Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
COMPOSITIONS AND METHODS FOR THE TREATMENT OF DISEASE ASSOCIATED WITH TRP-P8 EXPRESSION
Document Type and Number:
WIPO Patent Application WO/2005/020897
Kind Code:
A2
Abstract:
Provided are small-molecule Trp-p8 modulators, including Trp-p8 agonists and Trp-p8 antagonists, and compositions comprising small-molecule Trp-p8 agonists as well as methods for identifying and characterizing novel small-molecule Trp-p8 modulators and methods for decreasing viability and/or inhibiting growth of Trp-p8 expressing cells, methods for activating Trp-p8-mediated cation influx, methods for stimulating apoptosis and/or necrosis, and related methods for the treatment of diseases, including cancers such as lung, breast, colon, and/or prostate cancers as well as other diseases, such as benign prostatic hyperplasia, that are associated with Trp-p8 expression.

Inventors:
NATARAJAN SATEESH K (US)
MORENO OFIR (US)
GRADDIS THOMAS J (US)
DUNCAN DAVID (US)
LAUS REINER (US)
CHEN FENG (US)
Application Number:
PCT/US2004/026931
Publication Date:
March 10, 2005
Filing Date:
August 20, 2004
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
DENDREON CORP (US)
NATARAJAN SATEESH K (US)
MORENO OFIR (US)
GRADDIS THOMAS J (US)
DUNCAN DAVID (US)
LAUS REINER (US)
CHEN FENG (US)
International Classes:
A61K31/498; A61K31/517; A61K45/06; C07C311/20; C07C311/37; C07C323/40; C07C323/44; C07C333/06; C07D207/06; C07D207/09; C07D207/325; C07D209/08; C07D209/14; C07D209/20; C07D209/48; C07D213/40; C07D215/04; C07D215/08; C07D215/46; C07D217/06; C07D231/12; C07D231/14; C07D231/56; C07D235/18; C07D235/26; C07D239/42; C07D241/18; C07D241/44; C07D251/54; C07D263/34; C07D263/57; C07D279/12; C07D295/096; C07D295/135; C07D295/26; C07D307/52; C07D317/66; C07D333/34; C07D401/04; C07D405/12; C07D413/04; C07D413/12; C07D417/04; C07D471/04; C07D487/04; C07D493/08; C07D495/04; C07D513/04; G01N33/68; C07D207/32; C07D263/56; (IPC1-7): A61K/
Foreign References:
US4459425A1984-07-10
Other References:
See also references of EP 1663962A4
Attorney, Agent or Firm:
Myles, Gary M. (P.O. Box 5257 New York, NY, US)
Download PDF:
Claims:
We claim:
1. A compound of Formula VIII wherein R22 is a linker moiety, which may be selected from the group consisting of oxyacetamide, urea, carbamate, thiourea, sulfonamide, amine, amide; R23 is selected from the group consisting of H, an aliphatic group of up to 25 carbons, and an aryl group of up to 10 carbons selected from the group consisting of substituted phenyl, phenalkyl, substituted phenalkyl, naphthyl, substituted naphthyl, and pyridyl; and R24 is selected from the group consisting of H, OH, and an aliphatic group containing up to 25 carbon atoms; and R25 is selected from the group consisting of H.
2. The compound of claim 1 wherein R23 is selected from the group consisting of H, tetrahydro isoquinolinyl, tetrahydro quinolinyl, 3methyl indolinyl, indolinyl, 2 (Nmethyl, Nphenylethyl) amino ethyl, 3methyl indolinyl, 1phenyl ethyl, 2 chloro benzyl, 2methoxybenzyl, 2methoxyphenyl, 2cyclohex1enyl ethyl, (1phenyl cyclophentyl)methyl, 2 (tetrahydroquinolinyl)ethyl, 1propyl1, 2,3, 4tetrahydro pyrrolo [1, 2a] pyrazine, cycloheptyl, 3cyclohexylsulfanylpropyl, 2cyclohex1enyl ethyl, 2 (Nisopropyl, Nphenylethyl) amino ethyl, 1methyl1, 2,3, 4tetrhydropyrrolo [1, 2 a] pyrazine, 2cyclopentylethyl, 2phenylcyclopropyl, 1phenoxyethyl, 4butyloxyphenyl, (2nitrophenoxy) methyl, 4,7, 7trimethyl2oxabicyclo [2.2. 1] heptan3one, C (1phenyl 5propyllHpyrazol4yl)methyl, benzyl, 2chlorobenzyl, 1 [3 (6, 7dimethoxy1 methyl3, 4dihydrolHisoquinolin2ylmethyl)4methoxyphenyl]2, 3,4, 9tetrahydro <BR> <BR> <BR> <BR> 1Hbcarboline, C [3 (4butoxyphenyl)lHpyrazol4yl]methyl, 4 (azepane1 sulfonyl) phenyl, and 5 (7chloroquinolin4ylsulfanyl) [1, 3,4] thiadiazol2yl.
3. The compound of claim 1 wherein R24 is selected from the group consisting of H, tetrahydro isoquinolinyl, tetrahydro quinolinyl, 3methyl indolinyl, indolinyl, 3methyl indolinyl, 1propyl1, 2,3, 4tetrahydropyrrolo [1, 2a] pyrazine, 1 methyl1, 2,3, 4tetrhydropyrrolo [1,2a] pyrazine, and 1 [3 (6, 7dimethoxy1methyl3, 4 dihydrolHisoquinolin2ylmethyl)4methoxyphenyl]2, 3,4, 9tetrahydrolHb carboline.
4. The compound of claim 2 wherein R24 is selected from the group consisting of H, tetrahydro isoquinolinyl, tetrahydro quinolinyl, 3methyl indolinyl, indolinyl, 3methyl indolinyl, 1propyl1, 2,3, 4tetrahydropyrrolo [1, 2a] pyrazine, 1 methyl1, 2,3, 4tetrhydropyrrolo [1, 2a] pyrazine, and 1 [3 (6, 7dimethoxylmethyl3, 4 dihydrolHisoquinolin2ylmethyl)4methoxyphenyl]2, 3,4, 9tetrahydrolHb carboline.
5. A composition comprising a compound of claim 1, claim 2, claim 3, or claim 4 and a pharmaceutically acceptable excipient or diluent.
6. The composition of claim 5 further comprising a cancer therapeutic agent.
7. The composition of claim 6 wherein said cancer therapeutic agent is selected from the group consisting of an antimitotic agent, an alkylating agent, an antimetabolites, a cytotoxic antibiotic, an hormonal agent, an antiestrogen, an anti androgens, and a farnesyl transferase inhibitor.
8. A method for decreasing the viability of a Trpp8 expressing cell, said method comprising the step of contacting said cell with a smallmolecule Trpp8 modulator in a concentration and for a time required to decrease the viability of said cell.
9. The method of claim 8 wherein said smallmolecule Trpp8 modulator is selected from the group consisting of a compound of Formula I, a compound of Formula II, a compound of Formula in, a compound of Formula IV, a compound of Formula V, a compound of Formula VI, a compound of Formula VII, and a compound of Formula VIII.
10. The method of claim 9 wherein said smallmolecule Trpp8 modulator is a compound of Formula I wherein Rl is selected from the group consisting of H, OH, CH3, CH3CHCH3 (isopropyl), and CH3C=CH2 (isopropenyl); is selected from the group consisting of H ; is selected from the group consisting of O, OH, acetate, lactate, carboxamide, butanamide, sulphanamide, and propanetriol; and R4 is selected from the group consisting of CH3CHCH3 (isopropyl), isopropane 2ol, and CH3C=CH2 (isopropenyl).
11. The method of claim 9 wherein said smallmolecule Trpp8 agonist is a compound of Formula II wherein Rs is selected from the group consisting of H, OH, CH3, CH3CHCH3 (isopropyl), and CH3C=CH2 (isopropenyl); R6 is selected from the group consisting of N ; R7 is selected from the group consisting of O and N; R8 is selected from the group consisting of NH, O, and S; and R9 is selected from the group consisting of N02.
12. The method of claim 9 wherein said smallmolecule Trpp8 modulator is a compound of Formula III wherein Rl° is selected from the group consisting of H and a ClC5 alkyl including, but not limited to, CH3, C2H5, CH3CHCH3 (isopropyl) and CH3C=CH2 (isopropenyl); Rll is selected from the group consisting of OH, carboxamide, butanamide, propanetriol, and CONR'R", wherein R'is selected from the group consisting of H, CH3, C2H5, C4Hs (cyclobutyl), and C4H8O, and wherein R"is selected from the group consisting of C2H500CH2, C2H5, CH3CHCH3 (isopropyl), HOCH2G (CH3) 2, HOCH2CH2, C4H9 (tertbutyl), and C4H9 (secbutyl); R12 is selected from the group consisting of H and a ClC5 alkyl including, but not limited to, CH3, CH3CHCH3 (isopropyl), CH3C=CH2 (isopropenyl), C4H9 (secbutyl), C4H9 (isobutyl), C4H9 (nbutyl), and C5H11 (isohexyl) ; and R13 is selected from the group consisting of H and a ClC5 alkyl including, but not limited to, CH3, C2H5, CH3CHCH3 (isopropyl), CH3C=CH2 (isopropenyl), C4H9 (secbutyl), and C4H9 (isobutyl).
13. The method of claim 9 wherein said smallmolecule Trpp8 modulator is a compound of Formula IV wherein R14 is selected from the group consisting of H, an aliphatic group of up to 25 carbons, and an aryl group of up to 10 carbons selected from the group consisting of substituted phenyl, phenalkyl, substituted phenalkyl, naphthyl, substituted naphthyl, and pyridyl; and R15 is selected from the group consisting of H, OH, and an aliphatic group containing up to 25 carbon atoms.
14. The method of claim 13 wherein R"and R"together form a cyclic or heterocyclic group of up to 25 carbons and including the nitrogen atom.
15. The method of claim 14 wherein said cyclic or heterocyclic group is selected from the group consisting of 3phenylpiperidin1yl, 3phenylpyrrolidin1yl, 6, 7dimethoxylmethyl3, 4dihydrolHisoquinolin2yl, and 4pyrimidin2yl piperazin1yl.
16. The method of claim 13 wherein R14 and Rl5 are aliphatic groups selected from the group consisting of methyl, ethyl, propyl, butyl, isobutyl, ndecyl, cyclopropyl, cyclohexyl, cyclopentyl, cycloheptylmethyl, 2hydroxyethyl, 3hydroxynpropyl, 6 hydroxynhexyl, 2aminoethyl, 2acetoxyethyl, 2ethylcarboxyethyl, 4hydroxybut2 ynyl, and carboxymethyl.
17. The method of claim 13 wherein R 14 is an aryl group selected from the group consisting of benzyl, naphthyl, 4methoxyphenyl, 2methoxy4methoxyphenyl, 3 methoxy5methoxyphenyl, 4methyl5chlorophenyl, 4hydroxyphenyl, 4methylphenyl, 3methyl4methylphenyl, 3hydroxy4methylphenyl, 4fluorophenyl, 4chlorophenyl, 4 bromophenyl, 4ethylphenyl, 2fluoro4fluorophenyl, 4nitrophenyl, 2hydroxynaphthyl, pyridyl, [1carbamoyl2 (IHindol3yl)ethyl, 1,3dioxo2, 3dihydro1 Hisoindol4yl, 1, 3dioxo2, 3dihydrolHisoindol5yl, 1carbamoyl2 (4hydroxyphenyl) ethyl, 1 carbamoyl2phenylethyl, 1hydroxymethyl2 (lHindol3yl) ethyl, 1hydroxymethyl2 (4chlorophenyl) ethyl, 1hydroxymethyl2 (4hydroxyphenyl) ethyl, 1hydroxymethyl2 hydroxy2phenylethyl, 1hydroxymethyl2phenyl ethyl, 1methoxymethyl2 phenylethyl, 1methyl2 (4chlorophenyl)ethyl, 1methyl2 (5fluorolHindol3yl) ethyl, 1methyl2hydroxy2phenylethyl, 1oxo2phenylethyl, 1phenyl cyclopentylmethyl, 2 (lcyclopentylpyrrolidin3yl)ethyl, 2 (lHindol3yl) ethyl, 2 (2, 3 dimethoxyphenyl) ethyl, 2 (2, 4dichlorophenyl) ethyl, 2 (2, 4dimethylphenyl) ethyl, 2 (2, 5 dimethoxyphenyl) ethyl, 2 (2, 5dimethylphenyl) ethyl, 2(2, 6dimethylphenyl) ethyl, 2 (2 chloro6flourophenyl) ethyl, 2 (2chlorophenyl) ethyl, 2 (2flourophenyl) ethyl, 2 (2 furyl) ethyl, 2 (2methoxy5bromophenyl) ethyl, 2 (2methoxyphenyl)ethyl, 2 (2 methylphenyl) ethyl, 2 (3, 4dichlorophenyl) ethyl, 2 (3, 4dimethoxyphenyl) ethyl, 2 (3, 5 dimethoxyphenyl) ethyl, 2 (3bromo4methoxyphenyl) ethyl, 2 (3chlorophenyl) ethyl, 2 (3ethoxyphenyl) ethyl, 2 (3flourophenyl) ethyl, 2 (3hydroxy4methoxyphenyl)ethyl, 2 (3hydroxyphenyl) ethyl, 2 (3methoxy4ethoxyphenyl) ethyl, 2 (3methoxy4 hydroxyphenyl) ethyl, 2 (3methoxyphenyl) ethyl, 2 (3methylphenyl) ethyl, 2 (3 triflouromethylphenyl) ethyl, 2 (4bromophenyl)ethyl, 2 (4chlorophenyl) ethyl, 2 (4 ethylphenyl) ethyl, 2 (4fluorophenyl) ethyl, 2 (4hydroxyphenyl) ethyl, 2 (4methoxy phenyl) 2oxoethyl, 2 (4methoxyphenyl) ethyl, 2 (4methylphenyl) ethyl, 2 (4 methylphenyl) ethyl, 2 (4methylsulfanylphenyl) ethyl, 2 (4nitrophenyl) ethyl, 2 (4 <BR> <BR> <BR> sulfamoylphenyl)ethyl, 2 (5methoxylHindol3yl)ethyl, 2 (6fluorolHindol3yl) ethyl, 2(6methoxylHindol3yl)ethyl, 2(7methyllHindol3yl)ethyl, 2>, N dipropylamino) ethyl, 2 (pyridin2yl)ethyl, 2 (pyridin3yl)ethyl, 2 (pyridin4yl)ethyl, 2,2diphenylethyl, 2,3diflourobenzyl, 2,3dimethoxyphenyl, 2,4dichlorophenyl, 2,4 diflourophenyl, 2,4dimethoxyphenyl, 2,4dimethylphenyl, 2bromo4methylphenyl, 2 chloro4cyanophenyl, 2chloro4flourophenyl, 2chloro4iodophenyl, 2chloro4 nitrophenyl, 2chloro5nitrophenyl, 2chlorophenyl, 2cyclohex1enylethyl, 2flouro4 chlorophenyl, 2flouro5nitrophenyl, 2hydroxy2 (3hydroxyphenyl) ethyl, 2hydroxy2 (4hydroxyphenyl) ethyl, 2hydroxy2phenylethyl, 2iodophenyl, 2methyl1, 3dioxo2,3 dihydrolHisoindol5yl, 2methyl2phenylethyl, 2methyl4broophenyl, 2methyl5 nitrophenyl, 2methylphenyl, 2nitro4flourophenyl, 2nitrophenyl, 2oxo2,3dihydro 1Hbenzoimidazol5yl, 2oxo2phenylethyl, 2phenyllHbenzoimidazol5yl, 2phenyl <BR> <BR> <BR> 2 (4chlorophenyl) etliyl, 2phenyl2 (4flourophenyl)ethyl, 2phenyl2 (4 methoxyphenyl) ethyl, 2phenylethenyl, 2phenylethyl, 2pyridin2ylbenzooxazol5yl, 2pyridin3yl1Hbenzoimidazol5yl, 2thiophen2ylethyl), 2trilfouromethyllH benzoimidazol5yl, 3,4, 5triflourophenyl, 3,4, 5trimethoxyphenyl, 3,4 cyclopentanephenyl, 3,4dichlorophenyl, 3,4dimethylphenyl, 3, 5dimethoxyphenyl, 3 acetamidophenyl, 3bromo4methylphenyl, 3carboxamidophenyl, 3chloro4 methoxyphenyl, 3chloro4methylphenyl, 3chloro4morpholin4ylphenyl, 3 hydroxymethylphenyl, 3nitrophenyl, 3oxoindan5yl, 3phenylpropyl, 3yl acetophenone, 4 (1, 1dioxo116thiomorpholin4ylmethyl)phenyl, 4 (1 hydroxyethyl) phenyl, 4 (2hydroxyethyl) phenyl, 4 (4ethylpiperazin1yl)phenyl, 4 (4 methyllHbenzoimidazol2yl)phenyl, 4 (4methylpiperazin1ylmethyl)phenyl, 4 (morpholine4sulfonyl)phenyl, 4 [1, 3dioxo2 (2trifluoromethylphenyl)2, 3dihydro 1Hisoindol5yloxy]phenyl, 4 [2 (2methoxyphenyl)1, 3dioxo2, 3dihydrolH isoindol5yloxy]phenyl, 4 {2 [2 (3, 4dimethoxyphenyl)ethyl]1, 3dioxo2,3dihydro 1Hisoindol5yloxy}phenyl, 4acetylphenyl, 4azepan1ylmethylphenyl, 4 benzooxazol2ylphenyl, 4Biphenyl, 4bromophenyl, 4carboxamidophenyl, 4 chlorophenyl, 4cyanophenyl, 4ethoxyphenyl, 4ethylphenyl, 4fluorophenyl, 4 hydroxymethylphenyl, 4methoxyphenyl, 4methyl2oxo2Hchromen7yl, 4 methylcyclohexyl, 4methylphenyl, 4methylsulfanylphenyl, 4nitrobenzyl, 4pyrrolidin 1ylmethylphenyl, 4triflouromethylphenyl, benzo [1, 3] dioxol5yl, benzoylamino, benzyloxy, bicyclo [2.2. 1] hept2yl, ClHindazol5yl, cycloheptyl, indan2yl, N (2 diethylaminoethyl) benzamide4yl, N'quinoxalin2ylamino, and phenylcyclopropyl.
18. The method of claim 9 wherein said smallmolecule Trpp8 modulator is a compound of Formula V v wherein R16 is selected from the group consisting of a C2C6 alkylene group having at least one, but not more than three, hydroxyl groups; and Rl7 and Rl8, independently of one another, are selected from the group consisting of ClCl0alkyl groups, C5C7cycloalkyl, C6Cl2aryl, wherein the total of the C atoms of Rl7 and Rl8 is not less than 3, or RI and Rl8 together represent an alkylene group that, together with the carbon atom that carries the groups Rl7 and R'8, forms a 57membered ring.
19. The method of claim 18 wherein one or both of the alkyl groups of said Rl7 and Rl8 are substituted by 1 to 3 groups selected from the group consisting of hydroxyl, amino, thio, and halogen.
20. The method of claim 9 wherein said smallmolecule Trpp8 modulator is a compound of Formula VII VII wherein R17 is selected from the group consisting of 2pyridyl, 2nitro4 trifluoromethylphenyl, 2nitro4chlorophenyl, 2methoxyphenyl, 2chlorophenyl, phenyl, 2methylquinolin3yl, 4methoxyphenyl, 4fluorophenyl, 3azepanlyl5 (4 trifluoromethoxy) phenylamino [1, 3,5] triazyl, cyclohexyl, diphenylmethyl, 2phenylethyl, 4hydroxycyclohexyl, cycloheptyl, cyclopentyl, Cbenzo [1, 3] dioxol5ylmethyl, 2 pyridyl, and 4chlorobenzyl; Rl8 is selected from the group consisting of 1benzylIHpyrazolo [3,4 <BR> <BR> <BR> <BR> d] pyrimidin4yl, 3benzylamino2nitrophenyl, 5nitroquinolin8yl, 1yl3(2 isopropyl5methylcyclohexyloxy)propan2ol, 1phenyllHpyrazolo [3,4d] pyrimidin 4yl, benzyl2methylquinazolin4yl, 3methyl5morpholin4yl2nitrophenyl, 2nitro 5piperazin1ylethanol, 1yl3 (2isopropyl5methylcyclohexyloxy)propan2ol, 4 (2,5dimethylpyrrol1yl)2nitrophenyl, 2nitro3trifluoromethanesulfonylphenyl, 1 phenyllHpyrazolo [3,4d] pyrimidin4yl, 2 (2Fluorophenoxymethyl)2cyano oxazolyl, adamantly, 5 (benzo [1, 3] dioxol5ylamino)10b, 10cdihydroanthra [l, 9cd] isoxazol6 oneyl, 2methylthiazolo [3,2b] [1, 2,4] triazol6ol 4methylphenyl methyl, 3benzyl3H quinazolin4one2yl, cyclopentyl, tetrahydronapthyl, cyclooctyl, cyclohexyl, C [3 (4 chlorophenyl) 2, 5dimethylpyrazolo [1, 5a] pyrimidin7yl]methyl, C (2benzyl5, 6,7, 8 tetrahydrobenzo [4,5] thieno [2,3d] pyrimidin4yl) methyl, and 1yl3 (2isopropyl5 methylcyclohexyloxy)propan2ol ; Rl9 and Ro are each independently selected from the group consisting of H and O ; and Wl is selected from the group consisting of 4methylphenyl, 2chloro4 fluorophenyl, and 4chlorophenyl.
21. The method of claim 9 wherein said smallmolecule Trpp8 modulator is a compound of Formula VIII wherein R22 is a linker moiety, which may be selected from the group consisting of oxyacetamide, urea, carbamate, thiourea, sulfonamide, amine, amide; R23 is selected from the group consisting of H, tetrahydro isoquinolinyl, tetrahydro quinolinyl, 3methyl indolinyl, indolinyl, 2 (Nmethyl, Nphenylethyl) amino ethyl, 3 methyl indolinyl, 1phenyl ethyl, 2chloro benzyl, 2methoxybenzyl, 2methoxyphenyl, 2 cyclohex1enyl ethyl, (1phenylcyclophentyl)methyl, 2 (tetrahydroquinolinyl)ethyl, 1 propyl1, 2,3, 4tetrahydropyrrolo [1, 2a] pyrazine, cycloheptyl, 3 cyclohexylsulfanylpropyl, 2cyclohex1enyl ethyl, 2 (Nisopropyl, Nphenylethyl) amino ethyl, 1methyl1, 2,3, 4tetrhydropyrrolo [1, 2a] pyrazine, 2cyclopentylethyl, 2 phenylcyclopropyl, 1phenoxyethyl, 4butyloxyphenyl, (2nitrophenoxy) methyl, 4,7, 7 trimethyl2oxabicyclo [2.2. 1] heptan3one, C (Iphenyl5propylIHpyrazol4yl) methyl, benzyl, 2chlorobenzyl, 1 [3 (6, 7dimethoxy1methyl3, 4dihydrolH isoquinolin2ylmethyl)4methoxyphenyl]2, 3,4, 9tetrahydro1Hbcarboline, C [3 (4 butoxyphenyl)lHpyrazol4yl]methyl, 4(azepane1sulfonyl)phenyl, and 5 (7chloro quinolin4ylsulfanyl) [1, 3,4] thiadiazol2yl; R24 is selected from the group consisting of H, tetrahydro isoquinolinyl, tetrahydro quinolinyl, 3methyl indolinyl, indolinyl, 3methyl indolinyl, 1propyl1, 2,3, 4 tetrahydropyrrolo [1, 2a] pyrazine, 1methyl1, 2,3, 4tetrhydropyrrolo [1, 2a] pyrazine, and 1 [3 (6, 7dimethoxy1methyl3, 4dihydrolHisoquinolin2ylmethyl)4methoxy phenyl] 2, 3,4, 9tetrahydrolHbcarboline ; and R25 is selected from the group consisting of H.
22. A method for inducing apoptosis and/or necrosis in a cell expressing Trp p8, said method comprising the step of administering to said cell a smallmolecule Trpp8 modulator selected from the group consisting of a compound of Formula I, a compound of Formula II, a compound of Formula III, a compound of Formula IV, a compound of Formula V, a compound of Formula VI, a compound of Formula VII, and a compound of Formula Vin.
23. A method for treating a disease associated with Trpp8 expression, said method comprising the steps of administering to a mammal an efficacious amount of a composition comprising a compound having the formula: I in combination with a pharmaceutically acceptable carrier or diluent, wherein Rl is selected from the group consisting of H, OH, CH3, CH3CHCH3 (isopropyl), and CH3C=CH2 (isopropenyl); R2 is selected from the group consisting of OH, carboxamide, butanamide, and propanetriol; and R3 is selected from the group consisting of CH3CHCH3 (isopropyl), isopropane 2ol, and CH3C=CH2 (isopropenyl).
24. A method for treating a disease associated with Trpp8 expression, said method comprising the step of administering to a mammal an efficacious amount of a composition comprising a compound having the formula: in combination with a pharmaceutically acceptable carrier or diluent, wherein R5 is selected from the group consisting of H, OH, CH3, CH3CHCH3 (isopropyl), and CH3C=CH2 (isopropenyl); R6 is selected from the group consisting of N ; R is selected from the group consisting of O, CH2, and CH3CCH3 (isopropyl) ; R8 is selected from the group consisting of NH ; and R9 is selected from the group consisting of NO2.
25. A method for treating a disease associated with Trpp8 expression, said method comprising the step of administering to a mammal an efficacious amount of a composition comprising a compound having the formula: in combination with a pharmaceutically acceptable carrier or diluent, wherein Rl° is selected from the group consisting of H, CH3, CH3CHCH3 (isopropyl}, and CH3C=CH2 (isopropenyl); Rll is selected from the group consisting of OH, carboxamide, butanamide, and propanetriol; R12 is selected from the group consisting of H, CH3, CH3CHCH3 (isopropyl), and CH3C=CH2 (isopropenyl); and R13 is selected from the group consisting of H, CH3, CH3CHCH3 (isopropyl), and CH3C=CH2 (isopropenyl); and a pharmaceutically acceptable carrier or diluent.
26. A method for treating a disease associated with Trpp8 expression, said method comprising the step of administering to a mammal an efficacious amount of a composition comprising a compound having the formula: in combination with a pharmaceutically acceptable carrier or diluent, wherein R14 is selected from the group consisting of H, an aliphatic group of up to 25 carbons, and an aryl group of up to 10 carbons selected from the group consisting of substituted phenyl, phenalkyl, substituted phenalkyl, naphthyl, substituted naphthyl, and pyridyl; and RI5 is selected from the group consisting of H, OH, and an aliphatic group containing up to 25 carbon atoms.
27. The method of claim 26 wherein R14 and R15 together form a cyclic or heterocyclic group of up to 25 carbons and including the nitrogen atom.
28. The method of claim 27 wherein said cyclic or heterocyclic group is selected from the group consisting of 3phenylpiperidin1yl, 3phenylpyrrolidin1yl, 6, 7dimethoxy1methyl3, 4dihydrolHisoquinolin2yl, and 4pyrimidin2yl piperazin1yl.
29. The method of claim 26 wherein R14 and Rl5 are each aliphatic groups independently selected from the group consisting of methyl, ethyl, propyl, butyl, isobutyl, ndecyl, cyclopropyl, cyclohexyl, cyclopentyl, cycloheptylmethyl, 2hydroxyethyl, 3 hydroxynpropyl, 6hydroxynhexyl, 2aminoethyl, 2acetoxyethyl, 2ethylcarboxyethyl, 4hydroxybut2ynyl, and carboxymethyl.
30. The method of claim 26 wherein R14 is an aryl group selected from the group consisting of benzyl, naphthyl, 4methoxyphenyl, 2methoxy4methoxyphenyl, 3 methoxy5methoxyphenyl, 4methyl5chlorophenyl, 4hydroxyphenyl, 4methylphenyl, 3methyl4methylphenyl, 3hydroxy4methylphenyl, 4fluorophenyl, 4chlorophenyl, 4 bromophenyl, 4ethylphenyl, 2fluoro4fluorophenyl, 4nitrophenyl, 2hydroxynaphthyl, pyridyl, [lcarbamoyl2(lHindol3yl)ethyl, 1, 3dioxo2, 3dihydrolHisoindol4yl, 1,3dioxo2, 3dihydro1Hisoindol5yl, 1carbamoyl2 (4hydroxyphenyl) ethyl, 1 carbamoyl2phenylethyl, 1hydroxymethyl2(lHindol3yl) ethyl, 1hydroxymethyl2 (4chlorophenyl) ethyl, 1hydroxymethyl2 (4hydroxyphenyl) ethyl, 1hydroxymethyl2 hydroxy2phenylethyl, 1hydroxymethyl2phenyl ethyl, 1methoxymethyl2 phenylethyl, 1methyl2 (4chlorophenyl)ethyl, 1methyl2 (5fluorolHindol3yl) ethyl, lmethyl2hydroxy2phenylethyl, 1oxo2phenylethyl, 1phenyl cyclopentylmethyl, 2 (lcyclopentylpyrrolidin3yl)ethyl, 2 (lHindol3yl) ethyl, 2 (2, 3 dimethoxyphenyl) ethyl, 2 (2, 4dichlorophenyl) ethyl, 2 (2, 4dimethylphenyl) ethyl, 2 (2, 5 dimethoxyphenyl) ethyl, 2 (2, 5dimethylphenyl) ethyl, 2 (2, 6dimethylphenyl) ethyl, 2 (2 chloro6flourophenyl) ethyl, 2 (2chlorophenyl) ethyl, 2 (2flourophenyl) ethyl, 2 (2 furyl) ethyl, 2 (2methoxy5bromophenyl) ethyl, 2 (2methoxyphenyl)ethyl, 2 (2 methylphenyl) ethyl, 2 (3, 4dichlorophenyl) ethyl, 2 (3, 4dimethoxyphenyl) ethyl, 2 (3, 5 dimethoxyphenyl) ethyl, 2 (3bromo4methoxyphenyl) ethyl, 2 (3chlorophenyl) ethyl, 2 (3ethoxyphenyl) ethyl, 2 (3flourophenyl) ethyl, 2 (3hydroxy4methoxyphenyl)ethyl, 2 (3hydroxyphenyl) ethyl, 2 (3methoxy4ethoxyphenyl) ethyl, 2 (3methoxy4 hydroxyphenyl) ethyl, 2 (3methoxyphenyl) ethyl, 2 (3methylphenyl) ethyl, 2 (3 triflouromethylphenyl) ethyl, 2 (4bromophenyl)ethyl, 2 (4chlorophenyl) ethyl, 2 (4 ethylphenyl) ethyl, 2 (4fluorophenyl) ethyl, 2 (4hydroxyphenyl) ethyl, 2 (4methoxy phenyl) 2oxoethyl, 2 (4methoxyphenyl) ethyl, 2 (4methylphenyl) ethyl, 2 (4 methylphenyl) ethyl, 2 (4methylsulfanylphenyl) ethyl, 2 (4nitrophenyl) ethyl, 2 (4 sulfamoylphenyl) ethyl, 2 Hindol3yl)ethyl, 2(6fluoro1 Hindol3yl) ethyl, 2 (6methoxylHindol3yl)ethyl, 2 (7methyllHindol3yl)ethyl, 2 (N, N dipropylamino) ethyl, 2(pyridin2yl)ethyl, 2(pyridin3yl)ethyl, 2(pyridin4yl)ethyl, 2,2diphenylethyl, 2,3diflourobenzyl, 2,3dimethoxyphenyl, 2, 4dichlorophenyl, 2,4 diflourophenyl, 2,4dimethoxyphenyl, 2,4dimethylphenyl, 2bromo4methylphenyl, 2 chloro4cyanophenyl, 2chloro4flourophenyl, 2chloro4iodophenyl, 2chloro4 nitrophenyl, 2chloro5nitrophenyl, 2chlorophenyl, 2cyclohex1enylethyl, 2flouro4 chlorophenyl, 2flouro5nitrophenyl, 2hydroxy2 (3hydroxyphenyl) ethyl, 2hydroxy2 (4hydroxyphenyl) ethyl, 2hydroxy2phenylethyl, 2iodophenyl, 2methyl1, 3dioxo2,3 dihydrolHisoindol5yl, 2methyl2phenylethyl, 2methyl4broophenyl, 2methyl5 nitrophenyl, 2methylphenyl, 2nitro4flourophenyl, 2nitrophenyl, 2oxo2,3dihydro lHbenzoimidazol5yl, 2oxo2phenylethyl, 2phenyllHbenzoimidazol5yl, 2phenyl 2 (4chlorophenyl) ethyl, 2phenyl2 (4flourophenyl)ethyl, 2phenyl2 (4 methoxyphenyl) ethyl, 2phenylethenyl, 2phenylethyl, 2pyridin2ylbenzooxazol5yl, 2pyridin3yllHbenzoimidazol5yl, 2thiophen2ylethyl), 2trilfouromethyllH benzoimidazol5yl, 3,4, 5triflourophenyl, 3,4, 5trimethoxyphenyl, 3,4 cyclopentanephenyl, 3, 4dichlorophenyl, 3, 4dimethylphenyl, 3,5dimethoxyphenyl, 3 acetamidophenyl, 3bromo4methylphenyl, 3carboxamidophenyl, 3chloro4 methoxyphenyl, 3chloro4methylphenyl, 3chloro4morpholin4ylphenyl, 3 hydroxymethylphenyl, 3nitrophenyl, 3oxoindan5yl, 3phenylpropyl, 3yl acetophenone, 4 (1, 1dioxo116thiomorpholin4ylmethyl)phenyl, 4 (1 hydroxyethyl) phenyl, 4 (2hydroxyethyl) phenyl, 4 (4ethylpiperazin1yl)phenyl, 4 (4 methyllHbenzoimidazol2yl)phenyl, 4 (4methylpiperazin1ylmethyl)phenyl, 4 (morpholine4sulfonyl)phenyl, 4 [1, 3dioxo2 (2trifluoromethylphenyl)2, 3dihydro lHisoindol5yloxy]phenyl, 4 [2 (2methoxyphenyl)1, 3dioxo2, 3dihydrolH isoindol5yloxy] phenyl, 4 {2 [2 (3, 4dimethoxyphenyl)ethyl]1, 3dioxo2, 3dihydro lHisoindol5yloxy}phenyl, 4acetylphenyl, 4azepan1ylmethylphenyl, 4 benzooxazol2ylphenyl, 4Biphenyl, 4bromophenyl, 4carboxamidophenyl, 4 chlorophenyl, 4cyanophenyl, 4ethoxyphenyl, 4ethylphenyl, 4fluorophenyl, 4 hydroxymethylphenyl, 4methoxyphenyl, 4methyl2oxo2Hchromen7yl, 4 methylcyclohexyl, 4methylphenyl, 4methylsulfanylphenyl, 4nitrobenzyl, 4pyrrolidin 1ylmethylphenyl, 4triflouromethylphenyl, benzo [1, 3] dioxol5yl, benzoylamino, benzyloxy, bicyclo [2.2. 1] hept2yl, ClHindazol5yl, cycloheptyl, indan2yl, N (2 diethylaminoethyl) benzamide4yl, N'quinoxalin2ylamino, and phenylcyclopropyl.
31. A method for treating a disease associated with Trpp8 expression, said method comprising the step of administering to a mammal an efficacious amount of a composition comprising a compound having the formula : in combination with a pharmaceutically acceptable carrier or diluent, wherein R16 is selected from the group consisting of a C2C6 alkylene group having at least one, but not more than three, hydroxyl groups; and and R, independently of one another, are selected from the group consisting of ClCloalkyl groups, C5C7cycloalkyl, C6CI2aryl, wherein the total of the C atoms of Rl7 and Rl8 is not less than 3, or Rl7 and Rl8 together represent an alkylene group that, together with the carbon atom that carries the groups RI and R18, forms a 57membered ring.
32. The method of claim 31 wherein one or both of the alkyl groups of said Rl7 and Rl8 are substituted by 1 to 3 groups selected from the group consisting of hydroxyl, amino, thio, and halogen.
33. A method for treating a disease associated with Trpp8 expression, said method comprising the step of administering to a mammal an efficacious amount of a composition comprising a compound having the formula: in combination with a pharmaceutically acceptable carrier or diluent.
34. A method for treating a disease associated with Trpp8 expression, said method comprising the step of administering to a mammal an efficacious amount of a composition comprising a compound having the formula: VII in combination with a pharmaceutically acceptable carrier or diluent, wherein Rl7 is selected from the group consisting of 2pyridyl, 2nitro4 trifluoromethylphenyl, 2nitro4chlorophenyl, 2methoxyphenyl, 2chlorophenyl, phenyl, 2methylquinolin3yl, 4methoxyphenyl, 4fluorophenyl, 3azepanlyl5 (4 trifluoromethoxy) phenylamino [1, 3,5] triazyl, cyclohexyl, diphenylmethyl, 2phenylethyl, 4hydroxycyclohexyl, cycloheptyl, cyclopentyl, Cbenzo [1, 3] dioxol5ylmethyl, 2 pyridyl, and 4chlorobenzyl ; Rl8 is selected from the group consisting of 1benzyllHpyrazolo [3,4 d] pyrimidin4yl, 3benzylamino2nitrophenyl, 5nitroquinolin8yl, 1yl3 (2 isopropyl5methylcyclohexyloxy) propan2ol, 1phenyllHpyrazolo [3, 4d] pyrimidin 4yl, benzyl2methylquinazolin4yl, 3methyl5morpholin4yl2nitrophenyl, 2nitro 5piperazin1ylethanol, 1yl3 (2isopropyl5methylcyclohexyloxy)propan2ol, 4 (2, 5dimethylpyrrol1yl)2nitrophenyl, 2nitro3trifluoromethanesulfonylphenyl, 1 phenyllHpyrazolo [3,4d] pyrimidin4yl, 2 (2Fluorophenoxymethyl)2cyano oxazolyl, adamantly, 5 (benzo [1, 3] dioxol5ylamino)10b, lOcdihydroanthra [1, 9cd] isoxazol6 oneyl, 2methylthiazolo [3,2b] [1, 2,4] triazol6ol 4methylphenyl methyl, 3benzyl3H quinazolin4one2yl, cyclopentyl, tetrahydronapthyl, cyclooctyl, cyclohexyl, C [3 (4 chlorophenyl) 2, 5dimethylpyrazolo [1, 5a] pyrimidin7yl]methyl, C (2benzyl5, 6,7, 8 tetrahydrobenzo [4,5] thieno [2,3d] pyrimidin4yl) methyl, and 1yl3 (2isopropyl5 methylcyclohexyloxy)propan2ol ; Rl9 and Wo are each independently selected from the group consisting of H and O ; and Roi ils selected from the group consisting of 4methylphenyl, 2chloro4 fluorophenyl, and 4chlorophenyl.
35. A method for treating a disease associated with Trpp8 expression, said method comprising the step of administering to a mammal an efficacious amount of a composition comprising a compound having the formula: in combination with a pharmaceutically acceptable carrier or diluent, wherein R22 is a linker moiety, which may be selected from the group consisting of oxyacetamide, urea, carbamate, thiourea, sulfonamide, amine, amide; R23 is selected from the group consisting of H, tetrahydro isoquinolinyl, tetrahydro quinolinyl, 3methyl indolinyl, indolinyl, 2 (Nmethyl, Nphenylethyl) amino ethyl, 3 methyl indolinyl, 1phenyl ethyl, 2chloro benzyl, 2methoxybenzyl, 2methoxyphenyl, 2 cyclohex1enyl ethyl, (lphenylcyclophentyl)methyl, 2 (tetrahydroquinolinyl)ethyl, 1 propyl1, 2,3, 4tetrahydropyrrolo [1, 2a] pyrazine, cycloheptyl, 3 cyclohexylsulfanylpropyl, 2cyclohex1enyl ethyl, 2 (Nisopropyl, Nphenylethyl) amino ethyl, 1methyl1, 2,3, 4tetrhydropyrrolo [1, 2a] pyrazine, 2cyclopentylethyl, 2 phenylcyclopropyl, 1phenoxyethyl, 4butyloxyphenyl, (2nitrophenoxy) methyl, 4,7, 7 trimethyl2oxabicyclo [2.2. 1] heptan3one, C (lphenyl5propyllHpyrazol4yl) methyl, benzyl, 2chlorobenzyl, 1 [3 (6, 7dimethoxy1methyl3, 4dihydrolH isoquinolin2ylmethyl)4methoxyphenyl]2, 3,4, 9tetrahydrolHbcarboline, C [3 (4 butoxyphenyl)lHpyrazol4yl]methyl, 4(azepane1sulfonyl)phenyl, and 5 (7chloro quinolin4ylsulfanyl) [1,3, 4] thiadiazol2yl ; R4 is selected from the group consisting of H, tetrahydro isoquinolinyl, tetrahydro quinolinyl, 3methyl indolinyl, indolinyl, 3methyl indolinyl, 1propyl1, 2,3, 4 tetrahydropyrrolo [1, 2a] pyrazine, 1methyl1,2, 3,4tetrhydropyrrolo [1, 2a] pyrazine, and 1 [3 (6, 7dimethoxylmethyl3, 4dihydrolHisoquinolin2ylmethyl)4methoxy phenyl] 2, 3,4, 9tetrahydrolHbcarboline ; and R25 is selected from the group consisting of H.
36. A method for identifying a Trpp8 agonist, said method comprising the step of contacting a Trpp8 expressing cell and a nonTrpp8 expressing cell with a candidate Trpp8 agonist for a time and in an amount sufficient to decrease the viability of said Trpp8 expressing cell but not said nonTrpp8 expressing cell.
37. A method for identifying a Trpp8 antagonist, said method comprising the step of contacting a Trpp8 expressing cell with a Trpp8 agonist and with a candidate Trpp8 antagonist for a time and in an amount sufficient for said agonist to decrease the 0 viability of said Trpp8 expressing cell, wherein a Trpp8 antagonist is detectedby an increase in the viability of said Trpp8 expressing cell.
Description:
COMPOSITIONS AND METHODS FOR THE TREATMENT OF DISEASE ASSOCIATED WITH TRP-P8 EXPRESSION Cross-Reference to Related Application This application claims priority under 35 U. S. C. 119 (e) to U. S. Provisional Patent Application No. 60/497, 384 filed August 22,2003.

