piperazinyl, and can be substituted as noted above.

[0096] Some specific U-B contemplated include:

U-B

is selected from the group consisting of:

j

[0097] L 2 is Ci- 6 alkylene (e.g., Ci- 6 alkylene) or H c °- 2alkylene ~ 1 , wherein— indicates a double bond, a triple bond, or a fused or spiro cyclopropyl. In some

embodiments, L 2 is Co- 6 alkylene, e.g., Coalkylene (i.e. , a bond). In some embodiments, L 2 is Ci- 6 alkylene. For example, in some cases, L 2 is CH2, CH(CH 3 ), CH2CH2, CH2CH2CH2, or CH2CH2CH2CH2.

[0098] In various embodiments, L 2 is . in some cases wherein L 2 is ^ indicates a double bond. The double bond can be 1 ,1-

Y^ /=

substituted (e.g. ' ), or 1 ,2-substituted (e.g. ' 0 r '*'' ). In some cases, the double bond is tri- or tetra-substituted. The double bond can be substituted with substitutions independently selected from Ci-3alkyl and halo. For example, the double bond can be substituted with one or two groups independently selected from methyl, ethyl, n- propyl, isopropyl, F, Br, and Cl. The double bond orientation can be cis or trans, or when tri or tetra-substituted, E- or Z-. In some cases, the double bond is cis.

[0099] In various cases wherein L 2 is , = indicates a triple bond.

[00100] In various cases wherein L 2 is ^ indicates a fused cyclopropyl, e.g., . In some cases, = indicates a spiro cyclopropyl, e.g., The fused or spiro cyclopropyl can be further substituted with one to four substituents. Examples of suitable substituents include, but are not limited to Ci-3alkyl (e.g., methyl, ethyl, n-propyl, isopropyl), N(R N )C(0)Ci-3alkyl, and N(R N )2, where each R N is independently H or Ci-4alkyl.

[00101] In some cases

CH2CH2. In others, Co-2alkylene is null.

[00102] As provided herein, W is a bond, O, or C(0)N(R N ), and R N is H or C1-4 alkyl. In some embodiments, W is a bond. In some embodiments, W is O. In some embodiments, W is C(0)N(R N ), e.g., C(0)NH or C(0)N(Ci-4alkyl), and the alkyl can be, e.g., methyl, ethyl, propyl (n- or i-), or butyl (n-, s-, or t-). In some cases, W is C(0)N(Me).

[00103] As disclosed herein, D is C6-ioaryl or an aromatic or nonaromatic 5-10-membered heterocycle having 1-4 ring heteroatoms selected from N, O, and S. In embodiments, D is C6-ioaryl, such as phenyl or naphthyl.

[00104] In some embodiments, D is an aromatic or nonaromatic 5-10-membered heterocycle having 1-4 ring heteroatoms selected from N, O, and S. The heterocycle can be optionally substituted with 1-3 substituents independently selected from Ci-3alkyl, Ci-3alkoxy, halo, C1-3 haloalkyl, C^ haloalkoxy, N(R N ) 2 , Cs-ecycloalkyl, N0 2 , N(R N )C(0)Ci- 3 alkyl, C(0)Ci- 3 alkyl, NO2, C(0)N(R N ) 2 , CN, S02Ci-3alkyl, oxo (=0), O , NHCi-3alkylene-aryl, OCi-3alkylene- aryl, Ci-3alkylene-aryl, and 5-10 membered heterocycle having 1-3 ring heteroatoms selected from N, O, and S, and each R N is independently H or Ci-4alkyl.

[00105] In some cases, D is an aromatic membered heterocycle. Contemplated aromatic heterocycles include, but are not limited to pyridyl, indolyl, oxaxolyl, isoxazolyl, furanyl, pyranyl, thiophenyl, quinolinyl, and imidazolyl. For example, in some cases, D comprises pyridyl, and is optionally substituted with 1-3 substituents independently selected from Ci- 3alkyl, Ci-3alkoxy, halo, C1 -3 haloalkyl, Ci-3 haloalkoxy, N(R N )2, C3-6cycloalkyl, NO2,

N(R N )C(0)Ci- 3 alkyi, C(0)Ci- 3 alkyl, N0 2 , C(0)N(R N ) 2 , CN, S0 2 Ci- 3 alkyl, O , NHC^alkylene- aryl, OCi-3alkylene-aryl, Ci-3alkylene-aryl, and 5-10 membered heterocycle having 1-3 ring heteroatoms selected from N, O, and S, and each R N is independently H or Ci-4alkyl.

[00106] In some embodiments, D is a nonaromatic 5-10-membered heterocycle having 1- 4 ring heteroatoms selected from N, O, and S, and is optionally substituted. In some cases, the ring heteroatom is substituted. For example, the ring heteroatom can be substituted with an R 3 group. Contemplated non-aromatic heterocycles include, but are not limited to, tetrahydropyranyl, morpholinyl, piperazinyl, and piperidinyl. The heterocycle can be optionally substituted with 1-3 substituents independently selected from Ci-3alkyl, Ci-3alkoxy, halo, C1 -3 haloalkyl, C^ haloalkoxy, N(R N ) 2 , Cs-ecycloalkyl, N0 2 , N(R N )C(0)Ci- 3 alkyl, C(0)Ci- 3alkyl, NO2, C(0)N(R N ) 2 , CN, S0 2 Ci- 3 alkyl, and oxo (=0), and each R N is independently H or Ci-4alkyl.

[00107] In embodiments, the L 2 -W-D moiety is selected from the group consisting of:

Protein Secretion Inhibitors - Formula (II)

[00108] Also provided are compounds having a structure of Formula (II), or a

pharmaceutically acceptable salt thereof:

wherein one of Q and Q’ is U-B and the other is R 2 , or Q and Q’ and the atoms to which they are attached join together to form an aromatic or nonaromatic 5 or 6 membered carbocycle or a 5 or 6 membered heterocycle having 1 or 2 ring heteroatoms selected from N, O, and S;

U is a bond, indicates a double bond, a triple bond, or a fused or spiro cyclopropyl; B is Ci-e alkyl, C1 -3 haloalkyl, C1 -3 hydroxyalkyl, C1.3 haloalkoxy, Ci-3alkoxy, [0]o-i-Co- 3 alkylene-X or NR N Ci-3alkylene-X, X is an aromatic or nonaromatic C3-10 carbocycle, or an aromatic or nonaromatic 5-10 membered heterocycle having 1-4 ring heteroatoms selected from N, O, and S; L 2 is Ci- 6 alkylene or j ^ indicates a double bond, a triple bond, or a fused or spiro cyclopropyl; W is a bond, O, or C(0)N(R N ); D comprises pyridyl optionally substituted with 1-3 substituents independently selected from Ci-3alkyl, Ci-3alkoxy, halo, C1 -3 haloalkyl, C1.3 haloalkoxy, N(R N ) 2 , Cs-ecycloalkyl, N0 2 , C(0)N(R N ) 2 , N(R N )C(0)Ci- 3 alkyl, C(0)Ci- 3 alkyl, N0 2 , CN, S0 2 Ci- 3 alkyl, O , NHCi-3alkylene-aryl, OCi-3alkylene-aryl, Ci.

3alkylene-aryl, and 5-10 membered heterocycle having 1-3 ring heteroatoms selected from N, O, and S; each R N is independently H or Ci-4alkyl; R 1 is H or Ci-3alkyl; R 2 is H, Ci-3alkyl, or halo; and R 3 is H, Ci-3alkyl, Ci-3hydroxyalkyl, Ci-3haloalkyl, halo, or -C(0)N(R N ) 2 .

[00109] In some embodiments, Q is U-B and Q’ is R 2 . In some embodiments, Q is R 2 and Q’ is U-B. [00110] In some embodiments, Q and Q’ and the atoms to which they are attached join together to form an aromatic or nonaromatic 5 or 6 membered carbocycle or a 5 or 6 membered heterocycle having 1 or 2 ring heteroatoms selected from N, O, and S. For example, Q and Q’ can form a fused phenyl ring or cyclohexyl ring or a fused pyridyl, piperidinyl, or piperazinyl ring. The fused ring can be optionally substituted, e.g., with 1 or 2 substituents independently selected from Ci-3alkyl, Ci-3alkoxy, halo, Ci-3 haloalkyl, C1 -3 haloalkoxy, N(R N ) 2 , Cs-ecycloalkyl, N0 2 , N(R N )C(0)Ci- 3 alkyl, C(0)Ci- 3 alkyl, N0 2 , C(0)N(R N ) 2 , CN, SC>2Ci-3alkyl, oxo (=0), and O .

[00111] As disclosed herein, R 1 is H or Ci-3alkyl. In some embodiments, R 1 is H. In some embodiments, R 1 is Ci-3alkyl. Examples of contemplated R 1 groups include, but are not limited to, H, methyl, ethyl, n-propyl, and isopropyl. In various cases, R 1 is H or methyl.

[00112] As disclosed herein, R 2 is H, Ci-3alkyl, or halo. In some embodiments, R 2 is H. In some embodiments, R 2 is Ci-3alkyl. Examples of contemplated R 2 groups include, but are not limited to, H, methyl, ethyl, n-propyl, isopropyl, Br, Cl, and F. In various cases, R 2 is methyl. In various cases, R 2 is ethyl. In various cases, R 2 is n-propyl or isopropyl. In some embodiments, R 2 is halo. In some cases, R 2 is Br. In some cases, R 2 is F. In some cases, R 2 is Cl.

[00113] As disclosed herein, L 1 is a bond, Ci- 6 alkylene, or w j- ierejn ^ indicates a double bond, a triple bond, or a fused ( ) or spiro ( )cyclopropyl.

[00114] In some embodiments, L 1 is a bond. In some embodiments, U is a Ci- 6 alkylene. In various cases, is CH2, CH(CH3), CH2CH2, or C(CH3).

[00115] In some embodiments,

In some cases, the double bond is tri- or tetra-substituted. The double bond can be substituted with substitutions independently selected from Ci-3alkyl and halo. For example, the double bond can be substituted with one or two groups independently selected from methyl, ethyl, n-propyl, isopropyl, F, Br, and Cl. The double bond orientation can be cis or trans, or when tri- or tetra-substituted, E- or Z-. In some cases, the double bond is cis. [00116] In various cases when U is F c a ' k ''' a " a ^^ c °-* a ' k >' lan H ,„ = indicates a triple bond.

[00117] In various cases when U is F c ^ a ' k ''' a " a ^^ c °-* alk >' lan H ,„ = indicates a fused cyclopropyl, e.g., In some cases,— indicates a spiro cyclopropyl, e.g., . The fused or spiro cyclopropyl can be further substituted with one to four substituents. Examples of suitable substituents include, but are not limited to Ci-3alkyl (e.g., methyl, ethyl, n-propyl, isopropyl), N(R N )C(0)Ci- 3 alkyl, and N(R N )2, where each R N is independently H or Ci-4alkyl.

[00118] In some cases when

is CH2, CH(CH3) or CH2CH2 and the other is null (Co). In some cases, each is null. In some cases, each is independently CH2, CH(CH 3 ) or CH2CH2 .

[00119] As disclosed herein, B is C1 -6 alkyl, C1 -3 haloalkyl, C1.3 hydroxyalkyl, C1.3 haloalkoxy, Ci-3alkoxy, [0]o-i-Co- 3 alkylene-X or NR N Ci-3alkylene-X. X is an aromatic or nonaromatic C3-10 carbocycle, or an aromatic or nonaromatic 5-10 membered heterocycle having 1-4 ring heteroatoms selected from N, O, and S. R N is H or Ci-4alkyl, and in some cases, is H or methyl.

[00120] In various embodiments, B is C1 -6 alkyl. For example, in some cases, B is methyl, ethyl, n-propyl, isopropyl, n-butyl, sec-butyl, tert-butyl, n-pentyl, or n-hexyl.

[00121] In some embodiments, B is Ci-3haloalkyl or Ci-3haloalkoxy. For example, in some cases, B is CF 3 ,CF 2 CH 3 , CF 2 CF 3 , OCF 3 , OCH2CF3, or OCF2CF3.

[00122] In some cases, B is C1 -3 hydroxyalkyl or Ci-3alkoxy. For example, in some cases, B is CH2CH2OH, CH2OH, OMe, or OEt. .

[00123] In various embodiments, B is [0]o-i-Co- 3 alkylene-X, for example, O-X, O-C1- 3alkylene-X, Ci-3alkylene-X, or X.

[00124] In some cases, B is NR N Ci-3alkylene-X, for example, NHCi-3alkylene-X or N(CH 3 )Ci- 3 alkylene-X.

[00125] As disclosed herein, X is an aromatic or nonaromatic C3-10 carbocycle, or an aromatic or nonaromatic 5-10 membered heterocycle, having 1-4 ring heteroatoms selected from N, O, and S. X can be optionally substituted with 1-3 substituents. Contemplated substituents include Ci-3alkyl, Ci-3alkoxy, halo, Ci-3 haloalkyl, Ci-3 haloalkoxy, C(0)Ci- 3 alkyl, and S02Ci-3alkyl.

[00126] In some cases, X is an aromatic Ce-io carbocycle, e.g., phenyl or naphthyl.

[00127] In embodiments, X is an aromatic or nonaromatic 5-10 membered heterocycle. In some cases, X is a 5-10 membered nonaromatic heterocycle, such as morpholinyl, piperidinyl, tetrahydropyranyl, or piperazinyl. In some cases, X is a 5-10 membered heteroaryl, such as indolyl, or pyridyl.

[00128] Specifically contemplated X include phenyl, pyridyl, indolyl, tetrahydropyranyl, piperidinyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, and piperazinyl, and can be substituted as noted above.

[00129] Some specific U-B contemplated include:

, . In some cases, U-B is selected from the group

consisting of:

indicates a double bond, a triple bond, or a fused or spiro cyclopropyl. In some embodiments, L 2 is Ci- 6 alkylene. For example, in some cases, L 2 is CH2, CH(CH 3 ), CH2CH2, CH2CH2CH2, or CH2CH2CH2CH2.

[00131] In various embodiments, L 2 is In various embodiments,

L 2 is | n varjOUS embodiments, L 2 is In embodiments,— indicates a double bond. The double bond can be 1 ,1 -substituted (e.g. , or 1 ,2- substituted (e.g. In some cases, the double bond is tri- or tetra- substituted. The double bond can be substituted with substitutions independently selected from Ci-3alkyl and halo. For example, the double bond can be substituted with one or two groups independently selected from methyl, ethyl, n-propyl, isopropyl, F, Br, and Cl. The double bond orientation can be cis or trans, or when tri- or tetra-substituted, E- or Z-. In some cases, the double bond is cis.

[00132] In various embodiments, = indicates a triple bond.

[00133] In some embodiments, = indicates a fused cyclopropyl, e.g., . In some cases, = indicates a spiro cyclopropyl, e.g., . The fused or spiro cyclopropyl can be further substituted with one to four substituents. Examples of suitable substituents include, but are not limited to Ci-3alkyl (e.g., methyl, ethyl, n-propyl, isopropyl), N(R N )C(0)Ci- 3alkyl, and N(R N )2, where each R N is independently H or Ci-4alkyl. [00134] In some cases, when

is CH2, CH(CH3) or CH2CH2 and the other is null (Co). In some cases, each is null. In some cases, each is independently CH2, CH(CH 3 ) or CH2CH2 .

[00135] As provided herein, W is a bond, O, or C(0)N(R N ), wherein R N is H or C1 -4 alkyl.

In some embodiments, W is a bond. In some embodiments, W is O. In some embodiments, W is C(0)N(R N ), e.g., C(0)NH or C(0)N(Ci- 4 alkyl), and the alkyl can be methyl, ethyl, n- propyl, isopropyl, n-butyl, sec-butyl or tert-butyl. In some cases, W is C(0)N(Me).

[00136] As disclosed herein, D comprises pyridyl or quinolinyl. In some cases, D comprises pyridyl or quinolinyl substituted with 1-3 substituents independently selected from Ci-3alkyl, Ci-3alkoxy, halo, Ci-3 haloalkyl, Ci-3 haloalkoxy, N(R N )2, C3-6cycloalkyl, NO2, C(0)N(R n ) 2 , N(R N )C(0)Ci. 3 alkyl, C(0)Ci- 3 alkyl, N0 2 , CN, S0 2 C 1-3 alkyl, O , NHC^alkylene- aryl, OCi-3alkylene-aryl, Ci-3alkylene-aryl, and 5-10 membered heterocycle having 1-3 ring heteroatoms selected from N, O, and S, and each R N is independently H or Ci-4alkyl.

[00137] In embodiments, the L 2 -W-D moiety is selected from the group consisting of:

embodiments, the L 2 -W-D moiety is

Protein Secretion Inhibitors - Formula (III)

[00138] Also provided are compounds having a structure of Formula (III), or a

pharmaceutically acceptable salt thereof:

wherein L 1 is a bond, wherein = indicates a double bond, a triple bond, or a fused cyclopropyl; B is Co-3alkylene-X; X is an aromatic or nonaromatic C4-iocarbocycle, or an aromatic or nonaromatic 4-10 membered heterocycle having 1-3 ring heteroatoms selected from N, O, and S; R 2 is H or Ci-3alkyl; L 2 is Co-3alkylene; m is 0 to 2; and each R 4 independently is Ci-3alkyl, C^alkynyl, Ci-3haloalkyl, Ci-3alkoxy, halo, or NHCi-3alkylene-aryl.

[00139] As provided herein, L 1 is a bond, Ci- 6 alkylene, or

[00140] In some embodiments, L 1 is a bond.

[00141] In some embodiments, L 1 is Ci- 6 alkylene. In various cases, L 1 is CH2, CH2CH2, C(CH 3 ) , C(CH 3 )2CH 2 , or C(CH 3 )2CH 2 CH 2 .

[00142] In some embodiments, L 1 is | n various cases, various cases, various cases, U is · « . In various cases, = indicates a double bond. The double bond can be further substituted, for example, with Ci-3alkyl, e.g., methyl, ethyl, n-propyl, or isopropyl. In some embodiments, where = indicates a double bond, the double bond is substituted with a methyl. The double bond orientation can be cis or trans, or when substituted, E- or Z-. In various cases,— indicates a triple bond. In various cases, = indicates a fused cyclopropyl, e.g.,

[00143] As provided herein, B is Co-3alkylene-X, for example, X, Cialkylene-X, C2alkylene- X, or C3alkylene-X. In various cases, B is Ci-3alkylene-X. In various cases, B is X.

