Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
A METHOD OF TREATING CANCER
Document Type and Number:
WIPO Patent Application WO/1999/010525
Kind Code:
A1
Abstract:
The instant invention provides for a method of inhibiting prenyl-protein transferases and treating cancer which comprises administering to a mammal a prenyl-protein transferase inhibitor which is an inhibitor of cellular processing of the H-Ras and K4B-Ras proteins. The invention in particular provides for a method of inhibiting farnesyl-protein transferase and geranylgeranyl-protein transferase type I by administering a compound that is a dual inhibitor of both of those prenyl-protein transferases. The instant invention als provides for a method of identifying such a compound, the method comprising an assay whose readout is a consequence of the biological activity or inhibition of that activity of the Ras protein, thus providing convenient identification of compounds that inhibit cellular processing of the H-Ras and K4B-Ras proteins.

Inventors:
HEIMBROOK DAVID C (US)
DEFEO-JONES DEBORAH (US)
OLIFF ALLEN I (US)
STIRDIVANT STEVEN M (US)
Application Number:
PCT/US1998/017699
Publication Date:
March 04, 1999
Filing Date:
August 26, 1998
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
MERCK & CO INC (US)
HEIMBROOK DAVID C (US)
DEFEO JONES DEBORAH (US)
OLIFF ALLEN I (US)
STIRDIVANT STEVEN M (US)
International Classes:
A61K31/00; A61K31/4178; A61K31/4427; A61K31/4439; A61K31/445; A61K31/454; A61K31/495; A61K31/496; A61K31/5513; A61K38/00; A61K45/00; A61P35/00; A61P43/00; C07D233/64; C07D233/90; C07D241/04; C07D401/06; C07D401/12; C07D403/06; C07D403/14; C12N9/10; C12N15/09; C12Q1/48; C12Q1/68; G01N33/15; G01N33/50; (IPC1-7): C12Q1/48; A01N43/00; A01N43/40; A01N43/50; A61K31/415; A61K31/445; A61K31/55; C12N9/10
Foreign References:
US5185248A1993-02-09
Other References:
CHEMICAL ABSTRACTS, 1 January 1900, Columbus, Ohio, US; abstract no. 126:8133, XP002915013
See also references of EP 1019529A4
Attorney, Agent or Firm:
MERCK & CO., INC. (NJ, US)
MERCK & CO., INC. (NJ, US)
Download PDF:
Claims:
WHAT IS CLAIMED IS:
1. A method of inhibiting prenylprotein transferases which comprises administering to a mammal in need thereof a pharmaceutically effective amount of a compound that is a dual inhibitor of farnesylprotein transferase and geranylgeranylprotein transferase type I, the compound which is characterized by: a) an inhibitory activity (IC50) of less than (<) 12 uM against K4BRas dependent activation of MAP kinases in cells; and b) an inhibitory activity (IC50) against K4BRas dependent activation of MAP kinases in cells greater than 5 fold lower than the inhibitory activity (IC50) against MyrRas dependent activation of MAP kinases in cells.
2. The method according to Claim 1 wherein the compound is further characterized by: c) inhibitory activity (IC50) of < 10 nM against HRas dependent activation of MAP kinases in cells.
3. The method according to Claim 1 wherein the inhibitory activity (IC50) of the compound against K4BRas and MyrRas dependent activation of MAP kinases in cells is determined by an assay that comprises the steps of: a) cotransfecting cells with an expression plasmid for a ras gene and an expression plasmid for a reporter construct that encodes the product of a reporter gene; b) incubating the cells in the presence of test compound; and c) analyzing an aliquot of the assay medium or a lysate of the cells for the presence of the product of the reporter gene.
4. The method according to Claim 3 wherein the product of the reporter gene is secreted alkaline phosphatase.
5. The method according to Claim 3 wherein the compound is further characterized by: c) inhibitory activity (IC50) of < 10 nM against HRas dependent activation of MAP kinases in cells; and the inhibitory activity (IC50) of the compound against K4BRas and MyrRas dependent activation of MAP kinases in cells is determined by the assay.
6. The method according to Claim 3 wherein the cells are C33a cells.
7. The method according to Claim 4 wherein the cells are C33a cells.
8. The method according to Claim 3 wherein the expression of the reporter gene is controlled by a transcription factor which is activated by MAP kinases.
9. The method according to Claim 4 wherein the expression plasmid for a reporter construct that encodes secreted alkaline phosphatase is the pDSE101 plasmid.
10. The method according to Claim 7 wherein the expression plasmid for a reporter construct that encodes secreted alkaline phosphatase is the pDSE101 plasmid.
11. The method according to Claim 4 wherein the expression plasmid for a K4Bras gene is the pSMS640 plasmid or pZiprasK4B plasmid.
12. The method according to Claim 5 wherein the expression plasmid for a Myrras gene is the pSMS600 plasmid.
13. A method of inhibiting prenylprotein transferases which comprises administering to a mammal in need thereof a pharmaceutically effective amount of a compound that is a dual inhibitor of farnesylprotein transferase and geranylgeranylprotein transferase type I, the compound which is characterized by: a) an inhibitory activity (IC50) of less than (<) 12 RM against K4BRas dependent activation of MAP kinases in cells; and b) an inhibitory activity (IC50) against K4BRas dependent activation of MAP kinases in cells greater than 5 fold lower than the inhibitory activity (IC50) against expression of the SEAP protein in cells transfected with a pCMVSEAP plasmid that constitutively expresses the SEAP protein.
14. A method of inhibiting prenylprotein transferases which comprises administering to a mammal in need thereof a pharmaceutically effective amount of a compound that is a dual inhibitor of farnesylprotein transferase and geranylgeranylprotein transferase type I, the compound which is characterized by: a) an inhibitory activity (IC50) against HRas dependent activation of MAP kinases in cells greater than 2 fold lower but less than 20,000 fold lower than the inhibitory activity (IC50) against HrasCVLL (SEQ.ID.NO.: 1) dependent activation of MAP kinases in cells; and b) an inhibitory activity (IC50) against HrasCVLL dependent activation of MAP kinases in cells greater than 5 fold lower than the inhibitory activity (IC50) against expression of the SEAP protein in cells transfected with a pCMVSEAP plasmid that constitutively expresses the SEAP protein.
15. A method of inhibiting prenylprotein transferases which comprises administering to a mammal in need thereof a pharmaceutically effective amount of a compound that is a dual inhibitor of farnesylprotein transferase and geranylgeranylprotein transferase type I, the compound which is characterized by: a) an inhibitory activity (IC50) against HRas dependent activation of MAP kinases in cells greater than 2 fold lower but less than 20,000 fold lower than the inhibitory activity (IC50) against HrasCVLL (SEQ.ID.NO.: 1) dependent activation of MAP kinases in cells; and b) an inhibitory activity (IC50) against HrasCVLL dependent activation of MAP kinases in cells greater than 5 fold lower than the inhibitory activity (IC0) against MyrRas dependent activation of MAP kinases in cells.
16. A method of inhibiting farnesylprotein transferase and geranylgeranylprotein transferase type I which comprises administering to a mammal in need thereof a pharmaceutically effective amount of a compound that is a dual inhibitor of farnesylprotein transferase and geranylgeranylprotein transferase type I, the compound which is characterized by: a) an inhibitory activity (IC50) of less than 12 I1M against K4BRas dependent activation of MAP kinases in cells; and b) an inhibitory activity (IC50) against K4BRas dependent activation of MAP kinases in cells greater than 5 fold lower than the inhibitory activity (IC50) against MyrRas dependent activation of MAP kinases in cells.
17. The method according to Claim 16 wherein the compound is further characterized by: c) inhibitory activity (IC50) of < 10 nM against HRas dependent activation of MAP kinases in cells.
18. The method according to Claim 16 wherein the inhibitory activity (IC50) of the compound against K4BRas and MyrRas dependent activation of MAP kinases in cells is determined by an assay that comprises the steps of: a) cotransfecting cells with an expression plasmid for a ras gene and an expression plasmid for a reporter construct that encodes the product of a reporter gene; b) incubating the cells in the presence of test compound; and c) analyzing an aliquot of the assay medium or a lysate of the cells for the presence of the product of the reporter gene.
19. The method according to Claim 18 wherein the product of the reporter gene is secreted alkaline phosphatase.
