Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
TOLL-LIKE RECEPTOR SIGNALING INHIBITORS
Document Type and Number:
WIPO Patent Application WO/2019/136147
Kind Code:
A1
Abstract:
Di- and triaryl-substituted heteroaromatic compounds have Toll-like receptor inhibitory activity, including at TLR2, TLR4, TLR7, and/or TLR9. Compounds and compositions have applications in the treatment of diseases and conditions mediated by Toll-like receptors and related receptors, such as bacterial sepsis, autoimmune disease, lupus erythematosus, ischemia-reperfusion injury, stroke, metabolic disease, obesity-related metabolic inflammation, gout, and cancer.

Inventors:
KATZENELLENBOGEN JOHN (US)
POLLOCK JULIE (US)
HAECKER HANS (US)
SHARMA NAINA (US)
Application Number:
PCT/US2019/012181
Publication Date:
July 11, 2019
Filing Date:
January 03, 2019
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
UNIV ILLINOIS (US)
ST JUDE CHILDRENS RES HOSPITAL INC (US)
International Classes:
A61K31/05; C07D233/64; C07D261/08; C07D263/32
Domestic Patent References:
WO2000019994A12000-04-13
Foreign References:
US20110312925A12011-12-22
Other References:
DASGUPTA, S ET AL.: "Estrogen receptor-alpha mediates Toll-like receptor-2 agonist-induced monocyte chemoattractant protein-1 production in mesangial cells", RESULTS IN IMMUNOLOGY, vol. 2, 2012, pages 196 - 203, XP055621488
Attorney, Agent or Firm:
CARROLL, William, A. et al. (US)
Download PDF:
Claims:
WE CLAIM: 1. A compound of formula (I), or a pharmaceutically acceptable salt thereof,

wherein:

G1 is a 4- to 12-membered heterocyclyl containing 1-3 heteroatoms independently selected from N, O, and S,–NR1R2,–OH,–OC1-4alkyl, a C3-8cycloalkyl, or a 5- to 12-membered heteroaryl containing 1-3 heteroatoms independently selected from N, O, and S, wherein the cycloalkyl, the heterocyclyl, and the heteroaryl are optionally substituted with 1-4 substituents independently selected from halo, C1-4alkyl, C1-4haloalkyl,–OH,–OC1-4alkyl, and oxo;

R1 and R2 are each independently hydrogen or C1-4alkyl;

L1 is–C1-4alkylene–O–,–C1-5alkylene–,or–C(O)–CH=CH–, wherein the–C1-5alkylene– and the –C1-4alkylene–O– are optionally substituted with 1-2 halogens or one hydroxyl; or L1 is , wherein the C 1

1-4alkylene is bonded to G and the imidazole is fused at the meta and para positions on the phenyl relative to G2;

G2 is selected from (i) to (xiii)

X1 is O or S;

X2 is O, S, NH, or NC1-4alkyl;

R5 is or C1-4alkyl;

R3, R7, R13, R17, R19, R21, R27, R29, and R33 are each independently selected from hydrogen and C1-4alkyl;

R11 and R31 are each independently selected from hydrogen, C1-4alkyl, and phenyl optionally substituted with 1-3 substituents independently selected from halo, C1-4alkyl, C1-4haloalkyl,– OH, and–OC1-4alkyl;

R4a, R4b, R4c, R5a, R6, R8, R9, R10, R12, R14, R15, R16, R18, R20, R22, R23, R24, R25, R26, R28, R30, and R32, at each occurrence, are independently–OH, halo, nitro, cyano, C1-4alkyl, C1-4haloalkyl,– OC1-4alkyl,–OC1-4haloalkyl,–NH2,–NH(C1-4alkyl),–N(C1-4alkyl)(C1-4alkyl),–NHC(O)C1- 4alkyl,–N(C1-4alkyl)C(O)C1-4alkyl,–NHC(O)OC1-4alkyl,–N(C1-4alkyl)C(O)OC1-4alkyl,– C(O)OC1-4alkyl,–C(O)OH,–C(O)NH2,–C(O)NH(C1-4alkyl), or–C(O)N(C1-4alkyl)(C1- 15

4alkyl), and optionally two R4a, R4b, R5a, R6, R8, R9, R10, R12, R14, R , R16, R18, R20, R21, R22, R24, R25, R26, R28, R30, or R32, together with the atoms to which they are attached form a

fused ring ;

n1 and n2 are independently 1, 0, 2, 3, 4, or 5; and

n3 and n4 are independently 0, 1, 2, or 3. 2. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein the compound of formula (I) has formula (IA)

.

3. The compound of claim 2, or a pharmaceutically acceptable salt thereof, wherein G1 is a 4- to 12-membered heterocyclyl containing 1-3 heteroatoms independently selected from N, O, and S,–NR1R2, or a 5- to 12-membered heteroaryl containing 1-3 heteroatoms independently selected from N, O, and S, wherein the heterocyclyl and the heteroaryl are optionally substituted with 1-4 substituents independently selected from halo, C1-4alkyl, C1- 4haloalkyl,–OH,–OC1-4alkyl, and oxo; and

L1 is–C1-4alkylene–O– or–C1-5alkylene–. 4. The compound of any of claim 2, or a pharmaceutically acceptable salt thereof, wherein G1 is a 4- to 8-membered monocyclic heterocyclyl containing a first nitrogen atom and

optionally an additional heteroatom selected from N, O, and S, the heterocyclyl connecting to L1 at the first nitrogen atom and optionally containing a C1-3alkylene bridge between two non-adjacent ring atoms and/or a double bond, the heterocyclyl being optionally substituted with 1-4 substituents independently selected from halo, C1-4alkyl, C1-4haloalkyl,–OH,–OC1- 4alkyl, and oxo;

–NR1R2; or

a 5- to 6-membered monocyclic heteroaryl containing 1-3 nitrogen atoms, the heteroaryl being optionally substituted with 1-4 substituents independently selected from halo, C1- 4alkyl, C1-4haloalkyl,–OH, and–OC1-4alkyl; and

L1 is–C2-3alkylene–O– or–C2-4alkylene–. 5. The compound of claim 2, or a pharmaceutically acceptable salt thereof, wherein G1 is piperidin-1-yl,–N(CH3)2,–N(CH2CH3)2, pyrrolidin-1-yl, piperazin-1-yl, azepin-1-yl, pyrazol-1-yl, imidazol-1-yl, or triazol-1-yl. 6. The compound of claim 4, or a pharmaceutically acceptable salt thereof, wherein G2 is formula (i)

.

7. The compound of claim 6, or a pharmaceutically acceptable salt thereof, wherein . 8. The compound of claim 6, or a pharmaceutically acceptable salt thereof, wherein R5 is C1-4alkyl. 9. The compound of claim 7, or a pharmaceutically acceptable salt thereof, wherein

10. The compound of claim 9, or a pharmaceutically acceptable salt thereof, wherein

R4a, at each occurrence, is independently–OH, halo, cyano, C1-4alkyl, C1-4haloalkyl,–OC1-4alkyl, –OC1-4haloalkyl,–NH2,–NH(C1-4alkyl),–N(C1-4alkyl)(C1-4alkyl),–NHC(O)C1-4alkyl,–N(C1- 4alkyl)C(O)C1-4alkyl,–C(O)OC1-4alkyl,–C(O)NH2,–C(O)NH(C1-4alkyl), or–C(O)N(C1- 4alkyl)(C1-4alkyl), and optionally two R4a, together with the atoms to which they are attached,

form a fused ring ;

R5 is ;

R5a is–OH, halo, nitro, cyano, C1-4alkyl, C1-4haloalkyl,–OC1-4alkyl,–OC1-4haloalkyl,–NH2,– NH(C1-4alkyl),–N(C1-4alkyl)(C1-4alkyl),–NHC(O)C1-4alkyl,–N(C1-4alkyl)C(O)C1-4alkyl,– NHC(O)OC1-4alkyl, or–N(C1-4alkyl)C(O)OC1-4alkyl;

n1 is 1, 0, 2, or 3; and

n2 is 1 or 0. 11. The compound of claim 6, or a pharmaceutically acceptable salt thereof, wherein

R4 is

12. The compound of claim 10, or a pharmaceutically salt thereof, wherein R3 is hydrogen. 13. The compound of claim 10, or pharmaceutically acceptable salt thereof, provided that the compound is not 4,4'-(4-ethyl-3-(4-(2-(piperidin-1-yl)ethoxy)phenyl)-1H-pyrazole-1,5- diyl)diphenol, or a salt thereof. 14. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein G2 is formula (ii)

15. The compound of claim 14, or a pharmaceutically acceptable salt thereof, wherein n1 and n2 are each independently 0, 1, 2, or 3. 16. The compound of claim 15, or a pharmaceutically acceptable salt thereof, wherein G2 is formula (iia)

17. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein G2 is formula (iii)

18. The compound of claim 17, or a pharmaceutically acceptable salt thereof, wherein n1 and n2 are each independently 0, 1, 2, or 3. 19 The compound of claim 18, or a pharmaceutically acceptable salt thereof, wherein n1 and n2 are each independently 0 or 1. 20. The compound of claim 17, or a pharmaceutically acceptable salt thereof, wherein R9 and R10, at each occurrence, are independently halo, nitro, cyano, C1-4alkyl, C1-4haloalkyl,– OC1-4alkyl,–OC1-4haloalkyl,–NH2,–NH(C1-4alkyl),–N(C1-4alkyl)(C1-4alkyl),–NHC(O)C1- 4alkyl,–N(C1-4alkyl)C(O)C1-4alkyl,–NHC(O)OC1-4alkyl,–N(C1-4alkyl)C(O)OC1-4alkyl,– C(O)OC1-4alkyl,–C(O)OH,–C(O)NH2,–C(O)NH(C1-4alkyl), or–C(O)N(C1-4alkyl)(C1-4alkyl), and optionally two R9 or R10, together with the atoms to which they are attached form a fused . 21. The compound of claim 17, or a pharmaceutically acceptable salt thereof, wherein R9 and R10, at each occurrence, are independently halo, C1-4alkyl,–OH, or–OC1-4alkyl. 22. The compound of claim 21, or a pharmaceutically acceptable salt thereof, wherein: R9, at each occurrence, is independently halo, C1-4alkyl, or–OC1-4alkyl; and R10, at each occurrence, is independently halo, C1-4alkyl,–OH, or–OC1-4alkyl. 23. The compound of claim 22, or a pharmaceutically acceptable salt thereof, wherein R10, at each occurrence, is independently halo, C1-4alkyl, or–OC1-4alkyl. 24. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein G2 is formula (iiia)

and n1 and n2 are each independently 0 or 1. 25. The compound of claim 17, or a pharmaceutically acceptable salt thereof, wherein R11 is hydrogen. 26. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein G2 is formula (iv)

27. The compound of claim 26, or a pharmaceutically acceptable salt thereof, wherein n1 and n2 are each independently 0, 1, 2, or 3. 28. The compound of claim 27, or a pharmaceutically acceptable salt thereof, wherein n1 and n2 are each independently 0 or 1. 29. The compound of claim 26, or a pharmaceutically acceptable salt thereof, wherein R12 and R14, at each occurrence, are independently halo, nitro, cyano, C1-4alkyl, C1-4haloalkyl,– OC1-4alkyl,–OC1-4haloalkyl,–NH2,–NH(C1-4alkyl),–N(C1-4alkyl)(C1-4alkyl),–NHC(O)C1- 4alkyl,–N(C1-4alkyl)C(O)C1-4alkyl,–NHC(O)OC1-4alkyl,–N(C1-4alkyl)C(O)OC1-4alkyl,– C(O)OC1-4alkyl,–C(O)OH,–C(O)NH2,–C(O)NH(C1-4alkyl), or–C(O)N(C1-4alkyl)(C1-4alkyl), and optionally two R12 or R14, together with the atoms to which they are attached form a fused

ring ;

provided that the compound is not 1-(2-(4-(4-ethyl-3,5-bis(4-methoxyphenyl)-1H-pyrazol-1- yl)phenoxy)ethyl)piperidine, or a salt thereof. 30. The compound of claim 26, or a pharmaceutically acceptable salt thereof, wherein R12 and R14, at each occurrence, are independently halo,–OH, or–OC1-4alkyl.

31. The compound of claim 30, or a pharmaceutically acceptable salt thereof, wherein R12 and R14, at each occurrence, are independently halo or–OC1-4alkyl. 32. The compound of claim 30, or a pharmaceutically acceptable salt thereof, wherein the compound is not 1-(2-(4-(4-ethyl-3,5-bis(4-methoxyphenyl)-1H-pyrazol-1- yl)phenoxy)ethyl)piperidine, or a salt thereof. 33. The compound of claim 26, or a pharmaceutically acceptable salt thereof, wherein G2 is formula (iva)

;

and n1 and n2 are each independently 0 or 1. 34. The compound of claim 2, or a pharmaceutically acceptable salt thereof, wherein n1, n2, and n3 are each independently 0, 1, 2, or 3. 35. The compound of claim 2, or a pharmaceutically acceptable salt thereof, wherein R4a, R4b, R4c, R5a, R6, R8, R9, R10, R12, R14, R15, R16, R18, R20, R22, R23, R24, R25, R26, R28, R30, and R32, at each occurrence, are independently halo, cyano, C1-4alkyl,–OH, or–OC1-4alkyl.

36. The compound of claim 35, or a pharmaceutically acceptable salt thereof, wherein n1, n2, and n3 are each independently 0 or 1. 37. The compound of claim 34, or a pharmaceutically acceptable salt thereof, wherein G2 is (via)

.

38. The compound of claim 34, or a pharmaceutically acceptable salt thereof, wherein G2 is

.

39. The compound of claim 38, or a pharmaceutically acceptable salt thereof, wherein: R22 is halo, cyano, C1-4alkyl, or–OC1-4alkyl;

R23 is halo, cyano, C1-4alkyl,–OH, or–OC1-4alkyl; and

n1 and n2 are each 1. 40. The compound of claim 38, or a pharmaceutically acceptable salt thereof, wherein: R22 is halo, cyano, C1-4alkyl,–OH, or–OC1-4alkyl;

R23 is halo, cyano, C1-4alkyl, or–OC1-4alkyl; and

n1 and n2 are each 1. 41. The compound of claim 38, or a pharmaceutically acceptable salt thereof, wherein: R22 is halo, cyano, C1-4alkyl, or–OC1-4alkyl;

R23 is halo, cyano, C1-4alkyl, or–OC1-4alkyl; and

n1 and n2 are each 1. 42. The compound of claim 34, or a pharmaceutically acceptable salt thereof, wherein G2 is (ixa)

.

43. The compound of claim 34, or a pharmaceutically acceptable salt thereof, wherein G2 is (xa) or (xiiia)

44. The compound of claim 34, or a pharmaceutically acceptable salt thereof, wherein G2 is (va), (viiia), or (xia)

45. The compound of claim 1 selected from the group consisting of

4,4'-(3-(4-(2-(piperidin-1-yl)ethoxy)phenyl)-1H-pyrazole-1,5-diyl)diphenol;

4,4'-(5-(4-(2-(piperidin-1-yl)ethoxy)phenyl)-1H-pyrazole-1,3-diyl)diphenol;

(E)-3-(4-(1,3-bis(4-hydroxyphenyl)-4-methyl-1H-pyrazol-5-yl)phenyl)-1-(piperidin-1-yl)prop-2- en-1-one;

4,4'-(4-methyl-5-(4-(3-(piperidin-1-yl)propyl)phenyl)-1H-pyrazole-1,3-diyl)diphenol; 4,4'-(5-(4-(3-(piperidin-1-yl)propoxy)phenyl)-1H-pyrazole-1,3-diyl)diphenol;

4,4'-(5-(4-(3-cyclohexylpropoxy)phenyl)-1H-pyrazole-1,3-diyl)diphenol;

1-(2-(4-(1,3-bis(4-methoxyphenyl)-1H-pyrazol-5-yl)phenoxy)ethyl)piperidine;

1-(2-(4-(1,3-bis(4-methoxyphenyl)-4-methyl-1H-pyrazol-5-yl)phenoxy)ethyl)piperidine;

1-(3-(4-(1,3-bis(4-methoxyphenyl)-4-methyl-1H-pyrazol-5-yl)phenyl)propyl)piperidine;

1-(3-(4-(1,3-bis(4-methoxyphenyl)-1H-pyrazol-5-yl)phenoxy)propyl)piperidine;

1-(2-(3-(1-phenyl-3-(p-tolyl)-1H-pyrazol-5-yl)phenoxy)ethyl)piperidine;

1-(2-(4-(3-(4-methoxyphenyl)-1-phenyl-1H-pyrazol-5-yl)phenoxy)ethyl)piperidine;

4-(1-phenyl-5-(4-(2-(piperidin-1-yl)ethoxy)phenyl)-1H-pyrazol-3-yl)phenol;

1-(2-(4-(1,3-diphenyl-1H-pyrazol-5-yl)phenoxy)ethyl)piperidine;

1-(2-(4-(1-(4-chlorophenyl)-3-(4-methoxyphenyl)-1H-pyrazol-5-yl)phenoxy)ethyl)piperidine; 4-(1-(4-chlorophenyl)-5-(4-(2-(piperidin-1-yl)ethoxy)phenyl)-1H-pyrazol-3-yl)phenol;

1-(2-(4-(1-phenyl-3-(3,4,5-trimethoxyphenyl)-1H-pyrazol-5-yl)phenoxy)ethyl)piperidine;

4,4'-(4-ethyl-1-(4-(2-(pyrrolidin-1-yl)ethoxy)phenyl)-1H-pyrazole-3,5-diyl)diphenol;

4,4'-(5-ethyl-4-(4-(2-(piperidin-1-yl)ethoxy)phenyl)-1H-pyrazole-1,3-diyl)diphenol;

4,4'-(4-(4-(2-(piperidin-1-yl)ethoxy)phenyl)-5-propyl-1H-pyrazole-1,3-diyl)diphenol;

4-(1-phenyl-3-(4-(2-(piperidin-1-yl)ethoxy)phenyl)-1H-pyrazol-5-yl)phenol;

4-(5-phenyl-3-(4-(2-(piperidin-1-yl)ethoxy)phenyl)-1H-pyrazol-1-yl)phenol;

3-(1-(4-hydroxyphenyl)-3-(4-(2-(piperidin-1-yl)ethoxy)phenyl)-1H-pyrazol-5-yl)phenol;

4-(3-(4-(2-(piperidin-1-yl)ethoxy)phenyl)-1-(p-tolyl)-1H-pyrazol-5-yl)phenol;

4-(1-(4-chlorophenyl)-3-(4-(2-(piperidin-1-yl)ethoxy)phenyl)-1H-pyrazol-5-yl)phenol;

2,6-dimethoxy-4-(1-phenyl-3-(4-(2-(piperidin-1-yl)ethoxy)phenyl)-1H-pyrazol-5-yl)phenol; N-(4-(1-(4-hydroxyphenyl)-3-(4-(2-(piperidin-1-yl)ethoxy)phenyl)-1H-pyrazol-5- yl)phenyl)acetamide;

1-(2-(4-(1,5-bis(4-methoxyphenyl)-1H-pyrazol-3-yl)phenoxy)ethyl)piperidine;

1-(2-(4-(5-(4-methoxyphenyl)-1-phenyl-1H-pyrazol-3-yl)phenoxy)ethyl)piperidine;

1-(2-(4-(1-(3-methoxyphenyl)-5-(4-methoxyphenyl)-1H-pyrazol-3-yl)phenoxy)ethyl)piperidine; 1-(2-(4-(5-(4-methoxyphenyl)-1-(p-tolyl)-1H-pyrazol-3-yl)phenoxy)ethyl)piperidine;

1-(2-(4-(5-(4-methoxyphenyl)-1-(4-(trifluoromethoxy)phenyl)-1H-pyrazol-3- yl)phenoxy)ethyl)piperidine;

1-(2-(4-(1-(4-chlorophenyl)-5-(4-methoxyphenyl)-1H-pyrazol-3-yl)phenoxy)ethyl)piperidine; 1-(2-(4-(5-(4-methoxyphenyl)-1-(4-nitrophenyl)-1H-pyrazol-3-yl)phenoxy)ethyl)piperidine; tert-butyl (4-(5-(4-methoxyphenyl)-3-(4-(2-(piperidin-1-yl)ethoxy)phenyl)-1H-pyrazol-1- yl)phenyl)carbamate;

4-(5-(4-methoxyphenyl)-3-(4-(2-(piperidin-1-yl)ethoxy)phenyl)-1H-pyrazol-1-yl)aniline;

1-(2-(4-(1-(4-methoxyphenyl)-5-phenyl-1H-pyrazol-3-yl)phenoxy)ethyl)piperidine;

1-(2-(4-(5-(2,4-dimethoxyphenyl)-1-(4-methoxyphenyl)-1H-pyrazol-3- yl)phenoxy)ethyl)piperidine;

N-(4-(1-(4-methoxyphenyl)-3-(4-(2-(piperidin-1-yl)ethoxy)phenyl)-1H-pyrazol-5- yl)phenyl)acetamide;

1-(2-(4-(1-(4-methoxyphenyl)-5-(3,4,5-trimethoxyphenyl)-1H-pyrazol-3- yl)phenoxy)ethyl)piperidine;

1-(2-(4-(5-(3,4-dimethoxyphenyl)-1-(4-methoxyphenyl)-1H-pyrazol-3- yl)phenoxy)ethyl)piperidine;

1-(2-(4-(5-(3-chloro-4-methoxyphenyl)-1-(4-methoxyphenyl)-1H-pyrazol-3- yl)phenoxy)ethyl)piperidine;

1-(2-(4-(5-(4-methoxy-3,5-dimethylphenyl)-1-(4-methoxyphenyl)-1H-pyrazol-3- yl)phenoxy)ethyl)piperidine;

methyl 4-(1-phenyl-3-(4-(2-(piperidin-1-yl)ethoxy)phenyl)-1H-pyrazol-5-yl)benzoate;

4-(1-phenyl-3-(4-(2-(piperidin-1-yl)ethoxy)phenyl)-1H-pyrazol-5-yl)benzoic acid;

4-(1-phenyl-3-(4-(2-(piperidin-1-yl)ethoxy)phenyl)-1H-pyrazol-5-yl)benzamide;

4-(1-phenyl-3-(4-(2-(piperidin-1-yl)ethoxy)phenyl)-1H-pyrazol-5-yl)benzonitrile;

4-(1-phenyl-3-(4-(2-(piperidin-1-yl)ethoxy)phenyl)-1H-pyrazol-5-yl)aniline;

methyl (4-(1-phenyl-3-(4-(2-(piperidin-1-yl)ethoxy)phenyl)-1H-pyrazol-5-yl)phenyl)carbamate; N-(4-(1-phenyl-3-(4-(2-(piperidin-1-yl)ethoxy)phenyl)-1H-pyrazol-5-yl)phenyl)acetamide; N-(4-(1-phenyl-3-(4-(2-(piperidin-1-yl)ethoxy)phenyl)-1H-pyrazol-5-yl)phenyl)propionamide; 1-(2-(4-(5-(3,4-dimethoxyphenyl)-1-phenyl-1H-pyrazol-3-yl)phenoxy)ethyl)piperidine;

1-(2-(4-(1-phenyl-5-(3,4,5-trimethoxyphenyl)-1H-pyrazol-3-yl)phenoxy)ethyl)piperidine;

1-(2-(4-(1-phenyl-5-(2,4,5-trimethoxyphenyl)-1H-pyrazol-3-yl)phenoxy)ethyl)piperidine;

1-(2-(4-(1-phenyl-5-(2,4,6-trimethoxyphenyl)-1H-pyrazol-3-yl)phenoxy)ethyl)piperidine;

1-(2-(4-(1-phenyl-5-(2,3,4-trimethoxyphenyl)-1H-pyrazol-3-yl)phenoxy)ethyl)piperidine;

1-(2-(4-(5-(4-ethoxy-3,5-dimethoxyphenyl)-1-phenyl-1H-pyrazol-3-yl)phenoxy)ethyl)piperidine; 1-(2-(4-(1-phenyl-5-(3,4,5-triethoxyphenyl)-1H-pyrazol-3-yl)phenoxy)ethyl)piperidine; 1-(2-(4-(5-(4-ethoxy-3-methoxyphenyl)-1-phenyl-1H-pyrazol-3-yl)phenoxy)ethyl)piperidine; 1-(2-(4-(5-(7-methoxybenzo[d][1,3]dioxol-5-yl)-1-phenyl-1H-pyrazol-3- yl)phenoxy)ethyl)piperidine;

1-(2-(4-(5-(4-methoxy-3,5-dimethylphenyl)-1-phenyl-1H-pyrazol-3- yl)phenoxy)ethyl)piperidine;

1-(2-(4-(5-(3-chloro-4-methoxyphenyl)-1-phenyl-1H-pyrazol-3-yl)phenoxy)ethyl)piperidine; 1-(2-(4-(5-(6-methoxynaphthalen-2-yl)-1-(4-methoxyphenyl)-1H-pyrazol-3- yl)phenoxy)ethyl)piperidine;

1-(2-(4-(1-(4-methoxyphenyl)-5-(thiophen-3-yl)-1H-pyrazol-3-yl)phenoxy)ethyl)piperidine; 4,4'-(3-(4-(3-(piperidin-1-yl)propyl)phenyl)-1H-pyrazole-1,5-diyl)diphenol;

1-(3-(4-(1,5-bis(4-methoxyphenyl)-1H-pyrazol-3-yl)phenyl)propyl)piperidine;

1-(3-(4-(1,5-bis(4-methoxyphenyl)-1H-pyrazol-3-yl)phenoxy)propyl)piperidine;

3-(4-(2-(1H-pyrazol-1-yl)ethoxy)phenyl)-1,5-bis(4-methoxyphenyl)-1H-pyrazole;

4-(1-methyl-3-(4-(2-(piperidin-1-yl)ethoxy)phenyl)-1H-pyrazol-5-yl)phenol;

1-(2-(4-(5-(4-methoxyphenyl)-1-methyl-1H-pyrazol-3-yl)phenoxy)ethyl)piperidine;

1-(2-(4-(4,5-bis(4-methoxyphenyl)-1H-imidazol-2-yl)phenoxy)ethyl)piperidine;

1-(2-(4-(4,5-bis(4-methoxyphenyl)-1-methyl-1H-imidazol-2-yl)phenoxy)ethyl)piperidine; 1-(2-(4-(1-ethyl-4,5-bis(4-methoxyphenyl)-1H-imidazol-2-yl)phenoxy)ethyl)piperidine; 1-(2-(4-(4,5-di-p-tolyl-1H-imidazol-2-yl)phenoxy)ethyl)piperidine;

1-(2-(4-(4,5-bis(4-bromophenyl)-1H-imidazol-2-yl)phenoxy)ethyl)piperidine;

1-(2-(4-(4,5-bis(4-fluorophenyl)-1H-imidazol-2-yl)phenoxy)ethyl)piperidine;

1-(2-(4-(1-ethyl-4,5-diphenyl-1H-imidazol-2-yl)phenoxy)ethyl)piperidine;

4,4'-(2-(4-(2-(piperidin-1-yl)ethoxy)phenyl)-1H-imidazole-4,5-diyl)diphenol;

1-(2-(4-(1,4-bis(4-methoxyphenyl)-1H-imidazol-2-yl)phenoxy)ethyl)piperidine;

1-phenyl-2-(4-(2-(piperidin-1-yl)ethoxy)phenyl)-1H-benzo[d]imidazole;

1-methyl-2-(4-(2-(piperidin-1-yl)ethoxy)phenyl)-1H-benzo[d]imidazole;

5-(4-methoxyphenyl)-3-(4-(2-(piperidin-1-yl)ethoxy)phenyl)isoxazole;

5-phenyl-3-(4-(2-(piperidin-1-yl)ethoxy)phenyl)isoxazole;

3-phenyl-5-(4-(2-(piperidin-1-yl)ethoxy)phenyl)isoxazole;

5-(4-(2-(piperidin-1-yl)ethoxy)phenyl)-3-(p-tolyl)isoxazole; 3-(4-fluorophenyl)-5-(4-(2-(piperidin-1-yl)ethoxy)phenyl)isoxazole;

3-(naphthalen-2-yl)-5-(4-(2-(piperidin-1-yl)ethoxy)phenyl)isoxazole;

3-(4-bromophenyl)-5-(4-(2-(piperidin-1-yl)ethoxy)phenyl)isoxazole;

5-(4-methoxyphenyl)-3-(1-(2-(piperidin-1-yl)ethyl)-1H-benzo[d]imidazol-5-yl)-1,2,4- oxadiazole;

4-(3-(1-(2-(pyrrolidin-1-yl)ethyl)-1H-benzo[d]imidazol-5-yl)-1,2,4-oxadiazol-5-yl)benzonitrile; 4,5-bis(4-methoxyphenyl)-2-(4-(2-(piperidin-1-yl)ethoxy)phenyl)thiazole; and

4,5-bis(4-methoxyphenyl)-2-(4-(2-(piperidin-1-yl)ethoxy)phenyl)oxazole;

or a pharmaceutically acceptable salt thereof. 46. A pharmaceutical composition comprising a compound of claim 2, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier. 47. A method of treating a disease or condition mediated by Toll-like receptor activation comprising administering to a subject, in need thereof, a therapeutically effective amount of a compound of claim 2, or a pharmaceutically acceptable salt thereof. 48. The method of claim 47, wherein the disease or condition is an inflammatory disease or condition. 49. The method of claim 47, wherein the disease or condition is selected from bacterial sepsis, autoimmune disease, lupus erythematosus, ischemia-reperfusion injury, stroke, metabolic disease, obesity-related metabolic inflammation, gout, and cancer. 50. A kit comprising the compound of claim 2, or a pharmaceutically acceptable salt thereof.

