Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
AMIDO COMPOUNDS AS RORγt MODULATORS AND USES THEREOF
Document Type and Number:
WIPO Patent Application WO/2011/112263
Kind Code:
A1
Abstract:
Amido compounds are disclosed that have a formula represented by the following (I) and wherein n l, n2, Rla, Rlb, R2, R3, R4, R5, and R6 are as described herein. The compounds may be prepared as pharmaceutical compositions, and may be used for the prevention and treatment of a variety of conditions in mammals including humans, including by way of non-limiting example, inflammatory conditions, autoimmune disorders, cancer, and graft-versus-host disease.

Inventors:
LITTMAN DAN (US)
HUH JUN R (US)
HUANG RUILI (US)
HUANG WENWEI (US)
ENGLUND ERIKA ELAINE (US)
Application Number:
PCT/US2011/000459
Publication Date:
September 15, 2011
Filing Date:
March 11, 2011
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
UNIV NEW YORK (US)
US GOV HEALTH & HUMAN SERV (US)
LITTMAN DAN (US)
HUH JUN R (US)
HUANG RUILI (US)
HUANG WENWEI (US)
ENGLUND ERIKA ELAINE (US)
International Classes:
A61K31/33; A61K31/12; A61K31/535
Domestic Patent References:
WO2008031227A12008-03-20
WO2006007486A22006-01-19
Foreign References:
US20040147529A12004-07-29
EP2181710A12010-05-05
US4603145A1986-07-29
Other References:
KUMAR ET AL.: "The benzensulfonamide T0901317 [N-(2,2,2-trifuoroethyl)-N-[4-[2,2,2-trifluoro-1- hydroxy-(trifluoromethyl)ethyl]phenyl]-benzenesulfonamide] is a novel retinoic acid receptor- related orphan receptor-alpha/gamma inverse agonist", MOLECULAR PHARMACOLOGY, vol. 77, no. 2, 3 November 2009 (2009-11-03), pages 228 - 236, XP002645391
WANG ET AL.: "A second class of nuclear receptors for oxysterols: regulation of RORalpha and RORgamma activity by 24S-hydroxycholesterol (cerebrosterol)", BICHIMIICA ET BIOPHYSICA ACTA, vol. 1801, 6 March 2010 (2010-03-06), pages 917 - 923, XP027086280
JIN ET AL.: "Structural basis for hydroxycholesterols as natural ligands of orphan nuclear receptor for RORgamma", MOL ENDOCRINOLOGY, vol. 24, 4 March 2010 (2010-03-04), pages 923 - 929, XP055098204
IVANOV, II ET AL., CELL, vol. 126, 2006, pages 1121 - 33
WEAVER, C. ET AL., ANN. REV. IMMUNOL., vol. 25, 2007, pages 821 - 52
KRYCZEK, 1. ET AL., J. IMMUNOL., vol. 178, 2007, pages 6730 - 3
CUA, D. J. ET AL., NATURE, vol. 421, 2003, pages 744 - 8
J. EXP. MED., vol. 201, 2005, pages 233 - 40
YEN, D. ET AL., J. CLIN. INVEST., vol. 116, 2006, pages 1310 - 6
CARLSON, M.J. ET AL., BLOOD, 28 October 2008 (2008-10-28)
KAPPEL, L.W. ET AL., BLOOD, 17 October 2008 (2008-10-17)
PATHOLOGY,, vol. 172, 2008, pages 146 - 55
YU, J.J.; GAFFEN, S.L, FRONT. BIOSCI., vol. 13, 2008, pages 170 - 77
ZHENG, Y. ET AL., NATURE, vol. 445, 2007, pages 648 - 51
DUERR, R.H. ET AL., SCIENCE, vol. 314, 2006, pages 1461 - 63
IVANOV, I.I., CELL HOST & MICROBE, vol. 4, 2008, pages 337 - 49
SATO, K. ET AL., J. EX. MED., vol. 203, 2008, pages 2673 - 82
IVANOV, I.I. ET AL., CELL, vol. 126, 2006, pages 1121 - 33
UEDA, H. R. ET AL., NATURE, vol. 418, 2002, pages 534 - 39
SATO, T. K. ET AL., NEURON, vol. 43, 2004, pages 527 - 37
KUMAKI, Y. ET AL., PNAS, vol. 105, 2008, pages 14946 - 51
LIU, C. ET AL., NATURE, vol. 447, 2007, pages 477 - 81
YANG, X. ET AL., CELL, vol. 126, 2006, pages 801 - 10
KUMAR ET AL., MOL. PHARMACOL., vol. 77, no. 2, 3 November 2009 (2009-11-03), pages 228 - 236
BUNDGARD, H.: "Design of Prodrugs", 1985, ELSEVIER, pages: 7 - 9,21-24
CHEN ET AL., J INNATE IMMUN., vol. 2, no. 4, 7 May 2010 (2010-05-07), pages 325 - 33
T. W. GREENE; P. G. M. WUTS: "Protecting Groups in Organic Synthesis", 1991, WILEY
XIE, L. ET AL., J. MED. CHEM., vol. 42, 1999, pages 2662
BRAND ET AL., DEVELOPMENT, vol. 118, no. 2, 1993, pages 401 - 415
IWAKI; FIGUERA ET AL.: "Rapid selection of Drosophila S2 cells with the puromycin resistance gene", BIOTECHNIQUES, vol. 35, no. 3, 2003, pages 482 - 484,486
SCHNEIDER, I., JEMBRYOL EXPMORPHOL, vol. 27, 1972, pages 353 - 65
ARMKNECHT, S. ET AL., METHODS ENZYMOL, vol. 392, 2005, pages 55 - 73
KING-JONES, K.; THUMMEL, C. S, NAT REV GENET,, vol. 6, 2005, pages 311 - 23
STEHLIN, C. ET AL., EMBO J, vol. 20, 2001, pages 5822 - 31
WHITE, K. P.; HURBAN, P.; WATANABE, T.; HOGNESS, D. S., SCIENCE, vol. 276, 1997, pages 114 - 17
REINKING, J. ET AL., CELL, vol. 122, 2005, pages 195 - 207
INGLESE ET AL., PROC. NATL. ACAD. SCI., vol. 103, 2006, pages 11473 - 8
WANG, Y. ET AL., CURRENT CHEMICAL GENOMICS, 2010, pages 57 - 66
See also references of EP 2558087A4
Attorney, Agent or Firm:
FASHENA, Sarah, J. (411 Hackensack AvenueHackensack, NJ, US)
Download PDF:
Claims:
WHAT IS CLAIMED IS:

1. A method for preventing, treating or ameliorating in a mammal a disease or condition that is causally related to RORyt activity in vivo, which comprises administering to the mammal an effective disease-treating or condition-treating amount of a compound according to formula I:

I

wherein

each nl and n2 is independently 1 , 2, 3, 4 or 5;

each Rla and Rlb is independently H, substituted or unsubstituted Ci-C6 alkyl, or CN; or

Rl a and Rlb joined together to form cycloalkyl ring;

R2 is H, substituted or unsubstituted Ci-C6 alkyl, or aryl;

or one of Rla and Rlb is joined to the C of CR2 to form a cyclopropyl ring; or R2 is joined to the C of CRlaRlb to form a cyclopropyl ring;

each R3 and R4 is independently selected from H, OH, substituted or unsubstituted alkyl, substituted or unsubstituted alkoxy, substituted or unsubstituted acyl, substituted or unsubstituted acylamino, substituted or unsubstituted alkylamino, substituted or unsubstituted alkythio, substituted or unsubstituted alkoxycarbonyl, substituted or unsubstituted alkylarylamino, substituted or unsubstituted amino, substituted or unsubstituted arylalkyl, sulfo, substituted sulfo, substituted sulfonyl, substituted sulfinyl, substituted sulfanyl, substituted or unsubstituted aminosulfonyl, substituted or unsubstituted alkylsulfonyl, substituted or unsubstituted arylsulfonyl, azido, substituted or unsubstituted carbamoyl, carboxyl, cyano, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted dialkylamino, halo, nitro, and thiol; or any two adjacent R3 groups, or any two adjacent R4 groups may joined together to form a substituted or unsubstituted carbocyclic or heterocyclic ring; each R3 and R is independently selected from H, substituted or unsubstituted alkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, substituted or unsubstituted aralkyl, and substituted or unsubstituted heteroarylalkyl; or

R5 and R6, together with the N they are attached to, form a 4- 12 membered substituted or unsubstituted heterocycloalkyl;

or a pharmaceutically acceptable salt, solvate or prodrug thereof;

and stereoisomers, isotopic variants and tautomers thereof.

2. The method according to claim 1 , wherein each R5 and R6 is independently selected from H, substituted or unsubstituted alkyl, substituted and unsubstituted cycloalkyl, and substituted and unsubstituted phenyl.

3. The method according to claim 1 , wherein one of R5 and R6 is H or Me; and the other is independently selected from H, substituted or unsubstituted alkyl, substituted and unsubstituted cycloalkyl, and substituted and unsubstituted phenyl.

4. The method according to claim 1 , wherein each R5 and R6 is independently selected from substituted or unsubstituted alkyl.

5. The method according to claim 1 , wherein each R5 and R6 is independently selected from unsubstituted alkyl.

6. The method according to claim 1 , wherein each R5 and R6 is independently selected from H, Me, Et, n-Pr, i-Pr, n-Bu, and t-Bu.

7. The method according to claim 1 , wherein each R5 and R6 is independently selected from H, and alkyl substituted with hydroxyl, amino, alkylamino, dialkylamino, cylcloalkyl, heterocycloalkyl, aryl, and heteroaryl.

8. The method according to claim 1, wherein each R5 and R6 is independently selected from H, Me, Et, hydroxyethyl, hydroxypropyl, aminoethyl, aminopropyl, dimethylaminoethyl, dimethylaminopropyl, piperidinoethyl, morpholinoethyl, cyclopropylmethyl, benzyl, phenethyl, and furanyl methyl.

9. The method according to claim 1 , wherein each R5 and R6 is independently selected from H, and cycloalkyl.

10. The method according to claim 1 , wherein each R5 and R6 is independently selected from H, and cyclopropyl, cyclobutyl, cyclopentyl, and cyclohexyl.

1 1. The method according to claim 1 , wherein R5 is H; and R6 is selected from

hydroxyethyl, hydroxypropyl, aminoethyl, aminopropyl, dimethylaminoethyl, dimethylaminopropyl, piperidinoethyl, morpholinoethyl, cyclopropylmethyl, cyclohexyl, benzyl, phenethyl, and furanylmethyl.

12. The method according to claim 1 , wherein R and R are joined together to form a 4-

12 membered substituted or unsubstituted heterocycloalkyl.

13. The method according to claim 1 , wherein the compound is according to formula la:

la

wherein

Cy is substituted or unsubstituted 4- 12 membered substituted or unsubstituted heterocycloalkyl;

and wherein nl , n2, Rla, Rlb, R2, R3, and R4 are as in claim 1 ;

or a pharmaceutically acceptable salt, solvate or prodrug thereof;

and stereoisomers, isotopic variants and tautomers thereof.

14. The method according to Claim 13, wherein Cy is azetidinyl, pyrrolidinyl,

piperidinyl, piperizinyl, morpholinyl, tetrahydroquinolinyl, indolinyl, or azepinyl, unsubstituted or substituted with alkyl, substituted alkyl, haloalkyl, alkoxyalkyl, hydroxyalkyl,alkoxy, hydroxyl, CN, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, amido, acyl, aroyl, or -CO-alkoxy.

15. The method according to Claim 1 , wherein the compound is according to formula Ila, lib, lie, Hd, He, or Ilf:

Me or ||f

and wherein n l , n2, Rla, Rlb, R2, R3, R4, R5, and R6 are as in claim 1 ; and each R5a and R5b is independently H, phenyl, or substituted or unsubstituted Ci-C6 alkyl.

16. The method according to any one of claims 1 -15, wherein n2 is 1 , 2, or 3.

17. The method according to any one of claims 1 -15, wherein n2 is 1 , 2, or 3; and each R4 is independenly selected from halo, Ci-C6 alkyl, CN, OH, and Ci-C6 alkoxy.

18. The method according to any one of claims 1 -15, wherein n2 is 1 , 2, or 3; and each R4 is independenly selected from CI, Me, OH, and Ci-C6 alkoxy.

19. The method according to Claim 1 , wherein the compound is according to formula III:

III

wherein

and wherein nl , Rla, Rlb, R2, R3, and R4, are as in claim 1 ; n2 is 1 , 2, or 3;

A is C, N, O, or S; m is 0 or 1 ;

each R5a is alkyl, substituted alkyl, halo, haloalkyl, hydroxyalkyl, aryl, heteroaryl, CN, alkoxyalkyl, amido, hydroxyl, alkoxy or substitituted alkoxy; and t is 0, 1 , 2, or 3;

R7a is OH, substituted or unsubstituted alkyl, substituted or unsubstituted alkoxy, substituted or unsubstituted acyl, substituted or unsubstituted acylamino, substituted or unsubstituted alkylamino, substituted or unsubstituted alkythio, substituted or unsubstituted alkoxycarbonyl, substituted or unsubstituted alkylarylamino, substituted or unsubstituted amino, substituted or unsubstituted arylalkyl, sulfo, substituted sulfo, substituted sulfonyl, substituted sulfinyl, substituted sulfanyl, substituted or unsubstituted aminosulfonyl, substituted or unsubstituted alkylsulfonyl, substituted or unsubstituted arylsulfonyl, azido, substituted or unsubstituted carbamoyl, carboxyl, cyano, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted dialkylamino, halo, nitro, and thio; and

R7b is R4; or any two adjacent R4 and R7b groups, may joined together to form a substituted or unsubstituted carbocyclic or heterocyclic ring;

or a pharmaceutically acceptable salt, solvate or prodrug thereof;

and stereoisomers, isotopic variants and tautomers thereof.

20. A compound according to formula III:

111

wherein

A is C, N, O, or S; m is 0 or 1 ;

nl is 1 , 2, 3, 4 or 5; n2 is 1 , 2, or 3;

each Rl a and Rlb is independently H, substituted or unsubstituted Ci-Ce alkyl, or CN; or

Rl a and Rl b joined together to form cycloalkyl ring;

R2 is H, substituted or unsubstituted C|-C6 alkyl, or aryl;

or one of Rla and Rl b is joined to the C of CR2 to form a cyclopropyl ring; or R2 is joined to the C of CRlaRlb to form a cyclopropyl ring;

each R3 and R4 is independently selected from H, OH, substituted or unsubstituted alkyl, substituted or unsubstituted alkoxy, substituted or unsubstituted acyl, substituted or unsubstituted acylamino, substituted or unsubstituted alkylamino, substituted or unsubstituted alkythio, substituted or unsubstituted alkoxycarbonyl, substituted or unsubstituted alkylarylamino, substituted or unsubstituted amino, substituted or unsubstituted arylalkyl, sulfo, substituted sulfo, substituted sulfonyl, substituted sulfinyl, substituted sulfanyl, substituted or unsubstituted aminosulfonyl, substituted or unsubstituted alkylsulfonyl, substituted or unsubstituted arylsulfonyl, azido, substituted or unsubstituted carbamoyl, carboxyl, cyano, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted dialkylamino, halo, nitro, and thiol; or any two adjacent R3 groups, any two adjacent R4 groups, or any two adjacent R4 and R7b groups, may joined together to form a substituted or unsubstituted carbocyclic or heterocyclic ring;

each R5a is alkyl, substituted alkyl, halo, haloalkyl, hydroxyalkyl, aryl, heteroaryl, CN, alkoxyalkyl, amido, hydroxyl, alkoxy or substitituted alkoxy; and t is 0, 1 , 2, or 3;

R7a is OH, substituted or unsubstituted alkyl, substituted or unsubstituted alkoxy, substituted or unsubstituted acyl, substituted or unsubstituted acylamino, substituted or unsubstituted alkylamino, substituted or unsubstituted alkythio, substituted or

unsubstituted alkoxycarbonyl, substituted or unsubstituted alkylarylamino, substituted or unsubstituted amino, substituted or unsubstituted arylalkyl, sulfo, substituted sulfo, substituted sulfonyl, substituted sulfinyl, substituted sulfanyl, substituted or unsubstituted aminosulfonyl, substituted or unsubstituted alkylsulfonyl, substituted or unsubstituted arylsulfonyl, azido, substituted or unsubstituted carbamoyl, carboxyl, cyano, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted dialkylamino, halo, nitro, and thio; and

R7b is H, OH, substituted or unsubstituted alkyl, substituted or unsubstituted alkoxy, substituted or unsubstituted acyl, substituted or unsubstituted acylamino, substituted or unsubstituted alkylamino, substituted or unsubstituted alkythio, substituted or

unsubstituted alkoxycarbonyl, substituted or unsubstituted alkylarylamino, substituted or unsubstituted amino, substituted or unsubstituted arylalkyl, sulfo, substituted sulfo, substituted sulfonyl, substituted sulfinyl, substituted sulfanyl, substituted or unsubstituted aminosulfonyl, substituted or unsubstituted alkylsulfonyl, substituted or unsubstituted arylsulfonyl, azido, substituted or unsubstituted carbamoyl, carboxyl, cyano, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted dialkylamino, halo, nitro, and thio;

or a pharmaceutically acceptable salt, solvate or prodrug thereof;

and stereoisomers, isotopic variants and tautomers thereof;

provided that

i) when t is 1 , m is 1 , A is C, each of Rla, Rlb and R2 is H, R4 is 4-OMe, and one of R7a and R7b is OH and the other is OMe; then R5 is other than 2- Me;

ii) when t is 1 , m is 1 , A is N, and each of Rl a, Rl b and R2 is H, R4 is OMe, one of R7a and R7b is OH and the other is OMe; then R5a is other than 4-Ph; iii) when each of Rl a, Rl b, R2 and R3 is H, one of R7a and R7b is OH, CI, Br, Me, or F, and the other is H; then t is other than 0, and R5a is other than benzyl, acetyl, cyclopentyl, Me, OH, CH2OMe, CH2OH, CH2CH2OMe, CH2CH2OH, OMe, C02Me, C02Et, or CONH2;

iv) when t is 0, each of Rla, Rlb and R2 is H, R4 is 4-OMe or 4-Me, and one of R7a and R7b is OH and the other is OMe or Me; then R3 is other than 4- OMe, 4-NMe2, or 3,4-methylenedioxy; the (R )ni-Ph- group is other than substituted or unsubstituted

benzopyranyl ;

when each of Rla, Rlb, R2 and R3 is H, one of R7a and R7b is OH, OMe, CI, Br, Me, or F, and the other is H; then one of (R3)ni-Ph- group is other than unsubstituted phenyl; and

when each of Rl a, Rlb, R2 and R3 is H, one of R7a and R7b is OMe, and the other is H; then one of (R3)n|-Ph- group is other than unsubstituted naphthyl.

21. The method or compound according to any one of Claims 19-20, wherein each of R a and R7b is independently OH, substituted or unsubstituted alkyl, substituted or unsubstituted alkoxy, substituted or unsubstituted acyl, substituted or unsubstituted acylamino, substituted or unsubstituted alkylamino, substituted or unsubstituted alkythio, substituted or unsubstituted alkoxycarbonyl, substituted or unsubstituted alkylarylamino, substituted or unsubstituted amino, substituted or unsubstituted arylalkyl, sulfo, substituted sulfo, substituted sulfonyl, substituted sulfinyl, substituted sulfanyl, substituted or unsubstituted aminosulfonyl, substituted or unsubstituted alkylsulfonyl, substituted or unsubstituted arylsulfonyl, azido, substituted or unsubstituted carbamoyl, carboxyl, cyano, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted

dialkylamino, halo, nitro, and thio.

22. The method or compound according to any one of Claims 19-20, wherein each of R7a and R7b is independently OH, substituted or unsubstituted alkyl, substituted or unsubstituted alkoxy, CN, halo, amido, or haloalkyl.

23. The method or compound according to any one of Claims 19-20, wherein each of R7a and R7b is independently halo, C| -C6 alkyl, CN, OH, or C)-C6 alkoxy.

24. The method or compound according to any one of Claims 19-20, wherein each of R7a and R7b is independently CI, F, Me, CF3, CN, OH, or OMe.

25. The method or compound according to any one of Claims 19-20, wherein one of R7a and R7b is OH; and the other is alkoxy.

26. The method or compound according to any one of Claims 19-20, wherein each of R7a and R7b is OH or alkoxy.

27. The method or compound according to any one of Claims 19-20, wherein one of R7a and R7b is OH; and the other is OMe.

28. The method or compound according to any one of Claims 1 -27, wherein R'd and R is independently H, CN, or Me.

29. The method or compound according to any one of Claims 1 -27, wherein each of Rla and Rlb is H or Me.

30. The method or compound according to any one of Claims 1 -27, wherein one of Rl a and Rlb is H; and the other is CN.

3 1. The method or compound according to any one of Claims 1 -27, wherein m is 0.

32. The method or compound according to any one of Claims 1 -27, wherein m is 1 ; and A is C or N.

33. The method or compound according to any one of Claims 19-32, wherein R4 is H, alkyl, substituted alkyl, halo, haloalkyl, hydroxyalkyl, alkoxyalkyl, amido, hydroxyl, cyano, or alkoxy.

34. The method or compound according to any one of Claims 19-32, wherein R4 is H, halo, C,-C6 alkyl, CN, OH, or CrC6 alkoxy.

35. The method or compound according to any one of Claims 19-32, wherein R4 is H, CI, F, Me, CF¾ CN, OH, or OMe.

36. The method or compound according to any one of Claims 19-35, wherein the

compound is according to formula IVa, IVb, IVc, IVd, IVe, or IVf:

and wherein nl , R2, R3, R5, and R6 are as in claim 1 ; and t and R5a are as in claims 19 or 20.

37. The method or compound according to any one of Claims 1 -36, wherein n l is 1 , 2 or 3.

38. The method or compound according to any one of Claims 1 -37, wherein each R3 is independently selected from halo, amino, substituted amino, substituted or unsubstituted CpCe alkyl, CN, OH, and substituted or unsubstituted C 1-C6 alkoxy.

39. The method or compound according to any one of Claims 1 -37, wherein each R3 is independently selected from halo, substituted or unsubstituted Ci-C6 alkyl, CN, OH, and substituted or unsubstituted C1-C6 alkoxy.

40. The method or compound according to any one of Claims 1 -37, wherein nl is 1 ; and R3 is NMe2.

41. The method or compound according to any one of Claims 1 -37, wherein nl is l ; and R3 is CI, F, Me, Et, i-Pr, OMe, CF3, CN or OH.

42. The method or compound according to any one of Claims 19-41 , wherein the compound is according to formula Va, Vb, Vc, Vd, Ve, or Vf:

Ve

and wherein R2, R5, and R6 are as in claim 1 ; t and R5a are as in claims 19 or 20.; and n3 is 1 , 2 or 3.

43. The method or compound according to any one of Claims 1 -42, wherein R2 is H, OH, substituted or unsubstituted Ci-C6 alkyl, or substituted or unsubstituted phenyl.

44. The method or compound according to any one of Claims 1 -42, wherein R2 is H, Me, OH, or Ph.

45. The method or compound according to any one of Claims 1 -42, wherein R2 is H.

46. The method or compound according to any one of Claims 1 -42, wherein R2 is OH.

47. The method or compound according to any one of Claims 19-46, wherein t is 0.

48. The method or compound according to any one of Claims 19-46, wherein t is 1 or 2; and each R5a is independently OH, Ph, benzyl, or Me.

49. The method or compound according to any one of Claims 19-46, wherein t is 1 ; and

' R5a is Me, Et, n-Pr, or n-Bu.

50. The method or compound according to any one of Claims 19-46, wherein t is 1 ; and R5a is 3-Me, 3-Et, 3-n-Pr, or 3-n-Bu.

51. The method or compound according to any one of Claims 19-46, wherein t is 2; and one R a is 3-Me and the other is 5-Me.

52. The method according to any one of Claims 19-51 , wherein R5 is Me, or Et.

53. The method according to any one of Claims 19-52, wherein R6 is Me, Et, n-Pr, n-Bu, n-pentyl, n-hexyl, or n-heptyl.

54. The method according to claim 1 or the compound according to Claim 20, wherein the compound is according to formula Via, VIb, Vic, VId, Vie, or Vlf:

and wherein R is H or Me;

or a pharmaceutically acceptable salt, solvate or prodrug thereof;

and stereoisomers, isotopic variants and tautomers thereof.

The method according to claim 1 or the compound according to Claim 20, wherein the compound is according to formula Vila, Vllb, or VIIc:

Vila Vllb Vile or a pharmaceutically acceptable salt, solvate or prodrug thereof;

and stereoisomers, isotopic variants and tautomers thereof.

The method according to claim 1 or the compound according to Claim 20, wherein the compoun is according to formula Villa, Vlllb, VIIlc, Vllld, or Vllle:

Villa Vlllb

VIIlc Vllld or Vl l |e

or a pharmaceutically acceptable salt, solvate or prodrug thereof;

and stereoisomers, isotopic variants and tautomers thereof.

57. The method according to claim 1 or the compound according to Claim 20, wherein the compound is according to formula IXa, IXb, or IXc:

IXa IXb IXc

or a pharmaceutically acceptable salt, solvate or prodrug thereof;

and stereoisomers, isotopic variants and tautomers thereof.

58. The method according to Claim 1 or the compound accound according to claim 20, wherein the compound is any one of compounds listed in Table 1.

59. A compound selected from compounds listed in Table 1.

60. The method according to Claim 1 wherein the compound is any one of compounds listed in Table 1 with Compound ID 7, 8, 22, 41 , 62, 63, 65, 70, 72, 73, 74, 75, 76, 1 18, 121 , 134, 135, 136, 138, or 149.

61 . A compound selected from compounds listed in Table 1 , wherein compound ID is 7, 8, 22, 41 , 62, 63, 65, 70, 72, 73, 74, 75, 76, 1 18, 121 , 134, 135, 136, 138, or 149.