[00144] As provided herein, X is an aromatic or nonaromatic C4-iocarbocycle, or an aromatic or nonaromatic 4-10 membered heterocycle having 1-3 ring heteroatoms selected from N, O, and S. In some embodiments, X is aromatic or nonaromatic C4-7 carbocycle, or an aromatic or nonaromatic 4-9 membered heterocycle having 1 ring heteroatom. In various cases, X is an aromatic C 6 -iocarbocycle, e.g., phenyl or naphthyl. In various cases, X is an aromatic C6-7 carbocycle, e.g., phenyl. In various cases, X is a nonaromatic C4-iocarbocycle, e.g., cyclobutyl, cyclohexanyl, or cyclohexenyl. In various cases, X is a nonaromatic C4- 7carbocycle. In various cases, X is an aromatic 6-10 membered heterocycle having 1-3 ring heteroatoms selected from N, O, and S. In various cases, X is a nonaromatic 4-10 membered heterocycle having 1-3 ring heteroatoms selected from N, O, and S. In various cases, X is an aromatic 6-9 membered heterocycle having 1 ring heteroatom. In various cases, X is a nonaromatic 4-9 membered heterocycle having 1 ring heteroatom.

[00145] Examples of suitable X include, but are not limited to, pyrrolidinyl, cyclobutyl, cyclopentyl, cyclohexyl, cyclohexenyl, phenyl, piperidinyl, pyridinyl, piperazinyl,

tetrahydrofuranyl, furanyl, tetrahydropyranyl, quinolinyl, morpholinyl, pyrrolidonyl, pyrimidinyl, pyridazinyl, indenyl, dihydroindenyl, dihydrobenzofuranyl, chromanyl, isochromanyl, dihydroisoquinolinyl, or indolyl.

[00146] In some embodiments, X can be substituted with 1-3 G. In some embodiments, X is not substituted. In various cases, X is substituted with 1 G. In various cases, X is substituted with 2 G. In various cases, X is substituted with 3 G. Each G can be

independently selected from the group consisting of halo, OH, =0, CN, NO2, N(R N )2, N(R N )C(0)Ci- 3 alkyl, Ci-3alkyl, Ci-3alkoxy, Ci-3 haloalkyl, Ci-3 haloalkoxy, C(0)Ci- 3 alkyl, S(0 2 )- Z, C(0)-Z, C(0)N(R N ) 2 , silyl ether, and [0]o-i-Co- 3 alkylene-Z. As provided herein, each R N independently is H or Ci-4alkyl. [00147] In various cases, G is halo, e.g., F, Cl, or Br. In various cases, G is OH. In various cases, G is =0. In various cases, G is CN. In various cases, G is NO2. In various cases, G is N(R N )2, e.g., NH2, NHCi-3alkyl, or N(Ci- 3 alkyl) 2 . In various cases, G is

N(R N )C(0)Ci- 3 alkyl, e.g., NHC(0)CH 3 . In various cases, G is Ci-3alkyl, e.g., methyl, ethyl, n- propyl, or isopropyl. In various cases, G is Ci-3alkoxy, e.g., methoxy, ethoxy, n-propoxy, or isopropoxy. In various cases, G is Ci-3 haloalkyl, such as CF3, CHF2, or CH2F. In various cases, G is Ci-3 haloalkoxy, e.g., OCF3, OCH2CF3, or OCF2CF3. In various cases, G is C(0)Ci. 3 alkyl, e.g., C(0)CH 3 , C(0)CH 2 CH 3 , or C(0)CH 2 CH 2 CH 3 . In various cases, G is S(0 2 )-Z. In various cases, G is C(0)-Z. In various cases, G is C(0)N(R N ) 2 , e.g., C(0)NH 2 , C(0)NHCi- 3 alkyl, or C(0)N(Ci- 3 alkyl) 2 . In various cases, G is silyl ether, e.g. tert- butyldiphenylsilyl ether. In various cases, G is [0]o-i-Co- 3 alkylene-Z, e.g., 0-Ci- 3 alkylene-Z, O-Z, Ci-3alkylene-Z, or Z.

[00148] As provided herein, Z is aromatic or nonaromatic C3-iocarbocycle, or aromatic or nonaromatic 4-10 membered heterocycle having 1-3 heteroatoms selected from the group consisting of N, O, and S. In some embodiments, Z is aromatic C 6 -iocarbocycle, e.g., phenyl or naphthyl. In some embodiments, Z is nonaromatic C3-iocarbocycle, e.g., cyclopropyl, cyclobutyl, cyclopropyl, or cyclohexanyl. In some embodiments, Z is aromatic 6-10 membered heterocycle having 1-3 heteroatoms selected from the group consisting of N, O, and S. In some embodiments, Z is nonaromatic 4-10 membered heterocycle having 1-3 heteroatoms selected from the group consisting of N, O, and S.

[00149] Examples of suitable Z include, but are not limited to, pyrrolidinyl, cyclobutyl, cyclopentyl, cyclohexyl, cyclohexenyl, phenyl, piperidinyl, pyridinyl, piperazinyl,

tetrahydrofuranyl, furanyl, tetrahydropyranyl, quinolinyl, morpholinyl, pyrrolidonyl, pyrimidinyl, pyridazinyl, indenyl, dihydroindenyl, dihydrobenzofuranyl, chromanyl, isochromanyl, dihydroisoquinolinyl, or indolyl.

[00150] In some embodiments, Z is substituted with 1-3 E. In some embodiments, Z is not substituted. In various cases, Z is substituted with 1 E. In various cases, Z is substituted with 2 E. In various cases, Z is substituted with 3 E. Each E can be independently selected from Ci-3alkyl, Ci-3alkoxy, =0, Ci-3haloalkoxy, CN, and halo.

[00151] In various cases, E is Ci-3alkyl, e.g., methyl, ethyl, n-propyl, or isopropyl. In various cases, E is Ci-3alkoxy, e.g., methoxy, ethoxy, n-propoxy, or isopropoxy. In various cases, E is =0. In various cases, E is Ci-3haloalkoxy, e.g., OCF3, OCH2CF3, or OCF2CF3. In various cases, E is CN. In various cases, E is halo, e.g., F, Cl, or Br. x

[00152] In some embodiments, U-B is selected from the group consisting of

x

T 7 X

, and , wherein X is as described herein. For example, in embodiments, X is aromatic or nonaromatic C4-7 carbocycle, or an aromatic or nonaromatic 4-9 membered heterocycle having 1 ring heteroatom. In some embodiments, X is substituted with 1-3 G, wherein G can be as described herein.

[00153] As provided herein, R 2 is H or Ci-3alkyl. In some embodiments, R 2 is H. In some embodiments, R 2 is Ci-3alkyl. In various cases, R 2 is methyl, ethyl, n-propyl, or isopropyl. In various cases, R 2 is methyl.

[00154] As provided herein, L 2 is Co-3alkylene. In various cases, L 2 is a bond (/.e., Coalkylene). In various cases, L 2 is Cialkylene. In various cases, L 2 is C2alkylene. In various cases , L 2 is Csalkylene.

[00155] As provided herein, m is 0 to 2. In various cases, m is 0. In various cases, m is 1. In various cases, m is 2. In various cases, m is 1 or 2.

[00156] As provided herein, each R 4 independently is Ci-3alkyl, C^alkynyl, Ci-3haloalkyl, Ci-3alkoxy, halo, or NHCi-3alkylene-aryl.

[00157] In some embodiments, at least one R 4 is Ci-3alkyl, e.g., methyl, ethyl, n-propyl, or isopropyl. In various cases, at least one R 4 is methyl or ethyl. In some embodiments, at least one R 4 is C^alkynyl. In various cases, at least one R 4 is C2alkynyl. In various cases, at least one R 4 is Csalkynyl. In some embodiments, at least one R 4 is Ci-3haloalkyl, e.g.,

CF3, CHF2, or CH2F. In various cases, at least one R 4 is CF3. In some embodiments, at least one R 4 is Ci-3alkoxy, e.g., methoxy, ethoxy, n-propoxy, or isopropoxy. In various cases, at least one R 4 is methoxy. In some embodiments, at least one R 4 is halo, e.g., F, Cl, or Br. In various cases, at least one R 4 is F. In various cases, at least one R 4 is Cl. In some embodiments, NHCi-3alkylene-aryl. In various cases, at least one R 4 is NH-CFh-phenyl.

[00158] In some embodiments, one R 4 is halo, and the other R 4 is halo or methyl. In various cases, one R 4 is halo, and the other R 4 is halo. For example, in various cases, one R 4 is F, and the other R 4 is F. In various cases, one R 4 is halo, and the other R 4 is methyl. For example, in various cases, one R 4 is F, and the other R 4 is methyl.

[00159] In some embodiments, the compound has a structure of Formula (IIIA):

wherein each of U-B and R 2 are as described herein.

[00160] In some embodiments, U-B is selected from the group consisting of

[00162] In some embodiments, U-B is selected from the group consisting of

[00163] In some embodiments, U-B is selected from the group consisting of

[00164] In some embodiments, U-B is selected from the group consisting

[00165] Examples of compounds according to Formulae (I), (II), and (III) of the disclosure are shown in Table A, below, as compounds A1-A403. In some embodiments, a compound of the disclosure is one of A1-A210. Additional compounds of the disclosure are shown in Table B, below, as compounds B1-B29. In some embodiments, a compound of the disclosure is one of B1-B29.

[00166] In embodiments, the compound is selected from a compound listed in Table A, or a pharmaceutically acceptable salt thereof. In some embodiments, the compound or salt is selected from A1-A210. In some embodiments, the compound or salt is selected from A211- A403. In some cases, the compound is selected from the group consisting of

[00167] In some embodiments, the compound is selected from a compound listed in Table B, or a pharmaceutically acceptable salt thereof. In some embodiments, the compound or salt is selected from B1-B29.

[00168] The chemical structures having one or more stereocenters depicted with dashed and bold wedged bonds (i.e. , . and— ) are meant to indicate absolute stereochemistry of the stereocenter(s) present in the chemical structure. Bonds symbolized by a simple line do not indicate a stereo-preference. Bonds symbolized by dashed or bold straight bonds

(i.e., . and— ) are meant to indicate a relative stereochemistry of the stereocenter(s) present in the chemical structure. Unless otherwise indicated to the contrary, chemical structures that include one or more stereocenters which are illustrated herein without indicating absolute or relative stereochemistry, encompass all possible stereoisomeric forms of the compound (e.g., diastereomers, enantiomers) and mixtures thereof. Structures with a single bold or dashed wedged line, and at least one additional simple line, encompass a single enantiomeric series of all possible diastereomers. Similarly, the chemical structures having alkenyl groups are meant to encompass both cis and trans orientations, or when substituted, E- and Z-isomers of the chemical structure.

Synthesis of Protein Secretion Inhibitors

[00169] The compounds provided herein can be synthesized using conventional techniques readily available starting materials known to those skilled in the art. In general, the compounds provided herein are conveniently obtained via standard organic chemistry synthesis methods. [00170] Although not limited to any one or several sources, classic texts such as Smith,

M. B., March, J., Marchs Advanced Organic Chemistry: Reactions, Mechanisms, and Structure, 5 th edition, John Wiley & Sons: New York, 2001 ; and Greene, T.W., Wuts, P.G.

M., Protective Groups in Organic Synthesis, 3 rd edition, John Wiley & Sons: New York, 1999, are useful and recognized reference textbooks of organic synthesis known to those in the art. The following descriptions of synthetic methods are designed to illustrate, but not to limit, general procedures for the preparation of compounds of the present disclosure.

[00171] The synthetic processes disclosed herein can tolerate a wide variety of functional groups; therefore, various substituted starting materials can be used. The processes generally provide the desired final compound at or near the end of the overall process, although it may be desirable in certain instances to further convert the compound to a pharmaceutically acceptable salt thereof.

[00172] In general, the compounds of Formula (I), (II) or (III) can be synthesized in line with the examples shown below. For example, the compounds can be prepared by alkylation of the appropriate amine having a carboxyl group, with appropriate protecting groups as necessary. The intermediate can be saponified, for example, to expose a reactive carboxylate. Then, amide coupling between the appropriate amine and the free carboxylate can occur.

[00173] The amine for the amide coupling noted above can be prepared via known synthetic techniques using appropriate starting materials and protecting groups, as necessary.

[00174] Further modifications can be performed, e.g., to introduce additional substituents such as halo groups or alkyl groups.

Methods of Use

[00175] The compounds disclosed herein (e.g., the compounds of Formulae (I), (II), and (III), the compounds listed in Tables A and B, and pharmaceutically acceptable salts of any of the foregoing) can inhibit protein secretion of a protein of interest. The compounds disclosed herein can interfere with the Sec61 protein secretion machinery of a cell. In some cases, a compound as disclosed herein inhibits secretion of one or more of TNFa, IL2, Her3, and PD-1 , or each of TNFa, IL2, Her3, and PD-1. Protein secretion activity can be assessed in a manner as described in the Examples section below.

[00176] As used herein, the term“inhibitor” is meant to describe a compound that blocks or reduces an activity of a pharmacological target (for example, a compound that inhibits Sec61 function in the protein secretion pathway). An inhibitor can act with competitive, uncompetitive, or noncompetitive inhibition. An inhibitor can bind reversibly or irreversibly, and therefore, the term includes compounds that are suicide substrates of a protein or enzyme. An inhibitor can modify one or more sites on or near the active site of the protein, or it can cause a conformational change elsewhere on the enzyme. The term inhibitor is used more broadly herein than scientific literature so as to also encompass other classes of pharmacologically or therapeutically useful agents, such as agonists, antagonists, stimulants, co-factors, and the like.

[00177] Thus, provided herein are methods of inhibiting protein secretion in a cell. In these methods, a cell is contacted with a compound described herein (e.g., a compound of Formula (I), (II), or (III), or a compound listed in Tables A or B, and pharmaceutically acceptable salts of any of the foregoing), or pharmaceutical composition thereof, in an amount effective to inhibit secretion of the protein of interest. In some embodiments, the cell is contacted in vitro. In various embodiments, the cell is contacted in vivo. In various embodiments, the contacting includes administering the compound or pharmaceutical composition to a subject.

[00178] The biological consequences of Sec61 inhibition are numerous. For example, Sec61 inhibition has been suggested for the treatment or prevention of inflammation and/or cancer in a subject. Therefore, pharmaceutical compositions for Sec61 specific compounds, provide a means of administering a drug to a subject and treating these conditions. As used herein, the terms "treat," "treating," "treatment," and the like refer to eliminating, reducing, or ameliorating a disease or condition, and/or symptoms associated therewith. Although not precluded, treating a disease or condition does not require that the disease, condition, or symptoms associated therewith be completely eliminated. As used herein, the terms "treat," "treating," "treatment," and the like may include "prophylactic treatment," which refers to reducing the probability of redeveloping a disease or condition, or of a recurrence of a previously-controlled disease or condition, in a subject who does not have, but is at risk of or is susceptible to, redeveloping a disease or condition or a recurrence of the disease or condition. The term "treat" and synonyms contemplate administering a therapeutically effective amount of a compound of the disclosure to an individual in need of such treatment. Within the meaning of the disclosure, "treatment" also includes relapse prophylaxis or phase prophylaxis, as well as the treatment of acute or chronic signs, symptoms and/or

malfunctions. The treatment can be orientated symptomatically, for example, to suppress symptoms. It can be effected over a short period, be oriented over a medium term, or can be a long-term treatment, for example within the context of a maintenance therapy. As used herein, the terms“prevent,”“preventing,”“prevention,” are art-recognized, and when used in relation to a condition, such as a local recurrence (e.g., pain), a disease such as cancer, a syndrome complex such as heart failure or any other medical condition, is well understood in the art, and includes administration of a composition which reduces the frequency of, or delays the onset of, symptoms of a medical condition in a subject relative to a subject which does not receive the composition. Thus, prevention of cancer includes, for example, reducing the number of detectable cancerous growths in a population of patients receiving a prophylactic treatment relative to an untreated control population, and/or delaying the appearance of detectable cancerous growths in a treated population versus an untreated control population, e.g., by a statistically and/or clinically significant amount. As used herein, the terms“patient” and“subject” may be used interchangeably and mean animals, such as dogs, cats, cows, horses, and sheep (i.e. , non-human animals) and humans. Particular patients are mammals (e.g., humans). The term patient includes males and females.

[00179] Inhibition of Sec61 -mediated secretion of inflammatory proteins (e.g., TNFa) can disrupt inflammation signaling. Thus, provided herein is a method of treating inflammation in a subject by administering to the subject a therapeutically effective amount of a compound described herein, (i.e., a compound of Formula (I), (II), or (III), or a compound listed in Tables A or B), or a pharmaceutically acceptable salt thereof.

[00180] Further, the viability of cancer cells relies upon increased protein secretion into the ER for survival. Therefore, non-selective or partially selective inhibition of Sec61 mediated protein secretion may inhibit tumor growth. Alternatively, in the immune-oncology setting, selective secretion inhibitors of known secreted immune checkpoints proteins (e.g., PD-1 , TIM-3, LAG3, etc.) can result in activation of the immune system to against various cancers.

[00181] Accordingly, also provided herein are methods of treating cancer in a subject by administering to the subject a therapeutically effective amount of a compound described herein, (e.g., a compound of Formula (I), (II), or (III), or a compound listed in Table A or B), or a pharmaceutically acceptable salt thereof. Specifically contemplated cancers that can be treated using the compounds and compositions described herein include, but are not limited to melanoma, multiple myeloma, prostate, lung, non small cell lung carconimoa (NSCLC), squamous cell carcinoma, leukemia, acute myelogenous leukemia, chronic myelogenous leukemia, lymphoma, NPM/ALK-transformed anaplastic large cell lymphoma, renal cell carcinoma, rhabdomyosarcoma, ovarian cancer, endometrial cancer, small cell carcinoma, adenocarcinoma, gastric carcinoma, hepatocellular carcinoma, pancreatic cancer, thyroid carcinoma, anaplastic large cell lymphoma, hemangioma, head and neck cancer, bladder, and colorectal cancers.