Description:
TOLL-LIKE RECEPTOR SIGNALING INHIBITORS RELATED APPLICATIONS

[0001] This application claims priority to U.S. provisional application no.62/613,137, filed January 3, 2018, which is incorporated herein by reference in its entirety. TECHNICAL FIELD

[0002] The present invention relates to Toll-like receptor signaling inhibitors useful in the treatment of diseases or conditions mediated by Toll-like receptors (e.g., inflammatory diseases or conditions). BACKGROUND [0003] Toll-like receptors (TLRs), comprising a family of 10 functional receptors in humans, are a critical part of the innate pathogen recognition system (Kawai et al. Nature immunology 11, 373-384 (2010)). TLRs recognize different pathogen- and host-derived molecules and mediate cell activation and inflammation in order to initiate immune responses (Kumar et al.,

Biochemical and biophysical research communications 388, 621-625 (2009); Foster et al., Clinical immunology 130, 7-15 (2009)). TLRs are a key means by which mammals recognize and mount an immune response to foreign molecules and also provide a means by which the innate and adaptive immune responses are linked.

[0004] TLRs are expressed on different cell types, in particular on innate immune cells, which represent a first line host defense. TLR-induced effector mechanisms include a large number of cytotoxic and inflammatory mediators, e.g. nitric oxide, tumor necrosis factor Į (TNFĮ) and interleukin-1. These mediators act on various cell types, including endothelial cells and other immune cells, collectively supporting the ensuing inflammatory immune response (R. Medzhitov, Origin and physiological roles of inflammation. Nature 454, 428-435 (2008)).

[0005] TLRs have been shown to play a role in the pathogenesis of many diseases, including autoimmunity, infectious diseases, and inflammatory diseases. Exaggerated or prolonged TLR- mediated inflammation can lead to etiologically diverse diseases, including acute diseases, such as bacterial sepsis and ischemia reperfusion injury during stroke, but also chronic diseases, such as systemic lupus erythematosus (SLE) or obesity-related metabolic inflammation (Poltorak et al., Science 282, 2085-2088 (1998); Knapp, Wiener medizinische Wochenschrift 160, 107-111 (2010); Vilahur and Badimon, Frontiers in physiology 5, 496 (2014); Joosten et al., Nature reviews. Rheumatology 12, 344-357 (2016); Tall and Yvan-Charvet, Nature reviews.

Immunology 15, 104-116 (2015)).

[0006] Some TLRs are located on the cell surface to detect and initiate a response to extracellular pathogens and other TLRs are located in the endosomal compartment of the cell to detect and initiate a response to engulfed pathogens. The table below summarizes the TLRs, their cellular location, and known agonists.

TLR Molecule Agonist Cell Surface TLRs:

TLR2 bacterial lipopeptides

TLR4 gram negative bacteria

TLR5 motile bacteria

TLR6 gram positive bacteria

Endosomal TLRs:

TLR3 double stranded RNA viruses TLR7 single stranded RNA viruses TLR8 single stranded RNA viruses TLR9 unmethylated DNA

[0007] Key signaling proteins of TLRs are shared with members of the IL-1R family, including the IL-1R and IL-18R, owing to a common cytoplasmic domain, the TLR-IL-1R domain (TIR) (Gay and Keith, Nature 351, 355-356 (1991); Wesche et al., Immunity 7, 837-847 (1997); Adachi et al., Immunity 9, 143-150 (1998)). Members of the IL-1 family are important inflammatory mediators of TLRs, but also of other inflammatory receptors, such as Nod-like receptors (NLR) (He et al., Trends in biochemical sciences 41, 1012-1021 (2016)). As such, possible therapeutic indications of small molecule inhibitors of TLR/IL-1R signaling pathways extend beyond TLR-mediated inflammation to other diseases, e.g. NLR-mediated gout (Chen et al., The Journal of clinical investigation 116, 2262-2271 (2006); Martinon et al., Nature 440, 237-241 (2006); Pope and Tschopp, Arthritis and rheumatism 56, 3183-3188 (2007)).

[0008] One factor impeding TLR drug development is the signaling mechanism involved, which relies largely on protein-protein interactions (PPI) that are inherently difficult for drug targeting. Homotypic protein interactions between the TLR TIR-domain and the TIR-containing intracellular adaptor protein MyD88 transduce signaling (Wesche et al., Immunity 7, 837-847 (1997); Muzio et al., Science 278, 1612-1615 (1997); Medzhitov et al., Molecular cell 2, 253- 258 (1998)). In most cases, MyD88 binds directly to the intracellular TLR moiety, while in case of TLR2 and TLR4 the TIR-containing protein TIRAP/MAL appears to act as molecular bridge between TLR and MyD88 (Yamamoto et al., Nature 420, 324-329 (2002); Fitzgerald et al., Nature 413, 78-83 (2001); Horng et al., Nature immunology 2, 835-841 (2001)). An exception to this rule is the double stranded RNA-receptor TLR3, whose signaling does not depend on MyD88, but is mediated by two other TIR-containing proteins, i.e. TRAM and TRIF (Yamamoto et al., S. Science 301, 640-643 (2003); Oshiumi et al., Nature immunology 4, 161-167 (2003); Oshiumi et al., The Journal of biological chemistry 278, 49751-49762 (2003); Fitzgerald et al., The Journal of experimental medicine 198, 1043-1055 (2003); Yamamoto et al., Nature immunology 4, 1144-1150 (2003)). This‘TRIF-pathway’ is also used by TLR4, in addition to the MyD88-pathway; however, the MyD88-pathway is still important for inflammation mediated by TLR4, e.g. LPS-induced sepsis, highlighting the pivotal role of the MyD88-pathway as anti- inflammatory TLR/ IL-1R-target (Kawai et al., Immunity 11, 115-122 (1999)).

[0009] TLR activation induces oligomerization of MyD88, which results in sequential recruitment of IRAK family members via death domain interactions (Wesche et al., Immunity 7, 837-847 (1997); Lin et al., Nature 465, 885-890 (2010)). IRAK family members, in turn, bind and oligomerize TRAF6, which transduces and diversifies signal transduction towards downstream signaling pathways, such as the NF-kB and mitogen-activated protein kinase (MAPK) pathways (Hacker et al., Nature 439, 204-207 (2006); Cao et al., Nature 383, 443-446 (1996); Gohda et al., Journal of immunology 173, 2913-2917 (2004)). The function of all mentioned signaling proteins acting upstream of TRAF6 appears largely restricted to TLR/IL-1R members, while TRAF6 itself and proteins downstream thereof are used by various receptors and signaling pathways, e.g. members of the TNFR family (Walsh et al., Immunological reviews 266, 72-92 (2015)). Consistent with the restricted function of TLR/IL-1R-specific proteins in immune defense and inflammation, mice deficient in key molecules, such as MyD88, do not show overt adverse symptoms unless challenged with infectious agents. In turn, these mice are protected from inflammation pathology, e.g. during septic shock. MyD88-deficient humans have been identified. Such individuals are more susceptible to bacterial infections during childhood, but largely unaffected as adults, suggesting compensatory mechanisms via other parts of the immune system.

[0010] Still, despite the pressing need, no TLR-inhibitors are currently clinically available, at least in part due to the specific mode of signal transduction involved. SUMMARY

[0011] The present invention provides compounds or a pharmaceutically acceptable salt thereof and the methods, compositions and kits disclosed herein for treating a disease or condition mediated by Toll-like receptors and other receptors that activate cells through proteins containing TIR domains. In one aspect, the invention provides compounds of formula (I), or a pharmaceutically acceptable salt thereof,

wherein:

[0012] G 1 is–NR 1 R 2 ,–OH,–OC 1-4 alkyl, a C 3-8 cycloalkyl, a 4- to 12-membered heterocyclyl containing 1-3 heteroatoms independently selected from N, O, and S, or a 5- to 12-membered heteroaryl containing 1-3 heteroatoms independently selected from N, O, and S, wherein the cycloalkyl, the heterocyclyl, and the heteroaryl are optionally substituted with 1-4 substituents independently selected from halo, C1-4alkyl, C1-4haloalkyl,–OH,–OC1-4alkyl, and oxo;

[0013] R 1 and R 2 are each independently hydrogen or C1-4alkyl;

[0014] L 1 is–C 1-5 alkylene–,–C 1-4 alkylene–O–, or–C(O)–CH=CH–, wherein the–C 1- 5 alkylene– and the–C 1-4 alkylene–O– are optionally substituted with 1-2 halogens or one

hydroxyl; or L 1 is , wherein the C1-4alkylene is bonded to G 1 and the imidazole is fused at the meta and para positions on the phenyl relative to G 2 ;

[0015] G 2 is selected from (i) to (xiii)

R 11 and R 31 are each independently selected from hydrogen, C 1-4 alkyl, and phenyl optionally substituted with 1-3 substituents independently selected from halo, C 1-4 alkyl, C 1- d–OC1-4alkyl;

R 4c , R 5a , R 6 , R 8 , R 9 , R 10 , R 12 , R 14 , R 15 , R 16 , R 18 , R 20 , R 22 , R 23 , R 24 , R 25 , R 26 , each occurrence, are independently halo, nitro, cyano, C1-4alkyl, C1- 4haloalkyl,–OH,–OC1-4alkyl,–OC1-4haloalkyl,–NH2,–N H(C1-4alkyl),–N(C1-4alkyl)(C1-4alkyl), –NHC(O)C 1-4 alkyl,–N(C 1-4 alkyl)C(O)C 1-4 alkyl,–NHC(O)OC 1-4 alkyl,–N(C 1-4 alkyl)C(O)OC 1- 4 alkyl,–C(O)OC 1-4 alkyl,–C(O)OH,–C(O)NH 2 ,–C(O)NH(C 1-4 alkyl), or–C(O)N(C 1-4 alkyl)(C 1- 4 alkyl), and optionally two R 4a , R 4b , R 5a , R 6 , R 8 , R 9 , R 10 , R 12 , R 14 , R 15 , R 16 , R 18 , R 20 , R 21 , R 22 , R 24 , R 25 , R 26 , R 28 , R 30 , or R 32 , together with the atoms to which they are attached form a fused ring

[0023] n1 and n2 are independently 0, 1, 2, 3, 4, or 5; and

[0024] n3 and n4 are independently 0, 1, 2, or 3.

[0025] Another aspect of the present invention provides pharmaceutical compositions comprising a pharmaceutically acceptable carrier and therapeutically effective amounts of a compound of formula (I), or a pharmaceutically acceptable salt thereof.

[0026] Another aspect of the invention provides a method of treating a disease or condition mediated by Toll-like receptor activation comprising administering to a subject, in need thereof, a therapeutically effective amount of a compound of formula (I), or a pharmaceutically acceptable salt or a pharmaceutical composition thereof.

[0027] In another aspect, the invention provides compounds of formula (I), or a

pharmaceutically acceptable salt thereof, for use in treating a disease or condition mediated by Toll-like receptor activation.

[0028] In another aspect, the invention provides the use of a compound of formula (I), or a pharmaceutically acceptable salt thereof, for the manufacture of a medicament for the treatment a disease or condition mediated by Toll-like receptor activation.

[0029] The invention also provides kits comprising compounds of formula (I). BRIEF DESCRIPTION OF THE DRAWINGS

[0030] FIG.1A shows a model of the MyD88 signaling pathway activated by different TLR and IL-1R family members.

[0031] FIG.1B shows Coumermycin (CM)-induced NF-kB activity of HEK293T reporter cells equipped with NF-kB luciferase reporter and indicated GyrB-fusion proteins.

[0032] FIG.1C shows MyD88 expression in TIRAP-GyrB-expressing HEK293T cells that were infected with lentivirus expressing CAS9 and control sgRNA (ctrl) or sgRNA against MyD88 (MyD) and analyzed by immuno-blotting with antibodies against MyD88 and ȕACTIN.

[0033] FIG.1D shows NF-kB activity of TIRAP-GyrB-expressing HEK293T cells with loss of MyD88 expression (shown in (C)) that were stimulated with CM, IL-1ȕ or TNFĮ.

[0034] FIG.1E shows Calculation of Z’ values reflecting within plate- and between-plate variation of HEK293T reporter cell lines that were treated on HTS robots on independent plates at different time points with CM in the presence of PS1145 and staurosporine, followed by analysis of pathway-specific (luciferase activity) and non-specific activity (Alamar blue conversion).

[0035] FIG.2A shows a screening paradigm for HTS based on inducible TLR signaling proteins.

[0036] FIG.2B shows HTS-derived dose response curve of MPP (Methyl Piperidino Pyrazole, a non-steroidal ligand for the estrogen receptor beta) (Stauffer et al., J. Med. Chem. 2000, 43, 4934-4947) for NF-kB activity and AB-based toxicity using indicated reporter cell lines.

[0037] FIG.2C shows TNFĮ release of bone marrow-derived macrophages (BMM) (from C57BL/6 mice) stimulated with TLR agonists for six hours in the presence of different concentrations of MPP.

[0038] FIG.2D shows Immuno-blot analysis of CpG-DNA-, R848- and Curdlan-induced NF-kB and MAPK activity in BMM in the presence or absence of MPP (10 mM) as reflected by IkBĮ degradation and phosphorylation of p38, JNK1/2 and ERK1/2, respectively. Antibodies against total p38 were used as loading control.

[0039] FIG.2E shows TNFĮ release from BMM derived from wildtype (WT) and ERĮ- deficient mice that were treated for six hours with CpG-DNA in the presence of different concentrations of MPP. [0040] FIG.2F shows Immuno-blot analysis of IkBĮ and MAPK-phosphorylation in BMM derived from ERĮ-deficient mice that were treated for 20 min with CpG-DNA in the presence or absence of MPP (10 mM). Antibodies against total p38 were used as loading control.

[0041] FIG.3A shows quantitative MS analysis of proteins co-purifying with MyD88 in non-stimulated and CpG-DNA-stimulated RAW264.7 cells in the absence or presence of 10 mM MPP. The fold-ratio of MyD88-associated proteins purified from CpG-DNA-treated vs. control cells (open bars) and CpG-DNA plus MPP-treated vs. control cells (closed bars) is shown. Numbers in parenthesis indicate the number of unique peptides identified.

[0042] FIG.3B shows an immuno-blot (IB) analysis of indicated proteins co-immuno- purifying with MyD88 in MyD88-GyrB-F (Flag)-expressing RAW264.7 cells treated with CpG- DNA (45 min) ± 10 mM MPP (Endo = endogenous).

[0043] FIG.3C shows an immuno-blot (IB) analysis of indicated proteins co-immuno- purifying with MyD88 in MyD88-GyrB-F (Flag)-expressing RAW264.7 cells treated with CM (20 min) ± 10 mM MPP (Endo = endogenous).

[0044] FIG.3D shows co-IP analyses of different forms of MyD88 in the absence or presence of 10 mM MPP (IP, IP samples; Ly, total lysates).

[0045] FIG.3E shows CETSA (Cellular Thermal Shift Assay) of endogenous MyD88 in HEK293T cells that were exposed to indicated temperatures at 50 mM TSI-13-57 (TSI refers to Toll-like receptor Signaling Inhibitor).

[0046] FIG.3F shows CETSA of endogenous MyD88 in HEK293T cells that were exposed to indicated temperatures at 50 mM TSI-13-57.

[0047] FIG.3G shows CETSA of endogenous MyD88 in HEK293T cells that were exposed to various indicated TSI-13-57 concentrations at 43.8 °C.

[0048] FIG.3H shows CETSA of endogenous MyD88 in HEK293T cells that were exposed to various indicated TSI-13-57 concentrations at 43.8 °C.

[0049] FIG.4A shows activity of different TSI against TLR- and Curdlan-induced TNFĮ release. IC50 values for inhibition of TNFĮ release and AB conversion (toxicity) are shown.

[0050] FIG.4B shows mammalian two-hybrid (M2H) assay based on indicated M2H protein pairs during exposure to TSI-13-48 and TSI-13-57 at indicated concentrations. Inhibition of Gal4/VP16-mediated luciferase activity and AB conversion (toxicity) are shown.100% inhibition corresponds to values obtained in the presence of staurosporine. [0051] FIG.5A shows plasma TNFĮ levels of mice that were pre-treated for 60 minutes with TSI-2013-57 or vehicle control, followed by LPS challenge for 90min. TNFĮ levels in vehicle control mice (100%) were 0.432 ± 0.09 ng/ml. N = 5 mice per group. Data represent mean ± s.d.; **P < 0.01, ***P < 0.001 two-tailed Student's t test.

[0052] FIG.5B shows TSI-13-57 plasma concentration of the mice described in FIG.5A. DETAILED DESCRIPTION

1. Definitions

[0053] As described herein, compounds of the invention can optionally be substituted with one or more substituents, such as are illustrated generally above, or as exemplified by particular classes, subclasses, and species of the invention. As described herein, the variables in formula I encompass specific groups, such as, for example, alkyl and cycloalkyl. As one of ordinary skill in the art will recognize, combinations of substituents envisioned by this invention are those combinations that result in the formation of stable or chemically feasible compounds. The term "stable," as used herein, refers to compounds that are not substantially altered when subjected to conditions to allow for their production, detection, and preferably their recovery, purification, and use for one or more of the purposes disclosed herein. In some embodiments, a stable compound or chemically feasible compound is one that is not substantially altered when kept at a temperature of 40°C or less, in the absence of moisture or other chemically reactive conditions, for at least a week.

[0054] The term "alkyl" as used herein, means a straight or branched chain saturated hydrocarbon. Representative examples of alkyl include, but are not limited to, methyl, ethyl, npropyl, isopropyl, n-butyl, sec-butyl, isobutyl, tert-butyl, n-pentyl, isopentyl, neopentyl, n- hexyl, 3-methylhexyl, 2,2-dimethylpentyl, 2,3-dimethylpentyl, n-heptyl, n-octyl, n-nonyl, and n- decyl.

[0055] The term "alkylene," as used herein, means a divalent group derived from a straight or branched chain saturated hydrocarbon. Representative examples of alkylene include, but are not limited to, -CH 2 -, -CH 2 CH 2 -, -CH 2 CH 2 CH 2 -, -CH 2 CH(CH 3 )CH 2 -, and

CH 2 CH(CH 3 )CH(CH 3 )CH 2 -.

[0056] The term "aryl," as used herein, means phenyl or a bicyclic aryl. The bicyclic aryl is naphthyl, dihydronaphthalenyl, tetrahydronaphthalenyl, indanyl, or indenyl. The phenyl and bicyclic aryls are attached to the parent molecular moiety through any carbon atom contained within the phenyl or bicyclic aryl.

[0057] The term "halogen" means a chlorine, bromine, iodine, or fluorine atom.

[0058] The term "haloalkyl," as used herein, means an alkyl, as defined herein, in which one, two, three, four, five, six, or seven hydrogen atoms are replaced by halogen. For example, representative examples of haloalkyl include, but are not limited to, 2-fluoroethyl,

difluoromethyl, trifluoromethyl, 2,2,2-trifluoroethyl, 2,2,2-trifluoro-1, 1-dimethylethyl, and the like.

[0059] The term "heteroaryl," as used herein, means an aromatic heterocycle, i.e., an aromatic ring that contains at least one heteroatom selected from O, N, or S. A heteroaryl may contain from 5 to 12 ring atoms. A heteroaryl may be a 5- to 6-membered monocyclic heteroaryl or an 8- to 12-membered bicyclic heteroaryl. A 5-membered monocyclic heteroaryl ring contains two double bonds, and one, two, three, or four heteroatoms as ring atoms. Representative examples of 5-membered monocyclic heteroaryls include, but are not limited to, furanyl, imidazolyl, isoxazolyl, isothiazolyl, oxadiazolyl, oxazolyl, pyrazolyl, pyrrolyl, tetrazolyl, thiadiazolyl, thiazolyl, thienyl, and triazolyl. A 6-membered heteroaryl ring contains three double bonds, and one, two, three or four heteroatoms as ring atoms. Representative examples of 6-membered monocyclic heteroaryls include, but are not limited to, pyridinyl, pyridazinyl, pyrimidinyl, pyrazinyl, and triazinyl. The bicyclic heteroaryl is an 8- to 12-membered ring system having a monocyclic heteroaryl fused to an aromatic, saturated, or partially saturated carbocyclic ring, or fused to a second monocyclic heteroaryl ring. Representative examples of bicyclic heteroaryl include, but are not limited to, benzofuranyl, benzoxadiazolyl, 1,3- benzothiazolyl, benzimidazolyl, benzothienyl, indolyl, indazolyl, isoquinolinyl, naphthyridinyl, oxazolopyridine, quinolinyl, thienopyridinyl, 5 ,6, 7 ,8-tetrahydroquinolinyl, and 6, 7-dihydro- 5H-cyclopenta[b Jpyridinyl. The heteroaryl groups are connected to the parent molecular moiety through any substitutable carbon atom or any substitutable nitrogen atom contained within the groups.

[0060] The term "cycloalkyl" as used herein, means a monocyclic all-carbon ring containing zero heteroatoms as ring atoms, and zero double bonds. Examples of cycloalkyls include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl. The cycloalkyl groups described herein can be appended to the parent molecular moiety through any substitutable carbon atom.

[0061] The terms "heterocycle" or "heterocyclic" refer generally to ring systems containing at least one heteroatom as a ring atom where the heteroatom is selected from oxygen, nitrogen, and sulfur. In some embodiments, a nitrogen or sulfur atom of the heterocycle is optionally substituted with oxo. Heterocycles may be a monocyclic heterocycle, a fused bicyclic heterocycle, or a spiro heterocycle. The monocyclic heterocycle is generally a 4, 5, 6, 7, or 8- membered non-aromatic ring containing at least one heteroatom selected from O, N, or S. The 4- membered ring contains one heteroatom and optionally one double bond. The 5-membered ring contains zero or one double bond and one, two or three heteroatoms. The 6, 7, or 8-membered ring contains zero, one, or two double bonds, and one, two, or three heteroatoms. Representative examples of monocyclic heterocycle include, but are not limited to, azetidinyl, azepanyl, diazepanyl, 1,3-dioxanyl, 1,4-dioxanyl, 1,3-dioxolanyl , 4,5-dihydroisoxazol-5-yl, 3,4- dihydropyranyl, 1,3-dithiolanyl, 1,3-dithianyl, imidazolinyl, imidazolidinyl, isothiazolinyl, isothiazolidinyl, isoxazolinyl, isoxazolidinyl, morpholinyl, oxadiazolinyl, oxadiazolidinyl, oxazolinyl, oxazolidinyl, oxetanyl, piperazinyl, piperidinyl, pyranyl, pyrazolinyl, pyrazolidinyl, pyrrolinyl, pyrrolidinyl, tetrahydrofuranyl, tetrahydropyranyl, tetrahydrothienyl, thiadiazolinyl, thiadiazolidinyl, thiazolinyl, thiazolidinyl, thiomorpholinyl, 1,1-dioxidothiomorpholinyl, thiopyranyl, and trithianyl. The fused bicyclic heterocycle is a 7-12-membered ring system having a monocyclic heterocycle fused to a phenyl, to a saturated or partially saturated carbocyclic ring, or to another monocyclic heterocyclic ring, or to a monocyclic heteroaryl ring. Representative examples of fused bicyclic heterocycle include, but are not limited to, 1,3- benzodioxol-4-yl, 1,3-benzodithiolyl, 3-azabicyclo[3.1.0]hexanyl, hexahydro-1H-furo[3,4- c]pyrrolyl, 2,3-dihydro-1,4-benzodioxinyl, 2,3-dihydro-1-benzofuranyl, 2,3-dihydro-1- benzothienyl, 2,3-dihydro-1H-indolyl, 5,6,7,8-tetrahydroimidazo[1,2-a]pyrazinyl, and 1,2,3,4- tetrahydroquinolinyl. Spiro heterocycle means a 4-, 5-, 6-, 7-, or 8-membered monocyclic heterocycle ring wherein two of the substituents on the same carbon atom form a second ring having 3, 4, 5, 6, 7, or 8 members. Examples of a spiro heterocycle include, but are not limited to, 1,4-dioxa-8-azaspiro[4.5]decanyl, 2-oxa-7-azaspiro[3.5]nonanyl, 2-oxa-6- azaspiro[3.3]heptanyl, and 8-azaspiro[4.5]decane. The monocyclic heterocycle groups of the present invention may contain an alkylene bridge of 1, 2, or 3 carbon atoms, linking two nonadjacent atoms of the group. Examples of such a bridged heterocycle include, but are not limited to, 2,5-diazabicyclo[2.2.1]heptanyl, 2-azabicyclo[2.2.1]heptanyl, 2- azabicyclo[2.2.2]octanyl, and oxabicyclo[2.2.1]heptanyl. The monocyclic, fused bicyclic, and spiro heterocycle groups are connected to the parent molecular moiety through any substitutable carbon atom or any substitutable nitrogen atom contained within the group.

[0062] The term "oxo" as used herein refers to an oxygen atom bonded to the parent molecular moiety. An oxo may be attached to a carbon atom or a sulfur atom by a double bond. Alternatively, an oxo may be attached to a nitrogen atom by a single bond, i.e., an N-oxide.

[0063] Terms such as "alkyl," "cycloalkyl," "alkylene," etc. may be preceded by a designation indicating the number of atoms present in the group in a particular instance ( e.g., "C1-4alkyl," "C3-6cycloalkyl," "C1-4alkylene"). These designations are used as generally understood by those skilled in the art. For example, the representation "C" followed by a subscripted number indicates the number of carbon atoms present in the group that follows. Thus, "C3alkyl" is an alkyl group with three carbon atoms (i.e., n-propyl, isopropyl). Where a range is given, as in "C 1-4 ," the members of the group that follows may have any number of carbon atoms falling within the recited range. A "C 1-4 alkyl," for example, is an alkyl group having from 1 to 4 carbon atoms, however arranged (i.e., straight chain or branched).

[0064] Unless otherwise stated, structures depicted herein are also meant to include all isomeric (e.g., enantiomeric, diastereomeric, and geometric (or conformational)) forms of the structure; for example, the R and S configurations for each asymmetric center, (Z) and (E) double bond isomers, and (Z) and (E) conformational isomers. Therefore, single stereochemical isomers as well as enantiomeric, diastereomeric, and geometric ( or conformational) mixtures of the present compounds are within the scope of the invention. Unless otherwise stated, all tautomeric forms of the compounds of the invention are within the scope of the invention. Thus, included within the scope of the invention are tautomers of compounds of formula I. The structures also include zwitterioinc forms of the compounds or salts of formula I where appropriate. 2. Compounds

[0065] A first aspect of the invention provides compounds of formula (I), or a

pharmaceutically acceptable salt thereof, wherein G 1 , G 2 , and L 1 are as defined herein. The embodiments below include all combinations of the variables G 1 , G 2 , and L 1 and their sub- variables (e.g., R 1 , R 2 , R 3 , etc.).

[0066] In some embodiments, the compounds of formula (I) have formula (IA),

wherein G 1 , G 2 , and L 1 are as defined herein.

[0067] In some embodiments, G 1 is–NR 1 R 2 , a 4- to 12-membered heterocyclyl containing 1- 3 heteroatoms independently selected from N, O, and S, or a 5- to 12-membered heteroaryl containing 1-3 heteroatoms independently selected from N, O, and S, wherein the heterocyclyl and the heteroaryl are optionally substituted with 1-4 substituents independently selected from halo, C 1-4 alkyl, C 1-4 haloalkyl,–OH,–OC 1-4 alkyl, and oxo; and L 1 is–C 1-5 alkylene–or–C 1- 4alkylene–O–.

[0068] In some embodiments, R 1 and R 2 are hydrogen. In some embodiments, R 1 and R 2 are C1-4alkyl. In some embodiments, one of R 1 and R 2 is hydrogen and the other is C1-4alkyl.

[0069] In some embodiments, L 1 is–C1-5alkylene–, wherein the–C1-5alkylene– is optionally substituted with 1-2 halogens or one hydroxyl. In some embodiments, L 1 is–C 1-5 alkylene–. In some embodiments, L 1 is–C 2-4 alkylene–.

[0070] In some embodiments, L 1 is–C 1-4 alkylene–O–, wherein the–C 1-4 alkylene–O– is optionally substituted with 1-2 halogens or one hydroxyl. In some embodiments, L 1 is–C 1- 4alkylene–O–. In some embodiments, L 1 is–C2-3alkylene–O–. When L 1 is–C1-4alkylene–O– or –C2-3alkylene–O–, the alkylene portion of the–C1-4alkylene–O– or–C2-3alkylene–O– is bonded to G 1 and the oxygen atom is bonded to the phenyl of formula (I) or (IA), e.g., .

[0071] In some embodiments, L 1 is–C(O)–CH=CH–. In these embodiments, the–C(O)– portion of the–C(O)–CH=CH– is bonded to G 1 and the =CH– bonded to the phenyl of formula

(I) or (IA), e.g., . , wherein the C 1-4 alkylene is bonded to G 1 and the imidazole is fused at the meta and para positions on the phenyl relative

. In other words, L 1 is linked by ring fusion to the phenyl of formula (I) or (IA).

[0073] In some embodiments, L 1 is–C 1-5 alkylene– or–C 1-4 alkylene–O–. In some embodiments, L 1 is–C2-4alkylene–or–C2-3alkylene–O–.