62. A pharmaceutical composition of compound according to any one of claims 1 -61.

63. The method of Claim 1 , wherein the disease or condition is autoimmune disease.

64. The method of Claim 1 , wherein the disease or condition is inflammatory disease.

65. The method of Claim 1 , wherein the disease or condition is selected from arthritis, diabetes, multiple sclerosis, uveitis, rheumatoid arthritis, psoriasis, asthma, bronchitis, allergic rhinitis, chronic obstructive pulmonary disease, atherosclerosis, H. pylori infections and ulcers resulting from such infection, and inflammatory bowel diseases.

66. The method of Claim 1 , wherein the disease or condition is selected from Crohn's disease, ulcerative colitis, sprue and food allergies.

67. The method of Claim 1 , wherein the disease or condition is selected from multiple sclerosis and experimental autoimmune encephalomyelitis (EAE).

68. The method of Claim 1 , wherein the disease or condition is selected from rheumatoid arthritis or collagen-induced arthritis (CIA).

Description:
AMIDO COMPOUNDS AS RORyt MODULATORS AND USES THEREOF

FIELD OF THE INVENTION

[0001] This invention relates to amido compounds capable of modulating RORyt activity and uses of such compounds to treat diseases or conditions related to RORyt activity. More particularly, the amido compounds may be used to diminish inflammation associated with an inflammatory disease or condition or to reduce symptoms associated with an autoimmune disorder. Also encompassed herein, are compositions of amido compounds, pharmaceutical compositions of amido compounds, assays and methods for using same to identify compounds capable of modulating RORyt activity.

BACKGROUND OF THE INVENTION

[0002] The retinoic acid receptor-related orphan nuclear receptor (ROR) RORy and its isoform RORyt (collectively "RORy/yt") play a major role in regulation of a variety of biological systems. To illustrate, RORyt has a central role in immune system development, homeostasis, and responses to microbial pathogens. For example, RORyt is required for the differentiation of Thl 7 cells (Ivanov, II et al. Cell, 2006, 126, 1 121 -33), a subset of T helper cells that protect the host from infection by secreting inflammatory cytokines such as IL- 17, IL- 17 (also called IL- 17A), IL- 17F, IL-22, and TNFa. These cytokines are signaling proteins that have been shown to be essential in regulating numerous immune responses, including inflammatory responses to antigens. Th l 7 cells have also recently been shown to have important roles in activating and directing immune responses in a variety of autoimmune diseases, such as experimental autoimmune encephalomyelitis (EAE), collagen-induced arthritis (CIA), inflammatory bowel di sease (IBD), cancer (Weaver, C. et al. Ann. Rev. Immunol., 2007, 25, 821 -52; Kryczek, I. et al. J. Immunol., 2007, 178, 6730-3; Cua, D. J. et al. Nature, 2003, 421, 744-8; Langrish, C. L. et al. J. Exp. Med., 2005, 201, 233-40; Yen, D. et al. J. Clin. Invest., 2006, 116, 13 10-6), and graft- versus-host disease (Carlson, M.J. et al. Blood, 28 October 2008. [Epub ahead of print]; Kappel, L.W. et al. Blood, 17 October 2008. [Epub ahead of print]). Th l 7 cells have also been implicated in asthma, psoriasis, rheumatoid arthritis, multiple sclerosis (Tzartos, J.S., et al. Am. J.

Pathology, 2008, 172, 146-55; Yu, J.J., and Gaffen, S.L. Front. Bioscl, 2008, 13, 170-77; and Zheng, Y. et al. Nature, 2007, 445, 648-51), and Crohn's disease (Duerr, R.H., et al. Science, 2006, 314, 1461 -63). Additionally, it has been shown that mice defective for expression of RORyt lack Th l cells and are resistant to a variety of autoimmune diseases and that the absence of Th l 7-producing microbiota in the small intestine of mice alters the Thl 7: regulatory T (Treg) cell balance with implications for intestinal immunity, tolerance, and susceptibility to

inflammatory bowel diseases (Ivanov, I.I. Cell Host & Microbe, 2008, 4, 337-49).

[0003] The formation of immune cell aggregates, such as cryptopatches (CP) and isolated lymphoid follicles (ILF), which contain RORyt expressing cells, is known to be a vital step in many immune responses. For example, CPs and ILFs are required for mucosal immunity and for production of the intestinal antibody IgA. Such immune responses can result in inflammation in various diseases, such as Crohn's disease. The ability to inhibit such immune responses by inhibiting the formation of immune cell aggregates may offer another way to treat diseases associated with such responses.

[0004] T-cells have also been demonstrated to play a role in diseases characterized by bone loss and degradation, such as osteoarthritis. For example, in autoimmune arthritis, activation of T cells results in bone destruction mediated by osteoclasts. Th l 7, whose differentiation is regulated by RORyt, has been shown to be osteoclastogenic, thus linking T cell activation and bone resorption (Sato, K. et al. J. Ex. Med., 2008, 203, 2673-82). Thus, the ability to regulate Th 17 cell differentiation via RORyt modulation may offer a way to treat bone loss and degradation, such as that associated with autoimmune disease. Furthermore, interferon gamma (IFN-γ) suppresses osteoclast formation by rapidly degrading the RANK adaptor protein TRAF6 in the RANK-RANKL signaling pathway, and RORyt has been shown to down-regulate the production of IFN-y (Ivanov, I.I. et al. Cell, 2006, 126, 1 121 -33). Thus, the ability to regulate osteoclast formation through modulation of RORyt-mediated IFN-y osteoclast suppression may provide additional methods to treat bone loss and degradation, such as that associated with autoimmune disease (e.g., osteoarthritis).

[0005] Circadian rhythm relates to an approximately daily periodicity in the biochemical, physiological or behavioral processes of living beings, including plants, animals, fungi and some bacteria. Members of the ROR family of orphan nuclear receptors have been implicated in regulation of control of circadian clock function by regulation of clock genes (Ueda, H. R. et al. Nature, 2002, 418, 534-39; Sato, T. K. et al. Neuron, 2004, 43, 527-37), and RORy/yt has been implicated in the regulation of genes that govern circadian metabolism (Kumaki, Y. et al. PNAS, 2008, 705, 14946-51 ; Liu, C. et al. Nature, 2007, 447, 477-81 ). Moreover, RORy gene expression is known to oscillate in a circadian manner in metabolically active tissues such as liver and brown adipose tissue (Yang, X. et al., Cell, 2006, 126, 801 - 10), which further confirms that a role exists for RORy in regulating circadian function. Hence, the ability to modulate RORy/yt expression may also result in circadian rhythm regulation and treatment of disorders associated with disruption of circadian rhythm. Since circadian rhythm is integral in maintaining metabolic levels, whose imbalance is linked to obesity and diabetes, modulators of RORy/yt may also be useful in treating obesity and diabetes through regulation of circadian rhythm.

[0006] In view of the above, a need exists for therapeutic agents, and corresponding pharmaceutical compositions and related methods of treatment that address conditions causally related to RORyt activity, and it is toward the fulfillment and satisfaction of that need, that the present invention is directed.

SUMMARY OF THE INVENTION

[0007] The present invention provides a method for preventing, treating or ameliorating in a mammal a disease or condition that is causally related to RORy or RORyt activity in vivo, which comprises administering to the mammal an effective disease-treating or condition-treating amount of a compound according to formula I:

I

wherein

each n l and n2 is independently 1 , 2, 3, 4 or 5;

each R la and R l b is independently H, substituted or unsubstituted Ci-C 6 alkyl, or CN; or

R la and R lb joined together to form cycloalkyl ring;

R 2 is H, substituted or unsubstituted Ci-C 6 alkyl, or aryl;

or one of R la and R l b is joined to the C of CR 2 to form a cyclopropyl ring; or R 2 is joined to the C of CR la R l b to form a cyclopropyl ring;

each R 3 and R 4 is independently selected from H, OH, substituted or unsubstituted alkyl, substituted or unsubstituted alkoxy, substituted or unsubstituted acyl, substituted or unsubstituted acylamino, substituted or unsubstituted alkylamino, substituted or unsubstituted alkythio, substituted or unsubstituted alkoxycarbonyl, substituted or unsubstituted alkylarylamino, substituted or unsubstituted amino, substituted or unsubstituted arylalkyl, sulfo, substituted sulfo, substituted sulfonyl, substituted sulfinyl, substituted sulfanyl, substituted or unsubstituted aminosulfonyl, substituted or unsubstituted alkylsulfonyl, substituted or unsubstituted arylsulfonyl, azido, substituted or unsubstituted carbamoyl, carboxyl, cyano, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted dialkylamino, halo, nitro, and thiol; or any two adjacent R 3 groups, or any two adjacent R 4 groups may joined together to form a substituted or unsubstituted carbocyclic or heterocyclic ring;

each R 5 and R 6 is independently selected from H, substituted or unsubstituted alkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, substituted or unsubstituted aralkyl, and substituted or unsubstituted heteroarylalkyl; or

R 5 and R 6 , together with the N they are attached to, form a 4-12 membered substituted or unsubstituted heterocycloalkyl;

or a pharmaceutically acceptable salt, solvate or prodrug thereof;

and stereoisomers, isotopic variants and tautomers thereof.

[0008] In one embodiment, with respect to the compounds of formula I, the compound is according to formula la:

la

wherein

Cy is substituted or unsubstituted 4-12 membered substituted or unsubstituted heterocycloalkyl;

and wherein nl , n2, R l a , R lb , R 2 , R 3 , and R 4 are as described for formula I;

or a pharmaceutically acceptable salt, solvate or prodrug thereof;

and stereoisomers, isotopic variants and tautomers thereof.

[0009] In one embodiment, with respect to the compounds of formula la, Cy is substituted or unsubstituted azetidinyl, pyrrolidinyl, piperidinyl, piperizinyl, morpholinyl, tetrahydroquinolinyl, indolinyl, or azepinyl. In another embodiment, Cy is unsubstituted pyrrolidinyl, piperidinyl, piperizinyl, morpholinyl, or azepinyl. In a particular embodiment, Cy is piperidinyl or piperizinyl. [0010] . In a further aspect, the present invention provides a compound according to formula III:

III

wherein

A is C, N, O, or S; m is 0 or 1 ;

nl is 1 , 2, 3, 4 or 5; n2 is 1 , 2, or 3;

each R la and R lb is independently H, substituted or unsubstituted Ci-C 6 alkyl, or CN; or

R la and R lb joined together to form cycloalkyl ring;

R 2 is H, substituted or unsubstituted C1 -C6 alkyl, or aryl;

or one of R la and R l b is joined to the C of CR 2 to form a cyclopropyl ring; or R 2 is joined to the C of CR l a R lb to form a cyclopropyl ring;

each R 3 and R 4 is independently selected from H, OH, substituted or unsubstituted alkyl, substituted or unsubstituted alkoxy, substituted or unsubstituted acyl, substituted or unsubstituted acylamino, substituted or unsubstituted alkylamino, substituted or unsubstituted alkythio, substituted or unsubstituted alkoxycarbonyl, substituted or unsubstituted alkylarylamino, substituted or unsubstituted amino, substituted or unsubstituted arylalkyl, sulfo, substituted sulfo, substituted sulfonyl, substituted sulfinyl, substituted sulfanyl, substituted or unsubstituted aminosulfonyl, substituted or unsubstituted alkylsulfonyl, substituted or unsubstituted arylsulfonyl, azido, substituted or unsubstituted carbamoyl, carboxyl, cyano, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted dialkylamino, halo, nitro, and thiol; or any two adjacent R 3 groups, or any two adjacent R 4 groups, or any two adjacent R 4 and R 7b groups, may joined together to form a substituted or unsubstituted carbocyclic or heterocyclic ring;

each R 5a is alkyl, substituted alkyl, halo, haloalkyl, hydroxyalkyl, aryl, heteroaryl, CN, alkoxyalkyl, amido, hydroxyl, alkoxy or substitituted alkoxy; and t is 0, 1 , 2, or 3; R a .is OH, substituted or unsubstituted alkyl, substituted or unsubstituted alkoxy, substituted or unsubstituted acyl, substituted or unsubstituted acylamino, substituted or unsubstituted alkylamino, substituted or unsubstituted alkythio, substituted or

unsubstituted alkoxycarbonyl, substituted or unsubstituted alkylarylamino, substituted or unsubstituted amino, substituted or unsubstituted arylalkyl, sulfo, substituted sulfo, substituted sulfonyl, substituted sulfinyl, substituted sulfanyl, substituted or unsubstituted aminosulfonyl, substituted or unsubstituted alkylsulfonyl, substituted or unsubstituted arylsulfonyl, azido, substituted or unsubstituted carbamoyl, carboxyl, cyano, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted dialkylamino, halo, nitro, and thio; and

R 7b is H, OH, substituted or unsubstituted alkyl, substituted or unsubstituted alkoxy, substituted or unsubstituted acyl, substituted or unsubstituted acylamino, substituted or unsubstituted alkylamino, substituted or unsubstituted alkythio, substituted or

unsubstituted alkoxycarbonyl, substituted or unsubstituted alkylarylamino, substituted or unsubstituted amino, substituted or unsubstituted arylalkyl, sulfo, substituted sulfo, substituted sulfonyl, substituted sulfinyl, substituted sulfanyl, substituted or unsubstituted aminosulfonyl, substituted or unsubstituted alkylsulfonyl, substituted or unsubstituted arylsulfonyl, azido, substituted or unsubstituted carbamoyl, carboxyl, cyano, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted dialkylamino, halo, nitro, and thio; '

or a pharmaceutically acceptable salt, solvate or prodrug thereof;

and stereoisomers, isotopic variants and tautomers thereof;

provided that

i) when t is 1 , m is 1 , A is C, each of R la , R lb and R 2 is H, R 4 is 4-OMe, and one of R 7a and R 7b is OH and the other is OMe; then R 5a is other than 2- Me;

ii) when t is 1, m is 1 , A is N, and each of R la , R l b and R 2 is H, R 4 is OMe, one of R 7a and R 7b is OH and the other is OMe; then R 5a is other than 4-Ph; iii) when each of R la , R l b , R 2 and R 3 is H, one of R 7a and R 7b is OH, CI, Br, Me, or F, and the other is H; then t is other than 0, and R 5a is other than benzyl, acetyl, cyclopentyl, Me, OH, CH 2 OMe, CH 2 OH, CH 2 CH 2 OMe, CH 2 CH 2 OH, OMe, C0 2 Me, C0 2 Et, or CONH 2 ; .. iv) . when t is 0, each of R l a , R lb and R 2 is H, R 4 is 4-OMe or 4-Me, and one of

R 7a and R 7b is OH and the other is OMe or Me; then R 3 is other than 4-

OMe, 4-NMe 2 , or 3,4-methylenedioxy;

v) the (R 3 ) n i-Ph- group is other than substituted or unsubstituted benzopyranyl ;

vi) when each of R la , R lb , R 2 and R 3 is H, one of R 7a and R 7b is OH, OMe, CI, Br, Me, or F, and the other is H; then one of (R 3 ) n i-Ph- group is other than unsubstituted phenyl; and

vii) when each of R la , R lb , R 2 and R 3 is H, one of R 7a and R 7b is OMe, and the other is H; then one of (R 3 ) n i-Ph- group is other than unsubstituted naphthyl.

[0011] In one particular embodiment, with respect to the compound of formula III, R 7b is other than H.

[0012] In one particular embodiment, with respect to the compound of formula III, when m is 1 , A is N; then the N can be substitituted or unsubstituted. In one embodiment A is NH. In another embodiment, A is N-R 5a and R 5a is as described herein.

[0013] In a further aspect, the present invention provides pharmaceutical compositions comprising a heterocyclic compound of the invention, and a pharmaceutical carrier, excipient or diluent. In this aspect of the invention, the pharmaceutical composition can comprise one or more of the compounds described herein. Moreover, the compounds of the present invention useful in the pharmaceutical compositions and treatment methods disclosed herein, are all pharmaceutically acceptable as prepared and used.

[0014] In a further aspect, this invention provides a method of treating a mammal susceptible to or afflicted with a condition from among those listed herein, and particularly, such condition as may be associated with RORyt. Such conditions include, without limitation, multiple sclerosis (and the animal model thereof, EAE), rheumatoid arthritis (and the animal model thereof, CIA), inflammatory bowel disease (IBD), cancer, graft-versus-host disease, asthma, psoriasis, diabetes, uveitis, bronchitis, allergic rhinitis, chronic obstructive pulmonary disease, arteriosclerosis, and H. pylori infections and ulcers resulting from such infection.

[0015] In a further aspect, an assay for screening to identify modulators of RORy/yt transcriptional activity is envisioned, the assay comprising a first insect cell line that expresses a fusion protein (SEQ ID NO: 2; encoded by SEQ ID NO: 1 ) comprising a RORy/yt sequence, wherein the RORy/yt sequence does not comprise the DNA binding domain (DBD) of full length RORy/yt and a yeast GAL4 DBD, wherein expression of the fusion protein is transcriptionally regulated by an inducible promoter, and wherein the first insect cell line further comprises a reporter, whose expression is upregulated in the presence of the fusion protein. Accordingly, the component of the fusion protein representative of RORy/yt sequences is a DBD-deleted RORy/yt sequence. The fusion protein is essentially a chimeric protein wherein the RORy/yt DBD is deleted from the RORy/yt sequences and replaced with the yeast GAL4 DNA binding domain

DBD. In a particular embodiment, the Gal4 DNA binding domain (G4DBD) corresponds to amino acids 1 through 147 of Gal4 protein and the DBD-deleted RORy/yt sequence is amino acids 79 to the carboxyl terminal end. Accordingly, the G4DBD corresponds to amino acids 1 -

147 of SEQ ID NO: 2 and the DBD-deleted RORy/yt sequence corresponds to amino acids 148-

564 of SEQ ID NO: 2. In a particular embodiment, the inducible promoter is a copper inducible promoter.

[0016] In an embodiment of the assay, the reporter is transcriptionally regulated by a plurality of copies of the GAL4 binding site enhancer (UAS) operatively linked to nucleic acid sequences encoding the reporter. In a particular embodiment of the assay, the plurality of copies of the GAL4 binding site enhancer (UAS) is between 1 and 5 copies. In a more particular embodiment, the reporter is the firefly luciferase reporter.

[0017] In an aspect of the assay, the first insect cell line is the S2 cell line. In another aspect of the assay, the fusion protein is encoded by nucleic acids that are integrated into the genome of the first insect cell line or encoded by extrachromosomal nucleic acids incorporated into the first insect cell line. Accordingly, the assay may relate to stably transfected cell line or a transiently transfected cell line. The choice of stably or transiently transfected cell line depends, in part, on the number of compounds to be tested and availability and cost of assay reagents required.

[0018] As described herein, the assay may further comprise a second insect cell line that expresses a second fusion protein (SEQ ID NO: 4; encoded by SEQ ID NO: 3) comprising a RORa sequence, wherein the RORa sequence does not comprise the DBD of full length RORa sequence and a yeast GAL4 DBD; a third insect cell line that expresses a third fusion protein (SEQ ID NO: 6; encoded by SEQ ID NO: 5) comprising a DHR3 sequence, wherein the DHR3 sequence does not comprise the DNA binding domain (DBD) of full length DHR3 sequence and a yeast GAL4 DBD, and a fourth insect cell line that expresses a fourth fusion protein (SEQ ID NO: 8; encoded by SEQ ID NO: 7) comprising a transcriptionally active domain of general transcriptional activator VP 16 and a yeast GAL4 DBD, wherein expression of the second, third and fourth fusion proteins is transcriptionally regulated by inducible promoters. In a particular embodiment, the DBD-deleted mouse RORa sequence is amino acids 142 to the carboxyl terminal end of mouse RORa and the DBD-deleted Drosophila DHR3 is amino acids 120 to the carboxyl terminal end of Drosophila DHR3. Accordingly, the G4DBD corresponds to amino acids 1 - 147 of SEQ ID NOs: 4, 6, and 8 and the DBD-deleted RORa sequence corresponds to amino acids 148-529 of SEQ ID NO: 4, the DBD-deleted DHR3 sequence corresponds to amino acids 148-513 of SEQ ID NO: 6, and the VP 16 sequence corresponds to amino acids 148-231 of

SEQ ID NO: 8. In an embodiment, inducible promoters regulating expression of the first, second, third, and fourth fusion proteins are identical promoters. In yet another embodiment, the inducible promoters regulating expression of the first, second, third, and fourth fusion proteins are copper inducible promoters.

[0019] Methods for using the assay of the invention are also encompassed herein. Such methods include those involving use of the cell-based assay systems described herein to screen diverse compound libraries, such as those available from research institutions, federal agencies, and/or commercial vendors, to identify modulators of RORy/yt transcriptional activity.

Modulators of RORy/yt transcriptional activity may be identified based on results determined using the first insect cell-based assay system described herein alone or in combination with at least one of the second, third, and fourth insect cell-based assay systems described herein to identify compounds that are specific modulators of RORy/yt transcriptional activity.

Modulators identified using assays described herein may be identified as inhibitors or agonists of RORy/yt transcriptional activity. Compounds identified using the cell based systems described herein may be assessed in secondary screens, also described herein and understood in the art, to validate their identity as bona fide modulators of RORy/yt transcriptional activity.

[0020] Other objects and advantages will become apparent to those skilled in the art from a consideration of the ensuing detailed description.

BRIEF DESCRIPTION OF THE DRAWINGS

[0021] Figure 1 shows the structures of RORy specific compounds with inverse agonist or antagonist activity.

[0022] Figure 2 illustrates the RORy specific inhibitory activity of the compounds shown therein and the structures of related compounds that fail to inhibit RORy activity.

[0023] Figure 3 shows a fluorescence activated cell sorter (FACS) plot analysis revealing the effects of RORy inhibitory compounds on mouse Thl 7 differentiation.

[0024] Figure 4 shows a FACS plot analysis revealing that RORy inhibitory compounds do not inhibit mouse Thl differentiation.

[0025] Figure 5 shows a FACS plot analysis revealing that RORy inhibitory compounds

166547 and 166488 inhibit RORy overexpression dependent, but not RORa dependent, IL17a production in human CD4 T cells.

[0026] Figure 6A and B show a FACs plot showing that (A) Compound NCGC00238427 selectively inhibits RORy. Flow cytometry of IL-17a and IFN-y production by cord blood naive CD4" T cells (CD45ROXD45RA + CD3 + CD4 + CD25 " HLA-DR " ) transduced with RORad-IRES-

GEP or RORyt-IRES-GFP on day 1 and analyzed on day 6. GFP expressing cells were gated for analysis. DMSO or N2 (3 μΜ) was added 6-8h after viral transduction; and (B) Compound

NCGC00238427 inhibits human Thl 7 cell differentiation at as low as 1 μΜ. Flow cytometry of

IL- 17a and IFN-γ production by human naive cord blood T cells (CD45RCT

D45RA + CD3 + CD4 + CD25 " HLA-DR " ), which were cultured for six days in presence of IL2, IL23 and IL1 β, and with various concentrations of TGFp (ng/ml). DMSO or N2 was added 16 hours after the cytokine addition.

[0027] Figure 7A and B depict the (A) the structure of NCGC00238427 and (B) plots demonstrating that RORyt activity is important for maintenance of human Thl 7 cells. Human memory (CD45RO + CD45RA " CD3 + CD4 + CCR6 + CD161 + ) cells were purified from healthy donor peripheral blood samples and were cultured in the presence of IL- Ι β, IL-23 and IL-2 for 6 days with or without NCGC00238427 (3 μΜ). Intracellular staining for IFN-γ or IL-17a in memory CD4 + T cells from multiple donors, assessed on day 6. Each symbol (n=l 1 or 9, respectively) indicates a separate donor. Statistical analysis was by a two-tailed unpaired Student's t test; IL- 17a ~ IFN-y + , not significant and IL- 17a + IFN-y +/_ , p=0.02.

DETAILED DESCRIPTION OF THE INVENTION

Definitions

[0028] When describing the compounds, pharmaceutical compositions containing such compounds and methods of using such compounds and compositions, the following terms have the following meanings unless otherwise indicated. It should also be understood that any of the moieties defined forth below may be substituted with a variety of substituents, and that the respective definitions are intended to include such substituted moieties within their scope. It should be further understood that the terms "groups" and "radicals" can be considered interchangeable when used herein.

[0029] "Acyl" refers to a radical -C(0)R 20 , where R 20 is hydrogen, alkyl, cycloalkyl, cycloheteroalkyl, aryl, arylalkyl, heteroalkyl, heteroaryl, heteroarylalkyl as defined herein. Representative examples include, but are not limited to, formyl, acetyl, cyclohexylcarbonyl, cyclohexylmethylcarbonyl, benzoyl, benzylcarbonyl and the like.

[0030] "Acylamino" refers to a radical -NR 21 C(0)R 22 , where R 21 is hydrogen, alkyl, cycloalkyl, cycloheteroalkyl, aryl, arylalkyl, heteroalkyl, heteroaryl, heteroarylalkyl and R 22 is hydrogen, alkyl, alkoxy, cycloalkyl, cycloheteroalkyl, aryl, arylalkyl, heteroalkyl, heteroaryl or heteroarylalkyl, as defined herein. Representative examples include, but are not limited to, formylamino, acetylamino, cyclohexylcarbonylamino, cyclohexylmethyl-carbonylamino, benzoylamino, benzylcarbonylamino and the like. [0031] . "Acyloxy" refers to the group -OC(0)R where R is hydrogen, alkyl, aryl or cycloalkyl.