20. The method according to Claim 18 wherein the compound is further characterized by: c) inhibitory activity (IC50) of < 10 nM against HRas dependent activation of MAP kinases in cells; and the inhibitory activity (IC50) of the compound against K4BRas and MyrRas dependent activation of MAP kinases in cells is determined by the assay.
21. The method according to Claim 18 wherein the cells are C33a cells.
22. The method according to Claim 19 wherein the cells are C33a cells.
23. The method according to Claim 19 wherein the expression of the reporter gene is controlled by a transcription factor which is activated by MAP kinases.
24. The method according to Claim 19 wherein the expression plasmid for a reporter construct that encodes secreted alkaline phosphatase is the pDSE101 plasmid.
25. The method according to Claim 23 wherein the expression plasmid for a reporter construct that encodes secreted alkaline phosphatase is the pDSE101 plasmid.
26. The method according to Claim 18 wherein the expression plasmid for a K4Bras gene is the pSMS640 plasmid or pZiprasK4B plasmid.
27. The method according to Claim 18 wherein the expression plasmid for a Myrras gene is the pSMS600 plasmid.
28. A method of treating cancer which comprises administering to a mammal in need thereof a pharmaceutically effective amount of a compound that is a dual inhibitor of farnesylprotein transferase and geranylgeranylprotein transferase type I, the compound which is characterized by: a) an inhibitory activity (IC50) of less than 12 uM against K4BRas dependent activation of MAP kinases in cells; and b) an inhibitory activity (IC50) against K4BRas dependent activation of MAP kinases in cells greater than 5 fold lower than the inhibitory activity (IC50) against MyrRas dependent activation of MAP kinases in cells.
29. The method according to Claim 28 wherein the compound is further characterized by: c) inhibitory activity (IC50) of < 10 nM against HRas dependent activation of MAP kinases in cells.
30. The method according to Claim 28 wherein the inhibitory activity (IC50) of the compound against K4BRas and MyrRas dependent activation of MAP kinases in cells is determined by an assay that comprises the steps of: a) cotransfecting cells with an expression plasmid for a ras gene and an expression plasmid for a reporter construct that encodes the product of a reporter gene; b) incubating the cells in the presence of test compound; and c) analyzing an aliquot of the assay medium or a lysate of the cells for the presence of the product of the reporter gene.
31. The method according to Claim 30 wherein the product of the reporter gene is secreted alkaline phosphatase.
32. The method according to Claim 30 wherein the compound is further characterized by: c) inhibitory activity (IC50) of < 10 nM against HRas dependent activation of MAP kinases in cells.
33. The method according to Claim 30 wherein the cells are C33a cells.
34. The method according to Claim 31 wherein the cells are C33a cells.
35. The method according to Claim 30 wherein the expression of the reporter gene is controlled by a transcription factor which is activated by MAP kinases.
36. The method according to Claim 31 wherein the expression plasmid for a reporter construct that encodes secreted alkaline phosphatase is the pDSE101 plasmid.
37. The method according to Claim 35 wherein the expression plasmid for a reporter construct that encodes secreted alkaline phosphatase is the pDSE101 plasmid.
38. The method according to Claim 30 wherein the expression plasmid for a K4Bras gene is the pSMS640 plasmid or pZiprasK4B plasmid.
39. The method according to Claim 30 wherein the expression plasmid for a Myrras gene is the pSMS600 plasmid or the pSMS621 plasmid.
40. The method according to Claim 28 wherein the cancer is cancer characterized by a mutated K4Bras gene.
41. A method of treating cancer which comprises administering to a mammal in need thereof a pharmaceutically effective amount of a compound that is a dual inhibitor of farnesylprotein transferase and geranylgeranylprotein transferase type I, the compound which is characterized by: a) an inhibitory activity (IC50) of less than (<) 12 uM against K4BRas dependent activation of MAP kinases in cells; and b) an inhibitory activity (IC50) against K4BRas dependent activation of MAP kinases in cells greater than 5 fold lower than the inhibitory activity (IC50) against expression of the SEAP protein in cells transfected with a pCMVSEAP plasmid that constitutively expresses the SEAP protein.
42. A method of treating cancer which comprises administering to a mammal in need thereof a pharmaceutically effective amount of a compound that is a dual inhibitor of farnesylprotein transferase and geranylgeranylprotein transferase type I, the compound which is characterized by: a) an inhibitory activity (IC50) against HRas dependent activation of MAP kinases in cells greater than 2 fold lower but less than 20,000 fold lower than the inhibitory activity (IC50) against HrasCVLL (SEQ.ID.NO.: 1) dependent activation of MAP kinases in cells; and b) an inhibitory activity (IC50) against HrasCVLL dependent activation of MAP kinases in cells greater than 5 fold lower than the inhibitory activity (IC50) against expression of the SEAP protein in cells transfected with a pCMVSEAP plasmid that constitutively expresses the SEAP protein.
43. A method of treating cancer which comprises administering to a mammal in need thereof a pharmaceutically effective amount of a compound that is a dual inhibitor of farnesylprotein transferase and geranylgeranylprotein transferase type I, the compound which is characterized by: a) an inhibitory activity (IC0) against HRas dependent activation of MAP kinases in cells greater than 2 fold lower but less than 20,000 fold lower than the inhibitory activity (IC50) against HrasCVLL (SEQ.ID.NO.: 1) dependent activation of MAP kinases in cells; and b) an inhibitory activity (ICso) against HrasCVLL dependent activation of MAP kinases in cells greater than 5 fold lower than the inhibitory activity (IC50) against MyrRas dependent activation of MAP kinases in cells.
44. The method according to Claim 28 wherein the compound is selected from: 1[2(R)Amino3mercaptopropyl]2(S)[3pyridyl)methoxyethyl)]4 (1 naphthoyl)piperazine 1[2(R)Amino3mercaptopropyl]2(S)(benzyloxymethyl)4(1 naphthoyl)piperazine 1 [2(R)Amino3mercaptopropyl] 2(S)(benzyloxymethyl)4 [7 (2,3 dihydrobenzofuroyl)]piperazine 1[2(R)Amino3mercaptopropyl]2(S)(benzamido)4(1 naphthoyl)piperazine 1[2(R)Amino3mercaptopropyl]2(S)[4(5dimethylamino1 naphyhalenesulfonamido)1butyl]4(1naphthoyl)piperazine N[2(S)(1 (4Nitrophenylmethy1) 1Himidazol5ylacetyl)amino3 (S) methylpentyl] N 1 naphthylmethylglycylmethionine N[2(S)(1 (4Nitrophenylmethyl) 1 Himidazol5ylacetyl)amino3 (S) methylpentyl]N1naphtylmethylglycylmethionine methyl ester N[2(S)([1(4cyanobenzyl)1Himidazol5yl]acetylamino)3(S) methylpenytl]N(1napthylmethyl)glycylmethionine N[2(S)([1(4cyanobenzyl)1Himidazol5yl]acetylamino)3(S) methylpenytl]N(1napthylmethyl)glycylmethionine methyl ester 2(S)nButyl4(1naphthoyl)1[1(2naphthylamethyl)imidazol5 ylmethyl]piperazine 2(S)nButyl1[1(4cyanobenzyl)imidazol5ylmethyl]4(1 naphthoyl)piperazine 1{[1(4cyanobenzyl)1Himidazol5yl]acetyl}2(S)n butyl4(1naphtoyl)piperazine 1(3chlorophenyl)4[1(4cyanobenzyl)imidazolylmethyl]2 piperazinone 1phenyl4[1(4cyanobenzyl)1Himidazol5ylethyl]piperazin2one 1(3trifluoromethylphenyl)4[1(4cyanobenzyl)1Himidazol5 ylmethyl]piperazin2one 1(3bromophenyl)4[1(4cyanobenzyl)1Himidazol5ylmethyl] piperazin2one 5(S)(2[2,22trifluoroethoxy]ethyl)1(3trifluoromethylphenyl)4[1 (4cyanobenzyl)4imidazolylmethyl]piperazin2one 1(5,6,7,8tetrahydronaphthyl)4[1(4cyanobenzyl)1Himidazol5 ylmethyl]piperazin2one 1(2methyl3chlorophenyl)4[1(4cyanobenzyl)4 imidazolylmethyl)piperazin2one 2(RS) { [1 (Naphth2ylmethyl) 1Himidazol5yl)] acetyl 1 amino3(t butoxycarbonyl)amino N (2methylbenzyl) propionamide N{1(4Cyanobenzyl)1Himidazol5ylmethyl}4(R)benzyloxy2(S) ( N'acetylN'3chlorobenzyl ) aminomethylpyrrolidine N{1(4Cyanobenzyl)1Himidazol5ylethyl}4(R)benzyloxy2(S) {N'acetylN'3chlorobenzyl}aminomethyl pyrrolidine 1[1(4Cyanobenzyl)1Himidazol5ylacetyl] pyrrolidin2(S) ylmethyl](N2methylbenzyl)glycine N'(3chlorophenylmethyl) amide 1[1(4Cyanobenzyl)1Himidazol5ylacetyl] pyrrolidin2(S) ylmethyl](N2methylbenzyl)glycine N'methylN'(3 chlorophenylmethyl) amide <BR> <BR> <BR> <BR> <BR> (S)2[(1(4Cyanobenzyl)5imidazolylmethyl) amino]N <BR> <BR> <BR> <BR> (benzyloxycarbonyl)N (3 chlorobenzyl)4 (methane sulfonyl)butanamine 1(3,5Dichlorobenzenesulfonyl)3(S)[N(1(4cyanobenzyl)1H imidazol5ylethyl) carbamoyl] piperidine N ([1 (4Cyanobenzyl) 1 Himidazol5y1]methyl 1 4(3 methylphenyl) 4hydroxy