Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
COMPOUNDS AND USES THEREOF
Document Type and Number:
WIPO Patent Application WO/2018/187614
Kind Code:
A1
Abstract:
The present invention features compounds useful in the treatment of BAF complex related disorders.

Inventors:
ANTHONY NEVILLE (US)
KUMARAVEL GNANASAMBANDAM (US)
CHAN HO (US)
RAY MRIDULA (US)
Application Number:
PCT/US2018/026319
Publication Date:
October 11, 2018
Filing Date:
April 05, 2018
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
FOGHORN THERAPEUTICS (US)
International Classes:
A61K31/4412; A61K31/4545; A61K31/5377
Foreign References:
US20160152617A12016-06-02
Other References:
DATABASE PubChem [O] 28 September 2012 (2012-09-28), "Compound Summary for CID 60187139", XP055544327, Database accession no. CID-60187139
DATABASE PubChem [O] 28 September 2012 (2012-09-28), "Compound Summary for CID 60185043", XP055544331, Database accession no. CID-60185043
DATABASE PubChem [O] 28 September 2012 (2012-09-28), "Compound Summary for CID 60184108", XP055544334, Database accession no. CID-60184108
Attorney, Agent or Firm:
ELBING, Karen, L. (US)
Download PDF:
Claims:
What is claimed is:

CLAIMS

1 . A compound having the struc I :

Formula

wherein m is 1 , 2, 3, or 4;

n and o are, independently, 0, 1 , or 2;

each R1 is, independently, halo;

R2 and R3 are, independently, selected from the group consisting of H and C1 -C6 alkyl, wherein the alkyl group is optionally substituted with at least one independently selected from the group consisting of Ci-Ce alkyl, C3-C8 cycloalkyi, halo, -OH, Ci-C6 alkoxy, Ci-C6 haloalkoxy, -NH2, alkyl), -NH(Ci-C6 alkyl) and -N(Ci-C6 alkyl)(Ci-C6 alkyl);

R4 is hydrogen, C1 -C6 alkyl, C1 -C6 perfluoroalkyl, C6-C10 aryl C1 -C6 alkyl, C1 -C6 heteroaryl C1 -C6 alkyl, C2-C9 heterocyclyl C1 -C6 alkyl, or C3-C10 carbocyclyl C1 -C6 alkyl, wherein the alkyl group is optionally substituted with at least one independently selected from the group consisting of C1 -C6 alkyl, Cs-Cs cycloalkyi, halo, -OH, Ci-C6 alkoxy, Ci-C6 haloalkoxy, -NH2, alkyl), -NH(Ci-C6 alkyl) and -N(Ci-C6 alkyl)(Ci-C6 alkyl) and the aryl, heteroaryl, heterocyclyl, or carbocyclyl group is optionally substituted with at least one independently selected from the group consisting of C1 -C6 alkyl, Ci-Ce haloalkyl, C3-C8 cycloalkyi, halo, -OH, Ci-C6 alkoxy, Ci-C6 haloalkoxy, -NH2, -NH(Ci-C6 alkyl), -N(Ci-C6 alkyl)(Ci-C6 alkyl), -NO2, -C(=0)OH, -C(=0)OR, and alkyl);

R5 is selected from the group consisting of H, C1 -C6 alkyl, C2-C9 heteroaryl, C6-C10 aryl C1 -C6 alkyl, C1 -C6 heteroaryl C1 -C6 alkyl, C2-C9 heterocyclyl C1 -C6 alkyl, C2-C9 heteroaryl C1 -C6 alkyl, or C3-C10 carbocyclyl Ci-C6 alkyl, -S(=0)2R, -C(=0)R, -C(=0)OR, -S(=0)2NHR and -C(=0)NR2, wherein the alkyl group is optionally substituted with at least one independently selected from the group consisting of C1 -C6 alkyl, C3-C8 cycloalkyi, halo, -OH, Ci-C6 alkoxy, Ci-C6 haloalkoxy, -NH2, alkyl), -NH(Ci- C6 alkyl) and -N(Ci-C6 alkyl)(Ci-C6 alkyl) and the aryl, heteroaryl, heterocyclyl, or carbocyclyl group is optionally substituted with at least one independently selected from the group consisting of C1 -C6 alkyl, Ci-Ce haloalkyl, C3-C8 cycloalkyi, halo, -OH, Ci-C6 alkoxy, Ci-C6 haloalkoxy, -NH2, -NH(Ci-C6 alkyl), -N(Ci-C6 alkyl)(Ci-C6 alkyl), -NO2, -C(=0)OH, -C(=0)OR, and alkyl); and

each R is, independently, selected from the group consisting of hydrogen, C1 -C6 alkyl, C1 -C6 heteroaryl, C6-C10 aryl, C3-C8 cycloalkyi, C2-C9 heterocyclyl, C6-C10 aryl C1 -C6 alkyl, C1 -C6 heteroaryl C1 -C6 alkyl, wherein the alkyl, cycloalkyi, aryl, heteroaryl, or heterocyclyl group is optionally substituted with at least one independently selected from the group consisting of C1 -C6 alkyl, -OH, -(C1 -C6 alkoxy), halo, -NH2, alkyl), -NH(Ci-C6 alkyl) and -N(Ci-C6 alkyl)(Ci-C6 alkyl).

or a pharmaceutically acceptable salt thereof.

2. The compound of claim 1 , wherein R1 is bromo.

3. The compound of claim 1 or 2, wherein the compound has the structure of Formula la:

Formula la

4. A compound having the struc I :

Formula

wherein m is 1 , 2, 3, or 4;

n and o are, independently, 0, 1 , or 2;

each R1 is, independently, C1 -C6 alkyi, C1 -C6 haloalkyl, C3-C8 cycloalkyl, halo, -OH, C1 -C6 alkoxy, Ci-C6 haloalkoxy, -NH2, -NH(Ci-C6 alkyi), -N(Ci-C6 alkyl)(Ci-C6 alkyi), -NO2, -C(=0)OH, -C(=0)OR, and -NHC(=0)(Ci-C6 alkyi) ;

R2 is hydrogen and R3 is unsubstituted C1 -C6 alkyi, C1 -C6 alkyi substituted with one or more C1 -C6 alkyi, or C1 -C6 alkyi C1 -C6 alkoxy;

R4 is hydrogen, C1 -C6 alkyi, C1 -C6 perfluoroalkyl, C6-C10 aryl C1 -C6 alkyi, C1 -C6 heteroaryl C1 -C6 alkyi, C2-C9 heterocyclyl C1 -C6 alkyi, or C3-C10 carbocyclyl C1 -C6 alkyi, wherein the alkyi group is optionally substituted with at least one independently selected from the group consisting of C1 -C6 alkyi, Cs-Cs cycloalkyl, halo, -OH, Ci-C6 alkoxy, Ci-C6 haloalkoxy, -NH2, -NHC(=0)(Ci-Ce alkyi), -NH(Ci-Ce alkyi) and -N(Ci-C6 alkyl)(Ci-C6 alkyi) and the aryl, heteroaryl , heterocyclyl, or carbocyclyl group is optionally substituted with at least one independently selected from the group consisting of C1 -C6 alkyi, Ci-Ce haloalkyl, C3-C8 cycloalkyl, halo, -OH, Ci-C6 alkoxy, Ci-C6 haloalkoxy, -NH2, -NH(Ci-Ce alkyi), -N(Ci-C6 alkyl)(Ci-C6 alkyi), -NO2, -C(=0)OH, -C(=0)OR, and -NHC(=0)(Ci-C6 alkyi) ;

R5 is selected from the group consisting of H, C1 -C6 alkyi, C2-C9 heteroaryl, C6-C10 aryl C1 -C6 alkyi, C1 -C6 heteroaryl C1 -C6 alkyi, C2-C9 heterocyclyl C1 -C6 alkyi, C2-C9 heteroaryl C1 -C6 alkyi, or C3-C10 carbocyclyl Ci-C6 alkyi, -S(=0)2R, -C(=0)R, -C(=0)OR, -S(=0)2NHR and -C(=0)NR2, wherein the alkyi group is optionally substituted with at least one independently selected from the group consisting of C1 -C6 alkyi, C3-C8 cycloalkyl, halo, -OH, Ci-C6 alkoxy, Ci-C6 haloalkoxy, -NH2, -NHC(=0)(Ci-Ce alkyi), -NH(Ci-C6 alkyi) and -N(Ci-C6 alkyl)(Ci-C6 alkyi) and the aryl, heteroaryl, heterocyclyl, or carbocyclyl group is optionally substituted with at least one independently selected from the group consisting of C1 -C6 alkyi, Ci-C6 haloalkyl, C3-C8 cycloalkyi, halo, -OH, Ci-C6 alkoxy, Ci-C6 haloalkoxy, -NH2, -NH(Ci-C6 alkyi), -N(Ci-C6 alkyl)(Ci-C6 alkyi), -NO2, -C(=0)OH, -C(=0)OR, and alkyi); and

each R is, independently, selected from the group consisting of hydrogen, C1 -C6 alkyi, C1 -C6 heteroaryl, C6-C10 aryl, C3-C8 cycloalkyi, C2-C9 heterocyclyl, C6-C10 aryl C1 -C6 alkyi, C1 -C6 heteroaryl C1 -C6 alkyi, wherein the alkyi, cycloalkyi, aryl, heteroaryl, or heterocyclyl group is optionally substituted with at least one independently selected from the group consisting of C1 -C6 alkyi, -OH, -(C1 -C6 alkoxy), halo, -NH2, alkyi), -NH(Ci-C6 alkyi) and -N(Ci-C6 alkyl)(Ci-C6 alkyi),

or a pharmaceutically acceptable salt thereof.

5. The compound of claim 4, wherein the compound has the compound has the structure of Formula V:

Formula V

wherein R9 is hydrogen, C1 -C5 alkyi, or C1 -C6 alkoxy,

or a pharmaceutically acceptable salt thereof.

6. The compound of claim 4 or 5, wherein the compound has the structure of Formula Va:

Formula Va

or a pharmaceutically acceptable salt thereof.

7. The compound of claim 5 or 6, wherein R9 is methoxy or methyl.

8. The compound of claim 5 or 6, wherein R9 is hydrogen.

9. The compound of any one of claims 4 to 8, wherein the compound has the structure of Formula Vb:

Formula Vb

10. A compound having the structure of Formula I:

Formula

wherein m is 1 , 2, 3, or 4;

n and o are, independently, 0, 1 , or 2;

each R1 is, independently, C1-C6 alkyl, C1-C6 haloalkyl, C3-C8 cycloalkyl, halo, -OH, C1-C6 alkoxy, Ci-C6 haloalkoxy, -NH2, -NH(Ci-C6 alkyl), -N(Ci-C6 alkyl)(Ci-C6 alkyl), -NO2, -C(=0)OH, -C(=0)OR, and -NHC(=0)(Ci-C6 alkyl);

R2 and R3 are, independently, selected from the group consisting of H and C1-C6 alkyl, wherein the alkyl group is optionally substituted with at least one independently selected from the group consisting of Ci-Ce alkyl, C3-C8 cycloalkyl, halo, -OH, Ci-C6 alkoxy, Ci-C6 haloalkoxy, -NH2, -NHC(=0)(Ci-Ce alkyl), -NH(Ci-C6 alkyl) and -N(Ci-C6 alkyl)(Ci-C6 alkyl);

R4 is unsubstituted C1-C6 alkyl; C1-C6 alkyl C6-C10 aryl wherein the alkyl is unsubstituted and the aryl is unsubstituted or substituted with bromo, C1-C6 perfluoroalkyi, C1-C6 heteroalkyi; or C1-C6 alkyl Ce- C10 aryl wherein the alkyl is substituted with at least one -OH or C1-C6 alkyl and the aryl is substituted with Ci-Ce alkyl, Ci-C6 haloalkyl, C3-C8 cycloalkyl, halo, -OH, Ci-C6 alkoxy, Ci-C6 haloalkoxy, -NH2, -NH(Ci-C6 alkyl), -N(Ci-C6 alkyl)(Ci-C6 alkyl), -NO2, -C(0)OH, -C(0)OR, or -NHC(0)(Ci-C6 alkyl);

R5 is selected from the group consisting of H, C1-C6 alkyl, C2-C9 heteroaryl, C6-C10 aryl C1-C6 alkyl, C1-C6 heteroaryl C1-C6 alkyl, C2-C9 heterocyclyl C1-C6 alkyl, C2-C9 heteroaryl C1-C6 alkyl, or C3-C10 carbocyclyl Ci-C6 alkyl, -S(=0)2R, -C(=0)R, -C(=0)OR, -S(=0)2NHR and -C(=0)NR2, wherein the alkyl group is optionally substituted with at least one independently selected from the group consisting of C1-C6 alkyl, C3-C8 cycloalkyl, halo, -OH, Ci-C6 alkoxy, Ci-C6 haloalkoxy, -NH2, -NHC(=0)(Ci-Ce alkyl), -NH(Ci- C6 alkyl) and -N(Ci-C6 alkyl)(Ci-C6 alkyl) and the aryl, heteroaryl, heterocyclyl, or carbocyclyl group is optionally substituted with at least one independently selected from the group consisting of C1 -C6 alkyi, Ci-Ce haloalkyl, C3-C8 cycloalkyi, halo, -OH, Ci-C6 alkoxy, Ci-C6 haloalkoxy, -NH2, -NH(Ci-C6 alkyi), -N(Ci-C6 alkyl)(Ci-C6 alkyi), -NO2, -C(=0)OH, -C(=0)OR, and alkyi); and

each R is, independently, selected from the group consisting of hydrogen, C1 -C6 alkyi, C1 -C6 heteroaryl, C6-C10 aryl, C3-C8 cycloalkyi, C2-C9 heterocyclyl, C6-C10 aryl C1 -C6 alkyi, C1 -C6 heteroaryl C1 -C6 alkyi, wherein the alkyi, cycloalkyi, aryl, heteroaryl, or heterocyclyl group is optionally substituted with at least one independently selected from the group consisting of C1 -C6 alkyi, -OH, -(C1 -C6 alkoxy), halo, -NH2, alkyi), -NH(Ci-C6 alkyi) and -N(Ci-C6 alkyl)(Ci-C6 alkyi),

or a pharmaceutically acceptable salt thereof.

1 1 . The compound of clai has the structure of Formula II :

Formula II

wherein p is 1 or 2;

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

R6 is OH or Ci-Ce alkyi ; and

R7 is Ci-Ce alkyi, Ci-C6 haloalkyl, C3-C8 cycloalkyi, halo, -OH, Ci-C6 alkoxy, Ci-C6 haloalkoxy, -NH(Ci-C6 alkyi), -N(Ci-C6 alkyl)(Ci-C6 alkyi), -NO2, -C(0)OH, -C(0)OR, or -NHC(0)(Ci-C6 alkyi), or a pharmaceutically acceptable salt thereof.

12. The compound of claim 1 1 , wherein R7 is methoxy or -O-CH2CH2-O-CH3.

13. The compound of clai has the structure of Formula lla:

Formula lla

or a pharmaceutically acceptable salt thereof.

14. The compound of any one of claims 1 1 to 13, wherein R6 is methyl.

15. The compound of any one of claims 1 1 to 13, wherein R6 is OH.

16. The compound of any one of claims 1 1 to 15, wherein the compound has the structure of Formula lib:

Formula Mb

wherein R3 is hydrogen or -CH2OH.

17. A compound having the structure of Formula I :

Formula

wherein m is 1 , 2, 3, or 4;

n and 0 are, independently, 0, 1 , or 2;

each R1 is, independently, C1 -C6 alkyl, C1 -C6 haloalkyl, C3-C8 cycloalkyl, halo, -OH, C1 -C6 alkoxy, Ci-C6 haloalkoxy, -NH2, -NH(Ci-C6 alkyl), -N(Ci-C6 alkyl)(Ci-C6 alkyl), -NO2, -C(=0)OH, -C(=0)OR, and -NHC(=0)(Ci-C6 alkyl);

R2 and R3 are, independently, selected from the group consisting of H and C1 -C6 alkyl, wherein the alkyl group is optionally substituted with at least one independently selected from the group consisting of Ci-Ce alkyl, C3-C8 cycloalkyl, halo, -OH, Ci-C6 alkoxy, Ci-C6 haloalkoxy, -NH2, -NHC(=0)(Ci-Ce alkyl), -NH(Ci-C6 alkyl) and -N(Ci-C6 alkyl)(Ci-C6 alkyl);

R4 is hydrogen, C1 -C6 alkyl, C1 -C6 perfluoroalkyl, C6-C10 aryl C1 -C6 alkyl, C1 -C6 heteroaryl C1 -C6 alkyl, C2-C9 heterocyclyl C1 -C6 alkyl, or C3-C10 carbocyclyl C1 -C6 alkyl, wherein the alkyl group is optionally substituted with at least one independently selected from the group consisting of C1 -C6 alkyl, Cs-Cs cycloalkyl, halo, -OH, Ci-C6 alkoxy, Ci-C6 haloalkoxy, -NH2, -NHC(=0)(Ci-Ce alkyl), -NH(Ci-Ce alkyl) and -N(Ci-C6 alkyl)(Ci-C6 alkyl) and the aryl, heteroaryl, heterocyclyl, or carbocyclyl group is optionally substituted with at least one independently selected from the group consisting of C1 -C6 alkyl, Ci-Ce haloalkyl, C3-C8 cycloalkyl, halo, -OH, Ci-C6 alkoxy, Ci-C6 haloalkoxy, -NH2, -NH(Ci-Ce alkyl), -N(Ci-C6 alkyl)(Ci-C6 alkyl), -NO2, -C(=0)OH, -C(=0)OR, and -NHC(=0)(Ci-C6 alkyl);

R5 is hydrogen, C2-C9 heteroaryl, C1 -C6 alkyl C3-C8 cycloalkyl, -SO2R, or -C(0)R, wherein R is C2-C9 heteroaryl, C1 -C6 alkyl, C1 -C6 heteroalkyl, -NR2, or C6-C10 aryl; and

each R is, independently, selected from the group consisting of hydrogen, C1 -C6 alkyl, C1 -C6 heteroaryl, C6-C10 aryl, C3-C8 cycloalkyl, C2-C9 heterocyclyl, C6-C10 aryl C1 -C6 alkyl, C1 -C6 heteroaryl C1 -C6 alkyi, wherein the alkyi, cycloalkyi, aryl, heteroaryl, or heterocyclyl group is optionally substituted with at least one independently selected from the group consisting of C1 -C6 alkyi, -OH, -(C1 -C6 alkoxy), halo, -NH2, -NHC(=0)(Ci -C6 alkyi), -NH(Ci -C6 alkyi) and -N(Ci -C6 alkyl)(Ci -C6 alkyi),

or a pharmaceutically acceptable salt thereof.

18. The compound of claim 17, as the structure of Formula III:

Formula III

wherein R8 is C2-C9 heteroaryl, C1 -C6 alkyi, C1 -C6 heteroalkyl, -NR2, or C6-C10 aryl, or a pharmaceutically acceptable salt thereof.

19. The compound of claim 17 or 18, wherein the compound has the structure of Formula Ilia:

Formula Ilia

wherein R8 is C2-C9 heteroaryl, C1 -C6 alkyi, C1 -C6 heteroalkyl, -NR2, or C6-C10 aryl, or a pharmaceutically acceptable salt thereof.

20. The compound of claim 18 or 19, wherein R8 is 2-pyridinyl.

21 . The compound of claim 18 or 19, wherein R8 is methyl.

22. The compound of claim 18 or 19, wherein R8 is methoxy or methoxyethyl.

23. The compound of claim 18 or 19, wherein R8 is -NH(CH3) or -N(CH3)2.

24. The compound of claim 18 or 19, wherein R8 is phenyl, 4-methoxy-phenyl, or 4- methoxyethoxy-phenyl.

25. The compound of any one of claims 17 to 24, wherein the compound has the structure of Formula 1Mb:

Formula 1Mb

26. The compound of claim 17, wherein the compound has the structure of Formula IV:

Formula IV

wherein R5 is hydrogen, C2-C9 heteroaryl, C1 -C6 alkyl C3-C8 cycloalkyl, -SO2R,

or a pharmaceutically acceptable salt thereof.

The compound of claim 17 compound has the structure of Formula IVa:

or a pharmaceutically acceptable salt thereof.

28. The compound of claim 26 or 27, wherein R5 is 2-pyridinyl.

29. The compound of claim 26 or 27, wherein R5 is -Ch -cyclobutyl.

30. The compound of claim 26 or 27, wherein R5 is -S02-cyclobutyl.

31 . The compound of any one of claims 26 to 30, wherein the compound has the structure of Formula IVb:

Formula IVb

or a pharmaceutically acceptable salt thereof.

32. A compound having the structure of any one of compounds 1 -61 in Table 1 , or a pharmaceutically acceptable salt thereof.

33. A pharmaceutical composition comprising the compound of any one of claims 1 to 32 and a pharmaceutically acceptable excipient.

34. A method of inhibiting the activity of an SS18-SSX fusion protein, SS1 8 wild type protein, or SSX wild type protein in a cell, the method comprising contacting the cell with an effective amount of a compound of any one of claims 1 to 32 or a pharmaceutical composition of claim 33.

35. A method of decreasing the level of an SS18-SSX fusion protein, SS18 wild type protein, or SSX wild type protein in a cell, the method comprising contacting the cell with an effective amount of a compound of any one of claims 1 to 32 or a pharmaceutical composition of claim 33.

36. A method of increasing the activity of a BAF complex in a cell, the method comprising contacting the cell with an effective amount of a compound of any one of claims 1 to 32 or a

pharmaceutical composition of claim 33.

37. A method of increasing the level of BAF47 in a cell, the method comprising contacting the cell with an effective amount of a compound of any one of claims 1 to 32 or a pharmaceutical composition of claim 33.

38. A method of decreasing Wnt/p-catenin signaling in a cell, the method comprising contacting the cell with an effective amount of a compound of any one of claims 1 to 32 or a pharmaceutical composition of claim 33.

39. A method of inducing apoptosis in a cancer cell, the method comprising contacting the cell with an effective amount of a compound of any one of claims 1 to 32 or a pharmaceutical composition of claim 33.

40. The method of any one of claims 34 to 39, wherein the cell is a mammalian cell.

41 . A method of reducing tumor growth in a subject in need thereof, the method comprising administering to the subject an effective amount of a compound of any one of claims 1 to 32 or a pharmaceutical composition of claim 33.

42. A method treating a disorder related to a SS1 8-SSX fusion protein, SS18 wild type protein, or SSX wild type protein in a subject in need thereof, the method comprising administering to the subject an effective amount of a compound of any one of claims 1 to 32 or a pharmaceutical composition of claim 33.

43. The method of claim 42, wherein the disorder related to a SS18-SSX fusion protein, SS1 8 wild type protein, or SSX wild type protein is cancer.

44. The method of claim 43, wherein the cancer is a sarcoma.

45. The method of claim 44, wherein the sarcoma is synovial sarcoma or Ewing's sarcoma.

46. The method of any one of claims 42 to 45, wherein the disorder related to a SS18-SSX fusion protein, SS1 8 wild type protein, or SSX wild type protein is synovial sarcoma.

47. A method treating a disorder related to BAF47 in a subject in need thereof, the method comprising administering to the subject an effective amount of a compound of any one of claims 1 to 32 or a pharmaceutical composition of claim 33.

48. The method of claim 47, wherein the disorder related to BAF47 is cancer or a viral infection.

49. The method of claim 48, wherein the cancer is a malignant, rhabdoid tumor, a CD8+ T-cell lymphoma, endometrial carcinoma, ovarian carcinoma, bladder cancer, stomach cancer, pancreatic cancer, esophageal cancer, prostate cancer, renal cell carcinoma, melanoma, or colorectal cancer.

50. The method of claim 48, wherein the viral infection is an infection with a virus of the

Retroviridae family, Hepadnaviridae family, Flaviviridae family, Adenoviridae family, Herpesviridae family, Papillomaviridae family, Parvoviridae family, Polyomaviridae family, Paramyxoviridae family, or

Togaviridae family.

51 . A method for treating cancer in a subject in need thereof, the method comprising

administering to the subject an effective amount of a compound of any one of claims 1 to 32 or a pharmaceutical composition of claim 33.

52. The method of claim 51 , wherein the cancer is a malignant, rhabdoid tumor, a CD8+ T-cell lymphoma, endometrial carcinoma, ovarian carcinoma, bladder cancer, stomach cancer, pancreatic cancer, esophageal cancer, prostate cancer, renal cell carcinoma, melanoma, colorectal cancer, a sarcoma, non-small cell lung cancer, stomach cancer, or breast cancer.

53. The method of claim 51 or 52, wherein the cancer is a malignant, rhabdoid tumor, a CD8+ T- cell lymphoma, endometrial carcinoma, ovarian carcinoma, bladder cancer, stomach cancer, pancreatic cancer, esophageal cancer, prostate cancer, renal cell carcinoma, melanoma, or colorectal cancer.

54. The method of claim 51 or 52, wherein the cancer is a sarcoma, non-small cell lung cancer, stomach cancer, or breast cancer.

55. The method of claim 54, wherein the cancer is synovial sarcoma.

56. A method of treating a viral infection, the method comprising administering to the subject an effective amount of a compound of any one of claims 1 to 32 or a pharmaceutical composition of claim 33.