(0032] "Substituted alkenyl" includes those groups recited in the definition of

"substituted" herein, and particularly refers to an alkenyl group having 1 or more substituents, for instance from 1 to 5 substituents, and particularly from 1 to 3 substituents, selected from the group consisting of acyl, acylamino, acyloxy, alkoxy, substituted alkoxy, alkoxycarbonyl, alkoxycarbonylamino, amino, substituted amino, aminocarbonyl, aminocarbonylamino, aminocarbonyloxy, aryl, aryloxy, azido, carboxyl, cyano, cycloalkyl, substituted cycloalkyl, halogen, hydroxyl, keto, nitro, thioalkoxy, substituted thioalkoxy, thioaryloxy, thioketo, thiol, alkyl-S(O)-, aryl-S(O)-, alkyl-S(0) 2 - and aryl-S(0) 2 -.

[0033] "Alkoxy" refers to the group -OR 24 where R 24 is alkyl. Particular alkoxy groups include, by way of example, methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, tert-butoxy, sec-butoxy, n-pentoxy, n-hexoxy, 1 ,2-dimethylbutoxy, and the like.

[0034] "Substituted alkoxy" includes those groups recited in the definition of

"substituted" herein, and particularly refers to an alkoxy group having 1 or more substituents, for instance from 1 to 5 substituents, and particularly from 1 to 3 substituents, selected from the group consisting of acyl, acylamino, acyloxy, alkoxy, substituted alkoxy, alkoxycarbonyl, alkoxycarbonylamino, amino, substituted amino, aminocarbonyl, aminocarbonylamino, aminocarbonyloxy, aryl, aryloxy, azido, carboxyl, cyano, cycloalkyl, substituted cycloalkyl, halogen, heteroaryl, hydroxyl, keto, nitro, thioalkoxy, substituted thioalkoxy, thioaryloxy, thioketo, thiol, alkyl-S(O)-, aryl-S(O)-, alkyl-S(0) 2 - and aryl-S(0) 2 -.

[0035] "Alkoxycarbonylamino" refers to the group -NR 25 C(0)OR 26 , where R 25 is hydrogen, alkyl, aryl or cycloalkyl, and R 26 is alkyl or cycloalkyl.

[0036] "Alkyl" refers to monovalent saturated alkane radical groups particularly having up to about 1 1 carbon atoms, more particularly as a lower alkyl, from 1 to 8 carbon atoms and still more particularly, from 1 to 6 carbon atoms. The hydrocarbon chain may be either straight- chained or branched. This term is exemplified by groups such as methyl, ethyl, n-propyl, isopropyl, «-butyl, /so-butyl, iert-butyl, «-hexyl, n-octyl, iert-octyl and the like. The term "lower alkyl" refers to alkyl groups having 1 to 6 carbon atoms. The term "alkyl" also includes

"cycloalkyls" as defined below.

[0037] "Substituted alkyl" includes those groups recited in the definition of "substituted" herein, and particularly refers to an alkyl group having 1 or more substituents, for instance from 1 to 5 substituents, and particularly from 1 to 3 substituents, selected from the group consisting of acyl, acylamino, acyloxy, alkoxy, substituted alkoxy, alkoxycarbonyl, alkoxycarbonylamino, amino, substituted amino, aminocarbonyl, aminocarbonylamino, aminocarbonyloxy, aryl, aryloxy, aaido, carboxyl, cyano, cycloalkyl, substituted cycloalkyl, halogen, hydroxyl, heteroaryl, keto, nitro; thioalkoxy, substituted thioalkoxy, thioaryloxy, thioketo, thiol, alkyl-S(O)-, aryl- S(O)-, alkyl-S(0) 2 -, and aryl-S(0) 2 -.

[0038] Alkylene" refers to divalent saturated alkene radical groups having 1 to 1 1 carbon atoms and more particularly 1 to 6 carbon atoms which can be straight-chained or branched. This term is exemplified by groups such as methylene (-CH 2 -), ethylene (-CH 2 CH 2 -), the propylene isomers (e.g., -CH 2 CH 2 CH 2 - and -CH(CH 3 )CH 2 -) and the like.

[0039] Substituted alkylene" includes those groups recited in the definition of

"substituted" herein, and particularly refers to an alkylene group having 1 or more substituents, for instance from 1 to 5 substituents, and particularly from 1 to 3 substituents, selected from the group consisting of acyl, acylamino, acyloxy, alkoxy, substituted alkoxy, alkoxycarbonyl, alkoxycarbonylamino, amino, substituted amino, aminocarbonyl, amino-carbonylamino, aminocarbonyloxy, aryl, aryloxy, azido, carboxyl, cyano, halogen, hydroxyl, keto, nitro, thioalkoxy, substituted thioalkoxy, thioaryloxy, thioketo, thiol, alkyl-S(O)-, aryl-S(O)-, alkyl- S(0) 2 - and aryl-S(0) 2 -.

[0040] "Alkenyl" refers to monovalent olefinically unsaturated hydrocarbyl groups preferably having 2 to 1 1 carbon atoms, particularly, from 2 to 8 carbon atoms, and more particularly, from 2 to 6 carbon atoms, which can be straight-chained or branched and having at least 1 and particularly from 1 to 2 sites of olefinic unsaturation. Particular alkenyl groups include ethenyl (-CH=CH 2 ), rc-propenyl (-CH 2 CH=CH 2 ), isopropenyl (-C(CH 3 )=CH 2 ), vinyl and substituted vinyl, and the like.

[0041] "Alkenylene" refers to divalent olefinically unsaturated hydrocarbyl groups particularly having up to about 1 1 carbon atoms and more particularly 2 to 6 carbon atoms which can be straight-chained or branched and having at least 1 and particularly from 1 to 2 sites of olefinic unsaturation. This term is exemplified by groups such as ethenylene (-CH=CH-), the propenylene isomers (e.g., -CH=CHCH 2 - and -C(CH 3 )=CH- and -CH=C(CH 3 )-) and the like.

[0042] "Alkynyl" refers to acetylenically or alkynically unsaturated hydrocarbyl groups particularly having 2 to 1 1 carbon atoms, and more particularly 2 to 6 carbon atoms which can be straight-chained or branched and having at least 1 and particularly from 1 to 2 sites of alkynyl unsaturation. Particular non-limiting examples of alkynyl groups include acetylenic, ethynyl (- C≡CH), propargyl (-CH 2 C≡CH), and the like.

[0043] "Substituted alkynyl" includes those groups recited in the definition of

"substituted" herein, and particularly refers to an alkynyl group having 1 or more substituents, for instance from 1 to 5 substituents, and particularly from 1 to 3 substituents, selected from the group consisting of acyl, acylamino, acyloxy, alkoxy, substituted alkoxy, alkoxycarbonyl, alkoxycarbonylamino, amino, substituted amino, aminocarbonyl, aminocarbonylamino, aminocarbonyloxy, aryl, aryloxy, azido, carboxyl, cyano, cycloalkyl, substituted cycloalkyl, halogen, hydroxyl, keto, nitro, thioalkoxy, substituted thioalkoxy, thioaryloxy, thioketo, thiol, alkyl-S(O)-, aryl-S(O)-, alkyl-S(0) 2 - and aryl-S(0) 2 -.

[0044] "Alkanoyl" or "acyl" as used herein refers to the group R 27 -C(0)-, where R 27 is hydrogen or alkyl as defined above.

[0045] "Aryl" refers to a monovalent aromatic hydrocarbon group derived by the removal of one hydrogen atom from a single carbon atom of a parent aromatic ring system. Typical aryl groups include, but are not limited to, groups derived from aceanthrylene, acenaphthylene, acephenanthrylene, anthracene, azulene, benzene, chrysene, coronene, fluoranthene, fluorene, hexacene, hexaphene, hexalene, as-indacene, s-indacene, indane, indene, naphthalene, octacene, octaphene, octalene, ovalene, penta-2,4-diene, pentacene, pentalene, pentaphene, perylene, phenalene, phenanthrene, picene, pleiadene, pyrene, pyranthrene, rubicene, triphenylene, trinaphthalene and the like. Particularly, an aryl group comprises from 6 to 14 carbon atoms.

[0046] "Substituted Aryl" includes those groups recited in the definition of "substituted" herein, and particularly refers to an aryl group that may optionally be substituted with 1 or more substituents, for instance from 1 to 5 substituents, particularly 1 to 3 substituents, selected from the group consisting of acyl, acylamino, acyloxy, alkenyl; substituted alkenyl, alkoxy, substituted alkoxy, alkoxycarbonyl, alkyl, substituted alkyl, alkynyl, substituted alkynyl, amino, substituted amino, aminocarbonyl, aminocarbonylamino, aminocarbonyloxy, aryl, aryloxy, azido, carboxyl, cyano, cycloalkyl, substituted cycloalkyl, halogen, hydroxyl, nitro, thioalkoxy, substituted thioalkoxy, thioaryloxy, thiol, alkyl-S(O)-, aryl-S(O)-, alkyl-S(0) 2 - and aryl-S(0) 2 -.

[0047] "Fused Aryl" refers to an aryl having two of its ring carbon in common with a second aryl ring or with an aliphatic ring.

[0048] "Alkaryl" refers to an aryl group, as defined above, substituted with one or more alkyl groups, as defined above. ^

[0049] "Aralkyl" or "arylalkyl" refers to an alkyl group, as defined above, substituted with one or more aryl groups, as defined above.

[0050] "Aryloxy" refers to -O-aryl groups wherein "aryl" is as defined above.

[0051] "Alkylamino" refers to the group alkyl-NR 28 R 29 , wherein each of R 28 and R 29 are independently selected from hydrogen and alkyl.

[0052] "Arylamino" refers to the group aryl-NR 30 R 31 , wherein each of R 30 and R 31 are independently selected from hydrogen, aryl and heteroaryl.

[0053] "Alkoxyamino" refers to a radical -N(H)OR 32 where R 32 represents an alkyl or cycloalkyl group as defined herein. [0054] . "Alkoxycarbonyl" refers to a radical -C(0)-alkoxy where alkoxy is as defined herein. ■■

[0055] "Alkylarylamino" refers to a radical -NR 33 R 34 where R 33 represents an alkyl or cycloalkyl group and R 34 is an aryl as defined herein.

[0056] "Alkylsulfonyl" refers to a radical -S(0) 2 R 35 where R 35 is an alkyl or cycloalkyl group as defined herein. Representative examples include, but are not limited to, methylsulfonyl, ethylsulfonyl, propylsulfonyl, butylsulfonyl and the like.

[0057] "Alkylsulfinyl" refers to a radical -S(0)R 35 where R 35 is an alkyl or cycloalkyl group as defined herein. Representative examples include, but are not limited to, methylsulfinyl, ethylsulfinyl, propylsulfinyl, butylsulfinyl and the like.

[0058] "Alkylthio" refers to a radical -SR 35 where R 35 is an alkyl or cycloalkyl group as defined herein that may be optionally substituted as defined herein. Representative examples include, but are not limited to, methylthio, ethylthio, propylthio, butylthio, and the like.

[0059] "Amino" refers to the radical -NH 2 .

[0060] "Substituted amino" includes those groups recited in the definition of

"substituted" herein, and particularly refers to the group -N(R 36 ) 2 where each R 36 is

independently selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, cycloalkyl, substituted cycloalkyl, and where both R groups are joined to form an alkylene group. When both R groups are hydrogen, - N(R 36 ) 2 is an amino group.

[0061] "Aminocarbonyl" refers to the group -C(0)NR 37 R 37 where each R 37 is

independently hydrogen, alkyl, aryl and cycloalkyl, or where the R 37 groups are joined to form an alkylene group.

[0062] "Aminocarbonylamino" refers to the group -NR 38 C(0)NR 38 R 38 where each R 38 is independently hydrogen, alkyl, aryl or cycloalkyl, or where two R groups are joined to form an alkylene group.

[0063] "Aminocarbonyloxy" refers to the group -OC(0)NR 39 R 39 where each R 39 is independently hydrogen, alkyl, aryl or cycloalky, or where the R groups are joined to form an alkylene group.

[0064] "Arylalkyloxy" refers to an -O-arylalkyl radical where arylalkyl is as defined herein.

[0065] "Arylamino" means a radical -NHR 40 where R 40 represents an aryl group as defined herein.

[0066] "Aryloxycarbonyl" refers to a radical -C(0)-0-aryl where aryl is as defined herein. [0067] . "An lsulfonyl" refers to a radical -S(0) 2 R 41 where R 41 is an aryl or heteroaryl group as defined herein.

[0068] "Azido" refers to the radical -N 3 .

"Bicycloaryl" refers to a monovalent aromatic hydrocarbon group derived by the removal of one hydrogen atom from a single carbon atom of a parent bicycloaromatic ring system. Typical bicycloaryl groups include, but are not limited to, groups derived from indane, indene, naphthalene, tetrahydronaphthalene, and the like. Particularly, an aryl group comprises from 8 to 1 1 carbon atoms.

[0069] "Bicycloheteroaryl" refers to a monovalent bicycloheteroaromatic group derived by the removal of one hydrogen atom from a single atom of a parent bicycloheteroaromatic ring system. Typical bicycloheteroaryl groups include, but are not limited to, groups derived from benzofuran, benzimidazole, benzindazole, benzdioxane, chromene, chromane, cinnoline, phthalazine, indole, indoline, indolizine, isobenzofuran, isochromene, isoindole, isoindoline, isoquinoline, benzothiazole, benzoxazole, naphthyridine, benzoxadiazole, pteridine, purine, benzopyran, benzpyrazine, pyridopyrimidine, quinazoline, quinoline, quinolizine, quinoxaline, benzomorphan, tetrahydroisoquinoline, tetrahydroquinoline, and the like. Preferably, the bicycloheteroaryl group is between 9-1 1 membered bicycloheteroaryl, with 5-10 membered heteroaryl being particularly preferred. Particular bicycloheteroaryl groups are those derived from benzothiophene, benzofuran, benzothiazole, indole, quinoline, isoquinoline, benzimidazole, benzoxazole and benzdioxane.

[0070] "Carbamoyl" refers to the radical -C(0)N(R 42 ) 2 where each R 42 group is independently hydrogen, alkyl, cycloalkyl or aryl, as defined herein, which may be optionally substituted as defined herein.

[0071] "Carboxy" refers to the radical -C(0)OH.

[0072] "Carboxyamino" refers to the radical -N(H)C(0)OH.

[0073] "Cycloalkyl" refers to cyclic hydrocarbyl groups having from 3 to about 10 carbon atoms and having a single cyclic ring or multiple condensed rings, including fused and bridged ring systems, which optionally can be substituted with from 1 to 3 alkyl groups. Such cycloalkyl groups include, by way of example, single ring structures such as cyclopropyl, cyclobutyl, cyclopentyl, cyclooctyl, 1 -methylcyclopropyl, 2-methylcyclopentyl, 2- methylcyclooctyl, and the like, and multiple ring structures such as adamantanyl, and the like.

[0074] "Substituted cycloalkyl" includes those groups recited in the definition of

"substituted" herein, and particularly refers to a cycloalkyl group having 1 or more substituents, for instance from 1 to 5 substituents, and particularly from 1 to 3 substituents, selected from the group consisting of acyl, acylamino, acyloxy, alkoxy, substituted alkoxy, alkoxycarbonyl, alkoxycarhonylamino, amino, substituted amino, aminocarbonyl, aminocarbonylamino, aminocarbonyloxy, aryl, aryloxy, azido, carboxyl, cyano, cycloalkyl, substituted cycloalkyl, halogen, hydroxyl, keto, nitro, thioalkoxy, substituted thioalkoxy, thioaryloxy, thioketo, thiol, alkyl-S(O)-, aryl-S(O)-, alkyl-S(0) 2 - and aryl-S(0) 2 -.

[0075] "Cycloalkoxy" refers to the group -OR 43 where R 43 is cycloalkyl. Such cycloalkoxy groups include, by way of example, cyclopentoxy, cyclohexoxy and the like.

[0076] "Cycloalkenyl" refers to cyclic hydrocarbyl groups having from 3 to 10 carbon atoms and having a single cyclic ring or multiple condensed rings, including fused and bridged ring systems and having at least one and particularly from 1 to 2 sites of olefinic unsaturation. Such cycloalkenyl groups include, by way of example, single ring structures such as

cyclohexenyl, cyclopentenyl, cyclopropenyl, and the like.

[0077] "Substituted cycloalkenyl" includes those groups recited in the definition of

"substituted" herein, and particularly refers to a cycloalkenyl group having 1 or more

substituents, for instance from 1 to 5 substituents, and particularly from 1 to 3 substituents, selected from the group consisting of acyl, acylamino, acyloxy, alkoxy, substituted alkoxy, alkoxycarbonyl, alkoxycarhonylamino, amino, substituted amino, aminocarbonyl,

aminocarbonylamino, aminocarbonyloxy, aryl, aryloxy, azido, carboxyl, cyano, cycloalkyl, substituted cycloalkyl, halogen, hydroxyl, keto, nitro, thioalkoxy, substituted thioalkoxy, thioaryloxy, thioketo, thiol, alkyl-S(O)-, aryl-S(O)-, alkyl-S(0) 2 - and aryl-S(0) 2 -.

[0078] "Fused Cycloalkenyl" refers to a cycloalkenyl having two of its ring carbon atoms in common with a second aliphatic or aromatic ring and having its olefinic unsaturation located to impart aromaticity to the cycloalkenyl ring.

[0079] "Cyanato" refers to the radical -OCN.

[0080] "Cyano" refers to the radical -CN.

[0081] "Dialkylamino" means a radical -NR 44 R 45 where R 44 and R 45 independently represent an alkyl, substituted alkyl, aryl, substituted aryl, cycloalkyl, substituted cycloalkyl, cycloheteroalkyl, substituted cycloheteroalkyl, heteroaryl, or substituted heteroaryl group as defined herein.

[0082] "Ethenyl" refers to substituted or unsubstituted -(C=C)-.

[0083] "Ethylene" refers to substituted or unsubstituted -(C-C)-.

[0084] "Ethynyl" refers to -(C≡C)-.

[0085] "Halo" or "halogen" refers to fluoro, chloro, bromo and iodo. Preferred halo groups are either fluoro or chloro.

[00861 "Hydroxy" refers to the radical -OH.

[0087] "Nitro" refers to the radical -N0 2 . [0088] * "Substituted" refers to a group in which one or more hydrogen atoms are each independently replaced with the same or different substituent(s). Typical substituents include, but are not limited to, -X, -R 46 , -O " , =0, -OR 46 , -SR 46 , -S " , =S, -NR 46 R 47 , =NR 46 , -CX 3 , -CF 3 , -

CN, -OCN, -SCN, -NO, -N0 2 , =N 2 , -N 3 , -S(0) 2 0 " , -S(0) 2 OH, -S(0) 2 R 46 , -OS(0 2 )0 " , -

OS(0) 2 R 46 , -P(0)(0 ) 2 , -P(0)(OR 6 )(0 ), -OP(0)(OR 46 )(OR 47 ), -C(0)R 46 , -C(S)R 46 , -C(0)OR 46 ,

-C(0)NR 46 R 47 , -C(0)0 " , -C(S)OR 46 , -NR 48 C(0)NR 46 R 47 , -NR 8 C(S)NR 46 R 47 , -

NR 49 C(NR 8 )NR 46 R 47 and -C(NR 48 )NR 46 R 47 , where each X is independently a halogen; each R 46 ,

R 47 , R 48 and R 49 are independently hydrogen, alkyl, substituted alkyl, aryl, substituted alkyl, arylalkyl, substituted alkyl, cycloalkyl, substituted alkyl, cycloheteroalkyl, substituted cycloheteroalkyl, heteroalkyl, substituted heteroalkyl, heteroaryl, substituted heteroaryl, heteroarylalkyl, substituted heteroarylalkyl, -NR 50 R 51 , -C(0)R 50 or -S(0) 2 R 50 or optionally R 50 and R 51 together with the atom to which they are both attached form a cycloheteroalkyl or substituted cycloheteroalkyl ring ; and R 50 and R 51 are independently hydrogen, alkyl, substituted alkyl, aryl, substituted alkyl, arylalkyl, substituted alkyl, cycloalkyl, substituted alkyl, cycloheteroalkyl, substituted cycloheteroalkyl, heteroalkyl, substituted heteroalkyl, heteroaryl, substituted heteroaryl, heteroarylalkyl or substituted heteroarylalkyl.

[0089] Examples of representative substituted ar ls include the following

[0090] In these formulae one of R and R may be hydrogen and at least one of R and

R 53 is each independently selected from alkyl, alkenyl, alkynyl, cycloheteroalkyl, alkanoyl, alkoxy, aryloxy, heteroaryloxy, alkylamino, arylamino, heteroarylamino, NR 54 COR 55 ,

NR 54 SOR 55 , NR 54 S0 2 R 57 , COOalkyl, COOaryl, CONR 54 R 55 , CONR 54 OR 55 , NR 54 R 55 ,

S0 2 NR 5 R 55 , S-alkyl, S-alkyl, SOalkyl, S0 2 alkyl, Saryl,'SOaryl, S0 2 aryl; or R 52 and R 53 may be joined to form a cyclic ring (saturated or unsaturated) from 5 to 8 atoms, optionally containing one or more heteroatoms selected from the group N, O or S. R 54 , R 55 , and R 56 are independently hydrogen, alkyl, alkenyl, alkynyl, perfluoroalkyl, cycloalkyl, cycloheteroalkyl, aryl, substituted aryl, heteroaryl, substituted or hetero alkyl or the like.

[0091] "Hetero" when used to describe a compound or a group present on a compound means that one or more carbon atoms in the compound or group have been replaced by a nitrogen, oxygen, or sulfur heteroatom. Hetero may be applied to any of the hydrocarbyl groups described above such as alkyl, e.g. heteroalkyl, cycloalkyl, e.g. cycloheteroalkyl, aryl, e.g.

heteroaryl, cycloalkenyl, cycloheteroalkenyl, and the like having from 1 to 5, and especially from 1 to 3 heteroatoms. [0092] ·* "Heteroaryl" refers to a monovalent heteroaromatic group derived by the removal of, one hydrogen atom from a single atom of a parent heteroaromatic ring system. Typical heteroaryl groups include, but are not limited to, groups derived from acridine, arsindole, carbazole, β-carboline, chromane, chromene, cinnoline, furan, imidazole, indazole, indole, indoline, indolizine, isobenzofuran, isochromene, isoindole, isoindoline, isoquinoline, isothiazole, isoxazole, naphthyridine, oxadiazole, oxazole, perimidine, phenanthridine, phenanthroline, phenazine, phthalazine, pteridine, purine, pyran, pyrazine, pyrazole, pyridazine, pyridine, pyrimidine, pyrrole, pyrrolizine, quinazoline, quinoline, quinolizine, quinoxaline, tetrazole, thiadiazole, thiazole, thiophene, triazole, xanthene, and the like. Preferably, the heteroaryl group is between 5- 15 membered heteroaryl, with 5-10 membered heteroaryl being particularly preferred. Particular heteroaryl groups are those derived from thiophene, pyrrole, benzothiophene, benzofuran, indole, pyridine, quinoline, imidazole, oxazole and pyrazine.

[0093] Exam les of representative heteroaryls include the following:

wherein each Y is selected from carbonyl, N, NR , O, and S; and R is independently hydrogen, alkyl, cycloalkyl, cycloheteroalkyl, aryl, heteroaryl, heteroalkyl or the like.

[0094] As used herein, the term "cycloheteroalkyl" refers to a stable heterocyclic non- aromatic ring and fused rings containing one or more heteroatoms independently selected from N, O and S. A fused heterocyclic ring system may include carbocyclic rings and need only include one heterocyclic ring. Examples of heterocyclic rings include, but are not limited to, piperazinyl, homopiperazinyl, piperidinyl and morpholinyl, and are shown in the following illustrative examples:

wherein each X is selected from CR 2 , NR , O and S; and each Y is selected from NR , O and

S; and R is independently hydrogen, alkyl, cycloalkyl, cycloheteroalkyl, aryl, heteroaryl, heteroalkyl or the like. These cycloheteroalkyl rings may be optionally substituted with one or more groups selected from the group consisting of acyl, acylamino, acyloxy, alkoxy, substituted alkoxy, alkoxycarbonyl, alkoxycarbonylamino, amino, substituted amino, aminocarbonyl, aminocarbonylamino, aminocarbonyloxy, aryl, aryloxy, azido, carboxyl, cyano, cycloalkyl, substituted cycloalkyl, halogen, hydroxyl, keto, nitro, thioalkoxy, substituted thioalkoxy, thioaryloxy, thioketo, thiol, alkyl-S(O)-, aryl-S(O)-, alkyl-S(0) 2 - and aryl-S(0) 2 -. Substituting groups include carbonyl or thiocarbonyl which provide, for example, lactam and urea derivatives.

[0095] Examples of representative cycloheteroalkenyls include the following:

wherein each X is selected from CR 2 , NR , O and S; and each Y is selected from carbonyl, N,

58 58

NR , O and S; and R is independently hydrogen, alkyl, cycloalkyl, cycloheteroalkyl, aryl, heteroaryl, heteroalkyl or the like.

[0096] Examples of representative aryl having hetero atoms containing substitution include the followin :

wherein each X is selected from C-R 2 NR , O and S; and each Y is selected from carbonyl, NR 58 , O and S; and R 58 is independently hydrogen, alkyl, cycloalkyl, cycloheteroalkyl, aryl, heteroaryl, heteroalkyl or the like. [0097] . "Hetero substituent" refers to a halo, O, S or N atom-containing functionality that may be present as an R 4 in a R C group present as substituents directly on A, B, W, Y or Z of the compounds of this invention or may be present as a substituent in the "substituted" aryl and aliphatic groups present in the compounds.

Examples of hetero substituents include:

-halo,

-NO2, -NH 2 , -NHR 59 , -N(R 59 ) 2 ,

-NRCOR, -NR 59 SOR 59 , -NR 59 S0 2 R 59 , OH, CN,

-C0 2 H,

-R 59 -OH, -O-R 59 , -COOR 59 ,

-CON(R 59 ) 2 , -CONROR 59 ,

-SO3H, -R 59 -S, -S0 2 N(R 59 ) 2 ,

-S(0)R 59 , -S(0) 2 R 59

wherein each R 59 is independently an aryl or aliphatic, optionally with substitution. Among hetero substituents containing R 59 groups, preference is given to those materials having aryl and alkyl R 59 groups as defined herein. Preferred hetero substituents are those listed above.