piperidine, N{[1(4Cyanobenzyl)1Himidazol5yl]methyl}4(3chlorophenyl)4 hydroxy piperidine, 4[1(4cyanobenzyl)5imidazolylmethyl]1(2,3dimethylphenyl) piperazine2,3dione 1(2(3Trifluoromethoxyphenyl)pyrid5ylmethyl)5(4 cyanobenzyl) imidazole 4{5[1(3Chlorophenyl)2oxo1,2dihydropyridin4ylmethyl] imidazol1ylmethyl}2methoxybenzonitrile 3(R)3[1(4Cyanobenzyl)imidazol5ylethylamino]5phenyl1(2,2,2 trifluorethyl)Hbenzo[e][1,4] diazepine 3(S)3[1(4Cyanobenzyl) imidazol5yl]ethylamino]5phenyl1 (2,2,2trifluroethyl)Hbenzo[e][1,4] diazepine N[1(4Cyanobeznyl)1Himidazol5ylacetyl)pyrrolidin2(S) ylmethyl]N(1napthylmethyl)glycylmethionine N[1(4Cyanobenzyl)1Himidazol5ylacetyl)pyrrolidin2(S) ylmethyl]N(1napthylmethyl)glycylmethionine methyl ester N[1(1HImidazol4ylpropionyl)pyrroldin2(S)ylmethyl] N(2 methoxybenzyl)glycylmethionine N[1(1HImidazol4ylpropionyl)pyrrolidin2(S)ylmethyl] N(2 methoxybenzyl)glycylmethionine methyl ester 2(S)(4Acetamido1butyl)1[2(R)amino3mercaptopropyl]4(1 naphthoyl)piperazine 2(RS){[1(Naphth2ylmethyl)1Himidazol5yl)] acetyl}amino3(t butoxycarbonyl)aminoNcyclohexylpropionamide 1{2(R,S)[1 (4cyanobenzyl) 1 Himidazol5yl]propanoyl }2(S)n butyl4(1naphthoyl)piperazine 1[1(4cyanobenzyl)imidazol5ylmethyl]4(diphenylmethyl)piperazine 1(Diphenylmethyl)3(S)[N(1(4cyanobeznyl)2methyl1Himidazol 5ylethyl)N(acetyl) aminomethyl] piperidine N[1(1HImidazol4ylpropionyl)pyrrolidin2(S)ylmethyl] N(2 chlorobenzyl)glycylmethionine N[1(1HImidazo4ylpropionyl)pyrrolidin2(S)ylmethyl] N(2 chlorobeznyl)glycylmethionine methyl ester 3(R)3[1(4Cyanobenzyl)imidazol5ylmethylamino]5phenyl1 (2,2,2trifluoroethyl)Hbenzo[e][1,4] diazepine <BR> <BR> <BR> <BR> <BR> <BR> 1 (3 trifluoromethoxyphenyl)4 [1 (4cyanobenzyl)imidazolylmethyll 2piperazinone 1(2,5dimethylphenyl)4[1(4cyanobenzyl)imidazolylmethyl]2 piperazinone 1(3methylphenyl)4[1(4cyanobenzyl)imidazolylmethyl]2 piperazinone <BR> <BR> <BR> <BR> <BR> <BR> 1 (3 iodophenyl)4 [1 (4cyanobenzyl)imidazolylmethyl] 2piperazinone 1 (3 chlorophenyl)4 [1 (4cyano3 methoxybenzyl)imidazolylmethyl] 2piperazinone 1(3trifluoromethoxyphenyl)4[1(4cyano3 methoxybenzyl)imidazolyl methyl]2piperazinone 4[((1(4cyanobenzyl)5imidazolyl)methyl)amino]benzophenone 1(1{[3(4cyanobenzyl)3Himidazol4yl]acetyl}pyrrolidin2(S) ylmethyl)3 (S)ethylpyrrolidine2(S)carboxylic acid 3 chloro benzylamide or the pharmaceutically acceptable salt thereof.
45. A method of inhibiting prenylprotein transferases which comprises administering to a mammal in need thereof a pharma ceutically effective amount of a compound, which is characterized by: a) an inhibitory activity (IC50) of less than 12 uM against K4BRas dependent activation of MAP kinases in cells.
46. An assay for identifying a prenylprotein transferase inhibitor which is efficacious in vivo as an inhibitor of the biological activity of a Ras protein which comprises the steps of: a) cotransfecting cells with an expression plasmid for a ras gene and an expression plasmid for a reporter construct that encodes the product of a reporter gene; b) incubating the cells in the presence of test compound; and c) analyzing an aliquot of the assay medium or a lysate of the cells for the presence of the product of the reporter gene.
47. The assay according to Claim 46 wherein the product of the reporter gene is secreted alkaline phosphatase.
48. The assay according to Claim 46 wherein the cells are C33a cells.
49. The assay according to Claim 47 wherein the expression plasmid for a reporter construct that encodes secreted alkaline phosphatase is the pDSE101 plasmid.
50. The assay according to Claim 46 wherein the Ras protein is the K4BRas.
51. The assay according to Claim 50 wherein the prenyl protein transferase inhibitor is a dual inhibitor of farnesylprotein transferase and geranylgeranylprotein transferase type I.
52. The assay according to Claim 50 wherein the expression plasmid for a reporter construct that encodes secreted alkaline phosphatase is the pDSE101 plasmid.
53. A method for identifying a prenylprotein transferase inhibitor which is efficacious in vivo as an inhibitor of the growth of cancer cells which comprises the steps of: a) evaluating the test compound in the assay according to Claim 46 wherein the ras gene is Kras b) evaluating the test compound in the assay according to Claim 46 wherein the ras gene is Myrras ; and c) comparing the activity of the test compound against MyrRas dependent activation of MAP kinases in the assay according to Claim 46 with the activity of the test compound against KRas dependent activation of MAP kinases in the assay according to Claim 46.
54. The method according to Claim 53 wherein the Kras gene is K4Bras.
55. The method according to Claim 54 wherein the prenylprotein transferase inhibitor is a dual inhibitor of farnesyl protein transferase and geranylgeranylprotein transferase type I.
56. The method according to Claim 54 which further comprises the additional step of: d) evaluating the test compound in an assay, which comprises the steps of: a) cotransfecting cells with an expression plasmid for a ras gene and an expression plasmid for a reporter construct that encodes the product of a reporter gene; b) incubating the cells in the presence of test compound; and c) analyzing an aliquot of the assay medium or a lysate of the cells for the presence of the product of the reporter gene. wherein the ras gene is Hras.
57. The method according to Claim 56 wherein the product of the reporter gene is secreted alkaline phosphatase.
58. The method according to Claim 54 which further comprises the additional step of: d) evaluating the test compound in an assay, which comprises the steps of: a) cotransfecting cells with an expression plasmid for a ras gene and an expression plasmid for a reporter construct that encodes the product of a reporter gene; b) incubating the cells in the presence of test compound; and c) analyzing an aliquot of the assay medium or a lysate of the cells for the presence of the product of the reporter gene. wherein the ras gene is HrasCVLL.
59. The method according to Claim 58 wherein the product of the reporter gene is secreted alkaline phosphatase.
60. The method according to Claim 56 which further comprises the additional steps of: d) evaluating the test compound in an assay, which comprises the steps of: a) cotransfecting cells with an expression plasmid for a ras gene and an expression plasmid for a reporter construct that encodes the product of a reporter gene; b) incubating the cells in the presence of test compound; and c) analyzing an aliquot of the assay medium or a lysate of the cells for the presence of the product of the reporter gene. wherein the ras gene is HrasCVLL.
61. The method according to Claim 60 wherein the product of the reporter gene is secreted alkaline phosphatase.
62. A method for identifying a prenylprotein transferase inhibitor which is efficacious in vivo as an inhibitor of the growth of cancer cells which comprises the steps of: a) evaluating the test compound in the assay according to Claim 46 wherein the ras gene is Hras; b) evaluating the test compound in the assay according to Claim 46 wherein the ras gene is HrasCVLL; c) evaluating the test compound in the assay according to Claim 46 wherein the ras gene is Myrras ; and d) comparing the activity of the test compound against MyrRas dependent activation of MAP kinases in the assay according to Claim 46 with the activity of the test compound against HRas dependent activation of MAP kinases in the assay according to Claim 46 and HRas CVLL dependent activation of MAP kinases in the assay according to Claim 46.
63. The method according to Claim 62 wherein the product of the reporter gene is secreted alkaline phosphatase.
64. The method according to Claim 62 wherein the prenylprotein transferase inhibitor is a dual inhibitor of farnesyl protein transferase and geranylgeranylprotein transferase type I.
65. A method for identifying a prenylprotein transferase inhibitor which is efficacious in vivo as an inhibitor of the growth of cancer cells which comprises the steps of: a) evaluating the test compound in the assay according to Claim 46 wherein the ras gene is Nras; b) evaluating the test compound in the assay according to Claim 46 wherein the ras gene is Myrras ; and c) comparing the activity of the test compound against Myr Ras dependent activation of MAP kinases in the assay according to Claim 46 with the activity of the test compound against N Ras dependent activation of MAP kinases in the assay according to Claim 46.
66. The method according to Claim 65 wherein the product of the reporter gene is secreted alkaline phosphatase.