[00182] The compounds described herein are also contemplated to be used in the prevention and/or treatment of a multitude of diseases including, but not limited to, proliferative diseases, neurotoxic/degenerative diseases, ischemic conditions, autoimmune and autoinflammatory disorders, inflammation, immune-related diseases, HIV, cancers, organ graft rejection, septic shock, viral and parasitic infections, conditions associated with acidosis, macular degeneration, pulmonary conditions, muscle wasting diseases, fibrotic diseases, bone and hair growth diseases.

[00183] Examples of proliferative diseases or conditions include diabetic retinopathy, macular degeneration, diabetic nephropathy, glomerulosclerosis, IgA nephropathy, cirrhosis, biliary atresia, congestive heart failure, scleroderma, radiation-induced fibrosis, and lung fibrosis (idiopathic pulmonary fibrosis, collagen vascular disease, sarcoidosis, interstitial lung diseases and extrinsic lung disorders).

[00184] Inflammatory diseases include acute (e.g., bronchitis, conjunctivitis, myocarditis, pancreatitis) and chronic conditions (e.g., chronic cholecstitis, bronchiectasis, aortic valve stenosis, restenosis, psoriasis and arthritis), along with conditions associated with

inflammation such as fibrosis, infection and ischemia.

[00185] Immunodeficiency disorders occur when a part of the immune system is not working properly or is not present. They can affect B lymophyctes, T lymphocytes, or phagocytes and be either inherited (e.g., IgA deficiency, severe combined immunodeficiency (SCID), thymic dysplasia and chronic granulomatous) or acquired (e.g., acquired

immunodeficiency syndrome (AIDS), human immunodeficiency virus (HIV) and drug-induced immunodeficiencies). Immune-related conditions include allergic disorders such as allergies, asthma and atopic dermatitis like eczema. Other examples of such immune-related conditions include lupus, rheumatoid arthritis, scleroderma, ankylosing spondylitis, dermatomyositis, psoriasis, multiple sclerosis and inflammatory bowel disease (such as ulcerative colitis and Crohn’s disease).

[00186] Tissue/organ graft rejection occurs when the immune system mistakenly attacks the cells being introduced to the host’s body. Graft versus host disease (GVHD), resulting from allogenic transplantation, arises when the T cells from the donor tissue go on the offensive and attack the host’s tissues. In all three circumstances, autoimmune disease, transplant rejection and GVHD, modulating the immune system by treating the subject with a compound or composition of the disclosure could be beneficial.

[00187] Also provided herein are methods of treating an autoimmune disease in a patient comprising administering a therapeutically effective amount of the compound described herein. An“autoimmune disease” as used herein is a disease or disorder arising from and directed against an individual’s own tissues. Examples of autoimmune diseases include, but are not limited to, inflammatory responses such as inflammatory skin diseases including psoriasis and dermatitis (e.g., atopic dermatitis); systemic scleroderma and sclerosis;

responses associated with inflammatory bowel disease (such as Crohn’s disease and ulcerative colitis); respiratory distress syndrome (including adult respiratory distress syndrome(ARDS)); dermatitis; meningitis; encephalitis; uveitis; colitis; glomerulonephritis; allergic conditions such as eczema and asthma and other conditions involving infiltration of T cells and chronic inflammatory responses; atherosclerosis; leukocyte adhesion deficiency; rheumatoid arthritis; systemic lupus erythematosus (SLE); diabetes mellitus (e.g., Type I diabetes mellitus or insulin dependent diabetes mellitus); multiple sclerosis; Reynaud’s syndrome; autoimmune thyroiditis; allergic encephalomyelitis; Sjorgen’s syndrome; juvenile onset diabetes; and immune responses associated with acute and delayed hypersensitivity mediated by cytokines and T-lymphocytes typically found in tuberculosis, sarcoidosis, polymyositis, granulomatosis and vasculitis; pernicious anemia (Addison’s disease);

diseases involving leukocyte diapedesis; central nervous system (CNS) inflammatory disorder; multiple organ injury syndrome; hemolytic anemia (including, but not limited to cryoglobinemia or Coombs positive anemia); myasthenia gravis; antigen-antibody complex mediated diseases; anti-glomerular basement membrane disease; antiphospholipid syndrome; allergic neuritis; Graves’ disease; Lambert-Eaton myasthenic syndrome;

pemphigoid bullous; pemphigus; autoimmune polyendocrinopathies; Reiter’s disease; stiff- man syndrome; Behcet disease; giant cell arteritis; immune complex nephritis; IgA nephropathy; IgM polyneuropathies; immune thrombocytopenic purpura (ITP) or

autoimmune thrombocytopenia. Compounds provided herein may be useful for the treatment of conditions associated with inflammation, including, but not limited to COPD, psoriasis, asthma, bronchitis, emphysema, and cystic fibrosis.

[00188] Also provided herein is the use of a compound as disclosed herein for the treatment of neurodegenerative diseases. Neurodegenerative diseases and conditions includes, but not limited to, stroke, ischemic damage to the nervous system, neural trauma (e.g., percussive brain damage, spinal cord injury, and traumatic damage to the nervous system), multiple sclerosis and other immune-mediated neuropathies (e.g., Guillain-Barre syndrome and its variants, acute motor axonal neuropathy, acute inflammatory

demyelinating polyneuropathy, and Fisher Syndrome), HIV/AIDS dementia complex, axonomy, diabetic neuropathy, Parkinson’s disease, Huntington's disease, multiple sclerosis, bacterial, parasitic, fungal, and viral meningitis, encephalitis, vascular dementia, multi-infarct dementia, Lewy body dementia, frontal lobe dementia such as Pick’s disease, subcortical dementias (such as Huntington or progressive supranuclear palsy), focal cortical atrophy syndromes (such as primary aphasia), metabolic- toxic dementias (such as chronic hypothyroidism or B12 deficiency), and dementias caused by infections (such as syphilis or chronic meningitis).

[00189] Further guidance for using compounds and compositions described herein (e.g., a compound of Formula (I), (II), or (III), a compound listed in Table A or B, or a

pharmaceutically acceptable salt thereof) for inhibiting protein secretion can be found in the Examples section, below.

Pharmaceutical Compositions and Administration

[00190] The methods provided herein include the manufacture and use of pharmaceutical compositions, which include one or more of the compounds provided herein. Also included are the pharmaceutical compositions themselves. Pharmaceutical compositions typically include a pharmaceutically acceptable carrier. Thus, provided herein are pharmaceutical compositions that include a compound described herein (e.g., a compound of Formula (I),

(II), or (III), a compound listed in Table A or B, or a pharmaceutically acceptable salt thereof), as previously described herein, and one or more pharmaceutically acceptable carriers.

[00191] The phrase“pharmaceutically acceptable” is employed herein to refer to those ligands, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.

[00192] The phrase“pharmaceutically acceptable carrier” as used herein means a pharmaceutically acceptable material, composition, or vehicle, such as a liquid or solid filler, diluent, excipient, solvent or encapsulating material. As used herein the language “pharmaceutically acceptable carrier” includes buffer, sterile water for injection, solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like, compatible with pharmaceutical administration. Each carrier must be“acceptable” in the sense of being compatible with the other ingredients of the composition and not injurious to the patient. Some examples of materials which can serve as pharmaceutically acceptable carriers include: (1) sugars, such as lactose, glucose, and sucrose; (2) starches, such as corn starch, potato starch, and substituted or unsubstituted b- cyclodextrin; (3) cellulose, and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose, and cellulose acetate; (4) powdered tragacanth; (5) malt; (6) gelatin; (7) talc; (8) excipients, such as cocoa butter and suppository waxes; (9) oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil, and soybean oil; (10) glycols, such as propylene glycol; (11) polyols, such as glycerin, sorbitol, mannitol, and polyethylene glycol; (12) esters, such as ethyl oleate and ethyl laurate; (13) agar; (14) buffering agents, such as magnesium hydroxide and aluminum hydroxide; (15) alginic acid; (16) pyrogen-free water; (17) isotonic saline; (18) Ringers solution; (19) ethyl alcohol; (20) phosphate buffer solutions; and (21) other non-toxic compatible substances employed in pharmaceutical compositions. In certain embodiments, pharmaceutical compositions provided herein are non-pyrogenic, i.e., do not induce significant temperature elevations when administered to a patient.

[00193] The term“pharmaceutically acceptable salt” refers to the relatively non-toxic, inorganic and organic acid addition salts of a compound provided herein. These salts can be prepared in situ during the final isolation and purification of a compound provided herein, or by separately reacting the compound in its free base form with a suitable organic or inorganic acid, and isolating the salt thus formed. Representative salts include the hydrobromide, hydrochloride, sulfate, bisulfate, phosphate, nitrate, acetate, valerate, oleate, palmitate, stearate, laurate, benzoate, lactate, phosphate, tosylate, citrate, maleate, fumarate, succinate, tartrate, naphthylate, mesylate, glucoheptonate, lactobionate, laurylsulphonate salts, and amino acid salts, and the like. (See, for example, Berge et al. (1977)“Pharmaceutical Salts”, J. Pharm. Sci. 66: 1-19.)

[00194] In some embodiments, a compound provided herein may contain one or more acidic functional groups and, thus, is capable of forming pharmaceutically acceptable salts with pharmaceutically acceptable bases. The term“pharmaceutically acceptable salts” in these instances refers to the relatively non-toxic inorganic and organic base addition salts of a compound provided herein. These salts can likewise be prepared in situ during the final isolation and purification of the compound, or by separately reacting the purified compound in its free acid form with a suitable base, such as the hydroxide, carbonate, or bicarbonate of a pharmaceutically acceptable metal cation, with ammonia, or with a pharmaceutically acceptable organic primary, secondary, or tertiary amine. Representative alkali or alkaline earth salts include the lithium, sodium, potassium, calcium, magnesium, and aluminum salts, and the like. Representative organic amines useful for the formation of base addition salts include ethylamine, diethylamine, ethylenediamine, ethanolamine, diethanolamine, piperazine, and the like (see, for example, Berge et al., supra).

[00195] Wetting agents, emulsifiers, and lubricants, such as sodium lauryl sulfate and magnesium stearate, as well as coloring agents, release agents, coating agents,

sweetening, flavoring, and perfuming agents, preservatives and antioxidants can also be present in the compositions.

[00196] Examples of pharmaceutically acceptable antioxidants include: (1) water soluble antioxidants, such as ascorbic acid, cysteine hydrochloride, sodium bisulfate, sodium metabisulfite, sodium sulfite, and the like; (2) oil-soluble antioxidants, such as ascorbyl palmitate, butylated hydroxyanisole (BHA), butylated hydroxytoluene (BHT), lecithin, propyl gallate, alpha-tocopherol, and the like; and (3) metal chelating agents, such as citric acid, ethylenediamine tetraacetic acid (EDTA), sorbitol, tartaric acid, phosphoric acid, and the like.

[00197] A pharmaceutical composition may also contain adjuvants such as preservatives, wetting agents, emulsifying agents, and dispersing agents. Prevention of the action of microorganisms may be ensured by the inclusion of various antibacterial and antifungal agents, for example, paraben, chlorobutanol, phenol sorbic acid, and the like. It may also be desirable to include tonicity-adjusting agents, such as sugars and the like into the

compositions. In addition, prolonged absorption of the injectable pharmaceutical form may be brought about by the inclusion of agents which delay absorption such as aluminum monostearate and gelatin.

[00198] In some cases, in order to prolong the effect of one or more compounds provided herein, it is desirable to slow the absorption of the compound from subcutaneous or intramuscular injection. For example, delayed absorption of a parenterally administered compound can be accomplished by dissolving or suspending the compound in an oil vehicle.

[00199] Compositions prepared as described herein can be administered in various forms, depending on the disorder to be treated and the age, condition, and body weight of the patient, as is well known in the art. For example, where the compositions are to be administered orally, they may be formulated as tablets, capsules, granules, powders, or syrups; or for parenteral administration, they may be formulated as injections (intravenous, intramuscular, or subcutaneous), drop infusion preparations, or suppositories. For application by the ophthalmic mucous membrane route, they may be formulated as eye drops or eye ointments. These compositions can be prepared by conventional means in conjunction with the methods described herein, and, if desired, the active ingredient may be mixed with any conventional additive or excipient, such as a binder, a disintegrating agent, a lubricant, a corrigent, a solubilizing agent, a suspension aid, an emulsifying agent, or a coating agent.

[00200] Compositions suitable for oral administration may be in the form of capsules (e.g., gelatin capsules), cachets, pills, tablets, lozenges (using a flavored basis, usually sucrose and acacia or tragacanth), powders, troches, granules, or as a solution or a suspension in an aqueous or non-aqueous liquid, or as an oil-in-water or water-in-oil liquid emulsion, or as an elixir or syrup, or as pastilles (using an inert matrix, such as gelatin and glycerin, or sucrose and acacia) and/or as mouthwashes, and the like, each containing a predetermined amount of a compound provided herein as an active ingredient. A composition may also be administered as a bolus, electuary, or paste. Oral compositions generally include an inert diluent or an edible carrier.

[00201] Pharmaceutically compatible binding agents, and/or adjuvant materials can be included as part of an oral composition. In solid dosage forms for oral administration

(capsules, tablets, pills, dragees, powders, granules, and the like), the active ingredient can be mixed with one or more pharmaceutically acceptable carriers, such as sodium citrate or dicalcium phosphate, and/or any of the following: (1) fillers or extenders, such as starches, cyclodextrins, lactose, sucrose, saccharin, glucose, mannitol, and/or silicic acid; (2) binders, such as, for example, carboxymethylcellulose, microcrystalline cellulose, gum tragacanth, alginates, gelatin, polyvinyl pyrrolidone, sucrose, and/or acacia; (3) humectants, such as glycerol; (4) disintegrating agents, such as agar-agar, calcium carbonate, potato, corn, or tapioca starch, alginic acid, Primogel, certain silicates, and sodium carbonate; (5) solution retarding agents, such as paraffin; (6) absorption accelerators, such as quaternary ammonium compounds; (7) wetting agents, such as, for example, acetyl alcohol and glycerol monostearate; (8) absorbents, such as kaolin and bentonite clay; (9) lubricants, such a talc, calcium stearate, magnesium stearate, Sterotes, solid polyethylene glycols, sodium lauryl sulfate, and mixtures thereof; (10) a glidant, such as colloidal silicon dioxide; (11) coloring agents; and (12) a flavoring agent such as peppermint, methyl salicylate, or orange flavoring. In the case of capsules, tablets, and pills, the pharmaceutical compositions may also comprise buffering agents. Solid compositions of a similar type may also be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugars, as well as high molecular weight polyethylene glycols, and the like.

[00202] A tablet may be made by compression or molding, optionally with one or more accessory ingredients. Compressed tablets may be prepared using binder (for example, gelatin or hydroxypropylmethyl cellulose), lubricant, inert diluent, preservative, disintegrant (for example, sodium starch glycolate or cross-linked sodium carboxymethyl cellulose), surface-active or dispersing agent. Molded tablets may be made by molding in a suitable machine a mixture of a powdered compound moistened with an inert liquid diluent.

[00203] Tablets, and other solid dosage forms, such as dragees, capsules, pills, and granules, may optionally be scored or prepared with coatings and shells, such as enteric coatings and other coatings well known in the pharmaceutical-formulating art. They may also be formulated so as to provide slow or controlled release of the active ingredient therein using, for example, hydroxypropylmethyl cellulose in varying proportions to provide the desired release profile, other polymer matrices, liposomes, microspheres, and/or

nanoparticles. They may be sterilized by, for example, filtration through a bacteria-retaining filter, or by incorporating sterilizing agents in the form of sterile solid compositions which can be dissolved in sterile water, or some other sterile injectable medium immediately before use. These compositions may also optionally contain opacifying agents and may be of a composition that they release the active ingredient(s) only, or preferentially, in a certain portion of the gastrointestinal tract, optionally, in a delayed manner. Examples of embedding compositions which can be used include polymeric substances and waxes. The active ingredient can also be in micro-encapsulated form, if appropriate, with one or more of the above-described excipients.

[00204] Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups, and elixirs. In addition to the active ingredient, the liquid dosage forms may contain inert diluents commonly used in the art, such as, for example, water or other solvents, solubilizing agents, and emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1 ,3-butylene glycol, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor, and sesame oils), glycerol, tetra hydrofury I alcohol, polyethylene glycols, and fatty acid esters of sorbitan, and mixtures thereof.

[00205] Besides inert diluents, the oral compositions can also include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, coloring, perfuming, and preservative agents.

[00206] Suspensions, in addition to the active compound(s) may contain suspending agents as, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar and tragacanth, and mixtures thereof.

[00207] Pharmaceutical compositions suitable for parenteral administration can include one or more compounds provided herein in combination with one or more pharmaceutically acceptable sterile aqueous or nonaqueous solutions, dispersions, suspensions or emulsions, or sterile powders which may be reconstituted into sterile injectable solutions or dispersions just prior to use, which may contain antioxidants, buffers, bacteriostats, solutes which render the composition isotonic with the blood of the intended recipient or suspending or thickening agents.

[00208] Examples of suitable aqueous and nonaqueous carriers which may be employed in the pharmaceutical compositions provided herein include water for injection (e.g., sterile water for injection), bacteriostatic water, ethanol, polyols (such as glycerol, propylene glycol, polyethylene glycol such as liquid polyethylene glycol, and the like), sterile buffer (such as citrate buffer), and suitable mixtures thereof, vegetable oils, such as olive oil, injectable organic esters, such as ethyl oleate, and Cremophor EL™ (BASF, Parsippany, NJ). In all cases, the composition must be sterile and should be fluid to the extent that easy

syringability exists. Proper fluidity can be maintained, for example, by the use of coating materials, such as lecithin, by the maintenance of the required particle size in the case of dispersions, and by the use of surfactants.