[0098] "Hydrogen bond donor" group refers to a group containg O-H, or N-H functionality. Examples of "hydrogen bond donor" groups include -OH, -NH 2 , and -NH-R 59a and wherein R 59a is alkyl, cycloalkyl, aryl, or heteroaryl.

[0099] "Dihydroxyphosphoryl" refers to the radical -PO(OH) 2 .

[00100] "Substituted dihydroxyphosphoryl" includes those groups recited in the definition of "substituted" herein, and particularly refers to a dihydroxyphosphoryl radical wherein one or both of the hydroxyl groups are substituted. Suitable substituents are described in detail below.

[00101 ] "Aminohydroxyphosphoryl" refers to the radical -PO(OH)NH 2 .

[00102] "Substituted aminohydroxyphosphoryl" includes those groups recited in the definition of "substituted" herein, and particularly refers to an aminohydroxyphosphoryl wherein the amino group is substituted with one or two substituents. Suitable substituents are described in detail below. In certain embodiments, the hydroxyl group can also be substituted.

[00103] "Thioalkoxy" refers to the group -SR 60 where R 60 is alkyl.

[00104] "Substituted thioalkoxy" includes those groups recited in the definition of

"substituted" herein, and particularly refers to a thioalkoxy group having 1 or more substituents, for instance from 1 to 5 substituents, and particularly from 1 to 3 substituents, selected from the group consisting of acyl, acylamino, acyloxy, alkoxy, substituted alkoxy, alkoxycarbonyl, alkoxycarbonylamino, amino, substituted amino, aminocarbonyl, aminocarbonylamino, aminocarbonyloxy, aryl, aryloxy, azido, carboxyl, cyano, cycloalkyl, substituted cycloalkyl, halogen, hydroxyl, keto, nitro, thioalkoxy, substituted thioalkoxy, thioaryloxy, thioketo, thiol, alkyl-S(a)-, aryl-S(O)-, alkyl-S(0) 2 - and aryl-S(0) 2 -.

[001051 "Sulfanyl" refers to the radical HS-. "Substituted sulfanyl" refers to a radical such as RS- wherein R is any substituent described herein.

[00106) "Sulfonyl" refers to the divalent radical -S(0 2 )-. "Substituted sulfonyl" refers to a radical such as R 6 l -(0 2 )S- wherein R 61 is any substituent described herein. "Aminosulfonyl" or "Sulfonamide" refers to the radical H 2 N(0 2 )S-, and "substituted aminosulfonyl" "substituted sulfonamide" refers to a radical such as R 62 2 N(0 2 )S- wherein each R 62 is independently any substituent described herein.

[00107] "Sulfone" refers to the group -S0 2 R 63 . In particular embodiments, R 63 is selected from H, lower alkyl, alkyl, aryl and heteroaryl.

[00108] "Thioaryloxy" refers to the group -SR 64 where R 64 is aryl.

[00109] "Thioketo" refers to the group =S.

[00110] "Thiol" refers to the group -SH.

[00111] One having ordinary skill in the art of organic synthesis will recognize that the maximum number of heteroatoms in a stable, chemically feasible heterocyclic ring, whether it is aromatic or non aromatic, is determined by the size of the ring, the degree of unsaturation and the valence of the heteroatoms. In general, a heterocyclic ring may have one to four heteroatoms so long as the heteroaromatic ring is chemically feasible and stable.

[00112] "Pharmaceutically acceptable" means approved by a regulatory agency of the

Federal or a state government or listed in the U.S. Pharmacopoeia or other generally recognized pharmacopoeia for use in animals, and more particularly in humans.

[00113] "Pharmaceutically acceptable salt" refers to a salt of a compound of the invention that is pharmaceutically acceptable and that possesses the desired pharmacological activity of the parent compound. Such salts include: (1 ) acid addition salts, formed with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like; or formed with organic acids such as acetic acid, propionic acid, hexanoic acid,

cyclopentanepropionic acid, glycolic acid, pyruvic acid, lactic acid, malonic acid, succinic acid, malic acid, maleic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, 3-(4- hydroxybenzoyl) benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid,

ethanesulfonic acid, 1 ,2-ethane-disulfonic acid, 2-hydroxyethanesulfonic acid, benzenesulfonic acid, 4-chlorobenzenesulfonic acid, 2-naphthalenesulfonic acid, 4-toluenesulfonic acid, camphorsulfonic acid, 4-methylbicyclo[2.2.2]-oct-2-ene-l -carboxylic acid, glucoheptonic acid, 3-phenylpropionic acid, trimethylacetic acid, tertiary butylacetic acid, lauryl sulfuric acid, gluconic acid, glutamic acid, hydroxynaphthoic acid, salicylic acid, stearic acid, muconic acid, and the like; or (2.) salts formed when an acidic proton present in the parent compound either is replaced by a metal ion, e.g., an alkali metal ion, an alkaline earth ion, or an aluminum ion; or coordinates with an organic base such as ethanolamine, diethanolamine, triethanolamine, N- methylglucamine and the like. Salts further include, by way of example only, sodium, potassium, calcium, magnesium, ammonium, tetraalkylammonium, and the like; and when the compound contains a basic functionality, salts of non toxic organic or inorganic acids, such as hydrochloride, hydrobromide, tartrate, mesylate, acetate, maleate, oxalate and the like. The term

"pharmaceutically acceptable cation" refers to a non toxic, acceptable cationic counter-ion of an acidic functional group. Such cations are exemplified by sodium, potassium, calcium, magnesium, ammonium, tetraalkylammonium cations, and the like.

[00114] "Pharmaceutically acceptable vehicle" refers to a diluent, adjuvant, excipient or carrier with which a compound of the invention is administered.

[00115] "Preventing" or "prevention" refers to a reduction in risk of acquiring a disease or disorder (i.e., causing at least one of the clinical symptoms of the disease not to develop in a subject that may be exposed to or predisposed to the disease but does not yet experience or display symptoms of the disease).

[00116] "Prodrugs" refers to compounds, including derivatives of the compounds of the invention,which have cleavable groups and become by solvolysis or under physiological conditions the compounds of the invention which are pharmaceutically active in vivo. Such examples include, but are not limited to, choline ester derivatives and the like, N- alkylmorpholine esters and the like.

[00117] "Solvate" refers to forms of the compound that are associated with a solvent, usually by a solvolysis reaction. Conventional solvents include water, ethanol, acetic acid and the like. The compounds of the invention may be prepared e.g. in crystalline form and may be solvated or hydrated. Suitable solvates include pharmaceutically acceptable solvates, such as hydrates, and further include both stoichiometric solvates and non-stoichiometric solvates.

[00118] "Subject" includes humans. The terms "human," "patient" and "subject" are used interchangeably herein.

[00119] "Therapeutically effective amount" means the amount of a compound that, when administered to a subject for treating a disease, is sufficient to effect such treatment for the disease. The "therapeutically effective amount" can vary depending on the compound, the disease and its severity, and the age, weight, etc., of the subject to be treated.

[00120] "Treating" or "treatment" of any disease or disorder refers, in one embodiment, to ameliorating the disease or disorder (i.e., arresting or reducing the development of the disease or at least one of the clinical symptoms thereof). In another embodiment "treating" or "treatment" refers to ameliorating at least one physical parameter, which may not be discernible by the subject. In yet another embodiment, "treating" or "treatment" refers to modulating the disease or disorder, either physically, {e.g., stabilization of a discernible symptom), physiologically, (e.g., stabilization of a physical parameter), or both.

[00121] As used herein, the term "operably linked" refers to a regulatory sequence capable of mediating the expression of a coding sequence and which is placed in a DNA molecule (e.g., an expression vector) in an appropriate position relative to the coding sequence so as to effect expression of the coding sequence. This same definition is sometimes applied to the arrangement of coding sequences and transcription control elements (e.g. promoters, enhancers, and termination elements) in an expression vector. This definition is also sometimes applied to the arrangement of nucleic acid sequences of a first and a second nucleic acid molecule wherein a hybrid nucleic acid molecule is generated.

[00122] A "vector" is a replicon, such as a plasmid, cosmid, bacmid, phage or virus, to which another genetic sequence or element (either DNA or RNA) may be attached so as to bring about the replication of the attached sequence or element.

[00123] An "expression vector" or "expression operon" refers to a nucleic acid segment that may possess transcriptional and translational control sequences, such as promoters, enhancers, translational start signals (e.g., ATG or AUG codons), polyadenylation signals, terminators, and the like, and which facilitate the expression of a polypeptide coding sequence in a host cell or organism.

[00124] The terms "transform", "transfect", or "transduce", shall refer to any method or means by which a nucleic acid is introduced into a cell or host organism and may be used interchangeably to convey the same meaning. Such methods include, but are not limited to, transfection, electroporation, microinjection, PEG-fusion and the like.

[00125] The introduced nucleic acid may or may not be integrated (covalently linked) into nucleic acid of the recipient cell or organism. In bacterial, yeast, plant and mammalian cells, for example, the introduced nucleic acid may be maintained as an episomal element or independent replicon such as a plasmid. Alternatively, the introduced nucleic acid may become integrated into the nucleic acid of the recipient cell or organism and be stably maintained in that cell or organism and further passed on or inherited to progeny cells or organisms of the recipient cell or organism. In other applications, the introduced nucleic acid may exist in the recipient cell or host organism only transiently.

[00126] The phrase "consisting essentially of when referring to a particular nucleotide or amino acid means a sequence having the properties of a given SEQ ID NO:. For example, when used in reference to an amino acid sequence, the phrase includes the sequence per se and molecular modifications that would not affect the basic and novel characteristics of the sequence.

[00127] Other derivatives of the compounds of this invention have activity in both their acid and acid derivative forms, but in the acid sensitive form often offers advantages of solubility, tissue compatibility, or delayed release in the mammalian organism (see, Bundgard,

H., Design of Prodrugs, pp. 7-9, 21 -24, Elsevier, Amsterdam 1985). Prodrugs include acid derivatives well know to practitioners of the art, such as, for example, esters prepared by reaction of the parent acid with a suitable alcohol, or amides prepared by reaction of the parent acid compound with a substituted or unsubstituted amine, or acid anhydrides, or mixed anhydrides.

Simple aliphatic or aromatic esters, amides and anhydrides derived from acidic groups pendant on the compounds of this invention are preferred prodrugs. In some cases it is desirable to prepare double ester type prodrugs such as (acyloxy)alkyl esters or

((alkoxycarbonyl)oxy)alkylesters. Preferred are the Ci to C 8 alkyl, C 2 -C 8 alkenyl, aryl, C 7 -Ci 2 substituted aryl, and C 7 -Ci2 arylalkyl esters of the compounds of the invention.

[00128] As used herein, the term "isotopic variant" refers to a compound that contains unnatural proportions of isotopes at one or more of the atoms that constitute such

compound. For example, an "isotopic variant" of a compound can contain one or more non-

2 13 *

radioactive isotopes, such as for example, deuterium ( H or D), carbon- 13 ( C), nitrogen- 15 ( l 5 N), or the like. It will be understood that, in a compound where such isotopic substitution is made, the following atoms, where present, may vary, so that for example, any hydrogen may be 2 H/D, any carbon may be 13 C, or any nitrogen may be l 5 N, and that the presence and placement of such atoms may be determined within the skill of the art. Likewise, the invention may include the preparation of isotopic variants with radioisotopes, in the instance for example, where the resulting compounds may be used for drug and/or substrate tissue distribution studies. The radioactive isotopes tritium, i.e. 3 H, and carbon- 14, i.e. ,4 C, are particularly useful for this purpose in view of their ease of incorporation and ready means of detection. Further, compounds may be prepared that are substituted with positron emitting isotopes, such as "C, 18 F, l 5 0 and 13 N, and would be useful in Positron Emission Topography (PET) studies for examining substrate receptor occupancy.

[00129] All isotopic variants of the compounds provided herein, radioactive or not, are intended to be encompassed within the scope of the invention.

[00130] It is also to be understood that compounds that have the same molecular formula but differ in the nature or sequence of bonding of their atoms or the arrangement of their atoms in space are termed "isomers". Isomers that differ in the arrangement of their atoms in space are termed "stereoisomers". [00131] ψ Stereoisomers that are not mirror images of one another are termed

"diastereomers" and those that are non-superimposable mirror images of each other are termed

"enantiomers". When a compound has an asymmetric center, for example, it is bonded to four different groups, a pair of enantiomers is possible. An enantiomer can be characterized by the absolute configuration of its asymmetric center and is described by the R- and S-sequencing rules of Cahn and Prelog, or by the manner in which the molecule rotates the plane of polarized light and designated as dextrorotatory or levorotatory (i.e., as (+) or (-)-isomers respectively). A chiral compound can exist as either individual enantiomer or as a mixture thereof. A mixture containing equal proportions of the enantiomers is called a "racemic mixture".

[00132] "Tautomers" refer to compounds that are interchangeable forms of a particular compound structure, and that vary in the displacement of hydrogen atoms and electrons. Thus, two structures may be in equilibrium through the movement of π electrons and an atom (usually

H). For example, enols and ketones are tautomers because they are rapidly interconverted by treatment with either acid or base. Another example of tautomerism is the aci- and nitro- forms of phenylnitromethane, that are likewise formed by treatment with acid or base.

[00133] Tautomeric forms may be relevant to the attainment of the optimal chemical reactivity and biological activity of a compound of interest.

[00134] The compounds of this invention may possess one or more asymmetric centers; such compounds can therefore be produced as individual (R)- or (S)- stereoisomers or as mixtures thereof. Unless indicated otherwise, the description or naming of a particular compound in the specification and claims is intended to include both individual enantiomers and mixtures, racemic or otherwise, thereof. The methods for the determination of stereochemistry and the separation of stereoisomers are well-known in the art.

THE COMPOUNDS

[00135] The present invention provides a method for preventing, treating or ameliorating in a mammal a disease or condition that is causally related to the activity of the RORy or RORyt in vivo, which comprises administering to the mammal an effective disease-treating or condition- treating amount of a compound according to formula I:

I

wherein

each n l and n2 is independently 1 , 2, 3, 4 or 5;

each R l a and R lb is independently H, substituted or unsubstituted Ci-C 6 alkyl, or CN; or

R l a and R lb joined together to form cycloalkyl ring;

R is H, substituted or unsubstituted Ci-C 6 alkyl, or aryl;

or one of R la and R l b is joined to the C of CR 2 to form a cyclopropyl ring; or R 2 is joined to the C of CR l a R l b to form a cyclopropyl ring;

each R 3 and R 4 is independently selected from H, OH, substituted or unsubstituted alkyl, substituted or unsubstituted alkoxy, substituted or unsubstituted acyl, substituted or unsubstituted acylamino, substituted or unsubstituted alkylamino, substituted or unsubstituted alkythio, substituted or unsubstituted alkoxycarbonyl, substituted or unsubstituted alkylarylamino, substituted or unsubstituted amino, substituted or unsubstituted arylalkyl, sulfo, substituted sulfo, substituted sulfonyl, substituted sulfinyl, substituted sulfanyl, substituted or unsubstituted aminosulfonyl, substituted or unsubstituted alkylsulfonyl, substituted or unsubstituted arylsulfonyl, azido, substituted or unsubstituted carbamoyl, carboxyl, cyano, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted dialkylamino, halo, nitro, and thiol; or any two adjacent R 3 groups, or any two adjacent R 4 groups may joined together to form a substituted or unsubstituted carbocyclic or heterocyclic ring;

each R 5 and R 6 is independently selected from H, substituted or unsubstituted alkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, substituted or unsubstituted aralkyl, and substituted or unsubstituted heteroarylalkyl; or

R 5 and R 6 , together with the N they are attached to, form a 4- 12 membered substituted or unsubstituted heterocycloalkyl;

or a pharmaceutically acceptable salt, solvate or prodrug thereof; and stereoisomers, isotopic variants and tautomers thereof..

[00136] , In one embodiment, with respect to the compounds of formula I, each R 5 and R 6 is independently selected from H, substituted or unsubstituted alkyl, substituted and unsubstituted cycloalkyl, and substituted and unsubstituted phenyl.

[00137] In another embodiment, with respect to the compounds of formula I, one of R 5 and

R b is H or e; and the other is independently selected from H, substituted or unsubstituted alkyl, substituted and unsubstituted cycloalkyl, and substituted and unsubstituted phenyl.

[00138] In another embodiment, with respect to the compounds of formula I, each R 5 and

R 6 is independently selected from substituted or unsubstituted alkyl.

[00139| In another embodiment, with respect to the compounds of formula I, each R 5 and

R 6 is independently selected from unsubstituted alkyl.

[00140] In one particular embodiment, with respect to the compounds of formula I, each

R 5 and R 6 is independently selected from H, Me, Et, n-Pr, i-Pr, n-Bu, and t-Bu.

[00141| In another embodiment, with respect to the compounds of formula I, each R 5 and

R 6 is independently selected from H, and alkyl substituted with hydroxyl, amino, alkylamino, dialkylamino. cylcloalkyl, heterocycloalkyl, aryl, and heteroaryl.

[00142] In another particular embodiment, with respect to the compounds of formula I, each R 5 and R 6 is independently selected from H, Me, Et, hydroxyethyl, hydroxypropyl, aminoethyl. aminopropyl, dimethylaminoethyl, dimethylaminopropyl, piperidinoethyl.

morpholinoethyl, cyclopropylmethyl, benzyl, phenethyl, and furanylmethyl.

|00143] In one embodiment, with respect to the compounds of formula I, each R 5 and R b is independently selected from H, and cycloalkyl.

|00144] In another embodiment, with respect to the compounds of formula I, each R 5 and

R 6 is independently selected from H, and cyclopropyl, cyclobutyl, cyclopentyl, and cyclohexyl.

[00145] In another particular embodiment, with respect to the compounds of formula I, R 5 is H; and R 6 is selected from hydroxyethyl, hydroxypropyl, aminoethyl, aminopropyl, dimethylaminoethyl, dimethylaminopropyl, piperidinoethyl, morpholinoethyl,

cyclopropylmethyl, cyclohexyl, benzyl, phenethyl, and furanylmethyl.

J00146] In yet another particular embodiment, with respect to the compounds of formula I,

R 5 and R 6 are joined together to form a 4- 12 membered substituted or unsubstituted

heterocycloalkyl.

[00147] In one embodiment, with respect to the compounds of formula I, the compound is according to formula la:

la

wherein

Cy is substituted or unsubstituted 4-12 membered substituted or unsubstituted

heterocycloalkyl;

and wherein n l , n2, R la , R l b , R 2 , R 3 , and R 4 are as described for formula I;

or a pharmaceutically acceptable salt, solvate or prodrug thereof;

and stereoisomers, isotopic variants and tautomers thereof.

[00148] In one embodiment, with respect to the compounds of formula la, Cy is azetidinyl, pyrrolidinyl, piperidinyl, piperizinyl, morpholinyl, tetrahydroquinolinyl, indolinyl, or azepinyl, unsubstituted or substituted with alkyl, substituted alkyl, haloalkyl, alkoxyalkyl,

hydroxyalkyl,alkoxy, hydroxyl, CN, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, amido, acyl, aroyl, or -CO-alkoxy.

[00149] In one embodiment, with respect to the compounds of formula la, Cy is substituted or unsubstituted azetidinyl, substituted or unsubstituted pyrrolidinyl, piperidinyl, piperizinyl, morpholinyl, or azepinyl. In another embodiment, Cy is unsubstituted pyrrolidinyl, piperidinyl, piperizinyl, morpholinyl, or azepinyl. In a particular embodiment, Cy is piperidinyl or piperizinyl.

[00150] In one embodiment, with respect to the compounds of formula I, the compound is according to formula Ila, lib, lie, lid, He, or Ilf:

l ie l id

He or | |f

and wherein n l , n2, R 1 , R 2 , R 3 , R 4 , R 5 , and R 6 are as described for formula I; and each R 5a and R 5b is independently H, alkyl, substituted alkyl, haloalkyl, alkoxyalkyl, hydroxyalkyl,alkoxy, hydroxyl, CN, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, amido, acyl, aroyl, or -CO-alkoxy.

[00151] In one embodiment, with respect to the compounds of formula I-IIf, n2 is 1 , 2, or

3.

[00152] In one embodiment, with respect to the compounds of formula I-IIf, n2 is 1 , 2, or

3; and each R 4 is independenly selected from halo, Ci-C 6 alkyl, CN, OH, and C|-C 6 alkoxy.

[00153] In one embodiment, with respect to the compounds of formulae I-IIf, n2 is 1 , 2, or

3; and each R 4 is independenly selected from CI, Me, OH, and Ci-C 6 alkoxy. In a particular embodiment, n2 is 3.

[00154] In one embodiment, with respect to the compounds of formula I, the compound is according to formula III:

wherein

and wherein n l , R l a , R lb , R 2 , R 3 , and R 4 , are as described for formula I; n2 is 1 , 2, or 3; A is C, N, O, or S; m is 0 or 1 ;

each R 5a is alkyl, substituted alkyl, halo, haloalkyl, hydroxyalkyl, aryl, heteroaryl, CN, alkoxyalkyl, amido, hydroxyl, alkoxy or substitituted alkoxy; and t is 0, 1 , 2, or 3;

R 7a is OH, substituted or unsubstituted alkyl, substituted or unsubstituted alkoxy, substituted or unsubstituted acyl, substituted or unsubstituted acylamino, substituted or unsubstituted alkylamino, substituted or unsubstituted alkythio, substituted or unsubstituted alkoxycarbonyl, substituted or unsubstituted alkylarylamino, substituted or unsubstituted amino, substituted or unsubstituted arylalkyl, sulfo, substituted sulfo, substituted sulfonyl, substituted sulfinyl, substituted sulfanyl, substituted or unsubstituted aminosulfonyl, substituted or unsubstituted alkylsulfonyl, substituted or unsubstituted arylsulfonyl, azido, substituted or unsubstituted carbamoyl, carboxyl, cyano, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted dialkylamino, halo, nitro, and thio; and

R 7b is R 4 ; or any two adjacent R 4 and R 7b groups, may joined together to form a substituted or unsubstituted carbocyclic or heterocyclic ring;

or a pharmaceutically acceptable salt, solvate or prodrug thereof;

and stereoisomers, isotopic variants and tautomers thereof.

[00155] In a further aspect, the present invention provides a compound according to formula III:

II I

wherein

A is C, N, O, or S; m is 0 or 1 ;

n l is independently 1 , 2, 3, 4 or 5; n2 is 1 , 2, or 3;

each R l a and R l b is independently H, substituted or unsubstituted C ! -C 6 alkyl, or CN; or

R l and R l b joined together to form cycloalkyl ring;

R 2 is H, substituted or unsubstituted C|-C6 alkyl, or aryl;

or one of R la and R , b is joined to the C of CR 2 to form a cyclopropyl ring: or R 2 is joined . to the C of CR l a R lb to form a cyclopropyl ring;

each R 3 and R 4 is independently selected from H, OH, substituted or unsubstituted alkyl, substituted or unsubstituted alkoxy, substituted or unsubstituted acyl, substituted or unsubstituted acylamino, substituted or unsubstituted alkylamino, substituted or unsubstituted alkythio, substituted or unsubstituted alkoxycarbonyl, substituted or unsubstituted alkylarylamino, substituted or unsubstituted amino, substituted or unsubstituted arylalkyl, sulfo, substituted sulfo, substituted sulfonyl, substituted sulfinyl, substituted sulfanyl, substituted or unsubstituted aminosulfonyl, substituted. or

unsubstituted alkylsulfonyl, substituted or unsubstituted arylsulfonyl, azido, substituted or unsubstituted carbamoyl, carboxyl, cyano, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, substituted or unsubstituted cycloalkyl, substituted or

unsubstituted heterocycioalkyl, substituted or unsubstituted dialkylamino, halo, nitro, and thiol; or any two adjacent R J groups, or any two adjacent R 4 , or any two adjacent R 4 and R 7b groups, may joined together to form a substituted or unsubstituted earbocyclic or heterocyclic ring;

each R 5a is alkyl, substituted alkyl. halo, haloalkyl, hydroxyalkyl, aryl, heteroaryl, CN, alkoxyalkyl, amido, hydroxyl, alkoxy or substitituted alkoxy; t is 0, 1 , 2, or 3;

R 7a is OH, substituted or unsubstituted alkyl, substituted or unsubstituted alkoxy, substituted or unsubstituted acyl, substituted or unsubstituted acylamino, substituted or unsubstituted alkylamino, substituted or unsubstituted alkythio, substituted or

unsubstituted alkoxycarbonyl, substituted or unsubstituted alkylarylamino, substituted or unsubstituted amino, substituted or unsubstituted arylalkyl, sulfo, substituted sulfo, substituted sulfonyl, substituted sulfinyl, substituted sulfanyl, substituted or unsubstituted aminosulfonyl, substituted or unsubstituted alkylsulfonyl, substituted or unsubstituted arylsulfonyl, azido, substituted or unsubstituted carbamoyl, carboxyl, cyano, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted dialkylamino, halo, nitro, and thio; and

R 7b is H, OH, substituted or unsubstituted alkyl, substituted or unsubstituted alkoxy, substituted or unsubstituted acyl, substituted or unsubstituted acylamino, substituted or unsubstituted alkylamino, substituted or unsubstituted alkythio, substituted or

unsubstituted alkoxycarbonyl, substituted or unsubstituted alkylarylamino, substituted or unsubstituted amino, substituted or unsubstituted arylalkyl, sulfo, substituted sulfo, substituted sulfonyl, substituted sulfinyl, substituted sulfanyl, substituted or unsubstituted aminosulfonyl, substituted or unsubstituted alkylsulfonyl, substituted or unsubstituted arylsulfonyl, azido, substituted or unsubstituted carbamoyl, carboxyl, cyano, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted dialkylamino, halo, nitro, and thio;

or a pharmaceutically acceptable salt, solvate or prodrug thereof;

and stereoisomers, isotopic variants and tautomers thereof;

provided that

i) when t is 1 , m is 1 , A is C, each of R la , R l b and R 2 is H, R 4 is 4-OMe, and one of R 7a and R 7b is OH and the other is OMe; then R 5a is other than 2- Me; '

ii) when t is 1 , m is 1 , A is N, and each of R l a , R l b and R 2 is H, R 4 is OMe, one of R 7a and R 7b is OH and the other is OMe; then R 5a is other than 4-Ph; iii) when each of R la , R lb , R 2 and R 3 is H, one of R 7a and R 7b is OH, CI, Br, Me, or F, and the other is H; then t is other than 0, and R 5a is other than benzyl, acetyl, cyclopentyl, Me, OH, CH 2 OMe, CH 2 OH, CH 2 CH 2 OMe, CH 2 CH 2 OH, OMe, C0 2 Me, C0 2 Et, or CONH 2 ;

iv) when t is 0, each of R l a , R lb and R 2 is H, R 4 is 4-OMe or 4-Me, and one of R 7a and R 7b is OH and the other is OMe or Me; then R 3 is other than 4- OMe, 4-NMe 2 , or 3,4-methylenedioxy; 3

v) the (R )ni-Ph- group is other than substituted or unsubstituted benzopyranyl ;

vi) when each of R la , R lb , R 2 and R 3 is H, one of R 7a and R 7b is OH, OMe, CI, Br, Me, or F, and the other is H; then one of (R 3 ) n |-Ph- group is other than unsubstituted phenyl; and

vii) when each of R l a , R lb , R 2 and R 3 is H, one of R 7a and R 7b is OMe, and the other is H; then one of (R 3 ) n |-Ph- group is other than unsubstituted naphthyl.