Technical Field of the Invention The present invention relates generally to the fields of cell biology, biochemistry, and organic chemistry. More specifically, the present invention provides small-molecule modulators of Trp-p8 activity, which include Trp-p8 agonists and Trp-p8 antagonists, as well as compositions comprising small-molecule Trp-p8 modulators. Also provided are methods for identifying and characterizing novel small-molecule Trp-p8 modulators as well as methods for modulating Trp-p8-mediated cation influx and/or apoptosis in a cell and related methods for the treatment of diseases associated with Trp-p8 expression, activation, and/or signaling. Exemplary diseases suitably treated by the compositions and methods of the present invention include cancers, such as lung, breast, colon, and/or prostate cancers.

Background of the Invention Prostate carcinoma is the most common cancer diagnosed in men in the United States and has the second highest cancer death rate yielding only to lung adenocarcinoma.

Parker et al., CA Cancer J. Clin. 46: 5-27 (1996). Although it is possible to effectively treat organ-confined prostate cancer, there are very limited treatment options for metastatic disease. Thus, it is of great importance to find novel ways to diagnose early stage disease and to closely monitor both progression and treatment of the disease, as well as to develop new therapeutic approaches. To achieve this, it is important to understand the molecular mechanisms of prostate cancer development and to identify new biochemical markers for disease diagnosis and progression.

To date there are very few prostate-specific markers available. The best-known and well-characterized markers of proven prostate cancer diagnostic value are the proteins prostatic acid phosphatase (PAP), prostate specific antigen (PSA), and prostate-specific membrane antigen (PSMA). Each of these proteins has also become the target for novel

immunotherapy approaches to the treatment of disease. Horoszewicz et al., Anticancer Res. 7: 927-935 (1987); Barren et al., Prostate 30 : 65-68 (1997); Murphy et al., Prostate 33: 281-285 (1997); Murphy et al., Prostate 26: 164-168 (1995); Rochon et al., Prostate 25: 219-223 (1995); Correale et al., J. Immunol. 161 : 3186-3194 (1998); and Murphy et al., Prostate 38: 73-78 (1999).

It has been reported that a cation channel protein, variously designated Trp-p8 (transient receptor potential-p8), TRPM8, and CMR1 (cold and menthol receptor 1), is preferentially expressed in prostate. Cloning of the full-length human trp-p8 cDNA revealed a transcript corresponding to an 1104 amino acid polypeptide sharing homology with the trp family of calcium channels. Clapham et al., Nature Reviews 2 : 387-396 (2001) and Clapham et al., IUPHAR Compendium, TRP Channels (2002). Trp-p8 shows particularly high homology with the human TRPC7 gene--a putative Ca2+ channel protein of the trp family that is highly expressed in brain tissue. Nagamine et al., Genomics 54 : 124-131 (1998). Trp-p8 also shows significant homology to human melastatin, another Trp family-related protein expressed in melanocytes and believed to be a tumor suppressor gene. Duncan et al., Cancer Res. 58 : 1515-1520 (1998) ; and Hunter et al., Genomics 54: 116-123 (1998). Perhaps of greatest interest is the observation that the trp-p8 gene appears to be expressed in a large spectrum of nonprostatic in addition to prostatic neoplastic lesions. Tsavaler et al., Cancer Res. 6l (9 ! : 3760-g (2001).

The Trp superfamily comprises more than 20 related cation channel proteins that have been implicated in processes including sensory physiology to vasorelaxation and male fertility. Defects in Trp channels have been associated with changes in growth control and tumor suppression. While all Trp proteins are calcium channels, they vary significantly in their selectivity and mode of activation. Members of the Trp superfamily share significant sequence homology and predicted structural similarities, such as size of predicted transmembrane segments.