[0074] In some embodiments, G 1 is–NR 1 R 2 ; a 4- to 8-membered monocyclic heterocyclyl containing a first nitrogen atom and optionally an additional heteroatom selected from N, O, and S, the heterocyclyl connecting to L 1 at the first nitrogen atom and optionally containing a C1- 3 alkylene bridge between two non-adjacent ring atoms and/or a double bond, the heterocyclyl being optionally substituted with 1-4 substituents independently selected from halo, C 1-4 alkyl, C 1-4 haloalkyl,–OH,–OC 1-4 alkyl, and oxo; or a 5- to 6-membered monocyclic heteroaryl containing 1-3 nitrogen atoms, the heteroaryl being optionally substituted with 1-4 substituents independently selected from halo, C1-4alkyl, C1-4haloalkyl,–OH, and–OC1-4alkyl; and L 1 is–C2- 4alkylene–or–C2-3alkylene–O–. [0075] In some embodiments, G 1 is–N(CH 3 ) 2 ,–N(CH 2 CH 3 ) 2 , pyrrolidin-1-yl, piperidin-1-yl, piperazin-1-yl, azepin-1-yl, pyrazol-1-yl, imidazol-1-yl, or triazol-1-yl.

[0076] In some embodiments, G 2 has formula (i)

[0077] wherein R 3 , R 4 , and R 5 are as defined herein.

[0078] In some embodiments, G 2 has formula (

and n2 are as defined herein.

[0079] In some embodiments, G 2 has formula (i) and R 5 is C1-4alkyl.

In embodiments wherein G 2 has formula (i), R 4 may

R 4a and n1 are as defined herein.

In further embodiments wherein G 2 has formula

at each occurrence, is independently halo, cyano, C1-4alkyl, C1-4haloalkyl,–OH,–OC1-4alkyl,– OC1-4haloalkyl,–NH2,–NH(C1-4alkyl),–N(C1-4alkyl)(C1-4a lkyl),–NHC(O)C1-4alkyl,–N(C1- 4alkyl)C(O)C1-4alkyl,–C(O)OC1-4alkyl,–C(O)NH2,–C(O)NH( C1-4alkyl), or–C(O)N(C1- 4alkyl)(C1-4alkyl), and optionally two R 4a , together with the atoms to which they are attached,

form a fused ring halo, nitro, cyano, C 1-4 alkyl, C 1- 4 haloalkyl,–OH,–OC 1-4 alkyl,–OC 1-4 haloalkyl,–NH 2 ,–NH(C 1-4 alkyl),–N(C 1-4 alkyl)(C 1-4 alkyl), –NHC(O)C 1-4 alkyl,–N(C 1-4 alkyl)C(O)C 1-4 alkyl,–NHC(O)OC 1-4 alkyl, or–N(C 1-4 alkyl)C(O)OC 1- 4alkyl; n1 is 0, 1, 2, or 3; and n2 is 0 or 1. [0082] In still further embodiments wherein G 2 has formula (i), R 4 is , ,

.

In any of the embodiments wherein G 2 has formula (i), are further embodiments where R 3 is hydrogen. In other embodiments, wherein G 2 has formula (i), the compound of formula (I) or (IA) is not 4,4'-(4-ethyl-3-(4-(2-(piperidin-1-yl)ethoxy)phenyl)-1H-pyra zole-1,5-diyl)diphenol.

In some embodiments, G 2 has formula (ii)

wherein R 6 , R 7 , R 8 , n1, and n2 are as defined herein.

[0086] In some embodiments wherein G 2 has formula (ii), n1 and n2 are each independently 0, 1, 2, or 3.

[0087] In some embodiments wherein G 2 has formula (ii), G 2 has formula (iia)

wherein R 7 is as defined herein.

In some embodiments, G 2 has formula (iii)

wherein R 9 , R 10 , R 11 , n1, and n2 are as defined herein.

[0089] In some embodiments wherein G 2 has formula (iii), n1 and n2 are each independently 0, 1, 2, or 3. In other embodiments, n1 and n2 are each independently 0 or 1.

[0090] In some embodiments wherein G 2 has formula (iii), R 9 and R 10 , at each occurrence, are independently halo, nitro, cyano, C 1-4 alkyl, C 1-4 haloalkyl,–OC 1-4 alkyl,–OC 1-4 haloalkyl,– NH 2 ,–NH(C 1-4 alkyl),–N(C 1-4 alkyl)(C 1-4 alkyl),–NHC(O)C 1-4 alkyl,–N(C 1-4 alkyl)C(O)C 1-4 alkyl, –NHC(O)OC 1-4 alkyl,–N(C 1-4 alkyl)C(O)OC 1-4 alkyl,–C(O)OC 1-4 alkyl,–C(O)OH,–C(O)NH 2 ,– C(O)NH(C1-4alkyl), or–C(O)N(C1-4alkyl)(C1-4alkyl), and optionally two R 9 or R 10 , together with

the atoms to which they are attached form a fused ring .

[0091] In other embodiments wherein G 2 has formula (iii), R 9 and R 10 , at each occurrence, are independently halo, C 1-4 alkyl,–OH, or–OC 1-4 alkyl. In further embodiments, R 9 , at each occurrence, is independently halo, C 1-4 alkyl, or–OC 1-4 alkyl; and R 10 , at each occurrence, is independently halo, C1-4alkyl,–OH, or–OC1-4alkyl. In still further embodiments, R 9 and R 10 , at each occurrence, are independently halo, C1-4alkyl, or–OC1-4alkyl.

[0092] In some embodiments wherein G 2 has formula (iii), G 2 has formula (iiia)

;

wherein n1 and n2 are each independently 0 or 1, and R 9 , R 10 , and R 11 are as defined herein. In formula (iiia), n1 or n2 being 0 refers to compounds wherein the R 9 or R 10 substituent, respectively, is replaced by hydrogen.

[0093] In any of the embodiments wherein G 2 has formula (iii) or (iiia), are further embodiments where R 11 is hydrogen.

[0094] In some embodiments, G 2 has formula (iv)

wherein R 12 , R 13 , R 14 , n1, and n2 are as defined herein.

[0095] In some embodiments wherein G 2 has formula (iv), n1 and n2 are each independently 0, 1, 2, or 3. In other embodiments, n1 and n2 are each independently 0 or 1. [0096] In some embodiments wherein G 2 has formula (iv), R 12 and R 14 , at each occurrence, are independently halo, nitro, cyano, C 1-4 alkyl, C 1-4 haloalkyl,–OC 1-4 alkyl,–OC 1-4 haloalkyl,– NH2,–NH(C1-4alkyl),–N(C1-4alkyl)(C1-4alkyl),–NHC(O)C1- 4alkyl,–N(C1-4alkyl)C(O)C1-4alkyl, –NHC(O)OC1-4alkyl,–N(C1-4alkyl)C(O)OC1-4alkyl,–C(O)OC1 -4alkyl,–C(O)OH,–C(O)NH2,– C(O)NH(C1-4alkyl), or–C(O)N(C1-4alkyl)(C1-4alkyl), and optionally two R 12 or R 14 , together

with the atoms to which they are attached form a fused ring ; provided that the compound is not 1-(2-(4-(4-ethyl-3,5-bis(4-methoxyphenyl)-1H-pyrazol-1- yl)phenoxy)ethyl)piperidine, or a salt thereof.

[0097] In other embodiments wherein G 2 has formula (iv), R 12 and R 14 , at each occurrence, are independently halo,–OH, or–OC1-4alkyl. In still other embodiments, R 12 and R 14 , at each occurrence, are independently halo or–OC 1-4 alkyl.

[0098] In some embodiments wherein G 2 has formula (iv), the compound of formula (I) or (IA) is not 1-(2-(4-(4-ethyl-3,5-bis(4-methoxyphenyl)-1H-pyrazol-1- yl)phenoxy)ethyl)piperidine, or a salt thereof.

[0099] In some embodiments, G 2 has formula (iva)

wherein n1 and n2 are each independently 0 or 1; and R 12 , R 13 , and R 14 are as defined herein. In formula (iva), n1 or n2 being 0 refers to compounds wherein the R 12 or R 14 substituent, respectively, is replaced by hydrogen. [00100] In some embodiments of formula (I) or (IA), are further embodiments where n1, n2, and n3 are each independently 0, 1, 2, or 3. In still further embodiments, n1, n2, and n3 are each independently 0 or 1.

[00101] In the embodiments of formula (I) or (IA), are further embodiments where R 4a , R 4b , R 4c , R 5a , R 6 , R 8 , R 9 , R 10 , R 12 , R 14 , R 15 , R 16 , R 18 , R 20 , R 22 , R 23 , R 24 , R 25 , R 26 , R 28 , R 30 , and R 32 , at each occurrence, may be independently halo, cyano, C1-4alkyl,–OH, or–OC1-4alkyl.

[00102] In some embodiments, G 2 has formula (v)

and R 15 , R 16 , R 17 , n1, and n2 are as defined herein.

[00103] In some embodiments, G 2 has formula (va)

wherein n1 and n2 are each independently 0 or 1; and R 15 , R 16 , and R 17 are as defined herein. In formula (va), n1 or n2 being 0 refers to compounds wherein the R 15 or R 16 substituent, respectively, is replaced by hydrogen.

[00104] In some embodiments, G 2 has formula (vi)

and R 18 , R 19 , R 20 , n1, and n2 are as defined herein.

[00105] In some embodiments wherein G 2 has formula (vi), G 2 has formula (via)

wherein n1 and n2 are each independently 0 or 1; and R 18 , R 19 , and R 20 are as defined herein. In formula (via), n1 or n2 being 0 refers to compounds wherein the R 18 or R 20 substituent, respectively, is replaced by hydrogen.

[00106] In some embodiments, G 2 has formula (vii)

and R 21 , R 22 , R 23 , n1, and n2 are as defined herein.

[00107] In some embodiments wherein G 2 has formula (vii), G 2 has formula (viia)

wherein n1 and n2 are each independently 0 or 1; and R 21 , R 22 , and R 23 are as defined herein. In formula (viia), n1 or n2 being 0 refers to compounds wherein the R 22 or R 23 substituent, respectively, is replaced by hydrogen.

[00108] In some embodiments where G 2 has formula (viia), are further embodiments where R 22 is halo, cyano, C1-4alkyl, or–OC1-4alkyl; R 23 is halo, cyano, C1-4alkyl,–OH, or–OC1-4alkyl; and n1 and n2 are each 1.

[00109] In some embodiments where G 2 has formula (viia), are further embodiments where R 22 is halo, cyano, C 1-4 alkyl,–OH, or–OC 1-4 alkyl; R 23 is halo, cyano, C 1-4 alkyl, or–OC 1-4 alkyl; and n1 and n2 are each 1.

[00110] In some embodiments where G 2 has formula (viia), are further embodiments where R 22 is halo, cyano, C1-4alkyl, or–OC1-4alkyl; R 23 is halo, cyano, C1-4alkyl, or–OC1-4alkyl; and n1 and n2 are each 1.

[00111] In some embodiments, G 2 has formula (viii)

and X 1 , R 24 , R 25 , n1, and n2 are as defined herein.

[00112] In some embodiments, G 2 has formula (viiia)

wherein n1 and n2 are each independently 0 or 1; and X 1 , R 24 , and R 25 are as defined herein. In formula (viiia), n1 or n2 being 0 refers to compounds wherein the R 24 or R 25 substituent, respectively, is replaced by hydrogen.

[00113] In some embodiments, G 2 has formula (ix)

and R 26 , R 27 , and n1 are as defined herein.

[00114] In some embodiments, G 2 has formula (ixa)

wherein n1 is 0 or 1; and R 26 and R 27 are as defined herein. In formula (ixa), n1 being 0 refers to compounds wherein the R 26 substituent is replaced by hydrogen.

[00115] In some embodiments, G 2 has formula (x)

and R 28 , R 29 , and n1 are as defined herein.

[00116] In some embodiments, G 2 has formula (xiii)

and R 32 , R 33 , and n3 are as defined herein.

[00117] In some embodiments, G 2 has formula (xa) or (xiiia)

wherein n1 and n3 are each independently 0 or 1; and R 28 , R 29 , R 32 , and R 33 are as defined herein. In formulas (xa) and (xiiia), n1 or n3 being 0 refers to compounds wherein the R 28 or R 32 substituent, respectively, is replaced by hydrogen.

[00118] In some embodiments, G 2 has formula (xi)

and R 30 and n1 are as defined herein.

[00119] In some embodiments, G 2 has formula (xia)

wherein n1 is 0 or 1; and R 30 is as defined herein. In formula (xia), n1 being 0 refers to compounds wherein the R 30 substituent is replaced by hydrogen. [00120] In some embodiments, G 2 has formula (xii)

and R 31 is as defined herein.

[00121] In some embodiments, the compound of formula (I) or (IA) is selected from

[00122] 4,4'-(5-(4-(2-(piperidin-1-yl)ethoxy)phenyl)-1H-pyrazole-1,3 -diyl)diphenol;

[00123] (E)-3-(4-(1,3-bis(4-hydroxyphenyl)-4-methyl-1H-pyrazol-5-yl) phenyl)-1-(piperidin- 1-yl)prop-2-en-1-one;

[00124] 4,4'-(4-methyl-5-(4-(3-(piperidin-1-yl)propyl)phenyl)-1H-pyr azole-1,3-diyl)diphenol;

[00125] 4,4'-(5-(4-(3-(piperidin-1-yl)propoxy)phenyl)-1H-pyrazole-1, 3-diyl)diphenol;

[00126] 4,4'-(5-(4-(3-cyclohexylpropoxy)phenyl)-1H-pyrazole-1,3-diyl )diphenol;

[00127] 1-(2-(4-(1,3-bis(4-methoxyphenyl)-1H-pyrazol-5-yl)phenoxy)et hyl)piperidine;

[00128] 1-(2-(4-(1,3-bis(4-methoxyphenyl)-4-methyl-1H-pyrazol-5- yl)phenoxy)ethyl)piperidine;

[00129] 1-(3-(4-(1,3-bis(4-methoxyphenyl)-4-methyl-1H-pyrazol-5- yl)phenyl)propyl)piperidine;

[00130] 1-(3-(4-(1,3-bis(4-methoxyphenyl)-1H-pyrazol-5-yl)phenoxy)pr opyl)piperidine;

[00131] 1-(2-(3-(1-phenyl-3-(p-tolyl)-1H-pyrazol-5-yl)phenoxy)ethyl) piperidine;

[00132] 1-(2-(4-(3-(4-methoxyphenyl)-1-phenyl-1H-pyrazol-5-yl)phenox y)ethyl)piperidine;

[00133] 4-(1-phenyl-5-(4-(2-(piperidin-1-yl)ethoxy)phenyl)-1H-pyrazo l-3-yl)phenol;

[00134] 1-(2-(4-(1,3-diphenyl-1H-pyrazol-5-yl)phenoxy)ethyl)piperidi ne;

[00135] 1-(2-(4-(1-(4-chlorophenyl)-3-(4-methoxyphenyl)-1H-pyrazol-5 - yl)phenoxy)ethyl)piperidine;

[00136] 4-(1-(4-chlorophenyl)-5-(4-(2-(piperidin-1-yl)ethoxy)phenyl) -1H-pyrazol-3- yl)phenol;

[00137] 1-(2-(4-(1-phenyl-3-(3,4,5-trimethoxyphenyl)-1H-pyrazol-5- yl)phenoxy)ethyl)piperidine;

[00138] 4,4'-(4-ethyl-1-(4-(2-(pyrrolidin-1-yl)ethoxy)phenyl)-1H-pyr azole-3,5-diyl)diphenol; [00139] 4,4'-(5-ethyl-4-(4-(2-(piperidin-1-yl)ethoxy)phenyl)-1H-pyra zole-1,3-diyl)diphenol;

[00140] 4,4'-(4-(4-(2-(piperidin-1-yl)ethoxy)phenyl)-5-propyl-1H-pyr azole-1,3-diyl)diphenol;

[00141] 4,4'-(3-(4-(2-(piperidin-1-yl)ethoxy)phenyl)-1H-pyrazole-1,5 -diyl)diphenol;

[00142] 4-(1-phenyl-3-(4-(2-(piperidin-1-yl)ethoxy)phenyl)-1H-pyrazo l-5-yl)phenol;

[00143] 4-(5-phenyl-3-(4-(2-(piperidin-1-yl)ethoxy)phenyl)-1H-pyrazo l-1-yl)phenol;

[00144] 3-(1-(4-hydroxyphenyl)-3-(4-(2-(piperidin-1-yl)ethoxy)phenyl )-1H-pyrazol-5- yl)phenol;

[00145] 4-(3-(4-(2-(piperidin-1-yl)ethoxy)phenyl)-1-(p-tolyl)-1H-pyr azol-5-yl)phenol;

[00146] 4-(1-(4-chlorophenyl)-3-(4-(2-(piperidin-1-yl)ethoxy)phenyl) -1H-pyrazol-5- yl)phenol;

[00147] 2,6-dimethoxy-4-(1-phenyl-3-(4-(2-(piperidin-1-yl)ethoxy)phe nyl)-1H-pyrazol-5- yl)phenol;

[00148] N-(4-(1-(4-hydroxyphenyl)-3-(4-(2-(piperidin-1-yl)ethoxy)phe nyl)-1H-pyrazol-5- yl)phenyl)acetamide;

[00149] 1-(2-(4-(1,5-bis(4-methoxyphenyl)-1H-pyrazol-3-yl)phenoxy)et hyl)piperidine;

[00150] 1-(2-(4-(5-(4-methoxyphenyl)-1-phenyl-1H-pyrazol-3-yl)phenox y)ethyl)piperidine;

[00151] 1-(2-(4-(1-(3-methoxyphenyl)-5-(4-methoxyphenyl)-1H-pyrazol- 3

[00152] yl)phenoxy)ethyl)piperidine;

[00153] 1-(2-(4-(5-(4-methoxyphenyl)-1-(p-tolyl)-1H-pyrazol-3-yl)phe noxy)ethyl)piperidine;

[00154] 1-(2-(4-(5-(4-methoxyphenyl)-1-(4-(trifluoromethoxy)phenyl)- 1H-pyrazol-3- yl)phenoxy)ethyl)piperidine;

[00155] 1-(2-(4-(1-(4-chlorophenyl)-5-(4-methoxyphenyl)-1H-pyrazol-3 - yl)phenoxy)ethyl)piperidine;

[00156] 1-(2-(4-(5-(4-methoxyphenyl)-1-(4-nitrophenyl)-1H-pyrazol-3- yl)phenoxy)ethyl)piperidine;

[00157] tert-butyl (4-(5-(4-methoxyphenyl)-3-(4-(2-(piperidin-1-yl)ethoxy)pheny l)-1H- pyrazol-1-yl)phenyl)carbamate;

[00158] 4-(5-(4-methoxyphenyl)-3-(4-(2-(piperidin-1-yl)ethoxy)phenyl )-1H-pyrazol-1- yl)aniline;

[00159] 1-(2-(4-(1-(4-methoxyphenyl)-5-phenyl-1H-pyrazol-3-yl)phenox y)ethyl)piperidine; [00160] 1-(2-(4-(5-(2,4-dimethoxyphenyl)-1-(4-methoxyphenyl)-1H-pyra zol-3- yl)phenoxy)ethyl)piperidine;

[00161] N-(4-(1-(4-methoxyphenyl)-3-(4-(2-(piperidin-1-yl)ethoxy)phe nyl)-1H-pyrazol-5- yl)phenyl)acetamide;

[00162] 1-(2-(4-(1-(4-methoxyphenyl)-5-(3,4,5-trimethoxyphenyl)-1H-p yrazol-3- yl)phenoxy)ethyl)piperidine;

[00163] 1-(2-(4-(5-(3,4-dimethoxyphenyl)-1-(4-methoxyphenyl)-1H-pyra zol-3- yl)phenoxy)ethyl)piperidine;

[00164] 1-(2-(4-(5-(3-chloro-4-methoxyphenyl)-1-(4-methoxyphenyl)-1H -pyrazol-3- yl)phenoxy)ethyl)piperidine;

[00165] 1-(2-(4-(5-(4-methoxy-3,5-dimethylphenyl)-1-(4-methoxyphenyl )-1H-pyrazol-3- yl)phenoxy)ethyl)piperidine;

[00166] methyl 4-(1-phenyl-3-(4-(2-(piperidin-1-yl)ethoxy)phenyl)-1H-pyrazo l-5- yl)benzoate;

[00167] 4-(1-phenyl-3-(4-(2-(piperidin-1-yl)ethoxy)phenyl)-1H-pyrazo l-5-yl)benzoic acid;

[00168] 4-(1-phenyl-3-(4-(2-(piperidin-1-yl)ethoxy)phenyl)-1H-pyrazo l-5-yl)benzamide;

[00169] 4-(1-phenyl-3-(4-(2-(piperidin-1-yl)ethoxy)phenyl)-1H-pyrazo l-5-yl)benzonitrile;

[00170] 4-(1-phenyl-3-(4-(2-(piperidin-1-yl)ethoxy)phenyl)-1H-pyrazo l-5-yl)aniline;

[00171] methyl (4-(1-phenyl-3-(4-(2-(piperidin-1-yl)ethoxy)phenyl)-1H-pyraz ol-5- yl)phenyl)carbamate;

[00172] N-(4-(1-phenyl-3-(4-(2-(piperidin-1-yl)ethoxy)phenyl)-1H-pyr azol-5- yl)phenyl)acetamide;

[00173] N-(4-(1-phenyl-3-(4-(2-(piperidin-1-yl)ethoxy)phenyl)-1H-pyr azol-5- yl)phenyl)propionamide;

[00174] 1-(2-(4-(5-(3,4-dimethoxyphenyl)-1-phenyl-1H-pyrazol-3- yl)phenoxy)ethyl)piperidine;

[00175] 1-(2-(4-(1-phenyl-5-(3,4,5-trimethoxyphenyl)-1H-pyrazol-3- yl)phenoxy)ethyl)piperidine;

[00176] 1-(2-(4-(1-phenyl-5-(2,4,5-trimethoxyphenyl)-1H-pyrazol-3- yl)phenoxy)ethyl)piperidine; [00177] 1-(2-(4-(1-phenyl-5-(2,4,6-trimethoxyphenyl)-1H-pyrazol-3- yl)phenoxy)ethyl)piperidine;

[00178] 1-(2-(4-(1-phenyl-5-(2,3,4-trimethoxyphenyl)-1H-pyrazol-3- yl)phenoxy)ethyl)piperidine;

[00179] 1-(2-(4-(5-(4-ethoxy-3,5-dimethoxyphenyl)-1-phenyl-1H-pyrazo l-3- yl)phenoxy)ethyl)piperidine;

[00180] 1-(2-(4-(1-phenyl-5-(3,4,5-triethoxyphenyl)-1H-pyrazol-3- yl)phenoxy)ethyl)piperidine;

[00181] 1-(2-(4-(5-(4-ethoxy-3-methoxyphenyl)-1-phenyl-1H-pyrazol-3- yl)phenoxy)ethyl)piperidine;

[00182] 1-(2-(4-(5-(7-methoxybenzo[d][1,3]dioxol-5-yl)-1-phenyl-1H-p yrazol-3- yl)phenoxy)ethyl)piperidine;

[00183] 1-(2-(4-(5-(4-methoxy-3,5-dimethylphenyl)-1-phenyl-1H-pyrazo l-3- yl)phenoxy)ethyl)piperidine;

[00184] 1-(2-(4-(5-(3-chloro-4-methoxyphenyl)-1-phenyl-1H-pyrazol-3- yl)phenoxy)ethyl)piperidine;

[00185] 1-(2-(4-(5-(6-methoxynaphthalen-2-yl)-1-(4-methoxyphenyl)-1H -pyrazol-3- yl)phenoxy)ethyl)piperidine;

[00186] 1-(2-(4-(1-(4-methoxyphenyl)-5-(thiophen-3-yl)-1H-pyrazol-3- yl)phenoxy)ethyl)piperidine;

[00187] 4,4'-(3-(4-(3-(piperidin-1-yl)propyl)phenyl)-1H-pyrazole-1,5 -diyl)diphenol;

[00188] 1-(3-(4-(1,5-bis(4-methoxyphenyl)-1H-pyrazol-3-yl)phenyl)pro pyl)piperidine;

[00189] 1-(3-(4-(1,5-bis(4-methoxyphenyl)-1H-pyrazol-3-yl)phenoxy)pr opyl)piperidine;

[00190] 3-(4-(2-(1H-pyrazol-1-yl)ethoxy)phenyl)-1,5-bis(4-methoxyphe nyl)-1H-pyrazole;

[00191] 4-(1-methyl-3-(4-(2-(piperidin-1-yl)ethoxy)phenyl)-1H-pyrazo l-5-yl)phenol;

[00192] 1-(2-(4-(5-(4-methoxyphenyl)-1-methyl-1H-pyrazol-3-yl)phenox y)ethyl)piperidine;

[00193] 1-(2-(4-(4,5-bis(4-methoxyphenyl)-1H-imidazol-2-yl)phenoxy)e thyl)piperidine;

[00194] 1-(2-(4-(4,5-bis(4-methoxyphenyl)-1-methyl-1H-imidazol-2- yl)phenoxy)ethyl)piperidine;

[00195] 1-(2-(4-(1-ethyl-4,5-bis(4-methoxyphenyl)-1H-imidazol-2- yl)phenoxy)ethyl)piperidine; [00196] 1-(2-(4-(4,5-di-p-tolyl-1H-imidazol-2-yl)phenoxy)ethyl)piper idine;

[00197] 1-(2-(4-(4,5-bis(4-bromophenyl)-1H-imidazol-2-yl)phenoxy)eth yl)piperidine;

[00198] 1-(2-(4-(4,5-bis(4-fluorophenyl)-1H-imidazol-2-yl)phenoxy)et hyl)piperidine;

[00199] 1-(2-(4-(1-ethyl-4,5-diphenyl-1H-imidazol-2-yl)phenoxy)ethyl )piperidine;

[00200] 4,4'-(2-(4-(2-(piperidin-1-yl)ethoxy)phenyl)-1H-imidazole-4, 5-diyl)diphenol;

[00201] 1-(2-(4-(1,4-bis(4-methoxyphenyl)-1H-imidazol-2-yl)phenoxy)e thyl)piperidine;

[00202] 1-phenyl-2-(4-(2-(piperidin-1-yl)ethoxy)phenyl)-1H-benzo[d]i midazole;

[00203] 1-methyl-2-(4-(2-(piperidin-1-yl)ethoxy)phenyl)-1H-benzo[d]i midazole;

[00204] 5-(4-methoxyphenyl)-3-(4-(2-(piperidin-1-yl)ethoxy)phenyl)is oxazole;

[00205] 5-phenyl-3-(4-(2-(piperidin-1-yl)ethoxy)phenyl)isoxazole;

[00206] 3-phenyl-5-(4-(2-(piperidin-1-yl)ethoxy)phenyl)isoxazole;

[00207] 5-(4-(2-(piperidin-1-yl)ethoxy)phenyl)-3-(p-tolyl)isoxazole;

[00208] 3-(4-fluorophenyl)-5-(4-(2-(piperidin-1-yl)ethoxy)phenyl)iso xazole;

[00209] 3-(naphthalen-2-yl)-5-(4-(2-(piperidin-1-yl)ethoxy)phenyl)is oxazole;

[00210] 3-(4-bromophenyl)-5-(4-(2-(piperidin-1-yl)ethoxy)phenyl)isox azole;

[00211] 5-(4-methoxyphenyl)-3-(1-(2-(piperidin-1-yl)ethyl)-1H-benzo[ d]imidazol-5-yl)-1,2,4- oxadiazole;

[00212] 4-(3-(1-(2-(pyrrolidin-1-yl)ethyl)-1H-benzo[d]imidazol-5-yl) -1,2,4-oxadiazol-5- yl)benzonitrile;

[00213] 4,5-bis(4-methoxyphenyl)-2-(4-(2-(piperidin-1-yl)ethoxy)phen yl)thiazole; and 4,5-bis(4-methoxyphenyl)-2-(4-(2-(piperidin-1-yl)ethoxy)phen yl)oxazole;

[00214] or a pharmaceutically acceptable salt thereof. [00215] In another embodiment, the compounds include isotope-labelled forms. An isotope- labelled form of a compound is identical to the compound apart from the fact that one or more atoms of the compound have been replaced by an atom or atoms having an atomic mass or mass number which differs from the atomic mass or mass number of the atom which usually occurs in greater natural abundance. Examples of isotopes which are readily commercially available and which can be incorporated into a compound by well-known methods include isotopes of hydrogen, carbon, nitrogen, oxygen, fluorine and chlorine, for example 2 H, 3 H, 13 C, 14 C, 15 N, 18 O, 17 O, 18 F and 36 Cl. 3. Methods of Treatment

[00216] Diseases or conditions that may be treated with compounds and/or compositions of the invention include sepsis (e.g., bacterial sepsis), autoimmune disease, lupus erythematosus, ischemia-reperfusion injury, stroke, metabolic disease, obesity-related metabolic inflammation, gout, and cancer. Other diseases or conditions that may be treated with

compounds/compositions of the invention include periodontal disease, mucositis, acne, cardiovascular disease, chronic obstructive pulmonary disease, arthritis, cystic fibrosis, bacterial- induced infections, viral-induced infections, mycoplasma-associated diseases, post herpetic neuralgia, asthma, brain injury, necrotizing enterocolitis, bed sores, leprosy, atopic dermatitis, psoriasis, trauma, neurodegenerative disease, amphotericin B-induced fever and nephritis, coronary artery bypass grafting, and atherosclerosis.

[00217] The invention in one aspect relates to a method for reducing an immune response. As used herein, an immune response refers to a response to an appropriate stimulus by a cell of the immune system, a population of cells of the immune system, or by an immune system. An immune system as used herein refers to an immune system of a subject (e.g., a mammal), specifically including but not limited to an immune system of a human. In some embodiments is a method of reducing an innate immune response in a subject.