57. The method of claim 56, wherein the viral infection is an infection with a virus of the

Retroviridae family, Hepadnaviridae family, Flaviviridae family, Adenoviridae family, Herpesviridae family, Papillomaviridae family, Parvoviridae family, Polyomaviridae family, Paramyxoviridae family, or

Togaviridae family.

58. The method of any one of claims 41 to 49 and 51 to 55, wherein the method further comprises administering to the subject an additional anticancer therapy.

59. The method of claim 58, wherein the additional anticancer therapy is a chemotherapeutic or cytotoxic agent or radiotherapy.

60. The method of claim 59, wherein the chemotherapeutic or cytotoxic agent is doxorubicin or ifosfamide.

61 . The method of any one of claims 58 to 60, wherein the additional anticancer therapy and the compound of any one of claims 1 to 32 or pharmaceutical composition of claim 33 are administered within 28 days of each other each in an amount that together are effective to treat the subject.

Description:
COMPOUNDS AND USES THEREOF

Background

The protein encoded by the SS18 gene on chromosome 18 is a member of the human SWI/SNF chromatin remodeling complex (also known as BAF complex). An SS1 8-SSX fusion protein is formed by a chromosomal translocation (X;18)(p1 1 .2;q1 1 /2) which results in the fusion of the SS18 gene on chromosome 18 to a SSX gene (e.g., SSX1 , SSX2, or SSX4) on the X chromosome. SS18-SSX fusion proteins have been found to be capable of replacing the native SS18 protein in the BAF complex, and in some circumstances displacing a portion of the BAF complex, e.g., the subunit BAF47 (SNF5), resulting in its degradation. Disruption of the BAF complex by SS18-SSX has been found to result in several downstream effects in cells, including up-regulation of Wnt/p-catenin signaling dysregulation of cellular differentiation and self-renewal program, and enhanced eviction of polycomb repressive complex.

Further, the SS18-SSX fusion protein has been detected in more than 95% of synovial sarcoma tumors and is often the only cytogenetic abnomarlity in synovial sarcoma.

Summary of the Invention

The present invention features compounds useful to modulate a BAF complex, e.g., in a subject in need thereof. In some embodiments, the compounds described herein are useful in the treatment of disorders associated with an alteration in a BAF complex, e.g., a disorder associated with an alteration in a SS18 protein, e.g., a disorder associated with a SS1 8-SSX fusion protein. The compounds of the invention, alone or in combination with other pharmaceutically active agents, can be used for treating such disorders.

In one aspect, this disclosure provides a compound, or a pharmaceutically acceptable salt thereof, having the structure of Formula I:

Formula I

wherein m is 1 , 2, 3, or 4;

n and o are, independently, 0, 1 , or 2;

each R 1 is, independently, C1 -C6 alkyl, C1 -C6 haloalkyl, C3-C8 cycloalkyl, halo, -OH, C1 -C6 alkoxy, Ci -C 6 haloalkoxy, -NH 2 , -NH(Ci -C 6 alkyl), -N(Ci -C 6 alkyl)(Ci -C 6 alkyl), -NO2, -C(=0)OH, -C(=0)OR, and -NHC(=0)(Ci -C 6 alkyl);

R 2 and R 3 are, independently, selected from the group consisting of H and C1 -C6 alkyl, wherein the alkyl group is optionally substituted with at least one independently selected from the group consisting of Ci -Ce alkyl, C 3 -C 8 cycloalkyl, halo, -OH, Ci -C 6 alkoxy, Ci -C 6 haloalkoxy, -NH 2 , -NHC(=0)(Ci -Ce alkyl), -NH(Ci -C 6 alkyl) and -N(Ci -C 6 alkyl)(Ci -C 6 alkyl); R 4 is hydrogen, C1 -C6 alkyi, C1 -C6 perfluoroalkyl, C6-C10 aryl C1 -C6 alkyi, C1 -C6 heteroaryl C1 -C6 alkyi, C2-C9 heterocyclyl C1 -C6 alkyi, or C3-C10 carbocyclyl C1 -C6 alkyi, wherein the alkyi group is optionally substituted with at least one independently selected from the group consisting of C1 -C6 alkyi, Cs-Cs cycloalkyi, halo, -OH, Ci-C 6 alkoxy, Ci-C 6 haloalkoxy, -NH 2 , alkyi), -NH(Ci-C 6 alkyi) and -N(Ci-C6 alkyl)(Ci-C6 alkyi) and the aryl, heteroaryl, heterocyclyl, or carbocyclyl group is optionally substituted with at least one independently selected from the group consisting of C1 -C6 alkyi, Ci-Ce haloalkyl, C 3 -C 8 cycloalkyi, halo, -OH, Ci-C 6 alkoxy, Ci-C 6 haloalkoxy, -NH 2 , -NH(Ci-C 6 alkyi), -N(Ci-C 6 alkyl)(Ci-C 6 alkyi), -NO2, -C(=0)OH, -C(=0)OR, and alkyi);

R 5 is selected from the group consisting of H, C1 -C6 alkyi, C2-C9 heteroaryl, C6-C10 aryl C1 -C6 alkyi, Ci-C 6 heteroaryl Ci-C 6 alkyi, C2-C9 heterocyclyl Ci-C 6 alkyi, C2-C9 heteroaryl Ci-C 6 alkyi, or C3-C10 carbocyclyl Ci-C 6 alkyi, -S(=0) 2 R, -C(=0)R, -C(=0)OR, -S(=0) 2 NHR and -C(=0)NR 2 , wherein the alkyi group is optionally substituted with at least one independently selected from the group consisting of C1 -C6 alkyi, C 3 -C 8 cycloalkyi, halo, -OH, Ci-C 6 alkoxy, Ci-C 6 haloalkoxy, -NH 2 , alkyi),

-NH(Ci-C6 alkyi) and -N(Ci-C6 alkyl)(Ci-C6 alkyi) and the aryl, heteroaryl, heterocyclyl, or carbocyclyl group is optionally substituted with at least one independently selected from the group consisting of C1 -C6 alkyi, Ci-C 6 haloalkyl, C 3 -C 8 cycloalkyi, halo, -OH, Ci-C 6 alkoxy, Ci-C 6 haloalkoxy, -NH 2 , -NH(Ci-C 6 alkyi), -N(Ci-C 6 alkyl)(Ci-C 6 alkyi), -NO2, -C(=0)OH, -C(=0)OR, and alkyi); and

each R is, independently, selected from the group consisting of hydrogen, C1 -C6 alkyi, C1 -C6 heteroaryl, C6-C10 aryl, C3-C8 cycloalkyi, C2-C9 heterocyclyl, C6-C10 aryl C1 -C6 alkyi, C1 -C6 heteroaryl C1 -C6 alkyi, wherein the alkyi, cycloalkyi, aryl, heteroaryl, or heterocyclyl group is optionally substituted with at least one independently selected from the group consisting of C1 -C6 alkyi, -OH, -(C1 -C6 alkoxy), halo, -NH2, alkyi), -NH(Ci-C 6 alkyi) and -N(Ci-C 6 alkyl)(Ci-C 6 alkyi),

or a pharmaceutically acceptable salt thereof.

In some embodiments, R 1 is halo (e.g., bromo).

In some embodiments, R 2 is hydrogen and R 3 is unsubstituted C1 -C6 alkyi (e.g., methyl) or C1 -C6 alkyi Ci-C 6 alkoxy (e.g., -CH2-O-CH3).

In some embodiments, R 4 is unsubstituted C1 -C6 alkyi; C1 -C6 alkyi C6-C10 aryl wherein the alkyi is unsubstituted and the aryl is unsubstituted or substituted with bromo, C1 -C6 perfluoroalkyl, C1 -C6 heteroalkyl (e.g., methoxy or -O-CH2CH2-O-CH3) ; or C1 -C6 alkyi C6-C10 aryl wherein the alkyi is substituted with at least one -OH or C1 -C6 alkyi (e.g., methyl) and the aryl is substituted with C1 -C6 alkyi, Ci-Ce haloalkyl, C 3 -C 8 cycloalkyi, halo, -OH, Ci-C 6 alkoxy, Ci-C 6 haloalkoxy, -NH 2 , -NH(Ci-C 6 alkyi), -N(Ci-C 6 alkyl)(Ci-C 6 alkyi), -NO2, -C(0)OH, -C(0)OR, or -NHC(0)(Ci-C 6 alkyi).

In some embodiments, R 5 is hydrogen, C2-C9 heteroaryl (e.g., 2-pyridinyl), C1 -C6 alkyi C3-C8 cycloalkyi (e.g., -CH2-cyclobutyl), -SO2R (e.g., -S02-cyclobutyl), or -C(0)R, wherein R is C2-C9 heteroaryl (e.g., 2-pyridinyl), C1 -C6 alkyi (e.g.., methyl), C1 -C6 heteroalkyl (e.g., methoxy or methoxyethyl), -NR2

(e.g., -N(CH3)2 or -NH(CH3), or C6-C10 aryl (e.g., phenyl, 4-methoxy-phenyl, or 4-methoxyethoxy-phenyl).

In some embodiments of any of the foregoing compounds, m is 1 . In some embodiments of any of the foregoing compounds, n is 1 . In some embodiments of any of the foregoing compounds, 0 is 1 . In some embodiments, R 2 is hydrogen and R 3 is -CH2OH. In some embodiments, R 1 is C1 -C6 alkoxy (e.g., methoxy). In some embodiments, R 5 is C6-C10 aryl C1 -C6 alkyi (e.g., 2-fluoro-benzyl). In some embodiments, R 5 is -C(=0)R, wherein R is C3-C8 cycloalkyi (e.g., cyclobutyl). In some embodiments, the compound has the structure of Formula la:

Formula la

or a pharmaceutically acceptable salt thereof.

In some embodiments, the ula II:

Formula II

wherein p is 1 or 2;

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

R 6 is OH or Ci-Ce alkyl (e.g., methyl); and

R 7 is Ci-Ce alkyl, Ci-C 6 haloalkyl, C 3 -C 8 cycloalkyl, halo, -OH, Ci-C 6 alkoxy, Ci-C 6 haloalkoxy, -NH(Ci-C 6 alkyl), -N(Ci-C 6 alkyl)(Ci-C 6 alkyl), -N0 2 , -C(0)OH, -C(0)OR, or -NHC(0)(Ci-C 6 alkyl), or a pharmaceutically acceptable salt thereof.

In some embodiments, the mula lla:

Formula lla

or a pharmaceutically acceptable salt thereof.

In some embodiments, the mula Mb:

Formula Mb wherein R 3 is hydrogen or -CH2OH,

or a pharmaceutically acceptable salt thereof.

In some embodiments, the compound has the structure of Formula III :

Formula III

wherein R 8 is C2-C9 heteroaryl (e.g., 2-pyridinyl), C1 -C6 alkyl (e.g., methyl), C1 -C6 heteroalkyl (e.g., methoxy or methoxyethyl), -NR2 (e.g., -N(CH3)2 or -NH(CH3), or C6-C10 aryl (e.g., phenyl, 4-methoxy-phenyl, or 4-methoxyethoxy-phenyl),

or a pharmaceutically acceptable salt thereof.

In some embodiments, the compound has the structure of Formula Ilia:

Formula Ilia

wherein R 8 is C2-C9 heteroaryl, C1 -C6 alkyl, C1 -C6 heteroalkyl, -NR2, or C

or a pharmaceutically acceptable salt thereof.

In some embodiments, the compound has the structure of Formula lllb:

Formula lllb

or a pharmaceutically acceptable salt thereof.

In some embodiments, the compound has the structure of Formula IV:

Formula IV wherein R 5 is hydrogen, C2-C9 heteroaryl (e.g., 2-pyridinyl), C1 -C6 alkyl C3-C8 cycloalkyi (e.g., -CH 2 -cyclobutyl), -SO2R (e.g., -S0 2 -cyclobutyl),

or a pharmaceutically acceptable salt thereof.

In some embodiments, the compound has the structure of Formula IVa:

or a pharmaceutically acceptable salt thereof.

In some embodiments, the compound has the structure of Formula IVb:

Formula IVb

or a pharmaceutically acceptable salt thereof.

In some embodiments, the compound has the structure of Formula V:

Formula V

wherein R 9 is hydrogen, C1 -C5 alkyl (e.g., methyl), or C1 -C6 alkoxy (e.g., methoxy), or a pharmaceutically acceptable salt thereof.

In some embodiments, the compound has the structure of Formula Va:

Formula Va

or a pharmaceutically acceptable salt thereof. In some embodiments, the compound has the structure of Formula Vb:

Formula Vb

or a pharmaceutically acceptable salt thereof.

In another aspect, the disclosure provides a compound, or pharmaceutically acceptable salt thereof, having the structure of any one of compounds 1 -61 in Table 1 .

Table 1 .













 In another aspect, the disclosure provides pharmaceutical composition including any of the foregoing compounds, or pharmaceutically acceptable salts thereof and a pharmaceutically acceptable excipient.

In another aspect, the disclosure provides a method of altering, e.g., inhibiting, the activity of an SS18-SSX fusion protein, SS18 wild type protein, or SSX wild type protein, in a cell (e.g., a mammalian cell). This method includes contacting the cell with an effective amount of any of the foregoing compounds, or pharmaceutically acceptable salts thereof, or any of the foregoing pharmaceutical compositions.

In another aspect, the disclosure provides a method of altering, e.g., decreasing, the level of an SS18-SSX fusion protein, SS18 wild type protein, or SSX wild type protein, in a cell (e.g., a mammalian cell). This method includes contacting the cell with an effective amount of any of the foregoing compounds, or pharmaceutically acceptable salts thereof, or any of the foregoing pharmaceutical compositions.

In another aspect, the disclosure provides a method of altering, e.g., increasing, the activity of a BAF complex in a cell (e.g., a mammalian cell). This method includes contacting the cell with an effective amount of any of the foregoing compounds, or pharmaceutically acceptable salts thereof, or any of the foregoing pharmaceutical compositions.

In another aspect, the disclosure provides a method of altering, e.g., increasing, the level of BAF47 in a cell (e.g., a mammalian cell). This method includes contacting the cell with an effective amount of any of the foregoing compounds, or pharmaceutically acceptable salts thereof, or any of the foregoing pharmaceutical compositions.

In another aspect, the disclosure provides a method of altering, e.g., decreasing Wnt/p-catenin signaling, in a cell (e.g., a mammalian cell). This method includes contacting the cell with an effective amount of any of the foregoing compounds, or pharmaceutically acceptable salts thereof, or any of the foregoing pharmaceutical compositions.

In another aspect, the disclosure provides a method of inducing cell differentiation, e.g., regulated differentiation program in a cancer cell (e.g., a mammalian cancer cell). This method includes contacting the cell with an effective amount of any of the foregoing compounds, or pharmaceutically acceptable salts thereof, or any of the foregoing pharmaceutical compositions.

In another aspect, the disclosure provides a method of inducing cell death, e.g., regulated cell death, e.g., apoptosis, in a cancer cell (e.g., a mammalian cancer cell). This method includes contacting the cell with an effective amount of any of the foregoing compounds, or pharmaceutically acceptable salts thereof, or any of the foregoing pharmaceutical compositions.

In another aspect, the disclosure provides a method of reducing tumor growth in a subject in need thereof. This method includes administering to the subject an effective amount of any of the foregoing compounds, or pharmaceutically acceptable salts thereof, or any of the foregoing pharmaceutical compositions.

In another aspect, the disclosure provides a method treating a disorder related to a SS18-SSX fusion protein, SS1 8 wild type protein, or SSX wild type protein (e.g., cancer) in a subject in need thereof. This method includes contacting a cell with an effective amount of any of the foregoing compounds, or pharmaceutically acceptable salts thereof, or any of the foregoing pharmaceutical compositions. In some embodiments, the disorder is a sarcoma (e.g., synovial sarcoma or Ewing's sarcoma), non-small cell lung cancer (e.g., squamous or adenocarcinoma), stomach cancer, or breast cancer. In some embodiments, the cancer is synovial sarcoma.

In another aspect, the disclosure provides a method treating a disorder related to a SS18-SSX fusion protein (e.g., cancer) in a subject in need thereof. This method includes administering to the subject an effective amount of any of the foregoing compounds, or pharmaceutically acceptable salts thereof, or any of the foregoing pharmaceutical compositions. In some embodiments, the disorder is a sarcoma (e.g., synovial sarcoma or Ewing's sarcoma). In some embodiments, the sarcoma is synovial sarcoma.

In another aspect, the disclosure provides a method treating a disorder related to BAF47 (e.g., cancer or viral infections) in a subject in need thereof. This method includes contacting a cell with an effective amount of any of the foregoing compounds, or pharmaceutically acceptable salts thereof, or any of the foregoing pharmaceutical compositions. In some embodiments, the disorder is a cancer, e.g., malignant, rhabdoid tumor, a CD8+ T-cell lymphoma, endometrial carcinoma, ovarian carcinoma, bladder cancer, stomach cancer, pancreatic cancer, esophageal cancer, prostate cancer, renal cell carcinoma, melanoma, colorectal cancer. In some embodiments, the disorder is a viral infection, for example, an infection with a virus of the Retroviridae family such as the lentiviruses (e.g. Human immunodeficiency virus (HIV) and deltaretroviruses (e.g., human T cell leukemia virus I (HTLV-I), human T cell leukemia virus II (HTLV-II)); Hepadnaviridae family (e.g. hepatitis B virus (HBV)), Flaviviridae family (e.g. hepatitis C virus (HCV)), Adenoviridae family (e.g. Human Adenovirus), Herpesviridae family (e.g. Human cytomegalovirus (HCMV), , Epstein-Barr virus, herpes simplex virus 1 (HSV-1 ), herpes simplex virus 2 (HSV-2), human herpesvirus 6 (HHV-6), Herpesvitus K * , CMV, varicella-zoster virus), Papillomaviridae family (e.g. Human Papillomavirus (HPV, HPV E1 )), Parvoviridae family (e.g. Parvovirus B19),

Polyomaviridae family (e.g. JC virus and BK virus), Paramyxoviridae family (e.g. Measles virus), Togaviridae family (e.g. Rubella virus). In some embodiments, the disorder is Coffin Siris,

Neurofibromatosis (e.g., NF-1 , NF-2, or Schwannomatosis), or Multiple Meningioma.

In another aspect, the disclosure provides a method for treating cancer in a subject in need thereof. This method includes administering to the subject an effective amount of any of the foregoing compounds, or pharmaceutically acceptable salts thereof, or any of the foregoing pharmaceutical compositions. In some embodiments, the cancer is a malignant, rhabdoid tumor, a CD8+ T-cell lymphoma, endometrial carcinoma, ovarian carcinoma, bladder cancer, stomach cancer, pancreatic cancer, esophageal cancer, prostate cancer, renal cell carcinoma, melanoma, colorectal cancer, a sarcoma (e.g., a soft tissue sarcoma, synovial sarcoma, Ewing's sarcoma, osteosarcoma,

rhabdomyosarcoma, adult fibrosarcoma, alveolar soft-part sarcoma, angiosarcoma, clear cell sarcoma, desmoplastic small round cell tumor, epithelioid sarcoma, fibromyxoid sarcoma, gastrointestinal stromal tumor, Kaposi sarcoma, liposarcoma, leiomyosarcoma, malignant mesenchymoma malignant peripheral nerve sheath tumors, myxofibrosarcoma, low-grade rhabdomyosarcoma), non-small cell lung cancer (e.g., squamous or adenocarcinoma), stomach cancer, or breast cancer. In some embodiments, the cancer is a malignant, rhabdoid tumor, a CD8+ T-cell lymphoma, endometrial carcinoma, ovarian carcinoma, bladder cancer, stomach cancer, pancreatic cancer, esophageal cancer, prostate cancer, renal cell carcinoma, melanoma, or colorectal cancer. In some embodiments, the cancer is a sarcoma (e.g., synovial sarcoma or Ewing's sarcoma), non-small cell lung cancer (e.g., squamous or adenocarcinoma), stomach cancer, or breast cancer.

In another aspect, the disclosure provides a method for treating a viral infection in a subject in need thereof. This method includes administering to the subject an effective amount of any of the foregoing compounds, or pharmaceutically acceptable salts thereof, or any of the foregoing

pharmaceutical compositions. In some embodiments, the viral infection is an infection with a virus of the Retroviridae family such as the lentiviruses (e.g. Human immunodeficiency virus (HIV) and

deltaretroviruses (e.g., human T cell leukemia virus I (HTLV-I), human T cell leukemia virus II (HTLV-II)); Hepadnaviridae family (e.g. hepatitis B virus (HBV)), Flaviviridae family (e.g. hepatitis C virus (HCV)), Adenoviridae family (e.g. Human Adenovirus), Herpesviridae family (e.g. Human cytomegalovirus

(HCMV), , Epstein-Barr virus, herpes simplex virus 1 (HSV-1 ), herpes simplex virus 2 (HSV-2), human herpesvirus 6 (HHV-6), Herpesvitus K * , CMV, varicella-zoster virus), Papillomaviridae family (e.g. Human Papillomavirus (HPV, HPV E1 )), Parvoviridae family (e.g. Parvovirus B19), Polyomaviridae family (e.g. JC virus and BK virus), Paramyxoviridae family (e.g. Measles virus), Togaviridae family (e.g. Rubella virus).

In another embodiment of any of the foregoing methods, the method further includes

administering to the subject an additional anticancer therapy (e.g., chemotherapeutic or cytotoxic agent or radiotherapy).

In particular embodiments, the additional anticancer therapy is: a chemotherapeutic or cytotoxic agent (e.g., doxorubicin or ifosfamide), a differentiation-inducing agent (e.g., retinoic acid, vitamin D, cytokines), a hormonal agent, an immunological agent, or an anti-angiogenic agent. Chemotherapeutic and cytotoxic agents include, but are not limited to, alkylating agents, cytotoxic antibiotics,

antimetabolites, vinca alkaloids, etoposides, and others (e.g., paclitaxel, taxol, docetaxel, taxotere, cis-platinum). A list of additional compounds having anticancer activity can be found in L. Brunton, B. Chabner and B. Knollman (eds). Goodman and Gilman's The Pharmacological Basis of Therapeutics, Twelfth Edition, 201 1 , McGraw Hill Companies, New York, NY.

In particular embodiments, the compound of the invention and the additional anticancer therapy and any of the foregoing compounds or pharmaceutical compositions are administered within 28 days of each other (e.g., within 21 , 14, 1 0, 7, 5, 4, 3, 2, or 1 days) or within 24 hours (e.g., 12, 6, 3, 2, or 1 hours; or concomitantly) each in an amount that together are effective to treat the subject.

In some embodiments of any of the foregoing methods, the SS18-SSX fusion protein is a

SS18-SSX1 fusion protein, a SS18-SSX2 fusion protein, or a SS18-SSX4 fusion protein.

Chemical Terms

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

The term "acyl," as used herein, represents a hydrogen or an alkyl group, as defined herein, that is attached to a parent molecular group through a carbonyl group, as defined herein, and is exemplified by formyl (i.e., a carboxyaldehyde group), acetyl, trifluoroacetyl, propionyl, and butanoyl. Exemplary unsubstituted acyl groups include from 1 to 6, from 1 to 1 1 , or from 1 to 21 carbons. The term "alkyl," as used herein, refers to a branched or straight-chain monovalent saturated aliphatic hydrocarbon radical of 1 to 20 carbon atoms (e.g., 1 to 16 carbon atoms, 1 to 10 carbon atoms, or 1 to 6 carbon atoms). An alkylene is a divalent alkyl group.

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

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

The term "amino," as used herein, represents -N(R N 1 )2, wherein each R N1 is, independently, H, OH, NO2, N(R N2 ) 2 , S0 2 OR N2 , S0 2 R N2 , SOR N2 , an /V-protecting group, alkyl, alkoxy, aryl, arylalkyl, cycloalkyl, acyl (e.g., acetyl, trifluoroacetyl, or others described herein), wherein each of these recited R N1 groups can be optionally substituted; or two R N1 combine to form an alkylene or heteroalkylene, and wherein each R N2 is, independently, H, alkyl, or aryl. The amino groups of the invention can be an unsubstituted amino (i.e., -NH2) or a substituted amino (i.e., -N(R N1 ) 2 ).

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

The term "arylalkyl," as used herein, represents an alkyl group substituted with an aryl group. Exemplary unsubstituted arylalkyl groups are from 7 to 30 carbons (e.g., from 7 to 16 or from 7 to 20 carbons, such as C1 -6 alkyl Ce-ιο aryl, C1-10 alkyl Ce-ιο aryl, or C1-20 alkyl Ce-ιο aryl), such as, benzyl and phenethyl. In some embodiments, the akyl and the aryl each can be further substituted with 1 , 2, 3, or 4 substituent groups as defined herein for the respective groups.

The term "azido," as used herein, represents a -N3 group.

The term "cyano," as used herein, represents a -CN group.

The terms "carbocyclyl," as used herein, refer to a non-aromatic C3-12 monocyclic, bicyclic, or tricyclic structure in which the rings are formed by carbon atoms. Carbocyclyl structures include cycloalkyl groups and unsaturated carbocyclyl radicals.

The term "cycloalkyl," as used herein, refers to a saturated, non-aromatic, monovalent mono- or polycarbocyclic radical of three to ten, preferably three to six carbon atoms. This term is further exemplified by radicals such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, norbornyl, and adamantyl.

The term "halogen," as used herein, means a fluorine (fluoro), chlorine (chloro), bromine (bromo), or iodine (iodo) radical.