Trp-p8 is over-expressed in a range of cancers including prostate, breast, lung and colon, while within normal tissues, it is predominantly expressed in human prostate [Tsavaler et al., supra] and dorsal root ganglia (DRG, Dendreon, unpublished observation). Fuessel et al. reported that Trp-p8 is a highly prostate-specific and prostate carcinoma-associated gene thus qualifying it as a potential target for specific therapies.

International J. of Oncology 23: 221-228 (2003). Among other species, Trp-p8 orthologues are reportedly expressed in a subset of DRG and trigerminal ganglia (TG)

neurons in rat [McKemy et al., Nature 416 (6876) : 52-8 (2002) ] and mouse [Peier et al., Cell 108 5 : 705-15 (2002) ] as well. Thus, Trp-p8 is a pantumor-expressed marker with significant potential use in disease diagnosis and monitoring of disease progression during treatment as well as a viable target for cancer therapy.

In two articles published concurrently, it was reported for the first time that Trp-p8 orthologues, in response to cold and certain cooling compounds, initiate an influx of cations, such as calcium, from the extracellular space. McKemy et al., supra ; and Peier et al., supra. Two of the best known modulators of Trp-p8 activity are the Trp-p8 agonists menthol and Icilin. Menthol is effective in inducing calcium influx at"10-100 uM while Icilin is more potent with an effective concentration range of 0.1-1 uM.

The higher temperature threshold reported for Trp-p8 activation by the most widely studied agonist, menthol (2-isopropyl-5-methyl-cyclohexanol), is about 30-32°C in a variety of cells (cold-sensitive neurons, Trp-p8 heterologously expressed in Xenopus oocytes, HEK293 and CHO cells). McKemy et al., Nature, supra ; Peier et al., Cell, supra ; Nealen et al., J Neuroplaysiol. 90 1 : 515-520 (2003); and Reid et al., JPhysiol.

545 (Pt 2 ! : 595-614 (2002).

Although certain agonist compounds have been shown to activate Trp-p8 expressing cells up to 32°C, there has been no report disclosing growth manipulation in cells at physiological temperature (i. e. 37°C), the temperature at which a compound must be active in order to be an efficacious in vivo therapeutic.

Association of Trp-p8 with prostate, lung, breast, and colon cancers and the important role various ion channels play in vital cell functions suggest that Trp-p8 channel may have a significant function in cancer cell signaling and/or proliferation.

Modulation of Trp-p8 activity, either by activating via an agonist or inhibiting via an antagonist, at a physiological temperature can be valuable as a therapeutic to manipulate the Trp-p8 expressing cells in a specific manner.

Accordingly, there remains a need in the art for small-molecule modulators of Trp-p8 activity, compositions comprising one or more small-molecule Trp-p8 modulators, and methods for the identification and use of small-molecules for modulating the activity of Trp-p8 in a cell and for the treatment of disease associated with the aberrant expression of Trp-p8.

Summary of the Invention The present invention fulfills these and other related needs by providing small molecule modulators of Trp-p8 activity, including Trp-p8 agonists and Trp-p8 antagonists, as well as compositions comprising such Trp-p8 modulators, and methods for identifying and using Trp-p8 modulators. Within certain embodiments, compounds of the present invention bind to and activate Trp-p8 and/or stimulate cation influx, including but not limited to calcium influx, in a cell wherein cation influx is correlative of Trp-p8 modulator induced toxicity. Thus, within these and other embodiments, Trp-p8 agonists of the present invention are effective in inhibiting growth of and/or inducing apoptosis and/or necrosis in a cell expressing Trp-p8. Within alternative embodiments are provided Trp-p8 antagonists that are effective in reducing the basal activity of Trp-p8 in a cell thereby reducing the viability of Trp-p8 expressing cells. Advantageously, therefore, agonists and antagonists of the present invention can be used to treat diseases including, but not limited to, cancers of the breast, lung, colon, and/or prostate, that are associated with Trp-p8 expression.

One or more Trp-p8 modulator can be formulated in compositions, including pharmaceutical compositions, comprising one or more pharmaceutically acceptable carrier or excipient and/or one or more additional therapeutic compound. Such compositions will find utility in methods for the treatment of one or more disease associated with Trp- p8 expression.

Thus, in one embodiment, the present invention provides the following Trp-p8 modulators and derivatives thereof : A compound of Formula I wherein Rl is selected from the group consisting of H, OH, CH3, CH3-CH-CH3 (isopropyl), and CH3-C=CH2 (isopropenyl);

R is selected from the group consisting of H ; R3 is selected from the group consisting of O, OH, acetate, lactate, carboxamide, butanamide, sulphanamide, and propanetriol ; and R4 is selected from the group consisting of CH3-CH-CH3 (isopropyl), isopropane- 2-ol, and CH3-C=CH2 (isopropenyl).

Exemplary compounds of Formula I include the following:

CH3 CH3 CH3 OH CH3 CH3 CH3 CH3 CH2 CH3 OH Menthol Coolact P Coolact 38D ? CHU OH H3 0 OH CH3 CH3 CH3 CH3 CoolingAgentlO CH3 CHUS 0-C-CH3 CH3 CH3 L-Menthyl Acetate These exemplary Formula I Trp-p8 modulators have the following trade and chemical names: Menthol (2-isopropyl-5-methyl-cyclohexanol) (Sigma-Aldrich, Inc.; St.

Louis, Missouri); Frescolat ML (Harris & Ford, LLC; Indianapolis, Indiana; Menthyl lactate) ; L-Menthyl Acetate (Millenium Chemicals; Olympia Fields, Illinois;

Cyclohexanol-5-methyl-2- (l-methylethyl)-acetate- [lR- (lalpha, 2beta, 5alpha) ]-) ; Cooling Agent 10 (Takasago International Corp.; Rockleigh, New Jersey; (l)-Menthoxypropane- 1,2-diol) ; Coolact P# (Takasago International Corp.; (-) -Isopulegol) ; and Coolact 38D (Takasago International Corp.).

In another embodiment, the present invention further provides the following small-molecule Trp-p8 modulators and derivatives thereof : A compound of Formula II

wherein Rs is selected from the group consisting of H, OH, CH3, CH3-CH-CH3 (isopropyl), and CH3-C=CH2 (isopropenyl); R6 is selected from the group consisting of N ; R7 is selected from the group consisting of O and N; R8 is selected from the group consisting of NH, O, and S; and R9 is selected from the group consisting of NO2.

Compounds of Formula II are exemplified herein by Icilin (1- (2-hydroxyphenyl)- 4-(3-nitrophenyl)-1, 2,3, 6-tetrahydropyrimidine-2-one aka 3, 4-dihydro-3- (2- hydroxyphenyl)-6-(3-nitrophenyl)-(lH)-pyrimidin-2-one).

In another embodiment, the present invention further provides the following acyclic carboxamide Trp-p8 agonists and derivatives thereof as presented in U. S. Patent No. 4,153, 679, incorporated herein by reference: A compound of Formula III wherein Rl° is selected from the group consisting of H and a Cl-ces alkyl including, but not limited to, CH3, C2Hs, CH3-CH-CH3 (isopropyl) and CH3-C=CH2 (isopropenyl); R"is selected from the group consisting of OH, carboxamide, butanamide, propanetriol, and CONR'R", wherein R'is selected from the group consisting of H, CH3, C2Hs, C4H8 (cyclobutyl), and C4H8O, and wherein R"is selected from the group consisting of C2H500CH2, C2H5, CH3-CH-CH3 (isopropyl), HOCH2C (CH3) 2, HOCH2CH2, C4H9 (tertbutyl), and C4H9 (secbutyl) ; RI2 is selected from the group consisting of H and a Cl-Cs alkyl including, but not limited to, CH3, CH3-CH-CH3 (isopropyl), CH3-C=CH2 (isopropenyl), C4H9 (secbutyl), C4H9 (isobutyl), C4H9 (n-butyl), and CsHll (isohexyl) ; and R13 is selected from the group consisting of H and a Cl-C5 alkyl including, but not limited to, CH3, C2Hs, CH3-CH-CH3 (isopropyl), CH3-C=CH2 (isopropenyl), C4H9 (secbutyl), and C4H9 (isobutyl).

Compounds of Formula III are exemplified herein by WS-23 (2-Isopropyl-N, 2,3- trimethylbutyramide aka N, 2, 3-trimethyl-2-isopropyl butamide).

In another embodiment, the present invention further provides the. following 3- substituted-p-menthane Trp-p8 modulators and derivatives thereof exemplified by those presented in U. S. Patent No. 4,150, 052, incorporated herein by reference: A compound of Formula IV wherein Rl4 is selected from the group consisting of H and an aliphatic group containing up to 25 carbon atoms; Rls is selected from the group consisting of H, OH and an aliphatic group containing up to 25 carbon atoms, with the proviso that when R15 is H, R14 may also be an aryl group of up to 10 carbon atoms and selected from the group consisting of substituted phenyl, phenalkyl, substituted phenalkyl, naphthyl, substituted naphthyl, and pyridyl; and R and R, when taken together with the nitrogen atom to which they are attached, may form a cyclic or heterocyclic group of up to 25 carbon atoms, e. g., a piperidino or a morpholino group. Exemplary such cyclical groups may be selected from the group consisting of 3-phenyl-piperidin-1-yl, 3-phenyl-pyrrolidin-1-yl, 6,7-dimethoxy- 1-methyl-3, 4-dihydro-lH-isoquinolin-2-yl, and 4-pyrimidin-2-yl-piperazin-1-yl.

Typical values for R14 and R15 when aliphatic are methyl, ethyl, propyl, butyl, isobutyl, n-decyl, cyclopropyl, cyclohexyl, cyclopentyl, cycloheptylmethyl, 2- hydroxyethyl, 3-hydroxy-n-propyl, 6-hydroxy-n-hexyl, 2-aminoethyl, 2-acetoxyethyl, 2- ethylcarboxyethyl, 4-hydroxybut-2-ynyl, and carboxymethyl.

When R14 is aryl, typical values are benzyl, naphthyl, 4-methoxyphenyl, 2- methoxy-4-methoxyphenyl, 3-methoxy-5-methoxyphenyl, 4-methyl-5-chlorophenyl, 4- hydroxyphenyl, 4-methylphenyl, 3-methyl-4-methylphenyl, 3-hydroxy-4-methylphenyl, 4- fluorophenyl, 4-chlorophenyl, 4-bromophenyl, 4-ethylphenyl, 2-fluoro-4-fluorophenyl, 4- nitrophenyl, 2-hydroxynaphthyl, pyridyl, [1-carbamoyl-2-(lH-indol-3-yl)-ethyl, 1,3- dioxo-2, 3-dihydro-lH-isoindol-4-yl, 1, 3-dioxo-2, 3-dihydro-lH-isoindol-5-yl, 1-

carbamoyl-2- (4-hydroxyphenyl) ethyl, 1-carbamoyl-2-phenylethyl, 1-hydroxymethyl-2- (lH-indol-3-yl) ethyl, 1-hydroxymethyl-2- (4-chlorophenyl) ethyl, 1-hydroxymethyl-2- (4- hydroxyphenyl) ethyl, 1-hydroxymethyl-2-hydroxy-2-phenylethyl, 1-hydroxymethyl-2- phenyl ethyl, 1-methoxymethyl-2-phenylethyl, 1-methyl-2- (4-chlorophenyl)-ethyl, 1- methyl-2- (5-fluoro-lH-indol-3-yl)-ethyl, 1-methyl-2-hydroxy-2-phenylethyl, 1-oxo-2- phenylethyl, 1-phenyl-cyclopentylmethyl, 2-(1-cyclopentyl-pyrrolidin-3-yl)-ethyl, 2-(1 H- indol-3-yl) ethyl, 2- (2, 3-dimethoxyphenyl) ethyl, 2- (2, 4-dichlorophenyl) ethyl, 2- (2, 4- dimethylphenyl) ethyl, 2-(2, 5-dimethoxyphenyl) ethyl, 2-(2, 5-dimethylphenyl) -ethyl, 2- (2,6-dimethylphenyl) ethyl, 2- (2-chloro-6-flourophenyl) ethyl, 2- (2-chlorophenyl) ethyl, 2- (2-flourophenyl) ethyl, 2- (2-furyl) ethyl, 2- (2-methoxy-5-bromophenyl) ethyl, 2- (2- methoxyphenyl) -ethyl, 2- (2-methylphenyl) ethyl, 2- (3, 4-dichlorophenyl) ethyl, 2- (3, 4- dimethoxyphenyl) ethyl, 2- (3, 5-dimethoxyphenyl) ethyl, 2- (3-bromo-4-methoxyphenyl) ethyl, 2- (3-chlorophenyl) ethyl, 2- (3-ethoxyphenyl) ethyl, 2- (3-flourophenyl) ethyl, 2- (3- hydroxy-4-methoxyphenyl)-ethyl, 2- (3-hydroxyphenyl) ethyl, 2- (3-methoxy-4- ethoxyphenyl) ethyl, 2- (3-methoxy-4-hydroxyphenyl) ethyl, 2- (3-methoxyphenyl) ethyl, 2- (3-methylphenyl) ethyl, 2- (3-triflouromethylphenyl) ethyl, 2- (4-bromophenyl)-ethyl, 2- (4- chlorophenyl) ethyl, 2- (4-ethylphenyl) ethyl, 2- (4-fluorophenyl) ethyl, 2- (4- hydroxyphenyl) ethyl, 2- (4-methoxy-phenyl)-2-oxo-ethyl, 2- (4-methoxyphenyl) ethyl, 2- (4- methylphenyl) ethyl, 2- (4-methylphenyl) ethyl, 2- (4-methylsulfanylphenyl) ethyl, 2- (4- nitrophenyl) ethyl, 2- (4-sulfamoyl-phenyl)-ethyl, 2- (5-methoxy-lH-indol-3-yl)-ethyl, 2- (6- <BR> <BR> <BR> <BR> fluoro-lH-indol-3-yl)-ethyl, 2- (6-methoxy-lH-indol-3-yl)-ethyl, 2- (7-methyl-lH-indol-3- yl) -ethyl, 2- (N, N-dipropylamino) ethyl, 2- (pyridin-2-yl)-ethyl, 2- (pyridin-3-yl)-ethyl, 2- (pyridin-4-yl)-ethyl, 2,2-diphenylethyl, 2,3-diflourobenzyl, 2,3-dimethoxyphenyl, 2,4- dichlorophenyl, 2,4-diflourophenyl, 2,4-dimethoxyphenyl, 2,4-dimethylphenyl, 2-bromo- 4-methylphenyl, 2-chloro-4-cyanophenyl, 2-chloro-4-flourophenyl, 2-chloro-4- iodophenyl, 2-chloro-4-nitrophenyl, 2-chloro-5-nitrophenyl, 2-chlorophenyl, 2-cyclohex- 1-enyl-ethyl, 2-flouro-4-chlorophenyl, 2-flouro-5-nitrophenyl, 2-hydroxy-2- (3- hydroxyphenyl) ethyl, 2-hydroxy-2- (4-hydroxyphenyl) ethyl, 2-hydroxy-2-phenylethyl, 2- iodophenyl, 2-methyl-1, 3-dioxo-2, 3-dihydro-lH-isoindol-5-yl, 2-methyl-2-phenylethyl, 2- methyl-4-broophenyl, 2-methyl-5-nitrophenyl, 2-methylphenyl, 2-nitro-4-flourophenyl, 2- nitrophenyl, 2-oxo-2, 3-dihydro-lH-benzoimidazol-5-yl, 2-oxo-2-phenylethyl, 2-phenyl- 1H-benzoimidazol-5-yl, 2-phenyl-2- (4-chlorophenyl) ethyl, 2-phenyl-2- (4-flourophenyl)- ethyl, 2-phenyl-2- (4-methoxyphenyl) ethyl, 2-phenylethenyl, 2-phenylethyl, 2-pyridin-2- 11311. 100PCT 9

yl-benzooxazol-5-yl, 2-pyridin-3-yl-lH-benzoimidazol-5-yl, 2-thiophen-2-yl-ethyl), 2- trilfouromethyl-1H-benzoimidazol-5-yl, 3,4, 5-triflourophenyl, 3,4, 5-trimethoxyphenyl, 3,4-cyclopentanephenyl, 3,4-dichlorophenyl, 3,4-dimethylphenyl, 3,5-dimethoxyphenyl, 3-acetamidophenyl, 3-bromo-4-methylphenyl, 3-carboxamidophenyl, 3-chloro-4- methoxyphenyl, 3-chloro-4-methylphenyl, 3-chloro-4-morpholin-4-yl-phenyl, 3- hydroxymethylphenyl, 3-nitrophenyl, 3-oxo-indan-5-yl, 3-phenylpropyl, 3-yl- acetophenone, 4-(1, 1-dioxo-116-thiomorpholin-4-ylmethyl)-phenyl, 4-(1- hydroxyethyl) phenyl, 4- (2-hydroxyethyl) phenyl, 4- (4-ethyl-piperazin-1-yl)-phenyl, 4- (4- methyl-lH-benzoimidazol-2-yl)-phenyl, 4- (4-methyl-piperazin-1-ylmethyl)-phenyl, 4- (morpholine-4-sulfonyl)-phenyl, 4- [1, 3-dioxo-2- (2-trifluoromethyl-phenyl)-2, 3-dihydro- lH-isoindol-5-yloxy]-phenyl, 4- [2- (2-methoxy-phenyl)-1, 3-dioxo-2, 3-dihydro-lH- isoindol-5-yloxy] -phenyl, 4- {2- [2- (3, 4-dimethoxy-phenyl)-ethyl]-1, 3-dioxo-2, 3-dihydro- lH-isoindol-5-yloxy}-phenyl, 4-acetylphenyl, 4-azepan-1-ylmethyl-phenyl, 4- benzooxazol-2-yl-phenyl, 4-Biphenyl, 4-bromophenyl, 4-carboxamidophenyl, 4- chlorophenyl, 4-cyanophenyl, 4-ethoxyphenyl, 4-ethylphenyl, 4-fluorophenyl, 4- hydroxymethylphenyl, 4-methoxyphenyl, 4-methyl-2-oxo-2H-chromen-7-yl, 4- methylcyclohexyl, 4-methylphenyl, 4-methylsulfanylphenyl, 4-nitrobenzyl, 4-pyrrolidin- 1-ylmethyl-phenyl, 4-triflouromethylphenyl, benzo [1, 3] dioxol-5-yl, benzoylamino, benzyloxy, bicyclo [2.2. 1] hept-2-yl, C-lH-indazol-5-yl, cycloheptyl, indan-2-yl, N- (2- diethylamino-ethyl) -benzamide-4-yl, N'-quinoxalin-2-yl-amino, and phenylcyclopropyl.

Trp-p8 modulators of Formula IV are exemplified herein by the Trp-p8 agonists WS-3 (Millenium Chemicals; N-Ethyl-p-menthane-3-carboxamide aka cyclohexanecarboxamide, N-ethyl-5-methyl-2 (1-methylethyl)) and by WS-12 (N- (4- methoxyphenyl) -p-menthan-3-carboxamide aka cyclohexanecarboxamide, N- (4- methoxyphenyl) -5-methyl-2 (1-methylethyl)), and the compounds presented in Table 1.

In a further embodiment, the present invention provides the following Trp-p8 modulators and derivatives thereof that comprise at least one ketal moiety, including, but not limited to the Trp-p8 agonists 1-menthone glycerol ketal and 3,3, 5- trimethylcyclohexaone glycerol ketal presented in U. S. Patent No. 5,266, 592, incorporated herein by reference: A compound comprising at least one ketal of Formula V wherein R16 is selected from the group consisting of a C2-C6 alkylene group having at least one, but not more than three, hydroxyl group (s), preferably one hydroxyl group; and either R17 and R", independently of one another, represent Cl-Clo-alkyl which is optionally substituted by 1 to 3 groups selected from the group consisting of hydroxyl, amino, thio, and halogen (e. g. , fluorine, chlorine, bromine, or iodine), Cs-C7-cycloalkyl, preferably cyclohexyl, C6-Cz2-aryl, preferably phenyl, with the proviso that the total of the C atoms of Rl7 and Rl8 is not less than 3; or Rl7 and RI8 together represent an alkylene group that, together with the carbon atom that carries the groups Rl7 and Rl8, forms a 5-7-membered ring, it being possible for this alkylene group, in turn, to be substituted by Cl-C6-alkyl groups.

Preferred groups Rl7 and Rl8 comprise methyl, isopropyl, and tert-butyl.

Trp-p8 modulators comprising a ketal of Formula V include the following compounds wherein R16 is as defined above:

Compounds comprising a ketal of Formula V are exemplified herein by the Trp-p8 agonist Frescolat MGA (Harris & Ford, LLC; Menthone Glycerin Acetal), CHa H- o C CH3 CH3 Frescolat MGA

In yet a further embodiment, the present invention provides Formula VI small- molecule Trp-p8 agonists and derivatives thereof exemplified herein by L-Carvone (Millenium Chemicals; (R) -5-Isopropenyl-2-methyl-2-cyclohexenone p-Mentha-6, 8-dien- 2-one), CH3 - CH3 CH2 L-Carvone

Other embodiments of the present invention provide Trp-p8 modulators of Formula VII. A compound of Formula VII

VII wherein RI is selected from the group consisting of 2-pyridyl, 2-nitro-4- trifluoromethylphenyl, 2-nitro-4-chlorophenyl, 2-methoxyphenyl, 2-chlorophenyl, phenyl, 2-methyl-quinolin-3-yl, 4-methoxyphenyl, 4-fluorophenyl, 3-azepanl-yl-5- (4- trifluoromethoxy) phenylamino [1, 3,5] triazyl, cyclohexyl, diphenylmethyl, 2-phenylethyl, 4-hydroxy-cyclohexyl, cycloheptyl, cyclopentyl, C-benzo [1, 3] dioxol-5-yl-methyl, 2- pyridyl, and 4-chlorobenzyl; Rl8 is selected from the group consisting of 1-benzyl-IH-pyrazolo [3,4- d] pyrimidin-4-yl, 3-benzylamino-2-nitrophenyl, 5-nitro-quinolin-8-yl, 1-yl-3- (2- isopropyl-5-methyl-cyclohexyloxy) -propan-2-ol, 1-phenyl-lH-pyrazolo [3,4-d] pyrimidin- 4-yl, benzyl-2-methyl-quinazolin-4-yl, 3-methyl-5-morpholin-4-yl-2-nitro-phenyl, 2-nitro- 5-piperazin-1-yl-ethanol, 1-yl-3- (2-isopropyl-5-methyl-cyclohexyloxy)-propan-2-ol, 4- (2, 5-dimethyl-pyrrol-1-yl)-2-nitro-phenyl, 2-nitro-3-trifluoromethanesulfonyl-phenyl, 1- phenyl-lH-pyrazolo [3,4-d] pyrimidin-4-yl, 2-(2-Fluoro-phenoxymethyl)-2-cyano oxazolyl, adamantly, 5- (benzo [1, 3] dioxol-5-ylamino)-10b, 10c-dihydro-anthra [l, 9-cd] isoxazol-6- one-yl, 2-methyl-thiazolo [3,2-b] [1, 2,4] triazol-6-ol 4-methylphenyl methyl, 3-benzyl-3H- quinazolin-4-one-2-yl, cyclopentyl, tetrahydronapthyl, cyclooctyl, cyclohexyl, C- [3- (4- chloro-phenyl)-2, 5-dimethyl-pyrazolo [1, 5-a] pyrimidin-7-yl]-methyl, C- (2-benzyl-5, 6,7, 8- tetrahydro-benzo [4,5] thieno [2,3-d] pyrimidin-4-yl) -methyl, and 1-yl-3- (2-isopropyl-5- methyl-cyclohexyloxy) -propan-2-ol ; Rl9 and R20 are each independently selected from the group consisting of H and O ; and R2'is selected from the group consisting of 4-methylphenyl, 2-chloro-4- fluorophenyl, and 4-chlorophenyl.