[00218] A cell of an immune system can be any cell that is classified as an immune cell. Such cells include B cells, T cells, natural killer (NK) cells, mast cells, basophils, granulocytes, monocytes, macrophages, bone marrow-derived dendritic cells, and other professional antigen- presenting cells, as well as subcategories and precursors thereof. In one embodiment a cell of the immune system can be an isolated cell of the immune system.

[00219] A population of cells of the immune system refers to at least two cells, and more typically at least one thousand cells, of the immune system. In one embodiment a population of cells of the immune system can be an isolated population of cells of the immune system. In one embodiment a population of cells of the immune system is an isolated population of PBMC.

[00220] In one embodiment the method involves contacting a population of immune cells expressing a TLR selected from TLR2, TLR4, TLR7, TLR8, and TLR9, with a compound or composition of the invention. Immune cells expressing TLR 7 can include B cells and dendritic cells, and immune cells expressing TLR8 can include myeloid cells. Immune cells expressing TLR9 can include B cells and pDC. [00221] The method involves measuring a reduced immune response compared to a control immune response. A control immune response is an immune response that occurs in the absence of contacting an immune cell, or a population of immune cells, with a compound or composition of the invention. For purposes of comparing treatment and control immune responses, conditions are generally selected such that the number or concentration of TLR-expressing cells, the amount or concentration of the TLR agonist, temperature, and other such variables are identical or at least comparable between treatment and control measurements, so as to isolate the effect of the composition of the invention. Treatment and control measurements can be made in parallel or they can be made independently. For example, in one embodiment the control is a historical control. In one embodiment the control is a concurrent, parallel control.

[00222] In one embodiment the method relates to a method for reducing an immune response in a subject. As used herein, a subject refers to a mammal. In one embodiment the subject is a human. In another embodiment the subject is a nonhuman primate. In yet another embodiment the subject is a mammal other than a primate, including but not limited to a mouse, rat, hamster, guinea pig, rabbit, cat, dog, goat, sheep, pig, horse, or cow.

[00223] In one embodiment the immune response is an immune response to an antigen.

Antigenic substances include, without limitation, peptides, proteins, carbohydrates, lipids, phospholipids, nucleic acids, autacoids, and hormones. Antigens specifically include allergens, autoantigens (i.e., self-antigens), cancer antigens, and microbial antigens. In respect of peptide antigens and protein antigens, antigens further include both antigens per se and nucleic acids encoding said antigens.

[00224] An allergen is a substance that can induce an allergic or asthmatic response in a susceptible subject. Allergens include pollens, insect venoms, animal dander, dust, fungal spores and drugs (e.g., penicillin).

[00225] Autoantigens include any antigen of host origin, but they specifically include antigens characteristic of an autoimmune disease or condition. Autoantigens characteristic of an autoimmune disease or condition can be associated with, but not necessarily established as causative of, an autoimmune disorder. Specific examples of autoantigens characteristic of an autoimmune disease or condition include but are not limited to insulin, thyroglobulin, glomerular basement membrane, acetylcholine receptor, DNA, and myelin basic protein. [00226] A cancer antigen as used herein is a compound, such as a peptide or protein, associated with a tumor or cancer cell surface and which is capable of provoking an immune response when expressed on the surface of an antigen-presenting cell in the context of a major histocompatibility complex (MHC) molecule. Cancer antigens can be prepared from cancer cells either by preparing crude extracts of cancer cells, for example, as described in Cohen PA et al. (1994) Cancer Res 54:1055-8, by partially purifying the antigens, by recombinant technology, or by de novo synthesis of known antigens. Cancer antigens include but are not limited to antigens that are recombinantly expressed, an immunogenic portion thereof, or a whole tumor or cancer cell. Such antigens can be isolated or prepared recombinantly or by any other means known in the art.

[00227] A microbial antigen can be an antigen that is or is derived from an infectious microbial agent, including a bacterium, a virus, a fungus, or a parasite.

[00228] The invention in one aspect relates to a method for treating an autoimmune condition in a subject. As used herein, an autoimmune condition refers to an autoimmune disease or disorder, i.e., an immunologically mediated acute or chronic process, directed by immune cells of a host subject against a tissue or organ of the host subject, resulting in injury to the tissue or organ. The term encompasses both cellular and antibody-mediated autoimmune phenomena, as well as organ-specific and organ-nonspecific autoimmunity.

[00229] Autoimmune conditions specifically include insulin-dependent diabetes mellitus, rheumatoid arthritis, systemic lupus erythematosus (SLE), multiple sclerosis, atherosclerosis, and inflammatory bowel disease. Inflammatory bowel disease includes Crohn's disease and ulcerative colitis. Autoimmune diseases also include, without limitation, ankylosing spondylitis, autoimmune chronic active hepatitis, autoimmune encephalomyelitis, autoimmune hemolytic anemia, autoimmune thrombocytopenic purpura, autoimmune-associated infertility, Behçet' s syndrome, bullous pemphigoid, Churg-Strauss disease, glomerulonephritis, Goodpasture's syndrome, Graves' disease, Guillain-Barré syndrome, Hashimoto's thyroiditis, idiopathic Addison's disease, idiopathic thrombocytopenia, insulin resistance, mixed connective tissue disease, myasthenia gravis, pemphigus, pernicious anemia, polyarteritis nodosa,

polymyositis/dermatomyositis, primary biliary sclerosis, psoriasis, Reiter's syndrome, sarcoidosis, sclerosing cholangitis, Sjögren's syndrome, systemic sclerosis (scleroderma and CREST syndrome), Takayasu's arteritis, temporal arteritis, and Wegener's granulomatosis. [00230] The method of treatment of an autoimmune condition in a subject specifically includes treatment of a human subject. In one embodiment the autoimmune condition is systemic lupus erythematosus. In one embodiment the autoimmune condition is rheumatoid arthritis. The method of treatment of an autoimmune condition in a subject optionally can further include administration of another treatment agent or treatment modality useful in the treatment of the autoimmune condition. For example, the method can include administration of a compound or composition of the invention, either alone or in combination with an agent such as a

corticosteroid (e.g., prednisone), a cytokine (e.g., IFN-Į), or other suitable immunomodulatory agent. In this context, "in combination with" can refer to simultaneous administration at a single site of administration, or at different sites of administration. Alternatively and in addition, "in combination with" can refer to sequential administration at a single site of administration, or at different sites of administration.

[00231] As will be evident from the foregoing, autoimmune diseases also include certain immune complex-associated diseases. The term "immune complex-associated disease" as used herein refers to any disease characterized by the production and/or tissue deposition of immune complexes, including, but not limited to systemic lupus erythematosus (SLE) and related connective tissue diseases, rheumatoid arthritis, hepatitis C- and hepatitis B-related immune complex disease (e.g., cryoglobulinemia), Behçet's syndrome, autoimmune

glomerulonephritides, and vasculopathy associated with the presence of LDL/anti-LDL immune complexes.

[00232] Non-limiting examples of neurodegenerative disorders include adrenal

leukodystrophy, aging-related disorders and dementias, alcoholism, Alexander's disease, Alper's disease, Alzheimer's disease, amyotrophic lateral sclerosis (Lou Gehrig's Disease), ataxia telangiectasia, Batten disease (also known as Spielmeyer-Vogt-Sjögren-Batten disease), bovine spongiform encephalopathy (BSE), canavan disease, cerebral palsy, Cockayne syndrome, corticobasal degeneration (CBD), Creutzfeldt-Jakob disease, familial fatal insomnia, frontotemporal lobar degeneration, frontal temporal dementias (FTDs), Huntington's disease, HIV-associated dementia, Kennedy's disease, Krabbe's disease, Lewy body disease,

neuroborreliosis, Machado-Joseph disease (spinocerebellar ataxia type 3), multiple system atrophy, multiple sclerosis, narcolepsy, Niemann Pick disease, Parkinson disease, Pelizaeus- Merzbacher disease, Pick's disease, primary lateral sclerosis, progressive supranuclear palsy (PSP), psychotic disorders, Refsum's disease, Sandhoff disease, Schilder's disease, schizoaffective disorder, schizophrenia, stroke, subacute combined degeneration of spinal cord secondary to pernicious anemia, spinocerebellar ataxia, spinal muscular atrophy, Steele- Richardson-Olszewski disease, Tabes dorsalis, and toxic encephalopathy.

[00233] The inflammatory disorder may be arthritis including, but not limited to, rheumatoid arthritis, spondyloarthropathies, gouty arthritis, osteoarthritis, systemic lupus erythematosus, or juvenile arthritis. In some embodiments, the inflammation may be associated with asthma, allergic rhinitis, sinus diseases, bronchitis, tuberculosis, acute pancreatitis, sepsis, infectious diseases, menstrual cramps, premature labor, tendinitis, bursitis, skin-related conditions such as psoriasis, eczema, atopic dermatitis, urticaria, dermatitis, contact dermatitis, and burns, or from post-operative inflammation including from ophthalmic surgery such as cataract surgery and refractive surgery. In a further embodiment, the inflammatory disorder may be a gastrointestinal condition such as inflammatory bowel disease, Crohn's disease, gastritis, irritable bowel syndrome, chronic cholecystitis, or ulcerative colitis. In yet another embodiment, the

inflammation may be associated with diseases such as vascular diseases, migraine headaches, periarteritis nodosa, thyroiditis, aplastic anemia, Hodgkin's disease, sclerodoma, rheumatic fever, type I diabetes, neuromuscular junction disease including myasthenia gravis, white matter disease including multiple sclerosis, sarcoidosis, nephrotic syndrome, Behçet's syndrome, polymyositis, gingivitis, nephritis, hypersensitivity, swelling occurring after injury, myocardial ischemic, allergic rhinitis, respiratory distress syndrome, systemic inflammatory response syndrome (SIRS), cancer-associated inflammation, reduction of tumor-associated angiogenesis, endotoxin shock syndrome, atherosclerosis, and the like. In an alternate embodiment, the inflammatory disorder may be associated with an ophthalmic disease, such as retinitis, retinopathies, uveitis, ocular photophobia, or of acute injury to the eye tissue. In still another embodiment, the inflammation may be a pulmonary inflammation, such as that associated with viral infections or cystic fibrosis, chronic obstructive pulmonary disease, or acute respiratory distress syndrome. The inflammatory disorder may also be associated with tissue rejection, graft v. host diseases, delayed-type hypersensitivity, as well as immune-mediated and inflammatory elements of CNS diseases such as Alzheimer's, Parkinson's, multiple sclerosis and the like. 4. [00234] In another aspect of the invention, pharmaceutically acceptable compositions are provided, wherein these compositions comprise any of the compounds as described herein, and optionally comprise a pharmaceutically acceptable carrier, adjuvant or vehicle. In certain embodiments, these compositions optionally further comprise one or more additional therapeutic agents. In one embodiment, the pharmaceutical composition comprises a therapeutically effective amount of a compound of the present invention or a pharmaceutically acceptable salt thereof and one or more pharmaceutically acceptable carriers or vehicles.

[00235] Pharmaceutical compositions of the present invention may be manufactured by processes well known in the art, e.g., by means of conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping or lyophilizing processes.

[00236] As used herein, the term "pharmaceutically acceptable salt" refers to those salts which are, within the scope of sound medical judgement, suitable for use in contact with the tissues of humans and lower animals without undue toxicity, irritation, allergic response and the like, and are commensurate with a reasonable benefit/risk ratio. Pharmaceutically acceptable salts are well known in the art. For example, S. M. Berge, et al. describe pharmaceutically acceptable salts in detail in J Pharmaceutical Sciences, 1977, 66, 1-19, incorporated herein by reference.

Pharmaceutically acceptable salts of the compounds of this invention include those derived from suitable inorganic and organic acids and bases. Examples of pharmaceutically acceptable, nontoxic acid addition salts are salts of an amino group formed with inorganic acids such as hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid and perchloric acid or with organic acids such as acetic acid, oxalic acid, maleic acid, tartaric acid, citric acid, succinic acid or malonic acid or by using other methods used in the art such as ion exchange. Other pharmaceutically acceptable salts include adipate, alginate, ascorbate, aspartate,

benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphorate, camphorsulfonate, citrate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, formate, fumarate, glucoheptonate, glycerophosphate, gluconate, hemisulfate, heptanoate, hexanoate, hydroiodide, 2-hydroxy-ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, malate, maleate, malonate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oleate, oxalate, palmitate, pamoate, pectinate, persulfate, 3-phenylpropionate, phosphate, picrate, pivalate, propionate, stearate, succinate, sulfate, tartrate, thiocyanate, p-toluenesulfonate, undecanoate, valerate salts, and the like. Salts derived from appropriate bases include alkali metal, alkaline earth metal, ammonium and N(C 1-4 alkyl) 4 salts. This invention also envisions the quaternization of any basic nitrogen-containing groups of the compounds disclosed herein. Water or oil-soluble or dispersable products may be obtained by such quaternization. Representative alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, and the like. Further pharmaceutically acceptable salts include, when appropriate, nontoxic ammonium, quaternary ammonium, and amine cations formed using counterions such as halide, hydroxide, carboxylate, sulfate, phosphate, nitrate, lower alkyl sulfonate and aryl (e.g., phenyl/substituted phenyl) sulfonate.

[00237] As described herein, the pharmaceutically acceptable compositions of the invention additionally comprise a pharmaceutically acceptable carrier, adjuvant, or vehicle, which, as used herein, includes any and all solvents, diluents, or other liquid vehicle, dispersion or suspension aids, surface active agents, isotonic agents, thickening or emulsifying agents, preservatives, solid binders, lubricants and the like, as suited to the particular dosage form desired. Remington's Pharmaceutical Sciences, Sixteenth Edition, E.W. Martin (Mack Publishing Co., Easton, Pa., 1980) discloses various carriers used in formulating pharmaceutically acceptable compositions and known techniques for the preparation thereof. Except insofar as any conventional carrier medium is incompatible with the compounds of the invention, such as by producing any undesirable biological effect or otherwise interacting in a deleterious manner with any other component(s) of the pharmaceutically acceptable composition, its use is contemplated to be within the scope of this invention. Some examples of materials which can serve as

pharmaceutically acceptable carriers include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, serum proteins, such as human serum albumin, buffer substances such as phosphates, glycine, sorbic acid, or potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, polyacrylates, waxes,

polyethylenepolyoxypropylene-block polymers, wool fat, sugars such as lactose, glucose and sucrose; starches such as com starch and potato starch; cellulose and its derivatives such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; powdered tragacanth; malt; gelatin; talc; excipients such as cocoa butter and suppository waxes; oils such as peanut oil, cottonseed oil; safflower oil; sesame oil; olive oil; com oil and soybean oil; glycols; such a propylene glycol or polyethylene glycol; esters such as ethyl oleate and ethyl laurate; agar; buffering agents such as magnesium hydroxide and aluminum hydroxide; alginic acid; pyrogen- free water; isotonic saline; Ringer's solution; ethyl alcohol, and phosphate buffer solutions, as well as other non-toxic compatible lubricants such as sodium lauryl sulfate and magnesium stearate, as well as coloring agents, releasing agents, coating agents, sweetening, flavoring and perfuming agents, preservatives and antioxidants can also be present in the composition, according to the judgment of the formulator.

[00238] The pharmaceutically acceptable compositions of this invention can be administered to humans and other animals orally, rectally, parenterally, intracistemally, intravaginally, intraperitoneally, topically (as by powders, ointments, or drops), bucally, as an oral or nasal spray, or the like, depending on the severity of the disease being treated.

[00239] Pharmaceutical compositions for parenteral injection comprise pharmaceutically acceptable sterile aqueous or nonaqueous solutions, dispersions, suspensions or emulsions as well as sterile powders for reconstitution into sterile injectable solutions or dispersions just prior to use. Examples of suitable aqueous and nonaqueous carriers, diluents, solvents or vehicles include water, ethanol, polyols (such as glycerol, propylene glycol, polyethylene glycol and the like), vegetable oils (such as olive oil), injectable organic esters (such as ethyl oleate) and suitable mixtures thereof. Proper fluidity can be maintained, for example, by the use of coating materials such as lecithin, by the maintenance of the required particle size in the case of dispersions and by the use of surfactants.

[00240] These compositions can also contain adjuvants such as preservatives, wetting agents, emulsifying agents and dispersing agents. Prevention of the action of microorganisms can be ensured by the inclusion of various antibacterial and antifungal agents, for example, paraben, chlorobutanol, phenol sorbic acid and the like. It can also be desirable to include isotonic agents such as sugars, sodium chloride and the like. Prolonged absorption of the injectable

pharmaceutical form can be brought about by the inclusion of agents which delay absorption such as aluminum monostearate and gelatin.

[00241] In some cases, in order to prolong the effect of the drug, it is desirable to slow the absorption of the drug from subcutaneous or intramuscular injection. This can be accomplished by the use of a liquid suspension of crystalline or amorphous material with poor water solubility. The rate of absorption of the drug then depends upon its rate of dissolution which, in turn, can depend upon crystal size and crystalline form. Alternatively, delayed absorption of a parenterally administered drug form is accomplished by dissolving or suspending the drug in an oil vehicle.

[00242] Liquid dosage forms for oral administration include, but are not limited to, pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs. In addition to the active compounds, the liquid dosage forms may contain inert diluents commonly used in the art such as, for example, water or other solvents, solubilizing agents and emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, dimethylformamide, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor, and sesame oils), glycerol, tetrahydrofurfuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof. Besides inert diluents, the oral compositions can also include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents.

[00243] Solid dosage forms for oral administration include capsules, tablets, pills, powders, cement, putty, and granules. In such solid dosage forms, the active compound can be mixed with at least one inert, pharmaceutically acceptable excipient or carrier, such as sodium citrate or dicalcium phosphate and/or a) fillers or extenders such as starches, lactose, sucrose, glucose, mannitol and silicic acid; b) binders such as carboxymethylcellulose, alginates, gelatin, polyvinylpyrrolidone, sucrose and acacia; c) humectants such as glycerol; d) disintegrating agents such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates and sodium carbonate; e) solution retarding agents such as paraffin; f) absorption accelerators such as quaternary ammonium compounds; g) wetting agents such as cetyl alcohol and glycerol monostearate; h) absorbents such as kaolin and bentonite clay and i) lubricants such as talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate and mixtures thereof. In the case of capsules, tablets and pills, the dosage form can also comprise buffering agents.

[00244] Solid compositions of a similar type may also be employed as fillers in soft and hardfilled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polyethylene glycols and the like. The solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings and other coatings well known in the pharmaceutical formulating art. They may optionally contain opacifying agents and can also be of a composition that they release the active ingredient(s) only, or preferentially, in a certain part of the intestinal tract, optionally, in a delayed manner. Examples of embedding compositions that can be used include polymeric substances and waxes. Solid compositions of a similar type may also be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polyethylene glycols and the like.

[00245] The active compounds can also be in microencapsulated form with one or more excipients as noted above. The solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings, release controlling coatings and other coatings well known in the pharmaceutical formulating art. In such solid dosage forms the active compound may be admixed with at least one inert diluent such as sucrose, lactose or starch. Such dosage forms may also comprise, as is normal practice, additional substances other than inert diluents, e.g., tableting lubricants and other tableting aids such a magnesium stearate and microcrystalline cellulose. In the case of capsules, tablets and pills, the dosage forms may also comprise buffering agents. They may optionally contain opacifying agents and can also be of a composition that they release the active ingredient(s) only, or preferentially, in a certain part of the intestinal tract, optionally, in a delayed manner.

Examples of embedding compositions that can be used include polymeric substances and waxes.

[00246] Compositions for rectal or vaginal administration are preferably suppositories which can be prepared by mixing the compounds of this invention with suitable non-irritating excipients or carriers such as cocoa butter, polyethylene glycol or a suppository wax which are solid at ambient temperature but liquid at body temperature and therefore melt in the rectum or vaginal cavity and release the active compound.

[00247] Dosage forms for topical or trans dermal administration of a compound of this invention include ointments, pastes, creams, lotions, gels, powders, solutions, sprays, inhalants or patches. The active component is admixed under sterile conditions with a pharmaceutically acceptable carrier and any needed preservatives or buffers as may be required. Ophthalmic formulation, eardrops, and eye drops are also contemplated as being within the scope of this invention. Additionally, the invention contemplates the use of transdermal patches, which have the added advantage of providing controlled delivery of a compound to the body. Such dosage forms are prepared by dissolving or dispensing the compound in the proper medium. Absorption enhancers can also be used to increase the flux of the compound across the skin. The rate can be controlled by either providing a rate controlling membrane or by dispersing the compound in a polymer matrix or gel.

[00248] Compounds described herein can be administered as a pharmaceutical composition comprising the compounds of interest in combination with one or more pharmaceutically acceptable carriers. The phrase "therapeutically effective amount" of the present compounds means sufficient amounts of the compounds to treat disorders, at a reasonable benefit/risk ratio applicable to any medical treatment. It is understood, however, that the total daily dosage of the compounds and compositions can be decided by the attending physician within the scope of sound medical judgment. The specific therapeutically effective dose level for any particular patient can depend upon a variety of factors including the disorder being treated and the severity of the disorder; activity of the specific compound employed; the specific composition employed; the age, body weight, general health and prior medical history, sex and diet of the patient; the time of administration, route of administration, and rate of excretion of the specific compound employed; the duration of the treatment; drugs used in combination or coincidental with the specific compound employed; and like factors well-known in the medical arts. For example, it is well within the skill of the art to start doses of the compound at levels lower than required to achieve the desired therapeutic effect and to gradually increase the dosage until the desired effect is achieved. Actual dosage levels of active ingredients in the pharmaceutical compositions can be varied so as to obtain an amount of the active compound(s) that is effective to achieve the desired therapeutic response for a particular patient and a particular mode of administration. In the treatment of certain medical conditions, repeated or chronic administration of compounds can be required to achieve the desired therapeutic response. "Repeated or chronic administration" refers to the administration of compounds daily (i.e., every day) or intermittently (i.e., not every day) over a period of days, weeks, months, or longer.

[00249] Combination therapy includes administration of a single pharmaceutical dosage formulation containing one or more of the compounds described herein and one or more additional pharmaceutical agents, as well as administration of the compounds and each additional pharmaceutical agent, in its own separate pharmaceutical dosage formulation. For example, a compound described herein and one or more additional pharmaceutical agents, can be administered to the patient together, in a single oral dosage composition having a fixed ratio of each active ingredient, such as a tablet or capsule; or each agent can be administered in separate oral dosage formulations. Where separate dosage formulations are used, the present compounds and one or more additional pharmaceutical agents can be administered at essentially the same time (e.g., concurrently) or at separately staggered times (e.g., sequentially).

[00250] For adults, the doses are generally from about 0.01 to about 100 mg/kg, desirably about 0.1 to about 1 mg/kg body weight per day by inhalation, from about 0.01 to about 100 mg/kg, desirably 0.1 to 70 mg/kg, more desirably 0.5 to 10 mg/kg body weight per day by oral administration, and from about 0.01 to about 50 mg/kg, desirably 0.1 to 1 mg/kg body weight per day by intravenous administration.

[00251] The disclosed compounds may be included in kits comprising the compound, or a pharmaceutically acceptable salt, a pharmaceutical composition, or both; and information, instructions, or both that use of the kit will provide treatment for medical conditions in mammals (particularly humans). The information and instructions may be in the form of words, pictures, or both, and the like. In addition or in the alternative, the kit may include the medicament, a composition, or both; and information, instructions, or both, regarding methods of application of medicament, or of composition, preferably with the benefit of treating or preventing medical conditions in mammals (e.g., humans). 5.

[00252] Materials and Methods. All reagents were used as purchased. Dichloromethane and dimethylformamide were dried using a solvent dispensing system (SDS, neutral alumina columns). Glassware was oven-dried, assembled while hot, and cooled under an inert atmosphere. Solvents used for extraction and flash chromatography were reagent or ultima grade, purchased from either Aldrich or Fisher Scientific. Flash column chromatography was performed on Silica P Flash Silica Gel (40-64 mm, 60 Å) from SiliCycle ® . Reaction progress was monitored using analytical thin-layer chromatography (TLC) F-254 silica gel aluminum plates.

Visualization was achieved by either UV light (254 nm) or iodine vapor. 1 H NMR and 13 C NMR spectra were obtained on a 400 or 500 MHz Varian ® FT-NMR spectrometers and multiplicities are reported as follows: singlet (s), doublet (d), doublet of doublet, triplet (t), and multiplet (m).The chemical shifts are reported in ppm and are referenced to either tetramethylsilane or the solvent. High resolution mass spectra were obtained using electrospray ionization on either a Micromass Q-Tof Ultima or Waters Quattro instrument. [00253] Certain compounds of the invention may be prepared using methods that have been described previously in: Huang, et al. Org. Lett.2000, 2 (18), 2833-2836; Stauffer et al., J. Med. Chem.2000, 43(26), 4934-4947; Stauffer et al., Bioorg. Med. Chem.2001, 9(1), 141-150;

Stauffer et al., Bioorg. Med. Chem.2001, 9(1), 151-161; and Sun et al., Endocrinology 2002, 143, 941-947., which are incorporated herein by reference.

[00254] ABBREVIATIONS. The following abbreviations are employed in the Examples and elsewhere herein:

BMIM-PF 6 = 1-butyl-3-methylimidazolium hexafluorophosphate

DCM = dichloromethane

DMAP = N,N-dimethylaminopyridine

DMF = dimethylformamide

MeOH = methanol

TBAB = tetrabutyl ammonium bromide

min = minute(s)

h or hr = hour(s)

r.t. = room temperature

HRMS-ESI = high resolution mass spectrometry-electrospray ionization

NMR = nuclear magnetic resonance

(M+H) = the protonated mass of the free base of the compound

[00255] Compounds of formula (I) where G 2 has formula (i) may be prepared as generally illustrated in Scheme 1.

Scheme 1. General scheme and procedure for the synthesis of substituted pyrazoles

[00256] STEP 1: To a solution of p-hydroxyacetophenone (7.34 mmol) and the appropriate benzaldehyde (8.81 mmol) in methanol (70 mL), KOH (29.38 mmol) and catalytic

tetrabutylammonium bromide (TBAB) is added. The reaction mixture is refluxed overnight. After the completion of reaction, the mixture is cooled and poured into iced cold water to which dilute HCl is added. The precipitate obtained is then filtered and washed with excess of water, methanol, dried in air and finally recrystallized from methanol to obtain pure chalcones in 65-90 % yield.

[00257] STEP 2: To the solution of the respective chalcone (2.36 mmol) in

dimethylformamide (12 mL), K 2 CO 3 (3.54 mmol) and Cs 2 CO 3 (2.36 mmol) are added, followed by 1-(2-chloroethyl)piperidine-hydrochloride (2.60 mmol). The reaction mixture is allowed to stir for overnight at room temperature. After completion of the reaction, extraction is done using ethyl acetate. The organic layer is washed with brine, dried over sodium sulfate, concentrated under vacuum, and partially purified by flash column chromatography using

methanol/dichloromethane to remove the nonpolar impurities or any traces of unreacted chalcone. The synthesized chalcones with basic side chains (85-95 % yield) are then directly taken to the next step.

[00258] STEP 3 & 4: A mixture of substituted chalcone (0.55 mmol) and appropriate phenylhydrazine hydrochloride (0.82 mmol) in 5 mL of dimethylformamide are heated under a N 2 atmosphere at 85 °C for 4-10 hours. The reaction solution is concentrated under vacuum and partitioned between ethyl acetate and water twice. The organic layers are combined, washed with brine, dried (Na2SO4) and concentrated under vacuum. The residue is then subjected to oxidation using MnO2 (7.15 mmol) in benzene (15 mL) under reflux for 5-10 hours. After completion, the reaction the mixture is allowed to cool to room temperature and then passed through a pad of Celite to remove particulates. The clear solution is concentrated under vacuum and purified by flash chromatography using dichloromethane/methanol to obtain the pure pyrazoles in 75-90 % yield. [00259] The following compounds may be prepared by the foregoing procedures. [00260] 1-(2-(4-(5-(4-methoxyphenyl)-1-(4-nitrophenyl)-1H-pyrazol-3- yl)phenoxy)ethyl)piperidine (TSI-13-48)

[00261] 1 H NMR (500 MHz, Chloroform-d) į 8.20 (d, J = 9.0 Hz, 2H), 7.84 (d, J = 8.6 Hz, 2H), 7.56 (d, J = 9.0 Hz, 2H), 7.23 (d, J = 8.4 Hz, 2H), 6.99 (d, J = 8.4 Hz, 2H), 6.93 (d, J = 8.5 Hz, 2H), 6.74 (s, 1H), 4.30 (t, J = 5.5 Hz, 2H), 3.86 (s, 3H), 3.02– 2.97 (m, 2H), 2.75 (s, 4H), 1.80– 1.73 (m, 4H), 1.54 (s, 2H). 13 C NMR (126 MHz, CDCl 3 ) į 160.4, 153.2, 145.8, 145.3, 145.0, 130.4, 127.5, 125.7, 124.6, 124.5, 122.6, 115.1, 114.6, 106.7, 65.3, 57.6, 55.6, 54.9, 25.2, 23.8. HRMS-ESI: m/z [M+H] + for C29H31N4O4, calculated 499.2345; observed 499.2345.