[00156] For the sake of clarity, the present invention does not include composition of matter for the com ounds according to formulae XXa-XXe: '

XXc XXd

Xe, R 4 = OMe or Me, R 7a = OH, R 7b = OMe or Me ·

wherein the coumarin or benzopyranyl (XXc or XXd) may be substituted or unsubstituted; and

A, m, R 3a , R 5a , t and n l are as described for formula III. [00157] ' In one embodiment, with respect to the compounds of formula III, t is 0. In another embodiment, t is 1 or 2.

[00158] In one embodiment, with respect to the compounds of formula III, each of R 7a and

R 7b is independently OH, substituted or unsubstituted alkyl, substituted or unsubstituted alkoxy, substituted or unsubstituted acyl, substituted or unsubstituted acylamino, substituted or unsubstituted alkylamino, substituted or unsubstituted alkythio, substituted or unsubstituted alkoxycarbonyl, substituted or unsubstituted alkylarylamino, substituted or unsubstituted amino, substituted or unsubstituted arylalkyl, sulfo, substituted sulfo, substituted sulfonyl, substituted sulfinyl, substituted sulfanyl, substituted or unsubstituted aminosulfonyl, substituted or unsubstituted alkylsulfonyl, substituted or unsubstituted arylsulfonyl, azido, substituted or unsubstituted carbamoyl, carboxyl, cyano, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted dialkylamino, halo, nitro, and thio.

[00159] In one embodiment, with respect to the compounds of formula III, each of R 7a and

R 7b is independently OH, substituted or unsubstituted alkyl, substituted or unsubstituted alkoxy, CN, halo, amido, or haloalkyl.

[00160] In one embodiment, with respect to the compounds of formula III, each of R 7a and

R 7b is independently halo, Ci-C 6 alkyl, CN, OH, or Ci-C 6 alkoxy.

[00161] In one embodiment, with respect to the compounds of formula III, each of R 7a and

R 7b is independently CI, F, Me, CF 3 , CN, OH, or OMe.

[00162] In one embodiment, with respect to the compounds of formula III, one of R /a and

R 7b is OH; and the other is alkoxy.

[00163] In one embodiment, with respect to the compounds of formula III, each of R 7a and

R 7b is OH or alkoxy.

[00164] In one embodiment, with respect to the compounds of formula III, one of R /a and

R 7b is OH; and the other is OMe.

[00165] In one embodiment, with respect to the compounds of formula I-III, R la and R l b is independently H, CN, or Me.

[00166] In one embodiment, with respect to the compounds of formula I-III, each of R la and R l b is H. In another embodiment each of R la and R lb is Me.

[00167] In one embodiment, with respect to the compounds of formula I-III, one of R l a and R l b is H; and the other is CN.

[00168] In one embodiment, with respect to the compounds of formula III, m is 0. [00169] In one embodiment, with respect to the compounds of formula III, m is 1 ; and A is

C or N. In -one embodiment, A is CH 2 , CHR 5a , or CR 5a R 5a . In another embodiment A is NH or

NR 5a . In a yet another embodiment, A is O. In a further embodiment, A is S.

[00170] In one embodiment, with respect to the compounds of formula III, R 4 is H, alkyl, substituted alkyl, halo, haloalkyl, hydroxyalkyl, alkoxyalkyl, amido, hydroxyl, cyano, or alkoxy.

[00171] In one embodiment, with respect to the compounds of formula III, R 4 is H, halo,

C|-C 6 alkyl, CN, OH, or C C 6 alkoxy.

[00172] In one embodiment, with respect to the compounds of formula III, R 4 is H, CI, F,

Me, CF 3 , CN, OH, or OMe.

[00173] In another particular embodiment, with respect to the compounds of formulae I-

III, n2 is 1 ; and R 4 is CI, F, Me, Et, i-Pr, OMe, CF 3 , CN or OH. In one embodiment R 4 is at 4- or para- position of the phenyl ring. In another embodiment, R 4 is 4-Cl, 4-F, 4-Me, 4-Et, 4-i-Pr, 4- OMe, 4-CF 3 , 4-CN or 4-OH. In yet another embodiment, R 4 is 3-Cl, 3-F, 3-Me, 3-Et, 3-i-Pr, 3- OMe, 3-CF 3 , 3-CN or 3-OH.

[00174] In one particular embodiment, with respect to the compounds of formulae I-III, n l is 1 ; R 4 is 4-NMe 2 . In another particular embodiment, R 4 is 4-Me. In another particular embodiment, R 4 is 2-Me. In another particular embodiment, R 4 is 4-OMe. In another particular embodiment, R 4 is 4-OEt.

[00175] In one particular embodiment, with respect to the compounds of formulae I-III, two adjacent R 4 groups joined together to form -0-CH 2 -0-, -0-CF 2 -0-, -O-CH2-CH2-O-, O- CH 2 -CH 2 -CH 2 -0-, or -CH=CH-CH=CH-.

[00176] In one particular embodiment, with respect to the compounds of formulae I-III, the group

[00177] In another particular embodiment, with respect to the compounds of formulae I-

III, the group

[00178] In another particular embodiment, with respect to the compounds of formulae I-

III,, the group

[00179] In another particular embodiment, with respect to the compounds of formulae I-

III the group

wherein the naphthyl is unsubstituted or substituted with one or more R 4 groups.

[00180] In one embodiment, with respect to the compounds of formula I-III, Cy or the group

is substituted or unsubstituted

or

[001811 In one embodiment, with respect to the compounds of formula III, when m is 1 and A is N, the N is substituted H or R 5a and the R 5a is as described herein. In one particular embodiment, A is is N-R 5a , and R 5a is alkyl, substituted alkyl, acyl, haloalkyl, hydroxyalkyl, or alkoxyalkyl. In one embodiment, R 5a is methoxyalkyl.

[00182] In one embodiment, with respect to the compounds of formula I, the compound is according to formula IVa, IVb, IVc, IVd, IVe, or IVf:

and wherein n 1 , R 2 , R 3 , R 4 , R 5 , and R 6 are as described for formula I; t and R 5a are as described for formula III.

[00183] In one embodiment, with respect to the formula IV f, NH is N-R 5a , and R 5a is alkyl, substituted alkyl, acyl, haloalkyi, hydroxyalkyl, or alkoxyalkyl. In one embodiment, R 5a is methoxyalkyl.

[00184] In one embodiment, with respect to the compounds of formulae I-IVf, n l is 1 , 2 or

3. In another embodiment, n l is 3. In a particular embodiment, n l is 1 .

[00185] In one embodiment, with respect to the compounds of formulae I-IVf, each R 3 is independently selected from halo, amino, substituted amino, substituted or unsubstituted C1-C6 alkyl, CN, OH, and substituted or unsubstituted C1-C6 alkoxy.

[00186] In another embodiment, with respect to the compounds of formulae I-IVf, each R 3 is independently selected from halo, substituted or unsubstituted Ci-C6 alkyl, CN, OH, and substituted or unsubstituted C |-C 6 alkoxy. [00187] . In one particular embodiment, with respect to the compounds of formulae I-IVf, n l ,is 1 ; and R 3 is NMe 2 .

[00188] In another particular embodiment, with respect to the compounds of formulae I-

IVf, nl is 1 ; and R 3 is CI, F, Me, Et, i-Pr, OMe, CF 3 , CN or OH. In one embodiment R 3 is at 4- or para- position of the phenyl ring. In another embodiment, R 3 is 4-Cl, 4-F, 4-Me, 4-Et, 4-i-Pr, 4- OMe, 4-CF 3 , 4-CN or 4-OH. In yet another embodiment, R 3 is 3-Cl, 3-F, 3-Me, 3-Et, 3-i-Pr, 3- OMe, 3-CF 3 , 3-CN or 3-OH.

[00189] In one particular embodiment, with respect to the compounds of formulae I-IVf, n l is 1 ; R 3 is 4-NMe 2 . In another particular embodiment, R 3 is 4-Me. In another particular embodiment, R 3 is 2-Me. In another particular embodiment, R 3 is 4-OMe. In another particular embodiment, R 3 is 4-OEt.

[00190] In one particular embodiment, with respect to the compounds of formulae I-IVf, two adjacent R 3 groups joined together to form -0-CH 2 -0-, -0-CF 2 -0-, -0-CH 2 -CH 2 -0-, O- CH 2 -CH 2 -CH 2 -0-, or -CH=CH-CH=CH-.

[00191] In one particular embodiment, with respect to the compounds of formulae I-IVf, the group

[00192] In another particular embodiment, with respect to the compounds of formulae I-

IVf, the group

[00193] In another particular embodiment, with respect to the compounds of formulae I-

IVf, the group

[00194] . In another particular embodiment, with respect to the compounds of formulae I-

IVf, the roup

wherein the naphthyl is unsubstituted or substituted with one or more R groups.

[00195] In one embodiment, with respect to the compounds of formula I-III, the compound is according to fo

and wherein R 2 , R 5 , and R 6 are as described for formula I; t and R 5a are as described for formula III; and n3 is 1 , 2, or 3. [00196] . In one embodiment, with respect to the formula Vf, NH is N-R 3a , and R 3a is alkyl, substituted alkyl, haloalkyl, hydroxyalkyl, or methoxyalkyl.

[00197] In one embodiment, with respect to the compounds of formulae I-Vf, R 2 is H, OH, substituted or unsubstituted Ci-C 6 alkyl, or substituted or unsubstituted phenyl.

[[0000119988]] I 1n another embodiment, with respect to the compounds of formulae I-Vf, R 2 is H, Me, OH, or Ph.

[00199] In one particular embodiment, with respect to the compounds of formulae I-Vf, R is H.

[00200] In another particular embodiment, with respect to the compounds of formulae I-

Vf, R 2 is OH.

[00201] In another particular embodiment, with respect to the compounds of formulae III-

Vf, t is 0. In another embodiment t is 1 or 2.

[00202] In one embodiment, with respect to the compounds of formulae II I-Vf, t is 1 ; and

R 5a is OH, Ph, benzyl, or Me. In another embodiment, t is 1 ; and R 5a is Me, Et, n-Pr, or n-Bu. In one particular embodiment, t is 1 ; and R 5a is 3-Me, 3-Et, 3-n-Pr, or 3-n-Bu.

[00203] In one embodiment, with respect to the compounds of formulae III-Vf, t is 2; and each R 5a is independently OH, Ph, benzyl, or Me. In another embodiment, t is 2; and one R 5a is 3-

Me and the other is 5-Me. In another embodiment, t is 2; and one R 5a is 3-Me and the other is 3-

Me.

[00204] In one embodiment, with respect to the compounds of formulae III-Vf, R 5a is OH, or Me.

100205] In one particular embodiment, with respect to the compounds of formulae III-Vf,

R 5a is Me.

[00206] In one particular embodiment, with respect to the compounds of formulae III-Vf,

R 5a is Ph.

[00207] In one particular embodiment, with respect to the compounds of formulae III-Vf,

R 5a is benzyl.

[00208] In another particular embodiment, with respect to the compounds of formulae III-

Vf, R 5a is 3-Me.

[00209] In one particular embodiment, with respect to the compounds of formulae I-Vf,

R 5 , when present, is H. In another embodiment, R 5 , when present, is Me or Et.

[00210] In one particular embodiment, with respect to the compounds of formulae I-Vf,

R 5 , when present, is H. In another embodiment, R 5 , when present, is Me, Et, n-Pr, or n-Bu. In yet another embodiment, R 5 , when present, is n-pentyl, n-hexyl, or n-heptyl.

[00211] In one embodiment, with respect to the compounds of formulae I-Vf, n3 is 1. [00212] In one embodiment, with respect to the compounds of formulae I-Vf, n3 is 2.

[0Q213] In one embodiment, with respect to the compounds of formulae I-Vf, n3 is 3.

[00214] In one particular embodiment, with respect to the compounds of formula I-II1, the compound is accordin to formula Via, VIb, VIc, VId, Vie, or Vlf:

and wherein R 5b is H or Me;

or a pharmaceutically acceptable salt, solvate or prodrug thereof;

and stereoisomers, isotopic variants and tautomers thereof.

[00215] In one particular embodiment, with respect to the compounds of formulae Vla-

Vlf, R 5b is H. In another embodiment, R 5b is Me.

[00216] In another particular embodiment, with respect to the compounds of formula I, the compound is accordin to formula Vila, Vllb, or VIIc:

or a pharmaceutically acceptable salt, solvate or prodrug thereof;

and stereoisomers, isotopic variants and tautomers thereof.

[00217] In one particular embodiment, with respect to the compounds of formula I-III, the compound is according to formula Villa VHIb, VIIIc, Vllld, or VHIe:

Villa

Vlllc Vllld llle

and wherein R is H or Me;

or a pharmaceutically acceptable salt, solvate or prodrug thereof;

and stereoisomers, isotopic variants and tautomers thereof.

[00218] In one particular embodiment, with respect to the compounds of formula I-III, the compound is according to formula IXa IXb, or IXc:

IXa IXb IXc

and wherein R is H or e;

or a pharmaceutically acceptable salt, solvate or prodrug thereof;

and stereoisomers, isotopic variants and tautomers thereof.

[00219] In one particular embodiment, with respect to the compounds of formula I, the compound is any one of the compounds listed in Table 1.

[00220) In one particular embodiment, with respect to the compounds of formula I, the compound is any one of compounds listed in Table 1 with Compound ID 7, 8, 22, 41 , 62, 63, 65, 70, 72, 73, 74, 75, 76, 1 18, 121 , 134, 135, 136, 138, or 149. [002211 . - In another aspect, the present invention provides pharmaceutical composition of a compound according to formulae I-IXc.

[002221 In one embodiment, with respect to the method of treatment, the disease or condition is autoimmune disease.

[00223] In one embodiment, with respect to the method of treatment, the disease or condition is inflammatory disease.

[00224] In one embodiment, with respect to the method of treatment, the disease or condition is selected from arthritis, diabetes, multiple sclerosis (and the animal model thereof, EAE), uveitis, rheumatoid arthritis (and the animal model thereof, CIA), psoriasis, asthma, bronchitis, allergic rhinitis, chronic obstructive pulmonary disease, atherosclerosis, cancer, graft- versus-host disease, H. pylori infections and ulcers resulting from such infection, and inflammatory bowel diseases.

[00225] In one embodiment, with respect to the method of treatment, the disease or condition is selected from Crohn's disease, ulcerative colitis, sprue and food allergies.

[00226] In certain aspects, the present invention provides prodrugs and derivatives of the compounds according to the formulae above. Prodrugs are derivatives of the compounds of the invention, which have metabolically cleavable groups and become by solvolysis or under physiological conditions the compounds of the invention, which are pharmaceutically active, in vivo. Such examples include, but are not limited to, choline ester derivatives and the like, N- alkylmorpholine esters and the like.

[00227] Other derivatives of the compounds of this invention have activity in both their acid and acid derivative forms, but the acid sensitive form often offers advantages of solubility, tissue compatibility, or delayed release in the mammalian organism (see, Bundgard, Η., Design of Prodrugs, pp. 7-9, 21 -24, Elsevier, Amsterdam 1985). Prodrugs include acid derivatives well know to practitioners of the art, such as, for example, esters prepared by reaction of the parent acid with a suitable alcohol, or amides prepared by reaction of the parent acid compound with a substituted or unsubstituted amine, or acid anhydrides, or mixed anhydrides. Simple aliphatic or aromatic esters, amides and anhydrides derived from acidic groups pendant on the compounds of this invention are preferred prodrugs. In some cases it is desirable to prepare double ester type prodrugs such as (acyloxy)alkyl esters or ((alkoxycarbonyl)oxy)alkylesters. Preferred are the Ci to C 8 alkyl, C 2 -C 8 alkenyl, aryl, C 7 -Ci 2 substituted aryl, and C 7 -Ci 2 arylalkyl esters of the compounds of the invention.

PHARMACEUTICAL COMPOSITIONS [00228] " When employed as pharmaceuticals, the compounds of this invention are typically administered in the form of a pharmaceutical composition. Such compositions can be prepared in a manner well known in the pharmaceutical art and comprise at least one active compound.

[00229] Generally, the compounds of this invention are administered in a pharmaceutically effective amount. The amount of the compound actually administered will typically be determined by a physician, in the light of the relevant circumstances, including the condition to be treated, the chosen route of administration, the actual compound -administered, the age, weight, and response of the individual patient, the severity of the patient's symptoms, and the like.

[00230] The pharmaceutical compositions of this invention can be administered by a variety of routes including oral, rectal, transdermal, subcutaneous, intravenous, intramuscular, and intranasal. Depending on the intended route of delivery, the compounds of this invention are preferably formulated as either injectable or oral compositions or as salves, as lotions or as patches all for transdermal administration.

[00231] The compositions for oral administration can take the form of bulk liquid solutions or suspensions, or bulk powders. More commonly, however, the compositions are presented in unit dosage forms to facilitate accurate dosing. The term "unit dosage forms" refers to physically discrete units suitable as unitary dosages for human subjects and other mammals, each unit containing a predetermined quantity of active material calculated to produce the desired therapeutic effect, in association with a suitable pharmaceutical excipient. Typical unit dosage forms include prefilled, premeasured ampules or syringes of the liquid compositions or pills, tablets, capsules or the like in the case of solid compositions. In such compositions, the furansulfonic acid compound is usually a minor component (from about 0.1 to about 50% by weight or preferably from about 1 to about 40% by weight) with the remainder being various vehicles or carriers and processing aids helpful for forming the desired dosing form.

[00232] Liquid forms suitable for oral administration may include a suitable aqueous or nonaqueous vehicle with buffers, suspending and dispensing agents, colorants, flavors and the like. Solid forms may include, for example, any of the following ingredients, or compounds of a similar nature: a binder such as microcrystalline cellulose, gum tragacanth or gelatin; an excipient such as starch or lactose, a disintegrating agent such as alginic acid, Primogel, or corn starch; a lubricant such as magnesium stearate; a glidant such as colloidal silicon dioxide; a sweetening agent such as sucrose or saccharin; or a flavoring agent such as peppermint, methyl salicylate, or orange flavoring.

[00233] Injectable compositions are typically based upon injectable sterile saline or phosphate-buffered saline or other injectable carriers known in the art. As before, the active compound in such compositions is typically a minor component, often being from about 0.05 to

10% by weight with the remainder being the injectable carrier and the like.

[00234] . Transdermal compositions are typically formulated as a topical ointment or cream containing the active ingredient(s), generally in an amount ranging from about 0.01 to about 20% by weight, preferably from about 0.1 to about 20% by weight, preferably from about 0.1 to about

10% by weight, and more preferably from about 0.5 to about 15% by weight. When formulated as a ointment, the active ingredients will typically be combined with either a paraffinic or a water-miscible ointment base. Alternatively, the active ingredients may be formulated in a cream with, for example an oil-in-water cream base. Such transdermal formulations are well-known in the art and generally include additional ingredients to enhance the dermal penetration of stability of the active ingredients or the formulation. All such known transdermal formulations and ingredients are included within the scope of this invention.

[00235] The compounds of this invention can also be administered by a transdermal device. Accordingly, transdermal administration can be accomplished using a patch either of the reservoir or porous membrane type, or of a solid matrix variety.

[00236] The above-described components for orally administrable, injectable or topically administrable compositions are merely representative. Other materials as well as processing techniques and the like are set forth in Part 8 of Remington's Pharmaceutical Sciences, 17th edition, 1985, Mack Publishing Company, Easton, Pennsylvania, which is incorporated herein by reference.

[00237] The compounds of this invention can also be administered in sustained release forms or from sustained release drug delivery systems. A description of representative sustained release materials can be found in Remington's Pharmaceutical Sciences.

[00238] The following formulation examples illustrate representative pharmaceutical compositions of this invention. The present invention, however, is not limited to the following pharmaceutical compositions.

Formulation 1 - Tablets

[00239] A compound of the invention is admixed as a dry powder with a dry gelatin binder in an approximate 1 :2 weight ratio. A minor amount of magnesium stearate is added as a lubricant. The mixture is formed into 240-270 mg tablets (80-90 mg of active amide compound per tablet) in a tablet press.

Formulation 2 - Capsules

[00240] A compound of the invention is admixed as a dry powder with a starch diluent in an approximate 1 : 1 weight ratio. The mixture is filled into 250 mg capsules ( 125 mg of active amide compound per capsule). Formulation 3 - Liquid

[00241] ' A compound of the invention (125 mg), sucrose (1 .75 g) and xanthan gum (4 mg) are blended, passed through a No. 10 mesh U.S. sieve, and then mixed with a previously made solution of microcrystalline cellulose and sodium carboxymethyl cellulose ( 1 1 :89, 50 mg) in water. Sodium benzoate (10 mg), flavor, and color are diluted with water and added with stirring. Sufficient water is then added to produce a total volume of 5 mL.

Formulation 4 - Tablets

[00242] A compound of the invention is admixed as a dry powder with a dry gelatin binder in an approximate 1 :2 weight ratio. A minor amount of magnesium stearate is added as a lubricant. The mixture is formed into 450-900 mg tablets (150-300 mg of active amide compound) in a tablet press.

Formulation 5 - Injection

[00243] A compound of the invention is dissolved or suspended in a buffered sterile saline injectable aqueous medium to a concentration of approximately 5 mg/ml.

Formulation 6 - Topical

[00244] Stearyl alcohol (250 g) and a white petrolatum (250 g) are melted at about 75°C and then a mixture of a compound of the invention (50 g) methylparaben (0.25 g), propylparaben (0.15 g), sodium lauryl sulfate (10 g), and propylene glycol (120 g) dissolved in water (about 370 g) is added and the resulting mixture is stirred until it congeals.

METHODS OF TREATMENT

[00245] The present compounds are used as therapeutic agents for the treatment of conditions in mammals that are causally related or attributable to RORyt activity. Accordingly, the compounds and pharmaceutical compositions of this invention find use as therapeutics for preventing and/or treating a variety of inflammatory conditions and autoimmune disorders in mammals, including humans.

[00246] In a method of treatment aspect, this invention provides a method of treating ' a mammal susceptible to or afflicted with a condition associated with an inflammatory condition and/or an autoimmune disorder, which method comprises administering an effective amount of one or more of the pharmaceutical compositions just described.

[00247] In additional method of treatment aspects, this invention provides methods of treating a mammal susceptible to or afflicted with an inflammatory condition or autoimmune disorder causally related or attributable to RORyt activity. Such condition and disorders include, without limitation, arthritis, diabetes, multiple sclerosis (and the animal model thereof, EAE), uveitis, rheumatoid arthritis (and the animal model thereof, CIA), psoriasis, asthma, bronchitis, allergic rhinitis, chronic obstructive pulmonary disease, atherosclerosis, cancer, graft-versus-host disease, H. ylori infections and ulcers resulting from such infection, and inflammatory bowel diseases. Such methods comprise administering an effective condition-treating or condition- preventing amount of one or more of the pharmaceutical compositions just described.

[00248] The present inventors have shown that treatment of wildtype cells with RORyt inhibitor digoxin resulted in changes in gene expression that were very similar to those observed in RORyt-deficient cells. See Huh et al. (201 1 ) Digoxin and its derivatives suppress Thl 7 cell differentiation by antagonizing RORyt activity; Nature, in press, the entire contents of which is incorporated herein by reference. That being the case, RORyt inhibitors can be used to treat any diseases caused by RORy or RORyt expressing cells, including Th l 7, NK22, and other innate lymphoid cells. The pro-atherogenic contribution of IL- 17 to atherosclerotic lesions, for example, suggests that atherosclerosis can be treated efficaciously with the compounds and compositions described herein. See Chen et al. J Innate Immun. 2010;2(4):325-33. Epub 2010 May 7, the entire contents of which is incorporated herein by reference.

[00249] As a further aspect of the invention there is provided the present compounds for use as a pharmaceutical especially in the treatment or prevention of the aforementioned conditions and diseases. Also provided herein is the use of the present compounds in the manufacture of a medicament for the treatment or prevention of one of the aforementioned conditions and diseases.

[00250] ' Injection dose levels range from about 0.1 mg/kg/hour to at least 10 mg/kg/hour, all for from about 1 to about 120 hours and especially 24 to 96 hours. A preloading bolus of from about 0.1 mg/kg to about 10 mg/kg or more may also be administered to achieve adequate steady state levels. The maximum total dose is not expected to exceed about 2 g/day for a 40 to 80 kg human patient.