[00209] The composition should be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms such as bacteria and fungi. Prevention of the action of microorganisms can be achieved by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, ascorbic acid, thimerosal, and the like. In many cases, it will be preferable to include isotonic agents, for example, sugars, polyalcohols such as mannitol, sorbitol, and sodium chloride in the composition. Prolonged absorption of the injectable compositions can be brought about by including in the composition an agent that delays absorption, for example, aluminum monostearate and gelatin.

[00210] Sterile injectable solutions can be prepared by incorporating the active compound in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, followed by filtered sterilization. Generally, dispersions are prepared by incorporating the active compound into a sterile vehicle, which contains a basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile powders for the preparation of sterile injectable solutions, the methods of preparation are freeze-drying (lyophilization), which yields a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.

[00211] Injectable depot forms can be made by forming microencapsule or

nanoencapsule matrices of a compound provided herein in biodegradable polymers such as polylactide-polyglycolide. Depending on the ratio of drug to polymer, and the nature of the particular polymer employed, the rate of drug release can be controlled. Examples of other biodegradable polymers include poly(orthoesters) and poly(anhydrides). Depot injectable compositions are also prepared by entrapping the drug in liposomes, microemulsions or nanoemulsions, which are compatible with body tissue.

[00212] For administration by inhalation, the compounds can be delivered in the form of an aerosol spray from a pressured container or dispenser that contains a suitable propellant, e.g., a gas such as carbon dioxide, or a nebulizer. Such methods include those described in U.S. Patent No. 6,468,798. Additionally, intranasal delivery can be accomplished, as described in, inter alia, Hamajima et al., Clin. Immunol. Immunopathol., 88(2), 205-10 (1998). Liposomes (e.g., as described in U.S. Patent No. 6,472,375, which is incorporated herein by reference in its entirety), microencapsulation and nanoencapsulation can also be used. Biodegradable targetable microparticle delivery systems or biodegradable targetable nanoparticle delivery systems can also be used (e.g., as described in U.S. Patent No.

6,471 ,996, which is incorporated herein by reference in its entirety).

[00213] Systemic administration of a therapeutic compound as described herein can also be by transmucosal or transdermal means. Dosage forms for the topical or transdermal administration of a compound provided herein include powders, sprays, ointments, pastes, creams, lotions, gels, solutions, patches, and inhalants. The active component may be mixed under sterile conditions with a pharmaceutically acceptable carrier, and with any preservatives, buffers, or propellants which may be required. For transmucosal or transdermal administration, penetrants appropriate to the barrier to be permeated are used in the composition. Such penetrants are generally known in the art, and include, for example, for transmucosal administration, detergents, bile salts, and fusidic acid derivatives. Transmucosal administration can be accomplished through the use of nasal sprays or suppositories. For transdermal administration, the active compounds are formulated into ointments, salves, gels, or creams as generally known in the art.

[00214] The ointments, pastes, creams, and gels may contain, in addition to one or more compounds provided herein, excipients, such as animal and vegetable fats, oils, waxes, paraffins, starch, tragacanth, cellulose derivatives, polyethylene glycols, silicones, bentonites, silicic acid, talc, and zinc oxide, or mixtures thereof.

[00215] Powders and sprays can contain, in addition to a compound provided herein, excipients such as lactose, talc, silicic acid, aluminum hydroxide, calcium silicates, and polyamide powder, or mixtures of these substances. Sprays can additionally contain customary propellants, such as chlorofluorohydrocarbons and volatile unsubstituted hydrocarbons, such as butane and propane.

[00216] A compound provided herein can be administered by aerosol. This is

accomplished by preparing an aqueous aerosol, liposomal preparation, or solid particles containing a compound or composition provided herein. A nonaqueous (e.g., fluorocarbon propellant) suspension could be used. In some embodiments, sonic nebulizers are used because they minimize exposing the agent to shear, which can result in degradation of the compound.

[00217] Ordinarily, an aqueous aerosol can be made by formulating an aqueous solution or suspension of the agent together with conventional pharmaceutically acceptable carriers and stabilizers. The carriers and stabilizers vary with the requirements of the particular composition, but typically include nonionic surfactants (TWEEN® (polysorbates),

PLURONIC® (poloxamers), sorbitan esters, lecithin, CREMOPHOR® (polyethoxylates)), pharmaceutically acceptable co-solvents such as polyethylene glycol, innocuous proteins like serum albumin, sorbitan esters, oleic acid, lecithin, amino acids such as glycine, buffers, salts, sugars, or sugar alcohols. Aerosols generally are prepared from isotonic solutions.

[00218] Transdermal patches have the added advantage of providing controlled delivery of a compound provided herein to the body. Such dosage forms can be made by dissolving or dispersing the agent in the proper medium. Absorption enhancers can also be used to increase the flux of the compound across the skin. The rate of such flux can be controlled by either providing a rate controlling membrane or dispersing the compound in a polymer matrix or gel.

[00219] The pharmaceutical compositions can also be prepared in the form of

suppositories or retention enemas for rectal and/or vaginal delivery. Compositions presented as a suppository can be prepared by mixing one or more compounds provided herein with one or more suitable nonirritating excipients or carriers comprising, for example, cocoa butter, glycerides, polyethylene glycol, a suppository wax or a salicylate, which is solid at room temperature, but liquid at body temperature and, therefore, will melt in the rectum or vaginal cavity and release the active agent. Compositions which are suitable for vaginal administration also include pessaries, tampons, creams, gels, pastes, foams, or spray compositions containing such carriers as are known in the art to be appropriate.

[00220] In one embodiment, the therapeutic compounds are prepared with carriers that will protect the therapeutic compounds against rapid elimination from the body, such as a controlled release composition, including implants and microencapsulated delivery systems. Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid. Such compositions can be prepared using standard techniques, or obtained commercially, e.g., from Alza Corporation and Nova Pharmaceuticals, Inc. Liposomal suspensions (including liposomes targeted to selected cells with monoclonal antibodies to cellular antigens) can also be used as pharmaceutically acceptable carriers. These can be prepared according to methods known to those skilled in the art, for example, as described in U.S. Patent No. 4,522,811 , which is incorporated herein by reference in its entirety.

[00221] As described above, the preparations of one or more compounds provided herein may be given orally, parenterally, topically, or rectally. They are, of course, given by forms suitable for each administration route. For example, they are administered in tablets or capsule form, by injection, inhalation, eye lotion, ointment, suppository, infusion; topically by lotion or ointment; and rectally by suppositories. In some embodiments, administration is oral. [00222] The phrases“parenteral administration” and“administered parenterally” as used herein means modes of administration other than enteral and topical administration, usually by injection, and includes, without limitation, intravenous, intramuscular, intraarterial, intrathecal, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intraarticular, subcapsular, subarachnoid, intraspinal and intrastemal injection, and infusion.

[00223] The phrases“systemic administration”,“administered systemically”,“peripheral administration”, and“administered peripherally” as used herein mean the administration of a ligand, drug, or other material via route other than directly into the central nervous system, such that it enters the patient’s system and thus, is subject to metabolism and other like processes, for example, subcutaneous administration.

[00224] A compound provided herein may be administered to humans and other animals for therapy by any suitable route of administration, including orally, nasally, as by, for example, a spray, rectally, intravaginally, parenterally, intracistemally, and topically, as by powders, ointments or drops, including buccally and sublingually. Regardless of the route of administration selected, a compound provided herein, which may be used in a suitable hydrated form, and/or the pharmaceutical compositions provided herein, is formulated into a pharmaceutically acceptable dosage form by conventional methods known to those of skill in the art. In another embodiment, the pharmaceutical composition is an oral solution or a parenteral solution. Another embodiment is a freeze-dried preparation that can be reconstituted prior to administration. As a solid, this composition may also include tablets, capsules or powders.

[00225] Actual dosage levels of the active ingredients in the pharmaceutical compositions provided herein may be varied so as to obtain“therapeutically effective amount,” which is an amount of the active ingredient effective to achieve the desired therapeutic response for a particular patient, composition, and mode of administration, without being toxic to the patient.

[00226] The concentration of a compound provided herein in a pharmaceutically acceptable mixture will vary depending on several factors, including the dosage of the compound to be administered, the pharmacokinetic characteristics of the compound(s) employed, and the route of administration. In some embodiments, the compositions provided herein can be provided in an aqueous solution containing about 0.1-10% w/v of a compound disclosed herein, among other substances, for parenteral administration. Typical dose ranges can include from about 0.01 to about 50 mg/kg of body weight per day, given in 1-4 divided doses. Each divided dose may contain the same or different compounds. The dosage will be a therapeutically effective amount depending on several factors including the overall health of a patient, and the composition and route of administration of the selected compound(s).

[00227] Dosage forms or compositions containing a compound as described herein in the range of 0.005% to 100% with the balance made up from non-toxic carrier may be prepared. Methods for preparation of these compositions are known to those skilled in the art. The contemplated compositions may contain 0.001 %-100% active ingredient, in one embodiment 0.1-95%, in another embodiment 75-85%. Although the dosage will vary depending on the symptoms, age and body weight of the patient, the nature and severity of the disorder to be treated or prevented, the route of administration and the form of the drug, in general, a daily dosage of from 0.01 to 2000 mg of the compound is recommended for an adult human patient, and this may be administered in a single dose or in divided doses. The amount of active ingredient which can be combined with a carrier material to produce a single dosage form will generally be that amount of the compound which produces a therapeutic effect.

[00228] The pharmaceutical composition may be administered at once, or may be divided into a number of smaller doses to be administered at intervals of time. It is also noted that the dose of the compound can be varied over time. It is understood that the precise dosage and duration of treatment is a function of the disease being treated and may be determined empirically using known testing protocols or by extrapolation from in vivo or in vitro test data. It is to be noted that concentrations and dosage values may also vary with the severity of the condition to be alleviated. It is to be further understood that for any particular patient, specific dosage regimens should be adjusted over time according to the individual need and the professional judgment of the person administering or supervising the administration of the compositions, and that the concentration ranges set forth herein are exemplary only and are not intended to limit the scope or practice of the claimed compositions.

[00229] The precise time of administration and/or amount of the composition that will yield the most effective results in terms of efficacy of treatment in a given patient will depend upon the activity, pharmacokinetics, and bioavailability of a particular compound, physiological condition of the patient (including age, sex, disease type and stage, general physical condition, responsiveness to a given dosage, and type of medication), route of

administration, etc. However, the above guidelines can be used as the basis for fine-tuning the treatment, e.g., determining the optimum time and/or amount of administration, which will require no more than routine experimentation consisting of monitoring the patient and adjusting the dosage and/or timing.

[00230] The pharmaceutical compositions can be included in a container, pack, or dispenser together with instructions for administration. [00231] In jurisdictions that forbid the patenting of methods that are practiced on the human body, the meaning of“administering” of a composition to a human subject shall be restricted to prescribing a controlled substance that a human subject will self-administer by any technique (e.g., orally, inhalation, topical application, injection, insertion, etc.). The broadest reasonable interpretation that is consistent with laws or regulations defining patentable subject matter is intended. In jurisdictions that do not forbid the patenting of methods that are practiced on the human body, the“administering” of compositions includes both methods practiced on the human body and also the foregoing activities.

[00232] It is to be understood that while the disclosure is read in conjunction with the detailed description thereof, the foregoing description is intended to illustrate and not limit the scope of the disclosure, which is defined by the scope of the appended claims. Other aspects, advantages, and modifications are within the scope of the following claims.

EXAMPLES

[00233] The following examples are provided for illustration and are not intended to limit the scope of the disclosure in any way.

[00234] As used throughout these examples, common organic abbreviations are defined as follows:

Example 1 : Synthesis of Intermediate Compounds via Alkylation

Procedure 1 - Intermediate for A91

[00235] A 100 mL roundbottom flask with stir bar was charged with methyl 1 H-pyrrole-2- carboxylate (581 g, 4.65 mmol, 1.1 equiv), 4-(2-bromoethyl)pyridine (786 mg, 4.22 mmol, 1.0 equiv), K 2 CO 3 (1.75 g, 12.7 mmol, 3.0 equiv) and DMF (20 mL). The resulting reaction mixture was stirred at room temperature for 2 days. After this time, the reaction mixture was diluted with DCM (200 mL) and washed with water (3 x 200 mL). The organic layer was dried over Na2SC>4, filtered and concentrated in vacuo. The resulting crude material was purified via silica gel chromatography (50-100% EtOAc in hexanes) to yield the desired product.

[00236] The following intermediate compounds were synthesized in a similar manner:

Procedure 2 - Intermediate for A64

[00237] A roundbottom flask with stir bar was charged with NaH (210 mg, 60 wt%, 5.13 mmol, 1.2 equiv) and DMF (20 ml_). Methyl 1 H-pyrrole-2-carboxylate (535 mg, 4.27 mmol, 1.0 equiv) was slowly added to the reaction mixture, and the solution was stirred at room temperature for 30 min. After 30 min, (3-bromopropyl)benzene (936 mg, 4.70 mmol, 1.1 equiv) was added, and the reaction mixture was allowed to stir at room temperature overnight. The next morning, the reaction mixture was diluted with DCM (200 ml_) and washed with water (3 x 200 ml_). The organic layer was dried over Na2SC>4, filtered and concentrated in vacuo. The resulting crude material was purified via silica gel

chromatography (0-15% EtOAc in hexanes) to yield the desired product.

[00238] The following intermediate compounds were synthesized in a similar manner:

Procedure 3 - Intermediate for A90

[00239] A 40 mL scintillation vial with stir bar was charged with quinolin-4-ylmethanol (500 mg, 3.14 mmol, 1.1 equiv) and DMF (7 mL). Triethylamine (0.65 mL, 4.71 mmol, 1.5 equiv) was added to the reaction mixture, followed by the slow addition of mesyl chloride (0.269 mL, 3.45 mmol, 1.2 equiv) at room temperature. The reaction mixture was allowed to stir at room temperature overnight. The next morning, in a separate flask, a slurry of NaH (140 mg, 3.43 mmol, 60 wt%, 1.2 equiv) and DMF (7 mL) was prepared. Methyl 1 H-pyrrole-2- carboxylate (357 mg, 2.86 mmol, 1.0 equiv) was added, and the reaction mixture was allowed to stir at room temperature for 30 min. After 30 min, the solution of crude activated alcohol was slowly added to the pyrrole solution in four portions over 1 h. The resulting solution was allowed to stir at room temperature overnight. The next morning, the reaction mixture was diluted with DCM (150 mL) and washed with water (3 x 150 mL). The organic layer was dried over Na2SC>4, filtered and concentrated in vacuo. The resulting crude material was purified via silica gel chromatography (10-40% EtOAc in hexanes) to yield the desired product.

[00240] The following intermediate compounds were synthesized in a similar manner:

A36 methyl 1-((2,6-difluoropyridin-4-yl) ethyl)-1 H-pyrrole-2-carboxylate

Procedure 4 - Intermediate for A111

[00241] Methyl 4-formyl- 1 H-pyrrole-2-carboxylate (2 g, 13 mmol, 1 equiv) was dissolved in DMF (65 ml_), and CS2CO3 (6.37 g, 19 mmol, 1.5 equiv) and (bromomethyl)pyridine hydrobromide (3.3g, 13 mmol, 1.0 equiv) were added. The resulting reaction mixture was stirred at room temperature overnight. The next morning, the reaction mixture was diluted with water (20 ml_) and extracted with EtOAc (4 x 30ml_). The organic layer was dried over Mg2SC>4, filtered and concentrated in vacuo. The resulting crude product was purified via silica gel chromatography (25-100% EtOAc in hexanes) to yield the desired product.

[00242] The following intermediate compounds were synthesized in a similar manner:

Procedure 5 - Intermediate for A74 and A49

i ci

[00243] 2-(trichloroacetyl)pyrrole (1.0 g, 4.71 mmol, 1.0 eq), triphenylphosphine (0.527 g, 6.12 mmol, 1.3 equiv) and 1-(4-pyridinyl)ethanol (0.638 g, 5.18 mmol, 1.10 eq) were dissoled in anhydrous THF (15.8 ml_, 0.3 M). Then di-tertbutylazodicarboxylate (DBAD) (1.463 g, 6.35 mmol, 1.35 eq) dissolved in anhydrous THF (2 ml_) was added under argon atmosphere.

The reaction was allowed to stir at room temperature overnight. The next morning, the THF was evaporated under reduced preassure and the resulting crude material was purified via silica gel chromatography (0-100% EtOAc in DCM) to give the desired product. Procedure 6 - Intermediate for A93

[00244] Methyl 1 H-pyrrole-2-carboxylate (1 g, 8 mmol, 1 equiv) and 4- (hydroxymethyl)pyrrolidin-2-one (1.38 g, 12 mmol, 1.5 equiv) were dissolved in dry THF (16 ml_) followed by addition of CMBP (2.85g, 11.6 mmol, 1.45 equiv) under Ar. The tube was sealed and heated to 60°C overnight. The next morning, the THF was removed under reduced pressure and the crude mixture was purified via silica gel chromatography (1-5% methanol in DCM), which yielded the desired product mixed with tributylphosphine oxide. This mixture washed with hexane and filtered to provide the pure desired product.

Procedure 7 - Intermediate for B26

[00245] A 40 ml_ vial with stir bar was charged with methyl prolinate hydrochloride (500 mg, 3.02 mmol, 1.0 equiv) and DCM (15 ml_, 0.2 M). Triethylamine (1.7 ml_, 12.1 mmol, 4.0 equiv) was slowly added at room temperature. 4-(bromomethyl)pyridine hydrobromide (840 mg, 3.32 mmol, 1.1 equiv) was added portion-wise over 1 h. The resulting reaction mixture was allowed to stir at room temperature overnight. The next morning, the reaction mixture was concentrated in vacuo and the resulting crude product was purified via silica gel chromatography (50-100% EtOAc in hexanes) to yield the desired product.

Procedure 8 - Intermediate for A210

[00246] Methyl 1-((5-oxopyrrolidin-3-yl)methyl)-1 H-pyrrole-2-carboxylate (100 mg, 0.45 mmol, 1.0 equiv) was dissolved in THF, and cooled to 0 °C. NaH (21.6 mg, 0.54 mmol, 1.2 equiv) was added, and the reaction stirred at 0 °C for 15 min. After this time, Mel (127.8 mg, 0.9 mmol, 2.0 equiv) was added, and the reaction mixture was warmed to room temperature for 30 min. After 30 min at room temperature, the reaction was quenched with water (5 ml_) and concentrated in vacuo to yield the desired product.