[00263] 1 H NMR (499 MHz, Chloroform-d) į 7.83 (d, J = 8.8 Hz, 2H), 7.31 (d, J = 8.9 Hz, 2H), 6.97 (d, J = 8.8 Hz, 2H), 6.90 (d, J = 9.0 Hz, 2H), 6.73 (s, 1H), 6.47 (s, 2H), 4.17 (t, J = 6.1 Hz, 2H), 3.86 (s, 3H), 3.82 (s, 3H), 3.69 (s, 6H), 2.83 (d, J = 6.0 Hz, 2H), 2.56 (s, 4H), 1.63 (t, J = 8.4 Hz, 4H), 1.47 (s, 2H). 13 C NMR (126 MHz, CDCl3) į 159.1, 159.0, 153.3, 151.6, 144.4, 138.3, 133.7, 127.2, 127.2, 126.2, 126.1, 115.0, 114.3, 106.2, 103.9, 66.2, 61.2, 58.2, 56.2, 55.8, 55.3, 26.1, 24.4. HRMS-ESI: m/z [M+H] + for C 32 H 38 N 3 O 5 , calculated 544.2811; observed 544.2797. [00264] 1-(2-(4-(5-(7-methoxybenzo[d][1,3]dioxol-5-yl)-1-phenyl-1H-p yrazol-3-

[00265] 1 H NMR (500 MHz, Chloroform-d) į 7.84 (d, J = 8.8 Hz, 2H), 7.42– 7.36 (m, 4H), 7.34– 7.30 (m, 1H), 7.28 (s, 1H), 6.98 (d, J = 8.8 Hz, 2H), 6.71 (s, 1H), 6.48 (d, J = 1.5 Hz, 1H), 6.41 (d, J = 1.4 Hz, 1H), 6.00 (s, 2H), 4.20 (t, J = 5.9 Hz, 2H), 3.72 (s, 3H), 2.88 (t, J = 5.9 Hz, 2H), 2.61 (s, 4H), 1.68 (p, J = 5.5 Hz, 4H), 1.49 (s, 2H). 13 C NMR (126 MHz, CDCl 3 ) į 158.9, 151.9, 149.0, 144.2, 143.6, 140.4, 135.6, 130.8, 129.2, 127.7, 126.1, 125.0, 114.9, 114.5, 108.9, 104.7, 103.3, 102.0, 65.9, 58.1, 56.6, 55.2, 25.9, 24.2. HRMS-ESI: m/z [M+H] + for C 30 H 32 N 3 O 4 , calculated 498.2393; observed 498.2389. Dealkylation of aromatic alkoxy groups may be accomplished as generally shown in Scheme 2. Scheme 2. General scheme and procedure for the synthesis of different demethylated

[00267] To a mixture of dodecanethiol (0.55 mmol) and dry dichloromethane (3 mL) is added aluminum chloride (1.1 mmol) at 0 °C. In another flask, the methylated pyrazole (0.22 mmol) is dissolved in dichloromethane, cooled to 0 °C and then slowly added with stirring in the above mixture. The resulting solution is warmed to room temperature, and stirred for 4-5 hours. After the completion of the reaction, the mixture is poured into methanol and concentrated under vacuum. Crude product is extracted from the residue using ethyl acetate, and the organic layer is again concentrated under vacuum and then purified by flash chromatography using

dichloromethane/methanol to get pure product in 80-90% yield. [00268] The following compounds may be prepared by the foregoing procedure. [00269] 4-(5-phenyl-3-(4-(2-(piperidin-1-yl)ethoxy)phenyl)-1H-pyrazo l-1-yl)phenol (TSI- 13-50)

[00270] 1 H NMR (500 MHz, Methanol-d4) į 7.77 (d, J = 8.6 Hz, 2H), 7.29– 7.20 (m, 5H), 7.14 (d, J = 8.7 Hz, 2H), 6.97 (d, J = 8.7 Hz, 2H), 6.79 (d, J = 8.7 Hz, 2H), 6.75 (s, 1H), 4.19 (t, J = 5.5 Hz, 2H), 2.92 (t, J = 5.4 Hz, 2H), 2.68 (s, 4H), 1.67 (dt, J = 10.7, 5.5 Hz, 4H), 1.50 (s, 2H). 13 C NMR (126 MHz, CD 3 OD) į 158.7, 157.2, 151.7, 145.1, 132.1, 130.4, 128.7, 128.5, 128.4, 127.4, 127.3, 126.1, 115.7, 114.8, 104.1, 64.9, 57.6, 54.8, 25.0, 23.6. HRMS-ESI: m/z [M+H] + for C 28 H 30 N 3 O 2 , calculated 440.2338; observed 440.2339.

[00272] 1 H NMR (500 MHz, Methanol-d4) į 7.83 (d, J = 6.3 Hz, 2H), 7.44– 7.27 (m, 5H), 7.07 (d, J = 6.5 Hz, 4H), 6.78 (s, 1H), 6.71 (d, J = 6.8 Hz, 2H), 4.41 (s, 2H), 3.57 (s, 4H), 3.09 (s, 2H), 1.90 (s, 5H), 1.54 (s, 1H). 13 C NMR (126 MHz, CD 3 OD) į 158.0, 151.8, 145.5, 140.3, 130.1, 128.9, 127.8, 127.3, 127.0, 125.8, 121.4, 115.2, 114.9, 104.0, 62.3, 56.1, 53.8, 22.9, 21.4. HRMS-ESI: m/z [M+H] + for C 28 H 30 N 3 O 2 , calculated 440.2338; observed 440.2342 [00273] 4'-(3-(4-(2-(piperidin-1-yl)ethoxy)phenyl)-1H-pyrazole-1,5-d iyl)diphenol (TSI-

[00274] 1 H NMR (500 MHz, Methanol-d 4 ) į 7.77 (d, J = 8.4 Hz, 2H), 7.13 (d, J = 8.6 Hz, 2H), 7.09 (d, J = 8.4 Hz, 2H), 6.99 (d, J = 8.4 Hz, 2H), 6.80 (d, J = 8.6 Hz, 2H), 6.73 (s, 1H), 6.71 (d, J = 8.3 Hz, 2H), 4.19 (t, J = 5.4 Hz, 2H), 2.91 (t, J = 5.3 Hz, 2H), 2.69 (s, 4H), 1.67 (p, J = 5.5 Hz, 4H), 1.52 (s, 2H). 13 C NMR (126 MHz, CD3OD) į 158.0, 157.8, 157.5, 151.3, 145.5, 132.3, 130.0, 127.5, 127.2, 127.0, 121.5, 115.5, 115.2, 114.8, 103.2, 62.5, 56.2, 53.9, 23.1, 21.6. HRMS-ESI: m/z [M+H] + for C28H30N3O3, calculated 456.2287; observed 456.2305. [00275] 3-(1-(4-hydroxyphenyl)-3-(4-(2-(piperidin-1-yl)ethoxy)phenyl )-1H-pyrazol-5- yl)phenol (TSI-13-58) [00276] 1 H NMR (500 MHz, Methanol-d4) į 7.85 (s, 2H), 7.28– 7.20 (m, 2H), 7.13 (s, 3H), 6.83 (s, 2H), 6.80– 6.69 (m, 4H), 4.56– 4.37 (m, 2H), 3.62 (s, 4H), 3.10 (s, 2H), 2.04– 1.79 (m, 6H). 13 C NMR (126 MHz, CD3OD) į 158.8, 158.6, 157.6, 146.7, 130.1, 129.9, 129.8, 128.0, 128.0, 124.4, 120.1, 116.2, 115.8, 115.6, 115.3, 63.3, 57.0, 54.7, 47.9, 47.7, 23.3, 21.8. HRMS- ESI: m/z [M+H] + for C 28 H 30 N 3 O 3 , calculated 456.2287; observed 456.2282. Compounds wherein G 1 is a pyrazole may be prepared as shown in Scheme 3. Scheme 3. Procedure for the synthesis of TSI-15-16.

[00278] The procedure for the synthesis of pyrazole (A, 60% yield) is similar to that described in Scheme 1, except 1,2-dibromoethane is used for the initial phenol alkylation. [00279] For the synthesis of final compound (TSI-15-16, 65% yield) from A, the same procedure is followed as shown in step 1 (Scheme 3), and instead of 1,2-dibromoethane, pyrazole is used as reactant.

[00281] 1 H NMR (499 MHz, Chloroform-d) į 7.84 (d, J = 8.7 Hz, 2H), 7.58 (t, J = 2.1 Hz, 2H), 7.30 (d, J = 8.9 Hz, 2H), 7.21 (d, J = 8.6 Hz, 2H), 6.94 (d, J = 8.7 Hz, 2H), 6.89 (d, J = 8.9 Hz, 2H), 6.86 (d, J = 8.7 Hz, 2H), 6.70 (s, 1H), 6.29 (t, J = 2.0 Hz, 1H), 4.57 (t, J = 5.2 Hz, 2H), 4.39 (t, J = 5.2 Hz, 2H), 3.83 (d, J = 4.6 Hz, 6H). 13 C NMR (126 MHz, CDCl 3 ) į 159.7, 159.0, 158.3, 151.4, 144.4, 139.9, 133.7, 130.5, 130.2, 127.3, 127.0, 126.9, 123.2, 114.9, 114.3, 114.1, 105.9, 103.9, 67.0, 55.7, 55.5, 51.7. HRMS-ESI: m/z [M+H] + for C28H27N4O3 calculated 467.2083; observed 467.2080. [00282] Compounds of formula (I) where G 2 is (x) or (xiii) may be prepared as generally shown in Scheme 4. Scheme 4: General Chemical Synthesis Scheme of Target Isoxazoles

[00283] General Procedure for Chalcone Synthesis (1a-d): In an oven dried round bottom flask under nitrogen, substituted acetophenone (~10 mmol) is dissolved into 25 mL methanol. Next, a catalytic amount (20-30mg) of tetra-n-butylammonium bromide is added to the solution. Then, anisaldehyde (10 mmol) and potassium hydroxide (40 mmol) are added sequentially. A condenser is added onto the round bottom flask and the reaction is refluxed overnight. Following the completion of the reaction, it is then cooled to room temperature added onto 30 mL of ice- cold deionized water. The cool solution is acidified to a pH~2 with 12N HCl. If a precipitate forms, it is collected by vacuum filtration, and for those that do not have a lot of precipitate form, the organic product is extracted with 3x20 mL of diethyl ether, washed with 2x20 mL H 2 O, 1x20 mL brine, dried with magnesium sulfate, and concentrated.

[00284] 3-(4-fluorophenyl)-5-(4-(2-(piperidin-1-yl)ethoxy)phenyl)iso xazole (1c). In an oven dried round bottom flask, 4’-fluoroacetophenone was dissolved into 25 mL of a 5% NaOH solution. Then, one equivalent of anisaldehyde was added to the solution. The round bottom flask was then placed inside of a beaker to keep it upright, and it was covered by placing a small funnel into the neck of the flask. The mixture was placed into a domestic microwave oven and was reacted via microwave irradiation. A beaker of ice was placed near the beaker with the round bottom flask. The reaction mixture was irradiated for 10 second intervals for about 5-7 minutes with cooling of the mixture in between. The progress of the reaction was monitored by thin-layer chromatography. Following the completion of the reaction, it was then cooled to room temperature added onto 30 mL of ice-cold deionized water. The cool solution was acidified to a pH~2 with 12N HCl. A precipitate formed, and it was collected by vacuum filtration.

[00285] 3-(4-bromophenyl)-5-(4-methoxyphenyl)isoxazole (2a). In an oven dried round bottom flask, chalcone 1a [(E)-1-(4-bromophenyl)-3-(4-methoxyphenyl)prop-2-en-1-one], was dissolved in 100% ethanol. To the solution, 2 equivalents of hydroxylamine hydrochloride (1 mmol) were added. Then, potassium hydroxide was added to the solution and the reaction was refluxed for ~12 hours. Next, the reaction was quenched with deionized water and the organic product was extracted. The crude concentrated organic material was then added to a larger round bottom flask and it was dissolved in benzene. To the solution, manganese (IV) oxide was added and it was refluxed via a Dean-Stark apparatus for ~10 hours. The reaction was then vacuum filtered over celite and concentrated.

[00286] General Procedure of 3-(R-phenyl)-5-(4-methoxyphenyl)isoxazoles---isoxazole formation (2b-d). In an oven-dried round bottom flask, chalcone (1 mmol) is dissolved in 2- propanol. To the solution, 4 equivalents of hydroxylamine hydrochloride (4 mmol) are added. Then, ~1 mL of pyridine is added to the solution and the reaction is refluxed overnight. Next, the reaction is quenched with deionized water and the organic product is extracted. The crude concentrated organic material is then added to a larger round bottom flasked and it is dissolved in benzene. To the solution, manganese (IV) oxide is added and it is refluxed via a Dean-Stark apparatus for ~10 hours. The reaction is then vacuum filtered over celite and concentrated.

[00287] General procedure of 4-(3-(R-phenyl)isoxazol-5-yl)phenol-Deprotection of the methoxy- group (3a-d). The isoxazoles(2a-d)(~0.4 mmol) are dissolved in anhydrous dichloromethane (3 mL) in an oven-dried round bottom flask under nitrogen gas. Then, the solution is cooled to 0 °C using an ice water bath. Next, 3 equivalents of boron tribromide (BBr 3 ) are added to the round bottom flask dropwise very slowly over the course of 10 minutes. The reaction runs overnight warming to room temperature. The reaction is then poured onto ~20 mL of ice cold deionized water. The organic product is extracted using 3x10 mL diethyl ether. Then the phenol compound is extracted into an aqueous layer using 2x10 mL of 2M NaOH. The pH of this aqueous layer containing the phenol compound is adjusted to around 2 using 12N HCl, which is added dropwise. From this solution, the organic product is extracted using 2x10 mL of diethyl ether, washed with brine and dried with magnesium sulfate.

[00288] General procedure of 3-(R-phenyl)-5-(4-(2-(piperidin-1- yl)ethoxy)phenyl)isoxazole---addition of the basic side chain to the phenol (4a-d). Under nitrogen gas in an oven dried round bottom, the demethylated isoxazole (3a-d) are dissolved in 1 mL dimethylformamide. Next, the 1-(2-chloroethyl)piperidine hydrochloride (1 equivalent) is added to the solution. Then, potassium carbonate (1.6 equivalents) and cesium carbonate (1.1 equivalents) are added sequentially to the stirring solution. The reaction runs overnight for about 16 hours at room temperature. The reaction is then quenched with deionized water. The organic product is extracted with 3x10 mL of diethyl ether, washed with 1x5 mL 10%NaOH, 3x10 mL H 2 O, 1x10 mL brine, dried with magnesium sulfate, and concentrated.

[00289] The following compounds were prepared using the foregoing procedures.

[00290] 1a: (E)-1-(4-bromophenyl)-3-(4-methoxyphenyl)prop-2-en-1-one. 1 H NMR (500 MHz, Chloroform-d) į 7.90– 7.86 (d, 2H), 7.79 (d, J = 15.6 Hz, 1H), 7.66– 7.57 (m, 4H), 7.36 (d, J = 15.6, 1H), 6.94 (d, J = 8.8 Hz, 2H), 3.86 (s, 3H). 13 C NMR (126 MHz, CDCl3) į 189.48, 161.95, 145.39, 137.31, 131.95, 130.46, 130.06, 127.72, 127.49, 119.18, 114.57, 77.41, 77.16, 76.90, 55.55.

[00291] 1b: (E)-3-(4-methoxyphenyl)-1-(naphthalen-2-yl)prop-2-en-1-one. 1 H NMR (500 MHz, Chloroform-d) į 8.54 (s, 1H), 8.13– 7.84 (m, 5H), 7.71– 7.55 (m, 5H), 6.96 (d, J = 8.6 Hz, 2H), 3.88 (s, 3H). 13 C NMR (126 MHz, CDCl3) į 190.53, 161.89, 144.87, 136.01, 135.59, 132.77, 130.51, 129.94, 129.70, 128.70, 128.47, 128.01, 127.86, 126.93, 124.74, 119.93, 114.63, 77.52, 77.26, 77.01, 55.63. [00292] 1c: (E)-1-(4-fluorophenyl)-3-(4-methoxyphenyl)prop-2-en-1-one. 1 H NMR (499 MHz, Chloroform-d) į 8.09– 8.00 (m, 2H), 7.79 (d, J = 15.6 Hz, 1H), 7.60 (d, J = 8.7 Hz, 2H), 7.38 (d, J = 15.6 Hz, 1H), 7.17 (t, J = 8.6 Hz, 2H), 6.94 (d, J = 8.7 Hz, 2H), 3.86 (s, 3H). 13 C NMR (126 MHz, CDCl3) į 189.00, 161.92, 145.04, 131.14, 131.06, 130.40, 127.65, 119.42, 115.89, 115.71, 114.60, 77.40, 77.15, 76.89, 55.58.

[00293] 1d: (E)-3-(4-methoxyphenyl)-1-(p-tolyl)prop-2-en-1-one. 1 H NMR (499 MHz, Chloroform-d) į 7.91 (d, J = 8.0 Hz, 2H), 7.75 (d, J = 15.6, 1H), 7.57 (d, J = 8.6 Hz, 2H), 7.39 (d, J = 15.7, 1H), 7.26 (d, J = 15.8 Hz, 2H), 6.91 (d, J = 8.9 Hz, 2H), 3.82 (s, 3H), 2.40 (s, 3H). 13 C NMR (126 MHz, CDCl 3 ) į 190.02, 161.64, 144.26, 143.40, 135.97, 130.22, 129.33, 128.62, 127.78, 119.82, 114.46, 77.41, 77.15, 76.90, 55.44, 21.71.

[00294] 2a: 3-(4-bromophenyl)-5-(4-methoxyphenyl)isoxazole. 1 H NMR (499 MHz, Chloroform-d) į 7.79– 7.70 (m, 4H), 7.61 (d, J = 8.6 Hz, 2H), 7.00 (d, J = 8.9 Hz, 2H), 6.68 (s, 1H), 3.87 (s, J = 2.5 Hz, 3H). 13 C NMR (126 MHz, CDCl3) į 216.31, 160.80, 157.48, 139.44, 134.06, 131.76, 130.97, 129.16, 128.80, 123.61, 114.60, 77.40, 77.15, 76.89, 55.50.

[00295] 2b: 5-(4-methoxyphenyl)-3-(naphthalen-2-yl)isoxazole. 1 H NMR (499 MHz, Chloroform-d) į 8.31 (s, 1H), 7.92– 7.82 (m, 4H), 7.55 (ddt, J = 9.8, 6.8, 4.0 Hz, 4H), 7.02 (d, J = 8.8 Hz, 2H), 6.86 (s, 1H), 3.89 (s, 3H).

[00296] 2c: 3-(4-fluorophenyl)-5-(4-methoxyphenyl)isoxazole. 1 H NMR (500 MHz, Chloroform-d) į 7.49 (dd, J = 6.6, 2.0 Hz, 2H), 7.44 (d, J = 8.7 Hz, 2H), 7.17– 7.09 (m, 2H), 6.90– 6.86 (m, 2H), 6.72 (s, 1H), 3.83 (s, 3H). 13 C NMR (126 MHz, CDCl3) į 162.58, 160.87, 140.07, 131.30, 131.23, 129.24, 128.73, 115.72, 115.55, 114.77, 114.39, 55.50.

[00297] 2d: 5-(4-methoxyphenyl)-3-(p-tolyl)isoxazole. 1 H NMR (400 MHz, Chloroform-d) į 7.74 (t, J = 8.5 Hz, 4H), 7.29– 7.23 (m, 2H), 6.97 (d, J = 8.8 Hz, 2H), 6.66 (s, 1H), 3.85 (s, 3H),

56 2.39 (s, 3H). 13 C NMR (101 MHz, CDCl 3 ) į 140.16, 129.77, 129.70, 128.30, 127.53, 126.80, 126.53, 125.87, 120.50, 114.49, 96.19, 55.53, 21.57.

[00298] 3a: 4-(3-(4-bromophenyl)isoxazol-5-yl)phenol. 1 H NMR (499 MHz, Chloroform-d) į 7.73 (d, J = 8.6 Hz, 4H), 7.61 (d, J = 8.4 Hz, 2H), 6.95 (d, J = 8.6 Hz, 2H), 6.68 (s, 1H). 13 C NMR (126 MHz, CD 3 OD) į 172.62, 163.48, 161.06, 133.26, 129.51, 128.61, 125.19, 120.06, 116.93, 99.19, 96.78, 49.51, 49.34, 49.17, 49.00, 48.83, 48.66, 48.48.

[00299] 3b: 4-(3-(naphthalen-2-yl)isoxazol-5-yl)phenol.

[00300] 3c: 4-(3-(4-fluorophenyl)isoxazol-5-yl)phenol. 1 H NMR (499 MHz, Chloroform-d) į 7.84 (dd, J = 8.6, 5.3 Hz, 2H), 7.74 (d, J = 8.6 Hz, 2H), 7.17 (t, J = 8.6 Hz, 2H), 6.94 (d, J = 8.5 Hz, 2H), 6.67 (s, 1H). 13 C NMR (126 MHz, CD3OD) į 172.47, 163.49, 161.03, 130.00, 129.93, 129.35, 128.60, 126.93, 120.12, 116.92, 96.82, 49.51, 49.34, 49.17, 48.99, 48.82, 48.65, 48.48.

[00301] 3d: 4-(3-(p-tolyl)isoxazol-5-yl)phenol. 1 H NMR (499 MHz, Chloroform-d) į 7.76 (q, J = 6.9, 6.4 Hz, 4H), 7.30 (d, J = 7.5 Hz, 2H), 6.96 (d, J = 7.1 Hz, 2H), 6.70 (d, J = 1.2 Hz, 1H). 1 3 C NMR (126 MHz, CDCl3) į 170.28, 157.46, 140.24, 129.74, 128.60, 127.83, 126.84, 126.49, 125.92, 116.08, 96.26, 21.58. [00302] 4a: 3-(4-bromophenyl)-5-(4-(2-(piperidin-1-yl)ethoxy)phenyl)isox azole. 1 H NMR (499 MHz, Chloroform-d) į 7.78– 7.68 (m, 4H), 7.61 (t, J = 7.1 Hz, 2H), 7.00 (d, J = 8.7 Hz, 2H), 6.67 (s, 1H), 4.18 (t, J = 6.0 Hz, 2H), 2.82 (t, J = 5.9 Hz, 2H), 2.55 (s, 4H), 1.63 (dt, J = 11.1, 5.5 Hz, 4H), 1.46 (s, 2H). 13 C NMR (126 MHz, CDCl 3 ) į 170.87, 162.14, 160.62, 132.26, 128.44, 127.58, 127.40, 124.33, 120.27, 115.21, 96.04, 77.40, 77.15, 76.89, 66.24, 57.86, 29.84, 25.95, 24.23. HRMS-ESI: m/z [M+H] + for C22H23BrN2O2, calculated 427.09; observed 427.1024. [00303] 4b: 3-(naphthalen-2-yl)-5-(4-(2-(piperidin-1-yl)ethoxy)phenyl)is oxazole. 1 H NMR (499 MHz, Chloroform-d) į 8.30 (s, 1H), 8.01 (d, J = 8.5 Hz, 1H), 7.91 (dd, J = 26.5, 6.1 Hz, 3H), 7.80 (d, J = 7.7 Hz, 2H), 7.54 (dd, J = 5.7, 2.7 Hz, 2H), 7.01 (d, J = 7.8 Hz, 2H), 6.85 (s, 1H), 4.22 (t, J = 5.6 Hz, 2H), 2.90 (t, J = 5.7 Hz, 2H), 2.63 (s, 4H), 1.71– 1.63 (m, 4H), 1.48 (s, 2H). 13 C NMR (126 MHz, CDCl3) į 170.55, 163.07, 160.33, 134.15, 133.35, 128.84, 128.59, 127.96, 127.58, 127.05, 126.80, 126.74, 126.62, 124.14, 120.58, 115.16, 96.40, 77.40, 77.14, 76.89, 65.79, 57.49, 54.84, 25.48, 23.97. HRMS-ESI: m/z [M+H] + for C26H26N2O2, calculated 399.20; observed 399.2073. [00304] 4c: 3-(4-fluorophenyl)-5-(4-(2-(piperidin-1-yl)ethoxy)phenyl)iso xazole. 1 H NMR (500 MHz, Chloroform-d) į 7.89– 7.80 (m, 2H), 7.75 (d, J = 8.8 Hz, 2H), 7.16 (t, J = 8.6 Hz, 2H), 7.00 (d, J = 8.8 Hz, 2H), 6.66 (s, 1H), 4.18 (t, J = 6.0 Hz, 2H), 2.82 (t, J = 5.9 Hz, 2H), 2.55 (s, 4H), 1.63 (dt, J = 11.0, 5.4 Hz, 4H), 1.48 (s, 2H). 13 C NMR (126 MHz, CDCl 3 ) į 162.14, 160.55, 128.86, 128.79, 128.30, 127.55, 120.32, 116.21, 116.03, 115.17, 96.09, 77.40, 77.14, 76.89, 66.22, 57.87, 55.19, 25.96, 24.23. HRMS-ESI: m/z [M+H] + for C22H23FN2O2, calculated 367.17; observed 367.1823.

[00305] 4d: 5-(4-(2-(piperidin-1-yl)ethoxy)phenyl)-3-(p-tolyl)isoxazole. 1 H NMR (400 MHz, Chloroform-d) į 7.86– 7.68 (m, 4H), 7.28 (s, 2H), 6.98 (d, J = 8.7 Hz, 2H), 6.67 (s, 1H), 4.16 (t, J = 5.9 Hz, 2H), 2.80 (t, J = 5.9 Hz, 2H), 2.53 (s, 4H), 2.40 (s, 3H), 1.70– 1.57 (m, 4H), 1.46 (s, 2H). 13 C NMR (101 MHz, CDCl 3 ) į 170.29, 160.41, 140.14, 129.69, 128.26, 127.49, 126.78, 125.85, 120.48, 115.09, 96.17, 77.47, 77.15, 76.83, 66.18, 57.89, 55.18, 25.98, 24.24, 21.56. HRMS-ESI: m/z [M+H] + for C23H26N2O2, calculated 362.20; observed 363.2076.

[00306] Compounds of formula (I) where G 2 is (vii) may be prepared as generally shown in Scheme 5.

Scheme 5: General Chemical Synthesis Scheme of Target Imidazoles

[00307] (i) To a solution of 4-hydroxybenzaldehyde (2.36 mmol) in dimethylformamide (12 ml), K 2 CO 3 (3.54 mmol) and Cs 2 CO 3 (2.36 mmol) are added, followed by 1-(2- chloroethyl)piperidine-hydrochloride (2.60 mmol). The reaction mixture is allowed to stir overnight at room temperature. After completion of the reaction, extraction is done using ethyl acetate. The organic layer is washed with brine, dried over sodium sulfate, concentrated under vacuum, and partially purified by flash column chromatography using methanol/dichloromethane to remove the nonpolar impurities. The benzaldehyde with basic side chain (at para position) is then directly taken to the next step. [00308] (ii) To 4,4’-disubstitutedbenzil (0.74 mmol) and benzaldehyde with the basic side chain from step (i) (1.1l mmol) is added formamide (5 mL). The bright yellow suspension is heated to reflux (220 °C) for 2 hours. The reaction mixture is then cooled to room temperature then to 0 °C. The reaction mixture is poured into water and extracted with ethyl acetate. The organic layer is washed with brine and dried over Na2SO4 then concentrated under vacuum and purified by flash chromatography using methanol/dichloromethane to provide the product in 46- 55% yield.

[00309] The following compounds were prepared using the foregoing procedures.

[00310] 1-(2-(4-(4,5-bis(4-methoxyphenyl)-1H-imidazol-2-yl)phenoxy)e thyl)piperidine (TSI-14-14)

[00311] 1 H NMR (500 MHz, Chloroform-d) į 7.77 (d, J = 8.7 Hz, 2H), 7.41 (d, J = 7.8 Hz, 4H), 6.83 (dd, J = 14.3, 8.7 Hz, 6H), 4.09 (t, J = 5.8 Hz, 2H), 3.79 (s, 6H), 2.76 (t, J = 5.8 Hz, 2H), 2.48 (s, 4H), 1.56 (p, J = 5.6 Hz, 4H), 1.43 (s, 2H). 13 C NMR (126 MHz, CDCl 3 ) į 163.3, 159.2, 158.9, 146.0, 129.4, 127.1, 123.5, 114.9, 114.1, 65.9, 58.1, 55.5, 55.2, 25.9, 24.3. HRMS- ESI: m/z [M+H] + for C30H34N3O3, calculated 484.2600; observed 484.2600.

[00312] 1-(2-(4-(4,5-di-p-tolyl-1H-imidazol-2-yl)phenoxy)ethyl)piper idine (TSI-14-51)

[00313] 1 H NMR (500 MHz, Chloroform-d) į 8.06 (d, J = 8.4 Hz, 2H), 7.30 (d, J = 7.9 Hz, 4H), 6.97 (d, J = 8.1 Hz, 4H), 6.63 (d, J = 8.4 Hz, 2H), 4.15 (s, 2H), 3.11 (s, 2H), 2.90 (s, 4H), 2.22 (s, 6H), 1.70 (s, 4H), 1.42 (s, 2H). 13 C NMR (126 MHz, CDCl 3 ) į 158.5, 145.3, 137.7, 131.3, 129.3, 128.6, 128.4, 128.1, 121.5, 114.6, 63.0, 56.2, 54.0, 23.1, 22.0, 21.5. HRMS-ESI: m/z [M+H] + for C 30 H 34 N 3 O, calculated 452.2702; observed 452.2702. [00314] 1-(2-(4-(4,5-bis(4-bromophenyl)-1H-imidazol-2-yl)phenoxy)eth yl)piperidine

[00315] 1 H NMR (500 MHz, Methanol-d 4 ) į 7.96 (d, J = 8.9 Hz, 2H), 7.50 (d, J = 8.5 Hz, 4H), 7.37 (d, J = 8.5 Hz, 4H), 7.14 (d, J = 8.9 Hz, 2H), 4.49– 4.41 (m, 2H), 3.61– 3.54 (m, 2H), 3.36 (s, 4H), 1.92 (q, J = 5.6 Hz, 4H), 1.70 (s, 2H). 13 C NMR (126 MHz, CD 3 OD) į 158.7, 147.2, 131.7, 129.9, 127.5, 121.3, 115.0, 62.3, 56.0, 53.8, 23.0, 21.5. HRMS-ESI: m/z [M+H] + for C28H28N3OBr2, calculated 580.0599; observed 580.0599. xy)ethyl)piperidine (TSI-14-19)

[00317] 1 H NMR (500 MHz, Chloroform-d) į 7.74 (d, J = 8.4 Hz, 2H), 7.47 (s, 4H), 7.24 (dt, J = 12.3, 7.0 Hz, 7H), 6.82 (d, J = 8.4 Hz, 2H), 4.08 (t, J = 5.6 Hz, 2H), 2.74 (t, J = 5.6 Hz, 2H), 2.44 (s, 4H), 1.56– 1.49 (m, 4H), 1.41 (s, 2H). 13 C NMR (126 MHz, CDCl 3 ) į 159.3, 146.7, 128.6, 128.2, 127.3, 123.3, 114.9, 65.9, 58.1, 55.3, 25.9, 24.3. HRMS-ESI: m/z [M+H] + for C28H30N3O, calculated 424.2389; observed 424.2391.