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

The term "heteroalkenyl," as used herein, refers to an alkenyl group, as defined herein, in which one or more of the constituent carbon atoms have been replaced by nitrogen, oxygen, or sulfur. In some embodiments, the heteroalkenyl group can be further substituted with 1 , 2, 3, or 4 substituent groups as described herein for alkenyl groups. Examples of heteroalkenyl groups are an "alkenoxy" which, as used herein, refers alkenyl-O-. A heteroalkenylene is a divalent heteroalkenyl group.

The term "heteroalkynyl," as used herein, refers to an alkynyl group, as defined herein, in which one or more of the constituent carbon atoms have been replaced by nitrogen, oxygen, or sulfur. In some embodiments, the heteroalkynyl group can be further substituted with 1 , 2, 3, or 4 substituent groups as described herein for alkynyl groups. Examples of heteroalkynyl groups are an "alkynoxy" which, as used herein, refers alkynyl-O-. A heteroalkynylene is a divalent heteroalkynyl group.

The term "heteroaryl," as used herein, refers to an aromatic mono- or polycyclic radical of 5 to 12 atoms having at least one aromatic ring containing one, two, or three ring heteroatoms selected from N, O, and S, with the remaining ring atoms being C. One or two ring carbon atoms of the heteroaryl group may be replaced with a carbonyl group. Examples of heteroaryl groups are pyridyl, pyrazoyl, benzooxazolyl, benzoimidazolyl, benzothiazolyl, imidazolyl, oxaxolyl, and thiazolyl.

The term "heteroarylalkyl," as used herein, represents an alkyl group substituted with a heteroaryl group. Exemplary unsubstituted heteroarylalkyl groups are from 7 to 30 carbons (e.g., from 7 to 16 or from 7 to 20 carbons, such as C1-6 alkyl C2-9 heteroaryl, C1-10 alkyl C2-9 heteroaryl, or C1-20 alkyl C2-9 heteroaryl). In some embodiments, the akyl and the heteroaryl each can be further substituted with 1 , 2, 3, or 4 substituent groups as defined herein for the respective groups.

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

The term "heterocyclylalkyl," as used herein, represents an alkyl group substituted with a heterocyclyl group. Exemplary unsubstituted heterocyclylalkyl groups are from 7 to 30 carbons (e.g., from 7 to 16 or from 7 to 20 carbons, such as C1-6 alkyl C2-9 heterocyclyl, C1-10 alkyl C2-9 heterocyclyl, or C1-20 alkyl C2-9 heterocyclyl). In some embodiments, the akyl and the heterocyclyl each can be further substituted with 1 , 2, 3, or 4 substituent groups as defined herein for the respective groups.

The term "hydroxyl," as used herein, represents an -OH group.

The term "/V-protecting group," as used herein, represents those groups intended to protect an amino group against undesirable reactions during synthetic procedures. Commonly used /V-protecting groups are disclosed in Greene, "Protective Groups in Organic Synthesis," 3 rd Edition (John Wiley & Sons, New York, 1999). /V-protecting groups include acyl, aryloyl, or carbamyl groups such as formyl, acetyl, propionyl, pivaloyl, t-butylacetyl, 2-chloroacetyl, 2-bromoacetyl, trifluoroacetyl, trichloroacetyl, phthalyl, o-nitrophenoxyacetyl, a-chlorobutyryl, benzoyl, 4-chlorobenzoyl, 4-bromobenzoyl, 4- nitrobenzoyl, and chiral auxiliaries such as protected or unprotected D, L or D, L-amino acids such as alanine, leucine, and phenylalanine; sulfonyl-containing groups such as benzenesulfonyl, and

p-toluenesulfonyl; carbamate forming groups such as benzyloxycarbonyl, p-chlorobenzyloxycarbonyl, p-methoxybenzyloxycarbonyl, p-nitrobenzyloxycarbonyl, 2-nitrobenzyloxycarbonyl,

p-bromobenzyloxycarbonyl, 3,4-dimethoxybenzyloxycarbonyl, 3,5-dimethoxybenzyloxycarbonyl, 2.4- dimethoxybenzyloxycarbonyl, 4-methoxybenzyloxycarbonyl, 2-nitro-4,5-dimethoxybenzyloxycarbonyl, 3,4,5-trimethoxybenzyloxycarbonyl, 1 -(p-biphenylyl)-l -methylethoxycarbonyl, α,α-dimethyl-

3.5- dimethoxybenzyloxycarbonyl, benzhydryloxy carbonyl, t-butyloxycarbonyl,

diisopropylmethoxycarbonyl, isopropyloxycarbonyl, ethoxycarbonyl, methoxycarbonyl, allyloxycarbonyl, 2,2,2,-trichloroethoxycarbonyl, phenoxycarbonyl, 4-nitrophenoxy carbonyl, fluorenyl-9-methoxycarbonyl, cyclopentyloxycarbonyl, adamantyloxycarbonyl, cyclohexyloxycarbonyl, and phenylthiocarbonyl, arylalkyl groups such as benzyl, triphenylmethyl, and benzyloxymethyl, and silyl groups, such as trimethylsilyl. Preferred /V-protecting groups are alloc, formyl, acetyl, benzoyl, pivaloyl, t-butyl acetyl, alanyl, phenylsulfonyl, benzyl, t-butyloxycarbonyl (Boc), and benzyloxycarbonyl (Cbz).

The term "nitro," as used herein, represents an -NO2 group.

The term "thiol," as used herein, represents an -SH group.

The alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, carbocyclyl (e.g., cycloalkyl), aryl, heteroaryl, and heterocyclyl groups may be substituted or unsubstituted. When substituted, there will generally be 1 to 4 substituents present, unless otherwise specified. Substituents include, for example: aryl (e.g., substituted and unsubstituted phenyl), carbocyclyl (e.g., substituted and unsubstituted cycloalkyl), halogen (e.g., fluoro), hydroxyl, heteroalkyl (e.g., substituted and unsubstituted methoxy, ethoxy, or thioalkoxy), heteroaryl, heterocyclyl, amino (e.g., NH2 or mono- or dialkyl amino), azido, cyano, nitro, or thiol. Aryl, carbocyclyl (e.g., cycloalkyl), heteroaryl, and heterocyclyl groups may also be substituted with alkyl (unsubstituted and substituted such as arylalkyl (e.g., substituted and unsubstituted benzyl)).

Compounds of the invention can have one or more asymmetric carbon atoms and can exist in the form of optically pure enantiomers, mixtures of enantiomers such as, for example, racemates, optically pure diastereoisomers, mixtures of diastereoisomers, diastereoisomeric racemates or mixtures of diastereoisomeric racemates. The optically active forms can be obtained for example by resolution of the racemates, by asymmetric synthesis or asymmetric chromatography (chromatography with a chiral adsorbents or eluant). That is, certain of the disclosed compounds may exist in various stereoisomeric forms. Stereoisomers are compounds that differ only in their spatial arrangement. Enantiomers are pairs of stereoisomers whose mirror images are not superimposable, most commonly because they contain an asymmetrically substituted carbon atom that acts as a chiral center. "Enantiomer" means one of a pair of molecules that are mirror images of each other and are not superimposable. Diastereomers are stereoisomers that are not related as mirror images, most commonly because they contain two or more asymmetrically substituted carbon atoms and represent the configuration of substituents around one or more chiral carbon atoms. Enantiomers of a compound can be prepared, for example, by separating an enantiomer from a racemate using one or more well-known techniques and methods, such as, for example, chiral chromatography and separation methods based thereon. The appropriate technique and/or method for separating an enantiomer of a compound described herein from a racemic mixture can be readily determined by those of skill in the art. "Racemate" or "racemic mixture" means a compound containing two enantiomers, wherein such mixtures exhibit no optical activity; i.e., they do not rotate the plane of polarized light. "Geometric isomer" means isomers that differ in the orientation of substituent atoms in relationship to a carbon-carbon double bond, to a cycloalkyl ring, or to a bridged bicyclic system. Atoms (other than H) on each side of a carbon- carbon double bond may be in an E (substituents are on opposite sides of the carbon- carbon double bond) or Z (substituents are oriented on the same side) configuration. "R," "S," "S * ," "FT," "E," "Z," "cis," and "trans," indicate configurations relative to the core molecule. Certain of the disclosed compounds may exist in atropisomeric forms. Atropisomers are stereoisomers resulting from hindered rotation about single bonds where the steric strain barrier to rotation is high enough to allow for the isolation of the conformers. The compounds of the invention may be prepared as individual isomers by either isomer-specific synthesis or resolved from an isomeric mixture. Conventional resolution techniques include forming the salt of a free base of each isomer of an isomeric pair using an optically active acid (followed by fractional crystallization and regeneration of the free base), forming the salt of the acid form of each isomer of an isomeric pair using an optically active amine (followed by fractional crystallization and regeneration of the free acid), forming an ester or amide of each of the isomers of an isomeric pair using an optically pure acid, amine or alcohol (followed by chromatographic separation and removal of the chiral auxiliary), or resolving an isomeric mixture of either a starting material or a final product using various well known chromatographic methods. When the stereochemistry of a disclosed compound is named or depicted by structure, the named or depicted stereoisomer is at least 60%, 70%, 80%, 90%, 99% or 99.9%) by weight relative to the other

stereoisomers. When a single enantiomer is named or depicted by structure, the depicted or named enantiomer is at least 60%, 70%, 80%, 90%, 99% or 99.9% by weight optically pure. When a single diastereomer is named or depicted by structure, the depicted or named diastereomer is at least 60%, 70%, 80%, 90%, 99% or 99.9% by weight pure. Percent optical purity is the ratio of the weight of the enantiomer or over the weight of the enantiomer plus the weight of its optical isomer. Diastereomeric purity by weight is the ratio of the weight of one diastereomer or over the weight of all the diastereomers. When the stereochemistry of a disclosed compound is named or depicted by structure, the named or depicted stereoisomer is at least 60%, 70%, 80%, 90%, 99% or 99.9% by mole fraction pure relative to the other stereoisomers. When a single enantiomer is named or depicted by structure, the depicted or named enantiomer is at least 60%, 70%, 80%, 90%, 99% or 99.9% by mole fraction pure. When a single diastereomer is named or depicted by structure, the depicted or named diastereomer is at least 60%, 70%, 80%, 90%, 99% or 99.9% by mole fraction pure. Percent purity by mole fraction is the ratio of the moles of the enantiomer or over the moles of the enantiomer plus the moles of its optical isomer.

Similarly, percent purity by moles fraction is the ratio of the moles of the diastereomer or over the moles of the diastereomer plus the moles of its isomer. When a disclosed compound is named or depicted by structure without indicating the stereochemistry, and the compound has at least one chiral center, it is to be understood that the name or structure encompasses either enantiomer of the compound free from the corresponding optical isomer, a racemic mixture of the compound or mixtures enriched in one enantiomer relative to its corresponding optical isomer. When a disclosed compound is named or depicted by structure without indicating the stereochemistry and has two or more chiral centers, it is to be understood that the name or structure encompasses a diastereomer free of other diastereomers, a number of diastereomers free from other diastereomeric pairs, mixtures of diastereomers, mixtures of diastereomeric pairs, mixtures of diastereomers in which one diastereomer is enriched relative to the other

diastereomer(s) or mixtures of diastereomers in which one or more diastereomer is enriched relative to the other diastereomers. The invention embraces all of these forms. Definitions

In this application, unless otherwise clear from context, (i) the term "a" may be understood to mean "at least one"; (ii) the term "or" may be understood to mean "and/or"; (iii) the terms "comprising" and "including" may be understood to encompass itemized components or steps whether presented by themselves or together with one or more additional components or steps; and (iv) the terms "about" and "approximately" may be understood to permit standard variation as would be understood by those of ordinary skill in the art; and (v) where ranges are provided, endpoints are included.

As used herein, the term "administration" refers to the administration of a composition (e.g., a compound or a preparation that includes a compound as described herein) to a subject or system.

Administration to an animal subject (e.g., to a human) may be by any appropriate route. For example, in some embodiments, administration may be bronchial (including by bronchial instillation), buccal, enteral, interdermal, intra-arterial, intradermal, intragastric, intramedullary, intramuscular, intranasal,

intraperitoneal, intrathecal, intravenous, intraventricular, mucosal, nasal, oral, rectal, subcutaneous, sublingual, topical, tracheal (including by intratracheal instillation), transdermal, vaginal and vitreal.

As used herein, the term "BAF complex" refers to the BRG1 - or HRBM-associated factors complex in a human cell.

As used herein, the term "BAF47" refers to a peptide having the sequence of SEQ ID NO:1 and/or a polypeptide with at least 90% identity (e.g., at least 95% identity, at least 98% identity, at least 99% identity) to SEQ ID NO:1 .

SEQ ID NO: 1

MMMMALSKTFGQKPVK 'QLEDDGE TMIGSEVGKYLRMFRGSLYKRYPSLWRRLATVEERKKIVASSHGKKTKPNTK DHGYTTL7\TSVTLLK7\SEVEEILDGNDEKYK7\VS I STEPPTYLREQK7\KRNSQWVPTLPNSSHHLDAVPCSTTINRN RMGRDKKRTFPLCFDDHDPAVIHENASQPEVLVPIRLDMEIDGQKLRDAFTWNMNEKLMT PEMFSEILCDDLDLNPL TFVPAIASAIRQQIESYPTDS ILEDQSDQRVI IKLN IHVGNI SLVDQFE DMSEKENSPEKFALKLCSELGLGGEFV TTIAYS IRGQLSWHQKTYAFSENPLPTVEIAIRNTGDADQWCPLLETLTDAEMEKKIRDQDRNTRR MRRLANTAPAW

The term "cancer" refers to a condition caused by the proliferation of malignant neoplastic cells, such as tumors, neoplasms, carcinomas, sarcomas, leukemias, and lymphomas.

As used herein, a "combination therapy" or "administered in combination" means that two (or more) different agents or treatments are administered to a subject as part of a defined treatment regimen for a particular disease or condition. The treatment regimen defines the doses and periodicity of administration of each agent such that the effects of the separate agents on the subject overlap. In some embodiments, the delivery of the two or more agents is simultaneous or concurrent and the agents may be co-formulated. In other embodiments, the two or more agents are not co-formulated and are administered together or in a sequential manner as part of a prescribed regimen. In some embodiments, administration of two or more agents or treatments in combination is such that the reduction in a symptom, or other parameter related to the disorder is greater than what would be observed with one agent or treatment delivered alone or in the absence of the other. The effect of the two treatments can be partially additive, wholly additive, or greater than additive (e.g., synergistic). Sequential or substantially simultaneous administration of each therapeutic agent can be effected by any appropriate route including, but not limited to, oral routes, intravenous routes, intramuscular routes, and direct absorption through mucous membrane tissues. The therapeutic agents can be administered by the same route or by different routes. For example, a first therapeutic agent of the combination may be administered by intravenous injection while a second therapeutic agent of the combination may be administered orally.

By "decreasing Wnt/p-catenin signaling," is meant decreasing the level of any protein or activity of any protein in the Wnt/ β-catenin signaling pathway. A non-limiting example of decreasing Wnt/ β-catenin signaling is decreasing the level β-catenin in a cell. Methods to determine the level of Wnt^-catenin signaling are known in the art, e.g., as described in MacDonald et al. Developmental Cell, 2009, 17, 9-26, the methods of which are incorporated herein by reference.

By "determining the level of a protein" is meant the detection of a protein, or an mRNA encoding the protein, by methods known in the art either directly or indirectly. "Directly determining" means performing a process (e.g., performing an assay or test on a sample or "analyzing a sample" as that term is defined herein) to obtain the physical entity or value. "Indirectly determining" refers to receiving the physical entity or value from another party or source (e.g., a third party laboratory that directly acquired the physical entity or value). Methods to measure protein level generally include, but are not limited to, western blotting, immunoblotting, enzyme-linked immunosorbent assay (ELISA), radioimmunoassay (RIA), immunoprecipitation, immunofluorescence, surface plasmon resonance, chemiluminescence, fluorescent polarization, phosphorescence, immunohistochemical analysis, matrix-assisted laser desorption/ionization time-of-flight (MALDI-TOF) mass spectrometry, liquid chromatography (LC)-mass spectrometry, microcytometry, microscopy, fluorescence activated cell sorting (FACS), and flow cytometry, as well as assays based on a property of a protein including, but not limited to, enzymatic activity or interaction with other protein partners. Methods to measure mRNA levels are known in the art.

As used herein, the term "disorder related to a SS18-SSX fusion protein," refers to any disease or disorder that may derive a therapeutic benefit from modulation (e.g., inhibition) of the activity of a SS18- SSX fusion protein, e.g., synovial sarcoma.

As used herein, the term "disorder related to BAF47," refers to any disease or disorder that may derive a therapeutic benefit from modulating the activity of BAF47, e.g., synovial sarcoma.

An "effective amount" of a compound may vary according to factors such as the disease state, age, sex, and weight of the individual, and the ability of the compound to elicit the desired response. A therapeutically effective amount encompasses an amount in which any toxic or detrimental effects of the compound are outweighed by the therapeutically beneficial effects. A therapeutically effective amount also encompasses an amount sufficient to confer benefit, e.g., clinical benefit.

By "increasing the activity of a BAF complex," is meant increasing the level of an activity related to a BAF complex, or a related downstream effect. A non-limiting example of increasing an activity of a BAF complex is Sox2 inactivation. The activity level of a BAF complex may be measured using any method known in the art, e.g., the methods described in Kadoch et al. Cell, 2013, 153, 71 -85, the methods of which are herein incorporated by reference.

By "inhibiting the activity of an SS18-SSX fusion protein," is meant decreasing the level of an activity related to an SS18-SSX fusion protein, or a related downstream effect. A non-limiting example of inhibition of an activity of an SS18-SSX fusion protein is increasing the level of wild-type BAF complex in a cell. The activity level of an SS18-SSX fusion protein may be measured using any method known in the art, e.g., the methods described in Kadoch et al. Cell, 2013, 153, 71 -85, the methods of which are herein incorporated by reference.

By "level" is meant a level of a protein, or mRNA encoding the protein, as compared to a reference. The reference can be any useful reference, as defined herein. By a "decreased level" or an "increased level" of a protein is meant a decrease or increase in protein level, as compared to a reference (e.g., a decrease or an increase by about 5%, about 1 0%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, about 100%, about 1 50%, about 200%, about 300%, about 400%, about 500%, or more; a decrease or an increase of more than about 10%, about 15%, about 20%, about 50%, about 75%, about 100%, or about 200%, as compared to a reference; a decrease or an increase by less than about 0.01 -fold, about 0.02-fold, about 0.1 -fold, about 0.3-fold, about 0.5-fold, about 0.8-fold, or less; or an increase by more than about 1 .2-fold, about 1 .4-fold, about 1 .5-fold, about 1 .8-fold, about 2.0-fold, about 3.0-fold, about 3.5-fold, about 4.5-fold, about 5.0-fold, about 10-fold, about 15-fold, about 20-fold, about 30-fold, about 40-fold, about 50-fold, about 100-fold, about 1000-fold, or more). A level of a protein may be expressed in mass/vol (e.g., g/dL, mg/mL, g/mL, ng/mL) or percentage relative to total protein or mRNA in a sample.

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

A "pharmaceutically acceptable excipient," as used herein, refers any ingredient other than the compounds described herein (for example, a vehicle capable of suspending or dissolving the active compound) and having the properties of being substantially nontoxic and non-inflammatory in a patient. Excipients may include, for example: antiadherents, antioxidants, binders, coatings, compression aids, disintegrants, dyes (colors), emollients, emulsifiers, fillers (diluents), film formers or coatings, flavors, fragrances, glidants (flow enhancers), lubricants, preservatives, printing inks, sorbents, suspensing or dispersing agents, sweeteners, and waters of hydration. Exemplary excipients include, but are not limited to: butylated hydroxytoluene (BHT), calcium carbonate, calcium phosphate (dibasic), calcium stearate, croscarmellose, crosslinked polyvinyl pyrrolidone, citric acid, crospovidone, cysteine, ethylcellulose, gelatin, hydroxypropyl cellulose, hydroxypropyl methylcellulose, lactose, magnesium stearate, maltitol, mannitol, methionine, methylcellulose, methyl paraben, microcrystalline cellulose, polyethylene glycol, polyvinyl pyrrolidone, povidone, pregelatinized starch, propyl paraben, retinyl palmitate, shellac, silicon dioxide, sodium carboxymethyl cellulose, sodium citrate, sodium starch glycolate, sorbitol, starch (corn), stearic acid, sucrose, talc, titanium dioxide, vitamin A, vitamin E, vitamin C, and xylitol.

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

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

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

By a "reference" is meant any useful reference used to compare protein or mRNA levels. The reference can be any sample, standard, standard curve, or level that is used for comparison purposes. The reference can be a normal reference sample or a reference standard or level. A "reference sample" can be, for example, a control, e.g., a predetermined negative control value such as a "normal control" or a prior sample taken from the same subject; a sample from a normal healthy subject, such as a normal cell or normal tissue; a sample (e.g., a cell or tissue) from a subject not having a disease; a sample from a subject that is diagnosed with a disease, but not yet treated with a compound of the invention; a sample from a subject that has been treated by a compound of the invention; or a sample of a purified protein

(e.g., any described herein) at a known normal concentration. By "reference standard or level" is meant a value or number derived from a reference sample. A "normal control value" is a pre-determined value indicative of non-disease state, e.g., a value expected in a healthy control subject. Typically, a normal control value is expressed as a range ("between X and Y"), a high threshold ("no higher than X"), or a low threshold ("no lower than X"). A subject having a measured value within the normal control value for a particular biomarker is typically referred to as "within normal limits" for that biomarker. A normal reference standard or level can be a value or number derived from a normal subject not having a disease or disorder (e.g., cancer); a subject that has been treated with a compound of the invention. In preferred embodiments, the reference sample, standard, or level is matched to the sample subject sample by at least one of the following criteria: age, weight, sex, disease stage, and overall health. A standard curve of levels of a purified protein, e.g., any described herein, within the normal reference range can also be used as a reference.

As used herein, the term "subject" refers to any organism to which a composition in accordance with the invention may be administered, e.g., for experimental, diagnostic, prophylactic, and/or therapeutic purposes. Typical subjects include any animal (e.g., mammals such as mice, rats, rabbits, non-human primates, and humans). A subject may seek or be in need of treatment, require treatment, be receiving treatment, be receiving treatment in the future, or be a human or animal who is under care by a trained professional for a particular disease or condition.

As used herein, the terms "treat," "treated," or "treating" mean both therapeutic treatment and prophylactic or preventative measures wherein the object is to prevent or slow down (lessen) an undesired physiological condition, disorder, or disease, or obtain beneficial or desired clinical results.

Beneficial or desired clinical results include, but are not limited to, alleviation of symptoms; diminishment of the extent of a condition, disorder, or disease; stabilized (i.e., not worsening) state of condition, disorder, or disease; delay in onset or slowing of condition, disorder, or disease progression; amelioration of the condition, disorder, or disease state or remission (whether partial or total), whether detectable or undetectable; an amelioration of at least one measurable physical parameter, not necessarily discernible by the patient; or enhancement or improvement of condition, disorder, or disease. Treatment includes eliciting a clinically significant response without excessive levels of side effects. Treatment also includes prolonging survival as compared to expected survival if not receiving treatment.

Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Methods and materials are described herein for use in the present disclosure; other, suitable methods and materials known in the art can also be used. The materials, methods, and examples are illustrative only and not intended to be limiting. All publications, patent applications, patents, sequences, database entries, and other references mentioned herein are incorporated by reference in their entirety. In case of conflict, the present specification, including definitions, will control.

The details of one or more embodiments of the invention are set forth in the description below. Other features, objects, and advantages of the invention will be apparent from the description and from the claims.

Detailed Description of the Invention

The invention features compounds useful for the inhibition of the activity of SS18-SSX fusion proteins, e.g., for the treatment of cancer such as synovial sarcoma. Exemplary compounds described herein include compounds having a structure according to formula I:

Formula I

or pharmaceutically acceptable salts thereof.

In some embodiments, the compound has the structure of any one of compounds 1 -61 in Table 1 Other embodiments, as well as exemplary methods for the synthesis or production of these pounds, are described herein.

Pharmaceutical Uses

The compounds described herein are useful in the methods of the invention and, while not bound by theory, are believed to exert their desirable effects through their ability to modulate the level, status, and/or activity of a BAF complex, for example, to inhibit the activity or decrease the level of SS1 8-SSX fusion proteins in a cell and/or stabilize BAF47 within the BAF complex in a mammal.

Another aspect of the present invention relates to methods of treating disorders related to SS18-SSX fusion proteins such as cancer (e.g., synovial sarcoma) in a subject in need thereof. In some embodiments, the compound is administered in an amount and for a time effective to result in one of (or more, e.g., 2 or more, 3 or more, 4 or more of): (a) reduced tumor size, (b) reduced rate of tumor growth, (c) increased tumor cell death (d) reduced tumor progression, (e) reduced number of metastases, (f) reduced rate of metastasis, (g) decreased tumor recurrence (h) increased survival of subject, or (i) increased progression free survival of subject.

Treating cancer can result in a reduction in size or volume of a tumor. For example, after treatment, tumor size is reduced by 5% or greater (e.g., 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or greater) relative to its size prior to treatment. Size of a tumor may be measured by any reproducible means of measurement. For example, the size of a tumor may be measured as a diameter of the tumor.