Procedure 9 - Intermediate for A212 & A213

[00247] A 20 ml_ vial with stir bar was charged with bromide (461 mg, 1.56 mmol, 1.0 equiv), CS2CO3 (611 mg, 1.87 mmol, 1.2 equiv), and BrettPhos Pd G3 (142 mg, 0.156 mmol, 0.1 equiv). The vial was evacuated and backflushed with nitrogen. Freshly-sparged tBuOH (7 ml_) was added, followed by benzylamine (0.2 ml_, 1.87 mmol, 1.2 equiv). The vial was capped, and the reaction mixture was allowed to stir at 80 C overnight. The next morning, the reaction mixture was cooled to room temperature and diluted with DCM (100 ml_). The organic layer was washed with brine (2 x 100 ml_), and the combined aqueous layers were extracted with DCM (1 x 100 ml_). The combined organic layers were dried over Na2SC>4, filtered and concentrated in vacuo. The resulting material was purified by silica gel chromatography to yield the desired product.

Procedure 10 - Intermediate for A226 polymer supported PPh 3

[00248] A roundbottom flask with stir bar was charged with silyl ether (1.74 g, 6.14 mmol, 1.0 equiv) and THF (20 ml_). The reaction mixture was cooled to 0 C, and triethylamine trihydrofluoride (5.00 ml_, 30.7 mmol, 5.0 equiv) was added at 0 C. The reaction mixture was allowed to warm to room temperature overnight. The next morning, the reaction mixture was diluted with DCM (150 ml_) and washed with saturated NaHCCh (2 x 150 ml_). The combined aqueous layers were extracted with DCM (1 x 150 ml_), and the combined organic layers were dried over Na2SC>4, filtered and concentrated in vacuo. The resulting material was used in the next step without further purification. [00249] A roundbottom flask with stir bar was charged with alcohol (2.30 g, 13.6 mmol,

1.0 equiv), pyridin-4-ol (1.29 g, 13.6 mmol, 1.0 equiv), and polymer-bound PPh3 (3 mmol/g loading, 9.05 g, 27.2 mmol, 2.0 equiv) and THF (30 mL). The reaction mixture was cooled to 0 C. DIAD (5.34 mL, 27.2 mmol, 2.0 equiv) was slowly added at 0 C, and the reaction mixture was allowed to warm to room temperature overnight. The next morning, the reaction mixture filtered through a plug of Celite and washed with EtOAc. The filtrate was

concentrated in vacuo, and the resulting crude material was purified via silica gel chromatography to yield the desired product.

Example 2: Synthesis of Intermediate Compounds via Saponification

Procedure 1 - Intermediate for A6

[00250] A 40 mL vial with stir bar was charged with methyl 1-(3-(pyridin-4-yl)propyl)-1 H- pyrrole-2-carboxylate (559 g, 2.29 mmol, 1.0 equiv). MeOH (4 mL) and THF (4 mL) were added, followed by NaOH (1.6 mL, 5.0 M in water, 3.5 equiv). The resulting solution was heated at 60 °C overnight. The next morning, the solvents were removed in vacuo and the resulting aqueous solution was acidified to ~ pH 3 via addition of 1 M HCI. The resulting precipitate was filtered and collected, and any residual water was removed in vacuo to yield the desired product.

[00251] The following compounds were prepared in a similar manner:

Procedure 2 - Intermediate for A102

[00252] To the solution of methyl ester (0.210 g, 0.85 mmol, 1.0 eq.) in THF/water and few drops of MeOH was added LiOH monohydrate (0.177 g, 4.00 mmol, 5.0 eq.). Reaction was continued at 40°C for 18 hours. The next morning, the THF was evaporated, and the water layer was acidified to ~ pH 4-5 via addition of 1 M HCI. The water layer was then evaporated, and the crude product was purified by ionic resin (Amberlite IR 120) to give the desired product.

[00253] The following compounds were prepared in a similar manner:

Procedure 3 - Intermediate for A112

[00254] To a solution of methyl ester (0.175 g, 0.76 mmol, 1.0 eq) THF (5.8 ml_), potassium trimethylsiolate (0.194 g, 1.51 mmol, 2.0 eq) was added portion-wise. The reaction mixture was stirred at room temperature for 4 h. After this time, the precipitate was filtered, collected and dried under vacuum. The desired product was obtained as the potassium salt.

[00255] The following compounds were prepared in a similar manner:

Procedure 4 - Intermediates for A114 and A99

[00256] Synthesis of the intermediates for A114 and A99 followed the scheme below:

Step 1 : Nitro group reduction

[00257] 1-(4-nitrobenzyl)-1 H-pyrrole-2-carboxylic acid (0.200 g, 0.81 mmol, 1.0 eq.) was dissolved in methanol (10 ml_). Then, Pd/C (10 wt%, 0.414 g, 0.41 mmol, 0.5 eq.) was added and reaction was continued under hydrogen atmosphere overnight at room temperature.

The next morning, the reaction mixture was filtered through Celite and washed with EtOAc. The filtrate was concentrated to give the desired product, which was used in the next step without further purification.

Step 2: Aniline acylation

[00258] 1-(4-acetamidobenzyl)-1 H-pyrrole-2-carboxylic acid (0.150 g, 0.69 mmol, 1.0 eq.) was dissolved in ethanol (5 mL). Then, acetic anhydride (0.069 mL, 0.73 mmol, 1.05 eq.) was added, and reaction was stirred overnight at room temperature. The next morning, the volatile materials were evaporated. Water was added, and solution was acidified to pH 4 with 4N HCI. The resulting precipitate was filtered off and dried to yield the desired product (Intermediate for A114).

Step 3: Amide methylation

[00259] A solution of 1-(4-acetamidobenzyl)-1 H-pyrrole-2-carboxylic acid (0.163 g, 0,63 mmol, 1.0 eq) in THF (4 ml_) was added dropwise to a slurry of NaH (0.032 g, 0.79 mmol,

1.25 eq) in THF (4 ml_) at 0 °C. The mixture was stirred at this temperature for 30 min. After this time, Mel (0.098 ml, 1.58 mmol, 2.5 eq) was added dropwise to the reactinon mixture at 0 °C. The reaction mixture was allowed to warm to room temperature overnight. The next morning, the THF was evaporated, and the resulting material was diluted with water. The aqueous layer was extracted with EtOAc (3 x 30 ml_). The aqueous layer was then acidified to pH 4 with 4 N HCI, and the resulting precipitate was filtered off and dried to yield the desired product (Intermediate for A99)

Example 3: Synthesis of Thiazole Amine Intermediate Compounds

Procedure 1 - Intermediate for A41

[00260] A 100 mL roundbottom flask with stir bar was charged with 1 ,3-dibromo-3- methylbutan-2-one (1.95 g, 7.99 mmol, 1.0 equiv) and isopropanol (40 mL, 0.2 M). Thiourea (669 mg, 8.79 mmol, 1.1 equiv) was added, and the reaction mixture was stirred at room temperature overnight. The next morning, the solvent was evaporated, and the reaction mixture was diluted with DCM (100 mL). The organic layer was washed with saturated NaHCCh (3 x 100 mL), and subsequently dried over Na2SC>4. The organic layer was filtered and concentrated in vacuo. The resulting crude material was purified via silica gel chromatography (0-50% EtOAc in hexanes) to yield the desired product.

Procedure 2 - Intermediate for A39

[00261] A 40 ml_ vial with stir bar was charged with 1 ,3-dibromo-3-methylbutan-2-one (600 mg, 2.46 mmol, 1.0 equiv) and acetone (8 ml_, 0.2 M). (Tetrahydro-2H-pyran-2- yl)methanol (4.17 ml_, 36.9 mmol, 15 equiv) was added, followed by thiourea (206 mg, 2.71 mmol, 1.1 equiv), and the reaction mixture was stirred at room temperature overnight. The next morning, the reaction mixture was diluted with DCM (100 ml_). The organic layer was washed with saturated NaHCCh (3 x 100 ml_), and subsequently dried over Na2SC>4. The organic layer was filtered and concentrated in vacuo. The resulting crude material was purified via silica gel chromatography (40-80% EtOAc in hexanes) to yield the desired product.

Procedure 3 - Intermediate for A14

[00262] Synthesis of these intermediates followed the scheme below:

[00263] A 100 mL roundbottom flask was charged with benzyltriphenylphosphonium chloride (3.41 g, 8.76 mmol, 2.0 equiv) and THF (20 mL, 0.2 M). Potassium tert-butoxide (1.03 g, 9.20 mmol, 2.1 equiv) was added, and the reaction mixture was allowed to stir at room temperature for 15 min. Tert-butyl (4-formylthiazol-2-yl)carbamate (1.00 g, 4.38 mmol, 1.0 equiv) was then added, and the reaction mixture was allowed to stir at room temperature overnight. The next morning, the solvent was removed in vacuo, and the resulting material was taken up in DCM (100 mL). The organic layer was washed with saturated NhUCI (3 x 100 mL). The organic layer was then dried over Na2SC>4, filtered and concentrated in vacuo. The resulting crude material was purified via silica gel chromatography (0-10% EtOAc in hexanes) to yield the desired cis- and trans- isomers separately.

Step 2: Hydrogenation

[00264] A 20 mL scintillation vial with stir bar was charged with tert-butyl (4-styrylthiazol-2- yl)carbamate (187 mg, 0.618 mmol, 1.0 equiv) and Pd/C (10 wt%, 66 mg, 0.0618 mmol, 0.1 equiv). The solids were evacuated and backflushed with hydrogen (1 atm, 3x). Methanol (5 mL, 0.1 M) was added to the reaction mixture, and the resulting suspension was stirred at room temperature overnight. The next morning, the solids were filtered off, and the resulting crude material was purified via silica gel chromatography (0-30% EtOAc in hexanes) to yield the desired product.

Step 3: Boc Deprotection

[00265] A 20 mL scintillation vial with stir bar was charged with tert-butyl (4-styrylthiazol-2- yl)carbamate (176 mg, 0.582 mmol). DCM (2.7 mL) and TFA (600 uL, 20 vol%) were added, and the reaction mixture was stirred at room temperature overnight. The next morning, the reaction mixture was diluted with DCM (50 mL) and washed with saturated NaHCCh (3 x 50 mL). The organic layer was dried over Na2SC>4, filtered and concentrated in vacuo. The resulting product was used in the next step without further purification.

[00266] The following compounds were prepared in a similar manner:

Procedure 4 - Intermediate for A29

[00267] Synthesis of this intermediate followed the scheme, below:

Boc Boc

Step 1 : Boc Protection

Boc [00268] 1-(2-aminothiazol-4-yl)ethan-1-one (1.0 g, 2.06 mmol, 1 equiv) was suspended in neat B0C2O (1.79g, 8.24mmol, 4 equiv) at 60°C, then a catalytic amount of DMAP (0.024g; 0.02 mmol, 0.01 equiv) was added. Gas evolution started. The reaction mixture was left stirring overnight at 60 °C. The reaction mixture was concentrated and directly used into column chromatography. The resulting crude product was purified via silica gel

chromatography (10-50% EtOAc in hexanes) to yield the desired product.

Step 2: Fluorination

Boc Boc

[00269] DAST (0.456 g, 2.83 mmol, 3.0 equiv) was mixed with ketone (0.323 g, 0.943 mmol, 1.0 equiv) and left stirring at 50°C for 2 days. After this time, the reaction mixture was diluted with DCM (15 ml_), and carefully washed with NaHCC>3 (2 x 15 ml_). The organic layer was dried over MgSCU, filtered and concentrated in vacuo. The resulting crude product was used in the next step without further purification.

Step 3: Boc deprotection

Boc

[00270] Tert-butyl (tert-butoxycarbonyl)(4-(1 ,1-difluoroethyl)thiazol-2-yl)carbamate (0.38 g, 1.04 mmol, 1.0 equiv) was dissolved in HCI in dioxane (4M, 20 ml_) and heated to 50 °C overnight. The next morning, the volatile materials were removed under reduced pressure and the crude product was used in the next step without further purification.

Procedure 5 - Intermediate for A132

[00271] Synthesis of the intermediate for A132 followed the scheme, below:

Step 1 : Sonoqashira coupling

Pd(PPh 3 )2CI 2 (10 mol%)

Cul (20 mol%)

[00272] A solution of tert-Butyl (4-bromo-5-formylthiazol-2-yl)carbamate (0.500 g, 1.79 mmol, 1.0 equiv), phenylacetylene (0.573 g, 3.13 mmol, 1.75 equiv) and DABCO (1.005 g, 8.96 mmol, 5.0 equiv) in anhydrous THF (15.0 ml_) was purged three times with Ar (g), then Cul (0.003 g, 0.02 mmol, 0.01 equiv) and Pd(PPh3)2Cl2 (0.025 g, 0.04 mmol, 0.02 eq) were added. The reaction mixture was again purged with Ar (g) and then tube was closed and heated overnight at 80 °C. The next morning, the solvent was evaporated and the resulting crude product was purified via silica gel chromatography (0-2% EtOAc in hexanes) to yield the desired product.

Step 2: General Boc Deprotection Procedure*

[00273] Tert-butyl (4-(phenylethynyl)thiazol-2-yl)carbamate (0.269 g, 0.96 mmol, 1.0 equiv) was dissolved in DCM (8.1 ml_), and the solution was cooled to 0 °C. TFA (1.22 ml_, 9.64 mmol, 10.0 eq) was added at 0 °C. TLC showed no reaction progress so another portion of TFA (1.22 ml_, 9.64 mmol, 10.0 eq) was added after 2h. The reaction mixture was stirred at room temperature for the next 17 h. After this time, the reaction mixture was was cooled down to 0 °C, basified with NaHCCh sat. solution (50 ml_) and extracted with DCM (3x30 ml_). The organic phases were combined, dried over MgSCU, filtered and evaporated to dryness. Solid residues were washed with pentane and drop of DCM to obtain the desired product, which was used in the next reaction without further purification.

[00274] The following compounds were deprotected in a similar manner:

Procedure 6 - Intermediate for B22

[00275] Synthesis of intermediate for B22 followed the scheme, below

Step 1 : General Boc Protection Procedure ^

[00276] Methyl 2-(2-amino-5-methylthiazol-4-yl)acetate (1.0 g, 5.37 mmol, 1.0 eq.) was dissolved in DCM (10 ml_) , then di-tert-butyl dicarbonate (1.41 g, 6.44 mmol, 1.0 eq.) was added followed by TEA (0.90 ml_, 6.44 mmol, 1.2 eq.) and DMAP (0.171 g, 1.34 mmol, 0.25 eq.). The reaction was stirred at room temterature overnight. The next morning, the reaction was diluted with DCM (50 ml_) and washed with water (2x20 ml_). The organic layer was dried over MgSCU , filtered and evaporated. The resulting crude material was purified by FC eluting with hexanes/EtOAc to yield the desired product.

[00277] The following compound was protected in a similar manner: Step 2: Ester Reduction

Boc

[00278] LiBhU in THF (0.3 ml_, 2 N, 0.6 mmol, 0.6 eq) was added via syringe to a stirred solution of methyl 2-(2-((tert-butoxycarbonyl)amino)-5-methylthiazol-4-yl)aceta te (0.285 g,

1.0 mmol, 1.0 eq.) in anhydrous THF (2.0 ml_). The reaction was then slowly heated to reflux (initial exotherm). Reaction was continued under reflux for 16 h. After this time, the reaction was cooled down to 0°C and quenched with water (10 ml_). The aqueous layer was extracted with EtOAc (4 x 30 ml_) and the organic layer was dried over MgSCU, filtered and evaporated. The resulting crude product was purified by silica gel chromatography (EtOAc: hexane 1 :9 to 2:1) to yield the desired product.

Step 3: Mitsunobu Reaction

[00279] Di-tert-butyl azodicarboxylate (0.319 g, 1.38 mmol, 1.5 eq.) was added portion- wise to a solution of starting material (0.207g, 0.92 mmol, 1.0 eq.), phthalimide (0.163g; 1.11 mmol, 1.2 eq.), and PPh3 (0.363g, 1.38 mmol, 1.5 eq.) in Me-THF (10 ml_) at room temperature under N2 (g). The reaction mixture was stirred at room temperature overnight. The next morning, the solution was diluted with DCM (10 ml_) and washed with 10 percent aqueous solution of K 2 CO 3 (2 x 20 ml_). The organic layer was dried over MgSCU, filtered and evaporated to dryness. The resulting crude product was purified by silica gel chromatography (EtOAc: hexane 1 :9 to 1 :4) to yield the desired product.

Step 4: Phthalimide hydrolysis

[00280] Tert-butyl (4-(2-(1 ,3-dioxoisoindolin-2-yl)ethyl)-5-methylthiazol-2-yl)carbamat e (0.170 g, 1.0 eq.) was dissolved in 2 ml_ of EtOH. Then, hydrazine monohydrate (0.617 g, 30.0 eq.) was added at 0°C. Reaction was continued at room temperature for 17 hours. The resulting mixture was concentrated and then diluted with DCM (20 ml_) and washed with water (2 x 10 ml_). The organic layers were combined, dried over MgSCU, filtered and concentrated in vacuo. The resulting crude product was used in the next step without further purification.

Step 5: Acetylation

[00281] Tert-butyl (4-(2-aminoethyl)-5-methylthiazol-2-yl)carbamate (0.077 g, 0.30 mmol, 1.0 eq.) was dissolved in 2 ml_ of DCM. Then, TEA (0.125 ml_, 0.90 mmol, 3.0 eq.) was added, followed by acetyl chloride (0.032 ml_, 0.45 mmol, 1.5 eq.) at 0°C. The reaction was continued at room temperature for 17 hours. The resulting mixture was diluted with water (3 ml_) and extracted with DCM (3 x 10 ml_). The combined organic layers were dried over MgSCU, filtered and concentrated in vacuo. The resulting crude product was used to the next step without further purification.

Step 6: Boc Deprotection

[00282] See General Boc Deprotection Procedure, above.