[00319] 1 H NMR (500 MHz, Chloroform-d) į 8.06 (d, J = 8.2 Hz, 2H), 7.38 (dd, J = 8.3, 5.5 Hz, 4H), 6.88 (t, J = 8.6 Hz, 4H), 6.68 (d, J = 8.1 Hz, 2H), 4.22 (s, 2H), 3.18 (s, 2H), 2.99 (s, 4H), 1.76 (s, 4H), 1.46 (s, 2H). 13 C NMR (126 MHz, CDCl3) į 163.3, 161.3, 158.2, 146.0, 131.4, 130.3, 130.2, 128.2, 128.0, 122.9, 115.7, 115.5, 114.7, 62.9, 56.4, 54.2, 22.9, 21.9. HRMS-ESI: m/z [M+H] + for C28H28N3OF2, calculated 460.2200; observed 460.2190. [00320] 4,4'-(2-(4-(2-(piperidin-1-yl)ethoxy)phenyl)-1H-imidazole-4, 5-diyl)diphenol (TSI- 14-54)

[00321] 1 H NMR (500 MHz, Methanol-d4) į 7.96 (d, J = 8.9 Hz, 2H), 7.50 (d, J = 8.5 Hz, 4H), 7.37 (d, J = 8.5 Hz, 4H), 7.14 (d, J = 8.9 Hz, 2H), 4.49– 4.41 (m, 2H), 3.61– 3.54 (m, 2H), 3.36 (s, 4H), 1.92 (q, J = 5.6 Hz, 4H), 1.70 (s, 2H). 13 C NMR (126 MHz, CD 3 OD) į 158.7, 147.2, 131.7, 129.9, 127.5, 121.3, 115.0, 62.3, 56.0, 53.8, 23.0, 21.5. [00322] General scheme and procedure for the synthesis of imidazoles TSI-14-15 & TSI- 14-20

[00323] To a solution of the imidazole with the basic side chain (2.36 mmol) in

dimethylformamide (12 mL), K2CO3 (3.54 mmol) and Cs2CO3 (2.36 mmol) are added, followed by ethyl iodide (2.60 mmol). The reaction mixture is allowed to stir overnight at room

temperature. After completion of the reaction, extraction is done using ethyl acetate. The organic layer is washed with brine, dried over sodium sulfate, concentrated under vacuum, and purified by flash column chromatography using methanol/dichloromethane to obtain the product in 85-88 % yield.

[00325] 1 H NMR (500 MHz, Chloroform-d) į 7.59 (d, J = 8.7 Hz, 2H), 7.45 (d, J = 8.8 Hz, 2H), 7.31 (d, J = 8.6 Hz, 2H), 6.99 (d, J = 8.6 Hz, 4H), 6.73 (d, J = 8.9 Hz, 2H), 4.16 (t, J = 6.1 Hz, 2H), 3.87 (d, J = 5.6 Hz, 5H), 3.74 (s, 3H), 2.80 (t, J = 6.1 Hz, 2H), 2.52 (s, 4H), 1.61 (p, J = 5.6 Hz, 4H), 1.45 (s, 2H), 1.00 (t, J = 7.1 Hz, 3H). 13 C NMR (126 MHz, CDCl 3 ) į 159.9, 159.5, 158.2, 147.0, 137.5, 132.6, 130.6, 128.1, 128.0, 127.9, 124.3, 124.1, 114.9, 114.7, 113.7, 66.4, 58.1, 55.5, 55.3, 39.7, 26.2, 24.4, 16.5. HRMS-ESI: calculated 512.2913; observed 512.2908.

[00327] 1 H NMR (500 MHz, Chloroform-d) į 7.63 (d, J = 8.7 Hz, 2H), 7.52 (d, J = 7.2 Hz, 2H), 7.47 (t, J = 7.1 Hz, 3H), 7.45– 7.41 (m, 2H), 7.19 (t, J = 7.5 Hz, 2H), 7.12 (t, J = 7.3 Hz, 1H), 7.02 (d, J = 8.7 Hz, 2H), 4.19 (t, J = 6.0 Hz, 2H), 3.92 (q, J = 7.1 Hz, 2H), 2.83 (t, J = 6.0 Hz, 2H), 2.56 (s, 4H), 1.65 (p, J = 5.6 Hz, 4H), 1.47 (s, 2H), 1.01 (t, J = 7.1 Hz, 3H). 13 C NMR (126 MHz, CDCl3) į 159.5, 147.5, 137.8, 135.0, 132.0, 131.3, 130.7, 129.3, 129.2, 128.2, 127.0, 126.3, 124.2, 115.0, 66.3, 58.1, 55.3, 39.8, 26.1, 24.4, 16.4. HRMS-ESI: m/z [M+H] + for C 30 H 34 N 3 O, calculated 452.2702; observed 452.2699. Scheme 6. Synthesis of TSI-14-56

[00328] To a mixture of 4,4’-dimethoxybenzil (0.74 mmol) and benzaldehyde with the basic side chain (0.74 mmol) in a sealed tube is added ammonium acetate (0.74 mmol), methylamine (0.74 mmol) and sodium dihydrogen phosphate (1.5 mmol). The mixture is heated at 150 o C for 5 hours. The reaction mixture is cooled to room temperature and partitioned between ethyl acetate and water. The organic layer is collected and washed with brine, dried over sodium sulfate, concentrated under vacuum, and purified by flash column chromatography using methanol/dichloromethane to obtain the pure product in 65 % yield. [00329] 1-(2-(4-(4,5-bis(4-methoxyphenyl)-1-methyl-1H-imidazol-2- yl)phenoxy)ethyl)piperidine TSI-14-56

[00330] 1 H NMR (500 MHz, Chloroform-d) į 7.64 (d, J = 8.7 Hz, 2H), 7.48 (d, J = 8.9 Hz, 2H), 7.31 (s, 2H), 7.00 (dd, J = 7.9, 5.9 Hz, 4H), 6.76 (d, J = 8.9 Hz, 2H), 4.19 (t, J = 5.9 Hz, 2H), 3.87 (s, 3H), 3.76 (s, 3H), 3.44 (s, 3H), 2.86 (t, J = 5.9 Hz, 2H), 2.59 (s, 4H), 1.65 (dt, J = 11.2, 5.5 Hz, 4H), 1.47 (s, 2H). 13 C NMR (126 MHz, CDCl3) į 159.9, 159.3, 158.3, 147.6, 137.3, 132.4, 130.6, 129.2, 128.2, 127.9, 124.0, 123.8, 114.9, 114.7, 113.7, 66.1, 57.9, 55.5, 55.4, 55.2, 33.2, 25.9, 24.3. HRMS-ESI: m/z [M+H] + for C31H36N3O3, calculated 498.2757; observed 498.2747.

[00331] Compounds of formula (I) where G 2 has formula (vi) may be prepared as generally illustrated in Scheme 7. Scheme 7. Synthesis of TSI-14-39

[00332] A mixture of 4-methoxyaniline (2 mmol) and 4-hydroxybezaldehyde (2 mmol), ammonium acetate (3 mmol) and 2-bromo-1-phenylethanone (2 mmol) is heated at 130 o C for 2 hours. After the completion of reaction, the reaction mixture is cooled to room temperature, the crude product is partitioned between ethyl acetate and water. The organic layer is washed with brine, dried over sodium sulfate, concentrated and purified by flash column chromatography using ethyl acetate/hexane. In the next step, the basic side chain is installed on the free phenol following a procedure analogous to that shown above to obtain the product in 70 % yield.

[00333] 1-(2-(4-(1,4-bis(4-methoxyphenyl)-1H-imidazol-2-yl)phenoxy)e thyl)piperidine

4H), 1.62 (p, J = 5.6 Hz, 4H), 1.47 (s, 2H). 13 C NMR (126 MHz, CDCl 3 ) į 159.3, 159.1, 158.9, 147.1, 141.3, 131.9, 130.3, 127.3, 127.2, 126.5, 123.3, 117.7, 114.8, 114.5, 114.2, 77.6, 77.3, 77.0, 66.2, 58.1, 55.8, 55.5, 55.3, 26.1, 24.4. HRMS-ESI: m/z [M+H] + for C30H34N3O3, calculated 484.2600; observed 484.2591.

[00335] Compounds of formula (I) where G 2 has formula (viii) and X 1 is S may be prepared as generally illustrated in Scheme 8. Scheme 8. Synthesis of 4,5-bis(4-methoxyphenyl)-2-(4-(2-(piperidin-1- yl)ethoxy)phenyl)thiazole, TSI-14-17

[00336] To a solution of sodium hydrosulfide hydrate in dimethylformamide (1.8 mL) and water (0.25 mL) is added 4-hydroxybenzonitrile and NH4Cl with stirring. The reaction mixture is stirred for 22 hours at 40 °C. To this reaction mixture under ice cooling is added 5 mL of 5N HCl followed by water and allowed to stir for a while. Solid formed after stirring is filtered, dried and used as such for thiazole synthesis.

[00337] To a solution of 4,4ƍ-Dimethoxybenzoin (1 equivalent) in dichloromethane at 0 °C is added 4-Toluenesulfonyl chloride (1 equivalent) followed by trimethylamine (2 equivalents) and catalytic N,N-dimethylaminopyridine with constant stirring. The reaction mixture is stirred at room temperature overnight. After completion of reaction, the crude product is partitioned between ethyl acetate and water. The ethyl acetate layer is washed with brine and then dried over Na2SO4 and evaporated under reduced pressure. The crude reaction mixture is purified by column chromatography using ethyl acetate/hexane.

[00338] In the next step, the tosylated-4,4ƍ-dimethoxybenzoin (1.0 mmol) and thiobenzamide (1.2 mmol) are added to 1-butyl-3-methylimidazolium hexafluorophosphate (BMIM-PF6) (2 mL). The resulting mixture is heated at 110 °C for 3-4 hours. Subsequently, the reaction mixture is extracted with diethylether multiple times. The combined etheral solution is vacuum evaporated. The residue is chromatographed on silica gel column using

methanol/dichloromethane to provide the product in 55 % yield. [00339] 4,5-bis(4-methoxyphenyl)-2-(4-(2-(piperidin-1-yl)ethoxy)phen yl)thiazole (TSI-14- 17)

[00340] 1 H NMR (500 MHz, Chloroform-d) į 7.92 (d, J = 8.8 Hz, 2H), 7.54 (d, J = 8.8 Hz, 2H), 7.31 (d, J = 8.8 Hz, 2H), 6.96 (d, J = 8.8 Hz, 2H), 6.87 (d, J = 8.8 Hz, 2H), 6.84 (d, J = 8.8 Hz, 2H), 4.19 (t, J = 5.8 Hz, 2H), 3.83 (s, 3H), 3.82 (s, 3H), 2.84 (t, J = 5.3 Hz, 2H), 2.58 (s, 4H), 1.70– 1.62 (m, 4H), 1.48 (s, 2H). 13 C NMR (126 MHz, CDCl3) į 164.8, 160.5, 159.6, 159.3, 150.0, 134.2131.0, 130.5, 128.1, 128.0, 127.2, 124.9, 115.1, 114.4, 113.9, 66.3, 58.1, 55.6, 55.5, 55.47, 55.3, 26.1, 24.4. HRMS-ESI: m/z [M+H] + for C30H33N2O3S, calculated 501.2212;

observed 501.2211.

[00341] Compounds of formula (I) where G 2 has formula (viii) and X 1 is O may be prepared as generally illustrated in Scheme 9. Scheme 9. Synthesis of 4,5-bis(4-methoxyphenyl)-2-(4-(2-(piperidin-1-

[00342] TSI-14-16 was prepared using procedures analogous to those used to prepare the thiazole TSI-14-17, except in place of 4-hydroxy thiobenzamide , 4-hydroxy benzamide was used to obtain the product in 70 % yield. [00343] 4,5-bis(4-methoxyphenyl)-2-(4-(2-(piperidin-1-yl)ethoxy)phen yl)oxazole (TSI-14- 16)

[00344] 1 H NMR (499 MHz, Chloroform-d) į 8.06 (dd, J = 8.9, 2.4 Hz, 2H), 7.64 (dd, J = 8.9, 2.5 Hz, 2H), 7.59 (dd, J = 9.0, 2.5 Hz, 2H), 6.99 (dd, J = 8.9, 2.4 Hz, 2H), 6.93 (td, J = 8.8, 2.4 Hz, 4H), 4.19 (t, J = 7.0 Hz, 2H), 3.85 (s, 6H), 2.84 (t, J = 6.9 Hz, 2H), 2.57 (s, 4H), 1.65 (p, J = 5.0 Hz, 4H), 1.48 (s, 2H). 13 C NMR (126 MHz, CDCl 3 ) į 160.7, 159.9, 159.8, 159.6, 144.6, 135.4, 129.5, 128.2, 125.6, 122.2, 120.7, 115.0, 114.3, 114.2, 66.3, 58.0, 55.6, 55.5, 55.3, 26.1, 24.3. HRMS-ESI: m/z [M+H] + for C30H33N2O4, calculated 485.2440; observed 485.2441. 6. Biological Data

[00345] TLR-mediated cell activation can be mimicked by chemically induced dimerization of the TLR signaling proteins MyD88 and TRAF6 (Hacker et al., Nature 439, 204-207 (2006); Zhou et al., Proceedings of the National Academy of Sciences of the United States of America 108, E998-1006 (2011)). To this end, MyD88 and TRAF6 were fused to the protein Gyrase B (GyrB), which inducibly dimerizes upon exposure to the bivalent antibiotic coumermycin (CM) (Farrar et al., Nature 383, 178-181 (1996)). Expression of such fusion proteins allows selective activation of the TLR pathway at specific signaling levels, recapitulating the hierarchy of signal transduction during physiological receptor activation (Hacker et al., Nature 439, 204-207 (2006)). This approach was extended to TIRAP, whose GyrB (CM)-mediated dimerization results in cell activation via MyD88. When stably expressed in HEK293T-cells that are equipped with an NF-kB luciferase reporter, this system of three inducible TLR-signaling proteins allows assignment of interference with signal transduction, e.g. by small molecule drugs, to distinct levels of the signaling cascade (Fig.1A). This system is used in a step-wise, phenotypic drug screening approach in order to identify compounds that target the TLR-specific signaling level upstream of TRAF6.

[00346] In order to recapitulate the TLR2-/TLR4-induced signaling pathway, a similar approach was used developing an inducible form of the TIR-domain containing protein TIRAP. Genetic and biochemical evidence suggests that TIRAP acts upstream of MyD88, providing a molecular bridge that transduces activation (oligomerization) of TLR2 and TLR4 to MyD88 (Yamamoto et al., Nature 420, 324-329 (2002); Fitzgerald et al., Nature 413, 78-83 (2001); Horng et al., Nature 420, 329-333 (2002)). Consistent with this concept, GyrB-TIRAP fusion proteins showed robust CM-inducible activity when expressed in HEK293T cells, similar to fusion proteins of GyrB and MyD88/ TRAF6 (Fig.1B). To confirm the hierarchical organization of TIRAP acting upstream of MyD88, MyD88 was deleted in GyrB-TIRAP cells by sgRNA-/ CAS9-mediated deletion, which resulted in complete loss of TIRAP-mediated NF-kB activation (Fig.1C,D). Consistent with the essential, yet selective function of MyD88 in the TLR/IL-1R pathway, IL-1ȕ-mediated NF-kB activity was also abolished, while TNFĮ was not affected (Fig. 1D). As such, GyrB-TIRAP can be used to activate the TLR-pathway upstream of MyD88, recapitulating TLR2- and TLR4-mediated signal transduction. [00347] Validation of cell lines for high throughput screening (HTS). The three inducible cell lines (TIRAP-GyrB, MyD88-GyrB, TRAF6-GyrB) were systematically optimized for large- scale high throughput screening (HTS), first based on manual experiments by 96-well (bench- top) format, followed by 384-well format on HTS robots. As pathway-specific reference compound PS1145 was used, a selective inhibitor of the IkBĮ-kinase complex (IKK), which is required for NF-kB activation downstream of TRAF6. As non-specific reference compound staurosporine was used, a kinase inhibitor with largely non-selective toxicity. PS1145 and staurosporine concentrations were titrated to establish PS1145 at 30 mM and Staurosporine at 40 mM, which resulted in 100% specific and non-specific pathway inhibition in the presence of 100 nM CM, respectively. Assay validation, including analysis of within-plate and between-plate variation, was performed on the HTS platform, resulting consistently in Z’-factors of > 0.5, demonstrating the robustness of this phenotypic screening platform (Fig.1E).

[00348] Screening paradigm. Based on the screening system described above, the screening paradigm depicted in Fig.2A was established. TIRAP-GyrB cells are used first to identify compounds with NF-kB-specific inhibitory activity ³50% and non-specific activity (toxicity) £20% (primary screen, all compounds at 12 mM, Fig.2A). Identified hits are moved forward to dose-response experiments based on all three cell lines (secondary screen). Results from these cell lines are interpreted primarily based on selectivity, identifying compounds with preferential activity against individual adaptor proteins. Compounds that inhibit TRAF6 are eliminated as TLR-‘non-specific’. Compounds with selective activity against TIRAP, but not MyD88 and TRAF6 suggest selectivity for the TLR2-/ TLR4-pathway, while activity against TIRAP and MyD88 suggest general or‘Pan’-activity against all TLRs (Fig.2A). Compounds displaying selectivity are then investigated in tertiary screens based on re-synthesized compounds by using physiological TLR ligand-mediated cell stimulation of macrophage cell lines or primary macrophages. As cell line RAW264.7 cells were used, a well-characterized mouse macrophage cell line that produces robust amounts of TNFĮ upon stimulation with various pathogen components, including TLR ligands (Hacker et al., The Journal of experimental medicine 192, 595-600 (2000)).

[00349] High Throughput Screening. TIRAP-GyrB, MyD88-GyrB and TRAF6-GyrB cell lines were maintained in growth medium containing Phenol red (DMEM (Invitrogen), supplemented with 10% (v/v) FCS (Hyclone), 50 mM 2-mercaptoethanol, antibiotics (penicillin G (100 IU/mL) and streptomycin sulfate (100 IU/mL)) and pyruvate (1 mM)), which was replaced by growth medium without Phenol red during HTS assays. In the primary high throughput screening, TIRAP-GyrB cells (10,000 cells in 25 mL assay medium) were seeded in white 384-well solid bottom tissue culture-treated plates (PerkinElmer) with a WellMate dispenser (Thermo Scientific Matrix) and plates were incubated in an automated tissue culture incubator (Liconic Instruments). After 24 hour incubation, DMSO stock solutions of test chemicals (the St. Jude Bioactive collection), along with coumermycin A1 (Sigma), PS-1145 (Sigma), staurosporine (LC Laboratories) or DMSO (Fisher Scientific) were transferred with a Pin Tool (V&P Scientific) equipped with a 10H pin head to give a final test chemical concentration of 12 mM, along with 100 nM coumermycin A1. In addition, groups of wells with 100 nM coumermycin A1 alone, and 100 nM coumermycin A1 with 30 mM PS-1145 or 40 mM staurosporine were included in each plate as well. The final DMSO concentration in each well was 0.24%.24 hours later, each plate was treated with 5 mL/well DPBS (Fisher Scientific) diluted (1-to-12 dilution) AlamarBlue (Invitrogen). After brief centrifugation, the plate was incubated at 37 °C for 1 hour and cooled down at room temperature for 15 minutes before the fluorescence signal from each well was determined with an Envision HTS microplate reader (PerkinElmer, Waltham, MA) with excitation wavelength of 492 nm and emission wavelength of 590 nm. Next, SteadyLite HTS luminescence assay reagent (25 mL/well, PerkinElmer) was dispensed into each well followed by 20 minute room temperature incubation. The luminescence signals for individual wells were then measured with the Envision plate reader. In the

AlamarBlue cell toxicity assay, the 40 mM staurosporine with 100 nM coumermycin A1 group and the 100 nM coumermycin A1 alone group were assigned as the positive (100% inhibition) and negative (0% inhibition) controls, respectively. In the luminescence assay, the 30 mM PS- 1145 with 100 nM coumermycin A1 group and the 100 nM coumermycin A1 alone group were assigned as the corresponding positive (100% inhibition) and negative (0% inhibition) controls. Test compound activity was normalized to those of positive and negative controls in the individual assays.213 unique chemicals with luminescence inhibitory activity ³ 50% as well as with AlamarBlue cytotoxic inhibitory activity £ 20% were selected for further dose response confirmation testing.

[00350] In the dose confirmation tests, the basic primary screening protocol was followed with minor modifications. Those modifications included that chemicals were tested in 10 concentration levels from 56 mM to 2.8 nM, along with 100 nM coumermycin A1 in triplicates. Compounds were transferred with 100H pins which transfer 140 nL liquid and the final DMSO concentration in each well was 1.12%. In addition, MyD88-GyrB cells (3,500 cells/well) and TRAF6-GyrB cells (5,000 cells/well) were also tested in the dose response confirmation tests with a similar assay setting described for the TIRAP-GyrB cell screening. The controls and data normalized for the AlamarBlue and luminescence assays in the MyD88-GyrB- and TRAF6- GyrB cell-based tests were the same as the TIRAP-GyrB cell-based test. The activity data for individual chemicals were fit into sigmoidal dose-response curves if applicable to derive IC 50 values with GraphPad Prism 7.00 (GraphPad Software).

[00351] Calculation of Z’ values. The Zƍ values were calculated using the equation:

where ı + is the standard deviation of the negative control group (negative control groups in the luciferase reporter assay and AlamarBlue cytotoxicity assay for each cell line as defined above), ı- is the standard deviation of the positive control group (positive control groups in the luciferase reporter assay and AlamarBlue cytotoxicity assay for each cell line as defined above); Mean + is the mean of the negative control group, and Mean- is the mean of the positive control group.

[00352] Bioactive compound library screening for TLR-inhibitory compounds. A total of 4364 unique compounds were contained in the bioactive compound library used (a total of 8,904 compounds including replicates), which was acquired from various sources and included 913 FDA (US Food and Drug Administration)-approved drugs or clinical candidates. Based on activity at 12 mM concentration, 213 unique hits were identified that inhibited TIRAP-mediated NF-kB activity more than 50% (with cytotoxicity £ 20%). These 213 hits were further analyzed in a dose-response experiment (2.8 nM– 56 mM) using all three cell lines. Only 8 compounds exhibited preferential activity against TIRAP or TIRAP/MyD88 over TRAF6 (ratio IC50 > 5- fold).

[00353] The activity of all 8 compounds was confirmed based on re-synthesized compounds using the HTS-screening system; however, seven of the eight compounds were excluded due to lack of activity on mouse RAW264.7 cells (3 compounds) or additional activity against cell activation by TNFĮ or Curdlan, a beta-1,3-glucan that activates cells TLR-independently via Dectin-1 (4 compounds). The remaining compound, methyl-piperidino-pyrazole (MPP), a basic side-chain-containing pyrazole (BSC-pyrazole) that had originally been developed as an estrogen receptor alpha (ERĮ)-selective inhibitor, exhibited promising properties (Sun et al.,

Endocrinology 143, 941-947 (2002)). MPP inhibited TIRAP- and MyD88-mediated NF-kB activity comparably at low mM concentrations, suggesting general (‘Pan’)-TLR-inhibitory activity (Fig.2B). Consistent with this interpretation, MPP interfered with TNFĮ release from primary macrophages that were stimulated with CpG-DNA (TLR9), Resiquimod (R848; TLR7), Pam3Cys (TLR2) or LPS (TLR4)(Fig.2C). In contrast, MPP did not significantly reduce TNFĮ release induced by Curdlan. As such, MPP interferes selectively with TLR-induced cell activation, specifically at the TLR-restricted signaling level‘between’ MyD88 and TRAF6.

[00354] MPP-dependent inhibition of major TLR signaling pathways. TLR signaling is diversified at the level of TRAF6 towards various well-defined pathways, including the NF-kB and mitogen-activated protein kinase (MAPK) pathways (Fig.1A). To investigate the effect of MPP on TLR-induced signaling pathways, we analyzed NF-kB and MAPK pathways in RAW264.7 cells upon CpG-DNA activation. At 10 mM concentration, MPP blocked degradation of IkBĮ, reflecting NF-kB activation, as well as phosphorylation of different MAPK almost completely (Fig.2D). No apparently preferential activity towards a specific pathway was observed, supporting the interpretation that MPP acts upstream of TRAF6. We extended these investigations to the TLR7 ligand R848, whose activation of the NF-kB- and MAPK-pathways was strongly reduced, albeit not completely blocked by MPP (Fig.2D). Curdlan-induced cell activation remained largely unaffected, as expected from the previous experiment (Fig.2C and D).

[00355] Given that MPP was developed as ERĮ inhibitor, we tested whether this activity was related to its TLR-inhibitory activity using macrophages derived from ERĮ-deficient mice. As shown in Fig.2E and F, MPP inhibited TLR9-induced TNFĮ release and activation of signaling pathways comparably in both wildtype and ERĮ-deficient bone marrow-derived macrophages (BMM), strongly suggesting TLR- and ERĮ inhibition are independent events. This

interpretation is further supported by structure activity relationship (SAR) analyses based on MPP analogs (see below). Together, the data suggest that MPP blocks the MyD88-pathway upstream of TRAF6, independent of ERĮ inhibition.

[00356] MPP interference with dimerization of the MyD88 TIR domain. Different lines of evidence suggested that MPP targets a molecular event upstream of TRAF6, which involves the step-wise assembly of the MyD88-marshalled signaling complex composed of members of the IRAK family, TRAF6 and other, at least partially characterized signaling proteins (Fig.1A). To delineate the impact of MPP on TLR signaling, we investigated the assembly of the MyD88 signaling complex upon CpG-DNA stimulation. To this end we used stable isotope labeling with amino acids in cell culture (SILAC) and immuno-purified MyD88 from unstimulated and CpG- DNA-stimulated cells, either in the absence or presence of MPP. Samples containing purified proteins were analyzed by quantitative mass spectrometry (MS). As expected, the bait protein MyD88 was detected at similar levels in all conditions, reflected by protein ratios close to 1 (Fig. 3A). Known inducible components of the TLR/MyD88 signaling complex were unequivocally identified at increased levels upon activation. For example, IRAK4 was identified with 17 unique peptides at 8.4 fold higher levels in CpG-DNA-stimulated samples in comparison to

unstimulated control samples (Fig.3A). Strikingly, MPP treatment almost completely blocked recruitment of all activation-dependent MyD88-interacting proteins, including IRAK4, which directly interacts with MyD88 (Suzuki et al., Nature 416, 750-756 (2002); Li et al., Proceedings of the National Academy of Sciences of the United States of America 99, 5567-5572 (2002)). We confirmed this finding in small-scale immuno-precipitation (IP)/immuno-blotting (IB) experiments with antibodies against IRAK4 and TRAF6 (Fig.3B). As such, it appeared that MPP blocked a very early event in MyD88 activation.

[00357] The currently favored model of MyD88 activation involves TLR-induced, TIR- domain-mediated dimerization of MyD88 as the first step of signal induction, followed by a spiral assembly of further MyD88 molecules (Lin et al., Nature 465, 885-890 (2010)). Assembly of this higher-order complex, which at least in part involves homotypic death domain

interactions, appears to be required for recruitment and oligomerization of IRAK4. This model is based on various observations, including (i) the fact that TLR-signaling can be recapitulated by GyrB-mediated dimerization of MyD88, (ii) structural data obtained from the death domains of MyD88, IRAK4 and IRAK1 exhibiting a spiral architecture and (iii) the recruitment of additional MyD88 molecules upon MyD88-dimerization, as apparent in IP/IB experiments based the GyrB- MyD88 fusion protein and the (smaller) endogenous MyD88 (Fig.3C) (Lin et al., Nature 465, 885-890 (2010)); Hacker et al., Nature 439, 204-207 (2006)). The latter experimental setting was used to see if MPP blocked homotypic MyD88 oligomerization. Indeed, MPP inhibited recruitment of endogenous MyD88 (along with IRAK4), strongly suggesting that MyD88- dimerization itself is blocked by this compound (Fig.3C). To test this idea, co-IP experiments in HEK293T cells were performed based on different forms of MyD88. Given that both the TIR- domain and the DD-domain were shown to contribute to homotypic MyD88-interaction, the individual domains were co-expressed along with full length MyD88 and analyzed their interaction by IP/IB experiments (Burns et al., The Journal of biological chemistry 273, 12203- 12209 (1998)). While both domains co-purified with full length MyD88, as expected, MPP strongly reduced co-IP of the TIR-domain, but not the DD-domain (Fig.3D). Together, these data suggest that MPP inhibits directly homotypic TIR-domain interaction of MyD88, thereby preventing assembly of a higher order MyD88 complex, which is required for consecutive recruitment and activation of IRAK4.