In other embodiments, the present invention provides Formula VIM small- molecule Trp-p8 modulators. A compound of Formula VIII

VIII wherein R22 is a linker moiety, which may be selected from the group consisting of oxyacetamide, urea, carbamate, thiourea, sulfonamide, amine, amide. Formula VIM antagonists are represented by the following sub-formulae (Formula VIII-A, Formula VIII-B, Formula VIII-C, Formula VE-D, Formula VIII-E, Formula VIII-F, and Formula VIII-G) :

VIII-A VIII-B

VIII-G Irrespective of which of the seven R22 linker moieties is employed, R23 is selected from the group consisting of H, tetrahydro isoquinolinyl, tetrahydro quinolinyl, 3-methyl indolinyl, indolinyl, 2- (N-methyl, N-phenylethyl) amino ethyl, 3-methyl indolinyl, 1- phenyl ethyl, 2-chloro benzyl, 2-methoxybenzyl, 2-methoxyphenyl, 2-cyclohex-1-enyl ethyl, (1-phenyl-cyclophentyl)-methyl, 2-(tetrahydroquinolinyl)-ethyl, 1-propyl-1, 2,3, 4- tetrahydro-pyrrolo [1, 2-a] pyrazine, cycloheptyl, 3-cyclohexylsulfanylpropyl, 2-cyclohex-1- enyl ethyl, 2- (N-isopropyl, N-phenylethyl) amino ethyl, 1-methyl-1, 2,3, 4-tetrhydro- pyrrolo [1, 2-a] pyrazine, 2-cyclopentylethyl, 2-phenylcyclopropyl, 1-phenoxyethyl, 4- butyloxyphenyl, (2-nitrophenoxy) methyl, 4,7, 7-trimethyl-2-oxa-bicyclo [2.2. 1] heptan-3- one, C-(l-phenyl-5-propyl-lH-pyrazol-4-yl)-methyl, benzyl, 2-chlorobenzyl, 1- [3- (6, 7- dimethoxy-l-methyl-3, 4-dihydro-lH-isoquinolin-2-ylmethyl)-4-methoxy-phenyl]-2, 3,4, 9- <BR> <BR> <BR> tetrahydro-1H-b-carboline, C- [3- (4-butoxy-phenyl)-1H-pyrazol-4-yl]-methyl, 4- (azepane- l-sulfonyl)-phenyl, and 5- (7-chloro-quinolin-4-ylsulfanyl)- [1, 3,4] thiadiazol-2-yl; R24 is selected from the group consisting of H, tetrahydro isoquinolinyl, tetrahydro quinolinyl, 3-methyl indolinyl, indolinyl, 3-methyl indolinyl, 1-propyl-1, 2,3, 4- tetrahydro-pyrrolo [1,2-a] pyrazine, 1-methyl-1,2, 3,4-tetrhydro-pyrrolo [1, 2-a] pyrazine, and 1- [3- (6, 7-dimethoxy-1-methyl-3, 4-dihydro-lH-isoquinolin-2-ylmethyl)-4-methoxy- phenyl] -2, 3,4, 9-tetrahydro-lH-b-carboline ; and R25 is selected from the group consisting of H.

Other aspects of the present invention provide compositions, including pharmaceutical compositions, comprising one or more small-molecule Trp-p8 modulators of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, and Formula VIII in combination with a pharmaceutically acceptable excipient, carrier

and/or diluent. Exemplified herein within the Examples are specific Trp-p8 agonists of Formula I, Formula II, Formula in, Formula IV, Formula V, and Formula VI and Trp-p8 antagonists of Formula VII and Formula VIII. Also provided are Trp-p8 antagonists of Formula I, Formula II, Formula m, Formula IV, Formula V, and Formula VI and Trp-p8 agonists of Formula VII and Formula VIII.

Within still further aspects, compositions of the present invention comprise one or more compound of Formula I, Formula II, Formula HI, Formula IV, Formula V, Formula VI, Formula VII, and/or Formula VIII formulated together with one or more cancer therapeutic agent. Alternatively, compositions of the present invention comprise a compound of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, and/or Formula VIII independently formulated with one or more cancer therapeutic agent. That is, one or more compound of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, and/or Formula VIII and the cancer therapeutic agent are separately formulated.

Suitable cancer therapeutic agents include, but are not limited to, antimitotic agents including, but not limited to, paclitaxel, vincristine, and etoposide; alkylating agents including, but not limited to, mechlorethamine, cyclophosphamide, and carmustine ; antimetabolites including, but not limited to, methotrexate, gemcitabine, lometrexol, 5-fluorouracil, and 6-mercaptopurine ; cytotoxic antibiotics including, but not limited to, doxorubicin, daunorubicin, bleomycin, mitomycin C, and streptozocin; platinum agents including, but not limited to, cisplatin and carboplatin ; hormonal agents including, but not limited to, anti-estrogens such as tamoxifen and d iethylstilbestrol as well as anti-androgens such as flutamide; antiangiogenesis agents; and farnesyl transferase inhibitors.

In certain aspects, compounds of Formula I, Formula II, Formula m, Formula IV, Formula V, Formula VI, Formula VII, and/or Formula VIII are administered in combination with cancer therapeutic agents that are themselves ineffective for modulating Trp-p8 activity in a cell expressing Trp-p8. Surprisingly, these types of combination therapy result in enhanced efficacy relative to the use of a single compound of Formula I, Formula II, Formula m, Formula IV, Formula V, Formula VI, Formula VII, and/or Formula VIII alone.

In other aspects, compounds of Formula I, Formula II, Formula m, Formula IV, Formula V, Formula VI, Formula VII, and/or Formula VIII are administered in 11311. 100PCT 17

combination with one or more additional Trp-p8 modulator including, but not limited to, a compound of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, and/or Formula VIII.

Within certain of these embodiments are provided small-molecule antagonists of the small-molecule Trp-p8 agonists presented herein. Thus, within certain embodiments are provided small-molecule Trp-p8 antagonists of Formula I, Formula II, Formula m, Formula IV, Formula V, Formula VI, Formula VII, and/or Formula VIII, and derivatives thereof, of one or more Formula I, Formula II, Formula m, Formula IV, Formula V, Formula VI, Formula VII, and/or Formula VIII Trp-p8 agonist.

Further embodiments of the present invention provide methods for decreasing cell viability and/or inhibiting cell growth, methods for stimulating cation influx, and methods for inducing apoptosis and/or necrosis in a cell expressing Trp-p8. Exemplary such methods comprise the step of contacting a cell with a compound of Formula I, Formula II, Formula in. Formula IV, Formula V, Formula VI, Formula VII, and/or Formula VIII in a concentration and for a time required to decrease cell viability and/or inhibit cell growth, to raise intracellular calcium, and/or to induce apoptosis and/or necrosis of the cell.

In still further embodiments, the present invention provides methods for treating a disease in a mammal, most typically a human, by administering one or more compound and/or composition of the present invention. In certain aspects, the methods include the administration of a composition comprising a combination of a compound of Formula I, Formula II, Formula m, Formula IV, Formula V, Formula VI, Formula VII, and/or Formula VIII with one or more cancer therapeutic agent delivered in a simultaneous manner, such as in a single formulation. In certain other aspects, the methods of the present invention include combination therapy wherein the compound of Formula I, Formula II, Formula m, Formula IV, Formula V, Formula VI, Formula VII, and/or Formula VIII is administered first in one formulation, followed by the cancer therapeutic agent in a separate formulation. The methods also include a cancer therapeutic agent being delivered first in one formulation, followed by a compound of Formula I, Formula II, Formula m, Formula IV, Formula V, Formula VI, Formula VII, and/or Formula VIII in a separate formulation.

Therapeutic methods of the present invention are particularly effective in the treatment of cancers associated with the expression of Trp-p8 including, but not limited to, certain colon, lung, breast, and prostate cancers.

11311. 100PCT 18

The above-mentioned and additional features of the present invention and the manner of obtaining them will become apparent, and the invention will be best understood by reference to the following more detailed description, read in conjunction with the accompanying figures.

Brief Description of the Figures Figure 1 is a graph depicting an exemplary ATP viability assay. Trp-p8 agonists were tested at 10 p^M and agonist-specific killing of Trp-p8 expressing CHO cells (CHO/Trp-p8) measured at 37°C.

Figures 2A-2C are graphs depicting Trp-p8 agonist-induced increases in intracellular calcium as determined by a calcium flux assay performed at 37°C. Figure 2A is a positive control demonstrating that CHO and CHO/Trp-p8 cells respond similarly to 1 pM Ionomycin at 37°C in the calcium flux assay. Figure 2B is a negative control demonstrating that parental CHO cells that do not express endogenous or exogenous Trp- p8 do not respond to Trp-p8 agonists even at a concentration of 10 J. M. Figure 2C demonstrates that the Trp-p8 agonist, designated herein as compound 1 603, induced a specific, concentration-dependent response in CHO/Trp-p8 cells at 37°C.

Figure 3 are plots of flow cytometry data demonstrating that a Trp-p8 agonist is capable of specifically inducing apoptosis in Trp-p8 expressing CHO cells at 37°C.

Figure 4 is a graph depicting exemplary results from a primary screen for Trp-p8 antagonists using the ATP viability assay, described herein, with CHO/Trp-p8 cells at 37°C. CHO/Trp-p8 cells were exposed to compounds, at different concentrations, in 1% DMSO or 1% DMSO in combination with a toxic concentration of the Trp-p8 agonist 1607. The viability of cells was measured after 24-26 hours at 37°C using the ATP assay.

The compounds D-2258 and D-2212 protected the cells from the toxic effect of the Trp- p8 agonist D-1607 and, consequently, are classified as Trp-p8 antagonists. D-2250 had no protective effect and is shown here for the purpose of illustration of the assay.

Figure 5 is a graph depicting the screening and characterization of Trp-p8 antagonists by the calcium flux assay performed at 37°C. CHO/Trp-p8 cells were loaded with the calcium indicator dye, Fura-2, and the increase in intracellular calcium in response to compounds was determined by the increase in fluorescence. Fura-2 dye loaded CHO/Trp-p8 cells were exposed to 1% DMSO or D-2258, at different concentrations, in 1% DMSO at 3 7°C. Three minutes later, D-1607 was added to the 11311. 100PCT 19

cells. When cells were exposed to effective concentrations of the antagonist, D-2258, their ability to respond to the agonist D-1607 was significantly reduced or eliminated altogether.

SEQ ID NO: 1 is the nucleotide sequence of a human Trp-p8 cDNA (GenBank Accession No. AY090109).

SEQ ID NO: 2 is the amino acid sequence encoded by the nucleotide sequence of SEQ ID NO: 1 (GenBank Accession No. Nu076985).

Detailed Description of the Invention The present invention is based upon the discovery that certain small-molecule agonists of Trp-p8 activity are capable of inhibiting the growth of and/or inducing apoptosis and/or necrosis in cells that express Trp-p8. Without wishing to be limited to any specific mode of action, it is believed that Trp-p8 agonist-mediated activation of the Trp-p8 receptor substantially increases cation influx, which is correlative of cellular toxicity. It is further believed that Trp-p8 antagonists can inhibit the basal level and/or native ligand-induced activity of endogenous Trp-p8 activation which, consequently, leads to reduced growth or death of cells expressing this cation channel protein.

Thus, the present invention provides small-molecule Trp-p8 modulators, including agonists and antagonists of Trp-p8 activity, as well as compositions, including pharmaceutical compositions, comprising one or more small-molecule Trp-p8 modulator in combination with one ormorepharmaceutically acceptable carrier a nd/or e xcipient.

The present invention also provides combination compositions comprising one or more Trp-p8 modulator and one or more additional therapeutic compound such as, for example, a cancer therapeutic agent. Trp-p8 modulators and compositions comprising Trp-p8 modulators will find utility in methods for activating Trp-p8-mediated cation influx in a cell, methods for inducing apoptosis and/or necrosis in a cell, as well as methods for the treatment of diseases associated with Trp-p8 expression including, but not limited to, cancers, such as breast, colon, lung, and prostate cancers.

Definitions The term"Trp-p8 modulators"refers collectively to small-molecule agonists and antagonists that bind to and either increase or decrease, respectively, the activity of Trp-p8 in a cell. Trp-p8 agonists include compounds of Formula I, Formula II, Formula m,

Formula IV, Formula V, Formula VI, Formula VII, and Formula VIII and are exemplified herein by various compounds of Formulas I-VI, and chemical derivatives thereof. Trp-p8 antagonists include compounds of Formula I, Formula II, Formula HI, Formula IV, Formula V, Formula VI, Formula VII, and Formula VIII and are exemplified herein by various compounds of Formulas VII-VIII, and chemical derivatives thereof. Additional Trp-p8 agonists or antagonists of Formula I, Formula II, Formula m, Formula IV, Formula V, Formula VI, Formula VII, and Formula VIII, not specifically exemplified herein, may be readily synthesized and characterized by a skilled artisan by employing the methodology expressly provided herein and/or as is readily available in the art.

The phrase"activate Trp-p8"means agonist-mediated activation of Trp-p8 expressed on the surface of a cell. For example, within certain embodiments, agonists of the present invention, when contacted with a cell and/or administered in vivo to a mammalian subject, activate Trp-p8 thereby facilitating the influx of cations, such as calcium ions, to such an intracellular level and/or for such a duration that is sufficient to cause toxicity to the cell as evidenced by a diminution in cell growth and/or an onset of necrotic and/or apoptotic cell death.

The term"aliphatic"is intended to include any straight-chained, branched- chained, or cyclic group free of aromatic unsaturation, and thus embraces alkyl, cycloalkyl, alkenyl, cycloalkenyl, alkynyl, hydroxyalkyl, acyloxyalkyl, alkoxy, alkoxyalkyl, aminoalkyl, acylaminoalkyl, carboxyalkyl, and similar combinations.

The term"alkyl,"by itself or as part of another substituent, means, unless otherwise stated, a straight or branched chain, or cyclic hydrocarbon group, or combination thereof, which may be fully saturated, mono or polyunsaturated and can include di and multivalent groups, having the number of carbon atoms designated (i. e. Cl- Cl0 means one to ten carbons). Examples of saturated hydrocarbon groups include groups such as methyl, ethyl, n-propyl, isopropyl, n-butyl, t-butyl, isobutyl, sec-butyl, cyclohexyl, (cyclohexyl) ethyl, cyclopropylmethyl, homologs and isomers of, for example, n-pentyl, n- hexyl, n-heptyl, n-octyl, and the like. An unsaturated alkyl group is one having one or more double bonds or triple bonds. Examples of unsaturated alkyl groups include vinyl, 2-propenyl, crotyl, 2-isopentenyl, 2- (butadienyl), 2,4-pentadienyl, 3- (1, 4-pentadienyl), ethynyl, 1-and 3-propynyl, 3-butynyl, and the higher homologs and isomers.

The term"alkenyl"denotes branched or unbranched hydrocarbon chains containing one or more carbon-carbon double bonds.

The term"alkynyl"refers to branched or unbranched hydrocarbon chains containing one or more carbon-carbon triple bonds.

The term"alkylene"by itself or as part of another substituent means a divalent group derived from an alkane, as exemplified by--CH2CH2CH2CH2--. Typically, an alkylene group will have from 1 to 24 carbon atoms, with those groups having 10 or fewer carbon atoms being preferred in the present invention. A"lower alkyl"or"lower alkylene"is a shorter chain alkyl or alkylene group, generally having eight or fewer carbon atoms.

The term"cycloalkylene"by itself or as part of another substituent means a divalent group derived from a cycloalkane, as exemplified by cyclohexylene. Typically, a cycloalkylene group will have from 5-8 carbon atoms, with those groups having 6 carbon atoms being preferred in the present invention.

The term"alkenylene"by itself or as part of another substituent means a divalent group derived from an alkenyl, as exemplified by-CH=CHCH2CH2--. Typically, alkenylene groups will have from 2 to 24 carbon atoms, with those groups having 10 or fewer carbon atoms being preferred in the present invention.

The terms"alkoxy, ""alkylamino"and"alkylthio"refer to those groups having an alkyl group attached to the remainder of the molecule through an oxygen, nitrogen or sulfur atom, respectively. Similarly, the term"dialkylamino"is used in a conventional sense to refer to--NR'R"wherein the R groups can be the same or different alkyl groups.

The term"heteroalkyl,"by itself or in combination with another term, means, unless otherwise stated, a stable straight or branched chain, or cyclic hydrocarbon, or combinations thereof, fully saturated or containing from 1 to 3 degrees of unsaturation, consisting of the stated number of carbon atoms and from one to three heteroatoms selected from the group consisting of O, N, Si and S, and wherein the nitrogen and sulfur atoms may optionally be oxidized and the nitrogen heteroatom may optionally be quaternized. The heteroatom (s) O, N and S may be placed at any interior position of the heteroalkyl group. The heteroatom Si may be placed at any position of the heteroalkyl group, including the position at which the alkyl group is attached to the remainder of the molecule. Examples include--CH2--CH2--O--CH3,--CH2--CH2--NH--CH3,--CH2--CH2-- N (CH3-)-CH3,-CH2-S-CH2-CH3,-CH2-CH2-S (0)-CH3,-CH2-CH2-S (0) 2-CH3, --CH=CH--O--CH3,--Si (CH3) 3,--CH2-CH=N--OCH-3, and-CH=CH--N (CH3)--CH3.

Up to two heteroatoms may be consecutive, such as, for example,--CH2--NH--OCH3 and 11311. 100PCT 22

--CH2--O--Si (CH3) 3. Also included in the term"heteroalkyl"are those groups described in more detail below as"heterocycloalkyl. "The term"heteroalkylene"by itself or as part of another substituent means a divalent group derived from heteroalkyl, as exemplified by --CH2--CH2--S--CH2CH2--and--CH2--S--CH2CH2--NH--CH2--. For heteroalkylene groups, heteroatoms can also occupy either or both of the chain termini. Still further, for alkylene and heteroalkylene linking groups, no orientation of the linking group is implied.

The term"acyl"refers to those groups derived from an organic acid by removal of the hydroxy portion of the acid. Accordingly, acyl is meant to include, for example, acetyl, propionyl, butyryl, decanoyl, pivaloyl, benzoyl and the like.

An"activated carbonyl"group is a carbonyl group whose electrophilicity is enhanced as a result of the groups attached to either side of the carbonyl. Examples of such activated carbonyl groups are (polyfluoroalkyl) ketones, (polyfluoroalkyl) aldehydes, alpha-keto esters, alpha-keto acids, alpha-keto amides, 1,2-diketones, 2-acylthiazoles, 2- acylimidazoles, and the like.

The terms"cycloalkyl"and"heterocycloalkyl", by themselves or in combination with other terms, represent, unless otherwise stated, cyclic versions of"alkyl"and "heteroalkyl", respectively. Additionally, for heterocycloalkyl, a heteroatom can occupy the position at which the heterocycle is attached to the remainder of the molecule.

Examples of cycloalkyl include cyclopentyl, cyclohexyl, 1-cyclohexenyl, 3-cyclohexenyl, cycloheptyl, and the like. Examples of heterocycloalkyl include 1- (1, 2,5, 6- tetrahydropyridyl), 1-piperidinyl, 2-piperidinyl, 3-piperidinyl, 4-morpholinyl, 3- morpholinyl, tetrahydrofuran-2-yl, tetrahydrofuran-3-yl, tetrahydrothien-2-yl, tetrahydrothien-3-yl, 1-piperazinyl, 2-piperazinyl, and the like.

The t erms"h alo"o r"halogen,"b y themselves o r as part of another substituent, mean, unless otherwise stated, a fluorine, chlorine, bromine, or iodine atom. Additionally, terms such as"fluoroalkyl,"are meant to include monofluoroalkyl and polyfluoroalkyl.

The term"aryl, "employed alone or in combination with other terms (e. g., aryloxy, arylthioxy, arylalkyl) means, unless otherwise stated, an aromatic substituent which can be a single ring or multiple rings (up to three rings) which are fused together or linked covalently. The term"heteroaryl"is meant to include those aryl rings which contain from zero to four heteroatoms selected from N, O, and S, wherein the nitrogen and sulfur atoms are optionally oxidized, and the nitrogen atom (s) are optionally quaternized. The "heteroaryl"groups can be attached to the remainder of the molecule through a 11311. 100PCT 23

heteroatom. Non-limiting examples of aryl and heteroaryl groups include phenyl, 1- naphthyl, 2-naphthyl, 4-biphenyl, 1-pyrrolyl, 2-pyrrolyl, 3-pyrrolyl, 3-pyrazolyl, 2- imidazolyl, 4-imidazolyl, pyrazinyl, 2-oxazolyl, 4-oxazolyl, 2-phenyl-4-oxazolyl, 5- oxazolyl, 3-isoxazolyl, 4-isoxazolyl, 5-isoxazolyl, 2-thiazolyl, 4-thiazolyl, 5-thiazolyl, 2- furyl, 3-furyl, 2-thienyl, 3-thienyl, 2-pyridyl, 3-pyridyl, 4-pyridyl, 2-pyrimidyl, 4- pyrimidyl, 5-benzothiazolyl, purinyl, 2-benzimidazolyl, 5-indolyl, 1-isoquinolyl, 5- isoquinolyl, 2-quinoxalinyl, 5-quinoxalinyl, 3-quinolyl, and 6-quinolyl. Substituents for each of the above noted aryl ring systems are selected from the group of acceptable substituents described below. The term"arylalkyl"is meant to include those groups in which an aryl or heteroaryl group is attached to an alkyl group (e. g., benzyl, phenethyl, pyridylmethyl and the like) or a heteroalkyl group (e. g., phenoxymethyl, 2- pyridyloxymethyl, 3- (l-naphthyloxy) propyl, and the like).

Each of the above terms (e. g.,"alkyl,""heteroalkyl"and"aryl") are meant to include both substituted and unsubstituted forms of the indicated group. Preferred substituents for each type of group are provided below.