67. A method for identifying a prenylprotein transferase inhibitor which is efficacious in vivo as an inhibitor of the growth of cancer cells which comprises the steps of: a) evaluating the test compound in the assay according to Claim 46 wherein the ras gene is Hras; b) evaluating the test compound in the assay according to Claim 46 wherein the ras gene is HrasCVLL; c) evaluating the test compound in the assay according to Claim 46 wherein the ras gene is Myrras ; and d) comparing the activity of the test compound against Hras CVLL dependent activation of MAP kinases in the assay according to Claim 46 with the activity of the test compound against HRas dependent activation of MAP kinases in the assay according to Claim 46; e) comparing the activity of the test compound against MyrRas dependent activation of MAP kinases in the assay according to Claim 46 with the activity of the test compound against Hras CVLL dependent activation of MAP kinases in the assay according to Claim 46.
68. A method for identifying a prenylprotein transferase inhibitor which is efficacious in vivo as an inhibitor of the growth of cancer cells which comprises the steps of: a) evaluating the test compound in the assay according to Claim 46 wherein the ras gene is Kras; b) evaluating the test compound in an assay, which comprises the steps of: a) transfecting cells with a pCMVSEAP plasmid; b) incubating the cells in the presence of test compound; and c) analyzing an aliquot of the assay medium or a lysate of the cells for the presence of the product of the reporter gene; c) comparing the activity of the test compound against expresion of the SEAP protein in cells transfected with the pCMVSEAP plasmid with the activity of the test compound against Kras dependent activation of MAP kinases in the assay according to Claim 46.
69. A method for identifying a prenylprotein transferase inhibitor which is efficacious in vivo as an inhibitor of the growth of cancer cells which comprises the steps of: a) evaluating the test compound in the assay according to Claim 46 wherein the ras gene is Hras; b) evaluating the test compound in the assay according to Claim 46 wherein the ras gene is HrasCVLL; c) evaluating the test compound in an assay, which comprises the steps of: a) transfecting cells with a pCMVSEAP plasmid; b) incubating the cells in the presence of test compound; and c) analyzing an aliquot of the assay medium or a lysate of the cells for the presence of the product of the reporter gene; d) comparing the activity of the test compound against Hras CVLL dependent activation of MAP kinases in the assay according to Claim 46 with the activity of the test compound against HRas dependent activation of MAP kinases in the assay according to Claim 46; e) comparing the activity of the test compound against expresion of the SEAP protein in cells transfected with the pCMVSEAP plasmid with the activity of the test compound against against HrasCVLL dependent activation of MAP kinases in the assay according to Claim 46.
70. The method according to Claim 65 wherein the prenylprotein transferase inhibitor is a dual inhibitor of farnesyl protein transferase and geranylgeranylprotein transferase type I.
71. A method for identifying a combination of a selective inhibitor of geranylgeranylprotein transferase type I and a selective inhibitor of farnesylprotein transferase which is efficacious in vivo in the inhibition of the growth of cancer cells which comprises the steps of: a) evaluating a test combination of a selective inhibitor of geranylgeranylprotein transferase type I and a selective inhibitor of farnesylprotein transferase in the assay according to Claim 46 wherein the ras gene is selected from K4Bras and Nras; b) evaluating the test combination of a selective inhibitor of geranylgeranylprotein transferase type I and a selective inhibitor of farnesylprotein transferase in the assay according to Claim 46 wherein the ras gene is Myrras ; and c) comparing the activity of the test combination of a selective inhibitor of geranylgeranylprotein transferase type I and a selective inhibitor of farnesylprotein transferase against Myr Ras dependent activation of MAP kinases in the assay according to Claim 46 with the activity of the test combination of a selective inhibitor of geranylgeranylprotein transferase type I and a selective inhibitor of farnesylprotein transferase against dependent activation of MAP kinases by the protein encoded by the gene of step a) in the assay according to Claim 46.
72. The method according to Claim 71 wherein the product of the reporter gene is secreted alkaline phosphatase.
73. The method according to Claim 71 wherein the ras gene is K4Bras.
74. The method according to Claim 71 wherein the ras gene is Nras.
75. The method according to Claim 71 which further comprises the additional step of: d) evaluating the test combination of a selective inhibitor of geranylgeranylprotein transferase type I and a selective inhibitor of farnesylprotein transferase in the assay according to Claim 46 wherein the ras gene is Hras.
76. The method according to Claim 71 which further comprises the additional step of: d) evaluating the test combination of a selective inhibitor of geranylgeranylprotein transferase type I and a selective inhibitor of farnesylprotein transferase in the assay according to Claim 46 wherein the ras gene is HrasCVLL.
77. The method according to Claim 71 which further comprises the additional steps of: d) evaluating the test combination of a selective inhibitor of geranylgeranylprotein transferase type I and a selective inhibitor of farnesylprotein transferase in the assay according to Claim 46 wherein the ras gene is Hras ; and e) evaluating the test combination of a selective inhibitor of geranylgeranylprotein transferase type I and a selective inhibitor of farnesylprotein transferase in the assay according to Claim 46 wherein the ras gene is HrasCVLL.
78. A method for identifying a combination of a selective inhibitor of geranylgeranylprotein transferase type I and a selective inhibitor of farnesylprotein transferase which is efficacious in vivo in the inhibition of the growth of cancer cells which comprises evaluating a test combination of a selective inhibitor of geranylgeranylprotein transferase type I and a selective inhibitor of farnesylprotein transferase in the assay according to Claim 46 by the steps of: a) evaluating the test combination in the assay according to Claim 46 wherein the ras gene is Kras; b) evaluating the test combination in an assay, which comprises the steps of: a) transfecting cells with a pCMVSEAP plasmid; b) incubating the cells in the presence of test combination; and c) analyzing an aliquot of the assay medium or a lysate of the cells for the presence of the product of the reporter gene; c) comparing the activity of the test combination against expresion of the SEAP protein in cells transfected with the pCMVSEAP plasmid with the activity of the test combination against against Kras dependent activation of MAP kinases in the assay according to Claim 46.
79. A method for identifying a combination of a selective inhibitor of geranylgeranylprotein transferase type I and a selective inhibitor of farnesylprotein transferase which is efficacious in vivo in the inhibition of the growth of cancer cells which comprises evaluating a test combination of a selective inhibitor of geranylgeranylprotein transferase type I and a selective inhibitor of farnesylprotein transferase in the assay according to Claim 46 by the steps of: a) evaluating the test combination in the assay according to Claim 46 wherein the ras gene is Hras; b) evaluating the test combination in the assay according to Claim 46 wherein the ras gene is HrasCVLL; c) evaluating the test combination in an assay, which comprises the steps of: a) transfecting cells with a pCMVSEAP plasmid; b) incubating the cells in the presence of test combination; and c) analyzing an aliquot of the assay medium or a lysate of the cells for the presence of the product of the reporter gene; d) comparing the activity of the test combination against Hras CVLL dependent activation of MAP kinases in the assay according to Claim 46 with the activity of the test combination against HRas dependent activation of MAP kinases in the assay according to Claim 46; e) comparing the activity of the test combination against expresion of the SEAP protein in cells transfected with the pCMVSEAP plasmid with the activity of the test combination against against HrasCVLL dependent activation of MAP kinases in the assay according to Claim 46.
Description:
TITLE OF THE INVENTION A METHOD OF TREATING CANCER RELATED APPLICATION The present patent application is a continuation-in-part application of copending provisional application Serial No.