[00251] For the prevention and/or treatment of long-term conditions, such as, e.g., arthritis, diabetes, multiple sclerosis (and the animal model thereof, EAE), rheumatoid arthritis (and the animal model thereof, CIA), psoriasis, or asthma, the regimen for treatment usually stretches over many months or years, so oral dosing is preferred for patient convenience and tolerance. With oral dosing, one to five and especially two to four and typically three oral doses per day are representative regimens. Using these dosing patterns, each dose provides from about 0.01 to about 20 mg/kg of the compound of the invention, with preferred doses each providing from about 0.1 to about 10 mg/kg and especially about 1 to about 5 mg/kg.

[00252] Transdermal doses are generally selected to provide similar or lower blood levels than are achieved using injection doses. Modes of administration suitable for mucosal sites are also envisioned herein and include without limitation: intra-anal swabs, enemas, intranasal sprays, and aerosolized or vaporized compounds and/or compositions for delivery to the lung mucosa. One of skill in the art would choose an appropriate delivery mode/s based on a variety of parameters, including the organ or tissue site in a patient with a disease or condition that is most severely affected by the disease or condition. A skilled practitioner could, for example, treat a patient afflicted with an inflammatory bowel disease (IBD) with a therapeutic regimen that included delivery of the compounds or compositions of the invention using an enema for direct delivery to the bowel.

100253] When used to prevent the onset of an inflammatory condition or autoimmune disorder, the compounds of this invention will be administered to a patient at risk for developing the condition or disorder, typically on the advice and under the supervision of a physician, at the dosage levels described above. Patients at risk for developing a particular condition generally include those that have a family history of the condition, or those who have been identified by genetic testing or screening to be particularly susceptible to developing the condition.

[00254] The compounds of this invention can be administered as the sole active agent or they can be administered in combination with other agents, including other compounds that demonstrate the same or a similar therapeutic activity and are determined to safe and efficacious for such combined administration.

GENERAL SYNTHETIC PROCEDURES

[00255] The amido compounds of this invention may be purchased from various commercial sources or can be prepared from readily available starting materials using the following general methods and procedures. It will be appreciated that where typical or preferred process conditions (i.e., reaction temperatures, times, mole ratios of reactants, solvents, pressures, etc.) are given, other process conditions can also be used unless otherwise stated. Optimum reaction conditions may vary with the particular reactants or solvent used, but such conditions can be determined by one skilled in the art by routine optimization procedures.

[00256] Additionally, as will be apparent to those skilled in the art, conventional protecting groups may be necessary to prevent certain functional groups from undergoing undesired reactions. The choice of a suitable protecting group for a particular functional group as well as suitable conditions for protection and deprotection are well known in the art. For example, numerous protecting groups, and their introduction and removal, are described in T. W. Greene and P. G. . Wuts, Protecting Groups in Organic Synthesis, Second Edition, Wiley, New York, 1991 , and references cited therein.

[00257] The compounds of the invention may be prepared from known or commercially available starting materials and reagents by one skilled in the art of organic synthesis.

General Materials and Methods: [00258] ' All commercially available reagents and solvents were purchased and used without further purification. All microwave reactions were carried out in a sealed microwave vial equipped with a magnetic stir bar and heated in a Biotage Initiator Microwave Synthesizer.

HPLC purification was performed using a Waters semi-preparative HPLC equipped with a

Phenomenex Luna® CI 8 reverse phase (5 micron, 30 x 75 mm) column (unless state otherwise) having a flow rate of 45 mL/min. The mobile phase was a mixture of acetonitrile and H 2 0 each containing 0.1 % trifluoroacetic acid. Ή spectra were recorded using either an Inova 400 MHz spectrometer (Varian) or an Inova 300 MHz spectrometer (Varian). Two LCMS methods were used to analyze samples' purity. Method 1 : Agilent 1200 series LC MS equipped with a

Zorbax™ Eclipse XDB-C 18 reverse phase (5 micron, 4.6 x 150 mm) column having a flow rate of 1 .1 mL/min. The mobile phase was a mixture of acetonitrile and H 2 0 each containing 0.05% trifluoroacetic acid. A gradient of 5% to 100% acetonitrile over 8 minutes was used during analytical analysis. Method 2: Acquity HPLC equipped with a Waters BEH CI 8, 1.7 micron, 2.1 x 50 mm column; Column Temperature: 45 degrees C; Flow: 0.5mL/min; Solvent A: 0.05% TFA in Water; Solvent B: 0.025% TFA in Acetonitrile; Gradient: 2% to 100% Solvent B over 1 .3 minutes; Run Time- 3 min. High-resolution mass spectroscopy measurements were performed on a Agilent 6210 Electrospray TOF.mass spectrometer.

[00259] The following general procedures were used to synthesize compounds having different but analogous structures. One skilled in the art of synthesis will recognize how to modify these general procedures if necessary to accomplish the desired transformations.

Representative Synthetic Methods Method A

[00260] The representati ve hydroxy compounds of the invention can be prepared using the general synthetic pathway depicted in Scheme 1.

Scheme 1

The following reaction conditions were used for step 1:

(1 ) TFA, 60-100 °C, 2-16 h

or (2) p-toluenesulfonic acid, 125 °C, 2h

The following reaction conditions were used for step 2:

(1 ) R 5 R 6 NH, 60-80 °C, DMA or THF, 2-16 h

or (2) R 5 R 6 NH, Me^AI, Toluene/DCE,50 °C, 4-12 h

Step 1 :

[00261] A mixture of phenol (1, 0.2 mmol) and cinnamic acid (2, 0.2 mmol) in TFA (2 mL) was heated at 60- 100 °C for 2-16 h. Upon completion, TFA was removed using a Genevac evaporator and the residue was dissolved in DCM (2 mL), washed with saturated NaHC0 3 (2 mL). After removing solvent, 3 was obtained. The compound 3 may be prepared using the following procedure.

[00262] A mixture of (1, 0.2 mmol), cinnamic acid (2, 0.2 mmol) and p-toluenesulfonic acid (0.2 mmol) was heated at 125 °C for 2 h. The reaction mixture was cooled to room temperature and dissolved in DCM (2 mL), washed with NaOH ( 1 M, 1 mL). After removing solvent, 3 was obtained and used for next step without further purification.

Step 2:

[00263] A mixture of 3 (0.2 mmol) and amine (R 5 R 6 NH, 0.3 mmol) in DMA or THF (2 mL) was heated at 60-80 °C for 2-16 h. Upon completion, the mixture was cooled to room temperature. The desired product 4 was obtained by HPLC purification. The compound 4 may be prepared using the following procedure.

[00264] To an 8 mL-vial, amine (R5R6NH, 0.3 mmol) was added. The vial was capped with a Teflon cap, and then Me 3 Al (0.4 mL, 1M in heptane) was added. The resulting mixture was shaken at room temperature for 1 h. This was followed by addition of 3 (0.2 mmol, dissolved in 1 mL dry toluene/or DCE). The resulting mixture was heated at 50 °C for 12 h. The reaction mixture was cooled to room temperature and concentrated in a GeneVac. The residue was dissolved in DCM (2 mL), and then Na 2 SO 4 0H 2 O (0.2 mmol, 32 mg) was added. The suspension was vortexed and centrifuged, the clean solution was separated and concentrated. The crude product was purified by HPLC to afford 4.

Method B

Reaction conditions: (i) BF,EtjO, 0 °C - r.L 1-2 h; (ii) UOH, THF/H^/MeOH, 150 °C, 1 h; (ii) DMC, W . Hunig base.

General procedure

[00265] To a solution of 5 (l mmol) and ( l -methoxy-2-methylprop- l - enyloxy)trimethylsilane (1.5 mmol) in 10 mL of DCM at 0 °C was added BF 3 Et 2 0 ( 1.5 mmol) with stirring. The mixture was warmed to r.t. and stirring was continued for 1 -2 h. Upon completion, 20 mL DCM was added and the organic layer was washed with sat Na 2 C0 3 solution (10 mL) and water (10 mL), dried over MgS0 4 and concentrated. The residue was purified by column chromatography over silica-gel to give 6. A solution of 6 (0.5 mmol) and LiOH (5 mmol) in a mixture of solvents (10 mL THF, 5 mL MeOH and 5 mL water) was heated in a microwave at 150 °C for 1 h. Upon completion, the solvent was removed. The residue was dissolved in 10 mL of water and the solution was acidified by 1 M HC1 to pH = 2. The resulting mixture was extracted with DCM (3 x 30 mL). The combined organic layers was dried over MgS0 , filtered and concentrated to give acid 7. To a solution of acid 7 (0.3 mmol) and Hunig base (1 mmol) in DCM (10 mL) was added 2-chloro-l ,3-dimethylimidazolinium chloride (1.5 mmol) at room temperature. After stirring for 10 min, a solution of amine (0.6 mmol Κ 3 ¾ΝΗ in DCM or DMA) was added. The resulted mixture was stirred for 4- 12 h. After concentrated, the residue was purified by HPLC to yield the desired 8.

Method C

General Procedure:

[00266] Compound 2 was either used without purification from commercial sources or synthesized following the literature procedure (Xie, L. et al;. J. Med. Chem. 1999, 42, 2662.)

[00267] A solution of 2 (1 mmol) in CH 2 C1 2 (0.9 rtiL) was treated with phenol 1 (1 n-imol), TFA (2.7 mL) and Pd(OAc) 2 (15 mg, 0.069 mmol), and stirred at 40 °C for 8 h. The reaction mixture was concentrated to dryness using the Genevac evaporator and used crude in the next step. * Crude 9 was dissolved in MeOH (1 mL), treated with R5R6NH (2 mmol), 10% Pd/C (8 mg, 0. 1 mmol) and ammonium formate (67 mg, 1 mmol) and heated to 50 °C for 8 h. The reaction mixture was diluted with MeOH, filtered through a solid supported thiol column, and concentrated under reduced pressure. The material was purified by HPLC to yield compound 4.

[00268] * The degree of oxidation to 9 was substrate dependent and sometimes the primary product was lactone 3 as determined by LCMS. If the level of oxidation was < 20%, amide 4 was synthesized following the same conditions described in Method A.

Method D

10 11

Reaction Conditions: (i) Pd(OAc) 2 , BIPY, AcOH, H 2 0, THF, 60 °C, 8 h; (ii) R 5 R 6 NH, DMA, 80 °C, 8 h General Procedure

[00269] A suspension of coumarin 10 (0.15 mmol), boronic acid 11 (0.4 mmol) BIPY ( 10 mg, 0.064 mmol), and Pd(OAc) 2 (7 mg, 0.03 mmol) in AcOH (0.6 mL), THF (0.2 mL) and H 2 0 (0.1 mL) was heated to 60 ° C for 8 h and concentrated to dryness in the Genevac evaporator to afford 3 which was used without purification in the following step. Crude lactone 3 was converted to amide 4 following the same protocol as Method A (NHR5R6, DMA, 80 °C) and purified with HPLC to afford product 4.

Method E

2 11 12 13

Reaction Conditions: (i) Pd(OAc) 2 , BIPY, AcOH, H 2 0, THF, 60 °C, 8 h; (ii) R 5 R 6 NH, 2,3,4,6,7,8-hexahydro-1 H- pyrimido[1 ,2-a]pyrimidine, 70 °C, 8 ' h

General Procedure:

[00270] A suspension of cinnamic ester 2 (0.15 mmol), boronic acid 11 (0.4 mmol) BIPY

( 10 mg, 0.064 mmol), and Pd(OAc) 2 (7 mg, 0.03 mmol) in AcOH (0.6 mL), THF (0.2 mL) and H 2 0 (0.1 mL) was heated to 60° C for 8 h and concentrated to dryness in the Genevac evaporator to afford 12 which was used without purification in the following step. Crude 12 was treated with 2, 3,4,6,7, 8-hexahydro- l H-pyrimido[ l ,2-a]pyrimidine (0.18 mmol) and R5R6NH and heated to 70 °C for 8 h. The reaction mixture was purified by HPLC to afford 13.

4 14

General Procedure:

[00271] A mixture of 4 (0.1 mmol) and NaH (0.2 mmol) was dissolved in dry THF (2 mL). After stirring at room temperature for 0.5 h, alkyl halide (0.4 mmol) was added. The resulting mixture was heated at 60 °C for 2-8 h. The solution was cooled to room temperature. A couple drops of water were added and the mixture was purified by HPLC to give compound 14.

Representative Examples:

Compound 35 (Method A)

[00272] (-) 3-(Benzo[d][ l ,3]dioxol-5-yl)-l -c5-3,5-dimethylpiperidin-l -yl)-3-(2-hydroxy-

4,6-dimethoxyphenyl)propan- l -one (4k(-)) (NCGC00238427, 35)

4k(-) 4k(+)

(i) TFA, 80 °C 6 ; (ii) Hunig base, 3,5-c s-dimethylpiperidine, 60 °C DMA, 16 h; (iii) chiral separation by HPLC

[00273] Prepared following method A: A mixture of 3,5-dimethylphenol (la, 10.0 mmol,

1.54 g) and 3,4-(methylenedioxy)cinnamic acid (2a, 10.0 mmol, 1.92 g) in TFA (30 mL) was heated at 70 °C for 3 h. After removing TFA, the residue was purified by silica-gel column chromatography using hexane/ethyl acetate (7-60%) to give 3a (2.13 g, 65%) as a white powder. 'H NMR (400 MHZ, DMSO-i/ 6 ) δ ppm 2.83 (dd ; J=15.8, 1.6 Hz, 1 H), 3.20 (dd, J=15.9, 6.9 Hz, 1 H), 3.74 (s, 3 H), 3.79 (s, 3 H), 4.43 (d, J=5.9 Hz, 1 H), 5.96 (s, 2 H), 6.33 - 6.50 (m, 3 H), 6.66 (d, J= 1.6 Hz, 1 H), 6.79 (d, J=8.0 Hz, 1 H); LC/MS: Retention time t = 5.78 min; Purity: UV 220 > 98%, UV 254 > 98%.

[00274] To a solution of 3a (1.25 1 g, 3.81 mmol) in 10.0 mL of DMA was added 3,5- dimethylpiperidine monotartrate salt (2.01 g, 7.62 mmol) and Hunig base (2.46 g, 19.0 mmol).

The reaction mixture was heated at 60 °C for overnight. The solvent was removed and the residue was dissolved in 50 mL of DCM. The solution was washed with sat. NaHC0 3 (30 mL x3). The organic layer was dried over MgS0 4 , filtered and concentrated. The crude product was purified by silica gel column chromatography eluting with 7-60% hexanes/ethyl acetate to afford amide

3-(Benzo[d][ l ,3]dioxol-5-yl)- l-c 5-3,5-dimethylpiperidin-l -yl)-3-(2-hydroxy-4,6- dimethoxyphenyl)propan- l -one (869 mg, 52%) as a solid. Chiral separation by HPLC (Column:

IA Preparatory 5 cm x 50 cm; Run Time: 40 minutes; Flow Rate: 35 mL/min; Mobile Phase:

60/40 EtOH/Hexane; Detectors: DAD (220 and 254 nm)) gave 4k(-) and 4k(+).

[00275] (-) 3-(Benzo[d][l ,3]dioxol-5-yl)- l -c 5-3,5-dimethylpiperidin-l -yl)-3-(2-hydroxy-

4,6-dimethoxyphenyl)propan- l -one (4k(-)): Ή NMR (400 MHz, DMSO-i 6, 60 °C) δ ppm 0.65-

0.74 (m, 1 H), 0.81 (d, 6H, J = 6.3 Hz), 1.20- 1.38 (m, 2H), 1.64-1.74 (m, 1 H), 1.80- 1.94 (m, 1 H),

2.34-2.50 (m, 1 H), 2.92-3.20 (m, 2H), 3.67 (s, 3H), 3.69 (s, 3H), 3.72-3.90 (m, 1 H), 4.28-4.38

(m, 1 H), 4.88 (t, 1 H, J = 7.2 Hz), 5.88-5.89 (m, 2H), 6.02-6.04 (m. 2H), 6.67-6.73 (m, 2H), 6.82

(s, 1 H), 9.21 (s, 1 H); LC/MS: Retention time t = 4.39 min; Purity: UV 220 > 95%, UV 254 > 95%; (Column: I A analytical, 0.46 cm x 25 cm; Run time: 15 min; Flow rate: 1 mL/min; Mobile phase;

60/40 EtOH/Hexane; Deterctors: DAD (220 and 254 nm) and PDR chiral detector); [a] D 23 = -

129.1 (c 1 .0, CHC1 3 ); HRMS (ESI): m/z calcd for C25H32NC 442.2224, found 442.2221.

Compound 149 (Method B)

[00276] l -(3,5-Dimethylpiperidin-l -yl)-2,2-dimethyl-3-phenyl-3-(2,4,6- trimethoxyphenyl)propan- l -one 149)

[00277] Prepared following method B: To a solution of 5a (0.50 g, 1.82 mmol) and (1 - methoxy-2-methylprop-l -enyloxy)trimethylsilane (0.477 g, 2.73 mmol) in 10 mL of DCM at 0 °C was added BF 3 Et 2 0 (0.388 g, 2.73 mmol) with stirring. The mixture was warmed to r.t. and stirring was continued for 2 h. Upon completion, 20 mL DCM was added and the organic layer was washed with sat Na 2 C0 3 solution ( 10 mL) and water ( 10 mL), dried over MgS0 4 and concentrated. The residue was purified by silica-gel column chromatography using 7- 100% DCM/Hexanes to give 6a (0.312 g, 48%). A solution of 6a (0.312g, 0.87 mmol) and LiOH (0.21 g, 8.70 mmol) in a mixture solvents (10 mL THF, 5 mL MeOH and 5 mL water) was heated in a microwave at 150 °C for 1 h. Upon completion, the solvent was removed. The residue was dissolved in 10 mL of water and the solution was acidified by 1 M HC1 to pH = 2. The resulting mixture was extracted with DCM (3 x 30 mL). The combined organic layers was dried over MgS0 4 , filtered and concentrated to give acid 7a (0.256 g, 85%). To a solution of acid 7a

(0.236, 0.685 mmol) and Hunig base (2.1 mmol) in DCM (15 mL) was added 2-chloro- l ,3- dimethylimidazolinium chloride (1 4 mg, 1.03 mmol) at room temperature. After stirred for 10 min, a solution of cis-3,5-dimethylpiperidine (1 16 mg, 1.03 mmol ) in 5 mL DMA was added. The resulted mixture was stirred for overnight. After concentrated, the residue was dissolved in 30 mL and washed with sat NaHC0 3 (15 mL x 3). The organic layer was dried over MgS0 , filtered and concentrated. The residue was purified by silica gel chromatography eluting with 7- 60% ethyl acetate/hexanes to afford 8a (256 mg, 85%) as a solid. Ή NMR (400 MHz, DMSO-i 6 , 80 °C) δ ppm 0.50 - 0.70 (m, 1 H), 0.77 (m, 6 H), 0.85 (m, 1 H), 0.99 (m, 1 H), 1 .09 (s, 3 H), 1.28 (s, 3 H), ' 1.60 (m, 1 H), 1.90 - 2.15 (m, 2 H), 3.58 (br. s., 6 H), 3.76 (s, 3 H), 4.17 - 4.38 (m,

2 H) 5.23 (s, 1 H); 6.21 (s, 2 H); 6.90 - 7.23 (m, 5 H); LCMS: (electrospray +ve), m/z 440.2

(MH) + ;HPLC: t R = 2.61 min, UV 254 = 90%; HRMS (ESI): m/z calcd for C 2 7H 3 7N04 + 440.2807, found 440.2805.

Compound 106 (Method C)

[00278] l -(3,5-Dimethylpiperidin-l -yl)-3-(2-hydroxy-4,6-dimethoxyphenyl)-3-(3,4,5- trifluorophenyl)propan-l -one (4p) (NCGC00242635, 109)

1a 2 P 9 P 4p

Reaction conditions: (i) TFA, CH 2 CI 2 Pd(OAc) 2 , 40 °C, 8 h; (ii) 3,5-dimethylpiperidine, MeOH, HC0 2 NH 4 , 10%

Pd/C, 50 °C, 8 h

[00279] Prepared following method C: A solution of (E)-methyl 3-(3,4,5- trifluorophenyl)acrylate (2p, 216 mg, 1 mmol) in CH 2 C1 2 (0.9 mL) was treated with phenol la ( 154 mg, 1 mmol), TFA (2.7 mL) and Pd(OAc) 2 (15 mg, 0.069 mmol), and stirred at 40 °C for 8 h. The reaction mixture was concentrated to dryness using the Genevac evaporator and used crude in the next step. Crude 9p was dissolved in MeOH (1 mL), treated with 3,5- dimethylpiperidine (2 mmol), 10% Pd/C (8 mg, 0.1 mmol) and ammonium formate (67 mg, 1 mmol) and heated to 50 °C for 8 h. The reaction mixture was diluted with MeOH, filtered through a solid supported thiol column, and concentrated under reduced pressure. The material was purified by HPLC to yield compound 4p. Ή NMR (400 MHz, DMSO-< 6 ) δ ppm 9.53 - 9.58 (m, 1 H) 6.99 - 7.09 (m, 2 H) 6.02-6.03 (m, 2 H) 4.88 - 4.95 (m, 1 H) 4.30 (d, J=12.32 Hz, 1 H) 3.73 - 3.84 (m, 1 H) 3.67 - 3.70 (m, 3 H) 3.66 (d, J=0.78 Hz, 3 H) 3.13 - 3.20 (m, 1 H) 3.05 (dd, J=15.36, 6.94 Hz, 1 H) 2.97 (dd, J=16.24, 6.85 Hz, 1 H) 2.36 - 2.46 (m, 1 H) 1.89 (t, J=12.03 Hz, 1 H) 1.63 - 1.76 (m, 1 H) 1 .19 - 1.29 (m, 2 H) 0.75 - 0.89 (m, 6 H); LCMS: (electrospray +ve), mlz 452.2 (MH) + ;HPLC: t R = 6.63 min, UV 254 = 99%.

Compound 87 (Method D)

[00280] 3-(3-(3,5-Dimethylpiperidin-l-yl)-l-(2-hydroxy-4,6-dimethoxy phenyl)-3- oxopropyl)benzonitrile (4n) (NCGC00242612, 87)

Reaction Conditions: (i) Pd(OAc) 2 . BIPY, AcOH, H 2 0, THF. 60 °C. 8 h; (ii) 3,5-dimethylpiperidine, DMA. 80 °C, 8 h

[00281] Prepared following method D: A suspension of coumarin 10a (30 mg, 0.15 mmol), boronic acid l ln (64 mg, 0.4 mmol) BIPY (10 mg, 0.064 mmol), and Pd(OAc) 2 (7 mg, 0.03 mmol) in AcOH (0.6 mL), THF (0.2 mL) and H 2 0 (0.1 mL) was heated to 60° C for 8 h and concentrated to dryness in the Genevac evaporator to afford 3 which was used without purification in the following step. Crude lactone 3n was dissolved in DMF (0.8 mL) and treated with 3,5-dimethylpiperidine (0.1 mL, 0.75 mmol) and heated to 80 °C for 2 h at which point the reaction was complete according to LCMS. The reaction mixture was diluted with MeOH (~2 mL), filtered through a solid supported thiol column and purified by HPLC to afford 4n. Ή NMR (400 MHz, DMSO-d 6 ) δ ppm 9.51 - 9.54 (m, 1 H) 7.60 (s, 1 H) 7.50 - 7.57 (m, 2 H) 7.40 (tt, J=7.70, 1 .59 Hz, 1 H) 6.03 (s, 1 H) 6.02 (s, 1 H) 4.99 (t, J=9.10 Hz, 1 H) 4.30 (d, J=12.52 Hz, 1 H) 3.77 (d, J= 17.61 Hz, 1 H) 3.68 (d, J=2.93 Hz, 3 H) 3.65 (d, J=0.98 Hz, 3 H) 3.23 (dd, J=l 5.94, 8.5 1 Hz, 1 H) 3.06 (dd, J=l 5.65, 6.26 Hz, 1 H) 2.93 (dd, J=l 5.26, 6.85 Hz, 1 H) 2.40 - 2.46 (m, 1 H) 1.89 (t, J=1 1.93 Hz, 1 H) 1.63 - 1 .75 (m, 1 H) 1.18 - 1 .31 (m, 2 H) 0.74 - 0.89 (m, 6 H); LCMS: (electrospray +ve), mlz 423.2 (MH) + ;HPLC: t R = 6.12 min, UV 254 = 90%.

Compound 141 (Method E)

[00282] 3-(4-(l H- l ,2,4-Triazol- l-yl)phenyl)-3-(benzo[d][l ,3]dioxol-5-yl)-l -(3,5- dimethylpiperi

11a 12m 13m

[00283] Prepared by method E: A suspension of cinnamic methyl ester (2m, 224 mg, 0.97 mmol), boronic acid 11a (315 mg, 1.89 mmol) BIPY (30 mg, 0.192 mmol), and Pd(OAc) 2 (30 mg, 0.134 mmol) in AcOH ( 1 .5 mL), THF (0.5 mL) and H 2 0 (0.25 mL) was heated to 60 ° C for 8 h and concentrated to dryness in the Genevac evaporator to afford 12m which was used without purification ' in the following step. The crude methyl ester was treated with 3,5-dimethylpiperidine

( 1.34 mL, 9.88 mmol) and 2,3,4,6,7,8-hexahydro- lH-pyrimido[l ,2-a]pyrimidine (0.134 g, 0.96 mmol) and heated to 70 °C for 8 h, the reaction mixture was purified by HPLC to afford the amide 13m. LCMS: (electrospray +ve), mlz 411.2 (MH) + ;HPLC: t R = 5.87 min, UV 254 = 99%.