[00358] To further substantiate MyD88 as MPP drug target, cellular thermal shift assays (CETSA) we performed based on HEK293T cells and endogenous MyD88. In these experiments an analog of MPP, TSI-13-57 was employed, which exhibited similar activity in the low micromolar range, but reduced toxicity in comparison to MPP. Exposure of cells with TSI-13-57 stabilized and protected MyD88 against increasing temperature as reflected by reduced loss of MyD88 in the soluble protein fraction (Fig.3E). Actin was not affected in the temperature range used and served as standard for quantification (Fig.3E and F). Of note, the concentrations of TSI-13-57 required for MyD88 stabilization and inhibition of TLR-mediated TNFĮ release corresponded well to each other, with EC50 values in the low micromolar range (Fig.3G and 3H). As such, all results, including functional data obtained from macrophages during TLR stimulation, data obtained from Co-IP studies and data based on CETSA, are consistent with the interpretation that MPP and its analog TSI-13-57 bind directly the TIR-domain of MyD88 to prevent TLR-mediated cell activation.

[00359] TIR-specific activity of MPP analogs as molecular basis for TLR-selective inhibitory activity. Given that not only MyD88, but also other proteins, including TLRs and TIRAP, contain TIR-domains, the apparent selectivity in TLR-inhibition might reflect preferential interference with specific TIR-domain interactions. To address this possibility, a mammalian two-hybrid (M2H) system was established that allowed analysis of specific protein interactions quantitatively. Interactions that were recapitulated based on this M2H system are MyD88 (TIR) - MyD88 (TIR), TLR9 (TIR) - MyD88 and TIRAP - MyD88 (Fig.4B). Two of the compounds with characteristic phenotypes were chosen for more detailed analysis, (i) the MPP-like TSI-13-57, which showed Pan-TLR inhibitory activity accompanied by stabilization of MyD88 in CETSA (Fig.3E-H) and (ii) TSI-13-48, which displayed TLR9-selective activity (Fig. 4A). Consistent with previous data, TSI-13-57 inhibited homodimerization of the TIR-domain of MyD88, but showed little activity against interaction between TLR9 and MyD88, and no activity against interaction between TIRAP and MyD88 (Fig.4B). In marked contrast, TSI-13-48 did not show measurable activity against homodimerization of MyD88 TIR (or interaction between TIRAP and MyD88), but inhibited interaction between TLR9 (TIR) and MyD88 (Fig.4B). Of note, the IC 50 values obtained in this M2H system matched closely those obtained during physiological TLR activation of macrophages, both with respect to inhibition of MyD88- dimerization by TSI-13-57 and inhibition of TLR9 (TIR) - MyD88 interaction by TSI-13-48 (Fig.4A, B). As such, modifications of the MPP scaffold can be used to generate selectivity against different TLRs, which is most likely explained by the property of the compounds to block specific, yet different TIR-domain interactions.

[00360] Reagents and plasmids. CpG-DNA refers to the phosphothioate backbone containing oligonucleotide 1668 (TCCATGACGTTCCTGATGCT) (TIB Molbiol). Other agonists used were LPS (Escherichia coli 0127:B8) (Sigma-Aldrich), coumermycin A1 (Sigma- Aldrich), R848 (GLSynthesis), tripalmitoyl cysteinyl lipopeptide (Pam3Cys) (EMC

Microcollections) and Curdlan (InvivoGen). Antibodies were sourced as follows: anti-FLAG (M2 [soluble and bead immobilized] and anti-ȕACTIN were from Sigma-Aldrich, anti-MyD88, anti-IkBĮ, anti-P-p38, anti-P-JNK, anti-P-ERK, anti-p38 and anti-Myc-Tag were from Cell Signaling Technology, anti-HA was from Roche, secondary antibodies conjugated to horseradish peroxidase were from GE Healthcare Life Sciences. Chemiluminescent substrate was from Thermo Scientific. ELISA kits were from eBiosciences (TNF-Į). Luciferase assay system was from Promega. Alamar blue assay system and Lipofectamine 2000 was from Invitrogen. Full length human ERĮ and ERȕ were obtained from PanVera/Invitrogen (Carlsbad, CA); tritiated estradiol was obtained from Perkin-Elmer (Waltham, MA).

[00361] Expression plasmids were established by conventional molecular biology techniques and verified by DNA sequencing. Epitope tags used consisted in tandem triple tags (HA, FLAG) or single tag (Myc) which were fused N-terminal to the cDNA of full length MyD88, the MyD88 death domain (aa 2-109) or the MyD88 TIR-domain (aa 157-296). FLAG-tagged TIRAP-Gyrase B was expressed as fusion protein consisting in triple-FLAG-tagged, full-length TIRAP and a C- terminal Gyrase B moiety using a lentiviral vector containing a PGK1-promoter. FLAG-tagged MyD88-GyrB was expressed as fusion protein consisting in triple-FLAG-tagged, full-length MyD88 and a C-terminal Gyrase B moiety using a MSCV-based retroviral vector (MSCV-puro (Clontech)). FLAG-tagged TRAF6-GyrB was expressed as fusion protein consisting in a triple- FLAG-tagged, N-terminal part of TRAF6 (aa 2-351) and a C-terminal Gyrase B moiety using a pcDNA3-based vector with EF1Į promoter.

[00362] Plasmids used for M2H assays were based on the Checkmate mammalian two-hybrid system (Promega). The TIR domain of MyD88 (aa 158-296) was cloned into the Gal4-vector (pBIND) and VP16-vector (pACT), full length MyD88 was cloned into pACT, the TIR domain of TLR9 (aa 867-1032) was cloned into pBind, and TIRAP (aa 1-241) was cloned into pBind.

[00363] Reporter cell lines. To establish NF-kB-responsive luciferase reporter cells lines, HEK293T cells were transduced with a lentiviral vector containing a luciferase reporter gene under control of three NF-kB-binding sites. Single cell clones of transduced cells were established by limiting dilution, and inducible NF-kB activity was confirmed by transfection with various TLR adaptor proteins. Stable reporter cell lines expressing GyrB-fusion proteins were established by viral transduction (MyD88-GyrB, TIRAP-GyrB) or lipofectamine transfection (TRAF6-GyrB), followed by antibiotic selection. Stably growing cells were cloned by limiting dilution, and clones that showed CM-mediated NF-kB activation were selected for further experiments.

[00364] Cell culture and retrovirus generation. HEK293T cells were maintained in growth medium containing Phenol red (DMEM (Life Technologies), supplemented with 10% (v/v) FCS (Hyclone), 50 mM 2-mercaptoethanol, antibiotics (penicillin G (100 IU/mL) and streptomycin sulfate (100 IU/mL)) and pyruvate (1 mM)). RAW264.7 cells were cultured in RPMI 1640 (Life Technologies), supplemented with 10% (v/v) FCS (Hyclone), 50 mM 2-mercaptoethanol and antibiotics (penicillin G (100 IU/mL) and streptomycin sulfate (100 IU/mL)), as described (Hacker et al., Nature 439, 204-207 (2006); Hacker et al., The EMBO journal 18, 6973-6982 (1999)). Bone marrow (BM)-derived macrophages (BMM) were generated by cultivating unfractionated BM cells (obtained from female C57BL/6 mice or ERĮ-deficient mice (Esr1 tm1Ksk ) , The Jackson Laboratories) and corresponding wildtype control mice, as indicated in the figure descriptions) for 6 days in DMEM (Invitrogen), supplemented with 10% (v/v) FCS (Hyclone), 50 mM 2-mercaptoethanol, antibiotics (penicillin G (100 IU/mL) and streptomycin sulfate (100 IU/mL); Invitrogen) and 30% L-cell–conditioned medium as described (Redecke et al., Nature methods 10, 795-803 (2013)).

[00365] For testing of compounds, RAW264.7 cells were seeded in complete RPMI 1640 (without phenol red) medium in 96 well plates at cell a density of 50,000 per well at least 12 hours before stimulation. Cells were treated with compounds or DMSO for 30 minutes followed by stimulation with various physiological TLR agonists (CpG DNA (1 PM), R848 (300 nM), Pam3Cys (100 ng/mL), LPS (10 ng/mL)) or Curdlan (100 Pg/mL). TNFD levels were determined in cell culture supernatants 6 hours post stimulation by ELISA, and cell viability was analyzed by the Alamar blue assay system (Invitrogen).

[00366] Replication-deficient lentivirus and murine stem cell virus (MSCV) was generated based on Lipofectamine 2000 (Invitrogen)-based transient transfection of HEK293T cells using a four-plasmid system that was generously provided by Dr. Inder Verma (for lentivirus), or an ecotropic, MSCV-based two-plasmid system. Cas9-mediated deletion of MyD88 was done by lentiviral delivery of MyD88-specific sgRNA (genomic target sequence:

GTTCTTGAACGTGCGGACAC) that was expressed by the lentiviral vector LentiCrisprV2 provided through Addgene (Sanjana et al., Nature methods 11, 783-784 (2014)). Transduced cells were selected with puromycin (10 mg/mL) and used as polyclonal cell population.

[00367] Estrogen Receptor Binding Assay. Competitive radiometric binding assays were performed on 96-well microtiter filter plates (Millipore), using full length human estrogen receptor Į and ȕ, with tritiated estradiol as tracer, as previously described (Carlson et al., Biochemistry 36, 14897-14905 (1997)). After incubation on ice for 18-24 hours, ERĮ-bound tracer was absorbed onto hydroxyapatite (BioRad), washed with buffer, and measured by scintillation counting. The affinities are expressed as relative binding affinity (RBA) values, where the RBA of estradiol is 100. RBA values are the average ±SD of 2-3 determinations.

[00368] Cytotoxicity studies. BJ (human foreskin fibroblasts), HEK293 (human embryonic kidney cells), HepG2 (human hepatocellular carcinoma cells), and Raji (human lymphoblast cells (Burkitt’s lymphoma)) cell lines were purchased from the American Type Culture

Collection (ATCC, Manassas, VA) and were cultured according to recommendations. Cell culture media were purchased from ATCC. Cells were routinely tested for mycoplasma contamination using the MycoAlert Mycoplasma Detection Kit (Lonza). Exponentially growing cells were plated in 384-well white custom assay plates (Corning) and incubated overnight at 37 ºC in a humidified 5% CO 2 incubator. DMSO inhibitor stock solutions were added the following day to a top final concentration of 25 mM of 0.25% DMSO and then diluted 1/3 for a total of ten testing concentrations. Cytotoxicity was determined following a 72 h incubation using Cell Titer Glo Reagent (Promega), according to the manufacturer’s recommendation. Luminescence was measured on an Envision plate reader (Perkin Elmer). [00369] Test results for TLR activity, cytotoxicity and estrogen receptor activity are shown in Tables 1A-20.

Table 2

Table 6

Table 8

Table 9

Table 12

Table 14

Table 15

[00370] In vivo LPS challenge model. Female C57BL/6J mice (Jackson Laboratory) were treated i.p. with TSI-13-57 formulated in 5% NMP, 5% Solutol HS 15, 90% normal saline or vehicle control.60 minutes later, the mice were i.p. administered with 2500 ng/kg body weight LPS in PBS.90 minutes later, mice were bled and plasma TNFĮ levels were analyzed by ELISA (eBioscience). [00371] To investigate if TSI-13-57 protected from LPS-mediated TNFĮ release, mice were treated with two doses of TSI-13-57 (100 mg/kg and 200 mg/kg bodyweight), followed by LPS administration and analysis of TNFĮ levels in the serum 90 min after challenge. While 100 mg/kg led to significant reduction, 200 mg/kg almost completely blocked LPS-induced TNFĮ release (Fig.5A). In vivo inhibitory activity correlated well with plasma levels of TSI-13-57 and its known in vitro IC50 of 6.73 mM for LPS-induced TNFĮ release from macrophages (Fig.4A). The data provide proof of principle that MPP analogs with TLR-inhibitory activity can be used to counteract TLR-induced effector functions in vivo.

[00372] In vitro ADMET. Solubility assays were carried out on a Biomek FX lab automation workstation (Beckman Coulter, Inc., Fullerton, CA) using PSOL Evolution software (pION Inc.). 10 PL of 10 mM compound stock (in DMSO) were added to 190 PL 1-propanol to make a reference stock plate.5 PL from this reference stock plate were mixed with 70 PL 1-propanol and 75 PL citrate phosphate-buffered saline (PBS; isotonic) to make the reference plate, and the UV spectrum (250-500 nm) of the reference plate was read.6 PL of 10 mM test compound stock were added to 594 PL buffer in a 96-well storage plate and mixed. The storage plate was sealed and incubated at RT for 18 hours. The suspension was then filtered through a 96-well filter plate (pION Inc.).75 PL of filtrate were mixed with 75 PL 1-propanol to make the sample plate, and the UV spectrum of the sample plate was read. Calculations were carried out with PSOL

Evolution software based on the area under the curve (AUC) of the UV spectrum of the sample and reference plates. All compounds were tested in triplicate.

[00373] A parallel artificial membrane permeability assay (PAMPA) was conducted with a Biomek FX lab automation workstation (Beckman Coulter) and PAMPA Evolution 96

Command software (pION Inc.).3 PL of 10 PM test compound stock in DMSO were mixed with 597 PL of citrate PBS (isotonic) to make a diluted test compound.150 PL of diluted test compound were transferred to a UV plate (pION Inc.), and the UV spectrum of this reference plate was read. The membrane, on a pre-loaded PAMPA Sandwich (pION Inc.), was painted with 4 PL of GIT lipid (pION Inc.). The acceptor chamber was then filled with 200 PL of ASB (acceptor solution buffer, pION Inc.), and the donor chamber was filled with 180 PL of diluted test compound. The PAMPA Sandwich was assembled, placed on the Gut-box, and stirred for 30 minutes. The aqueous boundary layer was set to 40 Pm for stirring. The UV spectrum (250-500 nm) of the donor and the acceptor were read. The permeability coefficient was calculated using PAMPA Evolution 96 Command software based on the AUC of the reference plate, the donor plate, and the acceptor plate. All compounds were tested in triplicate.

[00374] Mouse liver microsomes (0.73 mL) were mixed with EDTA solution (0.06 mL, 0.5M in water) and potassium phosphate buffer (22.31 mL, 0.1M, pH 7.4, 37°C) to make 23.1 mL of liver microsome solution (20 mg/mL liver microsome protein).10 mM stocks of compound in DMSO were diluted with DMSO and acetonitrile to three different intermediate concentrations: high (2 mM), medium (0.4 mM) and low (0.08 mM) concentration in DMSO:acetonitrile (1:4, v:v).10 mM stocks of controls in DMSO (diphenhydramine HCl, verapamil HCl, and

Ketoprofen) are diluted to 0.4 mM concentration in DMSO:acetonitrile (1:4, v:v). Each diluted compound stock (37.83 mL) was added to an aliquot of the liver microsomal solution (3 ml) and vortexed. The resulting solution was added to each of 3 wells of a master assay plate (pION Inc., MA, #110323). Each plate holds triplicate samples of two controls (0.4 mM) and two

compounds (2 mM, 0.4 mM and 0.08 mM) in mouse microsomes. Aliquots of each well of the plate (175 mL of each well) were transferred from the master plate into 5 assay plates. For 0-hour time point, pre-cooled (4 ºC) internal standard (437.5 mL, 2 mM warfarin in methanol) was added to the first plate before the reaction starts. NADPH regenerating system solution A (6.05 mL, Fisher Scientific, #NC9255727) was combined with NADPH regenerating system solution B (1.21 mL, Fisher Scientific, #NC9016235) in potassium phosphate buffer (15.84 mL, 0.1 M, pH 7.4, 37°C). The resulting NADPH solution (43.75 mL) was added to each well of all the 96-well assay plates and mixed with pipette briefly making the final protein and compound

concentrations respectively: 0.5 mg/mL, high (20 PM), medium (4 PM), and low (0.08 PM). The plates are sealed, and all plates except the 0-hr plate were incubated at 37 ºC, shaken at a speed of 60 rpm. A single assay plate was tested at each time point: 0.5 hr, 1 hr, 2 hr, and 4 hr. At each time point, 437.5 mL of pre-cooled internal standard was added each well of the plate to quench the reaction. The quenched plate was then centrifuged (model 5810R, Eppendorf, Westbury, NY) at 4000 rpm for 15 minutes.150 mL supernatant was transferred to a 96-well plate and analyzed by UPLC-MS (Waters Inc., Milford, MA). The compounds and internal standard were detected by selected ion recording (SIR). The amount of material was measured as a ratio of peak area to the internal standard and graphed. Using the slope from the most linear portion of this curve, the degradation rate constant is calculated. The rate constant was then used to calculate the compounds half-life is in plasma. Intrinsic clearance was calculated as CLint’ = (0.693 / (t 1/2 )) * (1/ microsomal concentration in the reaction solution) * (45 mg microsome/gram liver) * (gram liver/kg b.w.), where microsomal concentration in the reaction solution is 0.5 mg/mL, and gram liver/kg b.w. of mouse is 52. Table 21. In vitro early ADMET analysis of TSI-13-57. CLint, intrinsic clearance

[00375] In vivo pharmacokinetic study. Female C57BL/6 mice with average weight of 19 grams were purchased from Charles River Laboratories (Wilmington). Food and water were provided ad libitum.15 mice were divided into three dosage groups: 0, 10 and 20 mg/kg. For each mouse, 0.1 mL of compound suspension in formulation (0.5% CMC, 0.4% Tween 80) was given by intra-peritoneal (i.p.) injection.0.1 mL blood was collected retro-orbitally from a different mouse within each dosage group at 5 minutes, 15 minutes, 30 minutes, 1 hour, 4 hours, and 24 hours. Animals were euthanized via cardiac puncture at 48 hours post injection. Blood samples were treated with 10 mL of EDTA sodium solution to prevent coagulation. Blood was kept on ice and centrifuged for 3 minutes at 13,000 rpm in a desktop centrifuge to collect plasma. 25 mL plasma samples were combined with 75 mL internal standard (2 mM warfarin) in acetonitrile in a 96 well plate and centrifuged at 4000 rpm for 20 minutes at 4 °C. The supernatant (40 mL) was collected and mixed with 2 part of Milli-Q water (EMD Millipore) and centrifuged again at 3000 rpm for 20 minutes at 4 °C. Plasma concentration was determined with partially validated LC/MS-MS assay with MRM detection (AB Sciex). The assay limit of quantification (LLOQ) was 1.5 nM in plasma.

[00376] The processed plasma concentration-time data were analyzed using non- compartmental analysis (NCA) in WinNonlin 6.0 with the Plasma (200-202) model type; all standard NCA parameters were estimated via default software settings, using predicted parameter estimates. If more than two thirds of the observed concentrations were below Lower Limit of Quantification (LLOQ), the mean concentration was treated missing. The Area Under the Concentration-Time Curve (AUC) was calculated with the linear trapezoidal, linear interpolation rule using mean concentrations and nominal times. The terminal elimination rate (Lambda_z) and half-life (HL_Lambda_z) was determined using the default“Best Fit” method. The predicted AUC from the last time point to infinity (AUCINF_pred) was calculated as AUClast plus Clast(pred)/Lambda_z.

[00377] In vivo pharmacokinetics (PK) experiments were performed for TSI-13-57. The maximum serum concentration (C max ) showed a dose-dependent increase from 669 to 1260 nM for 10 and 20 mg/kg of intra-peritoneally (i.p.) administered compound, respectively. The plasma half-life was 9.8 hours (10 mg/kg) and 9.0 hours (20 mg/kg) (Table 22). The area under the curve (AUC) within 24 hours displayed a dose-dependent, non-linear mode, where 43.2 mM*hr was achieved at 20 mg/kg (Table 22). During the acute observation phase after dosing, no adverse reactions or compound-related side effects were observed, and all animals maintained normal feeding behavior and body weight during the 48 hour period following i.p. administration at all doses. No significant changes in hematological parameters, clinical chemistry, or gross organ anatomy were observed at the terminal point of the study, corresponding to the lack of apparent toxicity against various cell lines in vitro (Table 21). As such, TSI-13-57 exhibited overall favorable, drug-like properties without overt liabilities. Table 22. In vivo pharmacokinetics (PK) of TSI-13-57

purification and quantitative mass spectrometry. For stable isotope labeling by amino acids in cell culture (SILAC), RAW264.7 cells expressing stably a FLAG- tagged form of MyD88-GyrB were cultured in arginine- and lysine-free RPMI (Invitrogen) supplemented with 10% dialyzed FBS (Invitrogen), penicillin-streptomycin, and either L- arginine and L-lysine (light), L-arginine-HCl (13C6; CLM-2265 [R6]) and L-lysine-2HCl (4,4,5,5 D4; DLM-2640 [K4]) (medium) or L-arginine-HCl (13C6, 15N4; CLM-539 [R10]) and L-lysine-2HCl (13C6, 15N2; DLM-291 [K8]) (heavy) (Cambridge Isotope Labs)(73). For complete incorporation of labeled amino acids, cells were passaged three times in SILAC medium over a period of 5 days. The labeled cells were treated with 10 mM MPP for 20 minutes (heavy), followed by stimulation with 1 mM CpG-DNA for 60 minutes (medium and heavy). The medium was replaced by ice-cold PBS and cells were collected by cell scraping and

centrifugation. Cell pellets were incubated with lysis buffer (LB; 20 mM Hepes/NaOH (pH 7.5), 1.5 mM MgCl2, 150 mM NaCl, 1 mM EDTA, 10% glycerol, 10 mM ȕ-glycerophosphate, 5 mM 4-nitrophenyl-phosphate, 10 mM sodium fluoride, complete protease inhibitors (Roche)) supplemented with 0.5 % NP-40 for 20 min. Samples were cleared by centrifugation and loaded five times over anti-FLAG-bead-containing columns. Unbound proteins were removed by washing column with LB plus 0.1% NP-40, and proteins were eluted at pH 3.5 in water supplemented with 100 mM glycine, 50 mM NaCl, 0.1% NP-40, and Roche complete protease inhibitors. The proteins were concentrated by trichloroacetic acid precipitation and dissolved in SDS PAGE loading buffer (Bio-Rad). The dissolved proteins were combined, followed by separation on a 10 % Bis-Tris gel (Bio-Rad) and staining with SYPRO Ruby protein stain (Invitrogen). The entire lane was cut into individual bands and analyzed by LC-tandem MS (LC- MS/MS) using a nanoAcquity UPLC (Waters) coupled to an Orbitrap ELITE high resolution mass spectrometer (Thermo Fisher).

[00379] Sample preparation and ESI LC. The protein gel bands were reduced with DTT and alkylated with iodoacetamide and then digested overnight with trypsin (Promega). The digest was introduced into the instrument via on-line chromatography using reversed-phase (C18) ultra-high-pressure LC on a nanoACQUITY UPLC (Waters). The column used was a Waters BEHC18 with an inner diameter of 75 mm and a bed length of 10 cm. The particle size was 1.7 mm. Tryptic peptides were gradient eluted over a gradient (0 to 70% B for 60 minutes and 70 to 100% B for 10 minutes, where B was 70% [vol/vol] acetonitrile, 0.2% formic acid) using a flow rate of 250 nL/minute into the high resolution Orbitrap ELITE through a noncoated spray needle with voltage applied to the liquid junction.

[00380] MS/MS analysis with LTQ XL (Thermo Fisher) and database analysis. Data- dependent scanning was incorporated to select the 20 most abundant ions (one microscan per spectrum; precursor isolation width, 2.0 Da; 35% collision energy; 10-ms ion activation; 15-s dynamic exclusion duration; 5-s repeat duration; and a repeat count of 1 from a full-scan mass spectrum at 60,000 resolution for fragmentation by collision-activated dissociation). Database searches were performed using RAW files in combination with Andromeda search engine that is part of the MaxQuant software (version 1.1.1.32) developed at the Max Planck Institute (Cox et al., Nature biotechnology 26, 1367-1372 (2008)). The SwissProt 2012_08 ((537,505 sequences; 190,795,142 residues); Taxonomy: Mus musculus (16,605 sequences)) database was used for peptide and protein identification. MaxQuant was also used to quantitate peptides and proteins and to provide ratios generated in Excel format. Protein assignments were made on the basis of both MS and MS/MS spectra, whereas peptide quantitation was based solely on MS data. The following residue modifications were allowed in the search: carbamidomethylation on cysteine (fixed modification), oxidation on methionine (variable modification), label:13C(6) on arginine, label:13C(10) on arginine, label:13C(4) on lysine, and label:13C(8) on lysine. The MS1 mass tolerance was set to 15 ppm and the MS/MS tolerance was set to 0.5 Da and protein FDR was set to 0.01.The identifications from the automated search were verified by manual inspection of the raw data.

[00381] Immunoprecipitation. Immuno-precipitation (IP) studies were performed in HEK293T cells that were transfected using Lipofectamine (Life Technlogies) with epitope- tagged forms of MyD88. MPP or DMSO was added 7 hours after transfection.20 hours after transfection, cells were lyzed for 20 minutes at 4 °C in lysis buffer (20 mM Hepes/NaOH (pH 7.5), 1.5 mM MgCl2, 150 mM NaCl, 1 mM EDTA, 0.5% Nonidet P-40 and 10% glycerol) supplemented with complete protease inhibitors (Roche Applied Science). After clearance of lysates by centrifugation (10 min, 20,817 g, 4 °C), lysates were subjected to immunoprecipitation (IP) using anti-FLAG M2 resin (Sigma-Aldrich) for 1 hour at 4qC. IP samples and total cell lysates were analyzed by immuno-blotting.

[00382] Cellular thermal shift assay (CETSA). CESTA was performed as described (Jafari et al., H. Nature protocols 9, 2100-2122 (2014)). Briefly, HEK293T cells were treated with TSI- 13-57 or DMSO as control at 37°C with 5% CO2 for 60 minutes in cell culture dishes (TPP). Cells were resuspended in medium and spun down at 240 g for 5 minutes at room temperature. Medium was carefully removed and cells were resuspended in phosphate buffered saline (PBS). Cells were spun down again at 240 x g for 5 minutes at room temperature. PBS was carefully removed again and the cell pellet was resuspended in PBS supplemented with protease inhibitors to obtain a cell density of 8 × 10 6 cells/mL. The cell suspension (0.8 million in 100 mL volume) was transferred to multiple tubes in a real-time PCR plate (Applied Biosystems). The PCR plate was loaded to the heating block of a PTC-200 Gradient Thermocycler (MJ Research) at 25°C. Samples were heated to their desired temperatures in parallel by applying a temperature gradient covering a range between 40°C and 64°C. The respective temperatures were maintained for 3 min before the samples were cooled and maintained at 25°C for 3 minutes. Next, the tubes were immediately shock-frozen in liquid nitrogen. Cells were lysed by two alternating thaw-freeze cycles in a heating block (25°C) and in liquid nitrogen, respectively. The resulting suspensions were centrifuged at 20,000 g for 20 minutes at 4°C. For the following steps the lysates were kept on ice. Supernatants of each sample were carefully transferred to reaction tubes without touching or disturbing the pellets and were analyzed by sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE) followed by Immuno-blot analysis.

[00383] Mammalian two-hybrid assay (M2H assay). HEK293T cells were transiently transfected with bait- and prey plasmids in 96 well format using Lipofectamine (2000), along with a Gal4-driven firefly luciferase reporter plasmid (pGL5-luc, Promega) and Renilla luciferase control vector (Promega). DMSO or compound was added to cells 7 hours post transfection. Cells were harvested 13 hours later, and luciferase activity was determined using the dual luciferase kit (Promega). Firefly luciferase activity values were normalized to Renilla luciferase activity.

[00384] Statistics. Statistical analyses were performed using GraphPad Prism 7 software. Pairwise comparisons were analyzed with the Mann-Whitney U test or Paired t test (two-tailed). Multiple comparisons were analyzed by one way or two way analysis of variance (ANOVA) followed by post-tests as specified in Figure legends. [00385] For reasons of completeness, various aspects of the invention are set out in the following numbered clauses:

[00386] Clause 1. A compound of formula (I), or a pharmaceutically acceptable salt thereof,

wherein:

G 1 is–NR 1 R 2 ,–OH,–OC 1-4 alkyl, a C 3-8 cycloalkyl, a 4- to 12-membered heterocyclyl containing 1-3 heteroatoms independently selected from N, O, and S, or a 5- to 12-membered heteroaryl containing 1-3 heteroatoms independently selected from N, O, and S, wherein the cycloalkyl, the heterocyclyl, and the heteroaryl are optionally substituted with 1-4 substituents independently selected from halo, C1-4alkyl, C1-4haloalkyl,–OH,–OC1-4alkyl, and oxo;

R 1 and R 2 are each independently hydrogen or C 1-4 alkyl;

L 1 is–C 1-5 alkylene–,–C 1-4 alkylene–O–, or–C(O)–CH=CH–, wherein the–C 1-5 alkylene– and the –C 1-4 alkylene–O– are optionally substituted with 1-2 halogens or one hydroxyl; or L 1 is , wherein the C1-4alkylene is bonded to G 1 and the imidazole is fused at the meta and para positions on the phenyl relative to G 2 ;

G 2 is selected from (i) to (xiii)

X 1 is O or S;

X 2 is O, S, NH, or NC1-4alkyl;

R 5 is C1-4alkyl or

R 3 , R 7 , R 13 , R 17 , R 19 , R 21 , R 27 , R 29 , and R 33 are each independently selected from hydrogen and C1-4alkyl;

R 11 and R 31 are each independently selected from hydrogen, C1-4alkyl, and phenyl optionally substituted with 1-3 substituents independently selected from halo, C 1-4 alkyl, C 1-4 haloalkyl,– OH, and–OC 1-4 alkyl;

R 4a , R 4b , R 4c , R 5a , R 6 , R 8 , R 9 , R 10 , R 12 , R 14 , R 15 , R 16 , R 18 , R 20 , R 22 , R 23 , R 24 , R 25 , R 26 , R 28 , R 30 , and R 32 , at each occurrence, are independently halo, nitro, cyano, C 1-4 alkyl, C 1-4 haloalkyl,–OH,– OC1-4alkyl,–OC1-4haloalkyl,–NH2,–NH(C1-4alkyl),–N(C1 -4alkyl)(C1-4alkyl),–NHC(O)C1- 4alkyl,–N(C1-4alkyl)C(O)C1-4alkyl,–NHC(O)OC1-4alkyl,–N (C1-4alkyl)C(O)OC1-4alkyl,– C(O)OC1-4alkyl,–C(O)OH,–C(O)NH2,–C(O)NH(C1-4alkyl), or–C(O)N(C1-4alkyl)(C1- 4alkyl), and optionally two R 4a , R 4b , R 5a , R 6 , R 8 , R 9 , R 10 , R 12 , R 14 , R 15 , R 16 , R 18 , R 20 , R 21 , R 22 , R 24 , R 25 , R 26 , R 28 , R 30 , or R 32 , together with the atoms to which they are attached form a

fused ring

n1 and n2 are independently 0, 1, 2, 3, 4, or 5; and

n3 and n4 are independently 0, 1, 2, or 3.