60/057,102, filed August 27,1997.

BACKGROUND OF THE INVENTION The present invention relates to methods of inhibiting prenyl-protein transferases and treating cancer which utilize prenyl- protein transferase inhibitors which inhibit the cellular processing of both the H-Ras protein and the K4B-Ras protein. The present invention also relates to a method of identifying such compounds.

Prenylation of proteins by intermediates of the isoprenoid biosynthetic pathway represents a class of post- translational modification (Glomset, J. A., Gelb, M. H., and Farnsworth, C. C. (1990), Trends Biochem. Sci. 15, 139-142; Maltese, W. A. (1990), FASEB J. 4, 3319-3328). This modification typically is required for the membrane localization and function of these proteins. Prenylated proteins share characteristic C-terminal sequences including CAAX (C, Cys; A, an aliphatic amino acid; X, another amino acid), XXCC, or XCXC. Three post-translational processing steps have been described for proteins having a C-terminal CAAX sequence: addition of either a 15 carbon (farnesyl) or 20 carbon (geranylgeranyl) isoprenoid to the Cys residue, proteolytic cleavage of the last 3 amino acids, and methylation of the new C-terminal carboxylate (Cox, A. D. and Der, C. J. (1992a), Critical Rev. Oncogenesis 3:365-400; Newman, C. M.

H. and Magee, A. I. (1993), Biochem. Biophys. Acta 1155:79-96).

Some proteins may also have a fourth modification: palmitoylation of one or two Cys residues N-terminal to the farnesylated Cys.

While some mammalian cell proteins terminating in XCXC are carboxymethylated, it is not clear whether carboxy methylation

follows prenylation of proteins terminating with a XXCC motif (Clarke, S. (1992). Annu. Rev. Biochem. 61, 355-386). For all of the prenylated proteins, addition of the isoprenoid is the first step and is required for the subsequent steps (Cox, A. D. and Der, C. J.

(1992a), Critical Rev. Oncogenesis 3:365-400; Cox, A. D. and Der, C. J. (1992b) Current Opinion Cell Biol. 4:1008-1016).

Three enzymes have been described that catalyze protein prenylation: farnesyl-protein transferase (FPTase), geranylgeranyl- protein transferase type I (GGPTase-I), and geranylgeranyl-protein transferase type-II (GGPTase-II, also called Rab GGPTase). These enzymes are found in both yeast and mammalian cells (Clarke, 1992; Schafer, W. R. and Rine, J. (1992) Annu. Rev. Genet. 30:209-237).

Each of these enzymes selectively uses farnesyl diphosphate or geranylgeranyl diphosphate as the isoprenoid donor and selectively recognizes the protein substrate. FPTase farnesylates CAAX- containing proteins that end with Ser, Met, Cys, Gln or Ala. For FPTase, CAAX tetrapeptides comprise the minimum region required for interaction of the protein substrate with the enzyme. The enzymological characterization of these three enzymes has demonstrated that it is possible to selectively inhibit one with little inhibitory effect on the others (Moores, S. L., Schaber, M. D., Mosser, S. D., Rands, E., O'Hara, M. B., Garsky, V. M., Marshall, M. S., Pompliano, D. L., and Gibbs, J. B., J. Biol. Chem., 266:17438 (1991), U.S. Pat. No. 5,470,832).

The prenylation reactions have been shown genetically to be essential for the function of a variety of proteins (Clarke, 1992; Cox and Der, 1992a; Gibbs, J. B. (1991). Cell 65 : 1-4 ; Newman and Magee, 1993; Schafer and Rine, 1992). This requirement often is demonstrated by mutating the CAAX Cys acceptors so that the proteins can no longer be prenylated. The resulting proteins are devoid of their central biological activity. These studies provide a genetic "proof of principle" indicating that inhibitors of prenylation can alter the physiological responses regulated by prenylated proteins.

The Ras protein is part of a signaling pathway that links cell surface growth factor receptors to nuclear signals initiating cellular proliferation. Biological and biochemical studies of Ras action indicate that Ras functions like a G-regulatory protein. In the inactive state, Ras is bound to GDP. Upon growth factor receptor activation, Ras is induced to exchange GDP for GTP and undergoes a conformational change. The GTP-bound form of Ras propagates the growth stimulatory signal until the signal is terminated by the intrinsic GTPase activity of Ras, which returns the protein to its inactive GDP bound form (D.R. Lowy and D.M. Willumsen, Ann.

Rev. Biochem. 62:851-891(1993)). Activation of Ras leads to activation of multiple intracellular signal transduction pathways, including the MAP Kinase pathway and the Rho/Rac pathway (Joneson et al., Science 271:810-812). One consequence of activation of the MAP Kinase pathway is activation of transcription factors, for example Elk-l, and transcription of specific proteins (R. Treisman, Current Opinion in Genetics and Development (1994) 4:96-101, and references therein).

Mutated ras genes are found in many human cancers, including colorectal carcinoma, exocrine pancreatic carcinoma, and myeloid leukemias. The protein products of these genes are defective in their GTPase activity and constitutively transmit a growth stimulatory signal.

The Ras protein is one of several proteins that are known to undergo post-translational modification. Farnesyl-protein transferase utilizes farnesyl pyrophosphate to covalently modify the Cys thiol group of the Ras CAAX box with a farnesyl group (Reiss et al., Cell, 62:81-88 (1990); Schaber et al., J. Biol. Chem., 265:14701-14704(1990); Schafer et al., Science, 249:1133-1139 (1990); Manne et al., Proc. Natl. Acad. Sci USA, 87:7541-7545 (1990)).

Ras must be localized to the plasma membrane for both normal and oncogenic functions. At least 3 post-translational modi-

fications are involved with Ras membrane localization, and all 3 modifications occur at the C-terminus of Ras. The Ras C-terminus contains a sequence motif termed a "CAAX" or "Cys-Aaa-Aaa-Xaa" box (Cys is cysteine, Aaa is an aliphatic amino acid, the Xaa is any amino acid) (Willumsen et al., Nature 310:583-586 (1984)).

Depending on the specific sequence, this motif serves as a signal sequence for the enzymes farnesyl-protein transferase or geranyl geranyl-protein transferase, which catalyze the alkylation of the cysteine residue of the CAAX motif with a C15 or C20 isoprenoid, respectively. (S. Clarke., Ann. Rev. Biochem. 61:355-386 (1992); W.R. Schafer and J. Rine, Ann. Rev. Genetics 30:209-237 (1992)).

Other farnesylated proteins include the Ras-related GTP-binding proteins such as RhoB, fungal mating factors, the nuclear lamins, and the gamma subunit of transducin. James, et al., J. Biol. Chem. 269, 14182 (1994) have identified a peroxisome associated protein Pxf which is also farnesylated. James, et al., have also suggested that there are farnesylated proteins of unknown structure and function in addition to those listed above.