Treating cancer may further result in a decrease in number of tumors. For example, after treatment, tumor number is reduced by 5% or greater (e.g., 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or greater) relative to number prior to treatment. Number of tumors may be measured by any reproducible means of measurement, e.g., the number of tumors may be measured by counting tumors visible to the naked eye or at a specified magnification (e.g., 2x, 3x, 4x, 5x, 10x, or 50x).

Treating cancer can result in a decrease in number of metastatic nodules in other tissues or organs distant from the primary tumor site. For example, after treatment, the number of metastatic nodules is reduced by 5% or greater (e.g., 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or greater) relative to number prior to treatment. The number of metastatic nodules may be measured by any reproducible means of measurement. For example, the number of metastatic nodules may be measured by counting metastatic nodules visible to the naked eye or at a specified magnification (e.g., 2x, 10x, or 50x). Treating cancer can result in an increase in average survival time of a population of subjects treated according to the present invention in comparison to a population of untreated subjects. For example, the average survival time is increased by more than 30 days (more than 60 days, 90 days, or 120 days). An increase in average survival time of a population may be measured by any reproducible means. An increase in average survival time of a population may be measured, for example, by calculating for a population the average length of survival following initiation of treatment with the compound of the invention. An increase in average survival time of a population may also be measured, for example, by calculating for a population the average length of survival following completion of a first round of treatment with a pharmaceutically acceptable salt of the invention.

Treating cancer can also result in a decrease in the mortality rate of a population of treated subjects in comparison to an untreated population. For example, the mortality rate is decreased by more than 2% (e.g., more than 5%, 10%, or 25%). A decrease in the mortality rate of a population of treated subjects may be measured by any reproducible means, for example, by calculating for a population the average number of disease-related deaths per unit time following initiation of treatment with a pharmaceutically acceptable salt of the invention. A decrease in the mortality rate of a population may also be measured, for example, by calculating for a population the average number of disease-related deaths per unit time following completion of a first round of treatment with a pharmaceutically acceptable salt of the invention.

Combination Formulations and Uses Thereof

The compounds of the invention can be combined with one or more therapeutic agents. In particular, the therapeutic agent can be one that treats or prophylactically treats any cancer described herein. Combination Therapies

A compound of the invention can be used alone or in combination with an additional therapeutic agent, e.g., other agents that treat cancer or symptoms associated therewith, or in combination with other types of treatment to treat cancer. In combination treatments, the dosages of one or more of the therapeutic compounds may be reduced from standard dosages when administered alone. For example, doses may be determined empirically from drug combinations and permutations or may be deduced by isobolographic analysis (e.g., Black et al., Neurology 65:S3-S6, 2005). In this case, dosages of the compounds when combined should provide a therapeutic effect.

In some embodiments, the second therapeutic agent is a chemotherapeutic agent (e.g., a cytotoxic agent or other chemical compound useful in the treatment of cancer). These include alkylating agents, antimetabolites, folic acid analogs, pyrimidine analogs, purine analogs and related inhibitors, vinca alkaloids, epipodopyyllotoxins, antibiotics, L-Asparaginase, topoisomerase inhibitors, interferons, platinum coordination complexes, anthracenedione substituted urea, methyl hydrazine derivatives, adrenocortical suppressant, adrenocorticosteroides, progestins, estrogens, antiestrogen, androgens, antiandrogen, and gonadotropin-releasing hormone analog. Also included is 5-fluorouracil (5-FU), leucovorin (LV), irenotecan, oxaliplatin, capecitabine, paclitaxel and doxetaxel. Non-limiting examples of chemotherapeutic agents include alkylating agents such as thiotepa and cyclosphosphamide; alkyl sulfonates such as busulfan, improsulfan and piposulfan; aziridines such as benzodopa, carboquone, meturedopa, and uredopa; ethylenimines and methylamelamines including altretamine,

triethylenemelamine, trietylenephosphoramide, triethiylenethiophosphoramide and trimethylolomelamine; acetogenins (especially bullatacin and bullatacinone); a camptothecin (including the synthetic analogue topotecan); bryostatin; callystatin; CC-1065 (including its adozelesin, carzelesin and bizelesin synthetic analogues); cryptophycins (particularly cryptophycin 1 and cryptophycin 8); dolastatin ; duocarmycin (including the synthetic analogues, KW-2189 and CB1 -TM1 ); eleutherobin ; pancratistatin; a sarcodictyin; spongistatin; nitrogen mustards such as chlorambucil, chlornaphazine, cholophosphamide, estramustine, ifosfamide, mechlorethamine, mechlorethamine oxide hydrochloride, melphalan, novembichin, phenesterine, prednimustine, trofosfamide, uracil mustard; nitrosureas such as carmustine, chlorozotocin, fotemustine, lomustine, nimustine, and ranimnustine; antibiotics such as the enediyne antibiotics (e.g., calicheamicin, especially calicheamicin gammall and calicheamicin omegall (see, e.g., Agnew, Chem. Intl. Ed Engl. 33:1 83-186 (1994)); dynemicin, including dynemicin A; bisphosphonates, such as clodronate; an esperamicin; as well as neocarzinostatin chromophore and related chromoprotein enediyne antiobiotic chromophores), aclacinomysins, actinomycin, authramycin, azaserine, bleomycins, cactinomycin, carabicin, caminomycin, carzinophilin, chromomycinis, dactinomycin, daunorubicin, detorubicin, 6-diazo- 5-oxo-L-norleucine, ADRIAMYCIN.RTM. doxorubicin (including morpholino-doxorubicin,

cyanomorpholino-doxorubicin, 2-pyrrolino-doxorubicin and deoxydoxorubicin), epirubicin, esorubicin, idarubicin, marcellomycin, mitomycins such as mitomycin C, mycophenolic acid, nogalamycin, olivomycins, peplomycin, potfiromycin, puromycin, quelamycin, rodorubicin, streptonigrin, streptozocin, tubercidin, ubenimex, zinostatin, zorubicin; anti-metabolites such as methotrexate and 5-fluorouracil (5-FU); folic acid analogues such as denopterin, methotrexate, pteropterin, trimetrexate; purine analogs such as fludarabine, 6-mercaptopurine, thiamiprine, thioguanine; pyrimidine analogs such as ancitabine, azacitidine, 6-azauridine, carmofur, cytarabine, dideoxyuridine, doxifluridine, enocitabine, floxuridine; androgens such as calusterone, dromostanolone propionate, epitiostanol, mepitiostane, testolactone; anti-adrenals such as aminoglutethimide, mitotane, trilostane; folic acid replenisher such as frolinic acid; aceglatone; aldophosphamide glycoside; aminolevulinic acid; eniluracil; amsacrine; bestrabucil;

bisantrene; edatraxate; defofamine; demecolcine; diaziquone; elfomithine; elliptinium acetate; an epothilone; etoglucid; gallium nitrate; hydroxyurea; lentinan; lonidainine; maytansinoids such as maytansine and ansamitocins; mitoguazone; mitoxantrone; mopidanmol; nitraerine; pentostatin ;

phenamet; pirarubicin; losoxantrone; podophyllinic acid; 2-ethylhydrazide; procarbazine; PSK.RTM. polysaccharide complex (JHS Natural Products, Eugene, Oreg.); razoxane; rhizoxin; sizofuran;

spirogermanium ; tenuazonic acid; triaziquone; 2,2',2"-trichlorotriethylamine; trichothecenes (especially T-2 toxin, verracurin A, roridin A and anguidine); urethan; vindesine; dacarbazine; mannomustine;

mitobronitol; mitolactol; pipobroman; gacytosine; arabinoside ("Ara-C"); cyclophosphamide; thiotepa; taxoids, e.g., Taxol®, paclitaxel (Bristol-Myers Squibb Oncology, Princeton, N.J.), ABraxane®.

Cremophor-free, albumin-engineered nanoparticle formulation of paclitaxel (American Pharmaceutical Partners, Schaumberg, III.), and TAXOTERE® doxetaxel (Rhone-Poulenc Rorer, Antony, France);

chloranbucil; GEMZAR® gemcitabine; 6-thioguanine; mercaptopurine; methotrexate; platinum

coordination complexes such as cisplatin, oxaliplatin and carboplatin; vinblastine; platinum; etoposide (VP-16); ifosfamide; mitoxantrone; vincristine; NAVELBINE.RTM. vinorelbine; novantrone; teniposide; edatrexate; daunomycin; aminopterin; xeloda; ibandronate; irinotecan (e.g., CPT-1 1 ); topoisomerase inhibitor RFS 2000; difluoromethylornithine (DMFO); retinoids such as retinoic acid; capecitabine; and pharmaceutically acceptable salts, acids or derivatives of any of the above. Two or more

chemotherapeutic agents can be used in a cocktail to be administered in combination with the first therapeutic agent described herein. Suitable dosing regimens of combination chemotherapies are known in the art and described in, for example, Saltz et al. (1999) Proc ASCO 18:233a and Douillard et al.

(2000) Lancet 355:1041 -7.

In some embodiments, the second therapeutic agent is a therapeutic agent which is a biologic such a cytokine (e.g., interferon or an interleukin (e.g., IL-2)) used in cancer treatment. In other embodiments the biologic is an anti-angiogenic agent, such as an anti-VEGF agent, e.g., bevacizumab (Avastin®). In some embodiments the biologic is an immunoglobulin-based biologic, e.g., a monoclonal antibody (e.g., a humanized antibody, a fully human antibody, an Fc fusion protein or a functional fragment thereof) that agonizes a target to stimulate an anti-cancer response, or antagonizes an antigen important for cancer. Such agents include Rituxan (Rituximab); Zenapax (Daclizumab); Simulect (Basiliximab); Synagis (Palivizumab); Remicade (Infliximab); Herceptin (Trastuzumab); Mylotarg

(Gemtuzumab ozogamicin); Campath (Alemtuzumab); Zevalin (Ibritumomab tiuxetan); Humira

(Adalimumab); Xolair (Omalizumab); Bexxar (Tositumomab-l-131 ); Raptiva (Efalizumab); Erbitux (Cetuximab); Avastin (Bevacizumab); Tysabri (Natalizumab); Actemra (Tocilizumab); Vectibix

(Panitumumab); Lucentis (Ranibizumab); Soliris (Eculizumab); Cimzia (Certolizumab pegol); Simponi (Golimumab); llaris (Canakinumab); Stelara (Ustekinumab); Arzerra (Ofatumumab); Prolia (Denosumab); Numax (Motavizumab); ABThrax (Raxibacumab); Benlysta (Belimumab); Yervoy (Ipilimumab); Adcetris (Brentuximab Vedotin); Perjeta (Pertuzumab); Kadcyla (Ado-trastuzumab emtansine); and Gazyva (Obinutuzumab). Also included are antibody-drug conjugates.

The second agent may be a therapeutic agent which is a non-drug treatment. For example, the second therapeutic agent is radiation therapy, cryotherapy, hyperthermia and/or surgical excision of tumor tissue.

The second agent may be a checkpoint inhibitor. In one embodiment, the inhibitor of checkpoint is an inhibitory antibody (e.g., a monospecific antibody such as a monoclonal antibody). The antibody may be, e.g., humanized or fully human. In other embodiments, the inhibitor of checkpoint is a fusion protein, e.g., an Fc-receptor fusion protein. In some embodiments, the inhibitor of checkpoint is an agent, such as an antibody, that interacts with a checkpoint protein. In other embodiments, the inhibitor of checkpoint is an agent, such as an antibody, that interacts with the ligand of a checkpoint protein. In one embodiment, the inhibitor of checkpoint is an inhibitor (e.g., an inhibitory antibody or small molecule inhibitor) of CTLA-4 (e.g., an anti-CTLA4 antibody such as ipilimumab/Yervoy or tremelimumab). In on embodiment, the inhibitor of checkpoint is an inhibitor (e.g., an inhibitory antibody or small molecule inhibitor) of PD-1 (e.g., nivolumab/Opdivo®; pembrolizumab/Keytruda®; pidilizumab/CT-01 1 ). In one embodiment, the inhibitor of checkpoint is an inhibitor (e.g., an inhibitory antibody or small molecule inhibitor) of PDL1 (e.g., MPDL3280A/RG7446; MEDI4736; MSB0010718C; BMS 936559). In one embodiment, the inhibitor of checkpoint is an inhibitor (e.g., an inhibitory antibody or Fc fusion or small molecule inhibitor) of PDL2 (e.g., a PDL2/lg fusion protein such as AMP 224). In one embodiment, the inhibitor of checkpoint is an inhibitor (e.g., an inhibitory antibody or small molecule inhibitor) of B7-H3 (e.g., MGA271 ), B7-H4, BTLA, HVEM, TIM3, GAL9, LAG 3, VISTA, KIR, 2B4, CD160, CGEN-1 5049, CHK 1 , CHK2, A2aR, B-7 family ligands, or a combination thereof.

In any of the combination embodiments described herein, the first and second therapeutic agent are administered simultaneously or sequentially, in either order. The first therapeutic agent may be administered immediately, up to 1 hour, up to 2 hours, up to 3 hours, up to 4 hours, up to 5 hours, up to 6 hours, up to 7 hours, up to, 8 hours, up to 9 hours, up to 10 hours, up to 1 1 hours, up to 12 hours, up to 13 hours, 14 hours, up to hours 16, up to 17 hours, up 18 hours, up to 1 9 hours up to 20 hours, up to 21 hours, up to 22 hours, up to 23 hours up to 24 hours or up to 1 -7, 1 -14, 1 -21 or 1 -30 days before or after the second therapeutic agent.

Pharmaceutical Compositions

The compounds of the invention are preferably formulated into pharmaceutical compositions for administration to human subjects in a biologically compatible form suitable for administration in vivo. Accordingly, in another aspect, the present invention provides a pharmaceutical composition comprising a compound of the invention in admixture with a suitable diluent, carrier, or excipient.

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

A compound of the invention may be orally administered, for example, with an inert diluent or with an assimilable edible carrier, or it may be enclosed in hard or soft shell gelatin capsules, or it may be compressed into tablets, or it may be incorporated directly with the food of the diet. For oral therapeutic administration, a compound of the invention may be incorporated with an excipient and used in the form of ingestible tablets, buccal tablets, troches, capsules, elixirs, suspensions, syrups, and wafers.

A compound of the invention may also be administered parenterally. Solutions of a compound of the invention can be prepared in water suitably mixed with a surfactant, such as hydroxypropylcellulose. Dispersions can also be prepared in glycerol, liquid polyethylene glycols, DMSO and mixtures thereof with or without alcohol, and in oils. Under ordinary conditions of storage and use, these preparations may contain a preservative to prevent the growth of microorganisms. Conventional procedures and ingredients for the selection and preparation of suitable formulations are described, for example, in Remington's Pharmaceutical Sciences (2003, 20 th ed.) and in The United States Pharmacopeia: The National Formulary (USP 24 NF19), published in 1999.

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

Compositions for nasal administration may conveniently be formulated as aerosols, drops, gels, and powders. Aerosol formulations typically include a solution or fine suspension of the active substance in a physiologically acceptable aqueous or non-aqueous solvent and are usually presented in single or multidose quantities in sterile form in a sealed container, which can take the form of a cartridge or refill for use with an atomizing device. Alternatively, the sealed container may be a unitary dispensing device, such as a single dose nasal inhaler or an aerosol dispenser fitted with a metering valve which is intended for disposal after use. Where the dosage form comprises an aerosol dispenser, it will contain a propellant, which can be a compressed gas, such as compressed air or an organic propellant, such as fluorochlorohydrocarbon. The aerosol dosage forms can also take the form of a pump-atomizer.

Compositions suitable for buccal or sublingual administration include tablets, lozenges, and pastilles, where the active ingredient is formulated with a carrier, such as sugar, acacia, tragacanth, gelatin, and glycerine. Compositions for rectal administration are conveniently in the form of suppositories containing a conventional suppository base, such as cocoa butter.

The compounds of the invention may be administered to an animal, e.g., a human, alone or in combination with pharmaceutically acceptable carriers, as noted herein, the proportion of which is determined by the solubility and chemical nature of the compound, chosen route of administration, and standard pharmaceutical practice.

Dosages

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

Alternatively, the dosage amount can be calculated using the body weight of the patient. For example, the dose of a compound, or pharmaceutical composition thereof, administered to a patient may range from 0.1 -50 mg/kg (e.g., 0.25-25 mg/kg). In exemplary, non-limiting embodiments, the dose may range from 0.5-5.0 mg/kg (e.g., 0.5, 1 .0, 1 .5, 2.0, 2.5, 3.0, 3.5, 4.0, 4.5, or 5.0 mg/kg) or from 5.0-20 mg/kg (e.g., 5.5, 6.0, 6.5, 7.0, 7.5, 8.0, 8.5, 9.0, 9.5, 10, 1 1 , 12, 13, 14, 15, 16, 17, 18, 19, or 20 mg/kg). EXAMPLES

The synthesis of compounds of this invention can be synthesized according to one or more of the examples shown below. The variables recited in the general schemes below are as defined for Formulae l-V.

Example 1 . Synthesis of Compounds of the Invention

Preparation of 1 -(6-methoxy-1 H-indol-3-yl)-N,N-dimethylmethanamine

A B

To a chilled solution of compound C (50.1 g, 367 mmol, 1 .50 eq) and formaline (19.8 g, 245 mmol, 1 .00 eq) in AcOH (360 mL) was added compound A (36.0 g, 245 mmol, 1 .00 eq) in three equal portions at 0 °C. Then the reaction mixture was warmed to 25 °C and stirred for 12 hrs. The solution was poured into ice water (500 mL) and basified to pH 12 with 50 % NaOH, and extracted with ether (3 * 500 mL), dried over Na2S04 and concentrated. The compound B (50.0 g, crude) which was used into the next step without further purification.

1 H NMR: 400MHz CDC

δ 8.34 (s, 1 H), 7.48 (d, J = 9.2 Hz, 1 H), 6.87 (d, J = 2.0 Hz, 1 H), 6.69 - 6.73 (m, 2H), 3.73 (s, 3H), 3.52 (s, 2H), 2.20 (s, 6 H) ppm. Preparation of 6-methoxy-3-((trimethyl-l4-azaneyl)methyl)-1 H-indole iodide

B

CH3I (104 g, 734 mmol, 3.00 eq) was added to a solution of compound B (50.0 g, 245 mmol, 1 .00 eq) in toluene (500 mL) at 0 °C, and it was stirred at 25 °C for 12 hrs. The product was filtered, dried under vacuum. Compound D (35.0 g, 96.6 mmol) was obtained as a white solid. Which was used without purification

Preparation of 2- 6-methoxy-1 H-indol-3-yl)acetonitrile

D E

To a solution of compound D (36.0 g, 1 04 mmol, 1 .00 eq) in EtOH (360 mL) was added NaCN

(10.2 g, 208 mmol, 2.00 eq), and the reaction was stirred at 80 °C for 5hrs. The reaction was concentrated under reduced pressure to give a residue. The residue was purified by column

chromatography (S1O2, DCM). Compound E (10.0 g, 53.7 mmol) was obtained as a light yellow solid.

δ 7.98 (s, 1 H), 7.38 (d, J = 8.4 Hz, 1 H), 7.05 - 7.00 (m, 1 H), 6.72 - 6.86 (m, 2H), 3.77 (s, 3H), 3.72 (s, 2H) ppm

Preparation of -(6-methoxy-1 -((2-(trimethylsilyl)ethoxy)methyl)-1 H-indol-3-yl)acetonitrile.

E F

A solution of compound E (15.0 g, 80.5 mmol) in dry THF (80.0 mL) was added at 0 °C under N 2 atmosphere to a suspension of NaH (5.80 g, 145 mmol) in THF (35.0 mL). After 30 min stirring at 0 °C, was added drop-wise SEMCI (22.8 g, 137 mmol) in THF (35.0 mL) under N2 atmosphere. The orange suspension was stirred for 5 hr at 25 °C. The reaction mixture was poured into ice-water (200 mL). The aqueous phase was extracted with ethyl acetate (200 mL * 3). The combined organic phase was washed with brine (200 mL * 2), dried with anhydrous Na2S04, filtered and concentrated in vacuum. The residue was purified by column chromatography (S1O2, Petroleum ethenEthyl acetate = 10:1 to 1 :1 ). Compound F (17.0 g, 53.7 mmol) was obtained as a yellow oil. 1 H NMR: 400MHz CDC

δ 7.47 (d, J = 8.4 Hz, 1 H), 7.13 (s, 1 H), 7.00 (d, J = 2.0 Hz, 1 H), 6.91 (dd, = 2.4, 8.4 Hz, 1 H), 5.34 - 5.54 (m, 2H), 3.90 - 3.93 (m, 3H), 3.83 (s, 2H), 3.50 - 3.57 (m, 2H), 1 .04 - 0.85 (m, 2H), 0.00 (s, 9H)

Preparation of 1 -benzyl-4-(6-methoxy-1 -((2-(trimethylsilyl)ethoxy)methyl)-1 H-indol-3-yl)piperidine-4- carbonitrile

To a solution of compound F (21 .0 g, 66.4 mmol) in DMF (90.0 mL) was added NaH (6.64 g, 166 mmol, 2.50 eg) at 0 °C under N2 atmosphere. After 30 min stirring at 0 °C, compound G (18.5 g, 79.6 mmol, 1 .20 eq) was added drop-wise in DMF (60.0 mL) under N2 atmosphere. The suspension was stirred for 5 hr at 25 °C. The reaction mixture was poured into ice-water (300 mL). The aqueous phase was extracted with ethyl acetate (300 mL * 3). The combined organic phase was washed with brine (300 mL * 2), dried with anhydrous Na2S04, filtered and concentrated in vacuum. The residue was purified by column chromatography (S1O2, Petroleum ether : Ethyl acetate = 50 : 1 to 1 : 1 ). Compound H (20.0 g, 42.0 mmol) was obtained as yellow oil.

1 H NMR: 400MHz CDCb

δ 7.76 (d, J = 8.8 Hz, 1 H), 7.30 - 7.43 (m, 6H), 6.98 - 7.08 (m, 1 H), 6.91 (dd, J = 2.0, 8.8 Hz, 1 H), 5.43 (s, 2H), 3.91 (s, 3H), 3.66 (s, 2H), 3.48 - 3.55 (m, 2H), 3.02 - 3.05 (m, 2H), 2.51 -2.60 (m, 2H), 2.28 - 2.33 (m, 2H), 2.24 - 2.26 (m, 2H), 0.94 (t, J = 8.0 Hz, 2H), 0.00 (s, 9H)

Preparatio -(6-methoxy-1 -((2-(trimethylsilyl)ethoxy)methyl)-1 H-indol-3-yl)piperidine-4-carbonitrile

H I

To a solution of compound H (15.0 g, 31 .5 mmol) in EtOH (300 mL) was added Pd/C (150 mg), AcOH (1 .89 g, 31 .5 mmol, 1 .80 mL) and the mixture was stirred at 50 °C for 12 hr under H 2 (40 Psi) atmosphere. The reaction mixture was filtered and concentrated in vacuum. The residue was purified by column chromatography (S1O2, Dichloromethane : Methanol = 1 00 : 1 to 10 : 1 ). Compound I (8.50 g, 22.0 mmol) was obtained as yellow oil.

Preparation of 1 -(cyclobutanecarbonyl)-4-(6-methoxy-1 -((2-(trimethylsilyl)ethoxy)methyl)-1 H-indol-3- yl)piperidine-4-carbonitrile

To a suspension of compound I (8.50 g, 22.0 mmol) in DCM (21 .0 mL) was added EteN (4.46 g, 44.1 mmol), compound J (2.61 g, 22.0 mmol) was added drop-wise to the reaction mixture at 0 °C. The mixture was stirred at 25 °C for 2 hrs. The reaction mixture was poured into ice-water (20 mL). The aqueous phase was extracted with DCM (20 mL * 3). The combined organic phase was washed with brine (20 mL * 2), dried with anhydrous Na2S04, filtered and concentrated in vacuum. The residue was purified by column chromatography (S1O2, Petroleum ether : Ethyl acetate = 1 0 : 1 to 1 : 1 ). Compound K (6.00 g, 12.8 mmol) was obtained as yellow oil.

Preparation of 1 -(cyclobutanecarbonyl)-4-(6-methoxy-1 H-indol-3-yl)piperidine-4-carbonitrile

The suspension of compound K (6.00 g, 12.8 mmol) in TBAF (1 M, 64.1 mL) was stirred for 5 hrs at 90 °C. The reaction mixture was poured into water (100 mL). The aqueous phase was extracted with ethyl acetate (100 mL * 3). The combined organic phase was washed with brine (100 mL * 2), dried with anhydrous Na2S04, filtered and concentrated in vacuum. The residue was purified by column chromatography (S1O2, Petroleum ether : Ethyl acetate = 1 0 : 1 to 1 : 1 ). Compound L (2.80 g, 8.30 mmol) was obtained as yellow oil. LCMS (ESI) m/z: [M+H] + = 338.2 Preparat -(aminomethyl)-4-(6-methoxy-1 H-indol-3-yl)piperidin-1 -yl)(cyclobutyl)methanone

To a suspension of compound L (2.80 g, 8.30 mmol) in MeOH (5.00 mL) was added Raney Ni (100 mg) and NH3.H2O (349 mg, 9.96 mmol), and the mixture was stirred at 30 °C for 5 hrs under H 2 (50Psi) atmosphere. The reaction mixture was filtered and concentrated in vacuum. Compound M (2.80 g, 8.20 mmol) was obtained as yellow oil.