Procedure 7 - Intermediate for A92

[00283] Synthesis of intermediate for A92 followed the scheme, below.

BOC 2 0 (1.0 equiv)

Step 1 : Boc Protection

[00284] See General Boc Protection Procedure, above. Step 2: DIBAL Reduction

[00285] Dried starting material (0.780 g, 2.72 mmol, 1.0 eq.) was dissolved in DCM (8 ml_) and diisobutylaluminum hydride (1 M in DCM, 8.17 ml_, 8.17 mmol, 3.0 eq.) was added at - 78°C to the solution. It was stirred for 5 h. After 5 h, the solution was quenched with methanol and 1 N HCI. After the temperature of the reaction mixture was elevated to room temperature, water was added (15 ml_) and extracted with DCM (3 x 20 ml_). The organic layer was dried over MgSCU , filtered and evaporated. The resulting crude product was used in the next step without further purification.

Procedure 8 - Intermediate for A96

[00286] Synthesis of intermediate for A96 followed the scheme, below

R = H, Me

[00287] To a stirred solution of 1-(ethanesulfonyl)piperazine (0.500g, 2.8 mmol, 1.0 equiv), in 7 ml_ of DCM at room temperature was added (4-formylthiazol-2-yl)carbamic acid tert-butyl ester (0.650g, 3.65 mmol, 1.3 equiv). The mixture was then treated with sodium triacetoxyborohydride (0.773g, 3.65 mmol, 1.3 equiv) and a catalytic amount of acetic acid. The reaction was allowed to stir for about 15 hours. The resulting mixture was diluted with NaHCCh (40 ml_) and extracted with DCM (3 x 30 ml_). The organic layers were combined, dried over MgSCU, filtered and concentrated in vacuo. The resulting crude material was purified via silica gel chromatography (0-100% EtOAc in hexanes) to give desired product.

[00288] The following compounds were prepared in a similar manner:

Step 2: Boc deprotection

[00289] See General Boc Deprotection Procedure, above.

Procedure 9 - Intermediate for A209

[00290] Synthesis of the intermediate for A209 was prepared according to the scheme, below:

[00291] To a solution of NaOH solid (0.569 g, 14.23 mmol, 1.0 eq.) and acetone (4.22 ml_, 56.9 mmol, 4.0 eq.) was added water and ethanol. The aldehyde (2.0 g, 14.23 mmol,

1.0 eq.) was added dropwise within 20 min at 0°C. The reaction mixture was allowed to warm to room temperature and was stirred. TLC analysis indicated completion of the reaction after 2 h. After this time, the reaction mixture was quenched with aqueous HCI (1 N), adjusted the pH to 6, and then evaporated to remove the residual ethanol. The residue was extracted by ethyl acetate (3 x 40 ml_). The combined organic phase was washed with brine (2 x 15 ml_), dried over MgSCU and concentrated under reduced pressure to yield the desired product, which was used without further purification in the next step. Step 2: Bromination

[00292] To a solution of enone (1.91 g, 10.57 mmol, 1.0 eq.) in 60 ml_ ACN:toluene (1 :1) was added methanesulfonic acid (1.72 ml_, 26.44 mmol, 2.5 eq.) and NBS (1.98g, 11.1 mmol, 1.05 eq.) and heated to 85°C for 6 hours. Sat. NaHCCh (70 ml_) and EtOAc (4 x 30 ml_) were added to the above mixture. The organic layer was separated, dried over magnesium sulfate and evaporated. The resulting crude material was purified via silica gel chromatography (0-25% EtOAc in hexanes) to yield the desired product.

Step 3: Thiazole condensation

[00293] Thiourea (0.442g, 5.81 mmol, 1.1 eq.) was dissolved in absolute ethanol (20 ml_). To this solution were added starting material (1.37g, 5.28 mmol, 1.0 eq.). The mixture was refluxed for 3 hours. After this time, the solution was cooled down, and the ethanol was evaporated. Water was added and the resulting solution was basified to pH 10. The aqueous layer was extracted with EtOAc (4 x 30 ml_), and the combined organic layers were dried over MgS0 4, filtered and evaporated. The resulting crude product was used in the next step without further purification.

Procedure 10 - Synthesis of Intermediate for A21

[00294] Synthesis of intermediate for A21 followed the scheme, below

Step 1 : Diazoketone formation

[00295] To a stirred solusion of 2-phenylpropionoic acid (1.0 g, 6.62 mmol, 1.0 eq) in DCM (20 ml_) at 0 °C were added 4 A molecular sieves and 1-chloro-N,N, 2-trimethyl- 1- propenylamine (0.99 ml_, 7.54 mmol, 1.14 eq). After 15 min, the reaction mixture was cooled to -20 °C and slowly added to a solution of TMS-diazomethane (3.47 ml_, 21.9 mmol, 2.9 eq) in DCM. The reaction mixture was allowed to warm to room temperature and stirred overnight. The reaction mixture was filtered on a sintered funnel and the filtrate was quenched by the addition of saturated NH 4 CI (30 ml_). The mixture was extracted with DCM (4 x 40 ml_)and the organic layers were separated, and washed with brine. The organic layer was then dried with MgS04, filtered and concentrated to dryness. The resulting crude material was used in the next step without further purification.

Step 2: Bromination

[00296] HBr (40% solusion in water, 45 ml_) was poured into the round-bottom flask containing crude diazoketone (1.5g, 8.56 mmol, 1.0 eq). The reaction mixture was allowed to be stirred at room temperature for 17 h. After this time, the reaction mixture was quenched with NaHCCh sat. solution (100 ml_). The mixture was extracted with DCM (4 x 30 ml_) and the organic layers were separated, washed with brine, dried with MgSCU filtered and concentrated to dryness. The resulting crude material was used without further purification. Step 3: Thiazole condensation

[00297] To a solution of crude bromoketone (1.03g , 4.52 mmol, 1.0 eq) in MeOH (10.3 ml_), thiourea (0.344g, 4.52 mmol, 1.0 eq) was added. The reaction mixture was allowed to be stirred overnight. LCMS analysis showed consumption of SM and formation of a new product. The next morning, the MeOH was evaporated and crude product was purified by preparatory HPLC (C18 column-prep Mobile phase: ACN + 0.1 % FA, H2O + 0.1 % FA) to yield the desired product.

Procedure 11A - Precursor for Intermediate for A307

[00298] Cul (24.36 mg, 0.243 mmol, 0.05 eq.), XantPhos (140 mg, 0.243 mmol, 0.05 eq.), and NaOtBu (46.6 mg, 0.485 mmol, 0.10 eq.) were mixed in a dry THF (20 ml_) in a flame dried Schlenk under argon. The reaction mixture was stirred at room temperature for 30 min. Then B2pin2 (1.35g, 5.34 mmol, 1.1 eq.) solution in dry THF (10 ml_) was added to mixture and stirred for further 10 min. Then alkyne (500 mg, 4.85 mmol, 1.0 eq.), MeOH (0.441 ml, 9.709 mmol, 2 eq.), and dry THF (5 ml_) were added successively. The resulting mixture was stirred for 18 h at room temperature, filtered through Celite and evaporated to dryness. The crude product was purified by column chromatography on silica (Hex/EtOAc) to yield pure product.

Procedure 11 B - Precursor for Intermediate for A157

CrCI 2 , Lil

[00299] Anhydrous CrCh (950 mg, 7.73 mmol, 8 eq.) was suspended in THF (10 ml_) under an argon atmosphere. A solution of aldehyde (150 mg, 0.96 mmol, 1.0 eq.) and dichloromethylboronic ester (407 mg, 1.93 mmol, 2 eq.) in THF (5 ml_) and a THF solution of Lil (517 mg, 3.86 mmol, 4 eq.) were added at 25 °C to the suspension successively. After being stirred at 25 °C for 16 h, the reaction mixture was poured into water (25 mL) and extracted with ether (3x10 ml_). The combined extracts were dried over Na2SC>4 and concentrated. Crude product was pure enough to be used in the next step.

[00300] The following compounds were prepared via a similar method:

Procedure 11C - Precursor for Intermediate for A245

[00301] Aryl bromide (1.39 g, 6.09 mmol, 1.0 eq), vinyl boronic pinacol ester (1.06 ml, 6.09 mmol, 1.0 eq) and TEA (1.69 ml, 12.19 mmol, 2.0 eq) were dissolved in toluene (24.5 ml_). The mixture was purged with argon for 5 min. After that time Pd(tBu3P)2 (0.156 g, 0.30 mmol, 0.05 eq) was added, and the reaction mixture was purged for 2 min with argon. The pressure vessel was closed, and the reaction was heated at 90 C for two hours. LCMS analysis showed consumption of starting material and formation of desired product. The reaction was cooled down to the room temperature and filtered through a plug of Celite. The filtrate was evaporated, and the desired product was recrystallized from Et2<D and washed with pentane. In case of necessity, additional purification by flash chromatography on silica (Hex/EtOAc) was performed.

[00302] The following compounds were prepared via a similar method:

Procedure 11 D - Precursor for Intermediate for A343

was charged with bromide (1.3 g, 5.9 mmol, 1.0 equiv) and Pd(dtfp)Cl2 (380 mg, 0.59 mmol, 0.1 equiv). The vial was evacuated and backflushed with nitrogen. Freshly-sparged toluene (5 ml_) was added, followed by vinyl pinacol boronic ester (3.5 ml_, 21 mmol, 3.5 equiv) and NEt3 (1.6 ml_, 12 mmol, 2.0 equiv). The vial was capped, and the reaction mixture was allowed to stir at 100 C overnight. The next morning, the reaction mixture was cooled to room temperature and diluted with EtOAc (100 ml_). The organic layer was washed with brine (2 x 100 ml_), and the combined aqueous layers were extracted with EtOAc (1 x 100 ml_). The combined organic layers were dried over Na 2 S0 4 , filtered and concentrated in vacuo. The resulting crude material was purified by either silca gel chromatography or RP-silica gel chromatography (C18) to yield the desired product.

[00304] The following compounds were prepared via a similar method:

Procedure 11E Intermediate for A248 (Route A)

[00305] The reactions followed the general reaction scheme, below:

Step 1A: Suzuki Coupling Procedure

[00306] A vial with stir bar was charged with bromide (388 mg, 1.39 mmol, 1.0 equiv), boronic ester (437 mg, 1.53 mmol, 1.1 equiv), potassium phosphate (589 mg, 2.78 mmol, 2.0 equiv) and Pd(PPh3)2Cl2 (97.4 mg, 0.139 mmol, 0.1 equiv). The vial was evacuated and backflushed with nitrogen. 15% water in DMF (sparged with nitrogen for 1 h, 4 ml_, 0.4 M) was added. The vial was capped, and the reaction mixture was stirred at 100 C overnight. The next morning, the reaction mixture was poured into EtOAc (50 ml_) and washed with 1 : 1 waterbrine (2 x 50 ml_). The combined aqueous layers were extracted with EtOAc (1 x 50 ml_). The combined organic layers were dried over Na 2 S0 4 , filtered and concentrated in vacuo. The resulting crude material was purified by silica gel chromatography to yield the desired product.

[00307] The following compounds were prepared via the same method:

Step 1A: Reverse Suzuki Coupling

[00308] A vial with stir bar was charged with bromide (51.7 g, 0.325 mmol, 1.0 equiv), boronic ester (126 mg, 0.358 mmol, 1.1 equiv), KsP0 4 (138 mg, 0.650 mmol, 2.0 equiv) and Pd(PPh3)2Cl2 (22.8 mg, 0.0325 mmol, 0.1 equiv). The vial was evacuated and backflushed with nitrogen. Freshly-sparged 15% water in DMF (2 ml_) was added, and the vial was capped. The reaction mixture was stirred at 100 C for 2 h. After 2 h, the reaction mixture was cooled to room temperature and poured into EtOAc (50 ml_). The resulting solution was washed with brine (2 x 50 ml_), and the combined aqueous layers were extracted with EtOAc (1 x 50 ml_). The combined organic layers were dried over Na2SC>4, filtered and

concentrated in vacuo. The resulting crude material was purified via silica gel

chromatography.

[00309] The following compounds were prepared via a similar method:

Step 2: Boc Deprotection

[00310] A 20 ml_ vial with stir bar was charged with carbamate (270 mg, 0.753 mmol, 1.0 equiv) and DCM (4 ml_). TFA (1 ml_, 13 mmol, 17 equiv) was added, and the reaction mixture was stirred at room temperature overnight. The next morning, the reaction mixture was poured into DCM (50 ml_) and washed with saturated NaHCCh (2 x 50 ml_). The combined aqueous layers were extracted with DCM (1 x 50 ml_), and the combined organic layers were dried over Na2SC>4, filtered and concentrated in vacuo. The resulting material was used in the next step without further purification.

[00311] The following compounds were prepared via a similar method:

Procedure 12A - Precursor for Intermediate for A289

[00312] A 100 mL roundbottom flask with stir bar was charged with D-proline ester (1.00 g, 7.7 mmol, 1.00 equiv), iodobenzene (1.90 g, 9.3 mmol, 1.2 equiv), 2-(2- methylpropanoyl)cyclohexan-1-one (521 mg, 3.1 mmol, 0.4 equiv), CS2CO3 (7.57 g, 23.227 mmol, 3 equiv), Cul (147.45 mg, 0.774 mmol, 0.1 equiv) and DMF (30 mL, 0.26 M) under nitrogen atmosphere, the vial was capped and placed in an 50°C bath. The reaction mixture was stirred at 50 °C overnight. The next morning, the reaction mixture was poured into DCM (200 mL) and washed with brine (3 x 100 ml_). The organic layer was dried over Na2SC>4, filtered and concentrated in vacuo. The resulting crude material was purified via silica gel chromatography to yield the desired product.

[00313] The following compounds were prepared via a similar method:

Procedure 12B - Precursor for Intermediate for A359

[00314] A 40 mL vial with stir bar was charged with D-proline (1.43 g, 12.4 mmol, 2.5 equiv), aryl bromide (1.00 g, 4.97 mmol, 1.0 equiv), Cul (189 mg, 0.994 mmol, 0.2 equiv) and K 3 PO 4 (4.22 g, 19.9 mmol, 4.0 equiv). The vial was evacuated and backflushed with nitrogen. Freshly-sparged DMSO (6 mL) was added. The vial was capped, and the reaction mixture was allowed to stir at 100 C overnight. The next morning, the reaction mixture was cooled to 60 C and diluted with DMF (10 mL). Mel (1.55 mL, 24.8 mmol, 5.0 equiv) was added, and the reaction mixture was allowed to stir at 60 C for 2 h. After 2 h, the reaction mixture was poured into EtOAc (150 mL) and washed with brine (2 x 150 mL). The combined aqueous layers were extracted with EtOAc (1 x 50 mL). The combined organic layers were dried over Na 2 S0 4 , filtered and concentrated in vacuo. The resulting crude material was purified via silica gel chromatography to yield the desired product.

[00315] The following compounds were prepared via a similar method:

Procedure 12C - Precursor for Intermediate for A319

[00316] A 100 mL roundbottom flask with stir bar was charged with D-proline ester (500 mg, 3.2 mmol, 1.00 equiv), iodobenzene (649 mg, 3.181 mmol, 1.00 equiv), methyl[2- (methylamino)ethyl]amine (280 mg, 3.2 mmol, 1 equiv), Cul (303 mg, 1.6 mmol, 0.5 equiv), CS2CO3 (2.59 g, 8.0 mmol, 2.5 equiv) and dioxane (20 mL, 0.16 M) under nitrogen atmosphere, the vial was capped and placed in an 25°C bath. The reaction mixture was stirred at 25°C overnight. The next morning, the reaction mixture was poured into DCM (80 mL) and washed with brine (2 x 40 mL). The organic layer was dried over Na2SC>4, filtered and concentrated in vacuo. The resulting crude material was purified via silica gel chromatography to yield the desired product.

Procedure 12D - Procedure for Intermediate for A279

[00317] A 100 mL roundbottom flask with stir bar was charged with pyrrolidine ester (1.50 g, 11.6 mmol, 1.00 equiv), TEA (3.53 g, 34 mmol, 3.00 equiv) and DCM (20 mL, 0.58 M). Benzyl bromide (2.38 g, 14.0 mmol, 1.20 equiv) was added, and the vial was capped and placed in a 25°C bath. The reaction mixture was stirred at 25°C for 2 h. The reaction mixture was poured into DCM (50 mL) and washed with H2O (3 x 50 mL). The organic layer was dried over Na2SC>4, filtered and concentrated in vacuo. The resulting crude material was purified via silica gel chromatography to yield the desired product.

[00318] The following compounds were prepared via a similar method:

Procedure 12E - Precursor for Intermediate for A320

[00319] A 50 mL roundbottom flask with stir bar was charged with D-proline ester (880 mg, 6.1 mmol, 1.00 equiv), benzyl bromide (1.15 g, 6.7 mmol, 1.09 equiv) and THF (12 mL, 0.51 M). NaH (60%, 369 mg, 9.2 mmol, 1.5 equiv) was added in portions at 0°C. The vial was capped and placed in an 25°C bath. The reaction mixture was stirred at 25°C for 5h. The reaction mixture was quenched by NH4CI (aq) (25 mL). The resulting solution was extracted with ethyl acetate (3 x 30 mL) and washed with brine (2 x 30 mL). The combined organic layers were dried over Na2SC>4, filtered and concentrated in vacuo. The resulting crude material was purified via silica gel chromatography to yield the desired product.

Procedure 12F - Precursor for Intermediate for A323

[00320] A 100 mL roundbottom flask with stir bar was charged with pyrrolidine ester (2.00 g, 4.4 mmol, 1.00 equiv), phenyl boronic acid (1.59 g, 13.0 mmol, 3 equiv), Cu(OAc)2 (2.37 g, 13.0 mmol, 3 equiv), TEA (2.20 g, 21.8 mmol, 5 equiv) and DCM (30 mL, 0.15 M) under nitrogen atmosphere. The reaction flask was then vacuumed and flushed with oxygen, and the sequence was repeated twice. The vial was capped and placed in a 25°C bath. The reaction mixture was stirred at 25°C for 48 h under oxygen atmosphere using an oxygen balloon. The reaction mixture was poured into DCM (150 mL) and quenched by the addition of NH3.H2O (20 mL), washed with H2O (1 x 50 mL) and brine (3 x 50 mL). The organic layer was dried over Na2SC>4, filtered and concentrated in vacuo. The resulting crude material was purified via silica gel chromatography to yield the desired product.