Inhibitors of farnesyl-protein transferase (FPTase) have been described in two general classes. The first class includes analogs of farnesyl diphosphate (FPP), while the second is related to protein substrates (e.g., Ras) for the enzyme. The peptide derived inhibitors that have been described are generally cysteine containing molecules that are related to the CAAX motif that is the signal for protein prenylation. (Schaber et al., ibid; Reiss et. al., ibid; Reiss et al., PNAS, 88:732-736 (1991)). Such inhibitors may inhibit protein prenylation while serving as alternate substrates for the farnesyl-protein transferase enzyme, or may be purely competitive inhibitors (U.S. Patent 5,141,851, University of Texas; N.E. Kohl et al., Science, 260:1934-1937 (1993); Graham, et al., J. Med.

Chem., 37, 725 (1994)).

Mammalian cells express four types of Ras proteins (H-, N-, K4A-, and K4B-Ras) among which K4B-Ras is the most frequently mutated form of Ras in human cancers. The genes that

encode these proteins are abbreviated H-ras, N-ras, K4A-ras and K4B-ras respectively. H-ras is an abbreviation for Harvey-ras. K4A- ras and K4B-ras are abbreviations for the Kirsten splice variants of ras that contain the 4A and 4B exons, respectively. Inhibition of farnesyl-protein transferase has been shown to block the growth of H-ras-transformed cells in soft agar and to modify other aspects of their transformed phenotype. It has also been demonstrated that certain inhibitors of farnesyl-protein transferase selectively block the processing of the H-Ras oncoprotein intracellularly (N.E. Kohl et al., Science, 260:1934-1937 (1993) and G.L. James et al., Science, 260:1937-1942 (1993). Recently, it has been shown that an inhibitor of farnesyl-protein transferase blocks the growth of H-ras-dependent <BR> <BR> tumors in nude mice (N.E. Kohl et al., Proc. Natl. Acad. Sci U. S. A., 91:9141-9145 (1994) and induces regression of mammary and salivary carcinomas in H-ras transgenic mice (N.E. Kohl et al., Nature Medicine, 1:792-797 (1995).

Indirect inhibition of farnesyl-protein transferase in vivo has been demonstrated with lovastatin (Merck & Co., Rahway, NJ) and compactin (Hancock et al., ibid; Casey et al., ibid; Schafer et al., Science 245:379 (1989)). These drugs inhibit HMG-CoA reductase, the rate limiting enzyme for the production of polyiso- prenoids including farnesyl pyrophosphate. Inhibition of farnesyl pyrophosphate biosynthesis by inhibiting HMG-CoA reductase blocks Ras membrane localization in cultured cells.

It has been disclosed that the lysine-rich region and terminal CVIM sequence of the C-terminus of K4B-Ras confer resistance to inhibition of the cellular processing of that protein by certain selective FPTase inhibitors. (James, et al., J. Biol. Chem.

270, 6221 (1995)) Those FPTase inhibitors were effective in inhibiting the processing of H-Ras proteins. James et al., suggested that prenylation of the K4B-Ras protein by GGTase contributed to the resistance to the selective FPTase inhibitors. (Zhang et al, J. Biol.

Chem. 272 :10232-239 (1997); Rowell et al, J. Biol. Chem. 272

:14093-14097 (1997); Whyte et al, J. Biol. Chem. 272 :14459-14464 (1997)).

Several groups of scientists have recently disclosed compounds that are non-selective FPTase/GGTase inhibitors.

(Nagasu et al. Cancer Research, 55:5310-5314(1995); PCT application WO 95/25086).

Recently, an assay useful to identify inhibitors of FPTase which incorporates all or part of the K4B-Ras protein substrate has been disclosed (PCT Appln. WO 96/34113).

It is the object of the present invention to provide a method of inhibiting prenyl-protein transferase which utilizes compounds that are prenyl-protein transferase inhibitors and which inhibit cellular processing of the H-Ras and K4B-Ras proteins.

A composition which comprises such an inhibitor compound is also used in the present invention to treat cancer.

It is also the object of the instant invention to provide a method for identifying a prenyl-protein transferase inhibitor which is an inhibitor of cellular processing of the H-Ras and K4B-Ras proteins.

SUMMARY OF THE INVENTION The instant invention provides for a method of inhibiting prenyl-protein transferases and treating cancer which comprises administering to a mammal a prenyl-protein transferase inhibitor which is an inhibitor of cellular processing of the H-Ras and K4B-Ras proteins. The invention in particular provides for a method of inhibiting farnesyl-protein transferase and geranylgeranyl-protein transferase type I by administering a compound that is a dual inhibitor of both of those prenyl-protein transferases. The instant invention also provides for a method of identifying such a compound, the method comprising an assay whose readout is a consequence of the biological activity or inhibition of that activity of the Ras protein, thus providing convenient identifica-

tion of compounds that inhibit cellular processing of the H-Ras and K4B-Ras proteins.

BRIEF DESCRIPTION OF THE FIGURES FIGURE 1: Schematic Diagram of the SEAP Assay: A schematic drawing of the MAPK signal transduction pathway and activation of the transcription factor Elkl leading to activation of the fos promoter/reporter construct and expression of secreted alkaline phosphatase (SEAP).

DETAILED DESCRIPTION OF THE INVENTION The present invention relates to a method of inhibiting prenyl-protein transferases which comprises administering to a mammal in need thereof a pharmaceutically effective amount of a compound which has certain characteristics that are indicative of in vivo efficacy as an inhibitor of the growth of cancer cells. In particular the compound is characterized by: a) an inhibitory activity (IC50) of less than 12 RM against K4B-Ras dependent activation of MAP kinases in cells.

Preferably, the compound utilized in the instant method is characterized by: a) an inhibitory activity (IC50) of less than 5 RM against K4B-Ras dependent activation of MAP kinases in cells.

The compound may be further characterized by one or more of the following: b) an inhibitory activity (IC50) against K4B-Ras dependent activation of MAP kinases in cells greater than 5 fold lower than the inhibitory activity (IC50) against Myr-Ras dependent activation of MAP kinases in cells; c) an inhibitory activity (ICs0) against H-ras-CVLL dependent activation of MAP kinases in cells greater than 5 fold lower

than the inhibitory activity (IC50) against Myr-Ras dependent activation of MAP kinases in cells; d) an inhibitory activity (IC50) against H-Ras dependent activation of MAP kinases in cells greater than 2 fold lower but less than 20,000 fold lower than the inhibitory activity (IC50) against H-ras-CVLL (SEQ.ID.NO.: 1) dependent activation of MAP kinases in cells; d) an inhibitory activity (IC50) against H-ras-CVLL dependent activation of MAP kinases in cells greater than 5 fold lower than the inhibitory activity (IC50) against expression of the SEAP protein in cells transfected with the pCMV-SEAP plasmid that constitutively expresses the SEAP protein; and e) an inhibitory activity (IC50) against K4B-Ras dependent activation of MAP kinases in cells greater than 5 fold lower than the inhibitory activity (IC50) against expression of the SEAP protein in cells transfected with the pCMV-SEAP plasmid that constitutively expresses the SEAP protein.

Preferably, the compound is further characterized by: c) inhibitory activity (IC50) of < 10 nM against H-Ras dependent activation of MAP kinases in cells.

Preferably, the prenyl-protein transferases that are being inhibited by the instant method are both farnesyl-protein transferase and geranylgeranyl-protein transferase type I.

Preferably the compound that is being administered is a dual inhibitor of farnesyl-protein transferase and geranylgeranyl-protein transferase type I.

It has been surprisingly found that such a potent dual inhibitor is particularly useful as an in vivo inhibitor of the growth of cancer cells, particularly those cancers characterized by a mutated K4B-Ras protein, at concentrations of inhibitor that do not cause mechanism based toxicity. Mechanism-based toxicity of farnesyl-

protein transferase inhibitors can be anticipated in rapidly proliferating tissues, for example, the bone marrow.

The present invention further relates to a method of identifying a prenyl-protein transferase inhibitor which is efficacious in vivo as an inhibitor of cancer cell growth. The instant method comprises a novel in vitro assay (described in detail below) whose readout is a consequence of the biological activity of the Ras protein (or its inhibition) instead of a determination of the physical state of the Ras protein (whether or not the protein has been processed).