1 H NMR: 400MHz CDCI3 δ= 8.53 (s, 1 H), 7.53 (d, J = 8.8 Hz, 1 H), 6.91 - 6.99 (m, 1 H), 6.88 (d, J = 2.0 Hz, 1 H), 6.74 (dd, J = 2.0, 8.8 Hz, 1 H), 4.10-4.12 (m, 1 H), 3.80 (s, 3H), 3.44-3.47 (m, 1 H), 3.22 (t, J = 8.8 Hz, 1 H), 2.98 - 3.15 (m, 2H), 2.90 - 2.97 (m, 2H), 2.22 - 2.40 (m, 4H), 2.03 - 2.20 (m, 2H), 1 .80 - 2.03 (m, 2 H), 1 .50 - 1 .76 (m, 2H). LCMS (ESI) m/z: [M+H] + = 342.3

Preparation of (1 '-(((tert-butyldimethylsilyl)oxy)methyl)-7'-methoxy-1 ',2',3',9'-tetrahydrospiro[piperidine- '-pyrido[3,4-b]indol]-1 -yl)(cyclobutyl)methanone

A suspension of compound M (200 mg, 586 umol, 1 .00 eq) was dissolved in CHCI3 (20.0 mL) with MgS04 (1 .76 g, 14.6 mmol, 5.00 eq) under N2. The suspension was cooled to 0 °C, and the compound N (766 mg, 4.40 mmol, 1 .50 eq) was added. After 30 min, the solution was allowed to warm to 25 °C and was stirred 12 hrs. The suspension was then cooled to 0 °C, CH3COOH (352 mg, 5.86 mmol, 2.00 eq) was added, and the reaction mixture was stirred at 0 °C for 3 hrs. The reaction mixture was poured into sat.NaHCCb (40 mL). The aqueous phase was extracted with DCM (50 mL * 3). The combined organic phase was washed with brine (50 mL * 2), dried with anhydrous Na2S04, filtered and concentrated in vacuum. The residue was purified by column chromatography (S1O2, Ethyl acetate). Compound O (0.85 g, 1 .71 mmol) was obtained as yellow oil. LCMS (ESI) m/z: [M+H] + = 498.3 Preparation of cyclobutyl-[1 -(hydroxylmethyl)-7-methoxy-2 [2-(trifluoromethyl)phenyl]methyl]spiro[3,9- dihydro-1 H-pyrido[3,4-b]indole-4,4'-piperidine]-1 '-yl]methanone (Compound 5)

Step 1 : Preparation of [1 -[[tert-butyl(dimethyl)silyl]oxymethyl]-7-methoxy-2-[[2-

(trifluoromethyl)phenyl]methyl]spiro[3,9-dihydro-1 H-pyrido[3,4-b]indole-4,4'-piperidine]-1 '-yl]-cyclobutyl- methanone

To a mixture of [1 -[[tert-butyl(dimethyl)silyl]oxymethyl]-7-methoxy-spiro[1 ,2,3,9-tetrahydr opyrido[3,4-b]indole-4,4'-piperidine]-1 '-yl]-cyclobutyl-methanone (500.00 mg, 1 .00 mmol, 1 .00 eq) and 2-(trifluoromethyl)benzaldehyde (208.94 mg, 1 .20 mmol, 158.29 μΙ_, 1 .20 eq) in DCE (10.00 ml_) was added acetic acid (30.03 mg, 500.00 umol, 28.60 μΙ_, 0.50 eq) in one portion at 25 °C under N 2 . The mixture was stirred at 25 °C for 1 hour, then sodium triacetoxyboranuide (529.85 mg, 2.50 mmol, 2.50 eq) was added at 0°C. The reaction was stirred 25°C for 15 hours. The reaction mixture was poured into water(100.0 mL) and extrated with DCM (50.0 ml_ * 3). The combined organics were washed

with NaHC03 solution, water and brine, dried over anhydrous sodium sulfate, filtered and filtration was evaporated to dryness. The residue was purified by column chromatography (S1O2, DCM/MeOH=100/1 to 50/1 ). [1 -[[tert-butyl(dimethyl) silyl]oxymethyl]-7-methoxy-2-[[2-(trifluoromethyl)phenyl]met hyl]spiro[3,9- dihydro-1 H-pyrido[3,4-b]indole-4,4'-piperidine]-1 '-yl]-cyclobutyl-methanone (400.00 mg) was obtained as a yellow solid. LCMS (ESI) m/z: [M+H] + =656.3 (Method: 5-95AB_R_220 & 254).

Preparation of cyclobutyl-[1 -(hydroxylmethyl)-7-methoxy-2-[[2(trifluoromethyl)phenyl]met hyl]spiro [3,9-

To a solution of [1 -[[tert-butyl(dimethyl)silyl]oxymethyl]-7-methoxy-2-[[2-(tri fluoromethyl) phenyl]methyl]spiro[3,9-dihydro-1 H-pyrido[3,4-b]indole-4,4'-piperidine]-1 '-yl]-cyclobutyl-methanone (400 mg, 610 umol) in THF (30 mL) was added tetrabutylammonium fluoride (1 M, 1 .83 mL, 1 .83

mmol) at 25 °C. The mixture was stirred at 25 °C for 2 hours. The reaction mixture was poured into water (300 mL) and extracted with DCM (100 mL * 3). The combined organics were washed with water and brine, dried over anhydrous sodium sulfate, filtered and the filtrate evaporated to dryness. The residue was purified by prep-TLC (S1O2, DCM/MeOH=20/1 ). Cyclobutyl-[1 -(hydroxylmethyl)-7-methoxy-2-[[2- (trifluoromethyl)phenyl]methyl]spiro[3,9-dihydro-1 H-pyrido[3,4-b]indole-4,4'-piperidine]-1 '- yl]methanone(1 00 mg) was obtained as a yellow solid.

1 H NMR (400 MHz, DMSO-de) δ= 1 .49 (br d, J=13.0 Hz, 1 H), 1 .65-1 .79 (m, 2H), 1 .82-1 .91 (m, 2H), 1 .93- 2.03 (m, 2H), 2.05-2.26 (m, 5H), 2.56-2.72 (m, 2H), 3.00 (br dd, J=1 9.1 , 12.1 Hz, 1 H), 3.40-3.55 (m, 1 H), 3.73 (s, 3H), 3.76-4.01 (m, 4H), 4.1 7 (br t, J=15.5 Hz, 1 H), 4.26-4.40 (m, 1 H), 4.87 (t, J=4.8 Hz, 1 H), 6.58 (dd, J=8.6, 2.3 Hz, 1 H), 6.87 (s, 1 H), 7.31 (dd, J=8.6, 5.4 Hz, 1 H), 7.42-7.53 (m, 1 H), 7.63-7.77 (m, 2H), 7.89-7.99 (m, 1 H), 10.49 (br d, J=3.5 Hz, 1 H) ppm ; LCMS (ESI) m/z: [M+H] + =542.1 (Method: 5- 95AB_R_220 & 254).

Preparation of Compound Cyclobutyl-[1 -(hydroxymethyl)-7-methoxy-2-[[4-(2-methoxyethoxy)phenyl]

o

Preparation of Cyclobutyl-[1 -(hydroxymethyl)-7-methoxy-2-[[4-(2-methoxyethoxy)phenyl] methyl]spiro[3,9- dihydro-1 H-pyrido[3,4-b]indole-4,4'-piperidine]-1 '-yl]methanone

To a solution of [1 -[[tert-butyl(dimethyl)silyl]oxymethyl]-7-methoxy-spiro[1 ,2,3,9-tetrahydropy rido[3,4-b]indole-4,4'-piperidine]-1 '-yl]-cyclobutyl-methanone (500 mg, 1 .00 mmol) and 4-(2- methoxyethoxy)benzaldehyde (216 mg, 1 .20 mmol) in DCE (35 mL). Then triacetoxysodium borohydride (530 mg, 2.50 mmol) was added at 0°C. The reaction mixture was stirred at 25°C for 16 hrs then poured into water (100 mL) and extracted with EA (10 mL * 3). The combined organic layer was washed with water (10 mL * 3) and brine (10 mL), dried over Na2S04, concentrated under vacuum. The residue was diluted with THF (15 mL) and TBAF (1 M, 594 μί, 5.9 mmol) was added. The mixture was stirred at 30 °C for 3 hrs. The reaction mixture was diluted with EA (150 mL) and washed with H2O (30 mL * 6) and brine (30 ml_ * 2), dried over Na2S04, concentrated under vacuum. The residue was purified by Pre-HPLC (column: Phenomenex Synergi C18 150 * 25 * 1 Oum; mobile phase: [water(0.1 %TFA)-ACN];B%: 18%- 48%,12min) to obtain cyclobutyl-[1 -(hydroxymethyl)-7-methoxy-2-[[4-(2- methoxyethoxy)phenyl]methyl]spiro[3,9-dihydro-1 H-pyrido[3,4-b]indole-4,4'-piperidine]-1 '-yl]methanone (138 mg, 248 umol) as a light yellow solid.

1 H NMR (400 MHz, DMSO-d6) δ= 1 .48-1 .52 (m, 1 H), 1 .64-1 .78 (m, 2H), 1 .82-1 .94 (m, 2H), 1 .95-2.35 (m, 7H), 2.60-2.74 (m, 1 H), 2.99-3.10 (m, 1 H), 3.30 (s, 3H), 3.44-3.48 (m, 1 H), 3.62-3.69 (m, 3H), 3.72 (s, 3H), 3.73-3.80 (m, 2H), 3.91 -3.99 (m, 1 H), 4.04-4.1 0 (m, 3H), 4.16-4.20 (m, 1 H), 4.71 -4.76 (m, 1 H), 6.55- 6.58 (m, 1 H), 6.86 (s, 1 H), 6.84-6.87 (m, 1 H), 6.91 (d, J=8.6 Hz, 1 H), 7.26-7.34 (m, 3H), 10.42 (s, 1 H), 10.38-10.49 (m, 1 H) ppm ; LCMS (ESI) m/z: [M+H] + = 548.3 (Method: 5-95AB_R_210 & 254).

The following examples were made analogously: Preparation of [2-[(2-bromophenyl)methyl]-1 -(hydroxymethyl)-7-methoxy-spiro[3,9-dihydro-1 H-pyrido[3,4- b]indole-4,4'-piperidine]-1 '-yl]-cyclobutyl-methanone (Compound 12)

Preparation of [2-[(2-bromophenyl)methyl]-1 -(hydroxymethyl)-7-methoxy-spiro[3,9-dihydro-1 H-pyrido[3,4- b]indole-4,4'-piperidine]-1 '-yl]-cyclobutyl-methanone

12

1 H NMR (400 MHz, MeOD-d 4 ) δ= 1 .53 (br t, J=12.6 Hz, 1 H), 1 .74 (br s, 2H), 1 .90-2.08 (m, 3H), 2.1 6-2.29 (m, 4H), 2.55-2.68 (m, 2H), 3.08 (dd, J=12.1 , 1 .8 Hz, 1 H), 3.30-3.39 (m, 1 H), 3.43-3.57 (m, 1 H), 3.73 (s, 3H), 3.80-3.92 (m, 3H), 4.02-4.12 (m, 1 H), 4.13-4.27 (m, 2H), 5.44 (s, 1 H), 6.55 (dd, J=8.7, 2.4 Hz, 1 H), 6.83 (d, J=2.3 Hz, 1 H), 7.09-7.17 (m, 1 H), 7.24-7.35 (m, 2H), 7.50-7.58 (m, 2H) ppm; [M+H] + =554.1 (Method: 5-95AB_R_220 & 254). Preparation of cyclobutyl-[1 -(hydroxymethyl)-7-methoxy-2-[(4-methoxyphenyl)methyl]spiro[ 3,9-dihydro- 1 H-pyrido[3,4-b]indole-4,4'-piperidine]-1 '-yl]methanone (Compound 13)

Preparation of cyclobutyl-[1 -(hydroxymethyl)-7-methoxy-2-[(4-methoxyphenyl)methyl]spiro[ 3,9-dihydro- 1 H-pyrido[3,4-b]indole-4,4'-piperidine]-1 '-yl]methanone

13

1 HNMR(400 MHz, DMSO-de) 5=0.81-0.90 (m, 1H), 1.24(s, 1H), 1.51 (br d, J=13.0 Hz, 1H), 1.63-1.78 (m, 2H), 1.80-1.95 (m, 2H), 1.96-2.36 (m, 7H), 2.59-2.77 (m, 2H), 3.05 (brdd, J=19.7, 12.0 Hz, 1H), 3.65 (brt, J=14.0 Hz, 1H), 3.72-3.73 (m, 1H), 3.74 (s, 4H), 3.78 (brd, J=5.6 Hz, 1H), 3.92-3.99 (m, 1H), 4.03- 4.12 (m, 1H), 4.18 (brd, J=12.5 Hz, 1H), 4.74 (t, J=4.8 Hz, 1H), 5.76 (s, 1H), 6.57 (dd, J=8.6, 2.3 Hz, 1H), 6.83-6.97 (m, 3H), 7.24-7.37 (m, 3H), 10.43 (d, J=1.7 Hz, 1H) ppm; LCMS (ESI) m/z: [M+H] + =504.3 (Method: 5-95AB_R_220 & 254). Preparation of Cyclobutyl-[1 -(hydroxymethyl)-7-methoxy-2-methyl-spiro[3,9-dihydro -1 H-pyrido[3,4- b]indole-4,4'-piperidine]-1'-yl]methanone TFA salt (Compound 24)

o 24 Preparation of Cyclobutyl-[1 -(hydroxymethyl)-7-methoxy-2-methyl-spiro[3,9-dihydro -1 H-pyrido[3,4- b]indole-4,4'-piperidine]-1 '-yl]methanon

24

Ή NMR (400 MHz, Methanol-d4) δ = 1 .61 -1 .96 (m, 2H), 1 .97-2.13 (m, 2H), 2.1 5-2.49 (m, 6H), 2.58-3.09 (m, 2H), 3.24 (s, 3H), 3.41 -3.62 (m, 1 H), 3.82 (s, 3H), 3.87-3.89 (m, 2H), 4.1 9-4.35 (m, 3H), 4.52-4.72 (m, 2H), 6.74 (dd, J=8.7, 2.2 Hz, 1 H), 6.93 (d, J=2.0 Hz, 1 H), 7.45 (d, J=8.7 Hz, 1 H) ppm ; LCMS (ESI) m/z: [M+H] + = 398.3 (Method: 5-95AB_R_210 & 254). Preparation of Cyclobutyl-[2-[(2-fluorophenyl)methyl]-1 -(hydroxymethyl)-7 -methoxy-spiro[3,9-dihydro-1 H-

31

1 H NMR (400 MHz, Methanol-d4) δ = 1 .75-1 .79 (m, 2H), 1 .84-1 .96 (m, 2H), 1 .98-2.12 (m, 2H), 2.14-2.44 (m, 7H), 2.62-2.66 (m, 1 H), 3.03-3.07 (m, 1 H), 3.42-3.55 (m, 1 H), 3.82 (s, 3H), 4.00-4.02 (m, 1 H), 4.1 9- 4.39 (m, 2H), 4.47-4.51 (m, 0.5H), 4.73-4.77 (m, 2.5H), 6.74 (dd, J=8.8, 2.3 Hz, 1 H), 6.93 (d, J=2.2 Hz, 1 H), 7.44 (d, J=8.8 Hz, 1 H), 7.55-7.60(m, 3H), 7.62-7.66 (m, 2H) ppm ; LCMS (ESI) m/z: [M+H] + = 474.4 (Method: 5-95AB_R_210 & 254). Preparation of cyclobutyl 2 (2SR)-2-(2 luorophenyl)-2-hydroxy-ethyl]-7-methoxy-spiro[3,9-dihydro-1 H- pyrido[3,4-b]indole-4,4'-piperidine]-1 '-yl]methanone and cyclobutyl-[2-[(2RS)-2-(2-fluorophenyl)-2-hydroxy- ethyl]-7-methoxy-spiro[3,9-dihydro-1 H-pyrido[3,4-b]indole-4,4'-piperidine]-1 '-yl]methanone (Compounds 1 , 2, and 3)

Scheme

2 (Peak 1 ] 3 (Peak 2) Step 1 : Preparation of Intermediate 1 B 2-(2-fluorophenyl)oxirane

To a solution of 1 -fluoro-2-vinyl-benzene (2.50 g, 20.5 mmol) in acetic acid (1 .23 g, 20.47 mmol, 1 .17 ml_), dioxane (14.5 mL) and water (35 mL) at 0°C was added N-bromosuccinimide (4.37 g, 24.56 mmol). The reaction was allowed to warm to 25 °C and stirred 16h. Sodium carbonate (4.34 g, 40.94 mmol) was added to the mixture and then sodium hydroxide (1 M, 4.91 mL) was added and the resulting mixture was stirred at 25 °C for another 16h. The reaction was quenched with water (20 mL) and extracted by EA (3 * 5 mL). The combined organic phase was dried over anhydrous sodium sulfate and concentrated in vacuum. 2-(2-fluorophenyl)oxirane (2.50 g) was obtained as a yellow oil, which was used into next step directly without further purification.

1 H NMR (400 MHz, CDC ) 5=2.79-2.85 (m, 1 H), 3.1 6-3.20 (m, 1 H), 4.15-4.19 (m, 1 H), 7.02-7.1 1 (m, 1 H), 7.12-7.24 (m, 2H), 7.26-7.35 (m, 1 H) ppm. Step 2: Preparation of 2 cyclobutyl(2'-(2-(2-fluorophenyl)-2-hydroxyethyl)-7'-methoxy -1 ',2',3',9'- tetrahydrospiro[piperidine-4,4'-pyrido[3,4-b]indol]-1 -yl)methanone

Cyclobutyl-(7-methoxyspiro[1 ,2,3,9-tetrahydropyrido[3,4-b]indole-4,4'-piperidine]-1 '-yl)methanone

(200 mg, 566 umol) and 2-(2-fluorophenyl)oxirane (234 mg, 1 .70 mmol) were dissolved in ethyl alcohol (1 0 mL), and triethylamine (115 mg, 1 .13 mmol, 156.86 μί) was added. Then the mixture was stirred at 80°C for 3h. The mixture was concentrated. The residue was purified by Prep-TLC (DCM:MeOH=20:1 ). To afford cyclobutyl(2'-(2-(2-fluorophenyl)-2-hydroxyethyl)-7'-methoxy -1 ',2',3',9'-tetrahydrospiro[piperidine- 4,4'-pyrido[3,4-b]indol]-1 -yl)methanone (1 50 mg) as a yellow solid.

Step 3: Preparation of cyclobutyl-[2-[(2SR)-2-(2-fluorophenyl)-2-hydroxy-ethyl]-7-m ethoxy-spiro[3,9- dihydro-1 H-pyrido[3,4-b]indole-4,4'-piperidine]-1 '-yl]methanone and cyclobutyl-[2-[(2RS)-2-(2- fluorophenyl)-2-hydroxy-ethyl]-7-methoxy-spiro[3,9-dihydro-1 H-pyrido[3,4-b]indole-4,4'-piperidine]-1 '- yl]methanone (The absolute configuration of the two enantiomers are unknown )

2 (Peak 1 in OD) 3 (Peak 2 in OD)

Cyclobutyl(2'-(2-(2-fluorophenyl)-2-hydroxyethyl)-7'-meth oxy-1 ',2',3',9'-tetrahydrospiro[piperidine- 4,4'-pyrido[3,4-b]indol]-1 -yl)methanone (1 50mg, 0.31 mmol) was dissolved in MeCN (10 mL) at 20°C. The solution was further separated by SFC (condition : column: OD (250mm * 30mm,10um); mobile phase:

[0.1 % NH3H2O MEOH]; B%: 45%-45%, 2.4min; 90minmin).

The first eluted compound (Peak 1 , retention time: 3.349 min): cyclobutyl-[2-[(2SR)-2-(2- fluorophenyl)-2-hydroxy-ethyl]-7-methoxy-spiro[3,9-dihydro-1 H-pyrido[3,4-b]indole-4,4'-piperidine]-1 '- yl]methanone (65.28 mg) was obtained as a white solid. 1 H NMR (400 MHz, DMSO-d6) δ =1.37-1.64 (m, 2H), 1.77-1.79 (m, 1 H), 1.86-2.01 (m, 3H), 2.04-2.28 (m, 5H), 2.73-2.91 (m, 4H), 3.00-3.16 (m, 1H), 3.51-3.64 (m, 3H), 3.71 (s, 3H), 4.15-4.32 (m, 1H), 5.08-5.10 (m, 1 H), 5.31 -5.32 (m, 1 H), 6.56 (dd, J=8.6, 1.9 Hz, 1 H), 6.77 (d, J=1.8 Hz, 1 H), 7.10-7.12 (m, 1 H), 7.15- 7.33 (m, 3H), 7.52-7.56 (m, 1H), 10.53 (s, 1H) ppm; LCMS (ESI) m/z: [M+H] + = 492.2 (Method: 5- 95AB_R_220&254).

The second eluted compound (Peak 2, retention time: 3.598 min) cyclobutyl-[2-[(2RS)-2-(2- fluorophenyl)-2-hydroxy-ethyl]-7-methoxy-spiro[3,9-dihydro-1 H-pyrido[3,4-b]indole-4,4'-piperidine]-1'- yl]methanone (66.28 mg) was obtained as a white solid

1H NMR (400 MHz, DMSO-d6) 5=1.41-1.62 (m, 2H), 1.71-1.83 (m, 1H), 1.85-2.02 (m, 3H), 2.05-2.28 (m, 4H), 2.67-2.93 (m, 5H), 2.99-3.16 (m, 1H), 3.36-3.45 (m, 1H), 3.48-3.64 (m, 3H), 3.71 (s, 3H), 4.24-4.26 (m, 1H), 5.11-5.12 (m, 1H), 6.56 (dd, J=8.5, 1.71 Hz, 1H), 6.77 (d, J=1.6 Hz, 1H), 7.09-7.11 (m, 1H), 7.16- 7.33 (m, 3H), 7.54-7.56 (m, 1H), 10.53 (s, 1H) ppm; LCMS (ESI) m/z: [M+H] + = 492.2 (Method: 5- 95AB_R_220 & 254).

Synthesis of Cyclobutyl-[1 -ethyl-2-[(2-fluorophenyl)methyl]-7-methoxy-spiro[3,9-dihydr o-1 H-pyrido [3,4-

M 7

Step 1 : Preparation of cyclobutyl-(1-ethyl-7-methoxy-spiro[1 ,2,3,9-tetrahydropyrido[3,4-b]indole -4,4'- piperidine]-1'-yl)methanone.

To a solution of [4-(aminomethyl)-4-(6-methoxy-1H-indol-3-yl)-1-piperidyl]-cy clobutyl- methanone (0.25 g, 732 umol) in EtOH (3 mL) was added propanal (213 mg, 3.66 mmol, 266 μΐ) and HCI/dioxane (4 M, 366 iL). The mixture was stirred at 60°C for 16 hours. The mixture was diluted with DCM (100 mL) and washed by saturated NaHCCb solution (2*30 mL). The combined organic phase was dried over anhydrous sodium sulfate and concentrated in vacuo. The residue was purified by column

chromatography (Si0 2 , DCM/MeOH=9/1) to afford cyclobutyl-(1-ethyl-7-methoxy-spiro[1 ,2,3,9- tetrahydropyrido[3,4-b]indole-4,4'-piperidine]-1'-yl)methano ne (70 mg) as a yellow solid.

LCMS (ESI) m/z: [M+H] + =382.3 (Method: 5-95AB_R_220 & 254). Step 2: Preparation of Cyclobutyl-[1 -ethyl-2-[(2-fluorophenyl)methyl]-7-methoxy-spiro[3,9-dihydr o-1 H- pyrido [3,4-b]indole-4,4'-piperidine]- '-yl]methanone.

7

To a mixture of cyclobutyl-(1 -ethyl-7-methoxy-spiro[1 ,2,3,9-tetrahydropyrido[3,4-b]indole- 4,4'- piperidine]-1 '-yl)methanone (70 mg, 183 umol) and triethylamine (56mg, 550 umol, 77 μΙ_) in THF (1 mL) was added 1 -(bromomethyl)-2-fluoro-benzene (52 mg, 275 umol, 33 μΙ_) in one portion at 0°C. The mixture was stirred at 25 °C for 16 hours. The reaction mixture was poured into water (50 mL) and extracted with DCM (30 mL * 3). The combined organics were washed with water and brine, dried over anhydrous sodium sulfate, filtered and evaporated to dryness. The residue was purified by column chromatography (Si0 2 , DCM/MeOH=30/1 ) to afford Cyclobutyl-[1 -ethyl-2-[(2-fluorophenyl)methyl]-7- methoxy-spiro[3,9-dihydro-1 H-pyrido[3,4-b]indole-4,4'-piperidine]-1 '-yl]methanone (20 mg) as a yellow solid.