Substituents for the alkyl and heteroalkyl groups (including those groups often referred to as alkylene, alkenyl, heteroalkylene, heteroalkenyl, alkynyl, cycloalkyl, heterocycloalkyl, cycloalkenyl, and heterocycloalkenyl) can be a variety of groups selected from :--OR', =O, =NR', =N--OR',--NR'R",--SR',-halogen,--SiR'R"R"',-- OC (O) R',--C (O) R',--CO2R', CONR'R",--OC (O) NR'R",--NR"C (O) R',--NR'-- C (O) NR"R"',--NR"C (0) 2R',--NH--C (NH2) =NH,--NR'C (NH2) =NH,--NH-- C (NH2) =NR',--S (O) R', S (0) 2R',--S (0) 2NR'R",--CN and--NO2 in a number ranging from zero to (2N+1), where N is the total number of carbon atoms in such group. R', R" and R"'each independently refer to hydrogen, unsubstituted (Cl-C8) alkyl and heteroalkyl, unsubstituted aryl, aryl substituted with 1-3 halogens, unsubstituted alkyl, alkoxy or thioalkoxy groups, or aryl- (CI-C4) alkyl groups. When R'and R"are attached to the same nitrogen atom, they can be combined with the nitrogen atom to form a 5-, 6-, or 7- membered ring. For example,--NR'R"is meant to include 1-pyrrolidinyl and 4- morpholinyl. From the above discussion of substituents, one of skill in the art will understand that the term"alkyl"is meant to include groups such as haloalkyl (e. g.,--CF3 and--CH2CF3) and acyl (e. g.,--C (O) CH3,--C (O) CF3,--C (O) CH20CH3, and the like).

Similarly, substituents for the aryl groups are varied and are selected from:- <BR> <BR> <BR> <BR> halogen,--OR',--OC (O) R',--NR'R",--SR',--R',--CN,--N02,--CO2R',--CONR'R",-- 11311. 100PCT 24

C(CH2)=NH, --NR'C(NH2)=NH, --NH--C(NH2)=NR', --S(O)R', --S(O)2R', --S(O)2NR'R'', --NR''--S(O)2--R', --N3, --CH(Ph)2, perfluoro(C1-C4)alkoxy, and perfluoro(C1-C4)alkyl, in a number ranging from zero to the total number of open valences on the aromatic ring system; and where R', R"and R"'are independently selected from hydrogen, (Ci-Cs) alkyl and heteroalkyl, unsubstituted aryl, (unsubstituted aryl)- (CI-C4) alkyl, and (unsubstituted aryl) oxy- (CI-C4) alkyl- Two of the substituents on adjacent atoms of the aryl ring may optionally be replaced with a substituent of the formula--T--C (O)-- (CH2) q--U--, wherein T and U are independently-NH-,-0-,--CH2--or a single bond, and the subscript q is an integer of from 0 to 2. Alternatively, two of the substituents on adjacent atoms of the aryl ring may optionally be replaced with a substituent of the formula--A-- (CH2) r-B--, wherein A and B are independently--CH2--,--O--,--NH--,--S--,--S (O)--,--S (O) 2--,--S (O) 2NR'--or a single bond, and r is an integer of from 1 to 3. One of the single bonds of the new ring so formed may optionally be replaced with a double bond. Alternatively, two of the substituents on adjacent atoms of the aryl ring may optionally be replaced with a substituent of the formula--(CH2) s--X--(CH2) t--, where s and t are independently integers of from 0 to 3, and X is--O--,--NR'--,--S--,--S (O)--,--S (0) 2--, or--S (0) 2NR'--. The substituent R'in--NR'--and--S (0) 2NR'-- is selected from hydrogen or unsubstituted (Cl- C6) alkyl.

As used herein, the term"heteroatom"is meant to include oxygen (O), nitrogen (N), and sulfur (S).

The term"pharmaceutically acceptable salts"is meant to include salts of the active compounds of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, and/or Formula VIII that are prepared with relatively nontoxic acids or bases, depending on the particular substituents found on the compounds described herein.

Examples of pharmaceutically acceptable base addition salts include, but are not limited to, sodium, potassium, calcium, ammonium, organic amino, magnesium salt, or other similar salt. Examples of pharmaceutically acceptable acid addition salts include, but are not limited to, those derived from inorganic acids like hydrochloric, hydrobromic, nitric, carbonic, monohydrogencarbonic, phosphoric, monohydrogenphosphoric, dihydrophosphoric, sulfuric, monohydrogensulfuric, hydriodic, or phosphorous acids and the like, as well as the salts derived from relatively nontoxic organic acids like acetic, 11311. 100PCT 25

propionic, isobutyric, oxalic, maleic, malonic, benzoic, succinic, suberic, fumaric, mandelic, phthalic, benzenesulfonic, p-tolylsulfonic, citric, tartaric, methanesulfonic, and the like.

Small-molecule Modulators of Trp-p8 Activitv Small-molecule Trp-p8 modulators that are suitably employed in the compositions and methods of the present invention are exemplified herein by the following Trop-p8 agonist compounds: Menthol (Sigma-Aldrich, Inc.; St. Louis, Missouri; (2-isopropyl-5- methyl-cyclohexanol) ) ; Frescolat ML (Harris & Ford, LLC; Indianapolis, Indiana ; Menthyl lactate); Frescolat MGA (Harris & Ford, LLC; Menthone Glycerin Acetal) ; L- Menthyl Acetate (Millenium Chemicals; Olympia Fields, Illinois; Cyclohexanol-5- methyl-2- (1-methylethyl)-acetate- [lR- (lalpha, 2beta, 5alpha) ]-) ; L-Carvone (Millenium Chemicals; (R)-5-Isopropenyl-2-methyl-2-cyclohexenone p-Mentha-6, 8-dien-2-one); WS- 3 (Millenium Chemicals; N-Ethyl-p-menthane-3-carboxamide aka cyclohexanecarboxamide, N-ethyl-5-methyl-2 (1-methylethyl)) ; Cooling Agent 10 (Takasago International Corp.; Rockleigh, New Jersey; (l)-Menthoxypropane-1, 2-diol); Coolact P (Takasago International Corp.; (-) -Isopulegol) ; Coolact 38D (Takasago International Corp.) ; Icilin (1- (2-hydroxyphenyl)-4- (3-nitrophenyl)-1, 2,3, 6- tetrahydropyrimidine-2-one aka 3, 4-dihydro-3-(2-hydroxyphenyl)-6-(3-nitrophenyl)-(1X)- pyrimidin-2-one); WS-23 (2-Isopropyl-N, 2, 3-trimethylbutyramide aka N, 2,3-trimethyl-2- isopropyl butamide), and WS-12 (N- (4-methoxyphenyl)-p-menthan-3-carboxamide aka cyclohexanecarboxamide, N- (4-methoxyphenyl)-5-methyl-2 (1-methylethyl)).

The present invention further contemplates that additional Trp-p8 agonists, including derivatives of the compounds of Formulas I, II, m, IV, V, VI, VII, and/or VIII disclosed herein, may also be suitably employed in the compositions and methods of the present invention.

Thus, in one embodiment, the present invention provides the following Trp-p8 modulators and derivatives thereof : A compound of Formula I

wherein Rl is selected from the group consisting of H, OH, CH3, CH3-CH-CH3 (isopropyl), and CH3-C=CH2 (isopropenyl); R2 is selected from the group consisting of H ; R3 is selected from the group consisting of O, OH, acetate, lactate, carboxamide, butanamide, sulphanamide, and propanetriol ; and R4 is selected from the group consisting of CH3-CH-CH3 (isopropyl), isopropane- 2-ol, and CH3-C=CH2 (isopropenyl).

Exemplary compounds of Formula I include the following Trp-p8 agonists:

CH3 CH3 CH3 OH CH3 CH3 CH3 CH3 CH2 CH3 OH Menthol Coolact ) ' CH3 CH3 OH O OH 0-", OH CH3 CH3 CH3 CH3 CoolingAgentlO ,, and

CH3 O 11 0-C-CH3 k CH3 CH3 L-Menthyl Acetate These exemplary Formula I Trp-p8 agonists have the following trade and chemical names: Menthol (2-isopropyl-5-methyl-cyclohexanol) (Sigma-Aldrich, Inc. ; St. Louis, Missouri); Frescolat ML (Harris & Ford, LLC; Indianapolis, Indiana; Menthyl lactate); L- Menthyl Acetate (Millenium Chemicals; Olympia Fields, Illinois; Cyclohexanol-5- methyl-2- (l-methylethyl)-acetate- [lR- (lalpha, 2beta, 5alpha)]-) ; Cooling Agent 10 (Takasago International Corp.; Rockleigh, New Jersey; (l)-Menthoxypropane-1, 2-diol); Coolact P (Takasago International Corp.; (-) -Isopulegol) ; and Coolact 38D (Takasago International Corp.).

In another embodiment, the present invention further provides the following small-molecule Trp-p8 modulators and derivatives thereof : A compound of Formula II wherein Rs is selected from the group consisting of H, OH, CH3, CH3-CH-CH3 (isopropyl), and CH3-C=CH2 (isopropenyl); R6 is selected from the group consisting of N ; R7 is selected from the group consisting of O and N; R8 is selected from the group consisting of NH, O, and S; and R9 is selected from the group consisting of NO2.

Compounds of Formula n are exemplified herein by the Trp-p8 agonist Icilin (1- (2-hydroxyphenyl)-4- (3-nitrophenyl)-1, 2,3, 6-tetrahydropyrimidine-2-one aka 3,4-dihydro- 3- (2-hydroxyphenyl)-6- (3-nitrophenyl)- (lH)-pyrimidin-2-one).

In another embodiment, the present invention further provides the following acyclic carboxamide Trp-p8 modulators and derivatives thereof as presented in U. S.

Patent No. 4,153, 679, incorporated herein by reference: A compound of Formula III

wherein Rl° is selected from the group consisting of H and a Cl-C5 alkyl including, but not limited to, CH3, C2H5, CH3-CH-CH3 (isopropyl), and CH3-C=CH2 (isopropenyl); Rll is selected from the group consisting of OH, carboxamide, butanamide, propanetriol, and CONR'R", wherein R'is selected from the group consisting of H, CH3, C2H5, C4H8 (cyclobutyl), and C4H8O, and wherein R"is selected from the group consisting of C2H5OOCH2, C2H5, CH3-CH-CH3 (isopropyl), HOCH2C (CH3) 2, HOCH2CH2, C4H9 (tertbutyl), C4H9 (secbutyl); R12 is selected from the group consisting of H and a Cl-C5 alkyl including, but not limited to, CH3, CH3-CH-CH3 (isopropyl), and CH3-C=CH2 (isopropenyl), C4H9 (secbutyl), C4H9 (isobutyl), C4H9 (n-butyl), C5Hll (isohexyl); and Rl3 is selected from the group consisting of H and a Cl-C5 alkyl including, but not limited to, CH3, C2H5, CH3-CH-CH3 (isopropyl), CH3-C=CH2 (isopropenyl), C4H9 (secbutyl), C4H9 (isobutyl).

Compounds of Formula III are exemplified herein by the Trp-p8 agonist WS-23 (2-Isopropyl-N, 2,3-trimethylbutyramide aka N, 2, 3-trimethyl-2-isopropyl butamide).

In another embodiment, the present invention further provides the following 3- substituted-p-menthane Trp-p8 modulators and derivatives thereof as presented in U. S.

Patent No. 4,150, 052, incorporated herein by reference: A compound of Formula IV

wherein R14 is selected from the group consisting of H and an aliphatic group containing up to 25 carbon atoms; Rl5 is selected from the group consisting of H, OH and an aliphatic group containing up to 25 carbon atoms, with the proviso that when Rl5 is H, R14 may also be an aryl group of up to 10 carbon atoms and selected from the group consisting of substituted phenyl, phenalkyl, substituted phenalkyl, naphthyl, substituted naphthyl, and pyridyl; and R and R, when taken together with the nitrogen atom to which they are attached, may form a cyclic or heterocyclic group of up to 25 carbon atoms, e. g., a piperidino or a morpholino group. Exemplary such cyclical groups may be selected from the group consisting of 3-phenyl-piperidin-1-yl, 3-phenyl-pyrrolidin-1-yl, 6,7-dimethoxy- 1-methyl-3, 4-dihydro-1H-isoquinolin-2-yl, and 4-pyrimidin-2-yl-piperazin-1-yl.

Typical values for R14 and Rl5 when aliphatic are methyl, ethyl, propyl, butyl, isobutyl, n-decyl, cyclopropyl, cyclohexyl, cyclopentyl, cycloheptylmethyl, 2- hydroxyethyl, 3-hydroxy-n-propyl, 6-hydroxy-n-hexyl, 2-aminoethyl, 2-acetoxyethyl, 2- ethylcarboxyethyl, 4-hydroxybut-2-ynyl, and carboxymethyl.

When R14 is aryl, typical values are benzyl, naphthyl, 4-methoxyphenyl, 2- methoxy-4-methoxyphenyl, 3-methoxy-5-methoxyphenyl, 4-methyl-5-chlorophenyl, 4- hydroxyphenyl, 4-methylphenyl, 3-methyl-4-methylphenyl, 3-hydroxy-4-methylphenyl, 4- fluorophenyl, 4-chlorophenyl, 4-bromophenyl, 4-ethylphenyl, 2-fluoro-4-fluorophenyl, 4- nitrophenyl, 2-hydroxynaphthyl, pyridyl, [1-carbamoyl-2-(lH-indol-3-yl)-ethyl, 1,3- dioxo-2, 3-dihydro-lH-isoindol-4-yl, 1, 3-dioxo-2, 3-dihydro-lH-isoindol-5-yl, 1- carbamoyl-2- (4-hydroxyphenyl) ethyl, 1-carbamoyl-2-phenylethyl, 1-hydroxymethyl-2- (lH-indol-3-yl) ethyl, 1-hydroxymethyl-2- (4-chlorophenyl) ethyl, 1-hydroxymethyl-2- (4- hydroxyphenyl) ethyl, 1-hydroxymethyl-2-hydroxy-2-phenylethyl, 1-hydroxymethyl-2- phenyl ethyl, 1-methoxymethyl-2-phenylethyl, 1-methyl-2- (4-chlorophenyl)-ethyl, 1- methyl-2- (5-fluoro-lH-indol-3-yl)-ethyl, 1-methyl-2-hydroxy-2-phenylethyl, 1-oxo-2- phenylethyl, 1-phenyl-cyclopentylmethyl, 2- (1-cyclopentyl-pyrrolidin-3-yl)-ethyl, 2- (1H- indol-3-yl) ethyl, 2- (2, 3-dimethoxyphenyl) ethyl, 2- (2, 4-dichlorophenyl) ethyl, 2- (2, 4- dimethylphenyl) ethyl, 2- (2, 5-dimethoxyphenyl) ethyl, 2- (2, 5-dimethylphenyl) -ethyl, 2- (2, 6-dimethylphenyl) ethyl, 2- (2-chloro-6-flourophenyl) ethyl, 2- (2-chlorophenyl) ethyl, 2- (2-flourophenyl) ethyl, 2- (2-furyl) ethyl, 2- (2-methoxy-5-bromophenyl) ethyl, 2- (2- methoxyphenyl) -ethyl, 2- (2-methylphenyl) ethyl, 2- (3, 4-dichlorophenyl) ethyl, 2- (3, 4- dimethoxyphenyl) ethyl, 2- (3, 5-dimethoxyphenyl) ethyl, 2- (3-bromo-4-methoxyphenyl) ethyl, 2- (3-chlorophenyl) ethyl, 2- (3-ethoxyphenyl) ethyl, 2- (3-flourophenyl) ethyl, 2- (3- hydroxy-4-methoxyphenyl) -ethyl, 2- (3-hydroxyphenyl) ethyl, 2- (3-methoxy-4- ethoxyphenyl) ethyl, 2- (3-methoxy-4-hydroxyphenyl) ethyl, 2- (3-methoxyphenyl) ethyl, 2- (3-methylphenyl) ethyl, 2- (3-triflouromethylphenyl) ethyl, 2- (4-bromophenyl)-ethyl, 2- (4- chlorophenyl) ethyl, 2- (4-ethylphenyl) ethyl, 2- (4-fluorophenyl) ethyl, 2- (4- hydroxyphenyl) ethyl, 2- (4-methoxy-phenyl)-2-oxo-ethyl, 2- (4-methoxyphenyl) ethyl, 2- (4- methylphenyl) ethyl, 2- (4-methylphenyl) ethyl, 2- (4-methylsulfanylphenyl) ethyl, 2- (4- nitrophenyl) ethyl, 2- (4-sulfamoyl-phenyl)-ethyl, 2- (5-methoxy-lH-indol-3-yl)-ethyl, 2- (6- <BR> <BR> <BR> <BR> fluoro-lH-indol-3-yl)-ethyl, 2- (6-methoxy-lH-indol-3-yl)-ethyl, 2- (7-methyl-lH-indol-3- yl) -ethyl, 2- (N, N-dipropylamino) ethyl, 2- (pyridin-2-yl)-ethyl, 2- (pyridin-3-yl)-ethyl, 2- (pyridin-4-yl) -ethyl, 2,2-diphenylethyl, 2,3-diflourobenzyl, 2,3-dimethoxyphenyl, 2,4- 11311. 100PCT 31

dichlorophenyl, 2, 4-diflourophenyl, 2,4-dimethoxyphenyl, 2,4-dimethylphenyl, 2-bromo- 4-methylphenyl, 2-chloro-4-cyanophenyl, 2-chloro-4-flourophenyl, 2-chloro-4- iodophenyl, 2-chloro-4-nitrophenyl, 2-chloro-5-nitrophenyl, 2-chlorophenyl, 2-cyclohex- 1-enyl-ethyl, 2-flouro-4-chlorophenyl, 2-flouro-5-nitrophenyl, 2-hydroxy-2- (3- hydroxyphenyl) ethyl, 2-hydroxy-2- (4-hydroxyphenyl) ethyl, 2-hydroxy-2-phenylethyl, 2- iodophenyl, 2-methyl-1, 3-dioxo-2, 3-dihydro-lH-isoindol-5-yl, 2-methyl-2-phenylethyl, 2- methyl-4-broophenyl, 2-methyl-5-nitrophenyl, 2-methylphenyl, 2-nitro-4-flourophenyl, 2- nitrophenyl, 2-oxo-2, 3-dihydro-lH-benzoimidazol-5-yl, 2-oxo-2-phenylethyl, 2-phenyl- 1H-benzoimidazol-5-yl, 2-phenyl-2- (4-chlorophenyl) ethyl, 2-phenyl-2- (4-flourophenyl)- ethyl, 2-phenyl-2- (4-methoxyphenyl) ethyl, 2-phenylethenyl, 2-phenylethyl, 2-pyridin-2- yl-benzooxazol-5-yl, 2-pyridin-3-yl-lH-benzoimidazol-5-yl, 2-thiophen-2-yl-ethyl), 2- trilfouromethyl-lH-benzoimidazol-5-yl, 3,4, 5-triflourophenyl, 3,4, 5-trimethoxyphenyl, 3,4-cyclopentanephenyl, 3,4-dichlorophenyl, 3, 4-dimethylphenyl, 3,5-dimethoxyphenyl, 3-acetamidophenyl, 3-bromo-4-methylphenyl, 3-carboxamidophenyl, 3-chloro-4- methoxyphenyl, 3-chloro-4-methylphenyl, 3-chloro-4-morpholin-4-yl-phenyl, 3- hydroxymethylphenyl, 3-nitrophenyl, 3-oxo-indan-5-yl, 3-phenylpropyl, 3-yl- acetophenone, 4- (1, 1-dioxo-116-thiomorpholin-4-ylmethyl)-phenyl, 4- (1- hydroxyethyl) phenyl, 4- (2-hydroxyethyl) phenyl, 4- (4-ethyl-piperazin-1-yl)-phenyl, 4- (4- methyl-1H-benzoimidazol-2-yl)-phenyl, 4- (4-methyl-piperazin-1-ylmethyl)-phenyl, 4- (morpholine-4-sulfonyl)-phenyl, 4- [1, 3-dioxo-2- (2-trifluoromethyl-phenyl)-2, 3-dihydro- 1H-isoindol-5-yloxy]-phenyl, 4- [2- (2-methoxy-phenyl)-1, 3-dioxo-2, 3-dihydro-lH- isoindol-5-yloxy]-phenyl, 4- {2- [2- (3, 4-dimethoxy-phenyl)-ethyl]-1, 3-dioxo-2,3-dihydro- lH-isoindol-5-yloxy}-phenyl, 4-acetylphenyl, 4-azepan-1-ylmethyl-phenyl, 4- benzooxazol-2-yl-phenyl, 4-Biphenyl, 4-bromophenyl, 4-carboxamidophenyl, 4- chlorophenyl, 4-cyanophenyl, 4-ethoxyphenyl, 4-ethylphenyl, 4-fluorophenyl, 4- hydroxymethylphenyl, 4-methoxyphenyl, 4-methyl-2-oxo-2H-chromen-7-yl, 4- methylcyclohexyl, 4-methylphenyl, 4-methylsulfanylphenyl, 4-nitrobenzyl, 4-pyrrolidin- 1-ylmethyl-phenyl, 4-triflouromethylphenyl, benzo [1, 3] dioxol-5-yl, benzoylamino, benzyloxy, bicyclo [2.2. 1] hept-2-yl, C-lH-indazol-5-yl, cycloheptyl, indan-2-yl, N- (2- diethylamino-ethyl) -benzamide-4-yl, N'-quinoxalin-2-yl-amino, and phenylcyclopropyl.

Trp-p8 modulators of Formula IV are exemplified herein by the Trp-p8 agonists WS-3 (Millenium Chemicals; N-Ethyl-p-menthane-3-carboxamide aka cyclohexanecarboxamide, N-ethyl-5-methyl-2 (1-methylethyl)) and by WS-12 (N- (4- 11311. 100PCT 32

methoxyphenyl)-p-menthan-3-carboxamide aka cyclohexanecarboxamide, N-(4- methoxyphenyl) -5-methyl-2 (1-methylethyl)), and the compounds presented in Table 1.

WS-3 WS-12 In a further embodiment, the present invention provides the following Trp-p8 modulators and derivatives thereof that comprise at least one ketal moiety, including, but not limited to the Trp-p8 agonists 1-menthone glycerol ketal and 3,3, 5- trimethylcyclohexaone glycerol ketal presented in U. S. Patent No. 5,266, 592, incorporated herein by reference: A compound comprising at least one ketal of Formula V wherein R16 is selected from the group consisting of a C2-C6 alkylene group having at least one, but not more than 3, hydroxyl group (s), preferably 1 hydroxyl group, and Either R17 and R18 independently of one another represent C-Ct0-alkyl which is optionally substituted by 1 to 3 groups selected from the group consisting of hydroxyl, amino, thio, and halogen (e. g., fluorine, chlorine, bromine, or iodine), C5-C7-cycloalkyl, preferably cyclohexyl, C6-Cz2-aryl, preferably phenyl, with the proviso that the total of the C atoms of RI and R18 is not less than 3, or RI and RI$ together represent an alkylene group that, together with the carbon atom that carries the groups R 17 and R18, forms a 5-7-membered ring, it being possible for this alkylene group, in turn, to be substituted by Cl-C6-alkyl groups.

Preferred groups Rl7 and Rl8 comprise methyl, isopropyl, and tert-butyl.