[00387] Clause 2. The compound of clause 1, or a pharmaceutically acceptable salt thereof, wherein the compound of formula (I) has formula (IA) .

[00388] Clause 3. The compound of clause 1 or 2, or a pharmaceutically acceptable salt thereof, wherein

G 1 is–NR 1 R 2 , a 4- to 12-membered heterocyclyl containing 1-3 heteroatoms independently

selected from N, O, and S, or a 5- to 12-membered heteroaryl containing 1-3 heteroatoms independently selected from N, O, and S, wherein the heterocyclyl and the heteroaryl are optionally substituted with 1-4 substituents independently selected from halo, C 1-4 alkyl, C 1- 4haloalkyl,–OH,–OC1-4alkyl, and oxo; and

L 1 is–C1-5alkylene–or–C1-4alkylene–O–.

[00389] Clause 4. The compound of any of clauses 1-3, or a pharmaceutically acceptable salt thereof, wherein

G 1 is –NR 1 R 2 ;

a 4- to 8-membered monocyclic heterocyclyl containing a first nitrogen atom and optionally an additional heteroatom selected from N, O, and S, the heterocyclyl connecting to L 1 at the first nitrogen atom and optionally containing a C 1-3 alkylene bridge between two non-adjacent ring atoms and/or a double bond, the heterocyclyl being optionally substituted with 1-4 substituents independently selected from halo, C1-4alkyl, C1-4haloalkyl,–OH,–OC1-4alkyl, and oxo; or

a 5- to 6-membered monocyclic heteroaryl containing 1-3 nitrogen atoms, the heteroaryl being optionally substituted with 1-4 substituents independently selected from halo, C 1- 4 alkyl, C 1-4 haloalkyl,–OH, and–OC 1-4 alkyl; and

L 1 is–C 2-4 alkylene– or–C 2-3 alkylene–O–.

[00390] Clause 5. The compound of clause 4, or a pharmaceutically acceptable salt thereof, wherein

G 1 is–N(CH3)2,–N(CH2CH3)2, pyrrolidin-1-yl, piperidin-1-yl, piperazin-1-yl, azepin-1-yl,

pyrazol-1-yl, imidazol-1-yl, or triazol-1-yl. [00391] Clause 6. The compound of any of clauses 1-5, or a pharmaceutically acceptable salt thereof, wherein G 2 is formula (i)

.

[00392] Clause 7. The compound of clause 6, or a pharmaceutically acceptable salt thereof, wherein .

[00393] Clause 8. The compound of clause 6, or a pharmaceutically acceptable salt thereof, wherein

R 5 is C1-4alkyl.

[00394] Clause 9. The compound of clause 6 or 7, or a pharmaceutically acceptable salt thereof, wherein

ompound of clause 9, or a pharmaceutically acceptable salt thereof, wherein

R 4a , at each occurrence, is independently halo, cyano, C1-4alkyl, C1-4haloalkyl,–OH,–OC1-4alkyl, –OC1-4haloalkyl,–NH2,–NH(C1-4alkyl),–N(C1-4alkyl)(C1 -4alkyl),–NHC(O)C1-4alkyl,–N(C1- 4alkyl)C(O)C 1-4 alkyl,–C(O)OC 1-4 alkyl,–C(O)NH 2 ,–C(O)NH(C 1-4 alkyl), or–C(O)N(C 1- 4 alkyl)(C 1-4 alkyl), and optionally two R 4a , together with the atoms to which they are attached,

form a fused ring ;

R 5a is halo, nitro, cyano, C 1-4 alkyl, C 1-4 haloalkyl,–OH,–OC 1-4 alkyl,–OC 1-4 haloalkyl,–NH 2 ,– NH(C1-4alkyl),–N(C1-4alkyl)(C1-4alkyl),–NHC(O)C1-4alkyl, –N(C1-4alkyl)C(O)C1-4alkyl,– NHC(O)OC1-4alkyl, or–N(C1-4alkyl)C(O)OC1-4alkyl;

n1 is 0, 1, 2, or 3; and

n2 is 0 or 1.

[00396] Clause 11. The compound of clause 6, or a pharmaceutically acceptable salt thereof, wherein

R 5 is

[00397] Clause 12. The compound of any of clauses 6-11, or a pharmaceutically salt thereof, wherein R 3 is hydrogen.

[00398] Clause 13. The compound of any of clauses 6-11, or pharmaceutically acceptable salt thereof, provided that the compound is not 4,4'-(4-ethyl-3-(4-(2-(piperidin-1-yl)ethoxy)phenyl)- 1H-pyrazole-1,5-diyl)diphenol, or a salt thereof.

[00399] Clause 14. The compound of any of clauses 1-5, or a pharmaceutically acceptable salt thereof, wherein G 2 is formula (ii)

.

[00400] Clause 15. The compound of clause 14, or a pharmaceutically acceptable salt thereof, wherein n1 and n2 are each independently 0, 1, 2, or 3.

[00401] Clause 16. The compound of clause 15, or a pharmaceutically acceptable salt thereof, wherein G 2 is formula (iia)

.

[00402] Clause 17. The compound of any of clauses 1-5, or a pharmaceutically acceptable salt thereof, wherein G 2 is formula (iii)

.

[00403] Clause 18. The compound of clause 17, or a pharmaceutically acceptable salt thereof, wherein n1 and n2 are each independently 0, 1, 2, or 3.

[00404] Clause 19 The compound of clause 18, or a pharmaceutically acceptable salt thereof, wherein n1 and n2 are each independently 0 or 1.

[00405] Clause 20. The compound of any of clauses 17-19, or a pharmaceutically acceptable salt thereof, wherein

R 9 and R 10 , at each occurrence, are independently halo, nitro, cyano, C 1-4 alkyl, C 1-4 haloalkyl,– OC 1-4 alkyl,–OC 1-4 haloalkyl,–NH 2 ,–NH(C 1-4 alkyl),–N(C 1-4 alkyl)(C 1-4 alkyl),–NHC(O)C 1-

108 4 alkyl,–N(C 1-4 alkyl)C(O)C 1-4 alkyl,–NHC(O)OC 1-4 alkyl,–N(C 1-4 alkyl)C(O)OC 1-4 alkyl,– C(O)OC 1-4 alkyl,–C(O)OH,–C(O)NH 2 ,–C(O)NH(C 1-4 alkyl), or–C(O)N(C 1-4 alkyl)(C 1-4 alkyl), and optionally two R 9 or R 10 , together with the atoms to which they are attached form a fused

ring .

[00406] Clause 21. The compound of any of clauses 17-19, or a pharmaceutically acceptable salt thereof, wherein

R 9 and R 10 , at each occurrence, are independently halo, C1-4alkyl,–OH, or–OC1-4alkyl.

[00407] Clause 22. The compound of clause 21, or a pharmaceutically acceptable salt thereof, wherein:

R 9 , at each occurrence, is independently halo, C1-4alkyl, or–OC1-4alkyl; and

R 10 , at each occurrence, is independently halo, C 1-4 alkyl,–OH, or–OC 1-4 alkyl.

[00408] Clause 23. The compound of clause 22, or a pharmaceutically acceptable salt thereof, wherein R 10 , at each occurrence, is independently halo, C 1-4 alkyl, or–OC 1-4 alkyl.

[00409] Clause 24. The compound of any of clauses 1-5 or 20-23, or a pharmaceutically acceptable salt thereof, wherein G 2 is formula (iiia)

;

and n1 and n2 are each independently 0 or 1. [00410] Clause 25. The compound of any of clauses 17-24, or a pharmaceutically acceptable salt thereof, wherein R 11 is hydrogen.

[00411] Clause 26. The compound of any of clauses 1-5, or a pharmaceutically acceptable salt thereof, wherein G 2 is formula (iv)

.

[00412] Clause 27. The compound of clause 26, or a pharmaceutically acceptable salt thereof, wherein n1 and n2 are each independently 0, 1, 2, or 3.

[00413] Clause 28. The compound of clause 27, or a pharmaceutically acceptable salt thereof, wherein n1 and n2 are each independently 0 or 1.

[00414] Clause 29. The compound of any of clauses 26-28, or a pharmaceutically acceptable salt thereof, wherein

R 12 and R 14 , at each occurrence, are independently halo, nitro, cyano, C 1-4 alkyl, C 1-4 haloalkyl,– OC 1-4 alkyl,–OC 1-4 haloalkyl,–NH 2 ,–NH(C 1-4 alkyl),–N(C 1-4 alkyl)(C 1-4 alkyl),–NHC(O)C 1- 4 alkyl,–N(C 1-4 alkyl)C(O)C 1-4 alkyl,–NHC(O)OC 1-4 alkyl,–N(C 1-4 alkyl)C(O)OC 1-4 alkyl,– C(O)OC1-4alkyl,–C(O)OH,–C(O)NH2,–C(O)NH(C1-4alkyl), or–C(O)N(C1-4alkyl)(C1-4alkyl), and optionally two R 12 or R 14 , together with the atoms to which they are attached form a fused

ring ;

provided that the compound is not 1-(2-(4-(4-ethyl-3,5-bis(4-methoxyphenyl)-1H-pyrazol-1- yl)phenoxy)ethyl)piperidine, or a salt thereof.

[00415] Clause 30. The compound of any of clauses 26-28, or a pharmaceutically acceptable salt thereof, wherein R 12 and R 14 , at each occurrence, are independently halo,–OH, or–OC 1-4 alkyl.

[00416] Clause 31. The compound of clause 30, or a pharmaceutically acceptable salt thereof, wherein R 12 and R 14 , at each occurrence, are independently halo or–OC1-4alkyl.

[00417] Clause 32. The compound of clause 30 or 31, or a pharmaceutically acceptable salt thereof, wherein the compound is not 1-(2-(4-(4-ethyl-3,5-bis(4-methoxyphenyl)-1H-pyrazol-1- yl)phenoxy)ethyl)piperidine, or a salt thereof.

[00418] Clause 33. The compound of any of clauses 26-32, or a pharmaceutically acceptable salt thereof, wherein G 2 is formula (iva)

and n1 and n2 are each independently 0 or 1.

Clause 34. The compound of any of clauses 1-5, or a pharmaceutically acceptable salt thereof, wherein n1, n2, and n3 are each independently 0, 1, 2, or 3.

[00420] Clause 35. The compound of any of clauses 1-34, or a pharmaceutically acceptable salt thereof, wherein

R 4a , R 4b , R 4c , R 5a , R 6 , R 8 , R 9 , R 10 , R 12 , R 14 , R 15 , R 16 , R 18 , R 20 , R 22 , R 23 , R 24 , R 25 , R 26 , R 28 , R 30 , and R 32 , at each occurrence, are independently halo, cyano, C1-4alkyl,–OH, or–OC1-4alkyl.

[00421] Clause 36. The compound of clause 35, or a pharmaceutically acceptable salt thereof, wherein n1, n2, and n3 are each independently 0 or 1. [00422] Clause 37. The compound of any of clauses 34-36, or a pharmaceutically acceptable salt thereof, wherein G 2 is (via)

.

[00423] Clause 38. The compound of any of clauses 34-36, or a pharmaceutically acceptable salt thereof, wherein G 2 is (viia)

.

[00424] Clause 39. The compound of clause 38, or a pharmaceutically acceptable salt thereof, wherein:

R 22 is halo, cyano, C1-4alkyl, or–OC1-4alkyl;

R 23 is halo, cyano, C1-4alkyl,–OH, or–OC1-4alkyl; and

n1 and n2 are each 1. [00425] Clause 40. The compound of clause 38, or a pharmaceutically acceptable salt thereof, wherein:

R 22 is halo, cyano, C1-4alkyl,–OH, or–OC1-4alkyl;

R 23 is halo, cyano, C1-4alkyl, or–OC1-4alkyl; and

n1 and n2 are each 1.

[00426] Clause 41. The compound of clause 38, or a pharmaceutically acceptable salt thereof, wherein:

R 22 is halo, cyano, C 1-4 alkyl, or–OC 1-4 alkyl;

R 23 is halo, cyano, C 1-4 alkyl, or–OC 1-4 alkyl; and

n1 and n2 are each 1.

[00427] Clause 42. The compound of any of clauses 34-36, or a pharmaceutically acceptable salt thereof, wherein G 2 is (ixa)

.

[00428] Clause 43. The compound of any of clauses 34-36, or a pharmaceutically acceptable salt thereof, wherein G 2 is (xa) or (xiiia)

. [00429] Clause 44. The compound of any of clauses 34-36, or a pharmaceutically acceptable salt thereof, wherein G 2 is (va), (viiia), or (xia)

[00430] Clause 45. The compound of clause 1 selected from the group consisting of

4,4'-(5-(4-(2-(piperidin-1-yl)ethoxy)phenyl)-1H-pyrazole- 1,3-diyl)diphenol;

(E)-3-(4-(1,3-bis(4-hydroxyphenyl)-4-methyl-1H-pyrazol-5- yl)phenyl)-1-(piperidin-1-yl)prop-2- en-1-one;

4,4'-(4-methyl-5-(4-(3-(piperidin-1-yl)propyl)phenyl)-1H- pyrazole-1,3-diyl)diphenol;

4,4'-(5-(4-(3-(piperidin-1-yl)propoxy)phenyl)-1H-pyrazole -1,3-diyl)diphenol;

4,4'-(5-(4-(3-cyclohexylpropoxy)phenyl)-1H-pyrazole-1,3-d iyl)diphenol;

1-(2-(4-(1,3-bis(4-methoxyphenyl)-1H-pyrazol-5-yl)phenoxy )ethyl)piperidine;

1-(2-(4-(1,3-bis(4-methoxyphenyl)-4-methyl-1H-pyrazol-5-y l)phenoxy)ethyl)piperidine;

1-(3-(4-(1,3-bis(4-methoxyphenyl)-4-methyl-1H-pyrazol-5-y l)phenyl)propyl)piperidine;

1-(3-(4-(1,3-bis(4-methoxyphenyl)-1H-pyrazol-5-yl)phenoxy )propyl)piperidine;

1-(2-(3-(1-phenyl-3-(p-tolyl)-1H-pyrazol-5-yl)phenoxy)eth yl)piperidine;

1-(2-(4-(3-(4-methoxyphenyl)-1-phenyl-1H-pyrazol-5-yl)phe noxy)ethyl)piperidine;

4-(1-phenyl-5-(4-(2-(piperidin-1-yl)ethoxy)phenyl)-1H-pyr azol-3-yl)phenol;

1-(2-(4-(1,3-diphenyl-1H-pyrazol-5-yl)phenoxy)ethyl)piper idine;

1-(2-(4-(1-(4-chlorophenyl)-3-(4-methoxyphenyl)-1H-pyrazo l-5-yl)phenoxy)ethyl)piperidine; 4-(1-(4-chlorophenyl)-5-(4-(2-(piperidin-1-yl)ethoxy)phenyl) -1H-pyrazol-3-yl)phenol; 1-(2-(4-(1-phenyl-3-(3,4,5-trimethoxyphenyl)-1H-pyrazol-5-yl )phenoxy)ethyl)piperidine;

4,4'-(4-ethyl-1-(4-(2-(pyrrolidin-1-yl)ethoxy)phenyl)-1H- pyrazole-3,5-diyl)diphenol;

4,4'-(5-ethyl-4-(4-(2-(piperidin-1-yl)ethoxy)phenyl)-1H-p yrazole-1,3-diyl)diphenol;

4,4'-(4-(4-(2-(piperidin-1-yl)ethoxy)phenyl)-5-propyl-1H- pyrazole-1,3-diyl)diphenol;

4,4'-(3-(4-(2-(piperidin-1-yl)ethoxy)phenyl)-1H-pyrazole- 1,5-diyl)diphenol;

4-(1-phenyl-3-(4-(2-(piperidin-1-yl)ethoxy)phenyl)-1H-pyr azol-5-yl)phenol;

4-(5-phenyl-3-(4-(2-(piperidin-1-yl)ethoxy)phenyl)-1H-pyr azol-1-yl)phenol;

3-(1-(4-hydroxyphenyl)-3-(4-(2-(piperidin-1-yl)ethoxy)phe nyl)-1H-pyrazol-5-yl)phenol;

4-(3-(4-(2-(piperidin-1-yl)ethoxy)phenyl)-1-(p-tolyl)-1H- pyrazol-5-yl)phenol;

4-(1-(4-chlorophenyl)-3-(4-(2-(piperidin-1-yl)ethoxy)phen yl)-1H-pyrazol-5-yl)phenol;

2,6-dimethoxy-4-(1-phenyl-3-(4-(2-(piperidin-1-yl)ethoxy) phenyl)-1H-pyrazol-5-yl)phenol; N-(4-(1-(4-hydroxyphenyl)-3-(4-(2-(piperidin-1-yl)ethoxy)phe nyl)-1H-pyrazol-5- yl)phenyl)acetamide;

1-(2-(4-(1,5-bis(4-methoxyphenyl)-1H-pyrazol-3-yl)phenoxy )ethyl)piperidine;

1-(2-(4-(5-(4-methoxyphenyl)-1-phenyl-1H-pyrazol-3-yl)phe noxy)ethyl)piperidine;

1-(2-(4-(1-(3-methoxyphenyl)-5-(4-methoxyphenyl)-1H-pyraz ol-3-yl)phenoxy)ethyl)piperidine; 1-(2-(4-(5-(4-methoxyphenyl)-1-(p-tolyl)-1H-pyrazol-3-yl)phe noxy)ethyl)piperidine;

1-(2-(4-(5-(4-methoxyphenyl)-1-(4-(trifluoromethoxy)pheny l)-1H-pyrazol-3- yl)phenoxy)ethyl)piperidine;

1-(2-(4-(1-(4-chlorophenyl)-5-(4-methoxyphenyl)-1H-pyrazo l-3-yl)phenoxy)ethyl)piperidine; 1-(2-(4-(5-(4-methoxyphenyl)-1-(4-nitrophenyl)-1H-pyrazol-3- yl)phenoxy)ethyl)piperidine; tert-butyl (4-(5-(4-methoxyphenyl)-3-(4-(2-(piperidin-1-yl)ethoxy)pheny l)-1H-pyrazol-1- yl)phenyl)carbamate;

4-(5-(4-methoxyphenyl)-3-(4-(2-(piperidin-1-yl)ethoxy)phe nyl)-1H-pyrazol-1-yl)aniline;

1-(2-(4-(1-(4-methoxyphenyl)-5-phenyl-1H-pyrazol-3-yl)phe noxy)ethyl)piperidine;

1-(2-(4-(5-(2,4-dimethoxyphenyl)-1-(4-methoxyphenyl)-1H-p yrazol-3- yl)phenoxy)ethyl)piperidine;

N-(4-(1-(4-methoxyphenyl)-3-(4-(2-(piperidin-1-yl)ethoxy) phenyl)-1H-pyrazol-5- yl)phenyl)acetamide;

1-(2-(4-(1-(4-methoxyphenyl)-5-(3,4,5-trimethoxyphenyl)-1 H-pyrazol-3- yl)phenoxy)ethyl)piperidine; 1-(2-(4-(5-(3,4-dimethoxyphenyl)-1-(4-methoxyphenyl)-1H-pyra zol-3- yl)phenoxy)ethyl)piperidine;

1-(2-(4-(5-(3-chloro-4-methoxyphenyl)-1-(4-methoxyphenyl) -1H-pyrazol-3- yl)phenoxy)ethyl)piperidine;

1-(2-(4-(5-(4-methoxy-3,5-dimethylphenyl)-1-(4-methoxyphe nyl)-1H-pyrazol-3- yl)phenoxy)ethyl)piperidine;

methyl 4-(1-phenyl-3-(4-(2-(piperidin-1-yl)ethoxy)phenyl)-1H-pyrazo l-5-yl)benzoate;

4-(1-phenyl-3-(4-(2-(piperidin-1-yl)ethoxy)phenyl)-1H-pyr azol-5-yl)benzoic acid;

4-(1-phenyl-3-(4-(2-(piperidin-1-yl)ethoxy)phenyl)-1H-pyr azol-5-yl)benzamide;

4-(1-phenyl-3-(4-(2-(piperidin-1-yl)ethoxy)phenyl)-1H-pyr azol-5-yl)benzonitrile;

4-(1-phenyl-3-(4-(2-(piperidin-1-yl)ethoxy)phenyl)-1H-pyr azol-5-yl)aniline;

methyl (4-(1-phenyl-3-(4-(2-(piperidin-1-yl)ethoxy)phenyl)-1H-pyraz ol-5-yl)phenyl)carbamate; N-(4-(1-phenyl-3-(4-(2-(piperidin-1-yl)ethoxy)phenyl)-1H-pyr azol-5-yl)phenyl)acetamide; N-(4-(1-phenyl-3-(4-(2-(piperidin-1-yl)ethoxy)phenyl)-1H-pyr azol-5-yl)phenyl)propionamide; 1-(2-(4-(5-(3,4-dimethoxyphenyl)-1-phenyl-1H-pyrazol-3-yl)ph enoxy)ethyl)piperidine;

1-(2-(4-(1-phenyl-5-(3,4,5-trimethoxyphenyl)-1H-pyrazol-3 -yl)phenoxy)ethyl)piperidine;

1-(2-(4-(1-phenyl-5-(2,4,5-trimethoxyphenyl)-1H-pyrazol-3 -yl)phenoxy)ethyl)piperidine;

1-(2-(4-(1-phenyl-5-(2,4,6-trimethoxyphenyl)-1H-pyrazol-3 -yl)phenoxy)ethyl)piperidine;

1-(2-(4-(1-phenyl-5-(2,3,4-trimethoxyphenyl)-1H-pyrazol-3 -yl)phenoxy)ethyl)piperidine;

1-(2-(4-(5-(4-ethoxy-3,5-dimethoxyphenyl)-1-phenyl-1H-pyr azol-3-yl)phenoxy)ethyl)piperidine; 1-(2-(4-(1-phenyl-5-(3,4,5-triethoxyphenyl)-1H-pyrazol-3-yl) phenoxy)ethyl)piperidine;

1-(2-(4-(5-(4-ethoxy-3-methoxyphenyl)-1-phenyl-1H-pyrazol -3-yl)phenoxy)ethyl)piperidine; 1-(2-(4-(5-(7-methoxybenzo[d][1,3]dioxol-5-yl)-1-phenyl-1H-p yrazol-3- yl)phenoxy)ethyl)piperidine;

1-(2-(4-(5-(4-methoxy-3,5-dimethylphenyl)-1-phenyl-1H-pyr azol-3- yl)phenoxy)ethyl)piperidine;

1-(2-(4-(5-(3-chloro-4-methoxyphenyl)-1-phenyl-1H-pyrazol -3-yl)phenoxy)ethyl)piperidine; 1-(2-(4-(5-(6-methoxynaphthalen-2-yl)-1-(4-methoxyphenyl)-1H -pyrazol-3- yl)phenoxy)ethyl)piperidine;

1-(2-(4-(1-(4-methoxyphenyl)-5-(thiophen-3-yl)-1H-pyrazol -3-yl)phenoxy)ethyl)piperidine; 4,4'-(3-(4-(3-(piperidin-1-yl)propyl)phenyl)-1H-pyrazole-1,5 -diyl)diphenol; 1-(3-(4-(1,5-bis(4-methoxyphenyl)-1H-pyrazol-3-yl)phenyl)pro pyl)piperidine;

1-(3-(4-(1,5-bis(4-methoxyphenyl)-1H-pyrazol-3-yl)phenoxy )propyl)piperidine;

3-(4-(2-(1H-pyrazol-1-yl)ethoxy)phenyl)-1,5-bis(4-methoxy phenyl)-1H-pyrazole;

4-(1-methyl-3-(4-(2-(piperidin-1-yl)ethoxy)phenyl)-1H-pyr azol-5-yl)phenol;

1-(2-(4-(5-(4-methoxyphenyl)-1-methyl-1H-pyrazol-3-yl)phe noxy)ethyl)piperidine;

1-(2-(4-(4,5-bis(4-methoxyphenyl)-1H-imidazol-2-yl)phenox y)ethyl)piperidine;

1-(2-(4-(4,5-bis(4-methoxyphenyl)-1-methyl-1H-imidazol-2- yl)phenoxy)ethyl)piperidine; 1-(2-(4-(1-ethyl-4,5-bis(4-methoxyphenyl)-1H-imidazol-2-yl)p henoxy)ethyl)piperidine;

1-(2-(4-(4,5-di-p-tolyl-1H-imidazol-2-yl)phenoxy)ethyl)pi peridine;

1-(2-(4-(4,5-bis(4-bromophenyl)-1H-imidazol-2-yl)phenoxy) ethyl)piperidine;

1-(2-(4-(4,5-bis(4-fluorophenyl)-1H-imidazol-2-yl)phenoxy )ethyl)piperidine;

1-(2-(4-(1-ethyl-4,5-diphenyl-1H-imidazol-2-yl)phenoxy)et hyl)piperidine;

4,4'-(2-(4-(2-(piperidin-1-yl)ethoxy)phenyl)-1H-imidazole -4,5-diyl)diphenol;

1-(2-(4-(1,4-bis(4-methoxyphenyl)-1H-imidazol-2-yl)phenox y)ethyl)piperidine;

1-phenyl-2-(4-(2-(piperidin-1-yl)ethoxy)phenyl)-1H-benzo[ d]imidazole;

1-methyl-2-(4-(2-(piperidin-1-yl)ethoxy)phenyl)-1H-benzo[ d]imidazole;

5-(4-methoxyphenyl)-3-(4-(2-(piperidin-1-yl)ethoxy)phenyl )isoxazole;

5-phenyl-3-(4-(2-(piperidin-1-yl)ethoxy)phenyl)isoxazole;

3-phenyl-5-(4-(2-(piperidin-1-yl)ethoxy)phenyl)isoxazole;

5-(4-(2-(piperidin-1-yl)ethoxy)phenyl)-3-(p-tolyl)isoxazo le;

3-(4-fluorophenyl)-5-(4-(2-(piperidin-1-yl)ethoxy)phenyl) isoxazole;

3-(naphthalen-2-yl)-5-(4-(2-(piperidin-1-yl)ethoxy)phenyl )isoxazole;

3-(4-bromophenyl)-5-(4-(2-(piperidin-1-yl)ethoxy)phenyl)i soxazole;

5-(4-methoxyphenyl)-3-(1-(2-(piperidin-1-yl)ethyl)-1H-ben zo[d]imidazol-5-yl)-1,2,4- oxadiazole;

4-(3-(1-(2-(pyrrolidin-1-yl)ethyl)-1H-benzo[d]imidazol-5- yl)-1,2,4-oxadiazol-5-yl)benzonitrile; 4,5-bis(4-methoxyphenyl)-2-(4-(2-(piperidin-1-yl)ethoxy)phen yl)thiazole; and

4,5-bis(4-methoxyphenyl)-2-(4-(2-(piperidin-1-yl)ethoxy)p henyl)oxazole;

or a pharmaceutically acceptable salt thereof. [00431] Clause 46. A pharmaceutical composition comprising a compound of any of clauses 1-45, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier.

[00432] Clause 47. A method of treating a disease or condition mediated by Toll-like receptor activation comprising administering to a subject, in need thereof, a therapeutically effective amount of a compound of any of clauses 1-45, or a pharmaceutically acceptable salt thereof, or the pharmaceutical composition of clause 46.

[00433] Clause 48. The method of clause 47, wherein the disease or condition is an inflammatory disease or condition.

[00434] Clause 49. The method of clause 47, wherein the disease or condition is selected from bacterial sepsis, autoimmune disease, lupus erythematosus, ischemia-reperfusion injury, stroke, metabolic disease, obesity-related metabolic inflammation, gout, and cancer.

[00435] Clause 50. A kit comprising the compound of any of clauses 1-45, or a

pharmaceutically acceptable salt thereof, or the pharmaceutical composition of clause 46, and instructions for use thereof.

[00436] The foregoing discussion discloses and describes merely exemplary embodiments of the invention. One skilled in the art will readily recognize from such discussion and from the accompanying drawings and claims, that various changes, modifications and variations can be made therein without departing from the spirit and scope of the invention as defined in the following claims.