1 H NMR (400 MHz, Methanol-d 4 ) δ=0.80 (m, 3H), 1 .43-1 .54 (m, 1 H), 1 .80-1 .93 (m, 2H), 1 .95-2.07 (m, 3H), 2.13-2.35 (m, 6H), 2.43-2.62 (m, 2H), 2.86-2.97 (m, 1 H), 3.37-3.48 (m, 1 H), 3.49-3.59 (m, 2H), 3.66 (br d, J=13.9 Hz, 1 H), 3.81 (s, 3H), 4.15 (m 1 H), 4.23 (m, 1 H), 4.36 (m, 1 H), 6.62 (dd, J=8.7, 2.3 Hz, 1 H), 6.87 (d, J=2 Hz, 1 H), 7.12 (br t, J=9.2 Hz, 1 H), 7.17-7.24 (m, 1 H), 7.27-7.38 (m, 2H), 7.55 (br t, J=7.5 Hz, 1 H) ppm ; LCMS (ESI) m/z: [M+H] + =490.2 (Method: 5-95AB_R_220 & 254).

Preparation of cyclobutyl(2'-(2-fluorobenzyl)-7'-methoxy-1 '-(methoxymethyl)-l ',2',3',9'- tetrahydrospiro[piperidine-4,4'-pyrido[3,4-b]indol]-1 -yl)methanone. (Compound 4)

Scheme:

Step 1 . Preparation of cyclobutyl(7'-methoxy-1 '-(methoxymethyl)-1 ',2',3',9'-tetrahydrospiro[piperidine-4,4'- pyrido[3,4-b]indol]-1 -yl)methanone:

To a solution of 1 ,1 ,2-trimethoxyethane (141 mg, 1 .17 mmol, 151 μΙ_) in CHC (20ml_) was added HCI/dioxane (2 M, 131 .51 iL). The mixture was stirred at 20 °C for 1 hr, then [4-(aminomethyl)-4- (6-methoxy-1 H-indol-3-yl)-1 -piperidyl]-cyclobutyl-methanone (200 mg, 586 umol), was added and the mixture was stirred at 45 °C for 16h. Saturated aq NaHCCb (20 mL) was added to the mixture, and the organic layer was dried over Na2S04, and concentrated in vacuum. Compound cyclobutyl-[7- methoxy-1 - (methoxymethyl)spiro[1 ,2,3,9-tetrahydropyrido[3,4-b]indole-4,4'-piperidine]-1 '-yl]methanone (150 mg) was given as a yellow solid, and it was confirmed by LCMS 398.2. The crude was used in the next step without further purification.

Step 2. Preparation of cyclobutyl(2'-(2-fluorobenzyl)-7'-methoxy-1 '-(methoxymethyl)-1 ',2',3',9'- tetrahydrospiro[piperidine-4,4'-pyrid -b]indol]-1 -yl)methanone.

To a solution of cyclobutyl-[7-methoxy-1 -(methoxymethyl)spiro[1 ,2,3,9-tetrahydropyrido[3,4-b] indole-4,4'-piperidine]-1 '-yl]methanone (140 mg, 352 umol) in DMF (5 mL) was added EteN (107 mg, 1 .06 mmol, 146 μί) and 1 -(bromomethyl)-2-fluoro-benzene (133 mg, 704 umol, 85 μί) and the mixture was stirred at 45 °C for 20h. Water (50 mL) was added and the mixture was filtered. The filtered solid was dissolved in DCM, dried over Na2S04, and concentrated in vacuum to afford cyclobutyl-[2-[(2- fluorophenyl)methyl]-7-methoxy-1 -(methoxymethyl) spiro[3,9-dihydro-1 H-pyrido[3,4-b]indole-4,4'- piperidine]-1 '-yl]methanone (58.85 mg) as a white solid. LCMS (ESI) m/z: [M+H] + = 506.3 Preparation of Cyclobutyl-[(1 S)-2-[(1 S)-1 -(2-fluorophenyl)ethyl]-1 -(hydroxymethyl)-7-methoxy-spiro[3,9- dihydro-1 H-pyrido[3,4-b]indole-4,4'-piperidine]-1 '-yl]methanone and its 3 diastereomers (Compounds 15- 18)

Step 1 : Preparation of Intermediate 1 [(1 SR)-1 -[[tert-butyl(dimethyl)silyl]oxymethyl]-2-[1 -(SR)(2- fluorophenyl)ethyl]-7-methoxy-spiro[3,9-dihydro-1 H-pyrido[3,4-b]indole-4,4'-piperidine]-1 '-yl]-cyclobutyl- methanone and

2 [(1 SR)-1 -[[tert-butyl(dimethyl)silyl]oxymethyl]-2-[1 -(RS)(2-fluorophenyl) ethyl]-7-methoxy-spiro[3,9- dihydro-1 H-p -b]indole-4,4'-piperidine]-1 '-yl]-cyclobutyl-methanone

15 18

To a solution of [1 -[[tert-butyl(dimethyl)silyl]oxymethyl]-7-methoxy-spiro[1 ,2,3,9-tetrahydropy rido[3,4-b]indole-4,4'-piperidine]-1 '-yl]-cyclobutyl-methanone (500 mg, 1 .00 mmol) in MeCN (30 mL) was added 1 -(1 -bromoethyl)-2-fluoro-benzene (245 mg, 1 .20 mmol), K2CO3 (416 mg, 3.0 mmol) and Kl (33 mg, 201 umol). The reaction mixture was stirred at 85 for 16 hrs. The reaction mixture was poured into water (200 mL) and extracted with EA (50 mL * 3). The combined organic layer was washed with water (50 mL * 2) and brine (30ml), then dried over Na2S04 and concentrated in vacuo. The residue was separated by Pre-HPLC (column: Phenomenex Synergi C18 150 * 25 * 10um ; mobile phase: [water(0.1 %TFA)- ACN];B%: 40%-70%,2min). The first eluted compound (retention time: 0.945min): ((S,R)-1 '-(((tert- butyldimethylsilyl)oxy)methyl)-2'-((S,R)-1 -(2-fluorophenyl)ethyl)-7'-methoxy-1 ',2',3',9'- tetrahydrospiro[piperidine-4,4'-pyrido[3,4-b]indol]-1 -yl)(cyclobutyl)methanone (0.16 g, 248.82 umol) as a light yellow solid)

LCMS (ESI) m/z: [M+H] + = 620.5 (Method: 5-95AB_R_210&254).

The second eluted compound (retention time: 1 .027min((S,R)-1 '-(((tert- butyldimethylsilyl)oxy)methyl)-2'-((R,S)-1 -(2-fluorophenyl)ethyl)-7'-methoxy-1 ',2',3',9'- tetrahydrospiro[piperidine-4,4'-pyrido[3,4-b]indol]-1 -yl)(cyclobutyl)methanone (0.09 g, 1 19.48 umol) as a light yellow solid.

LCMS (ESI) m/z: [M+H] + = 620.5 (Method: 5-95AB_R_210&254).

Step 2: Preparation of cyclobutyl-[(1 S)-2-[(1 S)-1 -(2-fluoro phenyl)ethyl]-1 -(hydroxymethyl)-7-methoxy- spiro[3,9-dihydro-1 H-pyrido[3,4-b]indole-4,4'-piperidine]-1 '-yl]methanone, and cyclobutyl-[(1 R)-2-[(1 R)- 1 - (2-fluorophenyl)ethyl]-1 -(hydroxymethyl)-7-methoxy-spiro[3,9-dihydro-1 H-pyrido[3,4-b]indole-4,4'- piperidine]-1 '-y

15 16

To a solution of [1 -[[tert-butyl(dimethyl)silyl]oxymethyl]-2-[1 -(2-fluorophenyl)ethyl]-7-methoxy - spiro[3,9-dihydro-1 H-pyrido[3,4-b]indole-4,4'-piperidine]-1 '-yl]-cyclobutyl-methanone (0.16 g, 260 umol) in THF (15 mL) was added TBAF (1 M, 774 μΙ_). The reaction mixture was stirred at 30 °C for 3 hrs. The reaction was diluted with EA (100 mL) and washed with H2O (30 mL * 6), then dried over Na2S04 and concentrated under vacuum. The residue was separated by SFC (column: IC (250mm * 30mm, 10um); mobile phase: [0.1 %NH3H2O MEOH];B%: 55%-55%,3.7min;70min).

The first eluted compound (retention time: 2.273 min): cyclobutyl-[(1 S)-2-[(1 S)-1 -(2-fluoro phenyl)ethyl]-1 -(hydroxymethyl)-7-methoxy-spiro[3,9-dihydro-1 H-pyrido[3,4-b]indole-4,4'-piperidine]-1 '- yl]methanone (75 mg, 146 umol) as a light yellow solid).

15: 1 H NMR (400 MHz, Methanol-d4) δ =1 .54 (d, J=6.40 Hz, 3H), 1 .57-1 .67 (m, 2H), 1 .77-2.35 (m, 9H), 2.52-2.62 (m, 0.5H), 2.66 (dd, J=12.0, 6.5 Hz, 1 H), 3.04 (dd, J=12.0, 5.7 Hz, 1 H), 3.52-3.56 (m, 0.5H), 3.80 (s, 3H), 3.96-4.03 (m, 1 H), 4.02-4.09 (m, 0.5H), 4.20-4.29 (m, 2.5H), 4.75-4.82 (m, 1 H), 6.60 (dd, J=8.6, 2.1 Hz, 1 H), 6.89 (d, J=2.2 Hz, 1 H), 7.07-7.15 (m, 1 H), 7.19-7.27 (m, 1 H), 7.28-7.37 (m, 2H), 7.55- 7.63 (m, 1 H)ppm ; LCMS (ESI) m/z: [M+H]+ = 506.4 (Method: 5-95AB_R_220&254). The second eluted compound (retention time: 3.183 min): cyclobutyl-[(1 R)-2-[(1 R)- 1 -(2- fluorophenyl)ethyl]-1 -(hydroxymethyl)-7-methoxy-spiro[3,9-dihydro-1 H-pyrido[3,4-b]indole-4,4'-piperidine]- 1 '-yl]methanone (55 mg, 108.58 umol), as a light yellow solid) 16: 1 H NMR (400 MHz, Methanol-d4) δ= 1 .54 (d, J=6.40 Hz, 3H), 1 .57-1 .67 (m, 2H), 1 .77-2.35 (m, 9H), 2.52-2.62 (m, 0.5H), 2.66 (dd, J=12.0, 6.5 Hz, 1 H), 3.04 (dd, J=12.0, 5.7 Hz, 1 H), 3.52-3.56 (m, 0.5H), 3.80 (s, 3H), 3.96-4.03 (m, 1 H), 4.02-4.09 (m, 0.5H), 4.20-4.29 (m, 2.5H), 4.75-4.82 (m, 1 H), 6.60 (dd, J=8.6, 2.1 Hz, 1 H), 6.89 (d, J=2.2 Hz, 1 H), 7.07- 7.15 (m, 1 H), 7.19-7.27 (m, 1 H), 7.28-7.37 (m, 2H), 7.55-7.63 (m, 1 H) ppm; LCMS (ESI) m/z: [M+H] + =506.4 (Method: 5-95AB_R_220&254). Step 2.2: Preparation of cyclobutyl-[(1 S)-2-[(1 R)-1 -(2-fluoro phenyl)ethyl]-1 -(hydroxymethyl)-7-methoxy- spiro[3,9-dihydro-1 H-pyrido[3,4-b]indole-4,4'-piperidine]-1 '-yl]methanone P2A and cyclobutyl-[(1 R)-2- [(1 S)-1 - (2-fluorophenyl)ethyl]-1 -(hydroxymethyl)-7-methoxy-spiro[3,9-dihydro-1 H-pyrido[3,4-b]indole-4,4'- piperidine]-1 '-yl]methanone

18 17

To a solution of [1 -[[tert-butyl(dimethyl)silyl]oxymethyl]-2-[1 -(2-fluorophenyl)ethyl]-7-methoxy - spiro[3,9-dihydro-1 H-pyrido[3,4-b]indole-4,4'-piperidine]-1 '-yl]-cyclobutyl-methanone (90 mg, 145 umol) in THF (20 ml_) was added TBAF (1 M, 436 umol) . The reaction mixture was stirred at 30 °C for 3 hrs. Then diluted with EA (100 mL) and washed with water (30 ml_ * 6), then dried over Na2S04 and concentrated under vacuum. The residue was separated by SFC (column: IC(250mm * 30mm,1 Oum); mobile phase: [0.1 %NH3H2O MEOH];B%: 55%-55%,3.7min;70min min).

The first eluted compound (retention time: 3.037 min): cyclobutyl-[(1 S)-2-[(1 R)-1 -(2-fluoro phenyl)ethyl]-1 -(hydroxymethyl)-7-methoxy-spiro[3,9-dihydro-1 H-pyrido[3,4-b]indole-4,4'-piperidine]-1 '- yl]methanone (25 mg, 48 umol) as a light yellow solid. 18: 1 H NMR (400 MHz, Methanol-d4) δ =1 .51 - 1 .55 (m, 1 H), 1 .60 (d, J=6.8 Hz, 3H), 1 .81 -2.41 (m, 10H), 2.55-2.74 (m, 1 .5H), 2.83-3.02 (m, 0.5H), 3.02- 3.13 (m, 0.5H), 3.36-3.44 (m, 1 H), 3.46-3.55 (m, 1 H), 3.62-3.65 (m, 0.5H), 3.79 (s, 3H), 3.81 -3.85 (m, 1 H), 3.96-4.04 (m, 1 H), 4.05-4.1 1 (m, 1 H), 4.34-4.51 (m, 1 H), 4.75-4.79 m, 1 H), 6.60 (dd, J=8.7, 2.3 Hz, 1 H), 6.85 (d, J=2.3 Hz, 1 H), 7.08-7.15 (m, 1 H), 7.1 7-7.24 (m, 1 H), 7.26-7.33 (m, 1 H), 7.34 (d, J=8.7 Hz, 1 H), 7.60 (t, J=7.0 Hz, 1 H) ppm; LCMS (ESI) m/z: [M+H]+ = 506.4 (Method: 5-95AB_R_210 & 254).

The second eluted compound (retention time: 3.420 min): cyclobutyl-[(1 R)-2-[(1 S)-1 - (2- fluorophenyl)ethyl]-1 -(hydroxymethyl)-7-methoxy-spiro[3,9-dihydro-1 H-pyrido[3,4-b]indole-4,4'-piperidine]- 1 '-yl]methanone (40 mg, 76.34 umol) as a light yellow solid). 17: 1 H NMR (400 MHz, Methanol-d4) δ=1 .51 -1 .55 (m, 1 H), 1 .60 (d, J=6.8 Hz, 3H), 1 .81 -2.41 (m, 10H), 2.55-2.74 (m, 1 .5H), 2.83-3.02 (m, 0.5H), 3.02-3.13 (m, 0.5H), 3.36-3.44 (m, 1 H), 3.46-3.55 (m, 1 H), 3.62- 3.65 (m, 0.5H), 3.79 (s, 3H), 3.81 -3.85 (m, 1 H), 3.96-4.04 (m, 1 H), 4.05-4.1 1 (m, 1 H), 4.34-4.51 (m, 1 H), 4.75-4.79 m, 1 H), 6.60 (dd, J=8.7, 2.3 Hz, 1 H), 6.85 (d, J=2.3 Hz, 1 H), 7.08-7.1 5 (m, 1 H), 7.17-7.24 (m, 1 H), 7.26-7.33 (m, 1 H), 7.34 (d, J=8.7 Hz, 1 H), 7.60 (t, J=7.0 Hz, 1 H) ppm ; LCMS (ESI) m/z: [M+H] + = 506.4 (Method: 5-95AB_R_210 & 254).

Preparation of [7-Bromo-2-[(2-fluorophenyl)methyl]-1 -(hydroxymethyl)spiro [3,9-dihydro-1 H-pyrido[3,4- b]indole-4,4'-piperidine]-1 '-yl]-cyclobutyl-methanone (Compound 10)

S

Step 1 . Preparation of intermediate R. tert-Butyl 4-(6-bromo-1 H-indol-3-yl)-4-(nitromethyl)piperidine-1 - carboxylate

To a solution of 6-bromo-1 H-indole (10.8 g, 55.0 mmol,) in THF (200 mL) was added methylmagnesium iodide (3 M, 38.5 mL in THF) at 0°C under N2. The reaction was stirred at 0°C for 0.5h. A solution of tert-butyl 4-(nitromethylene)piperidine-1 -carboxylate (20.0 g, 82.6 mmol) in THF (200 mL) was added at 0°C. Then the reaction mixture was stirred at 40 °C for 16h. The reaction was quenched by water (300 mL) and NH4CI aqueous (200 mL). The mixture was extracted with EA (200 mL * 3). The combined organic layers were washed with brine (600 mL), dried over Na2S04, filtered and concentrated. The residue was purified by column chromatography (S1O2, Petroleum

ether/Ethylacetate=10/1 to 1 :1 ) to give tert-butyl 4-(6-bromo-1 H-indol-3-yl)-4-(nitromethyl) piperidine-1 - carboxylate (13.3 g, 28.8 mmol) as a light yellow solid. LCMS (ESI) m/z: [Br 81 M+H] + =462.1 (Method: 5- 95AB_R_220&254).

Step 2. Preparation of intermediate S. tert-Butyl 4-(aminomethyl)-4-(6-bromo-1 H-indol-3-yl)piperidine-1 - carboxylate

To a solution of tert-butyl-4-(6-bromo-1 H-indol-3-yl)-4-(nitromethyl) piperidine-1 -carboxylate (13.0 g, 29.7 mmol, 1 .00 eg) in MeOH (130 mL) and CHCI3 (130 mL) was added Nickel II Chloride hexahydrate (17.6 g, 74.2 mmol). Then NaBH4 (16.8 g, 445 mmol) was added in small portions at 0°C. The reaction was stirred at 20 °C for 12h. The reaction was quenched with water (100 mL) and NH4CI aqueous (1 00 mL). Then the mixture was extracted with EA (80 mL * 3). The combined organic layers were washed with brine (250 mL), dried over Na2S04, filtered and concentrated to give tert-butyl 4-(aminomethyl)-4-(6- bromo-1 H-indol-3-yl) piperidine-1 -carboxylate (12.0 g) as a light yellow solid, which was used in the next step without further purification. LCMS (ESI) m/z: [Br 79 M+H-56] + =352.1 (Method: 5-95AB_R_220&254).

Step 3. Preparation of intermediate U. tert-Butyl-7-bromo-1 -[[tert- butyl(dimethyl)silyl]oxymethyl]spiro[1 ,2, 3, 9-tetrahydropyrido[3,4-b]indole-4,4'-piperidine]-1 '-carboxylate

To a suspension of tert-butyl 4-(aminomethyl)-4-(6-bromo-1 H-indol-3-yl)piperidine-1 -carboxylate (5.0 g, 12 mmol) and anhydrous magnesium sulfate(7.4 g, 61 .0 mmol) in toluene (300 mL) was added 2- [tert-butyl(dimethyl)silyl]oxyacetaldehyde (4.0 g, 18 mmol) in one portion at 0°C. After the addition was complete, the reaction was stirred at 70°C for 36h. The reaction was filtered and concentrated. The residue was purified by column chromatography (S1O2, DCM/MeOH=100/1 to 30:1 ) to give tert-butyl 7- bromo-1 -[[tert-butyl(dimethyl)silyl]oxymethyl]spiro[1 ,2,3,9-tetrahydropyrido[3,4-b]indole-4,4'-piperidine]-1 '- carboxylate (2.0 g, 2.8 mmol) as a black brown solid. LCMS (ESI) m/z: [Br 81 M+H] + =566.2 (Method: 5- 95AB_R_220&254). Step 4. Preparation of intermediate W. tert-Butyl-7-bromo-1 -[[tert-butyl(dimethyl)silyl]oxymethyl]-2- [(2- fluorophenyl)methyl]spiro[3,9-dihydro-1 H-pyrido[3,4-b]indole-4,4'-piperidine]-1 '-carboxylate

To a suspension of tert-butyl-7-bromo-1 -[[tert-butyl(dimethyl)silyl]oxymethyl] spiro[1 ,2,3,9- tetrahydropyrido[3,4-b]indole-4,4'-piperidine]-1 '-carboxylate (2.0 g, 3.5 mmol) and TEA (895 mg, 8.85 mmol) in THF (30.00 mL) was added 1 -(bromomethyl)-2-fluoro-benzene (803 mg, 4.25 mmol) at 0°C. The reaction was stirred at 20°C for 1 h. Then the reaction was heated to 45 °C and stirred for 12h. The reaction was diluted with water (100 mL) and extracted with EA (50 mL * 3). The combined organic layers were washed with brine (150 mL), dried over Na2S04, filtered and concentrated. The residue was purified by column chromatography (S1O2, Petroleum ether/Ethyl acetate=50/1 to 10:1 ) to give tert-butyl-7-bromo- 1 -[[tert-butyl(dimethyl)silyl]oxymethyl]-2- [(2-fluorophenyl)methyl]spiro[3,9-dihydro-1 H-pyrido[3,4-b]indole- 4,4'-piperidine]-1 '-carboxylate (1 .45 g, 2.00 mmol) as a light yellow solid. LCMS (ESI) m/z:

[Br 81 M+H] + =674.2 (Method: 5-95AB_R_220&254).

Step 5. Preparation of intermediate X. [7-Bromo-2-[(2-fluorophenyl)methyl]spiro[3,9-dihydro-1 H- pyrido[3,4-b]indole-4,4'-piperidine]-1 -yl]methoxy-tert-butyl-dimethyl-silane

To a solution of tert-butyl 7-bromo-1 -[[tert-butyl(dimethyl)silyl]oxymethyl]-2-[(2-fluorophenyl) methyl]spiro[3,9-dihydro-1 H-pyrido[3,4-b]indole-4,4'-piperidine]-1 '-carboxylate (1 .4 g, 2.1 mmol, 1 .00 eg) in DCM (20.00 mL) was added TFA (6.16 g, 54.03 mmol, 4.00 mL) and the reaction was stirred at 20 °C for 3h. The reaction was diluted with DCM (50 mL) and washed with water (50 mL * 2). The organic layer was concentrated to give [7-bromo-2-[(2-fluorophenyl)methyl]spiro[3,9-dihydro-1 H-pyrido[3,4-b]indole- 4,4'-piperidine]-1 -yl]methoxy-tert-butyl-dimethyl-silane (1 .10 g, 1 .7 mmol) as a light yellow solid. LCMS (ESI) m/z: [Br 81 M+H] + =574.2 (Method: 5-95AB_R_220&254). Step 6. Preparation of intermediate Z. [7-Bromo-1 -[[tert-butyl(dimethyl)silyl]oxymethyl]-2-[(2- fluorophenyl)methyl]spiro[3,9-dihydro-1 H-pyrido[3,4-b]indole-4,4'-piperidine]-1 '-yl]-cyclobutyl-methanone

To a solution of [7-bromo-2-[(2-fluorophenyl)methyl]spiro[3,9-dihydro-1 H-pyrido[3,4-b]indole-4,4'- piperidine]-1 -yl]methoxy-tert-butyl-dimethyl-silane (1 .1 g, 1 .9 mmol, 1 .00 eg) and TEA (389 mg, 3.84 mmol, 533 μΙ_) in DCM (20 mL) was added cyclobutanecarbonyl chloride (273 mg, 2.3 mmol) at 0°C. Then the reaction was stirred at 20 °C for 1 h. The reaction was diluted with water (100 mL) and extracted with DCM (30 mL * 3). The combined organic layers were washed with NaHC03 (50 mL), dried over Na2S04, filtered and concentrated to give [7-bromo-1 -[[tert-butyl(dimethyl)silyl]oxymethyl]-2-[(2- fluorophenyl)methyl]spiro[3,9-dihydro-1 H-pyrido[3,4-b]indole-4,4'-piperidine]-1 '-yl]-cyclobutyl-methanone (1 .15 g, 1 .54 mmol) as a light yellow solid. LCMS (ESI) m/z: [Br 79 M+H] + =654.2 (Method: 5- 95AB_R_220&254).

Step 7. Preparation of [7-Bromo-2-[(2-fluorophenyl)methyl]-1 -(hydroxymethyl)spiro [3,9-dihydro-1 H- pyrido[3,4-b]indole-4,4'-piperidine]-1 '-yl]-cyclobutyl-methanone (Compound 10)

10

To a solution of [7-bromo-1 -[[tert-butyl(dimethyl)silyl]oxymethyl]-2-[(2-fluorophenyl)

methyl]spiro[3,9-dihydro-1 H-pyrido[3,4-b]indole-4,4'-piperidine]-1 '-yl]-cyclobutyl-methanone (80.0 mg,122 umol) in THF (1 .50 mL) was added TBAF (1 M, 244.38 iL). Then the reaction was stirred at 20°C for 2h. The reaction was diluted with water (20 mL) and extracted with EA (10 mL * 3). The combined organic layers were washed with brine (20 mL), dried over Na2S04, filtered and concentrated. The residue was washed by MeOH (10 mL) to give [7-bromo-2-[(2-fluorophenyl)methyl]-1 -(hydroxymethyl)spiro [3,9- dihydro-1 H-pyrido[3,4-b]indole-4,4'-piperidine]-1 '-yl]-cyclobutyl-methanone (58 mg, 99.8 umol) as a white solid. LCMS (ESI) m/z: [Br 79 M+H] + =540.2 (Method: 5-95AB_R_220&254). 1 H NMR (400 MHz, DMSO-de) δ =1 .45-1 .49 (m, 1 H), 1 .55-1 .71 (m, 2H), 1 .86-1 .90 (m, 2H), 1 .95-2.15 (m, 4H), 2.21 -2.48 (m, 2H), 2.50-2.70 (m, 3H), 3.08-3.12 (m, 1 H), 3.41 -3.50 (m, 1 H), 3.79-3.86 (m, 3H), 3.98- 4.02 (m, 1 H), 4.12-4.16 (m, 2H), 4.87-4.88 (m, 1 H), 7.01 -7.02 (m, 1 H), 7.02-7.04 (m, 2H), 7.17-7.40 (m, 2H), 7.51 -7.57 (m, 2H), 10.83 (s, 1 H).