Compounds comprising a ketal of Formula V include the following Trp-p8 modulator compounds wherein R16 is as defined above:

Compounds comprising a ketal of Formula V are exemplified herein by the Trp-p8 agonist Frescolat MGA (Harris & Ford, LLC; Menthone Glycerin Acetal), CH3 0 i O CH3 CH3 Frescolat MGA

In yet a further embodiment, the present invention provides Formula VI small- molecule Trp-p8 modulators and derivatives thereof exemplified herein by L-Carvone (Millenium Chemicals; (R)-5-Isopropenyl-2-methyl-2-cyclohexenone p-Mentha-6,8-dien- 2-one),

CH3 - , CH3 CH2 L-Carvone Still further embodiments of the present invention provide small-molecule antagonists of the small-molecule Trp-p8 agonists presented herein. Thus, within certain embodiments are provided Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, and/or Formula VIII small-molecule antagonists, and derivatives thereof, of the Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, and/or Formula VIII Trp-p8 agonists disclosed herein above.

Presented herein in the Examples are specific Trp-p8 antagonists that are exemplary of the Trp-p8 modulators of Formula VII wherein R' is selected from the group consisting of 2-pyridyl, 2-nitro-4- trifluoromethylphenyl, 2-nitro-4-chlorophenyl, 2-methoxyphenyl, 2-chlorophenyl, phenyl, 2-methyl-quinolin-3-yl, 4-methoxyphenyl, 4-fluorophenyl, 3-azepanl-yl-5- (4- trifluoromethoxy) phenylamino [1, 3,5] triazyl, cyclohexyl, diphenylmethyl, 2-phenylethyl, 4-hydroxy-cyclohexyl, cycloheptyl, cyclopentyl, C-benzo91, 3] dioxol-5-yl-methyl, 2- pyridyl, and 4-chlorobenzyl ; R'8 is selected from the group consisting of 1-benzyl-lH-pyrazolo [3,4- d] pyrimidin-4-yl, 3-benzylamino-2-nitrophenyl, 5-nitro-quinolin-8-yl, 1-yl-3- (2- isopropyl-5-methyl-cyclohexyloxy)-propan-2-ol, 1-phenyl-1 H-pyrazolo [3,4-d] pyrimidin-

4-yl, benzyl-2-methyl-quinazolin-4-yl, 3-methyl-5-morpholin-4-yl-2-nitro-phenyl, 2-nitro- 5-piperazin-1-yl-ethanol, 1-yl-3- (2-isopropyl-5-methyl-cyclohexyloxy)-propan-2-ol, 4- (2,5-dimethyl-pyrrol-1-yl)-2-nitro-phenyl, 2-nitro-3-trifluoromethanesulfonyl-phenyl, 1- phenyl-lH-pyrazolo [3,4-d] pyrimidin-4-yl, 2- (2-Fluoro-phenoxymethyl)-2-cyano oxazolyl, adamantly, 5- (benzo [1, 3] dioxol-5-ylamino)-1 Ob, 1 Oc-dihydro-anthra [l, 9-cd] isoxazol-6- one-yl, 2-methyl-thiazolo [3,2-b] [1, 2,4] triazol-6-ol 4-methylphenyl methyl, 3-benzyl-3H- quinazolin-4-one-2-yl, cyclopentyl, tetrahydronapthyl, cyclooctyl, cyclohexyl, C- [3- (4- chloro-phenyl) -2, 5-dimethyl-pyrazolo [1, 5-a] pyrimidin-7-yl]-methyl, C- (2-benzyl-5, 6,7, 8- tetrahydro-benzo [4,5] thieno [2,3-d] pyrimidin-4-yl)-methyl, and 1-yl-3- (2-isopropyl-5- methyl-cyclohexyloxy) -propan-2-ol ; Rl9 and R2° are each independently selected from the group consisting of H and O ; and R21 is selected from the group consisting of 4-methylphenyl, 2-chloro-4- fluorophenyl, and 4-chlorophenyl.

In other embodiments, the present invention provides Formula VIII small- molecule modulators, and derivatives thereof, including Formula VIII antagonists of the Formula IV Trp-p8 agonists disclosed herein above and in the Examples.

A compound of Formula VIII VIII wherein R22 is a linker moiety, which may be selected from the group consisting of oxyacetamide, urea, carbamate, thiourea, sulfonamide, amine, amide. Formula VIII antagonists are represented by the following sub-formulae (Formula VIII-A, Formula VIII-B, Formula VIII-C, Formula VE-D, Formula VE-E, Formula VIII-F, and Formula VE-G) :

VIII-A VIII-B

VIII-C VIII-D

VIII-E VIII-F VIII-G Irrespective of which of the seven R22 linker moieties is employed, R23 is selected from the group consisting of H, tetrahydro isoquinolinyl, tetrahydro quinolinyl, 3-methyl indolinyl, indolinyl, 2-(N-methyl, N-phenylethyl) amino ethyl, 3-methyl indolinyl, 1- phenyl ethyl, 2-chloro benzyl, 2-methoxybenzyl, 2-methoxyphenyl, 2-cyclohex-1-enyl ethyl, (1-phenyl-cyclophentyl)-methyl, 2-(tetrahydroquinolinyl)-ethyl, 1-propyl-1, 2,3, 4- tetrahydro-pyrrolo [1, 2-a] pyrazine, cycloheptyl, 3-cyclohexylsulfanylpropyl, 2-cyclohex-1- enyl ethyl, 2- (N-isopropyl, N-phenylethyl) amino ethyl, 1-methyl-1, 2,3, 4-tetrhydro- pyrrolo [1, 2-a] pyrazine, 2-cyclopentylethyl, 2-phenylcyclopropyl, 1-phenoxyethyl, 4- butyloxyphenyl, (2-nitrophenoxy) methyl, 4,7, 7-trimethyl-2-oxa-bicyclo [2.2. 1] heptan-3- one, C- (l-phenyl-5-propyl-lH-pyrazol-4-yl)-methyl, benzyl, 2-chlorobenzyl, 1- [3- (6, 7-

dimethoxy-l-methyl-3, 4-dihydro-lH-isoquinolin-2-ylmethyl)-4-methoxy-phenyl]-2, 3,4, 9- <BR> <BR> <BR> <BR> tetrahydro-lH-b-carboline, C- [3- (4-butoxy-phenyl)-lH-pyrazol-4-yl]-methyl, 4- (azepane- 1-sulfonyl)-phenyl, and 5- (7-chloro-quinolin-4-ylsulfanyl)- [1, 3,4] thiadiazol-2-yl ; R24 is selected from the group consisting of H, tetrahydro isoquinolinyl, tetrahydro quinolinyl, 3-methyl indolinyl, indolinyl, 3-methyl indolinyl, 1-propyl-1, 2,3, 4- tetrahydro-pyrrolo [1, 2-a] pyrazine, 1-methyl-1, 2,3, 4-tetrhydro-pyrrolo [1, 2-a] pyrazine, and 1- [3- (6, 7-dimethoxy-l-methyl-3, 4-dihydro-lH-isoquinolin-2-ylmethyl)-4-methoxy- phenyl] -2, 3,4, 9-tetrahydro-lH-b-carboline ; and R25 is selected from the group consisting of H.

Synthesis of Small-molecule Trp-p8 Modulators As noted above, compounds of the present invention include compounds of Formula I, Formula II, Formula HE, Formula IV, Formula V, Formula VI, Formula VII, and Formula VIII. Within certain aspects, compounds of the present invention can be obtained from commercial sources and/or can be made using commercially available starting materials by employing synthetic methodology readily available in the art.

Compounds of Formulae I-VIII may be isolated using typical isolation and purification techniques known in the art, including, for example, chromatographic and recrystallization methods.

Exemplary compositions and methodology for their synthesis are disclosed in the following patents, each of which is incorporated herein by reference: U. S. Patent No.

4,150, 052 discloses N-substituted p-menthane 3-carboxyamide compounds having a physiological cooling action on the skin; U. S. Patent No. 4,153, 679 discloses compositions comprising acyclic tertiary and secondary carboxamides that have a physiological cooling action on the skin; U. S. Patent No. 4,020, 153 discloses cyclic sulphonamides and sulphinamides having a physiological cooling action on the skin; European Patent Application No. 1 157 617 discloses alpha-keto enamine derivatives in a variety of food, cosmetic, pharmaceutical, and perfume compositions; U. S. Patent No.

4,296, 093 discloses alkyl substituted cyclohexanamides having a physiological cooling effect on the skin; U. S Patent No. 5,756, 857 discloses cyclohexanol derivatives having a cool feeling; U. S. Patent No. 4,248, 859 discloses alicyclic amides having a physiological cooling effect; U. S. Patent No. 5,266, 592 discloses ketals, such as glycerol ketals, for example 1-menthone glycerol ketal or 3,3, 5-trimethylcyclohexanone glycerol ketal, that 11311. 100PCT 39

have a physiological cooling effect; U. S. Patent No. 6,328, 982 discloses compositions comprising a cooling compound such as 1-menthol (2-isopropyl-5-methyl-cyclohexanol), 1-isopulegol, 3- (1-menthoxy) propane-1, 2-diol and p-menthane-3, 8-diol ; and U. S. Patent No. 4,459, 425 discloses 3-1-menthoxypropane-1, 2-diol and its associated cooling activity.

Those of skill in the art will readily recognize that compounds suitably included in the compositions and methods of the present invention can exist in a number of cis and trans isomers, E/Z forms, diastereomers, as well as optical isomers. Thus, compounds used in the compositions and methods of the present invention include all such combinations and variations.

In compounds of Formula I, Formula II, Formula III, Formula IV, Formula V, and Formula VI carbon atoms to which four non-identical substituents are bonded are asymmetric. Accordingly, compounds of Formula I, Formula II, Formula m, Formula IV, Formula V, or Formula VI may exist as enantiomers, diastereomers or a mixture thereof.

The enantiomers and diastereomers may be separated by chromatographic or crystallization methods, or by other methods known in the art. The asymmetric carbon atom may be in one of two configurations, R or S, both of which are within the scope of the present invention. The presence of small amounts of the opposing enantiomer or diastereomer in the final purified product does not affect the therapeutic application of such compounds.

Compounds of Formulae I-VI may be further treated to form pharmaceutically acceptable salts. Treatment of a compound of the invention with an acid or base may form, respectively, a pharmaceutically acceptable acid addition salt and a pharmaceutically acceptable base addition salt, each as defined above. Various inorganic and organic acids and bases known in the art, including those described herein above, may be used to effect the conversion to the salt.

The present invention also relates to pharmaceutically acceptable isomers, hydrates, and solvates of compounds of Formula I, Formula II, Formula ni. Formula IV, Formula V, and Formula VI. Compounds of these formulae may also exist in various isomeric and tautomeric forms including pharmaceutically acceptable salts, hydrates and solvates of such isomers and tautomers.

This invention also encompasses prodrug derivatives of the compounds of Formula I, Formula II, Formula m, Formula IV, Formula V, and Formula VI. The term "prodrug"refers to a pharmacologically inactive derivative of a parent drug molecule that 11311. 100PCT 40

requires biotransformation, either spontaneous or enzymatic, within the organism to release the active drug. Prodrugs are variations or derivatives of the compounds of Formulae I-VI of the present invention that have groups cleavable under metabolic conditions. Prodrugs become the compounds of the invention which are pharmaceutically active in vivo when they undergo solvolysis under physiological conditions or undergo enzymatic degradation. An exemplary prodrug technology that may be suitably employed with the compounds of the present invention is the protease activated cancer therapy (PACT) technology described in detail within U. S. Patent Application No. 10/156,214 and PCT Application Publication No. WO 02/095007, both of which are incorporated herein by reference.

Synthesis of compounds of Formula I, maybe achieved, as described below in reference to compounds of Formula IV, by reacting an acid chloride, obtained by reacting p-menthane-3-carboxylic acid with thionyl chloride, with the appropriate amine. As noted below, typically, the reaction is carried out in solution at room temperature in the presence of a hydrogen chloride receptor (e. g., sodium hydroxide).

Synthesis of (l)-Menthoxypropane-1, 2-diol (Cooling Agent 10) from 1-menthol (2-isopropyl-5-methyl-cyclohexanol) is described in U. S. Patent No. 4,459, 425, incorporated herein by reference. Briefly, 1-menthol and metallic sodium or sodium hydride are introduced into a solvent (e. g., toluene or xylene) and heated. When the temperature reaches 100°C, or higher, the reaction starts and generation of hydrogen gas occurs. After confirming that the generation of hydrogen has stopped, the mixture is further heated at the reflux temperature of the solvent to complete the reaction.

Allyl halide (e. g., allyl chloride or allyl bromide) is then added to the reaction mixture in small portions. As the reaction proceeds, sodium halide deposits and the reaction solution becomes slurry-like. After the reaction is completed, the reaction solution is cooled, and after addition of water, the resulting mixture is stirred. Then, a solvent, (e. g., benzene, toluene, ether, hexane, or petroleum ether) is added. The organic layer is separated and washed with saturated saline water. After the solvent is recovered, the residue is distilled under reduced pressure to obtain 3-1-menthoxypropate-1-ene.

The 3-1-menthoxypropane-1-ne is oxidized into the corresponding oxide by use of an organic peracid. The oxide is hydrolyzed to form the desired 3-1-methoxypropane-1, 2- diol. That is, an organic acid, (e. g. , formic acid or acetic acid) and aqueous hydrogen peroxide are mixed with 3-1-menthoxypropane-1-ene and gradually heated carefully

while stirring. The organic acid and the hydrogen peroxide react, forming an organic peracid that participates in the oxidation reaction. The reaction is exothermic, and rapid heating should be avoided. When the temperature of the reaction solution reaches near 50°C, the heating is stopped. It is, thereafter, necessary for the temperature of the reaction solution to be maintained at about 70°C by external cooling to prevent a further temperature increase caused by the heat of reaction. If the temperature of the reaction solution is excessively high, the organic peracid decomposes before it participates in the oxidation reaction, resulting in a reduction in yield.

After the reaction is completed, a solvent (e. g., benzene, toluene, xylene, or petroleum ether) is added to perform the extraction. The extracted liquid is washed with water. Upon recovery of the solvent by distillation, a crude oxide in the form of an organic acid ester is produced. The crude oxide thus formed is mixed with an about 20% aqueous solution or caustic soda, for example, and is hydrolyzed by boiling for about 1 hour to produce the desired 3-1-menthoxypropane-1, 2-diol.

Synthesis of compounds of Formula II, exemplified herein by 1- (2- hydroxyphenyl)-4- (3-nitrophenyl)-1, 2,3, 6-tetrahydropyrimidine-2-one (Icilin aka AG-3- 5), is disclosed in U. S. Patent No. 3,821, 221, incorporated herein by reference. Briefly, ß- diethlyamino-m-nitropropiophenone hydrochloride is added to 50% aqueous ethanol and the mixture refluxed with stirring until the hydrochloride is dissolved. 0-aminophenol is added and the solution refluxed for 30 minutes, then set aside to cool. The reaction product is crystallized out of solution to yield p- (o-hydroxyanilino)-m- nitrophropiophenone (mp of 107°C to 109°C). ß-(o-hydroxyanilino)-m- nitrophropiophenone is dissolved in ethanol and concentrated HC1 is added to acidify the solution. The solvent is evaporated in vacuo and ß-(o-hydroxyanilino-m- nitropropiophenone)-HCI (mp of 172°C to 173°C) is crystallized from methanol-acetone.

ß-(o-hydroxyanilino-m-nikopropiophenone)-HCl is dissolved in acetic acid at 60 °C.

Potassium cyanate is added and the reaction mixture cooled to room temperature. Water is added and the crystalline 1- (2-hydroxyphenyl)-4- (3-nitrophenyl)-1, 2,3, 6- tetrahydropyrimidine-2-one (mp of 228°C to 230°C) is recovered by filtration.

Synthesis of acyclic carboxamide compounds of Formula m, as exemplified herein by 2-isopropyl-N, 2, 3-trimethylbutyramide alca N, 2, 3-trimethyl-2-isopropyl butamide (WS-23), is disclosed in U. S. Patent No. 4,153, 679, incorporated herein by reference. Briefly, Formula III amides may be prepared by conventional techniques 11311. 100PCT 42

known to those of skill in the art, for example, by reacting an acid chloride of the formula RioRi2Rl3COCl with an amine (Rll), as indicated above, in the presence of hydrogen chloride acceptor.

N, 2, 3-trimethyl-2-isopropyl b utamide may be prepared, for example, by heating 2,3-Dimethyl-2-isopropylbutanic acid under reflux with thionyl chloride for 60 minutes.

The excess of thionyl chloride may be removed under reduced pressure and the 2,3- dimethyl-2-isopropylbutanoyl chloride distilled, bp. 73°C-75°C/15 mm.

The acid chloride in ether may be added dropwise to a stirred solution of methylamine (70% soln. i n w ater) in ether with stirring. The ether 1 ayer may t hen b e washed with water, dilute HC1 and water. The dried (MgS04) ether solution was concentrated, and the residue distilled to give N, 2, 3-trimethyl-2-isopropyl butamide (mp 58"C-61 °C, bp. 83 °C-85 °C/0. 35 mm.).

Synthesis of exemplary 3-substituted-p-menthane compounds that may be suitably employed as Trp-p8 modulators in the compositions and methods of the present invention is described in U. S. Patent No. 4,150, 052, incorporated herein by reference in its entirety.

For example, the corresponding acid chloride (obtained by reacting p-menthane-3- carboxylic acid with thionyl chloride) may be reacted with the appropriate amine. The reaction will usually be carried out at room temperature in solution in the presence of a hydrogen chloride receptor, e. g., sodium hydroxide.

The basic p-menthane structure is a chair-shaped molecule that can exist in cis or trans form. Substitution of the carboxyl or amide group into the 3-position gives rise to four configurational or geometric isomers depending upon whether the substitution is axially or equatorially into the cis or trans isomer, the four isomers are related as menthol is to neomenthol, isomenthol, and neoisomenthol.

In an exemplary reaction protocol, p-Menthane-3-carboxylic acid is heated under reflux with thionyl chloride. Excess thionyl chloride is distilled off in vacuo. The crude p-menth-3-oyl chloride is dissolved in diethyl ether and the ethereal solution added with stirring and cooling to a solution of ethylamine and sodium hydroxide in water. The mixture is stirred and the ethereal layer separated. The aqueous layer is washed with ether and the combined ethereal solution washed with dilute hydrochloric acid and water. The ether solution is dried with MgS04 and evaporated to give a white crystalline solid. The solid is recrystallised. from acetone: water (9: 1) by dissolving the crystals at room temperature and then cooling to produce N-ethyl-p-menthane-3-carbozamide as a white 11311. 100PCT 43

crystalline solid, mp. 82. 5°C-84. 5°C. Substitution of the amide group in the 3-position of the p-menthane structure gives rise to optical and geometric isomerism.

When either R13 or R14 is aliphatic, the preferred values are Cl-Cg straight or branched chain alkyl, Ci-C straight or branched chain hydroxyalkyl or aminoalkyl and Cl-C4 acylated derivatives thereof, and-CnH2nCORls or-CnH2nCOOR, where-CnH2n is a straight or branched chain alkylene in which n is an integer of from 1-6 and Rls is H or a Cl-C8 alkyl or hydroxyalkyl group, preferably a Cl-C4 straight chain alkyl group.

When R13 is H and R14 is OH or substituted phenyl, e. g., alkylphenyl, hydroxyphenyl, alkoxyphenyl, halophenyl of up to 10 carbon atoms, phenalkyl or substituted phenalkyl, e. g., benzyl, naphthyl, or substituted naphthyl, and compounds where R13 and R14 are joined to form a cyclic group. When so joined, R13 and Rl4 preferably represent an alkylene chain, optionally interrupted by oxygen, which together with the nitrogen atom to which R13 and RI4 are attached forms a 5-or 6-membered heterocyclic ring.

Synthesis of exemplary compounds comprising one or more ketal, including for example menthone glycerine ketals, that may be suitably employed as Trp-p8 modulators in the compositions and methods of the present invention is described in U. S. Patent No.

5,266, 592, incorporated herein by reference in its entirety.

For example, ketals of Formula V may be prepared by an acid-catalysed reaction of a ketone on which the ketal of Formula V is based and not less than the equivalent amount of aliphatic C3-C6-alcohol having not less than 3 and not more than 5, preferably 3, hydroxyl groups. The ketone on which the ketal of Formula V is based and an excess amount of the C3-C6 alcohol having 3 to 5 hydroxyl groups will be employed.

Exemplary acid catalysts that can be used are p-toluenesulphonic acid, phosphoric acid, or potassium hydrogen sulphate in catalytically effective amounts. The reaction will generally be carried out either in an organic solvent that together with water forms an azeotrope, so that the water, which is liberated during formation of the ketal, can be eliminated by azeotropic entrainment or water-consuming co-reagents such as, for example, trialkyl ortho esters are used. Exemplary organic solvents include benzene, toluene, x ylene, c liloroform, m ethylene chloride a nd trichloroethylene. T he reaction is complete when water no longer separates out or when the ester/alcohol mixture is no longer separated out. The products may be washed subsequently with dilute alkali and

with water, to separate and dry the organic phase, to strip off the solvent and, if appropriate, to purify the residue, for example by distillation.

An exemplary compound comprising a ketal of Formula V is 1-menthone glycerol ketal (Frescolat MGA, aka menthone glycerin acetal; Harris & Ford, LLC), which is synthesized as follows. 2 mol of 1-menthone, 3 mol of glycerol, and 5 g of potassium hydrogen sulphate are mixed in a 2 liter three-neck flask in the presence of toluene. This mixture is refluxed in a water separator. After 7 hours, water separates and the mixture is neutralized and distilled.

Synthesis of the exemplary Formula VI terpene compound L-Carvone (2- cyclohexen-l-one, 2-methyl-5- (l-methylethenyl)- (R) ; Millenium Chemicals) from D- limonene is described in Ikan, Natural Products-A Laboratory Guide pp. 151-155 (Academic Press, 1969), incorporated herein by reference. Briefly, a solution of D- limonene in isopropanol is cooled to below 10°C. Solutions of HCl in isopropanol and concentrated aqueous sodium nitrite arc added dropwise to the D-limonene solution to generate limonene nitrosochloride. The limonene nitrosochloride and dimethylformamide is refluxed with isopropanol. After cooling, crystallization is induced and the precipitate filtered and washed with water. The resulting L-carvoxime is refluxed with 0.5 M oxalic acid and the mixture is steam distilled. The distillate is extracted with ether, dried over anhydrous magnesium sulfate, and the excess ether evaporated to leave L-carvone.

Additional synthetic methods for the preparation of Trp-p8 modulators of the present invention are presented herein in Examples 1-5.

Compositions Comprising a Small-molecule Trp-p8 Modulators As discussed above, the present invention is directed to small-molecule Trp-p8 modulators, including Trp-p8 agonists and Trp-p8 antagonists that bind to and alter the activity of Trp-p8. Within certain embodiments, Trp-p8 modulators are agonists that are, in certain instances, capable of stimulating cation influx in, and toxicity of, a cell expressing the Trp-p8 channel protein. Within alternative embodiments, Trp-p8 modulators are antagonists of Trp-p8 activity that are capable of reducing the activity of Trp-p8 expressed in a cell. Thus, Trp-p8 modulators of the present invention will find utility in compositions, including pharmaceutical compositions, which are useful in the treatment of diseases associated with Trp-p8 expression. Suitable compositions, according to the present invention, comprise one or more Trp-p8 agonist of Formula I, 11311. 100PCT 45

Formula II, Formula m, Formula IV, Formula V, Formula VI, Formula VII, and/or Formula VIM and/or one or more Trp-p8 antagonist of Formula I, Formula II, Formula m, Formula IV, Formula V, Formula VI, Formula VII, and/or F onnula V III, as described above, in combination with one or more pharmaceutically acceptable carrier or excipient.