This assay differs from previously disclosed assays that measure the extent of inhibition of Ras processing in cells because the determination of the extent of processing may be performed with a high through-put luminometer and does not depend on time-intensive use of polyacrylamide gel electrophoresis. Compounds that inhibit the processing of the Ras protein but do not inhibit its biological activity may be improperly identified by previously disclosed assays of Ras processing in cells which merely measure the extent to which the protein was processed.

The instant assay that is useful in the identification of the prenyl-protein transferase inhibitors of the instant invention, comprises the steps of: a) co-transfecting cells with an expression plasmid for a ras gene and an expression plasmid for a reporter construct that encodes the product of a reporter gene; b) incubating the cells in the presence of test compound; and c) analyzing an aliquot of the assay medium or a lysate of the cells for the presence of the product of the reporter gene.

Preferably, the product of the reporter gene is secreted alkaline phosphatase. When secreted alkaline phosphatase is used as a reporter the assay is termed the SEAP assay. In the methods described herein below, use of the SEAP assay is described.

However, one of ordinary skill in the art would readily appreciate that other reporter systems can be utilized, including but not limited

to: luciferase, -galactosidase, chloramphenicol acetyl transferase and -glucuronidase.

Preferably, the expression of the reporter gene is controlled by a transcription factor which is activated by MAP kinases. The term MAP kinases includes but is not limited to ERK-1 (Extracellular-Regulated protein Kinase), ERK-2, SAPK/JNK (stress activated protein kinase/C-jun N-terminal kinase) and p38.

Plasmids that incorporate the SEAP reporter construct include but are not limited to those described by D. Defeo-Jones et al. (Mol. Cell. Biol. 11:2307-2310 (1991)), R. E. Jones et al.

(Oncogene, 6:745-751(1991)) and J. Berger et al. (J. Biol. Chem., 263:10016-10021 (1988)). The SEAP reporter plasmids pDSE100 and pDSE101 described in Example 15 hereinbelow is also useful in the instant assay. The SEAP reporter plasmid pCMV is also useful in the methods of the instant invention as a control plasmid to identify non-mechanism based toxicity of a test compound.

The term ras gene includes the H-ras , N-ras , K4B-ras, Myr-ras and H-ras -CVLL genes.

Expression plasmids for a ras gene of the instant invention include but are not limited to pZIP-rasH, pZIP-rasN, pZIP-rasK4B, pSMS600, pSMS601, pSMS620, pSMS62 1, pSMS622, pSMS630, pSMS640, pSMS650, pBW1423 (B.W. Williamsen et al.

Mol. Cell. Biol., 11:6026-6033 (1991)), pRcCMV-H-ras-V12 and pRcCMV-H-ras-vl2,L1 89 (G.L. James et al. J. Biol.

Chem. 269 : 27705-27714 (1994)).

Alternate expression vectors that can be utilized to create expression plasmids for a ras gene include, but are not limited to, pCI, pSI, pSport (Promega), pBK-CMV, pBK-RSV (Stratagene), pEUK-C1 (Clonetech), pCMV-L1C (Pharmingen) and pcDNA1.1/Amp (Invitrogen).

The assay medium used in the instant assay may be selected from medium useful for maintaining transiently transfected cells. Preferably, the medium will lack phenol red and will be low in serum. Preferably the assay medium comprises phenol red free

DMEM, 2% mammalian serum, Pen/Strep, glutamine and nonessential amino acids (NEAA).

It is contemplated that virtually any of the commonly employed transfectable host cells that arrest in low serum growth media can be used in connection with the instant assay. Examples include cell lines typically employed for eukaryotic expression such as C33a (ATCC: HTB-31), Ratl and 3T3 cell lines. A preferred line for use in the instant assay has been found to be the human cell line C33a.

Preferably the cancer cells are isolated after being co-transfected with the expression plasmids.

In a first embodiment of the instant invention, the method of identifying a prenyl-protein transferase inhibitor comprises the steps of: a) evaluating the test compound in the instant assay wherein the ras gene is K-ras; b) evaluating the test compound in the instant assay wherein the ras gene is Myr-ras ; and c) comparing the activity of the test compound against Myr-Ras dependent activation of MAP kinases in the instant assay with the activity of the test compound against K- Ras dependent activation of MAP kinases in the instant assay.

Preferably, the K-ras gene utilized in the instant method is the K4B-ras gene, although it is envisaged that the K4A-ras gene could also be utilized.

Preferably, in the first embodiment of the method of the instant invention comprises one or both of the additional steps of: d) evaluating the test compound in the instant assay wherein the ras gene is H-ras ; and e) evaluating the test compound in the instant assay wherein the ras gene is H-ras-CVLL.

In a second embodiment of the instant invention, the method of identifying a prenyl-protein transferase inhibitor comprises the steps of:

a) evaluating the test compound in the instant assay wherein the ras gene is H-ras; b) evaluating the test compound in the instant assay wherein the ras gene is H-ras-CVLL; c) evaluating the test compound in the instant assay wherein the ras gene is Myr-ras ; and d) comparing the activity of the test compound against Myr- Ras dependent activation of MAP kinases in the instant assay with the activity of the test compound against H- Ras dependent activation of MAP kinases in the instant assay and H-Ras-CVLL dependent activation of MAP kinases in the instant assay.

In a third embodiment of the instant invention, the method of identifying a prenyl-protein transferase inhibitor comprises the steps of: a) evaluating the test compound in the instant assay wherein the Ras gene is N-ras; b) evaluating the test compound in the instant assay wherein the Ras gene is Myr-ras ; and c) comparing the activity of the test compound against Myr- Ras dependent activation of MAP kinases in the instant assay with the activity of the test compound against N- Ras dependent activation of MAP kinases in the instant assay.

In a forth embodiment of the instant invention, the method of identifying a prenyl-protein transferase inhibitor comprises the steps of: a) evaluating the test compound in the instant assay wherein the ras gene is H-ras; b) evaluating the test compound in the instant assay wherein the ras gene is H-ras-CVLL; c) evaluating the test compound in the instant assay wherein the cells have been transfected with a pCMV-SEAP plasmid in the absence of transfection with a rasgene; and d) comparing the activity of the test compound against H-Ras- CVLL dependent activation of MAP kinases in the instant assay

with the activity of the test compound against H- Ras dependent activation of MAP kinases in the instant assay and the activity of the test compound against SEAP expression in part c) of this method.

Preferably the assay utilized in the above methods of identifying inhibitors is the SEAP assay.

Preferably, the pCMV-SEAP plasmid used in the instant assay is the pCMV-SEAP-A plasmid.

The inhibitor compounds identified by the instant method are useful in the inhibition of prenyl-protein transferase and the treatment of cancer and other proliferative disorders in mammals in need thereof. The above methods of identifying a single compound that is a prenyl-protein transferase inhibitor or a dual inhibitor of farnesyl-protein transferase and geranylgeranyl -protein transferase-type I may also be used to identify optimal ratios of the active components in a combination of a selective farnesyl-protein transferase inhibitor and a selective geranylgeranyl-protein transferase-type I inhibitor.

Preferably, the compounds of the invention have inhibitory concentrations (IC50) of < 100 nM against H-Ras dependent activation of MAP kinases in cells in the SEAP assay.

More preferably, the compounds of the invention have inhibitory concentrations (IC50) of < 10 nM against H-Ras dependent activation of MAP kinases in cells in the SEAP assay. Preferably, the ratio of inhibitory activity (IC50) against K-Ras4B dependent activation to inhibitory activity against H-Ras dependent activation is <2000.

Preferably, the compounds of the invention have inhibitory concentrations (IC50) of < 10 uM against H-Ras-CVLL dependent activation of MAP kinases in cells in the SEAP assay.

More preferably, the compounds of the invention have inhibitory concentrations (IC50) of < 1 uM against H-Ras-CVLL dependent activation of MAP kinases in cells in the SEAP assay. Preferably,

the ratio of inhibitory activity (IC50) against H-Ras-CVLL dependent activation to inhibitory activity against H-Ras dependent activation is from about 2 to about 20,000. More preferably, the ratio of inhibitory activity (IC50) against H-Ras-CVLL dependent activation to inhibitory activity (IC50) against H-Ras dependent activation is from about 20 to about 2,000.

Preferably, the compounds of the invention have inhibitory concentrations (ICs0) of < 5 ptM against cellular N-Ras dependent activation of MAP kinases in the SEAP assay.

More preferably, the compounds of the invention have inhibitory concentrations (IC50) of < 1 RM against cellular N-Ras dependent activation of MAP kinases in the SEAP assay.