Preparation of Cyclobutyl-[2-[(2-fluorophenyl)methyl]-7-methoxy-1 -methyl-spiro[3,9-dihydro-1 H-pyrido[3,4- b]indole-4,4'-piperidine]-1 '-yl]methanone TFA salt. (Compound 25)

To a solution of [4-(aminomethyl)-4-(6-methoxy-1 H-indol-3-yl)-1 -piperidyl]-cyclobutyl-methanone (0.05 g, 146 umol) in EtOH (1 mL) was added acetaldehyde (5 M, 87.86 μΙ_) and HCI/dioxane (4 M, 73 μΙ_). Then the mixture was stirred for 6 hours at 40 °C The mixture was diluted with DCM (30 mL) and washed by saturated NaHC03 solution (2 * 20 mL). The combined organic phase was dried over anhydrous sodium sulfate and concentrated in vacuum to afford cyclobutyl-(7-methoxy-1 -methyl- spiro[1 ,2,3,9-tetrahydropyrido[3,4-b]indole-4,4'-piperidine]-1 '-yl)methanone (40 mg) as a yellow solid, which was used in the next step without further purification.

LCMS (ESI) m/z: [M+H] + =368.2 (Method: 5-95AB_R_220 & 254). Step 2: Preparation of Cyclobutyl-[2-[(2-fluorophenyl)methyl]-7-methoxy-1 -methyl-spiro[3,9-dihydro-1 H- pyrido[3,4-b]indole-4,4'-piperidine]-1 '-yl]methanone TFA salt

25

To a mixture of cyclobutyl-(7-methoxy-1 -methyl-spiro[1 ,2,3,9-tetrahydropyrido[3,4-b] indole-4,4'- piperidine]-1 '-yl)methanone (40 mg, 108.85 umol) and triethylamine (33 mg, 327 umol, 45 μΙ_) in THF (1 mL) was added 1 -(bromomethyl)-2-fluoro-benzene (31 mg, 163 umol, 19.66 μΙ_) in one portion at 0°C . The mixture was stirred at 25°C for 16 hours. The reaction mixture was poured into water (20 mL), and extracted with DCM (1 0 mL * 3). The combined organics were washed with water and brine, dried over anhydrous sodium sulfate, filtered and filtrate was evaporated to dryness. The residue was purified by prep-HPLC (column: Phenomenex Synergi C18 150 * 25 * 10um ;mobile phase: [water(0.1 %TFA)- ACN];B%:27%-57%,12min). cyclobutyl-[2-[(2-fluorophenyl)methyl]-7-methoxy-1 -methyl-spiro[3,9-dihydro- 1 H-pyrido[3,4-b]indole-4,4'-piperidine]-1 '-yl]methanone TFA salt (5mg) was obtained as a yellow solid. 1 H NMR (400 MHz, Methanol-d 4 ) δ= 1 .49 (br t, J=14.4 Hz, 3H), 1 .84-1 .95 (m, 2H), 2.12-2.17 (m, 2H), 2.33-2.42 (m, 2H), 2.52-2.62 (m, 1 H), 2.64-2.75 (m, 1 H), 2.94-3.04 (m, 1 H), 3.23-3.35 (m, 2H), 3.40-3.60 (m, 3H), 3.65-3.82 (m, 5H), 3.90-4.02 (m, 1 H), 4.1 0-4.24 (m, 1 H), 4.47 (s, 1 H), 6.49 (dd, J=8.7, 2.3 Hz, 1 H), 6.75 (d, J=2.3 Hz, 1 H), 6.96-7.03 (m, 1 H), 7.04-7.10 (m, 1 H), 7.16-7.25 (m, 2H), 7.43 (br t, J=6.6 Hz, 1 H), 7.99 (s, 1 H) ppm ; LCMS (ESI) m/z: [M+H] + =476.2 (Method: 5-95AB_R_220 & 254). Preparation of Intermediate AE tert-butyl 4-(aminomethyl)-4-(6-methoxy-1 H-indol-3-yl)piperidine-1 - carboxylate

Scheme

Step 1 : Preparation of Intermediate AB tert-butyl 4-hydroxy-4-(nitromethyl) piperidine-1 - carboxylate.

To a solution of tert-butyl 4-oxopiperidine-1 -carboxylate (1 .50 kg, 7.53 mol, 1 .00 eq) in MeOH (12.00 L) was added CH3NO2 (1 .84 kg, 30.12 mol, 1 .63 L, 4.00 eq) dropwise in one portion under 1 0 . Followed by TEA (1 .52 kg, 15.06 mol, 2.09 L, 2.00 eq) was added dropwise at 10 °C over a 1 h period. Then the reaction was heated to 50 °C and stirred at 50 °C for 16hrs. The reaction solution was concentrated to 500 mL. The residue was diluted with water (5L) and extracted with EA (2L * 3). The combined organic layer was washed with saturated brine (1 L * 3). The separated organic layer was concentrated in vacuum. PE (1 L) was poured into the residue, and white solid formed. The resulting mixture was filtered, the filter cake was dried in vacuum to give tert-butyl4-hydroxy-4-(nitromethyl) piperidine-1 -carboxylate (720 g) as white solid.

1 H NMR (400 MHz, Methanol-d4) δ= 1 .48 (s, 9H), 1 .71 - 1 .62 (m, 4H), 3.19 (m, 2H), 3.87 (m, 2H), 4.52 (s, 2H) ppm.

Step 2: Preparation of Intermediate P -tert-but -(nitromethylene)piperidine-1 -carboxylate.

To a solution of DMSO (6.2 L) and Ac 2 0 (4.86 kg, 47.60 mol, 4.46 L) was added tert-butyl 4- hydroxy-4-(nitromethyl)piperidine-1 -carboxylate (620 g, 2.38 mol) at 10 over 1 h. The resulting mixture was stirred for 16h at 30°C. The mixture was quenched with water (20 L) and extracted with EA (3 * 5 L). The combined organic phase was washed by saturated NaCI solution (5 * 1 .5 L) and saturated Na2C03 solution (1 .5 L). The combined organic phase was dried over anhydrous Na2S04 and concentrated in vacuum. The residue was purified by column chromatography (PE:EA=1 00:1 -20:1 ) to afford tert-butyl 4- (nitromethylene)piperidine-l -carboxylate (168 g) as a yellow solid.

1 H NMR (400MHz, Methanol d4) δ= 1 .50 (s, 9H), 2.43-2.22 (m, 2H), 3.02-2.95 (m, 2H), 3.62-3.52 (m, 4H), 7.19 (s, 1 H) ppm. Step 3: Preparation of Intermediate AD tert-butyl 4-(6-methoxy-1 H-indol-3-yl) -4-(nitromethyl)piperidine-1 - carboxylate.

To a solution of 6-methoxy-1 H-indole (10 g, 68 mmol) in THF (200 mL) was added MeMgl (3 M, 25 mL, 75 mmol) dropwise at 0°C under N2. Then the reaction mixture was stirred at 0°C for 30 min. tert- Butyl 4-(nitromethylene)piperidine-1 -carboxylate (24.7 g, 102 mmol) dissolved in THF (200 mL) was added to the reaction at 0°C. Then the reaction was stirred at 40 °C for 16h. The reaction was quenched by water (500 mL) and extracted with EA (150 mL * 5). The combined organic phase was washed with brine (100 mL * 2), dried over Na2S04, filtered and concentrated. The residue was purified by column chromatography (S1O2, Petroleum ether/Ethyl acetate=20/1 to 5:1 ) to afford tert-butyl 4-(6-methoxy-1 H- indol-3-yl)-4-(nitromethyl)piperidine-1 -carboxylate (17.00 g) as white solid.

1 H NMR (400MHz, DMSO-d6) δ=1 .38 (s, 9H) , 1 .88 (m, 2H), 2.26 (m, 2H), 3.04 (br, 2H), 3.69-3.61 (m, 2H), 3.75 (s, 3H), 4.87 (s, 2H), 6.62 (dd, J=2.4, 8.8 Hz, 1 H), 6.85 (d, J=2.4 Hz, 1 H), 7.08 (d, J=2.5 Hz, 1 H), 7.58 (d, J=8.8 Hz, 1 H), 10.90-10.83 (s, 1 H) ppm; LCMS (ESI) m/z: [M+Na] + = 412.2 (Method: 5- 95AB_R_220 & 254).

Step 4: Preparation of Intermediate AE tert-butyl 4-(aminomethyl)-4-(6-methoxy-1 H-indol-3-yl)piperidine- 1 -carboxylate.

To a solution of tert-butyl 4-(6-methoxy-1 H-indol-3-yl)-4-(nitromethyl) piperidine-1 -carboxylate (10 g, 25.7 mmol) in CH3OH (100 mL) and Chloroform (100 mL) was added NiCI 2 .6H 2 0 (15.26 g, 64.20 mmol). Then NaBH4 (14.6 g, 385 mmol) was added at 0°C. The reaction was stirred at 20°C for 2.5h. The reaction was diluted with water (1 0 mL) and extracted with EA (10 mL * 3). The combined organic layers were washed with brine (20 mL), dried over anhydrous Na2S04, filtered and concentrated to afford tert-butyl 4-(aminomethyl)-4-(6-methoxy-1 H-indol-3-yl) piperidine-1 -carboxylate (9.00 g) as light yellow solid. 1 H NMR (400MHz, CDC ) δ= 1 .38 (s, 9H), 1 .47-1 .39 (m, 2H), 1 .53 (m, 2H), 2.28-2.07 (m, 2H), 2.93 ( s, 3H), 3.67 (s, 4H), 3.82-3.70 (m, 2H), 6.62 (d, J=7.2 Hz, 1 H), 6.91 -6.76 (m, 2H),7.35 (d, J=7.0 Hz, 1 H) ppm; LCMS (ESI) m/z: [M+H] + =360.2 (Method: 5-95AB_R_220 & 254).

Preparation of Intermediate AJ: 1 '-(((tert-butyldimethylsilyl)oxy)methyl)-2'-(2-fluorobenzyl) -7'-methoxy-

1 ',2',3',9'-tetrahydrospiro[piperidine-4,4'-pyrido[3,4-b]indo le] TFA salt

Scheme:

Step 1 . Preparation of Intermediate AG : tert-butyl-1 -[[tert-butyl(dimethyl)silyl]oxymethyl]-7-methoxy-spiro [1 ,2,3,9-tetrahydropyrido[3,4-b]indol -4,4'-piperidine]-1 '-carboxylate

To a solution of tert-butyl 4-(aminomethyl)-4-(6-methoxy-1 H-indol-3-yl)piperidine-1 -carboxylate (5.00 g, 13.91 mmol) and MgSC (8.37 g, 69.55 mmol, 254.03 μΙ_) in CHC (30.00 mL) at 0°C. 2-[tert- butyl(dimethyl)silyl]oxyacetaldehyde (5.39 g, 27.82 mmol, 5.86 mL) was added dropwise at 0°C. The reaction stirred at 0°C. After 30 min, the solution was allowed to warm to 25 °C and was stirred 12 hrs. Then acetic acid (1 .67 g, 27.82 mmol, 1 .59 mL) was added dropwise. After the addition was completed, the reaction was stirred at 0°C for 3h. The reaction was diluted with water (1 00 mL) and extracted with DCM (50 mL * 3). The combined organic phase was washed with water (30ml * 2) and brine (30ml), dried over Na2S04, filtered and concentrated. The residue was purified by column chromatography (S1O2, PE:EA=10:1 -3:1 ). tert-butyl-1 -[[tert-butyl(dimethyl)silyl]oxymethyl]-7-methoxy-spiro[1 ,2,3,9- tetrahydropyrido[3,4-b]indole-4,4'-piperidine]-1 '-carboxylate (3.70 g, 6.02 mmol) was obtained as yellow solid. LCMS (ESI) m/z: [M+H] + = 51 6.3 Step 2. Preparation of Compound Al: tert-butyl 1 '-(((tert-butyldimethylsilyl)oxy)methyl)-2'-(2-fluorobenzyl) - 7'-methoxy-1 ',2',3',9'-tetrahydrospiro[piperidine-4,4'-pyrido[3,4-b]indo le]-1 -carboxylate

To a solution of tert-butyl 1 -[[tert-butyl(dimethyl)silyl]oxymethyl]-7-methoxy-spiro [1 ,2,3,9- tetrahydropyrido[3,4-b]indole-4,4'-piperidine]-1 '-carboxylate (3.50 g, 6.79 mmol, 1 .00 eg) in EteN (2.06 g, 20.37 mmol) was added l -(bromomethyl) -2-fluoro-benzene (1 .28 g, 6.79 mmol, 817.48 μΙ_) at 0 °C. The suspension was stirred for 3hrs at 25 °C. The reaction mixture was concentrated under vacuum to give a residue. The residue was purified by column chromatography(SiO2,PE:EA=10:1 to 1 :1 ) to afford tert-butyl 1 -[[tert-butyl(dimethyl) silyl]oxymethyl]-2- [(2-fluorophenyl) methyl]-7-methoxy-spiro[3,9-dihydro-1 H- pyrido[3,4-b]indole-4,4'-piperidine]-1 '-carboxylate (2.20 g, 3.50 mmol) as light yellow oil. LCMS (ESI) m/z: [M+H] + = 624.4

Step 3. Preparation of Intermediate AJ: 1 '-(((tert-butyldimethylsilyl)oxy)methyl)-2'-(2-fluorobenzyl) -7'- methoxy-1 ',2',3',9'-tetrahydrospiro[piperidine-4,4'-pyrido[3,4-b]indo le] TFA salt

To a solution of tert-butyl 1 -[[tert-butyl(dimethyl)silyl]oxymethyl] -2-[(2-fluorophenyl)methyl]-7- methoxy-spiro[3,9-dihydro-1 H-pyrido[3,4-b]indole-4,4'-piperidine]-1 '-carboxylate (2.20 g, 3.53 mmol) in DCM (25.00 ml_) was added TFA (4.02 g, 35.30 mmol). The reaction solution was stirred at 20 °C for 2 hour. The reaction solution was diluted with DCM (50ml) and washed with aq NaHC03 (15m 3) and brine (15ml * 2), dried over NaS04 and concentrated under vacuum. The residue was purified by Prep-HPLC (TFA) to afford tert-butyl-[[2-[(2-fluorophenyl)methyl]-7-methoxy-spiro[3,9- dihydro-1 H-pyrido[3,4-b]indole- 4,4'-piperidine]-1 -yl]methoxy]-dimethyl-silane TFA salt (1 .20 g) as light yellow solid. Preparation of [2-[(2-fluorophenyl)methyl]-1 -(hydroxymethyl)-7-methoxy-spiro [3,9-dihydro-1 H-pyrido[3,4- '-piperidine]-1 '-yl]-(2-pyridyl)methanone (Compound 8)

8 Preparation of [2-[(2-fluorophenyl)methyl]-1 -(hydroxymethyl)-7-methoxy-spiro [3,9-dihydro-1 H-pyrido[3,4- b]indole-4,4'-piperidine]-1 '-yl]-(2-pyrid

8

To a solution of tert-butyl-[[2-[(2-fluorophenyl)methyl]-7-methoxy-spiro[3,9- dihydro-1 H-pyrido [3,4- b]indole-4,4'-piperidine]-1 -yl]methoxy]-dimethyl-silane (300 mg, 573 umol) in DCM (20 mL) was added HOBT (101 mg, 745 umol), EDCI (143 mg, 745 umol) and pyridine-2-carboxylic acid (85 mg, 687 umol). The reaction mixture was stirred at 25 °C for 16 hrs. The reaction mixture was diluted with DCM (200 mL) and washed with water (50 mL * 2) and brine (30 mL), then dried over Na2S04 and concentrated under vacuum. The residue was dissolved in THF (20 mL) and TBAF (1 M, 1 .47mL) was added. The reaction mixture was stirred at 25 °C for 3 hrs. The reaction mixture was diluted with EA (200 mL) and washed with water (50 mL * 6) and brine (30 mL), then dried over Na2S04 and concentrated under vacuum. The residue was purified by Pre-HPLC (column: Phenomenex Synergi C18 150 * 25 * 1 Oum; mobile phase: [water(0.1 %TFA)-ACN]; B%:18%-48%,2min) to obtained [2-[(2-fluorophenyl)methyl]-1 -(hydroxymethyl)-7- methoxy -spiro[3,9-dihydro-1 H-pyrido[3,4-b]indole-4,4'-piperidine]-1 '-yl]-(2-pyridyl)methanone (15 mg, 28.55 umol) as a light yellow solid).

1 H NMR (400 MHz, Methanol-d4) δ= 1 .61 -1 .91 (m, 1 .5H), 1 .93-2.02 (m, 0.5H), 2.44-2.73 (m, 2H), 2.77- 2.98 (m, 2H), 3.1 1 -3.24 (m, 1 H), 3.49-3.77 (m, 2H), 3.83 (s, 3H), 3.89-4.06 (m, 1 H), 4.25-4.28 (m, 2H), 4.61 -4.63 (m, 2H), 4.70-4.79 (m, 1 H), 6.76 (dd, J=8.8, 2.1 Hz, 1 H), 6.93 (d, J=2.3 Hz, 1 H), 7.27-7.41 (m, 2H), 7.50-7.62 (m, 2H), 7.63-7.76 (m, 3H), 8.01 (td, J=7.8, 1 .4 Hz, 1 H), 8.61 -8.65 (m, 1 H) ppm ; LCMS (ESI) m/z: [M+H] + = 551 .3 (Method: 5-95AB_R_210 & 254).

[2-[(2-fluorophenyl)methyl]-1 -(hydroxymethyl)-7-methoxy-spiro[3,9- dihydro-1 H-pyrido[3,4-b]indole-4,4'- piperidine]-1 '-yl]-[4-(2-methoxyethoxy)phenyl]methanone TFA salt (Compound 9)

Scheme

Preparation of [2-[(2-fluorophenyl)methyl]-1 -(hydroxymethyl)-7-methoxy-spiro[3,9- dihydro-1 H-pyrido[3,4- b]indole-4,4'-piperidine]-1 '-yl]-[4-(2-methoxyethoxy)phenyl]methanone TFA salt.

9

To a solution of tert-butyl-[[2-[(2-fluorophenyl)methyl]-7-methoxy-spiro[3,9- dihydro-1 H-pyrido [3,4- b]indole-4,4'-piperidine]-1 -yl]methoxy]-dimethyl-silane (300 mg, 573 umol) in DCM (30 mL) was added HOBT (101 mg, 745 umol), EDCI (143 mg, 745 umol), TEA (239 μΙ_, 1 .72 mmol) and 4-(2- methoxyethoxy)benzoic acid (135 mg, 687 umol). The reaction mixture was stirred at 25 °C for 16 hours. The reaction mixture was then diluted with DCM (200 mL) and washed with water (50 mL * 2) and brine (30 mL), then dried over Na2S04 and concentrated under vacuum. The residue was dissolved in THF (20 mL) and TBAF (1 M, 1 .71 9 mL 1 .72 mmol) was added. The reaction mixture was stirred at 25 °C for 3 hrs. The reaction mixture was diluted with EA (200 mL) and washed with H2O (50 mL * 6) and brine (30 mL * 1 ), then dried over Na2S04 and concentrated under vacuum. The residue was purified by Pre-HPLC (column: Phenomenex Synergi C18 150 * 25 * 1 Oum; mobile phase: [water(0.1 %TFA)-ACN];B%:18%- 48%,2min) to obtained [2-[(2-fluorophenyl)methyl] -1 -(hydroxymethyl)-7-methoxy-spiro[3,9-dihydro-1 H- pyrido[3,4-b]indole-4,4'-piperidine]-1 '-yl]-[4-(2-methoxyethoxy)phenyl]methanone TFA salt (76 mg, 125 umol) as a light yellow solid).

1 H NMR (400 MHz, Methanol-d4) δ= 1 .65-1 .97 (m, 2H), 2.46-2.49 (m, 2H), 2.77-3.27 (m, 2H), 3.44 (s, 3H), 3.61 -3.64 (m, 1 H), 3.74-3.80 (m, 2H), 3.83 (s, 3H), 4.01 -4.06 (m, 1 H), 4.16-4.21 (m, 2H), 4.22-4.39 (m, 2H), 4.62-4.76 (m, 3H), 6.78 (dd, J=8.8, 2.3 Hz, 1 H), 6.94 (d, J=2.3 Hz, 1 H), 7.07 (d, J=8.7 Hz, 2H), 7.32-7.42 (m, 2H), 7.47 (d, J=8.7 Hz, 2H), 7.55 (d, J=8.8 Hz, 1 H), 7.57-7.65 (m, 1 H), 7.70-7.76 (m, 1 H) ppm; LCMS (ESI) m/z: [M+H] + = 588.3 (Method: 5-95AB_R_21 0 & 254). Preparation of 1 -[2-[(2-fluorophenyl)methyl]-1 -(hydroxylmethyl)-7-methoxy-spiro [3,9-dihydro-1 H- pyrido[3,4-b]indole-4,4'-piperidine]-1 '-yl]-3-methoxy-propan-1 -one. (Compound 1 1 )

Preparation of 1 -[2-[(2-fluorophenyl)methyl]-1 -(hydroxylmethyl)-7-methoxy-spiro [3,9-dihydro-1 H- pyrido[3,4-b]indole-4,4'-piperidine]-1 '-yl]-3-methoxy-propan-1 -one

11

To a solution of tert-butyl-[[2-[(2-fluorophenyl)methyl]-7-methoxy-spiro[3,9- dihydro-1 H-pyrido [3,4- b]indole-4,4'-piperidine]-1 -yl]methoxy]-dimethyl-silane (200 mg, 382 umol) in DCM (20 mL) was added 3- methoxypropanoic acid (48 mg, 458 umol,), HOBT (67 mg, 496.41 umol), EDCI (95 mg, 496 umol) and TEA (159 μΙ_, 1 .15 mmol). The reaction was stirred at 25 °C for 16 hrs. The reaction mixture was diluted with DCM (200 mL) and washed with water (50 ml_ * 2) and brine (30 mL), then dried over Na2S04 and concentrated under vacuum. The residue was dissolved in THF (20 mL) and TBAF (1 M, 1 .47mL, 1 .47 mmol) was added. The reaction mixture was stirred at 25 °C for 3 hrs. The reaction mixture was diluted with EA (200 mL) and washed with water (50 mL * 6) and brine (30 mL * 1 ), then dried over Na2S04 and concentrated under vacuum. The residue was purified by Pre-HPLC (column: Phenomenex Synergi C18 150 * 25 * 1 Oum ; mobile phase: [water(0.1 %TFA)-ACN];B%:18%-48%,2min) to obtain 1 -[2-[(2- fluorophenyl)methyl]-1 -(hydroxymethyl)-7-methoxy-spiro[3,9-dihydro-1 H-pyrido[3,4-b]indole-4,4'- piperidine]-1 '-yl]-3-methoxy-propan-1 -one (1 10 mg, 212 umol) as a light yellow solid.

1 H NMR (400 MHz, DMSO-d6) δ= 1 .44-1 .52 (m, 1 .5H), 1 .62-1 .73 (m, 0.5H), 1 .79-1 .89 (m, 0.5H), 1 .96- 1 .99 (m, 0.5H), 2.03-2.13 (m, 1 H), 2.14-2.30 (m, 2H), 2.41 -2.47 (m, 1 H), 2.64-2.66 m, 2H), 2.73-2.83 (m, 1 H), 3.02-3.13 (m, 1 H), 3.25 (m, 2H), 3.49-3.63 (m, 2H), 3.73 (s, 3H), 3.76-3.86 (m, 3H), 4.12-4.26 (m, 2H), 4.78 (q, J=5.0 Hz, 1 H), 6.57 (d, J=8.8 Hz, 1 H), 6.87 (t, J=2.7 Hz, 1 H), 7.16-7.25 (m, 2H), 7.28-7.39 (m, 2H), 7.54-7.64 (m, 1 H), 10.47 (br d, J=5.5 Hz, 1 H) ppm ; LCMS (ESI) m/z: [M+H] + = 496.3 (Method: 5- 95AB_R_210&254). Preparation of [2-[(2-fluorophenyl)methyl]-1 -(hydroxymethyl)-7-methoxy-spiro[3,9- dihydro-1 H-pyrido[3,4- b]indole-4,4'-piperidine]-1 '-yl]-phenyl-methanone TFA salt. (Compound 19)

Scheme

Preparation of [2-[(2-fluorophenyl)methyl]-1 -(hydroxymethyl)-7-methoxy-spiro[3,9- dihydro-1 H-pyrido[3,4- b]indole-4,4'-piperidine]-1 '-yl]-phenyl-methanone TFA salt

19

To a solution of tert-butyl-[[2-[(2-fluorophenyl)methyl]-7-methoxy-spiro[3,9- dihydro-1 H-pyrido

[3,4-b]indole-4,4'-piperidine]-1 -yl]methoxy]-dimethyl-silane (300 mg, 573 umol) in DCM (30 mL) was added HOBt (1 01 mg, 745 umol), EDCI (143 mg, 745 umol), TEA (1 .72 mmol, 239.1 8 xL) and benzoic acid (84 mg, 687 umol). The reaction mixture was stirred at 25 °C for 1 6 hrs. The reaction mixture was diluted with DCM (200 mL) and washed with water (50 mL*2) and brine (30 mL), then dried over Na2SC>4 and concentrated under vacuum. The residue was dissolved in THF (20 mL) and TBAF (1 M, 1 .34 mL, 1 .34 mmol) was added. The reaction mixture was stirred at 25 °C for 3 hrs. The reaction mixture was diluted with EA (200 mL) and washed with H 2 0 (50 mL*6) and brine (30 mL), then dried over Na2SC>4 and concentrated under vacuum. The residue was purified by Pre-H PLC (column : Phenomenex Synergi C1 8 1 50*25*1 Oum ; mobile phase: [water(0.1 %TFA)-ACN];B%:1 8%-48%,2min) to obtain [2-[(2- fluorophenyl)methyl]-1 -(hydroxymethyl)-7-methoxy-spiro[3,9-dihydro-1 H-pyrido[3,4-b]indole-4,4'- piperidine]-1 '-yl]-phenyl-methanone (90 mg, 1 75 umol) as a yellow solid).