Compound 120 (Method F)

[00284] 3-(Benzo[d][ 1 ,3]dioxol-5-yl)- 1 -(3-methylpiperidin- 1 -yl)-3-(2,4,6- trimethoxyphenyl)propan-l -one (14a) (NCGC00189204, 120)

4a 14a

[00285] Prepared by method F: To a solution of 4a (0.1 mmol, 43 mg) in dry THF (2 mL) was added NaH (0.2 mmol, 5 mg). After stirring at room temperature for 0.5 h, Mel (56 mg, 0.4 mmol) was added and the mixture was heated at 60 °C for 2 h. The solution was cooled to room temperature. A couple drops of water were added and the mixture was purified by HPLC to give compound 14a. LCMS: (electrospray +ve), mlz 442.4 (MH) + ; HPLC: t R = 2.35 min, UV 254 = 99%.

Compound 105 (Method D)

[00286] l -(3,5-Dimethylpiperidin- l-yl)-3-(2-hydroxy-4,6-dimethoxyphenyl)-3-(4- (methylthio)phenyl)propan- 1 -one

[00287] Prepared by method D: DMSO-rf 6 ) δ ppm 9.38 (d, J=3.13 Hz,

1 H) 7.13 - 7.20 (m, 2 H) 7.06 - 7.13 (m, 2 H) 5.98 - 6.03 (m, 2 H) 4.87 - 4.92 (m, 1 H) 4.31 (d, J=12.72 Hz, 1 H) 3.73 - 3.79 (m, 1 H) 3.67 (d, J=2.54 Hz, 3 H) 3.65 (s, 3 H) 3.24 - 3.28 (m, 1 H) 3.06 - 3.12 (m, 1 H) 2.92 (dd, J=15.36, 6.16 Hz, 1 H) 2.53 - 2.57 (m, 1 H) 2.40 (s, 3 H) 1.83 - 1.91 (m, 1 H) 1.62 - 1.72 (m, 1 H) 1.14 - 1.37 (m, 2 H) 0.73 - 0.85 (m, 6 H); LCMS:

(electrospray +ve), mlz 444.2 (MH) + ;HPLC: t R = 6.47 min, UV 254 = 99% Compound 83 (Method D)

100288] l -(3,5-Dimethylpiperidin- l -yl)-3-(2-hydroxy-4,6-dimethoxyphenyl)-3-(3- (methylsulfonyl)phenyI)propan- 1 -one

[002891 Prepared by method D: Ή NMR (400 MHz, OMSO-d 6 ) δ ppm 9.54 (d, J=9.19

Hz, 1 H) 7.77 (s, 1 H) 7.64 - 7.68 (m, 1 H) 7.51 - 7.56 (m, 1 H) 7.47 (t, J=7.73 Hz, 1 H) 5.99 - 6.06 (m, 2 H) 5.02 - 5.10 (m, 1 H) 4.31 (d, J=13.1 1 Hz, 1 H) 3.74 - 3.82 (m, 1 H) 3.67 (d, J=1.76 Hz, 3 H) 3.65 (s, 3 H) 3.35 - 3.47 (m, 1 H) 3.19 - 3.26 (m, 1 H) 3.12 - 3.15 (m, 3 H) 2.99 - 3.10 (m, 1 H) 2.83 - 2.95 (m, 1 H) 1.84 - 1.93 (m, 1 H) 1.63 - 1.75 (m, 1 H) 1 .13 - 1.35 (m, 2 H) 0.69 - 0.90 (m, 6 H); LCMS: (electrospray +ve), mlz 476.2 (MH) + ;HPLC: t R = 5.63 min, UV 254 = 99%

Compound 86 (Method D)

[00290] l -(3,5-Dimethylpiperidin-l-yl)-3-(2-hydroxy-4,6-dimethoxyphen yl)-3-(4- (trifluoromethyl)phenyl)propan- 1 -one

[00291] Prepared by method D: Ή NMR (400 MHz, DMSO-rf 6 ) δ ppm 9.47 (d, J=3.91

Hz, 1 H) 7:54 (d, J=6.65 Hz, 2 H) 7.38 - 7.44 (m, 2 H) 6.03 (s, 1 H) 6.02 (s, 1 H) 5.03 (t, J=7.14 Hz, 1 H) 4.31 (d, J=13.1 1 Hz, 1 H) 3.77 (d, J=12.32 Hz, 1 H) 3.67 (d, J=3. 13 Hz, 3 H) 3.65 (d, J=0.78 Hz, 3 H) 3.25 (dd, J=15.45, 8.22 Hz, 1 H) 3.02 - 3.10 (m, 1 H) 2.92 (dd, J= 15.06, 5.67 Hz, 1 H) 2.35 - 2.47 (m, 1 H) 1.89 (t, J=12.13 Hz, 1 H) 1.63 - 1.73 (m, 1 H) 1.16 - 1.35 (m, 2 H) 0.71 - 0.89 (m, 6 H); LCMS: (electrospray +ve), mlz 466.2 (MH) + ;HPLC: t R = 6.72 min, UV 254 = 99%

Compound 89 (Method D) [00292] 3-(4-(ert-ButyIphenyl)- 1 -(3,5-dimethylpiperidin- 1 -yl)-3-(2-hydroxy-4,6- dimethoxyphenyl)propan- 1 -one

[00293] Prepared by method D: Ή NMR (400 MHz, DMSO-</ 6 ) δ ppm 9.32 - 9.38 (m, 1 H)7.11 -7.21 (m, 4 H) 5.97 - 6.04 (m, 2 H) 4.89 (dd, J= 12.03, 7.92 Hz, 1 H)4.31 (d,J=14.48 Hz, 1 H) 3.75 (d, J=l 8.00 Hz, 1 H) 3.68 (d, J=4.89 Hz, 3 H) 3.64 - 3.66 (m, 3 H) 3.32 - 3.40 (m, 1 H) 3.11 (d,J=7.24 Hz, 1 H) 2.87 (dd, J=14.18, 7.14 Hz, 1 H) 2.36 - 2.45 (m, 1 H) 1.86 (td, J=12.28, 5.77 Hz, 1 H) 1.65 (t,J=13.21 Hz, 1 H) 1.27 - 1.38 (m, 2 H) 1.22 (d,J=1.76 Hz, 9 H) 0.71 - 0.84 (m, 6 H) LCMS: (electrospray +ve), mlz 454.3 (MH) + ;HPLC: t R = 7.10 min, UV 254 = 98%.

Compound 85 (Method D)

[00294] 3-(3-Bromophenyl)-l -(3,5-dimethylpiperidin- l-yl)-3-(2-hydroxy-4,6- dimethoxyphenyl)propan- 1 -one

[00295] Prepared by method D: Ή NMR (400 MHz, DMSO-c/ 6 ) δ ppm 9.47 - 9.51 (m, 1

H)7.38(ddd,J=9.63, 1.71, 1.57 Hz, 1 H) 7.27 (dt,J=7.73, 1.03 Hz, 1 H) 7.19 -7.23 (m, 1 H) 7.16(d,J=7.83 Hz, 1 H) 6.03 (t, J- 2.18 Hz, 1 H) 6.02 (t,J=2.18Hz, 1 H)4.91 -4.96 (m, 1 H) 4.27- 4.35 (m, 1 H) 3.75 (d,J=15.65 Hz, 1 H) 3.68 (d, J=4.30 Hz, 3 H) 3.66 (s, 3 H) 3.17 (dd, J= 14.97, 6.55 Hz, 1 H) 2.95 - 3.09 (m, 1 H) 2.75 - 2.88 (m, 1 H) 2.37 - 2.47 (m, 1 H) 1.88 (t, J=12.32 Hz, 1 H) 1.62 - 1.75 (m, 1 H) 1.24 (s, 2 H) 0.74 - 0.86 (m, 6 H); LCMS: (electrospray +ve), mlz 478.1 (MH) + ;HPLC: t R = 6.66 min, UV 254 = 90%.

Compound 93 (Method D)

[00296] l-(3,5-Dimethylpiperidin-l-yl)-3-(2-hydroxy-4,6-dimethoxyphe nyl)-3-(3- (trifluoromethoxy)phenyl)propan- 1 -one

[002971 Prepared by method D: Ή NMR (400 MHz, DMSO-rf 6 ) δ ppm 9.50 (d, J=4.1 1

Hz, 1 H) 7.31 (t, J=7.83 Hz, 1 H) 7.22 (dd, J=8.12, 3.23 Hz, 1 H) 7.17 (br. s., 1 H) 7.06 (dd, J=8.12, 1 .27 Hz, 1 H) 6.00 - 6.03 (m, 2 H) 4.96 - 5.02 (m, 1 H) 4.31 (d, J=12.32 Hz, 1 H) 3.76 (d, J=14.28 Hz, 1 H) 3.67 (d, J=3.52 Hz, 3 H) 3.65 (s, 3 H) 3.20 (dd, J=15.06, 7.43 Hz, 1 H) 3.05 (dd, J=15.85, 6.85 Hz, 1 H) 2.87 (dd, J= 15.94, 5.77 Hz, 1 H) 2.37 - 2.46 (m, 1 H) 1.88 (td, J=12.62, 5.48 Hz, 1 H) 1.63 - 1.74 (m, 1 H) 1.18 - 1.33 (m, 2 H) 0.71 - 0.90 (m, 6 H); LCMS: (electrospray +ve), mlz 482.2 (MH) + ;HPLC: t R = 6.79 min, UV 254 = 95%.

Compound 96 (Method D)

[00298] 1 -(3,5-Dimethylpiperidin- 1 -yl)-3-(2-hydroxy-4,6-dimethoxyphenyl)-3-(4-

(trifluoromethoxy)phenyl)propan- 1 -one

[00299] Prepared by method D: DMSO-i/ 6 ) δ ppm 9.45 (s, 1 H) 7.32

(t, J=7.14 Hz, 2 H) 7.17 (d, J=8.41 Hz, 2 H) 6.02 (s, 1 H) 6.02 (s, 1 H) 4.96 (t, J=7.53 Hz, 1 H) 4.31 (d, J=13.1 1 Hz, 1 H) 3.75 (d, J=16.24 Hz, 1 H) 3.68 (d, J=4.30 Hz, 3 H) 3.65 (d, J=1.57 Hz, 3 H) 3.19 (dd, J=15.45, 7.04 Hz, 1 H) 3.06 (dd, J=l 5.65, 6.46 Hz, 1 H) 2.89 (dd, J=14.97, 5.58 Hz, 1 H) 2.36 - 2.44 (m, 1 H) 1.88 (t, J=l 1.25 Hz, 1 H) 1.63 - 1.71 (m, 1 H) 1.18 - 1.35 (m, 2 H) 0.80 (dd, J=6.46, 1 .57 Hz, 6 H); LCMS: (electrospray +ve), mlz 482.2 (MH) + ;HPLC: t R = 6.79

Compound 101 (Method D)

[00300] 3-(3-(3,5-Dimethylpiperidin- 1 -yl)- 1 -(2-hydroxy-4,6-dimethoxyphenyl)-3- oxopropyl)benzoic acid

[00301] ' Prepared by method D: Ή NMR (400 MHz, DMSO-i/ 6 ) δ ppm 12.73 (br. s., 1 Η)

9.44 ' - 9.48 (m, 1 H) 7.82 - 7.89 (m, 1 H) 7.65 (d, J=7.63 Hz, 1 Η) 7.46 (t, J=9.68 Hz, 1 H) 7.27 -

7.33 (m, 1 H) 6.00 - 6.04 (m, 2 H) 4.97 - 5.03 (m, 1 H) 4.31 (d, J=15.45 Hz, 1 H) 3.73 - 3.81 (m,

1 H) 3.67 (d, J=4.69 Hz, 3 H) 3.65 (s, 3 H) 3.12 - 3.24 (m, 1 H) 2.98 (dd, J=13.79, 7.92 Hz, 1 H)

2.76 - 2.89 (m, 1 H) 2.37 - 2.47 (m, 1 H) 1.84 - 1.92 (m, 1 H) 1 .62 - 1 .74 (m, 1 H) 1.24 (s, 2 H)

0.71 - 0.91 (m, 6 H); LCMS: (electrospray +ve), mlz 442.2 (MH) + ;HPLC: t R = 5.5 1 min, UV 254 =

98%.

Compound 81 (Method D)

[00302] N-(4-(3-(3,5-Dimethylpiperidin- l -yl)-l -(2-hydroxy-4,6-dimethoxyphenyl)-3- oxopropyl)phenyl)acetamide

[00303] Prepared by method D: , DMSO-i/ 6 ) δ ppm 9.74 (s, 1 H) 9.36

(d, J=2.35 Hz, 1 H) 7.29 - 7.44 (m, 2 H) 7.07 (t, J=7.92 Hz, 1 H) 6.93 (t, J=8.02 Hz, 1 H) 5.99 - 6.03 (m, 2 H) 4.85 - 4.95 (m, 1 H) 4.32 (d, J= 13.50 Hz, 1 H) 3.72 - 3.78 (m, 1 H) 3.66 - 3.68 (m, 3 H) 3.65 (d, J=0.78 Hz, 3 H) 2.90 - 3.02 (m, 1 H) 2.84 (dd, J=15.55, 6.16 Hz, 1 H) 2.71 (dd, J=12.91 , 6.46 Hz, 1 H) 2.37 - 2.46 (m, 1 H)1.98 (s, 3 H) 1.86 (t, J= 13.21 Hz, 1 H) 1 .61 - 1.73 (m, 1 H) 1 .1 1 - 1.25 (m, 2 H) 0.71 - 0.84 (m, 6 H); LCMS: (electrospray +ve), mlz 455.3

(MH) + ;HPLC: t R = 5.45 min, UV 254 = 99%.

Compound 82 (Method D)

[00304] N-(3-(3-(3,5-Dimethylpiperidin-l -yl)-l-(2-hydroxy-4,6-dimethoxyphenyl)-3- oxopropyl)phenyl)methanesulfonamide

[00305] Prepared by method D: Ή NMR (400 MHz, DMSO-rf 6 ) δ ppm 9.38 (s, 1 H) 7.08

(t, J=7.83 Hz, 1 H) 6.88 (d, J= 14.87 Hz, 1 H) 6.81 (d, J=6.65 Hz, 1 H) 6.73 (dd, J=8.51 , 1 .86 Hz, 1 H) 6.01 (d, J=1.37 Hz, 2 H) 5.00 (s, 1 H) 4.90 (t, J=7.63 Hz, 1 H) 4.32 (d, J=l 2.72 Hz, 1 H) 3.72 - 3.79 (m, 1 H) 3.62 - 3.68 (m, 6 H) 3.32 (s, 3 H) 3.28 (d, J=8.61 Hz, 1 H) 3.10 (d, J=8.41 Hz, 1 Η) 2.9 i (dd, J=l 5.06, 6.85 Hz, 1 H) 2.35 - 2.44 (m, 1 H) 1.86 (t, =12.13 Hz, 1 H) 1.61

1.70' (m, 1 H) 1 .13 - 1.35 (m, 2 H) 0.71 - 0.84 (m, 6 H); LCMS: (electrospray +ve), mlz 491.2

(MH) + ;HPLC: t R = 5.53 min, UV 254 = 85%.

Compound 84 (Method D)

100306] 4-(3-(3,5-Dimethylpiperidin- 1 -yl)- 1 -(2-hydroxy-4,6-dimethoxyphenyl)-3- oxopropyl)-N,N-dimethylbenzenesulfonamide

[00307] Prepared by method D: Ή NMR (400 MHz, DMSO-i/ 6 ) δ ppm 9.48 (d, J=2.93

Hz, 1 H) 7.54 (dd, J=8.31 , 2.05 Hz, 2 H) 7.38 - 7.44 (m, 2 H) 6.00 (s, 1 H) 5.99 (s, 1 H) 4.98 - 5.03 (m, 1 H) 4.29 (d, J=15.85 Hz, 1 H) 3.77 - 3. (m, 1 H) 3.63-3.62 (m, 6 H) 3.30 - 3.34 (m, 1 H) 3.18 (d, J=6.46 Hz, 1 H) 3.04 (dd, J=l 5.75, 7.14 Hz, 1 H) 2.88 (dd, J=16.82, 5.67 Hz, 1 H) 2.52 - 2.57 (m, 6 H) 1.82 - 1.90 (m, 1 H) 1.60 - 1.70 (m, 1 H) 1.16 - 1.26 (m, 2 H) 0.67 - 0.86 (m, 6 H); LCMS: (electrospray +ve), mlz 505.2 (MH) + ;HPLC: t R = 5.99 min, UV 254 = 85%.

Compound 88 (Method D)

[00308] 3-(3,4-Dichlorophenyl)-l -(3,5-dimethylpiperidin-l-yl)-3-(2-hydroxy-4,6- dimethoxyphenyl)propan- 1 -one

[00309] Prepared by method D: , DMSO-i¾) δ ppm 9.53 (d, J=5.28

Hz, 1 H) 7.41 - 7.46 (m, 1 H) 7.39 (t, J= 1.66 Hz, 1 H) 7.17 (dt, J=8.36, 1.59 Hz, 1 H) 6.00 - 6.04 (m, 2 H) 4.90 - 4.96 (m, 1 H) 4.30 (d, J=10.56 Hz, 1 H) 3.77 (br. s., 1 H) 3.68 (d, J=2.74 Hz, 3 H) 3.66 (s, 3 H) 3.19 (dd, J=14.97, 8.12 Hz, 1 H) 3.02 (dd, J=15.45, 6.65 Hz, 1 H) 2.87 (dd, J=14.67, 6.26 Hz, 1 H) 2.41-2.46 (m, 1 H) 1.89 (t, J=12.42 Hz, 1 H) 1.63 - 1.75 (m, 1 H) 1.18 - 1.30 (m, 2 H) 0.74 - 0.90 (m, 6 H); LCMS: (electrospray +ve), mlz 466.1 (MH) + ;HPLC: t R = 6.90 min, UV 254 = 99%.

Compound 94 (Method D)

[00310] l -(3,5-Dimethylpiperidin- l -yl)-3-(2-hydroxy-4,6-dimethoxyphenyl)-3-(3- isopropoxyphenyl)propan- 1 -one [00311] Prepared by method D: Ή NMR (400 MHz, DMSO-i 6 ) δ ppm 9.38 (s, 1 H) 7.05

(t,J=8.12 Hz, 1 H) 6.73 - 6.80 (m, 2 H) 6.59 - 6.64 (m, 1 H) 5.99 - 6.04 (m, 2 H) 4.86 - 4.92 (m, 1 H) 4.44 -4.51 (m, 1 H)4.31 (d,J=12.52 Hz, 1 H)3.71 -3.81 (m, 1 H) 3.67 (d,J=4.89 Hz, 3 H) 3.64-3.66 (m, 3 H) 3.28 (d, J=10.56 Hz, 1 H) 3.10 (d, J=9.00 Hz, 1 H) 2.88 (dd,J=16.24, 6.65 Hz, 1 H) 2.35 -2.44 (m, 1 H) 1.82- 1.90 (m, 1 H) 1.66 (d, J=14.87 Hz, 1 H) 1.14- 1.29 (m, 6 H) 0.73 - 0.82 (m, 6 H); LCMS: (electrospray +ve), mlz 456.2 (MH) + ;HPLC: t R = 6.61 min, UV 254 = 90%.

Compound 140 (Method D)

[00312] 3-(Benzo[d][l,3]dioxol-5-yl)-l-(3,5-dimethylpiperidin-l-yl)- 3-(2-hydroxy-6- methoxyphenyl)propan- 1 -one

[00313] Prepared by method D: H NMR (400 MHz, DMSO-rf 6 ) δ ppm 9.35 - 9.37 (m, 1

H) 6.93 (t,J=8.12Hz, 1 H) 6.85 (d,J=16.04 Hz, 1 H) 6.71 (d, J=1.17 Hz, 2 H) 6.41 (d,J=8.22 Hz, 2 H) 5.88 -5.91 (m, 2 H) 4.93 - 5.02 (m, 1 H) 4.31 (d,J=13.11 Hz, 1 H) 3.75 - 3.79 (m, 1 H) 3.70 (d,J=0.98 Hz, 3 H) 3.13 (dd, 7=15.85,8.22 Hz, 1 H) 2.96 (dd, J=15.55, 6.94 Hz, 1 H) 2.83 - 2.91 (m, 1 H) 2.35 - 2.46 (m, 1 H) 1.82 - 1.90 (m, 1 H) 1.62 - 1.76 (m, 1 H) 1.15 - 1.34 (m, 2 H) 0.72 - 0.91 (m, 6 H); LCMS: (electrospray +ve), mlz 412.1 (MH) + ;HPLC: t R = 6.11 min, UV 25 = 90%.

Compound 100 (Method D)

[00314] 3-(4-Acetylphenyl)-l-(3,5-dimethylpiperidin-l-yl)-3-(2-hydro xy-4,6- dimethoxyphenyl)propan- 1 -one

[00315] * Prepared by method D: Ή NMR (400 MHz, DMSO-rf 6 ) δ ppm 9.42 - 9.47 (m, 1

H) 7.78 (d, =8.22 Hz, 2 H) 7.33 (d, J=8.22 Hz, 2 H) 5.98 - 6.05 (m, 2 H) 4.99 - 5.04 (m, 1 H)

4.31 (d, J=15.06 Hz, 1 H) 3.74 - 3.80 (m, 1 H) 3.62 - 3.70 (m, 6 H) 3.14 - 3.25 (m, 1 H) 3.09 (d,

J=9.59 Hz, 1 H) 2.96 (d, J=9.00 Hz, 1 H) 2.55 (d, J=3.91 Hz, 1 H) 2.52 (s, 3 H) 1 .85 - 1.94 (m, 1

H) 1 .63 - 1 .76 (m, 1 H) 1.20 - 1.28 (m, 2 H) 0.64 - 0.90 (m, 6 H); LCMS: (electrospray +ve), m/z

440.2 (MH) + ;HPLC: t R = 5.92 min, UV 254 = 90%.

Compound 78 (Method D)

[00316] 3-(4-Chlorophenyl)- l -(3,5-dimethylpiperidin- l -yl)-3-(2-hydroxy-4,6- dimethoxyphenyl)propan- 1 -one

[00317] Prepared by method D: Ή NMR (400 MHz, DMSO-i/ 6 ) δ ppm 9.43 (d, J=4.30

Hz, 1 H) 7.22 (d, J=3.72 Hz, 4 H) 5.99 - 6.03 (m, 2 H) 4.90 - 4.95 (m, 1 H) 4.30 (d, J=\ 1.93 Hz, 1 H) 3.73- 3.77 (m, 1 H) 3.63 - 3.69 (m, 6 H) 3.1 1 - 3.18 (m, 1 H) 2.97 - 3.08 (m, 1 H) 2.56 (br. s., 1 H), 2.38- 2.46 (m, 1 H) 1.86 (d, J=l 1.74 Hz, 1 H) 1.61 - 1.75 (m, 1 H) 1 .15 - 1.27 (m, 2 H) 0.69 - 0.92 (m, 6 H); LCMS: (electrospray +ve), m/z 432.2 (MH) + ;HPLC: t R = 6.62 min, UV 254 = 99%.

Development of a Hish-Throushput Screening Assay

[00318] Methods: The Drosophila S2 cell line was originally purchased from Invitrogen and was maintained in Schneider's medium supplemented with 10% heat-inactivated bovine fetal calf serum and antibiotics (Invitrogen). Gal4 DNA binding domain (G4DBD) corresponding to amino acids 1 through 147 of Gal4 protein was PCR amplified to make a fusion construct with mouse RORyt (amino acids 79 to the carboxyl terminal end) lacking its DNA binding domain. The resulting chimeric gene was subcloned into the copper inducible pMT/V5-His A vector (available from Invitrogen). In a similar manner, Gal4 fusion constructs were prepared with mouse ROR (amino acids 142 to end) or Drosophila DHR3 (amino acids 120 to end). Coding sequences for firefly luciferase (available from Promega) were PCR amplified and subcloned into pUAST vector containing Gal4 binding enhancer sequences. pUAST is a commonly used Drosophila vector used by ordinarily skilled practitioners and has been described by Brand et al. (Development 1 18(2'):401 -415, 1993), the entire contents of which is incorporated herein in its entirdty.

[00319] Renilla luciferase construct under the pollll promoter was obtained from Dr.

Dasgupta's laboratory (NYU medical center). In order to generate stable cell lines, cells were co- transfected with pMT-G4DBD-RORyt, pMT-G4DBD-RORa, pMT-G4DBD-DHR3 , or pMT- G4DBDVP16 and pHygro plasmids (Invitrogen) and screened for resistant colonies in the presence of hygromycin (0.3 mg/mL). More than 150 independent RORy stable clones with luciferase reporters (pMt-RORy luc) were tested for their suitability for the high-throughput screening (HTS) based on the following criteria: high signal-to background ratio in 384- and 1 ,536- well plates, their high induction rate upon the addition of copper, and RORy/yt specificity probed by the dsRORy- or RORy/yt antagonist-mediated suppression. Positive clones were further transfected with pUAST-firefly luciferase, polIII-Renilla luciferase, and pCoPuro (Iwaki, Figuera et al. 2003) ("Rapid selection of Drosophila S2 cells with the puromycin resistance gene." Biotechniques 35(3): 482-4, 486.) and selected with puromycin (2.5 ug/ml). Seven clones were finally selected, and one of them (stable clone #25) was subsequently used for large-scale HTS. Using similar methods, stable clones with genomic integration of the luciferase reporters and other reporters were generated such as pMT-RORa_luc, pMT-DHR3_luc, or pMT- VP16_luc.