In one embodiment, the present invention provides small-molecule Trp-p8 modulators in combination with a pharmaceutically acceptable excipient such as sterile saline or other medium, water, gelatin, oil, etc. , to form pharmaceutically acceptable compositions. The compositions and/or agonists may be administered alone or in combination with any convenient carrier, diluent, etc. and such administration may be provided in single or multiple dosages. Useful carriers include, but are not limited to, solid, semi-solid, or liquid medium including water and non-toxic organic solvents.

Pharmaceutical compositions of the present invention may be prepared by mixing one or more Trp-p8 agonist of Formula I, Formula II, Formula m, Formula IV, Formula V, Formula VI, Formula VII, and/or Formula VIII with a pharmaceutically acceptable carrier or agent. Alternatively, pharmaceutical compositions may be prepared by mixing one or more Trp-p8 antagonist of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, and/or Formula VIH with a pharmaceutically acceptable carrier or agent. In addition, pharmaceutical compositions may further include excipients, stabilizers, diluents and the like and may be provided in sustained release or timed release formulations. Acceptable carriers, agents, excipients, stabilizers, diluents and the like for therapeutic use are well known in the pharmaceutical field, and are described, for example, in"Remington's Pharmaceutical Sciences, " (Mack Publishing Co. , ed. A. R. Gennaro, 1985), incorporated herein by reference. Such materials are nontoxic to the recipients at the dosages and concentrations employed and include buffers such as phosphate, citrate, acetate, and other organic acid salts, antioxidants such as ascorbic acid, low molecular weight peptides such as polyarginine, proteins, such as serum albumin, gelatin, or immunoglobulin, hydrophilic polymers such as serum albumin, gelatin, or immunoglobulin, hydrophilic polymers such as polyvinylpyrrolidinone, amino acids such as glycine, glutamic acid, aspartic acid, or arginine, monosaccharides, disaccharides, and other carbohydrates including cellulose or its derivatives, glucose, mannose or dextrins, chelating agents such as EDTA, sugar alcohols such as mannitol or sorbitol, counterions such as sodium and/or nonionic surfactants such as TWEEN, or polyethyleneglycol.

11311. 100PCT 46

Within still further aspects, the compositions of the present invention comprise a compound of Formula I, Formula II, Formula m, Formula IV, Formula V, Formula VI, Formula VII, and/or Formula VIII formulated together with one or more cancer therapeutic agent. Alternatively, the compositions of the present invention comprise a compound of Formula I, Formula II, Formula m, Formula IV, Formula V, Formula VI, Formula VII, and/or Formula VIII independently formulated with one or more cancer therapeutic agent. That is, the compound of Formula I, Formula II, Formula m, Formula IV, Formula V, Formula VI, Formula VII, and/or Formula VIII and the cancer therapeutic agent are separately formulated.

Suitable cancer therapeutic agents include, but are not limited to, antimitotic agents including, but not limited to, paclitaxel, vincristine, and etoposide; alkylating agents including, but not limited to, mechlorethamine, cyclophosphamide, and carmustine ; antimetabolites including, but not limited to, methotrexate, gemcitabine, lometrexol, 5-fluorouracil, and 6-mercaptopurine; cytotoxic antibiotics including, but not limited to, doxorubicin, daunorubicin, bleomycin, mitomycin C, and streptozocin ; platinum agents including, but not limited to, cisplatin and carboplatin; hormonal agents including, but not limited to, anti-estrogens such as tamoxifen and d iethylstilbestrol as well as anti-androgens such as flutamide; antiangiogenesis agents; and farnesyl transferase inhibitors.

In certain aspects, compounds of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, and/or Formula VIII are administered in combination with a cancer therapeutic agent that is ineffective in stimulating Trp-p8- mediated cation influx.

In other aspects, compounds of Formula I, Formula II, Formula in, Formula IV, Formula V, Formula VI, Formula VII, and/or Formula VIII are administered in combination with one or more additional Trp-p8 modulator including, but not limited to a compound of Formula I, Formula II, Formula m, Formula IV, Formula V, Formula VI, Formula VII, and/or Formula VIII.

Depending upon the particular treatment regimen contemplated, pharmaceutical compositions of the present invention may be administered parenterally, topically, orally, or locally. In certain aspects, the pharmaceutical compositions are administered parenterally, e. g., intravenously, subcutaneously, intradermally, or intramuscularly. In one embodiment, the present invention provides compositions for parenteral 11311. 100PCT 47

administration that comprise a compound of the present invention, dissolved or suspended in a carrier such as an aqueous carrier.

For solid formulations, compounds may be admixed with conventional nontoxic solid c arriers s uch a s, for example, pharmaceutical grades of m annitol, lactose, starch, magnesium stearate, sodium saccharin, talcum, cellulose, glucose, sucrose, magnesium carbonate, and the like.

For aerosol administration, compounds of the present invention may be supplied in finely divided form along with a nontoxic surfactant and propellant. Exemplary such agents are the esters or partial esters of fatty acids containing from 6 to 22 carbon atoms, such as caproic, actanoic, lauric, palmitic, stearic, linoleic, olesteric, and oleic acids.

Compositions of the present invention may be administered by injection, i. e. intravenously, intramuscularly, intracutaneously, subcutaneously, introaduodenally, or intraperitoneally. Alternatively, compositions may be administered by inhalation, such as intranasally, and may be administered transdermally, such as through a patch or the like.

It will be understood that the actual preferred formulation of compositions, including pharmaceutical compositions, will vary according to the mode of administration as well as the particular disease being treated. The optimal formulations and modes of administration will be routinely determined on a disease by disease and patient by patient basis by those of skill in the art.

Methods for Identifying and Characterizing the In vitro and Irz vivo Efficacy of Small-molecule Modulators of Trp-p8 As discussed above, the present invention is directed to small-molecule Trp-p8 modulators, including agonists and antagonists of Trp-p8 activity. Disclosed herein are Trp-p8 modulators exemplified by the compounds of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII, andlor Formula vm described herein above. The present invention further contemplates that additional Trp-p8 modulators may also be suitably employed in the compositions and methods of the present invention.

Additional or alternative Trp-p8 agonists and antagonists may be identified by the methodology disclosed in the accompanying Examples. For instance, Trp-p8 agonists having efficacy in the treatment of disease (s) associated with Trp-p8 expression include small molecules that result in one or more of the following: (1) inhibit the growth or decrease the viability of a cell expressing Trp-p8; (2) stimulate calcium and/or other

cation influx in a cell expressing Trp-p8; (3) induction of apoptosis and/or necrosis in a cell expressing Trp-p8; and/or (4) efficacy in one or more animal model systems of human disease. Trp-p8 antagonists having efficacy in the treatment of disease (s) associated with Trp-p8 expression include small molecules that that result in one or more of the following: (1) protect Trp-p8 expressing cells from toxic effect of agonists in in vitro model system (2) inhibit growth of and/or kill cancer cell line with endogenous Trp- p8 expression (3) are efficacious in one or more animal model systems of human disease.

Thus, within certain embodiments, the present invention provides methods for identifying Trp-p8 agonists comprising thestepofcontacting a Trp-p8 expressing cell with a candidate Trp-p8 agonist for a time and in an amount sufficient to inhibit the growth a nd/or d ecrease t he viability o f a T rp-p8 e xpressing c ell, w herein t he i nhibited growth and/or reduced viability indicate that the candidate Trp-p8 agonist is capable of activating Trp-p8 expressed by the cell.

Other embodiments provide methods for identifying Trp-p8 agonists, comprising the step of contacting a Trp-p8 expressing cell with a candidate Trp-p8 agonist for a time and in an amount sufficient to induce influx of calcium and/or other cations into the cell, wherein increased cation influx is correlative of increased cellular toxicity.

Still further embodiments provide methods for identifying Trp-p8 agonists comprising the step of administering a candidate Trp-p8 agonist to an animal having one or more neoplastic cell that expresses Trp-p8 for a time and in an amount sufficient to inhibit the growth of and/or induce apoptosis and/or necrosis in the cell thereby increasing the survival of the animal, wherein any one or more of inhibition of cell growth, induction of apoptosis, induction of necrosis, and/or increased survival of the animal indicate efficacy of the Trp-p8 agonist.

The present invention provides methods for the identification of Trp-p8 antagonists in addition to the Trp-p8 antagonists disclosed herein by the compounds of Formula VII and Formula VIII. Such method include (1) in vitro assay systems for detecting the protection of Trp-p8 expressing cells from toxicity induced by Trp-p8 agonists; (2) in vitro and in vivo assay systems of detecting the inhibition of growth of a cancer cell and/or cancer cell line endogenously expressing Trp-p8; (3) in vivo animal model systems whereby one or more candidate Trp-p8 antagonist is administered to an animal having one or more neoplastic cell that expresses Trp-p8 for a time and in an 11311. 100PCT 49

amount sufficient to inhibit the growth of and/or induce apoptosis and/or necrosis in the cell thereby increasing the survival of the animal.

Methods for Use of Trp-p8 Modulators Small-molecule Trp-p8 modulators of the present invention may be suitably employed in methods for modifying (i. e. activating or reducing) Trp-p8-mediated calcium influx in a cell and therapeutic methods for the treatment of one or more diseases associated with expression of Trp-p8. For example, and as noted above, it has been observed that abnormal Trp-p8 expression is associated with a neoplastic phenotype in a variety of cancerous tissues including breast, colon, lung, and prostate tissues. Tsavaler et al., Cancer Research, supra.

Thus, within certain embodiments are provided methods for activating Trp-p8- mediated calcium influx in a cell, such methods comprising the step of c ontacting the Trp-p8 expressing cell with an amount of a Trp-p8 agonist for a time sufficient to inhibit growth of the cell and/or to induce necrosis and/or apoptosis in the cell. Exemplary methods for activating Trp-p8 are provided within the Examples presented herein.

Other embodiments of the present invention provide therapeutic methods for the treatment of diseases associated with expression of Trp-p8, such methods comprising the step of administering to a mammal, typically a human, a therapeutically effective amount of a composition comprising a Trp-p8 agonist for a time sufficient to inhibit growth of the cell and/or to induce necrosis and/or apoptosis in the cell. As used herein, the phrase "therapeutically effective amount"refers to the amount of a compound that, when administered to a mammal for treating a disease, is sufficient to effect such treatment for the disease. The"therapeutically effective amount"will vary depending upon the compound, the disease, and its severity and the age, weight, etc., of the mammal to be treated.

As used herein, the terms"treat","treating", and"treatment"include : (1) preventing the disease, i. e. causing the clinical symptoms of the disease not to develop in a mammal that may be predisposed to the disease but does not yet experience any symptoms of the disease; (2) inhibiting the disease, i. e. arresting or reducing the development of the disease or its clinical symptoms; or (3) relieving the disease, i. e. causing regression of the disease or its clinical symptoms.

11311. 100PCT 50

While the frequency and dosage of treatment regimens will vary depending upon such factors as the disease and patient treated, compositions comprising one or more compound of Formula I, Formula II, Formula in, Formula IV, Formula V, Formula VI, Formula VII, and/or Formula VIII are typically administered in the range of about 0.001 mg compound/kg body mass to about 100 mg/kg. Typically, treatment is initiated with smaller dosages that are less than the optimum dose of the compound. Thereafter, the dosage may be increased until optimal effectiveness is achieved.

In most instances, administration of a composition (s) of the present invention is achieved by any method that ensures systemic exposure to the compound of Formula I, Formula II, Formula m, Formula IV, Formula V, Formula VI, Formula VII, and/or Formula VIII. Thus, compositions may be administered orally, parenterally, intraduodenally, and intranasally. Typically, such compositions comprise one or more such compound in combination with one or more pharmaceutically acceptable carrier or diluent, as described in further detail herein above.

Other embodiments of the present invention provide combination therapies wherein one or more compound of Formula I, Formula II, Formula m, Formula IV, Formula V, Formula VI, Formula VII, and/or Formula VIII is administered in conjunction with one or more cancer therapeutic agent, as described in further detail herein above, such as an antimitotic agent, an alkylating agent, an antimetabolite, a cytotoxic antibiotic, a platinum agent, a hormonal agent, and/or an antiandrogen. Still further embodiments of the present invention provide combination therapies wherein two or more compounds of Formula I, Formula II, Formula m, Formula IV, Formula V, Formula VI, Formula VII, and/or Formula VIII are administered either simultaneously or sequentially to achieve the desired therapeutic outcome.

Thus, as used herein, the term"combination"means that at least two compounds can be delivered in a simultaneous manner, in combination therapy wherein the first compound is administered first, followed by the second compound, as well as wherein the second compound is delivered first, followed by the first compound. The desired result can be either a subjective relief of a symptom (s) or an objectively identifiable improvement in the recipient of the dosages.

The following examples are offered by way of illustration and not by way of limitation.

11311. 100PCT 51

EXAMPLES Example 1 Synthesis of Menthane-3-ca7-boxa77zide Compounds This example discloses methodology for the synthesis of mentliane-3-carboxamide Trp-p8 modulators.

Mentlt. ane-3-carboxvlic acid (2). Water (300 ml) was placed in a 2-L Erlenmeyer flask with a large stir bar. Sulfuric acid (500 ml) was added carefully with stirring. The solution was allowed to cool to 75°C, and N-ethyl-p-menthane-3-carboxamide (1,62. 5 g) was added. The temperature was maintained at 75°C with a hot plate, and sodium nitrite (31 g) was added carefully. Two more 31 gram portions of NaNO2 were added at 1-hour intervals, and the mixture was stirred overnight at 75°C.

The mixture was cooled to room temperature, diluted with-IL of ice water, and extracted with-500 ml of ether. The ether layer was separated, washed with water, and extracted with 2 x 350 ml of 1M NaOH. The aqueous layer was made acidic with concentrated HC1 and extracted with ether. The ether layer was dried with MgS04 and evaporated to give menthane-3-carboxylic acid (33.2 g, 61%) as a crystalline solid, [#] =-50.3 deg (c=l, CHC13, 25 °C).

Merzthane-3-carbonvl chloride (3). Menthane-3-carboxylic acid (54.35 g) was refluxed with 80 ml of thionyl chloride for 3 hours. The SOC12 was removed by distillation, and the acid chloride was distilled at 114-115 °C (8 Torr). (Lit. b. p. 84-85 °C at 3.5 Torr). Yield: 50 g (84%).

General procedure for preparation of nzenthane-3-carboxamides (5). To a stirred solution of 0.2 mmol of the amine (4) in 1 ml of acetonitrile or NMP and 0.4 mmol of DIPEA was added 0.022 ml of menthane-3-carbonyl chloride (S. The reaction mixture was shaken for 3 hours. For less reactive amines, the mixture was heated (60°C) and shaken for 24 hours. The product was purified from the crude reaction mixture by HPLC (40-95% gradient over 10 minutes using 0.05% TFA in CH3CN and 0.05% TFA in H2O) and evaporated to dryness.

Example 2 Svnthesis ofMerathol Acetamide arad Carbanaate Compounds This example discloses methodology for the synthesis of Menthol Acetamide and Carbamate Trp-p8 modulators.

General procedures for the preparation of menthol acetamides (7) and menthol carbamates (9). To a two-phase ether-water system containing 0.07 mmol of amine (@ in 1 ml of ether and 1 ml of 0.1 M aqueous sodium hydroxide was added 0.1 mmol of acid

chloride U6 or chloroformate (S. The reaction mixture was shaken for 2-3 hours. The upper layer was removed and evaporated to dryness to afford the product.

Example 3 Synthesis of Menthyl-3-urea and Menthyl-3-thiourea Compounds This example discloses methodology for the synthesis of Menthyl-3-urea and Menthyl-3-thiourea Trp-p8 modulators.

12 13 Menthvl-3-isocyanate (10 ! To an ice-cooled solution of menthane-3-carboxylic acid (4 g, 22 mmol) and triethylamine (3.05 ml, 22 mmol) in dry DMF (45 ml) was added diphenylphosphoryl azide (4.72 ml, 22 mmol). The mixture was stirred at 0°C for 2 hours and at room temperature for 3 hours, then poured into a mixture of ether and ice water.

The ether layer was separated and washed with aqueous sodium bicarbonate, dried with MgS04, and evaporated. The residue was distilled (85°C, 10 Torr) to give menthyl-3- isocyanate (2.9 g, 73%) as a colorless liquid.

General procedure for the preparation of menthyl-3-ureas (12) and menthyl-3- thioureas (13). To a stirred solution of 0.07 mmol of the amine (4) or thiol (11) in 1 ml of dry ethyl acetate was added a solution of 0.07 mmol of menthyl-3-isocyanate in 0.5 ml of

ethyl acetate. The reaction mixture was shaken overnight then evaporated to dryness to afford the product.

Example 4 Synthesis of Menthane-3-amide, Menthane-3-sulfonamide and Menthane-3-carbamate Compounds This example discloses methodology for the synthesis of Menthane-3-amide, Menthane-3-sulfonamide and Menthane-3-carbamate Trp-p8 modulators.

18 19 20 Menthyl-3-amine hydrochloride (14). To a vigorously stirred mixture of diethyl ether (100 ml) and 6N aqueous hydrochloric acid (100 ml) was added menthyl-3- isocyanate (2.0 g, 11 mmol), and the mixture was stirred overnight at room temperature.

The aqueous phase was separated, made basic with an excess of aqueous sodium hydroxide, and extracted with ether. The ether phase was dried with MgS04 and filtered.

An excess of anhydrous HCl (3M) in ethanol was added, and a precipitate immediately formed. Filtration gave menthylamine hydrochloride (1.9 g, 90 %) as fine white crystals.

General procedure for the preparation of menthane-3-amides (18), menthane-3- sulfonamides (19) and menthane-3-carbamates (20). A solution of menthyl-3-amine hydrochloride (0. 078 mmol) in 0.7 ml of water was added to a solution of the acid

chloride, sulfonyl chloride, or chloroformate (0.078 mmol) in 1 ml Et2O, followed by the addition 0.3 ml of 0.5 M NaOH. The mixture was shaken at room temperature for 12-18 hours. The Et2O layer was then separated and the solvent removed under reduced pressure to afford the product.

Example 5 Synthesis o Menthvl-3-amine Conzpounds This example discloses methodology for the synthesis of Menthyl-3-amine Trp-p8 modulators. H2N E O 22 L Method Method 2 21 23 14 General procedures for the preparation o fmenthvl-3-amiraes 23).

Method 1 A mixture of the amine (, 0.1 mmol) and 15.4 mg (0.1 mmol) of menthone (2) in 1 ml of 1,2-dichloroethane was treated with sodium triacetoxyborohydride (32 mg, 0.15 mmol). The mixture was stirred for 24 h (periodically monitored by LCMS). The reaction mixture was quenched by adding 1N NaOH (1 ml) and the product was extracted with ether. The extract was washed with brine, dried (MgS04) and evaporated to dryness to afforf the product.

Method 2 A mixture of menthyl-3-amine (0.1 mmol) and 15.4 mg (0.1 mmol) of aldehyde (24) in 1 ml of 1, 2-dichloroethane was treated with sodium triacetoxyborohydride (32 mg, 0.15 mmol). The mixture was stirred for 24 h (periodically monitored by LCMS). The reaction mixture was quenched by adding 1N NaOH (1 ml) and the product was extracted with ether. The extract was washed with brine, dried (MgS04) and evaporated to dryness.

Example 6 Expression of Trp-p8 in CHO Cells Human trp-p8 transfected CHO cells (referred to herein as CHO/Trp-p8) were generated for use in experiments of the present invention. Expression of Trp-p8 polypeptide in this transfectant and the absence of any endogenous expression in the non- transfected CHO was confirmed by western blot and immunofluorescence using a Trp-p8 specific antibody (GS2.20 ; disclosed within copending U. S. Patent Application No.

10/_,, incorporated herein by reference in its entirety) as well as the calcium flux assay with Icilin (1- [2-hydroxyphenyl]-4- [3-nitrophenyl]-1, 2,3, 6-tetrahydropyrimidine-2- one) and menthol (2-isopropyl-5-methyl-cyclohexanol). Non-transfected CHO cells were used to establish the specificity of the effects of the compounds observed with CHO/Trp- p8.

Example 7 Trp-p8-mediated Decrease in Cell Viabilitv followingExposure of CHOlTrp-p8 Cells with Candidate Trp-P8 Agonist Co7npounds at 3 7°C This Example discloses an ATP viability assay suitable for screening for effective Trp-p8 agonists. The ATP viability assay described herein employs CHO cells expressing an exogenous Trp-p8 cDNA. This example further establishes that Trp-p8 agonists of the present invention are effective in decreasing the viability of Trp-p8 expressing cells.

The concentration of intracellular ATP declines very rapidly when metabolically active cells undergo necrosis and/or apoptosis. The ATP concentration and consequently the relative cell viability can be measured by established methods using commercially available reagents. In the agonist screening methodology disclosed herein, a compound that specifically decreases the viability of CHO/Trp-p8 cells is referred to as an agonist.

As a primary screen for efficacy and specificity for agonists, both the non- transfected CHO and CHO/Trp-p8 cells were exposed to 10 uM of test compounds in 1% dimethylsulfoxide (DMSO) or 1% DMSO (control) in a 96-well black walled, black- bottomed, cell-culture treated plate. DMSO was the solvent for all of the compounds tested. After 24-26 hours at 37°C, the cells were lysed and ATP concentration determined via a chemiluminescence assay using a commercially available reagent kit-- Cell Titer-Glo (Promega; Madison, WI). Relative viability (%), expressed as the ATP level in cells treated with compounds expressed as a percentage of ATP levels in cells

treated with the DMSO alone, was a measure of the agonist activity of the candidate compound--the lower the % viability, the more potent the Trp-p8 agonist. ECSO values were determined for the most active candidate Trp-p8 agonists at 37°C by measuring viability at 8-10 agonist concentrations. (EC50 is defined herein as the agonist concentration at which there is a 50% reduction in cell viability).

Exemplary Trp-p8 Agonists of Formula IV that were efficacious in the ATP viability assay are presented herein in Table 1. Table 1A presents Formula IV Trp-p8 agonists exhibiting an ECSO value within the range of 0.05 to 0. 20 µM; Table 1B presents Formula IV Trp-p8 agonists exhibiting an ECso value within the range of 0.20 to 0.50 pM ; Table 1 C presents Formula IV Trp-p8 agonists exhibiting an ECSO value within the range of 0.50 to 1. 00 uM ; and Table 1D presents Formula IV Trp-p8 agonists exhibiting an ECso value within the range of 1.00 to 7. 00 uM.

Viability of CHO/Trp-p8 cells following treatment with exemplary Trp-p8 agonists is presented in Figure 1.