1 H NMR (400 MHz, Methanol-d4) δ= 1 .60-2.09 (m , 2H), 2.30-2.72 (m , 2H), 2.75-3.03 (m, 1 H), 3.1 9-3.21 (m , 1 H), 3.58-3.78 (m, 2H), 3.83 (s, 3H), 3.99-4.1 0 (m , 1 H), 4.21 -4.37 (m, 2H), 4.66-4.68 (m , 2H), 4.72- 4.83 (m, 2H), 6.79 (dd, J=8.8, 2.3 Hz, 1 H), 6.94 (d, J=2.3 Hz, 1 H), 7.31 -7.42 (m, 2H), 7.47-7.53 (m, 5H), 7.56 (d, J=8.8 Hz, 1 H), 7.59-7.61 (m, 1 H), 7.71 -7.73 (m , 1 H) ppm ; LCMS (ESI) m/z: [M+H] + = 514.3 (Method: 5-95AB_R_21 0 & 254).

Preparation of[2-[(2-fluorophenyl)methyl]-1 -(hydroxymethyl)-7-methoxy-spiro [3,9-dihydro-1 H-pyrido[3,4- b]indole-4,4'-piperidine]-1 '-yl]-(4-methoxyphenyl)methanone TFA salt. (Compound 20)

Scheme

Preparation of [2-[(2-fluorophenyl)methyl]-1 -(hydroxymethyl)-7-methoxy-spiro [3,9-dihydro-1 H-pyrido[3,4- b]indole-4,4'-piperidine]-1 '-yl]-(4-methoxyphenyl)methanone TFA salt.

20

To a solution of tert-butyl-[[2-[(2-fluorophenyl)methyl]-7-methoxy-spiro[3,9- dihydro-1 H-pyrido [3,4- b]indole-4,4'-piperidine]-1 -yl]methoxy]-dimethyl-silane (300 mg, 573 umol) in DCM (2 ml_) was added HOBt (1 01 mg, 745 umol), EDCI (1 43 mg, 745 umol), TEA (238.1 9 μΙ_, 1 .72 mmol) and 4-methoxybenzoic acid (1 05 mg, 687 umol). The reaction mixture was stirred at 25 °C for 1 6 hrs. The reaction mixture was diluted with DCM (200 ml_) and washed with water (50 ml_*2) and brine (30 ml_), then dried over Na2SC>4 and concentrated under vacuum. The residue was dissolved in THF (20 mL) and TBAF (1 M, 1 .4 ml_, 1 .4 mmol)) was added. The reaction mixture was stirred at 25 °C for 3 hrs. The reaction mixture was diluted with EA (200 mL) and washed with water (50 ml_*6) and brine (30 mL), then dried over Na2S04 and concentrated under vacuum. The residue was purified by Pre-H PLC (column : Phenomenex Synergi C1 8 1 50*25*1 Oum ; mobile phase: [water(0.1 %TFA)-ACN];B%:1 8%-48%,2min) to obtained [2-[(2- fluorophenyl)methyl] -1 -(hydroxymethyl)-7-methoxy-spiro[3,9-dihydro-1 H-pyrido[3,4-b]indole-4,4'- piperidine]-1 '-yl]-(4-methoxyphenyl)methanone (1 06 mg, 1 95 umol), as a light yellow solid.

1 H NMR (400 MHz, Methanol-d4) δ= 1 .64-1 .98 (m , 2H), 2.40-5-2.71 (m , 2H), 2.75-3.28 (m, 2H), 3.66-3.76 (m , 1 H), 3.83 (s, 3H), 3.86 (s, 3H), 4.07-4.1 1 (m, 1 H), 4.22-4.40 (m , 2H), 4.54-4.58 (m, 1 H), 4.67-4.78 (m, 2H), 4.78-4.85 (m , 2H), 6.78 (dd, J=2.3, 8.8 Hz, 1 H), 6.94 (d, J=2.3 Hz, 1 H), 7.04 (d, J=8.8 Hz, 2H), 7.42 - 7.32 (m, 2H), 7.47 (d, J=8.7 Hz,2H), 7.55 (d, J=8.8 Hz, 1 H), 7.66 - 7.57 (m, 1 H), 7.73 (t, J=7.0 Hz, 1 H) ppm ; LCMS (ESI) m/z: [M+H] + = 544.4 (Method: 5-95AB_R_21 0 & 254).

Preparation of [1 '-(cyclobutylmethyl)-2-[(2-fluorophenyl)methyl] -7-methoxy-spiro [3,9-dihydro-1 H- pyrido[3,4-b]indole-4,4'-piperidine]-1 -yl]methanol TFA salt (Compound 21 )

AJ Preparation of [1 '-(cyclobutylmethyl)-2-[(2-fluorophenyl)methyl] -7-methoxy-spiro [3,9-dihydro-1 H- pyrido[3,4-b]indole-4,4'-piperidine]-1 -yl]methanol TFA salt.

21

To a solution of tert-butyl-[[2-[(2-fluorophenyl)methyl]-7-methoxy-spiro[3,9- dihydro -1 H-pyrido[3,4- b]indole-4,4'-piperidine]-1 -yl]methoxy]-dimethyl-silane (150 mg, 286 umol) in DMF (10 mL) was added bromomethylcyclobutane (51 mg, 344 umol) and K2CO3 (1 18.75 mg, 859.18 umol). The mixture was stirred for 16 hrs at 30 °C. The reaction mixture was quenched by addition water (50 mL) and then extracted with EA (20 mL * 3). The combined organic layers were washed with water (15 mL * 2), dried over Na2S04, filtered and concentrated under reduced pressure to give a residue. The residue was dissolved in THF (20 mL) and TBAF (1 M, 1 .12 mL 1 .12 mmol) was added. The mixture was stirred at 30 °C for 3 hr. The reaction mixture was diluted with EA (100 mL) and washed with water (20 mL * 6), then dried over anhydrous Na2S04 and concentrated under vacuum. The residue was purified by Pre-HPLC (column: Phenomenex Synergi C18 150 * 25 * 1 Oum; mobile phase: [water (0.1 % TFA)-ACN]; B%: 8%-38%, 2min). [1 '-(cyclobutylmethyl)-2-[(2-fluorophenyl)methyl]-7-methoxy-sp iro[3,9-dihydro- 1 H-pyrido[3,4- b]indole-4,4'-piperidine]-1 -yl]methanol TFA salt (80 mg) as a light yellow solid).

1 H NMR (400MHz, Methanol-d4), 1 .84-2.14 (m, 6H), 2.1 7-2.32 (m, 2H) , 2.64-2.95 (m, 3H), 2.98-3.1 1 (m, 2H), 3.22 (d, J=7.0 Hz, 2H), 3.45-3.51 (m, 3H), 3.81 (s, 3H), 3.89-4.02 (m, 1 H), 4.12-4.29 (m, 2H), 4.41 - 4.52 (m, 1 H), 4.60-4.65 (m, 2H), 6.70 (m, 1 H), 6.92 (d, J=2.1 Hz, 1 H), 7.24-7.38 (m, 2H), 7.50 - 7.58 (m, 1 H), 7.65 (d, J=8.8 Hz, 1 H), 7.73 (m, 1 H) ppm ; LCMS (ESI) m/z: [M+H] + = 478.3 (Method: 5-95AB_R_220 & 254).

Preparation of 1 -[2-[(2-fluorophenyl)methyl]-1 -(hydroxylmethyl)-7-methoxy-spiro [3,9-dihydro-1 H- pyrido[3,4-b]indole-4,4'-piperidine]-1 '-yl]ethanone TFA salt (Compound 22)

Preparation of 1 -[2-[(2-fluorophenyl)methyl]-1 -(hydroxylmethyl)-7-methoxy-spiro [3,9-dihydro-1 H- pyrido[3,4-b]indole-4,4'-piperidine]-1 '-yl]ethenone TFA salt

22

To a solution of tert-butyl-[[2-[(2-fluorophenyl)methyl]-7-methoxy-spiro[3,9- dihydro-1 H-pyrido [3,4- b]indole-4,4'-piperidine]-1 -yl]methoxy]-dimethyl-silane (200 mg, 381 .86 umol) in THF (20 mL) was added acetyl chloride (36 mg, 458 umol, 32.7 μΙ_,) and TEA (77 mg, 764 umol, 106 μΙ_). The mixture was stirred for 1 hr at 25 °C then poured into H2O (1 00 mL) and extracted with EA (15 mL * 3). The combined organic layer was washed with water (15 mL * 3) and brine (15mL * 2), dried over Na2S04, concentrated under vacuum. The residue was dissolved in THF (20 mL) and TBAF (1 M, 1 .2 mL) was added. The reaction mixture was stirred at 25 °C for 3 hrs. The reaction mixture was diluted with EA (200mL) and washed with water (50 mL * 6) and brine (30 mL), then dried over Na2S04 and concentrated under vacuum. The residue was purified by Pre-HPLC (column: Phenomenex Synergi C18 150 * 25 * 10um;mobile phase: [water(0.1 %TFA)-ACN];B%: 20%-50%,12min) to obtained 1 -[2-[(2-fluorophenyl)methyl] -1 - (hydroxymethyl)-7-methoxy-spiro[3,9-dihydro-1 H-pyrido[3,4-b]indole-4,4'-piperidine]-1 '-yl]ethanone (96 mg).

1 H NMR (400MHz, METHANOL-d4) 51 .94 - 1 .71 (m, 2H) ,2.18 (s, 3H), 2.47-2.51 (m, 2H), 2.74 - 2.60 (m, 1 H), 3.1 7-3.21 (m, 1 H), 3.64 (br s, 1 H) , 3.83 (s, 3H) ,4.08 - 3.88 (m, 2H), , 4.40 -4.23 (m, 2H), 4.50-4.54 (m, 1 H), 4.78 - 4.68 (m, 2H), 4.81 -4.83 (m, 2H) ,6.74 (dd, J=2.2, 8.8 Hz, 1 H), 6.93 (d, J=2.1 Hz, 1 H), 7.43 - 7.31 (m,2H), 7.47-7.49 (m, 1 H), 7.66 - 7.58 (m, 1 H), 7.71 -7.75 (br t, J=7.5 Hz, 1 H) ppm ; LCMS (ESI) m/z: [M+H]+ = 452.3 (Method: 5-95AB_R_ 210 & 254).

The following compounds were prepared analogously:

Preparation of Methyl 2-[(2-fluorophenyl)methyl]-1 -(hydroxymethyl)-7-methoxy -spiro[3,9-dihydro-1 H-

Preparation of Methyl 2-[(2-fluorophenyl)methyl]-1 -(hydroxymethyl)-7-methoxy -spiro[3,9-dihydro-1 H- pyrido[3,4-b]indole-4,4'-piperidine]-1 '-carboxylate TFA salt

23

Ή NMR (400MHz, Methanol-d4) δ= 1 .88 - 1 .66 (m, 2H), 2.42-2.46 (m, 2H), 2.87-2.91 (m, 1 H) ,3.70 - 3.58 (m, 1 H) ,3.76 (s, 3H), 3.83 (s, 3H), 4.00-4.03 (m, 1 H), 4.10-4.13(m, 2H), 4.40 - 4.22 (m, 2H), 4.72-4.75 (m, 2H), 4.81 -4.83 (m, 2H), 6.74 (dd, J=2.3, 8.8 Hz, 1 H), 6.93 (d, J=2.2 Hz, 1 H), 7.44 - 7.30 (m,2H), 7.49 (d, J=8.8 Hz, 1 H), 7.68 - 7.56 (m, 1 H), 7.70-7.74 (m, 1 H) ppm; LCMS (ESI) m/z: [M+H] + = 468.3 (Method: 5-95AB_R_210 & 254)

Preparation of A86 2-[(2-fluorophenyl)methyl]-1 -(hydroxymethyl)-7-methoxy-N-methyl -spiro[3,9-dihydro- 1 H-pyrido[3,4-b]indole-4,4'-piperidine]-1 '-carboxamide TFA salt (Compound 26)

26

1 H NMR (400 MHz, Methanol-d4) δ= 1 .57-1 .61 (m, 1 H), 1 .66-1 .69 (m, 1 H), 2.31 -2.36 (m, 2H), 2.67 (s, 3H),2.69-2.81 (m, 2H), 3.53-3.56 (m, 1 H), 3.70 (s, 3H), 3.81 -3.97 (m, 3H), 4.10-4.30 (m, 2H), 4.63-4.66 (m, 2H),4.73 (s, 1 H), 6.62 (dd, J=8.74, 2.26 Hz, 1 H), 6.81 (d, J=2.20 Hz, 1 H), 7.21 -7.32 (m, 2H), 7.36 (d, J=8.80 Hz, 1 H), 7.48-7.55 (m,1 H), 7.61 (td, J=7.55, 1 .41 Hz, 1 H) ppm; LCMS (ESI) m/z: [M+H] + = 467.4 (Method: 5-95AB_R_210 & 254). Preparation of Compound 2-[(2-fluorophenyl)methyl]-1 -(hydroxymethyl)-7-methoxy-N,N- dimethyl- -dihydro-1 H-pyrido[3,4-b]indole-4,4'-piperidine]-1 '-carboxamide TFA salt. (Compound 27)

Preparation of 2-[(2-fluorophenyl)methyl]-1 -(hydroxymethyl)-7-methoxy-N,N- dimethyl-spiro[3,9-dihydro- 1 H-pyrido[3,4-b]indole-4,4'-piperidin -1 '-carboxamide TFA salt

27

1 H NMR (400 MHz, Methanol-d4) δ= 0.91 (t, J=7.40 Hz, 1 H), 1 .31 -1 .35 (m,1 H), 1 .53-1 .73 (m, 3H), 2.41 - 2.44 (m, 2H), 2.69-2.78 (m, 2H), 2.80 (s, 6H), 3.00-3.06 (m, 1 H), 3.47-3.54 (m, 2H), 3.71 (s, 3H), 3.88- 3.91 (m,1 H), 4.1 1 -4.19 (m, 1 H), 4.20-4.28 (m, 1 H), 4.61 -4.64 (m, 2H), 4.71 (s, 1 H), 6.62 (dd, J=8.8, 2.3 Hz, 1 H), 6.82 (d, J=2.2 Hz, 1 H), 7.20-7.30 (m, 2H), 7.41 (d, J=8.8 Hz, 1 H), 7.46-7.54 (m, 1 H), 7.61 (t, J=7.03 Hz, 1 H) ppm ; LCMS (ESI) m/z: [M+H] + = 481 .4 (Method: 5-95AB_R_210 & 254).

Preparation of [2-[(2-fluorophenyl)methyl]-7-methoxy-1 '-(2-pyridyl)spiro[3,9-dihydro- 1 H-pyrido[3,4- b]indole-4,4'-piperidine]-1 -yl]methanol TFA salt. (Compound 28)

Scheme

Preparation of [2-[(2-fluorophenyl)methyl]-7-methoxy-1 '-(2-pyridyl)spiro[3,9-dihydro- 1 H-pyrido[3,4- b]indole-4,4'-piperidine]-1 -yl]methan

28

To a solution of tert-butyl-[[2-[(2-fluorophenyl)methyl]-7-methoxy-spiro[3,9- dihydro-1 H-pyrido [3,4- b]indole-4,4'-piperidine]-1 -yl]methoxy]-dimethyl-silane (100 mg, 191 umol) in N-methylmorpholine(5 mL) was added 2-fluoropyridine (49 μί, 573 umol) and Na2C03 (101 mg, 955 umol). The mixture was stirred for 16 hrs at 1 15 °C. The reaction was poured into water (50 mL) and extracted with EA (10 mL * 3). The combined organic layer was washed with water (10 mL * 3), brine (10 mL), dried over Na2S04, filtered and concentrated. The residue was diluted in THF (15 mL) and TBAF (1 M, 849 μί, 849 mmol) was added. The mixture was stirred for 3 hrs at 30 °C then diluted with EA (100 mL) and washed with water (20mL * 6), dried over Na2S04, concentrated under vacuum. The residue was purified by Pre-HPLC (column:

Phenomenex Synergi C18 150 * 25 * 10um ;mobile phase: [water (0.1 %TFA) -ACN];B%:5%-35%,min) to obtained [2-[(2-fluorophenyl)methyl]-7-methoxy-1 '-(2-pyridyl)spiro[3,9-dihydro-1 H-pyrido[3,4-b] indole-4,4'- piperidine]-1 -yl]methanol TFA salt (25 mg) as a light yellow solid.

1 H NMR (300 MHz, Methanol-d4) δ= 1 .91 -1 .93 (m, 1 H), 2.03-2.14 (m, 1 H), 2.45-2.80 (m, 2H), 3.18-3.28 (m, 2H), 3.83 (s, 3H), 4.1 1 -4.14 (m, 2H), 4.23-4.26 (m,2H), 4.46-4.49 (m, 2H), 4.64-4.67 (m, 1 H), 6.64- 6.70 (m, 1 H), 6.93 (d, J=2.1 Hz, 1 H), 7.01 (t, J=6.6 Hz, 1 H), 7.23-7.42 (m 4H), 7.50-7.54 (m, 1 H), 7.70- 7.73 (m, 1 H), 7.96-8.08 (m, 2H) ppm ; LCMS (ESI) m/z: [M+H] + = 487.5 (Method: 5-95AB_R_21 0&254).

Preparation of [1 '-cyclobutylsulfonyl-2-[(2-fluorophenyl)methyl]-7-methoxy-sp iro [3,9-dihydro-1 H- pyrido[3,4-b]indole-4,4'-piperidine]-1 -yl]methanol TFA salt. (Compound 30)

Preparation of [1 '-cyclobutylsulfonyl-2-[(2-fluorophenyl)methyl]-7-methoxy-sp iro [3,9-dihydro-1 H- pyrido[3,4-b]indole-4,4'-piperidine]-1 -yl]methanol.

30

To a solution of tert-butyl-[[2-[(2-fluorophenyl)methyl]-7-methoxy-spiro[3,9- dihydro-1 H -pyrido[3,4- b]indole-4,4'-piperidine]-1 -yl]methoxy]-dimethyl-silane (1 00 mg, 1 91 umol) in THF (2 mL) was added cyclobutanesulfonyl chloride (59.04 mg, 381 .86 umol ) and TEA (58 mg, 573 umol, 79.73 μΐ). The mixture was stirred at 30 °C for 3 hrs. The mixture was poured into water (1 00 mL) and extracted with EA (1 5 mL*3). The combined organic layer was washed with water (1 5ml*3) and brine (1 5 mL*2), dried over Na2S04, concentrated under vacuum . The residue was dissolved in THF (20 mL) and TBAF (1 M, 1 .47 mL, 1 .47 mmol) was added. The reaction mixture was stirred at 25 °C for 3 hrs. The reaction mixture was diluted with EA (200 mL) and washed with water (50 mL*6) and brine (30 mL), then dried over Na2S04 and concentrated under vacuum. The residue was purified by Pre-HPLC (column : Phenomenex Synergi C1 8 1 50*25*1 0um ;mobile phase: [water(0.1 %TFA)-ACN];B%: 20%-50%, 12min) to obtained [V- cyclobutylsulfonyl-2-[(2-fluorophenyl)methyl]-7-methoxy-spir o[3,9-dihydro-1 H-pyrido[3,4-b]indole-4,4'- piperidine]-1 -yl]methanol TFA salt (1 0 mg) as a light yellow solid.

1 H NMR (400 MHz, Methanol-d4) δ= 1 .71 -1 .75 (m , 1 H), 1 .84-1 .88 (m , 1 H), 1 .97-2.08 (m, 1 H), 2.09- 2.20(m , 1 H), 2.27-2.31 (m , 3H), 2.36-2.54 (m, 3H), 2.71 -2.74 (m , 3H), 3.65-3.68 (m, 2H), 3.83 (s, 3H), 3.91 -4.03 (m, 1 H), 4.21 -4.26 (m , 2H), 4.39-4.76 (m, 2H), 6.74 (dd, J=8.85, 2.32 Hz, 1 H), 6.93 (d, J=2.1 3 Hz, 1 H), 7.29-7.40 (m , 2H), 7.51 -7.55 (m, 2H), 7.67-7.69 (m, 1 H) ppm ; LCMS (ESI) m/z: [M+H] + = 528.4 (Method: 5-95AB_R_21 0 & 254).

Compounds 32-61 were synthesized using methods similar to the methods described above.

Experimental LCMS data for compounds 32-61 is provided in Table 2:

Table 2. Experimental LCMS data for compounds of the invention

37 518.3

38 475.4

39 475.3

40 521 .2

41 542.1

42 542.1

43 475.1

44 546.1

45 532.1

46 571 .1

47 516.1

48 558.1

49 585.5

50 531 .0

51 464.4

52 591 .8

53 515.2

54 560.0

55 643.8

56 515.0

57 528.0

58 521 .5

59 563.4

60 542.3

61 542.4

Example 2. Inhibition of Proliferation of Synovial Sarcoma Cells

Method: The effect of compounds on synovial sarcoma cell viability was measured by CellTiter Glo® assay. Yamato-SS cells were first plated under standard conditions in 96-well plate. Compounds were added 24 hours after cell seeding and Cell Titer Glo reagents was used to measure cell viability after 48 hours of compound treatment compared to DMSO as negative control. Staurosporine was used as a positive control. The results are shown in Table 3 below.

Table 3. Inhibition of Proliferation by Selected Compounds

13 ++

21 +++

26 ++

27 ++

28 ++

30 +++

31 ++

25 ++

24 +

22 ++

23 +++

19 ++

20 ++

15 +++

16 +++

17 ++

18 ++

7 ++

10 +++

8 ++

9 ++

'+' - 5-25% inhibition; '++' - 26-50% inhibition; '+++' - >50% inhibition

Example 3. SS18-SSX1 Partial Protein Thermal Shift Assay

All assays were carried out in 384 well plates. In a round bottom polypropylene tube, the DDB7 construct (human SS18 (294-379) + human SSX1 (1 1 1 -1 88)) was diluted with assay buffer, so that the final reaction concentration was 50 mM imidazole (pH 7), 450 mM NaCI, 3% glycerol, and 20 μΜ protein. The tube was centrifuged briefly to remove precipitate and 44.5 μΙ_ of buffered protein solution was then added to round bottom polypropylene plate (Corning #3657).

The plate had been prepared with 0.5 μΙ_ of the DMSO controls or compounds plated and diluted from 20 mM DMSO stocks to a final concentration range of 0 to 50 μΜ with 2-fold dilutions (1 .0% v/v DMSO). Finally, 5 μΙ_ of Sypro Orange (Life Technologies #S6651 ) dye was aliquoted per well, and mixed into the buffered protein and compound solution to a final dye concentration of 20X and reaction volume of 50 μΙ_. This reaction plate was sealed, centrifuged for 10 seconds at 800xg, and incubated in the dark at room temperature for 75 minutes.

Subsequently aliquots (15 μΙ_) of each well were transferred to a PCR plate (Brandtech #781347) in triplicate, sealed with optical polyolefin sealing film (VWR # 89134-428) and centrifuged (2 min,

1000xg) to be analyzed on a Bio-Rad CFX384 instrument using a temperature gradient of 25-80 °C and a scanning rate of 2 °C/min. The midpoint of the melting transitions (Tm) were assessed using Bio-Rad CFX Manager 3.1 instrument software, measuring the first derivative of the rate of fluorescence change as a function of temperature. Compound-induced changes in the melting temperature, dTm, were calculated relative to DMSO controls within the same plate.

Experimental DFS data for compounds 32-61 is provided in Table 4:

Table 4. DFS data for compounds of the invention

60 ++++ ++++ ++++

61 ++++ ++++ ++++

'+' -1 .07-0.99; '++' 1 .0 to 2.99; '+++' 3.0 to 4.99; '++++' 5.0 to 12.2

Other Embodiments

While the present invention has been described with reference to what are presently considered to be the preferred examples, it is to be understood that the invention is not limited to the disclosed examples. To the contrary, the invention is intended to cover various modifications and equivalent arrangements included within the spirit and scope of the appended claims.

All publications, patents and patent applications are herein incorporated by reference in their entirety to the same extent as if each individual publication, patent or patent application was specifically and individually indicated to be incorporated by reference in its entirety. Where a term in the present application is found to be defined differently in a document incorporated herein by reference, the definition provided herein is to serve as the definition for the term.

Other embodiments are in the claims.