[00320] Results and Discussion: An activity-based assay system that permits high- throughput screening (HTS) for chemical modulators of RORyt transcriptional activity was developed. Since RORyt is located exclusively in cell nuclei, even when mutations are introduced in the putative ligand binding pocket, RORyt-dependent transcriptional activation, rather than the often used ligand-induced cytoplasm to nucleus translocation of hormone receptor, served as a better read-out. A cell-based assay was used to eliminate cell-impermeable or toxic molecules and to perform screening in a biologically relevant setting. The system employed provided a high signal-to-noise ratio and was able to handle a large-scale screen, as well as be cost-effective. S2 cells were derived from late stage Drosophila melanogaster embryos with hemocyte-like morphology and gene expression profiles (Schneider, I. J Embryo! Exp Morphol, 1972, 27, 353-65). They grew at room temperature as a semi-adherent monolayer with no requirement for C0 2 , making it easy to apply large sets of small chemical molecules and to transfer cells without trypsin treatment.

[00321] Like other hormone receptors, RORyt contains both a DNA binding domain

(DBD) and a ligand binding domain (LBD). The DBD was replaced with the heterologous yeast GAL4 DBD, because the endogenous DNA binding sites for RORyt are less well characterized. [00322] ' The GAL4-RORyt fusion construct was placed under the control of a copper inducible promoter, and stable S2 cell lines with genomic integration of this construct were generated. The copper inducible promoter ensures a tight regulation of GAL4-RORyt expression and allows small molecules to get into the cells prior to its induction, thus increasing their effects on GAL4-RORyt. The reporter cells also carried the firefly luciferase reporter, whose expression is regulated by five copies of the GAL4 binding site enhancer (UAS) and a constitutive heat shock promoter, along with a control plasmid, pol Ill-driven Renilla luciferase. Pol III-Renilla luciferase was previously shown to serve as an excellent transfection and cell viability control in the S2 cell system. Use of Pol III-Renilla luciferase permitted normalization of RORyt-driven firefly luciferase activity and reduced cytotoxic effects and corrected for potential imprecise dispensation of cells in culture medium (Armknecht, S. et al. Methods Enzymol, 2005, 392, 55-

73). When Cu++ was added, the ratio of firefly to Renilla luciferase activity (FR ratio) in these cells increased more than 100-fold compared to control cells lacking GAL4-RORyt (~34 fold increase compared to GAL4-RORyt cells treated with dsROR). RORyt induces robust transcriptional activation in Drosophila S2 cells in 384-well plates. For test with transient transfection, firefly reporter under GAL4 binding sites and Pol III-Renilla control plasmids were transiently transfected along with dsRNA (75 ng), targeting EYFP or RORy, into both control or pMT-GAL4-RORyt stable S2 cell lines ( 10,000 cells/well). After three days, copper was added to induce GAL4-RORyt, and dual luciferase activity was measured following overnight incubation. The increase was also observed when the experiment was carried out in 384-well plates, demonstrating that it can be adopted as a high-throughput screen. Co-transfecting dsRNA that targets RORyt suppressed ROR-mediated firefly luciferase induction, demonstrating that this activity is ROR dependent.

[00323] In order to confirm that the RORyt function in S2 cells was physiologically relevant, it was first confirmed that Drosophila has a RORyt homologue. Mouse encodes three different ROR proteins, RORa, RORp, and RORy. RORy and RORyt are two isoforms that differ in non-translated N-terminal mRNA sequences. Indeed, Drosophila has one ROR homologue, Drosophila hormone receptor 3(DHR3) (King- Jones, K. & Thummel, C. S. Nat Rev Genet, 2005, 6, 3 1 1-23). Structure-based alignment using BLAST revealed 48% amino acid identity between DHR3 and RORyt.

[00324] Next it was confirmed that RORyt ligands are likely present in Drosophila S2 cells. Since many ligands for nuclear hormone receptors are found to be sterols or their derivatives, growth of cells in medium lacking FBS (fetal bovine serum) or medium

supplemented with fatty-acid stripped serum (charcoal treated) was attempted. Only a small decrease of the FR ratio was detected in cells grown in this condition, and the results were not conclusive. Previous studies have shown that the introduction of a single amino acid change inside the ligand binding pocket of RORP abrogates its function as a transcriptional activator, suggesting that RORP is a ligand-dependent hormone receptor. The crystal structure of the protein strongly suggested that replacement of alanine 269 with amino acids carrying bulkier side chains, such as valine and phenylalanine, would prevent binding of endogenous ligands without affecting the correct folding of the ligand binding domain (Stehlin, C. et al. Embo J, 2001, 20,

5822-3 1). When the corresponding alanine residue in the putative ligand binding pocket of

RORyt was replaced with phenylalanine, the mutant protein was no longer sufficient to induce

Th l 7 cell differentiation when transduced into naive mouse CD4+ T cells, consistent with the presence of cognate RORyt ligand(s) in these cells. The Ala to Phe mutation in RORyt also completely abrogated transcriptional activation of firefly luciferase expression without affecting the transcription of control Renilla luciferase, suggesting that RORyt ligand is present in the

Drosophila assay system. Indeed, introduction of alanine to phenylalanine (F) mutation in a putative ligand binding pocket abolishes activity of RORyt in this system. As confirmed by immunoblot, the alanine to phenylalanine mutation did not, however, affect protein stability.

[00325] DHR3 is transcriptionally regulated by 20-hydroxyecdysone (20E) and is essential for fly larval development (King-Jones, supra). It has been shown that E75, another 20E- dependent fly nuclear hormone receptor, negatively regulates the function of DHR3 (White, K.

P., Hurban, P., Watanabe, T. & Hogness, D. S. Science, 1997, 276, 1 14-17; Reinking, J. et al.

Cell, 2005, 122, 195-207. Indeed, co-expression of E75a or E75b (two Drosophila E75 isoforms) decreased the level of DHR3-mediated transcriptional activation in a dosage-dependent manner. E75 is a hormone receptor having antagonizing activities of DHR3 and RORyt.

Transfection of increased amount of E75a or E75b resulted in concomitant reduction of FR ratio.

Each well received the same amount of DNA in transfection mix. These fly genes also function as dosage-dependent negative regulators for mouse RORyt activity in S2 cells, without affecting the functions of an irrelevant transcriptional activator VP 16, strongly suggesting that the

ROR/DHR3 core regulatory mechanism is conserved between mouse and fly systems.

Collectively, these data confirm the accuracy and relevance of the above approach utilizing the heterologous S2 cell system in order to identify chemical agonists or antagonists of the mouse hormone receptor RORyt.

[00326] Measurement of Luciferase activity. Methods: Promega Dual-Glo system is widely used for HTS as the luciferase substrates. Cell culture medium was reduced to the final volume of 10 μΐ or less and 10 μΐ of Dual-glo and Ι ΟμΙ of Stop-glo luciferase substrates were added in a sequencial manner (Promega). Firefly and Renilla luciferase activity was determined by measunngluminescence signals using an automated 384-well plate reader equipped with multiple sta"ck units (such as the Analyst GT or Envision plate readers).

[00327] Results and Discussion: The Dual-Glo luciferase assay system from Promega facilitated measuring luciferase activity in HTS. First, it did not require a washing step, and the luciferase activities of both the firefly and the Renilla could be measured in the same well one after the other. Second, the signals that it produced remained stable for more than two hours, making it possible to measure activity from many different plates for a given experiment. Since the reagents are expensive, the volume of medium was reduced to one third prior to adding luciferase substrates, so that fewer substrates were used. However, when minimizing cost is less of a priority, the luciferase substrates used in the primary assay may be directly added to cells without going through this additional step.

[00328] HTS for identification of RORy/yt antagonists using RORy/yt-luc stable cell lines

[00329] Methods: 600 G4DBD-RORy/yt-luc reporter (#25) or G4DBD-VP 16-luc reporter cells were distributed into each well of 1 ,536-well white bottom plates in 4 μΐ S2 cell culture volume containing hygromicin (300 μg/ml) and puromycin (2 μg/ml). Small compounds in seven different concentrations range from 46 μΜ to 1.7 nM were pin-transferred (23 nl) by a robot (Inglese et al., 2006, Proc. Natl. Acad. Sci. 103: 1 1473-8). After one-hour incubation, 1 μΐ of culture medium solution containing copper sulfate (to final 0.7 mM) and 10 μΜ RORy/yt antagonists were added to each well. After 20-hour incubation at ambient temperature, 1.5 ul firefly luciferase detection mix was added and the luciferase activities were measured in 10 min. ViewLux luminometers were used for measuring luciferase signal.

[00330] Results and Discussion: Even though G4BD-RORyt stable cells with transient transfection of two luciferase constructs were successfully used for small-scale screening and led the present inventors to identify specific RORy/yt antagonists in Harvard LCCB, it became problematic to apply the same method to larger scale screens with chemical libraries covering more than 250,000 compounds. First, transient transfection method often produced well-to-well variation due to incomplete mixing and unequal distribution of transfection mix. There is also day-to-day variation when preparing the master transfection mix. Second, in order to save reagent and to handle large quantities of chemicals, it was necessary to do the screen with reduced numbers of cells in a smaller culture volume. Moreover, performing the screen with

1 ,536-well plates is more efficient than performing the screen with 384-well plates. Thus, new

RORy/yt-luc reporter stable cells were developed to eliminate the necessity of repeated transfection and to achieve increased well-to-well consistency. When tested in a 384-well plate, even small numbers of cells (400 cells/well) gave high firefly luciferase signal (thus high ,

RORy/yt activity), which is suppressed (22-fold reduction when 400 cells are used) by addition of a RORy/yt antagonist. The new stable cell line systems also turned out to be suitable for HTS in

1 ,536-well plates, because z' value is 0.75 when 600 cells are used per well with 10 μΐ total cell culture volume.

[00331] Using RORy/yt-luc reporter lines, the pilot screen with the LOP AC (Sigma) library was performed through a NIH roadmap program to identify RORy/yt antagonist compounds. LOPAC contains 1 ,280 pharmacologically active compounds. Among these, approximately 40 compounds were found as initial hits (-0.3%). These hits were tested against validation screens to identify RORy/yt specific compounds. To facilitate a large-scale validation, VP 16-luc reporter cell lines with genomic integrations of G4BD-VP16, UASt-ffluc, and polIII- Rluc were developed as discussed earlier. These cells also exhibited robust and consistent VP 16 activity in 1 ,536-well plates (data not shown), and thus can be used as a good control reporter to weed-out compounds inhibiting general transcription, the function of GAL4 DNA binding domain, and a nuclear import of GAL4 chimeric proteins. Indeed, many compounds were identified as initial hits to reduce RORy/yt activity from the pilot LOPAC screen, but later were found to be non-specific inhibitors, because they reduced both RORy/yt and VP 16 activities. Intriguingly, previously identified RORy/yt specific antagonists identified by small scale HTS, also selectively inhibited RORy/yt activity without affecting VP 16, confirming their specificity on RORy/yt. A large-scale screen covering more than 250,000 compounds was carried out against RORy/yt-luc and VP 16-luc reporter systems in parallel to identify RORy/yt specific antagonists in a systematic manner.

[00332] Secondary assays directed toward evaluating the ability of such compounds to suppress mouse or human Thl 7 cell differentiation are also encompassed herein. Exemplary secondary assays that serve to confirm bona fide RORy inhibitors include without limitation: Secondary screening lists

1) S2 cell reporter system: Identified hits from HTS are further screened and their specificity confirmed by testing against RORy-luc, VP16-luc, RORa-luc, and DHR3-luc reporter S2 cell lines. Compounds either having no activity on VP 16, RORa, and DHR3 or having ten times higher IC50 values on such reporters are selected for further tests.

2) Cytokine induced mouse Thl 7 cells differentiation. Effects on mouse endogenous RORyt in a relevant physiological setting were determined by testing compounds in the Th l 7 cell differentiation assay. Compounds having RORyt inhibitory activity are predicted to suppress Th l 7 cell differentiation. Th l or regulatory T cell differentiation was used as a counter-screen method to select specific compounds that only affect Thl 7 ,

cell differentiation without having pleiotrophic effects on general T cell proliferation or cytokine production.

3) ROR dependent Th l 7 cell differentiation. Compounds were further tested, by examining their effects on T cells expressing RORa or RORy. Compounds that directly inhibit RORy are expected to inhibit RORy- but not RORa- dependent Thl 7 cell differentiation. Compounds affecting IL17a production or ROR regulatory pathways, however, are expected to inhibit both.

4) Human Th l 7 cell differentiation. Compound effects on human RORyt will be tested by treating human cord blood CD4 T cells with select compounds to determine if such compounds alter differentiation into Th l 7 lineages.

[00333] With aspects of the claimed invention now being generally described, these will be more readily understood by reference to the following examples, which are included merely for purposes of illustration of certain features and embodiments of the presently claimed invention and are not intended to be limiting.

EXAMPLES

[00334] As detailed above, in order to identify small molecules to antagonize RORy/yt transcriptional activity, the present inventors developed insect cell line based reporter systems, expressing murine RORy/yt or closely related transcriptional activators. Since their cognate DNA binding sites were not well characterized, the DNA binding domains (DBD) of RORy/yt, RORa (mouse homolog for POPy), and DHR3 (Drosophila orthologue for ROR family proteins) were replaced with the heterologous yeast GAL4 DBD. The transcriptionally active domain of general transcriptional activator VP 16 was also fused with GAL4 DBD. The GAL4 fusion constructs were placed under the control of a copper inducible promoter, and stable S2 cell lines with genomic integration of these four reporter constructs were generated. The copper inducible promoter ensures tight regulation of GAL4-fusion protein expression and it allows small molecules to get into the cells prior to protein induction, thus increasing their effects on GAL4 reporters. The stable reporter cell lines also encode the firefly luciferase reporter, whose expression is regulated by five copies of the GAL4 binding site enhancer (UAS), along with the pol Ill-driven Renilla luciferase reporter. Pol III-Renilla luciferase was included to serve as cell viability control in the S2 cell system (Armknecht, S. et al. Methods Enzymol 392, 55-73 (2005). |00335] From screening a chemical compounds library consisting of 4,812 compounds, including known Bioactives and Prestwick collections, a number of compounds were identified as small molecule inhibitors for the RORy/yt transcriptional activity. ICgn Determinations

[00336] A cell-based reporter assay was used to detect RORyt-mediated activity. This assay, called RORyt employed Drosophila Schneider cells that were stably transfected with two vectors: a gene expressing a fusion of the Gal4 DNA binding domain and RORyt transactivation domain under the control of the metallothionine promoter and a Photinus luciferase reporter regulated by the Gal4 binding site enhancer, UAS. Copper addition to the medium induced expression of the Gal4-RORyt fusion, which subsequently induced expression of the UAS- luciferase reporter. Small molecule inhibitors of RORyt activity were detected by a decrease in luciferase reporter activity. Cells (600/well) were dispensed into white solid 1536-well plates (Greiner) using a solenoid-based dispenser. Following transfer of 23 nL compound or DMSO vehicle by a pin tool, the plates were incubated 1 hr at ambient temperature, and 1 uL/well copper sulfate (700 uM final concentration) was added. The plates were centrifuged 15 s at 1000 RPM and incubated 20 hr at ambient temperature. After addition of 1.5 uL Photinus luciferase detection reagent, the plates were incubated 10 min at ambient temperature and then read by a ViewLux (Perkin Elmer) to detect luminescence. The concentration-response data were fit using a reported algorithm (Wang, Y. et al. Current Chemical Genomics, 2010, 57-66). Efficacy is expressed as % of the maximal response of control inhibitor (Digoxin), set at 100%. IC50 is the concentration at which compound exhibits half-maximal efficacy.

[00337] A number of representative amido compounds of this invention were or can be tested for their inhibitory activity. The amido compounds of the invention along with their IC50 and Efficacy values, as determined using conventional methods to those skilled in the art, are listed below in Table 1. For the purpose of Table 1 , the IC50 values are expressed as follows: compound exhibited IC50 < 1 μΜ

compound exhibited IC50 1- 10 μΜ

compound exhibited IC50 1 1 -50 μΜ

compound exhibited IC50 >50 μΜ

[00338] TABLE 1 : IC50 Values for Exemplary Compounds

Compd MW MW Efficacy

Sample ID Structure ΙΟο(μΜ)

# (Calcd) (obsvd) (%)

NCGCOO 189203 68 417.51 418.20 + null

NCGCOO 189205 69 443.54 444.30 ++ -45.58

NCGC00238412 71 307.39 308.20 + null

NCGC00238415 72 440.59 441 .40 +++ -96.81

NCGC00238435 73 41 1.55 412.20 ++++ -93.64

NCGC00238436 74 427.55 428.20 +++ -94.10

NCGC00238437 75 413.52 414.20 +++ -93.76

NCGC00238443 76 455.56 456.20 ++++ -95.37

NCGC00242601 77 L 427.55 428.30 +++ -97.81

NCGC00242602 78 431.96 432.20 +++ -98.04

NCGC00242604 80 ¾ 469.58 470.30 ++++ -93.97 Compd MW MW Efficacy

S.ample I,D Structure ΙΟ 50 (μΜ)

# (Calcd) (obsvd) (%)

NCGC00242617 92 503.64 504.20 +++ -94.56

NCGC00242618 93 481.52 482.20 +++ -93.26

NCGC00242619 94 455.60 456.20 +++ -90.55

NCGC00242620 95 480.61 481.20 +++ -93.06

NCGC00242621 96 481.52 482.20 +++ -94.77

NCGC00242622 97 466.41 466.10 ++ -61.47

NCGC00242624 99 441.53 442.20 +++ -92.59

NCGC00242625 100 439.56 440.20 ++++ -93.60

NCGC00242626 101 441.53 442.20 +++ -88.72

NCGC00242627 102 415.51 416.20 +++ -99.94

NCGC00242628 103 J 431.96 432.20 +++ -95.80 Compd MW MW Efficacy

Sample ID Structure ICMGIM)

# (Calcd) (obsvd) (%)

NCGC00242636 140 41 1.50 412.10 H i t -96.47

NCGC00242637 141 432.53 433.20 ++ - 105.50

NCGC00242639 143 399.47 400.10 ++ -86.36

NCGC00242640 144 399.47 400.10 ++ -97.33

NCGC00242641 145 399.47 400.10 +++ -79.73

CM,

NCGC00242642 146 432.52 433.10 +++ -94.07

CM,

NCGC00242648 147 403.48 404.20 ++ -88.07

NCGC00238416 149 439.60 440.30 +++ -93.79

NCGC00242649 150 441.57 442.20 +++ -91.61

NCGC00242650 151 477.65 478.20 + - 1 18.13

NCGC00242652 152 423.56 424.20 ++ -50.66

* The stereochemistry is not confirmed.

Thl7 Assay

[00339] Effects of compound derivatives at various concentrations on mouse Thl 7 polarization. Percentage of DMSO treated IL- 17a producing cells was set at 100. . Naive mouse CD4 T cells (CD25 " , CD62L + , and CD44 int"hish ) were sorted and cultured with

CD3/CD28 stimulatory antibodies in the presence of TGF (O. lng/ml) and 1L6 (20ng/ml). Compounds were added on Day 1 and cells were analyzed on Day 4. For intracellular cytokine staining, cells were incubated for 5 h with phorbol ester (50 ng/ml; Sigma), ionomycin (500 ng/ml; Sigma) and GolgiStop (BD). When needed, surfaces were stained by incubation for 15 min on ice with PECy7-conjugated CD4 (BD Biosciences). The Cytofix/Cytoperm buffer set (BD) was used for intracellular staining. Cells were fixed and made permeable for 30 min on ice and were stained for 30 min on ice in permeabilization buffer with Alexa647-conjugated anti-IL 17 (eBioscience) and PE-conjugated anti-lFNg (eBioscience). An LSR II (BD

Biosciences) and FlowJo software (Tree Star) were used for flow cytometry. RORg ICso value acquired from the S2 cell reporter assay is listed for comparison.

[00340] A number of representative amido compounds of this invention were or can be tested for their Th l 7 inhibitory activity. The exemplary amido compounds of the invention along with their Th l 7 and S2 RORg IC50 values, as determined using conventional methods to those skilled in the art, are listed below in Table 2. For the purpose of Table 2, the IC50 values are expressed as follows: compound exhibited TH17 IC 50 < 1 μΜ

compound exhibited TH17 IC50 1 - 10 μΜ

compound exhibited TH17IC 50 1 1 -20 μΜ

compound exhibited TH 17 IC50 >20 μΜ

compound exhibited RORg IC50 < 1 μΜ

compound exhibited RORg IC50 1 -10 μΜ

compound exhibited RORg IC50 1 1 -20 μΜ

compound exhibited RORg IC50 >20 μΜ

[00341] Table 2: Thl7 and RORy S2 Cell IC 50 values for Exemplary Comp

[00342] The chemical structures of three related compounds that were identified as

RORy/RORyt inverse agonists using the S2 cell RORy reporter chemical screen are shown in Figure 1.

[00343] Figure 3 shows a FACS plot analysis demonstrating that RORy inhibitory compounds NCGCOO 166547 and NCGCOO 166488 (20 μΜ) suppress mouse Thl 7 cell differentiation, as measured by IL17a production, when compared to DMSO treated cells. Naive CD4 T cells (CD25 " , CD62L + , and CD44 int - high ) were sorted and cultured with CD3/CD28 stimulatory antibodies in the presence of TGFp (O. lng/ml) and IL6 (20ng/ml). Compounds were added on Day 1 and cells were analyzed on Day 4. For intracellular cytokine staining, cells were incubated for 5 h with phorbol ester (50 ng/ml; Sigma), ionomycin (500 ng/ml; Sigma) and GolgiStop (BD). When needed, surfaces were stained by incubation for 15 min on ice with PECy7-conjugated CD4 (BD Biosciences). The Cytofix/Cytoperm buffer set (BD) or FoxP3 staining kit (eBioscience) were used for intracellular staining. Cells were fixed and made permeable for 30 min on ice and were stained for 30 min on ice in permeabilization buffer with Alexa647-conjuga ' ted anti-IL17 (eBioscience), PE-conjugated anti-IFNg (eBioscience), and/or

PEiconjugated anti-FoxP3 (eBioscience). An LSR II (BD Biosciences) and FlowJo software

(Tree Star) were used for flow cytometry.

100344] Figure 4 shows a FACS plot analysis demonstrating that treatment with 20 μΜ

NCGC00166547 does not inhibit Thl differentiation, as measured by IFNy production, from naive CD4 T cells. These results suggest that NCGC00166547 does not inhibit general T cell proliferation or cytokine production. For Th l differentiation, IL12 (lOng/ml) and IL2 (10U) was added to naive CD4 T cell cultures instead of TGFp and IL6.

[00345] As shown in Figure 5, compounds 166547 and 166488 inhibit RORy

overexpression dependent, but not RORct dependent, IL17a production in human CD4 T cells. These findings demonstrate their specificity for inhibition of RORy activity. Briefly, CD3+ CD4+ CD45RA+ naive human T cells were isolated from peripheral blood derived from healthy donors. The naive T cells were cultured in XVIVO medium for 7 days in the presence of IL2 and stimulatory CD3 and CD28 antibodies. Cells were infected with lentivirus encoding human RORa or RORy and compounds or DMSO were added on day 2. FACS plot analysis is shown after gating on GFP expressing (virus infected) cells.

[00346] Figure 6 shows FACs plots revealing that Compound NCGC00238427 selectively inhibits human RORyt induced Thl 7 polarization. See Figure 6A. The results depict flow cytometry of I L- 17a and IFN-y production by cord blood naive CD4 T cells (CD45RCT

CD45RA + CD3 + CD4 + CD25 " HLA-DR " ) transduced with human RORad-IRES-GFP or human RORyt-IRES-GFP on day 1 and analyzed on day 6. GFP expressing cells were gated for analysis. DMSO or NCGC00238427 (3 μΜ) was added 6-8h after viral transduction. As shown in Figure 6B, Compound NCGC00238427 inhibits human Thl 7 cell differentiation at as low as 1 μΜ. The results depict flow cytometry of IL- 17a and IFN-y production by human naive cord blood T cells (CD45RO ~ D45RA + CD3 + CD4 + CD25 ~ HLA-DR ~ ), which were cultured for six days in the presence of IL2, IL23 and IL1 β, and with various concentrations of TGFp (ng/mL). DMSO or NCGC00238427 were added 16 hours after the cytokine addition. IL-17a-APC (eBio64CAP 17 eBioscience) and IFN-y-PECy7 (45.B3 eBioscience) were used for analysis.

[00347] Figure 7 shows that NCGC00238427 inhibits human Th l 7 cell memory cells expression of IL- 17a and is, therefore, important for maintenance of human Th l 7 cells. Human memory (CD45RO + CD45RA " CD3 + CD4 + CCR6 + CD161 + ) cells were purified from healthy donor peripheral blood samples and were cultured in the presence of IL- Ι β, IL-23 and IL-2 for 6 days with or without NCGC00238427 (3 uM). Intracellular staining for IFN-y or IL- 17a in memory CD4 + T cells from multiple donors, assessed on day 6. Each symbol (n=l 1 or 9, respectively) indicates a separate donor. Statistical analysis was by a two-tailed unpaired Student's t test; IL-

17a FN-y + , not significant and IL-17a + IFN-y +/~ , p=0.02.

[00348] In vitro competition assays have, furthermore, shown that compounds described herein, such as NCGCOO 166547, directly bind to human RORyt. To demonstrate this binding property, recombinant human RORy LBD was loaded with fluorescently-labeled 25- hydroxycholesterol in the presence of various concentrations of NCGCOO 166547, and fluorescence polarization was measured. The presence of increasing concentrations of

NCGCOO 166547 was correlated with displacement of the fluorescently-labeled 25- hydroxycholesterol from the LBD.

[00349] From the foregoing description, various modifications and changes in the compositions and methods of this invention will occur to those skilled in the art. All such modifications coming within the scope of the appended claims are intended to be included therein.

[00350] All publications, including but not limited to patents and patent applications, cited in this specification are herein incorporated by reference as if each individual publication were specifically and individually indicated to be incorporated by reference herein as though fully set forth.

[00351] The chemical names of compounds of invention given in this application are generated using Open Eye Software's Lexichem naming tool, Symyx Renassance Software's Reaction Planner or MDL's ISIS Draw Autonom Software tool and not verified. Preferably, in the event of inconsistency, the depicted structure governs.