Table 1A Exemplarv Trp-p8Agonists of FormulaIV Exhibiting an EC50 Value within the Range of 0.05 to 0. 20 µM Ref. Chemical Structure Rials Ris No. : CF 2905 J OH OU cl, O 3012 H, CH Con CM, OH 2-hydroxy-2- hydroxyphenyl) $ HaC 3006 phenylethyl

Ref. Chemical Structure Rials No. : CF 2926 H, c, CH 0 3014"T" 1 Benzoylamino CH2 Chez N 2963 c, , 4-acetylphenyl vs cH, Table 1B Exemplary Trp-p8 Agonists of Formula IV Exhibiting an EC50 Value within the Range of 0.20 to 0.50 µM Ref. Chemical Structure R14/Rl'R" No. : cl, fC N'-quinoxalin CH3 Cl, ..'k, °" 2- ethyl CFI c 2-hydroxy-2- H CACHO hydroxyphenyl) ethyl CF 2- ethyl if 2901 N" Y Ref. Chemical Structure Rials No. : 1906 () ethyl CH A 2920 N ethyl H, cl, f]/°°°''' CH lic H, N 2- CF CN 1603 xi N 2Z64 f '"Y°CH. 2261 KC CF , JJ 2904 CH, OH CH con, 2932 0 H, : CHO CH 2- H, C F Ref. Chemical Structure Rials Ris No. : CH 2942 CL3 CIi 2930-O ethyl N, CH Vs 1901 ethyl ZU cH, 1-methyl-2- 2944 e N- JUUo '.-< < benzamide-4-yl obc OH. 2966 i, a 2973 2-chloro-4-cyanophenyl Y M oH, c 2869 ) OFIC N CFi 0< chromen-7-yl 2832 t phenyl f H, Ref. Chemical Structure Rl41Rl5 No. : , 2836 tt 0 4- 2887 3-dioxo-2, 3-dihydro-lH- isoindol-5-yloxy]-phenyl 4- 2892 3-dioxo- 2, yloxy}-phenyl aNn 3-chloro-4-morpholin-4-yl- phenol () phenyi V 2958 l rv OHC GI CII 2864 2-chloro-4-iodophenyl ai°N v 2831 CH. e. Lr 2983 2-chloro-4-nitrophenyl Cl 2961 4-cyclopentanephenyl '% Q 2626 °

Ref. Chemical Structure Rl4/Rl5 No. : Oh, N -CF 2964/\ \-rut Table 1C Exemplary Trp-p8 Agonists of Formula IV Exhibiting an EC50 Value within the Range of 0.50 to 1.00 µM Ref. Chemical Structure Rials Ris No. 3040 N, CF Cl \ 2903 L CL, cet 191) e, c 1903 5 0' N 2679 ethyl I H3C CH H C/OH , hydroxyphenyl) CH3 A 2263 2- F CH 3041 l, HC H, C CHg Ref. Chemical Structure Rl4/Rl5 No. : CHO 3039 HC /N S 1619 3-phenyl-piperidin-1-yl . . OCH, N 2262 ethyl CH 2- 2940 C 8 bromophenyl) ethyl a, 'i 1-hydroxymethyl-2-phenyl 2270 ethyl cl HIC 301) CH c 29 methoxyphenyl) ethyl CH3 H, /CH, 2914 ethyl N CH, 3010 %, H, o° CH3 CH3 2912 N Ref. Chemical Structure Rl4/Rl5 No. : CF 2-phenyl-2- 2922 Chu 2950 29 ' '1f yl)-ethyl 2868 4-methylphenyl 0 HC 2891 trifluoromethyl-phenyl)- T yloxy]-phenyl 2998"'eH, ylmethyl)-phenyl lC-d V 2960 'N \ 2970 oh, , II, G 2979 ß 0 n 2-methyl-4-broophenyl UX B, FC 2987 4-Pyrrolidin-1-ylmethyl- phenyl P 2853 benzoimidazol-5-yl ka

Ref. Chemical Structure No. 2875 c phenyl C 2956 !"'roi) y N4 HCRCIS 2978 , 3-chloro-4-methoxyphenyl p o n, e 0'ite 2856 2-pyridin-3-yl-lH- benzoimidazol-5-yl Table 1D Exemplar Trp-p8 -p8 Agonists of Formula IV Exhibiting an EC50 Value within the Range of 1.00 to 7. 00 µM Ref. Chemical Structure No. : CH N ethyl c C c. OH methoxyphenyl)-ethyl CH, 0 2269 2269 2 Ref. Chemical Structure Rials Ris No. : zoo 1-methoxymethyl-2- 3007 cf . 1-methyl-2- 2898 y M cM 1627 3-phenyl-pyrrolidin-1-yl -. 2271 ethyl if CHO 2933 (2, 6- C, CH3. 2936 ethyl H, C n T 2-phenyl-2- y ! CHO CL chez H, CN cH, w 1-hydroxymethyl-2-phenyl ' CRI 29'29 triflouromethylphenyl) ethyl Ref. Chemical Structure Rl4/Rl5 R15 No. : razz 2- 2935 1591 oH, 3035) benzyloxy H, HCt1< 1568 2- CH, 1-hydroxymethyl-2- chlorophenyl) nS, Qs a 2265 2- ethyl I w 2924 2924 chlorophenyl) H, C CH, a 2677 6 1910 2- (4- methylsulfanylphenyl) ethyl m 2273 hydroxy-2-phenylethyl Ref. Chemical Structure Rl4/Rl5 Ris No. : 2- 29 dimethoxyphenyl) ethyl CH 2- 2949 2949 g} cN w cia° I i 2941 2- ethyl CH, CH, 2953 N H, ccH XN 2938 ci, c o, CF6 2934 Y'rcH, , dimethoxyphenyl) CF 1-methyl-2-hydroxy-2- H, Jd' M 6, 2647 isoquinolin-2-yl cr , 2915 zur nFl. '2- 1912 , pyrrolidin-3-yl)-ethyl Ref. Chemical Structure Rl4/Rl5 No. 2- ethoxyphenyl) ethyl 1-hydroxymethyl-2- (lH- 2947 2947 H, ethyl [1-carbamoyl-2- 2945 \ [1-caxbamoyl-2- (1H-indol- 2945 3-yl)-ethyl "r\ 1599 1--phenyl- H, 3008 "'1-hydroxymethyl-2-phenyl 3008 CCH, 1-hydroxymethyl-2- hydroxyphenyl) Y YP Y) Y 2598, c6 . "1 25 2, 3-diflourobenzyl 0 N I/ 2899 X Cl N 2279 2, 2-diphenylethyl 9 Ref. Chemical Structure RlaRl5 No. JtCCH, AU 2267 . 4-pyrimidin-2-yl- 1611 piperazin-1-yl 4% o Y dimethoxyphenyl) ethyl cit I% 1566 4-methylcyclohexyl HsC, tc 2260 CH2 0 "CL 1-carbamoyl-2- hydroxyphenyl) H, 1637 O-N CHj H3C-J." 1629 bicyclo 2. 1] hept-2-yl rf 'N 1614 dipropylamino) Cl 2272 ethyl 0 Ref. Chemical Structure Rl4iRl5 No. : N-\-CH, 00 4-Biphenyl 1617 3, 4-dimethylphenyl ONC ZU 4- 28152 phenyl phenyl i o 2844 foc OH, 2-oxo-2, 28'70 /N fut, N F 303 2-trilfouromethyl-lH- w benzoimidazol-5-yl ru o 2974 2, 3-dimethoxyphenyl zur fiv 2849 4-Azepan-1-ylmethyl- phenyl Si 2850 phenyl cl, H, 1631/QCH, 4-chlorophenyl cl Ref. Chemical Structure Rl4/Rl5 No. : 2841 3-dioxo-2, 3-dihydro-lH- 2841 isoindol-4-yl on, c, 2843 B 2843 H, c oc . 0 1607, H, c ch ru 2840 2, CF 0 2962 X H, , ° 0 2872 2-flouro-5-nitrophenyl lac y 2985 2-methyl-1, 3-dioxo-2, 3- dihydro-1H-isoindol-5-yl ct ttc 0 1586'"y\-"cn. 0 o" SNq 2833 3-nitrophenyl Nu 2957 'N 0 Ref. Chemical Structure RlaRls No. 0 1639 4-bromophenyl FA F O CH 2835 t'" 2982 2-chloro-5-nitrophenyl cet CH 4 2954 O o"\/ 2871 3-acetamidophenyl 1 , r 1575 4-ethylphenyl ordo OH. 2980 3, 4-dichlorophenyl cl cit . 1642 c6 benzo [1, 3] dioxol-5-yl 'r w 2873 1, 3-dioxo-2, 3-dihydro-lH- isoindol-5-yl ou O-N CH 2965 (35 H, C Ref. Chemical Structure Rl4/Rl5 No. : F oc 2976 "/ 1545 F 2990 4, 5-trimethoxyphenyl w en, cl CHO /II N C\ 2829 2837 r c6 2859 2-iodophenyl Nu F - -GH 2972 4, 5-triflourophenyl . M 4- 3002 "c) 2851 cil r-X 1616

Ref. Chemical Structure Rials-Ris No. : IF N benzooxazol-5-yl a n 28, Ka cl, °Q 1577 4-dimethoxyphenyl H, 0 1585 5-dimethoxyphenyl _o RlaRl5 RisExample 8 Screen and Characterization of Trp-p8 Agonist Compounds bv Measuring Calcium Infiux in CHO/Trp-pS cells at 37°C This example discloses a CHO/Trp-p8-based calcium influx assay used to further assess the activity of candidate Trp-p8 agonists of the present invention.

Calcium influx was measured using a Flexstation Microplate Fluorescence Plate Reader (Molecular Devices; Sunnyvale, CA). A typical assay for calcium flux was performed as follows. Cells in DMEM/Ham's F-12 based medium, typically at a density of 30,000 cells/well/100 al, were plated in a 96-well black-walled, clear bottomed tissue culture plate (Greiner Bio-one) and incubated for 16-20 hours at 37°C. Cells in each well were incubated for one hour at 37°C with a Fura2-AM Fluorescent Dye/Pluronic F-27 mixture (Molecular Probes ; Eugene, Oregon) and dissolved in the medium containing Probenecid. Typical final concentrations were: 5-8 uM of Fura2-AM, 0.01% Pluronic F- 27, and 2.5 mM Probenecid (an anion exchange inhibitor that reduces transport of the hydrolyzed dye from inside the cell thereby minimizing loss of dye during the experiment). After one hour, cells were washed in a buffered solution (20 mM HEPES and Hanks Balanced Salt Solution with 1.26 mM CaCl2), pH 7.4 containing Probenecid at

a final concentration of 2.5 mM and pre-incubated for at least 30 minutes at the assay temperature of 37°C.

Typically, the above described HEPES/HBSS-based buffer containing either no additional calcium or with calcium to increase the concentration to 2 mM and various concentrations of compounds (at 5-times the final concentrations) were added to each well using the robotic multi-channel pipettor. The compounds were preincubated at 37°C for at least 30 minutes before performing the assay (at 37°C). Signals were read with dual excitation wavelengths of 340 and 380 nm and emission wavelength of 510 nm with a cut-off filter at 495 nm. The signal was reported as the ratio of emission when excited at 340 nm to the emission when excited at 380 nm [Relative Fluorescence Units (RFU)].

Ionomycin was routinely used as a positive control.

In the case of the agonist assay, the compounds at different concentrations were added to the dye-loaded cells (as described above). The increase in RFU was a measure of potency of the compound as an agonist. Exemplary results are presented in Figure 2.

Example 9 <BR> <BR> <BR> <BR> Increase in Apoptosis following Exposure of CHO/Trp-p8 Cells <BR> <BR> <BR> with Trp-P8Agonist Co7npounds at 37°C This example discloses the effectiveness of Trp-p8 agonist compounds in inducing apoptosis in Trp-p8 expressing cells.

An Annexin V/Propidium Iodide (PI) flow cytometry assay was used to provide additional insights into the mechanism of cell death induced by Trp-p8 agonist compounds. Annexin V staining detects translocation of Phosphatidylserine to the outer layer of plasma membrane, an event characteristic of apoptosis, while PI staining indicates dead cells with compromised membranes.

Cells were treated with compounds in 1% DMSO or with a 1% DMSO (control) for 24-26 hours at 37°C. The cells were briefly trypsinized under controlled conditions and stained with an Annexin V/PI reagent kit following the methodology provided by the supplier (e. g., Southern Biotech; Birmingham, Alabama). Exemplary results are presented in Figure 3.

Example 10 In Vitro Screen using a Cell Viabilitv Assav for Trp-p8 Antagonist Compounds Based upon Protection o f Trp-p8-expressing Cells from Toxic Agonist Compounds This example discloses an assay system for identifying and characterizing candidate Trp-p8 antagonist compounds.

Trp-p8 antagonists were identified by employing a cell viability assay with CHO/Trp-p8 cells at 37°C (see Example 7) with the following modification. Within the context of the present invention, compounds that protect CHO/Trp-p8 cells from the toxic effect of a control agonist thereby maintaining the viability of the CHO/Trp-p8 cell exposed to a Trp-p8 agonist is defined as antagonist. As a primary screen for antagonists, CHO/Trp-p8 cells were exposed to 10 M of test compounds in 1% dimethylsulfoxide (DMSO) or 1% DMSO plus a toxic concentration of a control agonist, D-1607. The relative viability at 10 µM, determined as described in Example 7, was a measure of the potential of the compound as a Trp-p8 antagonist--the higher the viability, the more potent the antagonist. Exemplary results are presented in Figure 4.

Table 2 Exemplary Formula VIII Trp-p8 Antagonists of Formula IVAgonist Compounds Ref. Chemical Structure R3/R24 R24 Relative No. : Viability (%) at 10 ) nM M 1457-N \/isoquinolinyl 9 Y Tetrahydro quinolinyl n 0 1475 1504 90 Y Ref. Chemical Structure R231W4 R24 Relative No. : Viability (%) at 10 ut 1582 2- (N-methyl, N-103 Phenylethyl) amino ethyl CHO 0 1588 88 w v 1664 1-phenyl - 1669 r , 1673 2-methoxybenzyl 112 v 1688 H H, 1691 3-methyl 92 Y Y 1696 Tetrahydro quinolinyl 1709 2-methoxyphenyl 95 N , a 1743 ethyl 97 Ref. Chemical Structure R23/R24 Relative No. : Viability (%) at 10 UT 9 1745} methyl H/ Hic 1781 91 1815 2- ethyl o c, 1819 Tetrahydro Fl, etc NH 1-Propyl-1, 2, 3, 4- 1838 88 pyrrolo a] pyrazine 0 -N Ly 1876"cyclohepttyl 86 tAm 1882 / Cyctohexyfsutfanylpropyi Cyclohexylsulfanylpropyl 0 Os 1883 H, 84 c o S -N -c N-/-2- c Phenylethyl) Y Y) Y Ref. Chemical Structure R23/R24 R24 Relative No. : Viability (%) at ut 1-methyl-1, 2, 3, 4- 1918 tetrahydro-94 pyrrolo 2- a] pyrazine 1920 t) tr0 1923 116 <t/ 1925 94 "/ 1937 101 o /\ 1-methyl-1, 2, 3, 4- tetrahydro-110 pyrrolo a] pyrazine H, , 1-Propyl-1, 2, 3, 4- 1941 pyrrolo 2- a] 1996"' 89 Nu "&c. 2013 \... o 95 Ref. Chemical Structure R23/R24 R24 Relative No. : Viability (%) at 10 uM Cl 1 a H, C 2018 103 oc 2044-4-butyloxyphenyl 94 H r 2045 "0 methyl 191 Nu r\ CH /1\ ß 7, 7-trimethyl-2-oxa- 2046 bicyclo [2. 2. 1] heptan-3- one 2067 ¢t r 2291 Benzyl 87 U H, H, -TY 2306 92 CH, art A- 2639 ; R2 methyl) Ref. Chemical Structure R2324-Rza Relative No. : Viability (%) at 10 ! 1- 7- Dimethoxy-l- -F. methyl-3, 4- ", '' , 2676 ylmethyl)-4- methoxy-phenyl3- 2, 3, 4, 9-tetr ahydro-lH-b- carboline CH, C- 2777 ," XNÆN OH CHAI 2865 4- phenyl 11-M 5- 3026 a 3, 4] thiadiazol-2-yl H. CH CI 2131 2, 4, 6-trichlorophenyl 59 c CHEZ Ho 2134 4, 5-dibromothiophen-2-yl 63 buzz Br 2-hydroxy-5- methylphenyl tai Q 2745'''"t," 62 A. 2752 3- (4-flourophenyl)-lH- pyrazol-4-yl N

Ref. Chemical Structure R23/le4 No. : Viability (%) at 10 AM M 3- 2754"'°,," -pyrazol-4-yl N Tetrahydro 112Table 3 Exemplary Formula VII Trp-p8 Antagonists of Formula II Agonist Compounds Ref. Chemical Structure R17 No./R'Viab au atlO at t 13 91 . pyrazolo [3, 4- 2-Pyrid 27 /F 2-Nitro-4- triflourome th 34 0'$2 ci WN :-N' chlorophen yl 36 isopropyl-5- 2-methyl- metlioxyph cyclohexyloxy)- enyl 51 ''00 /N/N/ 2-1-Phenyl-lH- chlorophen pyrazolo [3, 4- yl 67 9 CCX-, Benzyl-2-methyl- phenyl Ref. Chemical Structure Ri 17 R'9 No./R (%) at 10 uM 69 106 o 3-Methyl-5- morpholin-4-yl-2- henyl 74 81 ) r 2-methyl-2-nitro-5- quinolin-3-piperazin-1-yl- yl 93 c. CA o"isopropyl-S- 4-methyl- methoxyph cyclohexyloxy)- ex propan-2-ol 103 FNgN'4 (2, 5-Dimethyl- pyrrol-1-yl)-2- phenyl 107 i Y" IJ 2-nitro-3- trifluoromethanes phenyt 159 ir 4-pyrazolo [3, 4- flou d] pyrimidin-4-yl H 711 N 2- phenoxymethyl)- phenyl 809 3-azepanl-89 yl-5- triflourome -" phen ylamino [l, 3, 5 812, Y° yl-5- triflourome 0'O-L thoxy) phen ylamino [l, adamantyl Ref. Chemical Structure R17 No./R'Viab 0 at m 881 Q (Benzo 3] dioxol -5-ylamino)- 10b, anthra [1, 9- cd] isoxazol-6- cyclohexyl 882 p 5-86 (Benzo tF-5-ylamino}- l anthra [1, 9- cd] isoxazol-6- c 1019 thiazolo [3, 2- b] 2, 4] triazol-6- diphenyle ol 4-methylphenyl thyl 1021 thiazolo [3, 2- b] 2, 4] triazol-6- diphenylme thyl 1026 thiazolo [3, 2- b] diphenyle ol 4-methylphenyl thyl 1027 thiazolo [3, 2- b] 2, 4] triazol-6- diphenylme 4-methylphenyl thyl 1028 2-Methyl-85 thiazolo [3, 2- b] 2, 4] triazol-6- diphenylme thyl methyl 1039 thiazolo [3, 2- b] 2, 4] triazol-6- diphenyle ol 4-methylphenyl P Y YP Y thyl 1069 f" 3-benzyl-3H- 2-quinazolin-4-one- phenylethyl

Ref. Chemical Structure R17 No./R'Viab 0 at uM 1262 86 4-hydroxy- cyclohexyl 1280 87 XF F 2-chloro-4- cycloheptyl 1283 100 0 6 cyclopentyl 1284 b °J'CL cyclopentyl 1313 phenyl)-2, 5- C-dimethyl- Benzo 3] pyrazolo [1, 5- dioxol-5-a] yl-methyl 1323, 5, 6, 7, 8- tetrahydro- benzo [4, 5] thieno 2, 3-d] pyrimidin-4- 2-Pyridyl 2496 Y° 82 GH 4-methyl- chlorobenz yl ropan-2-ol Example 11 I72 Vitro Screen using a Calcium Flux Assay for Trp-p8 Antagonist Compounds Based upon their Abilities to Suppress the Calcium Influx Induced by Trp-p8 Agonists in CHO/Trp-p8 Cells This example discloses an in vitro assay system employed to further screen and characterize candidate Trp-p8 antagonists.

Trp-p8 antagonists were also screened and characterized using a calcium flux assay at 37°C as described in Example 8 with the following two distinctions : (1) the compound was pre-mixed with the control agonist or only the control agonist is added to the cells and suppression of the response to the agonist is a measure of the potency of the compound as an antagonist and (2) the compound, at different concentrations, was added to the cells followed by addition of the control agonist after 2-3 minutes and the suppression of response induced by agonist was a measure of potency of the compound as an antagonist. Exemplary results are presented in Figure 5.

Example 12 An Animal Model Svstem forAssaving the In vivo Eff cacv of Candidate Trp-p8 Agonists and Antagonists for the Treat7nent of Cancer This Example provides an animal model system suitable for determining the in vivo efficacy of candidate Trp-p8 modulators-including both agonists and antagonists.

Human prostate cancer xenografts expressing Trp-P8 (from Dr. Robert Vessella's lab in University of Washington--as assessed by in situ hybridization and immunohistochemistry using a protein specific rabbit polyclonal antibody, T-904) as well as CHO (Chinese Hamster Ovary) and EL-4 (Mouse Thymoma) cell lines were engineered to express Trp-P8 and used to establish tumor models in mice. Trp-P8 expression in these transfectants was confirmed by western blots and immunofluorescence using a Trp-p8 specific antibody (GS 2.20) as well as by response to known agonists in a calcium influx functional assay. In addition, these transfected cell lines were susceptible to killing by Trp-p8 agonists as evident from the ATP viability and apoptosis assays (as described herein in Examples 7 and 8).

A tumor model in mice was established by subcutaneously injecting CHO/Trp-P8 cells in SCID mice. Trp-p8 expression in tumors excised from these mice was confirmed by RT-PCR and western blot analysis. Further tumor model development is carried out using the human prostate cancer xenografts described above in athymic nude o r S CID mice and using an EL4/Trp-p8 transfectant in normal mice. Prostate xenografts from other sources and other cell lines that may be engineered to express Trp-p8 are also potential candidates for building more model systems.

Based on results from in vitro and in vivo evaluations, a set of trp-p8 agonists will be chosen to determine efficacy in mice. The in vitro evaluations would include potency

in cell killing assay, aqueous solubility, plasma binding study and metabolic stability (potential for a compound to be metabolized by liver as determined by using hepatocytes and/or mouse microsomes). The ira vive evaluations would include pharmacokinetics and toxicity studies. The chosen compounds will be administered to mice with Trp-p8 expressing tumors by different routes [oral, intravenous, intraperitoneal, subcutaneous, intramuscular]. Tumor reduction and survival of these mice will be evaluated at different dosages of these compounds. The compound most effective in fighting tumor will be chosen for further investigations Although the present invention has been described in some detail by way of illustration and example for purposes of clarity of understanding, changes and modifications can be carried out without departing from the scope of the invention which is intended to be limited only by the scope of the appended claims.