[00321] The following compounds were prepared via a similar method:

Procedure 12G - Precursor for Intermediate for A285

Pd(OAc) 2 (10 mol%)

[00322] A vial with stir bar was charged with amine (1.4 g, 9.7 mmol, 1.2 equiv), CS2CO3 (3.2 g, 9.7 mmol, 1.2 equiv), Rά(OA (180 mg, 0.81 mmol, 0.1 equiv), and BINAP (510 mg, 0.81 mmol, 0.1 equiv). The vial was evacuated and backflushed with nitrogen. Freshly sparged toluene (15 ml_) was added, followed by bromobenzene (0.85 ml_, 8.1 mmol, 1.0 equiv). The reaction mixture was capped and allowed to stir at 100 C overnight. The next morning, the reaction mixture was cooled to room temperature and poured into EtOAc (100 ml_). The organic layer was washed with brine (2 x 100 ml_) and the combined aqueous layers were extracted with EtOAc (1 x 100 ml_). The combined organic layers were dried over Na 2 S0 4 , filtered and concentrated in vacuo. The resulting crude material was purified via silica gel chromatography to yield the desired product.

[00323] The following compounds were prepared via a similar method:

Procedure 12H - Precursor for Intermediate for A322

[00324] A 250 ml_ roundbottom flask with stir bar was charged with pyrrolidine ester (2.40 g, 6.257 mmol, 1.00 equiv), iodobenzene (1.91 g, 9.362 mmol, 1.50 equiv), CS2CO3 (6.10 g, 18.722 mmol, 2.99 equiv), XPhOS (595.00 mg, 1.248 mmol, 0.20 equiv), XPhOS Pd G3 (1.06 g, 1.252 mmol, 0.20 equiv) and dioxane (20 ml_, 0.16 M) under nitrogen atmosphere, the vial was capped and placed in an 90°C bath. The reaction mixture was stirred at 90°C overnight. The next morning, the reaction mixture was cooled to room temperature and quenched by H2O (50 ml_). The resulting solution was extracted with (3 x 50 ml_) of ethyl acetate. The combined organic layers were dried over Na2SC>4, filtered and concentrated in vacuo. The resulting crude material was purified via prep-TLC to yield the desired product. [00325] The following compounds were prepared via a similar method:

Procedure 121 - Precursor for Intermediate for A365

[00326] A vial with stir bar was charged with methyl D-prolinate hydrochloride (100 g, 0.604 mmol, 1.0 equiv), DIPEA (0.1 1 ml_, 0.604 mmol, 1.0 equiv), cyclohexanone (62.6 uL, 0.604 mmol, 1.0 equiv) and DMF (1 ml_). The reaction mixture was cooled to 0 C.

Na(OAc)3BH () was added, and the reaction mixture was allowed to warm to room

temperature overnight. The next morning, the reaction mixture was diluted with EtOAc (50 ml_) and washed with saturated NaHCCh (3 x 50 ml_). The combined aqueous layers were extracted with EtOAc (1 x 50 ml_), and the combined organic layers were dried over Na 2 S0 4 , filtered and concentrated in vacuo. The resulting material was used without further purification in the next step.

Procedure 12J - Precursor for Intermediate for A313

[00327] A 250 ml_ roundbottom flask with stir bar was charged with pyrrolidine ester (4.00 g, 24 mmol, 1.00 equiv), TEA (8.40 ml_, 60 mmol, 2.50 equiv) and DCM (100 ml_, 0.24 M). Benzoyl chloride (2.8 ml_, 24 mmol, 1.01 eq) was added, and the vial was capped and placed in a 0°C bath. The reaction mixture was stirred at 25°C overnight. The next morning, the reaction mixture was poured into DCM (50 ml_) and washed with H2O (2 x 70 ml_). The organic layer was dried over Na 2 S0 4 , filtered and concentrated in vacuo. The resulting crude material was purified via silica gel chromatography to yield the desired product. Procedure 12K - Synthesis of Intermediates via Halogenation (e.g., for A163, A154, A158, & A292) (Route B)

[00328] The reactions followed the general reaction scheme, below:

[00329] In accordance with this procedure, compounds having a single stereocenter were prepared and evaluated as a single enantiomer. Unless indicated otherwise, compounds having two stereocenters were prepared and evaluated as a single diastereomer, but as a mixture of enantiomers. Each of compounds A321-A323, A325, A326, A333, A334, A344- A346, and A351-A354 (having two stereocenters) where prepared and evaluated as pure diastereomers and pure enantiomers.

Step 1A: Route B-1 (Bromoketone Synthesis)

[00330] Route B-1 followed the general reaction scheme, below:

Condensation Procedure A

NaOH (1.0eq)

Water (10.0eq)

[00331] Solid NaOH (0.356g, 8.89mmol, 1.0eq) was dissolved in the mixture of acetone (2.63ml_, 35.57mmol, 4.0eq), water (12.5ml_) and EtOH (6.3mL). Then the 4- chlorobenzaldehyde (1.250g, 8.89mmol, 1.0eq) was added dropwise within 20 min at 0 °C. The reaction mixture was allowed to warm to room temperature and was stirred until TLC analysis indicated completion of the reaction (0.5-2 h). The reaction mixture was quenched with aqueous HCI (1 N), adjusted the pH to 6 and evaporated to remove the residual EtOH and acetone. The residue was extracted by EtOAc (3 c 40 ml_). The combined organic phases were washed with brine (50 ml_), dried over anhydrous Na 2 S0 4 and concentrated under reduced pressure. Crude product was purified by column chromatography on silica gel. [00332] The following compounds were prepared via a similar method:

Condensation Procedure B

[00333] Diethyl (2-oxopropyl)-phosphonate (1.34 mL, 6.97 mmol, 1.5 equiv) was added to a slurry of NaH (250 mg of a 60 % mineral oil suspension, 6.51 mmol, 1.4 equiv) in THF (10.3 mL) at 0°C, and stirred for 1 hr. Aldehyde (0.5ml, 4.65 mmol, 1.0 equiv) was added dropwise, the cooling bath was removed and the resulting solution stirred for an additional 1.5 hr at room temperature. H O (10 mL) was added and the layers were separated. The aqueous phase was extracted with Et <D (3 x 10 mL), and the combined organic extracts were dried (MgSCU) and concentrated under reduced pressure. The crude residue was purified by flash chromatography.

[00334] The following compounds were prepared via a similar method:

Bromination

[00335] Ketone (730mg, 4.15 mmol, 1.0 equiv) was dissolved in dry THF (10ml) followed by slow (1h) dropwise addition of pyrrolidone hydrotribromide (2468 mg, 4.98 mmol, 1.2 equiv) in THF (15 ml). Reaction was stirred overnight at rt, the solid residue was filtered off and filtrate was evaporated. The oily residue was dissolved in Et2<D (50ml), washed with saturated NaHCCh (20ml), water (20ml) and brine (20ml). Organic layer was separated, dried by MgS04 and evaporated to yield crude product which was further purified by flash chromatography on silica gel.

[00336] The following compounds were prepared via a similar method:

Step 1 B: Route B-2 (Chloroketone Synthesis)

[00337] Route B-2 followed the general reaction scheme, below:

Chloroketone

Procedure A or

Procedure B

Chloroketone Procedure A chloroiodomethane (4 equiv)

LDA (5 equiv)

[00338] A roundbottom flask with stir bar was charged with starting material (0.50 g, 2.4 5mmol, 1.0 equiv) dissolved in THF (6.1 ml_) under Ar atmosphere. Then to reaction mixture was added chloroiodomethane (0.71 ml_, 9.79 mmol, 4.0 equiv). When the flask was cooled to -72°C, solution of LDA (12.24 ml_, 12.24 mmol, 5.0 equiv, 1 M in THF) was added dropwise maintaining the temperature below -68°C. Upon completion of the addition, the solution was stirred at -72°C for 15 min. An acetic acid solution (1.4 ml_ in 1.4 ml_ of THF) was added dropwise at a rate sufficient to keep the internal temperature below -60°C and the reaction mixture was stirred for 1 hour while flask was slowly allowed to warm. Then toluene (6 ml_) was added and when temperature reached -20°C, a portion of water (15 ml_) was slowly added and the solution was stirred for 20 minutes. Layers were separated and the organic layer was washed with NaHCCh (2 x 100 ml), dried over MgSCL, filtered and concentrated. The crude product was purified by flash chromatography on silica gel using Hex/EtOAc as eluent.

[00339] The following compounds were prepared in a similar way:

Chloroketone Procedure B

tBuMgCI (3 equiv)

NEt 3 (1 equiv)

O I

Boc

[00340] A 20 mL vial with stir bar was charged with methyl ester (182 g, 0.887 mmol,

1.0 equiv), sodium 2-chloroacetate (155 mg, 1.33 mmol, 1.5 equiv), triethylamine (0.124 mL, 0.887 mmol, 1.0 equiv) and THF (1.0 mL, 0.2 M). The reaction mixture was cooled to 0 C, and tert-butylmagnesium chloride (1.0 M in THF, 2.7 mL, 2.70 mmol, 3.0 equiv) was added.

The reaction mixture was allowed to warm to room temperature overnight. The next morning, the reaction mixture was poured into EtOAc (50 mL). The organic layer was washed with saturated NaHCCh (2 x 50 mL). The combined aqueous layers were extracted with EtOAc (1 x 50 mL), and the combined organic layers were dried over Na 2 S0 4 , filtered and concentrated in vacuo. The resulting crude material was used in the next step without further purification.

[00341] The following compounds were prepared in a similar way:

Step 2: Standard Condensation

O thiourea H 2 N ^

B OU>" Ph EtOH, 80 C S ^>'- ph

[00342] A 20 mL vial with stir bar was charged with bromoketone (307 mg, 1.28 mmol, 1.0 equiv), thiourea (108 mg, 1.41 mmol, 1.1 equiv) and EtOH (7 mL). The resulting solution was stirred at 80 C overnight. The next morning, the resulting solution was cooled and poured into EtOAc (100 ml_). The mixture was washed with saturated NaHCCh (2 x 100 ml_), and the combined aqueous layers were extracted with EtOAc (1 x 100 ml_). The combined organic layers were dried over Na2S04, filtered and concentrated in vacuo. The resulting crude material was purified via silica gel chromatography to yield the desired product.

[00343] The following compounds were prepared via a similar method:

Procedure 13 - Intermediate for A267 (Route C)

[00344] The reactions followed the general reaction scheme, below:

[00345] A 50 ml_ vial with stir bar was charged with thiazole (300 g, 1.08 mmol, 1.0 equiv), amine (869 mg, 5.39 mmol, 5.0 equiv) and DMF (9.0 ml_, 0.12 M) under nitrogen atmosphere, the vial was capped and placed in an 100°C bath. The reaction mixture was stirred at 100°C for 4 h. The reaction mixture was cooled to room temperature and quenched by H2O (50 ml_). The resulting solution was extracted with ethyl acetate (3 x 40 ml_) and washed with brine (3 x 40 ml_). The organic layer was then dried over Na2SC>4, filtered and concentrated in vacuo. The resulting crude material was purified via prep-TLC to yield the desired product.

[00346] The following compounds were prepared via a similar method:

Step 2: Deprotection

[00347] A 20 ml_ vial with stir bar was charged with thiazole (90 mg, 0.25 mmol, 1.00 equiv) and DCM (2.0 ml_, 0.125 M). TFA (2 ml_, 27.0 mmol, 108 equiv) was added, and the vial was capped and placed in a 25°C bath. The reaction mixture was stirred at 25°C for 1 h. The resulting mixture was concentrated under vacuum and quenched by sat. NaHCCh (aq) (10 ml_). The combined aqueous layers were extracted with EtOAc (4 x 20 ml_), the combined organic layers were dried over Na2SC>4, filtered and concentrated in vacuo. The resulting crude material was used directly for next step.

[00348] The following compounds were prepared via a similar method:

Example 4: Amide Coupling Reactions

Procedure 1 - Synthesis of Compounds A74 and A49

[00349] Synthesis of A74 and A49 followed the scheme, below.

[00350] N-(2-(4-pyridinylethyl)-2-(trichloroacetyl)pyrrole (0.53 g, 1.67 mmol, 1.0 eq), amine (0.334 g, 1.67 mmol, 1.0 eq) and NaHCCh (0.911 g, 10.9 mmol, 6. 5eq) were taken up in NMP (6.5 ml_). The reaction mixture was heated at 150 °C for 17h. After this time, the NMP was evaporated, and the resulting crude material was purified on pTLC (60% EtOAc in hexanes). LCMS (+ESI): calc. [M+H] + = 396; found 397. The enantiomers were resolved via SFC (Chiralpak IF 3, 5% MeOH+TEA in C0 2 ).

Procedure 2 - Synthesis of Compound A71

[00351] Synthesis of A71 followed the scheme, below

[00352] A 20 mL vial with stir bar was charged with acid (71.3 mg, 0.341 mmol, 1.0 equiv), amine (115 mg, 0.574 mmol, 1.7 equiv), sodium bicarbonate (115 mg, 1.36 mmol,

4.0 equiv) and HATU (143 mg, 0.375 mmol, 1.1 equiv). DMF (2 mL) was added, and the vial was capped and placed in an 80 °C bath. The reaction mixture was stirred at 80 °C overnight. The next morning, the reaction mixture was poured into DCM (50 mL) and washed with 0.5 M NaOH (2 x 50 mL), followed by water (1 x 50 mL). The organic layer was then dried over Na2SC>4, filtered and concentrated in vacuo. The resulting crude material was purified via silica gel chromatography (20-50% EtOAc in hexanes) to yield the desired product. LCMS (+ESI): calc. [M+H] + = 392; found 392.

[00353] The following compounds were prepared in a similar manner:

Procedure 3 - Synthesis of Compound A103

[00354] Synthesis of A103 followed the scheme, below:

[00355] To the solution of potassium salt of acid (0.145 g, 0.57 mmol, 1.0 equiv), DIPEA (0.395 ml_, 2.27 mmol, 4.0 equiv) and HATU (0.266 g, 0.70 mmol, 1.23 equiv) in DMF (2.9 ml_) were added. After 15 min, amine (0.114 g, 0.57 mmol, 1.0 equiv) was added. The reaction was allowed to stir at 80 °C overnight. The next morning, the reaction mixture was diluted wth EtOAc (50 ml_) and washed with saturated NaHCCh (2 x 50 ml_), followed by water (1 x 50 ml_). The organic layer was then dried over MgSCU, filtered and concentrated in vacuo. The crude product was purified via silica gel chromatography eluting with EtOAc. LCMS (+ESI): calc. [M+H] + = 400; found 400.

[00356] The following compounds were prepared in a similar manner:

Procedure 4 - Synthesis of Compound A97

[00357] Synthesis of A97 followed the scheme, below:

[00358] To the solution of the acid (0.090 g, 0.39 mmol, 1.0 equiv) in NMP (1 ml_) were added HATU (0.163 g, 0.43 mmol, 1.1 equiv), DIPEA (0.272 ml_, 1.56 mmol, 4.0 equiv) and amine (0.117 g, 0.59 mmol, 1.5 eq.). Reaction was continued either with conventional heating or in the microwave at 130°C for 1 h. After that the reaction mixture was diluted with EtOAc (50 ml_) and washed with saturated NaHCCh (2 x 50 ml_), followed by water (1 x 50 ml_). The organic layer was then dried over MgSCU, filtered and concentrated in vacuo. Crude product was purified via silica gel chromatography (1-10% MeOH in DCM). LCMS (+ESI): calc. [M+H] + = 413; found 413.

[00359] The following compounds were prepared in a similar manner:

Procedure 5 - Synthesis of Compound A281

[00360] A 20 mL vial with stir bar was charged with acid (100 mg, 0.495 mmol, 1.1 equiv), amine (97.2 mg, 0.450 mmol, 1.0 equiv), BTFFH (156 mg, 0.495 mmol, 1.1 equiv) and DMF (1.0 mL, 0.4 M). DIPEA (160 uL, 0.899 mmol, 2.0 equiv) was added, and the vial was capped and placed in a 100 C bath. The reaction mixture was stirred at 100 C overnight.

The next morning, the reaction mixture was poured into EtOAc (50 mL) and washed with 1 :1 1 M NaOFLbrine (2 x 50 mL). The combined aqueous layers were extracted with EtOAc (1 x 50 mL), and the combined organic layers were dried over Na2S04, filtered and concentrated in vacuo. The resulting crude material was purified via silica gel chromatography to yield the desired product.

[00361] The following compounds were prepared via a similar method:

Procedure 7 - Synthesis of Compound A309

[00362] A 20 mL microwave vial (G30) with stir bar was charged with acid (109.95 g, 0.544 mmol, 1.3 equiv), amine (116 mg, 0.418 mmol, 1.0 equiv), BTFFH (224.83 mg, 0.711 mmol, 1.7 equiv) and DMF (8.0 mL, 0.052 M). DIPEA (0.474 mL, 2.719 mmol, 6.5 equiv) was added, and the vial was capped and placed in a microwave reactor at 150 C. The reaction mixture was stirred at 150 C for 1 h. The reaction mixture was poured into EtOAc (50 mL) and washed with 1 :1 a saturated NaHCCh: brine (2 x 20 mL). The combined aqueous layers were extracted with EtOAc (1 x 50 mL), and the combined organic layers were dried over Na 2 S0 4 , filtered and concentrated in vacuo. The resulting crude material was purified via silica gel chromatography to yield the desired product.

[00363] The following compounds were prepared via a similar method:

Procedure 8 - Synthesis of Compound A251

[00364] A 25 mL vial with stir bar was charged with acid (110.00 mg, 0.544 mmol, 1.00 equiv), amine (162.80 mg, 0.707 mmol, 1.30 equiv), NMI (156.30 mg, 1.904 mmol, 3.50 equiv) and ACN (4.0 mL, 0.14 M). TCFH (184.60 mg, 0.660 mmol, 1.21 equiv) was added, and the vial was capped and placed in a 25°C bath. The reaction mixture was stirred at 25°C overnight. The next morning, the reaction mixture was poured into EtOAc (30 mL) and washed with brine (2 x 30 mL). The combined aqueous layers were extracted with EtOAc (1 x 30 mL), and the combined organic layers were dried over Na 2 S0 4 , filtered and

concentrated in vacuo. The resulting crude material was purified via silica gel

chromatography & Prep-HPLC or RP column to yield the desired product.

[00365] The following compounds were prepared via a similar method:

Procedure 9 - Synthesis of A261

[00366] A 25 mL vial with stir bar was charged with acid (54.60 mg, 0.27 mmol, 1.00 equiv), EDCI (77.70 mg, 0.41 mmol, 1.50 equiv), DIEA (104.60 mg, 0.81 mmol, 3.00 equiv), DMAP (10.00 mg, 0.08 mmol, 0.30 equiv) and DCM (4.0 mL, 0.068 M). Amine (70.00 mg, 0.27 mmol, 1.00 equiv) was added, and the vial was capped and placed in a 25°C bath. The reaction mixture was stirred at 25°C for 4 h. The resulting solution was concentrated in vacuo. The resulting crude material was purified via silica gel chromatography & Prep-HPLC or RP column to yield the desired product.

[00367] The following compounds were prepared via a similar method:

Procedure 10 - Synthesis of Intermediate for A141-A144, A164, A229, & A237

[00368] A vial with stir bar was chaged with amine (1.0 g, 5.59 mmol, 1.0 equiv) and DCM

(10 mL). BTFFH (2.7 g, 8.4 mmol, 1.5 equiv), DIPEA (4.4 mL, 25 mmol, 4.5 equiv) and acid (1.5 g, 7.26 mmol, 1.1 equiv) were added. The reaction mixture was sealed and stirred at 80 C for 20 h. After this time, the reaction mixture was poured into water (100 ml_). The water layer was extracted with EtOAc (2 x 100 ml_), and the combined organic layers were washed with water (3 x 100 ml_). The combined organic layers were then dred over MgS04, filtered and concentrated in vacuo. The resulting crude material was purified via silica gel chromatography, followed by recrystallization from hot EtOH.

Example 5: Post-Amide Coupling Modifications

Procedure 1 - Synthesis of Compound A30

[00369] Synthesis of A30 followed the scheme, below:

[00370] A 20 ml_ scintillation vial with stir bar was charged with N-(4-(2-isopropoxypropan- 2-yl)thiazol-2-yl)-1-(pyridin-4-ylmethyl)-1 H-pyrrole-2-carboxamide (131 mg, 0.341 mmol, 1.0 equiv) and CCU (2 ml_, 0.2 M). N-bromosuccinimide (66.7 mg, 0.375 mmol, 1.1 equiv) was added, and the reaction mixture was stirred at room temperature overnight. The next morning, the solvent was evaporated, and the crude material was purified directly via silica gel chromatography (70-100% EtOAc in hexanes) to yield the desired product. LCMS (+ESI): calc. [M+H] + = 463; found 463.

Procedure 2 - Synthesis of Compound A126

[00371] Synthesis of A126 followed the scheme, below:

[00372] A 4 ml_ vial with stir bar was charged with benzyl 4-((2-((4-(2-isopropoxypropan-2- yl)thiazol-2-yl)carbamoyl)-1 H-pyrrol-1-yl)methyl)piperidine-1-carboxylate (28 mg, 0.053 mmol, 1.0 equiv) and Pd/C (10 wt%, 5.7 mg, 0.0053 mmol, 0.1 equiv). The solids were evacuated and backflushed with hydrogen (1 atm, 3x). Methanol (0.5 ml_, 0.1 M) was added, and the reaction mixture was heated to 60 °C over 3 days. After 3 days, the reaction mixture was filtered and concentrated in vacuo. The resulting crude material was purified via silica gel chromatography (5% MeOH in DCM with 1 % NEΐb) to yield the desired product. LCMS (+ESI): calc. [M+H] + = 391 ; found 391.

Procedure 3 - Synthesis of Compound B24

[00373] Synthesis of B24 followed the scheme, below:

Step 1 : Boc Deprotection

[00374] A 20 ml_ scintillation vial with stir bar was charged with tert-butyl (R)-2-((4-ethyl-5- methylthiazol-2-yl)carbamoyl)pyrrolidine-1-carboxylate (196 mg, 0.577 mmol, 1.0 equiv). DCM (2.4 ml_) and TFA (600 uL, 20 vol%) were added, and the reaction mixture was stirred at room temperature overnight. The next morning, the reaction mixture was concentrated in vacuo. The resulting product was used directly in the next step without further purification.

Step 2: Reductive amination

[00375] A vial with stir bar was charged with (R)-N-(4-ethyl-5-methylthiazol-2- yl)pyrrolidine-2-carboxamide 2,2,2-trifluoroacetate (204 mg, 0.577 mmol, 1.0 equiv) and methanol (2.4 ml_, 0.2 M). Isonicotinaldehyde (0.326 ml_, 6.46 mmol, 6.0 equiv) was added, followed by acetic acid (0.149 mL, 2.60 mmol, 4.5 equiv). The reaction mixture was cooled to 0 °C. Sodium cyanoborohydride (218 mg, 3.46 mmol, 6.0 equiv) was slowly added at 0 °C, and the reaction was warmed to room temperature overnight. The next morning, the reaction mixture was diluted with DCM (50 mL) and washed with saturated NaHCCh (3 x 50 mL). The organic layer was dried over Na2SC>4, filtered and concentrated in vacuo. The resulting crude material was purified via silica gel chromatography (5% MeOH in DCM with 1 % NEt 3 ) to yield the desired product. LCMS (+ESI): calc. [M+H] + = 331 ; found 331.

Procedure 4 - Synthesis of Compound A85

[00376] Synthesis of A85 followed the scheme, below:

[00377] Amide (100 mg, 0.26 mmol, 1 equiv) was dissolved in DCM (2.6mL). mCPBA (64.1 mg, 0.28 mmol, 1.1 equiv) was added at room temperature, and the reaction mixture was stirred overnight. The next morning, the solvent was evaporated, and the resulting crude material was purified via pTLC (3% NEt3 in EtOAc), followed by trituration with Et2<D and filtration, to yield the desired product. LCMS (+ESI): calc. [M+H] + = 401 ; found 401.

Procedure 5 - Synthesis of Compound A228

[00378] Synthesis of Compound A228 followed the scheme, below:

[00379] A vial with stir bar was charged with aryl bromide (127 mg, 0.248 mmol, 1.0 equiv), sodium carbonate (79 mg, 0.745 mmol, 3.0 equiv), and Pd/C (10 wt%, 26 mg, 0.1 equiv). The vial was evacuated and backflushed with hydrogen. EtOH (1 mL) was added, and the solution was allowed to stir at room temperature for 1 h. After 1 h, the solution was filtered through a plug of Celite, and the reaction mixture was concentrated in vacuo to yield the desired product (obs. [M+H] = 433).

Procedure 6 - Synthesis of Compound A231

[00380] Synthesis of Compound A231 followed the scheme, below:

[00381] A vial with stir bar was charged with nitrile (20 mg, 0.0486 mmol, 1.0 equiv) in 1 :1 DMSO:MeOH (1 ml_). NaOH (1.0 M in water, 97 uL, 0.097 mmol, 2.0 equiv) was added, followed by H202 (30 wt% in water, 83 uL, 0.729 mmol, 15 equiv). The reaction mixture was allowed to stir at room temperature overnight. The next morning, the reaction mixture was poured into DCM (50 ml_) and washed with saturated NaHCOs (2 x 50 ml_). The combined aqueous layers were extracted with DCM (1 x 50 ml_), and the combined organic layers were dried over Na2S04, filtered and concentrated in vacuo to yield the desired product (obs. [M+H] = 430).

Procedure 7 - Synthesis of Compound A317

[00382] Synthesis of Compound A317 followed the scheme, below:

[00383] A vial with stir bar was charged with piperidine (74 mg, 0.208 mmol, 1.0 equiv), K 2 CO 3 (201 mg, 1.46 mmol, 7 equiv), 2-fluoropyridine (36 uL, 0.416 mmol, 2.0 equiv) and DMSO (1 ml_). The vial was capped and stirred at 100 C overnight. The next morning, the reaction mixture was poured into 10% MeOH in DCM (50 ml_). The organic layer was washed with saturated NaHCOs (2 x 50 ml_), and the combined aqueous layers were extracted with 10% MeOH in DCM (1 x 50 ml_). The combined organic layers were dried over Na 2 S0 4 , filtered and concentrated in vacuo. The resulting crude material was purified via silica gel chromatography to yield the desired product (obs. [M+H] = 431).

Procedure 8 - Synthesis of Compound A344

[00384] Synthesis of Compound A344 followed the scheme, below:

[00385] A 50 ml_ roundbottom flask with stir bar was charged with TBDPS-SM (220 mg, 0.322 mmol, 1.00 equiv), TBAF (841 mg, 3.22 mmol, 10 equiv) and THF (10.0 ml_, 0.032 M). The vial was capped and placed in a 40°C bath. The reaction mixture was stirred at 40°C for 5h. The reaction mixture was cooled to room temperature and quenched by H2O (30 ml_). The resulting solution was extracted with (3 x 30 ml_) of ethyl acetate and washed with (2x 30 ml_) of brine. The organic layer was then dried over Na2SC>4, filtered and concentrated in vacuo. The resulting crude material was purified via silica gel chromatography & Prep-HPLC or RP column to yield the desired product (obs. [M+H] = 446).

[00386] The following compounds were prepared via a similar method:

Procedure 9 - Synthesis of Compound A143

[00387] Acetylene (0.082 g, 0.28 mmol, 2.0 eq.), Cul (0.0236 g, 0.12 mmol, 0.3 eq.) and Pd(PPh3)2Cl2 (0.029 g, 0.04 mmol, 0.1 eq.) were added in succession, under Ar, to the degassed solution of starting material (0.150 g, 0.41 mmol, 1.0 eq.) in TEA (1 ml_). Reaction was continued at 80°C for 3h. Solution was filtered through Celite and washed with ethyl acetate. Filtrate was evaporated and crude product purified via FCC with hexane/ethyl acetate as eluent. In case of necessity final product was repurified by prep. HPLC

[00388] The following compounds were prepared in an analogous manner:

Procedure 10 - Synthesis of Compound A233

equiv), bromide (93 mg, 0.26 mmol, 1.0 equiv), K 3 PO 4 OO9 mg, 0.51 mmol, 2.0 equiv) and solution of 15% water in DMF (3.0 mL). The mixture was purged with argon for 5 min. After that time, Pd(PPh 3 ) 2 Cl 2 (18 mg, 0.03 mmol, 0.10 equiv) was added, the vial was capped, purged with argon for 2 min and placed in a 90 C bath for 2 hours. Then reaction mixture was cooled down to the room temperature and palladium residues were filtered off through celite and washed with EtOAc. The resulting filtrate was extracted with water and brine. Organic phase was dried over MgS0 4 , filtered and evaporated to dryness. The resulting crude material was purified via preparative HPLC to yield the desired product.

[00390] The following compounds were prepared in an analogous manner:

Procedure 11 - Synthesis of Compound A180

[00391] Reaction was carried out in H-cube. Conditions: 10% Pd/C , 60°C, 30 bar, THF, dilution 0.05M. The resulting crude material was purified via preparative HPLC to yield the desired product.

[00392] The following compounds were prepared in an analogous manner: Hydrogenation 2

[00393] Starting material (0.055 g, 0.13 mmol, 1.0 eq), Pd (0.21g, 10% on activated carbon) were mixed in ethanol. Reaction mixture was flushed with Ar, connected to H2 ballon (1 atm) and stirred overnight at 40°C. The catalyst was removed by filtration, the filtrate was evaporated and the crude material was purified by preparative HPLC to yield the desired product.

[00394] The following compounds were prepared in an analogous manner:

Example 6: Biological Assays

Dox-lnduced PD1-ss-Gluc Assay

[00395] Flp-ln 293 T-REx™ cells were transfected with pcDNA™5/FRT/TO plasmid inserted with cDNA encoding Gaussia Luciferase fused to the 3’ end of cDNA encoding PD1 signal sequence plus 10 amino acids (N-MQIPQAPWPVVWAVLQLGWRPGWFLDSPDR-C) (SEQ ID NO: 1). Transfected cells were selected for resistance to the selectable markers Hygromycin and Blasticidin to create a stable cell line that contained the PD1- ss+10aa/Gaussia Luciferase cDNA insert whose expression was regulated under the T- REx™ system. The day before assay, cells were trypsinized and plated in 384-well tissue culture plates. The next day, compound dilutions in DMSO/media containing doxycycline were added to the wells and incubated at 37°C, 5% CO2. 24 hours later, coelenterazine substrate was added to each well and luciferase signal was quantified using Tecan Infinite M1000 Pro for potency determination.

[00396] Results for select compounds provided herein are shown in Table 1 , below. For chemical structures that include one or more stereoisomers, but are illustrated without indicating stereochemistry, the assay data refers to a mixture of stereoisomers.

Pox Induced TNFa-FL-Gluc Assay

[00397] Flp-ln 293 T-REx™ cells were transfected with pcDNA™5/FRT/TO plasmid inserted with cDNA encoding Gaussia Luciferase fused to the 3’ end of cDNA encoding full length TNFa (amino acids 1-233). Transfected cells were selected for resistance to the selectable markers Hygromycin and Blasticidin to create a stable cell line that contained the TNFa-FL/Gaussia Luciferase cDNA insert whose expression was regulated under the T- REx™ system. The day before assay, cells were trypsinized and plated in 384-well tissue culture plates. The next day, compound dilutions in DMSO/media containing doxycycline were added to the wells and incubated at 37°C, 5% CO2. 24 hours later, coelenterazine substrate was added to each well and luciferase signal was quantified using Tecan Infinite M1000 Pro for potency determination.

[00398] Results for select compounds provided herein are shown in Table 1 , below. For chemical structures that include one or more stereoisomers, but are illustrated without indicating stereochemistry, the assay data refers to a mixture of stereoisomers.

Dox-lnduced Her3-ss-Gluc Assay

[00399] Flp-ln 293 T-REx™ cells were transfected with pcDNA™5/FRT/TO plasmid inserted with cDNA encoding Gaussia Luciferase fused to the 3’ end of cDNA encoding HER3 signal sequence plus 4 amino acids (N- MRANDALQVLGLLFSLARGSEVG-C) (SEQ ID NO: 2). Transfected cells were selected for resistance to the selectable markers Hygromycin and Blasticidin to create a stable cell line that contained the HER3- ss+4aa/Gaussia Luciferase cDNA insert whose expression was regulated under the T-REx™ system. The day before assay, cells were trypsinized and plated in 384-well tissue culture plates. The next day, compound dilutions in DMSO/media containing doxycycline were added to the wells and incubated at 37°C, 5% CO2. 24 hours later, coelenterazine substrate was added to each well and luciferase signal was quantified using Tecan Infinite M1000 Pro for potency determination.

[00400] Results for select compounds provided herein are shown in Table 1. For chemical structures that include one or more stereoisomers, but are illustrated without indicating stereochemistry, the assay data refers to a mixture of stereoisomers.

Pox Induced IL2-FL-Gluc Assay

[00401] Flp-ln 293 T-REx™ cells were transfected with pcDNA™5/FRT/TO plasmid inserted with cDNA encoding Gaussia Luciferase fused to the 3’ end of cDNA encoding full length IL-2 (amino acids 1-153). Transfected cells were selected for resistance to the selectable markers Hygromycin and Blasticidin to create a stable cell line that contained the IL-2-FL/Gaussia Luciferase cDNA insert whose expression was regulated under the T-REx™ system. The day before assay, cells were trypsinized and plated in 384-well tissue culture plates. The next day, compound dilutions in DMSO/media containing doxycycline were added to the wells and incubated at 37°C, 5% CO2. 24 hours later, coelenterazine substrate was added to each well and luciferase signal was quantified using Tecan Infinite M1000 Pro for potency determination.

[00402] Results for select compounds provided herein are shown in Table 1 , below. For chemical structures that include one or more stereoisomers, but are illustrated without indicating stereochemistry, the assay data refers to a mixture of stereoisomers. H929 Cell Viability Assay

[00403] The human multiple myeloma cell line NCI-H929 was cultured in Advanced RPMI 1640 media (Gibco®) supplemented with 6% fetal bovine serum, 2mM Glutamine, and 1x Penicillin/Streptomycin. On the day of assay, cells were resuspended in RPMI 1640 media supplemented with 10% fetal bovine serum, 2mM Glutamine, and 1x Penicillin/Streptmycin and plated in 384-well tissue culture plates and treated with compound dilutions in

DMSO/media. Plates were incubated at 37°C, 5% C0 2 for 48 hours. After 48 hours, Celltiter-Glo® (Promega) was added to each well and luciferase signal was quantified using Tecan Infinite M1000 Pro for cell viability determination.

[00404] Results for select compounds provided herein are shown in Table 1 , below. For chemical structures that include one or more stereoisomers, but are illustrated without indicating stereochemistry, the assay data refers to a mixture of stereoisomers.

U266 Cell Viability Assay

[00405] The human multiple myeloma cell line U266B1 was cultured in RPMI 1640 media supplemented with 10% fetal bovine serum, 2mM Glutamine, and 1x Penicillin/Streptomycin. Cells were plated in 384-well tissue culture plates and treated with compound dilutions in DMSO/media. Plates were incubated at 37°C, 5% C0 2 for 48 hours. After 48 hours, Celltiter-Glo® (Promega) was added to each well and luciferase signal was quantified using Tecan Infinite M1000 Pro for cell viability determination.

[00406] Results for select compounds provided herein are shown in Table 1 , below. For chemical structures that include one or more stereoisomers, but are illustrated without indicating stereochemistry, the assay data refers to a mixture of stereoisomers.

Table 1

I.A. indicates IC50 > 25 mM; NT indicates not tested.