It is preferred that the compounds of the invention selectively inhibit processing of a Ras protein, and therefore inhibit the growth of cells transformed by a ras oncogene. In the instant method of identifying a prenyl-protein transferase inhibitor, the step of evaluating the activity of the test compound in the instant assay wherein the SEAP plasmid is selected from the mutated ras gene designated Myr-ras assesses whether the test compound inhibits signal transduction independent of inhibiting Ras prenylation, since the mutated gene Myr-ras enables the protein to bypass the requirement of prenylation for cellular activity (J. E. Buss et al. Science, 243:1600-1603 (1989)). If the IC50 of the test compound against cellular Myr-Ras dependent activation of MAP kinases in the instant assay is close in value to the IC50 of the test compound against K4B-Ras dependent activation of MAP kinases in the same assay it is difficult to determine the true effect of the test compound in inhibiting Ras prenylation. Such inhibition may represent non- specific cytotoxicity rather than selective inhibition of Ras prenylation.

Alternatively, non-specific cytotoxicity of a test compound may be evaluated by incubating a cell that has been

transfected with the pCMV-SEAP plasmid and analyzing the assay medium for the presence of the SEAP protein.

In an embodiment of the instant invention, it is therefore preferable that the ratio of the activity (as an IC50) of the test com- pound against cellular Myr-Ras dependent activation of MAP kinases in the SEAP assay to the activity (as an IC50) of the test compound against K4B-Ras dependent activation of MAP kinases in the SEAP assay is greater than 1. Most preferably, the ratio of inhibitory activity (IC50) against Myr- Ras dependent activation of MAP kinases (as measured in the SEAP assay) to the inhibitory activity (IC50) against K4B-Ras dependent activation of MAP kinases is >5.

It is understood that the phrase "an inhibitory activity (IC50) against K4B-Ras dependent activation of MAP kinases in cells greater than 5 fold lower than the inhibitory activity (IC50) against Myr-Ras dependent activation of MAP kinases in cells" (and similar phrases directed to other comparisons) has the same meaning as the phrase "the ratio of inhibitory activity (IC50) against Myr- Ras dependent activation of MAP kinases (as measured in the SEAP assay) to the inhibitory activity (IC50) against K4B-Ras dependent activation of MAP kinases is >5" (and other like phrases).

It is also preferable that the ratio of the activity (as an IC50) of the test compound against cellular Myr- Ras dependent activation of MAP kinases in the SEAP assay to the activity (as an IC50) of the test compound against H-Ras-CVLL dependent activation of MAP kinases in the SEAP assay is greater than 1.

Most preferably, the ratio of inhibitory activity against Myr- Ras dependent activation of MAP kinases (as measured in the SEAP assay) to the inhibitory activity against H-Ras-CVLL dependent activation of MAP kinases is >5.

It is further preferable that the ratio of the activity (as an IC50) of the test compound against cellular Myr- Ras dependent activation of MAP kinases in the SEAP assay to the activity (as an IC50) of the test compound against N-Ras dependent activation of MAP kinases in the SEAP assay is greater than 5. Most preferably,

the ratio of inhibitory activity against Myr-Ras dependent activation of MAP kinases (as measured in the SEAP assay) to the inhibitory activity against N-Ras dependent activation of MAP kinases is >20.

In another embodiment of the instant invention, it is preferable that the ratio of the activity (as an IC50) of the test compound against expression of the SEAP protein in cells transfected with the pCMV-SEAP plasmid to the activity (as an IC50) of the test compound against H-Ras-CVLL dependent activation of MAP kinases in the SEAP assay is greater than 1. Most preferably, the ratio of inhibitory activity against expression of the SEAP protein in cells transfected with the pCMV-SEAP plasmid to the activity (as an IC50) of the test compound against H-Ras-CVLL dependent activation of MAP kinases in the SEAP assay is >5.

When a particular Ras protein is referred to herein by a term such as "K4B-Ras", "N-Ras", "H-Ras" and the like, such a term represents both the protein arising from expression of the corresponding wild type ras gene and various proteins arising from expression of ras genes containing various point mutations. When a particular ras gene is referred to herein by a term such as "K4B- ras", "N-ras", "H-ras" and the like, such a term represents both the wild type ras gene and ras genes containing various point mutations.

The term prenyl-protein transferase inhibiting compound refers to compounds which antagonize, inhibit or counteract the activity of the genes coding farnesyl-protein transferase and geranylgeranyl-protein transferase type I or the proteins produced in response thereto.

The term selective as used herein refers to the inhibitory activity of the particular compound against one biological activity (such as inhibition of prenyl-protein transferases) when compared to the inhibitory activity of the compound against another biological activity. It is understood that the greater the selectivity of a prenyl- protein transferase inhibitor, the more preferred such a compound is for the methods of treatment described.

For example, when discussing the combination of a selective inhibitor of geranylgeranyl -protein transferase-type I and a selective inibitor of farnesyl-protein transferase, a compound is considered a selective inhibitor of geranylgeranyl protein transferase-type I, when its in vitro activity, as assessed by the assay described in Example 11, is at least 10 times greater that the in vitro activity of the same compound against farnesyl-protein transferase in the assay described in Example 10. A compound is considered a selective inhibitor of farnesyl-protein transferase, for example, when its in vitro farnesyl-protein transferase inhibitory activity, as assessed by the assay described in Example 10, is at least 10 times greater that the in vitro activity of the same compound against geranylgeranyl- protein transferase-type I in the assay described in Example 11.

Preferably, a selective compound exhibits at least 20 times greater activity against one of the enzymatic activities when comparing geranylgeranyl-protein transferase-type I inhibition and farnesyl- protein transferase inhibition. More preferably the selectivity is at least 100 times or more. It is understood that the greater the selectivity of a geranylgeranyl-protein transferase-type I inhibitor or farnesyl-protein transferase inhibitor, the more preferred such a compound is in the such a combination.

The preferred therapeutic effect provided by the instant composition is the treatment of cancer and specifically the inhibition of cancerous tumor growth and/or the regression of cancerous tumors. Cancers which are treatable in accordance with the invention described herein include cancers of the brain, breast, colon, genitourinary tract, prostate, skin, lymphatic system, pancreas, rectum, stomach, larynx, liver and lung.

More particularly, such cancers include histiocytic lymphoma, lung adenocarcinoma, pancreatic carcinoma, colo-rectal carcinoma, small cell lung cancers, bladder cancers, head and neck cancers, acute and chronic leukemias, melanomas, and neurological tumors.

The composition of this invention is also useful for inhibiting other proliferative diseases, both benign and malignant,

wherein Ras proteins are aberrantly activated as a result of oncogenic mutation in other genes (i.e., the ras gene itself is not activated by mutation to an oncogenic form) with said inhibition being accomplished by the administration of an effective amount of the instant composition to a mammal in need of such treatment.

For example, the composition is useful in the treatment of neurofibromatosis, which is a benign proliferative disorder.

The composition of the instant invention is also useful in the prevention of restenosis after percutaneous transluminal coronary angioplasty by inhibiting neointimal formation (C. Indolfi et al.

Nature medicine, 1:541-545(1995).

The instant composition may also be useful in the treatment and prevention of polycystic kidney disease (D.L.

Schaffner et al. American Journal of Pathology, 142:1051-1060 (1993) and B. Cowley, Jr. et al. FASEB Journal, 2:A3160 (1988)).

The instant compounds may also inhibit tumor angio- genesis, thereby affecting the growth of tumors (J. Rak et al. Cancer Research, 55:4575-4580 (1995)). Such anti-angiogenesis properties of the instant compounds may also be useful in the treatment of certain forms of vision deficit related to retinal vascularization.

The instant compounds may also be useful in the treatment of certain viral infections, in particular in the treatment of hepatitis delta and related viruses (J.S. Glenn et al. Science, 256:1331-1333 (1992).

The instant compounds may also be useful as inhibitors of proliferation of vascular smooth muscle cells and therefore useful in the prevention and therapy of arteriosclerosis and diabetic vascular pathologies.

The compounds of this invention may be administered to mammals, preferably humans, either alone or, preferably, in combination with pharmaceutically acceptable carriers, excipients or diluents, in a pharmaceutical composition, according to standard pharmaceutical practice. The compounds can be administered orally or parenterally, including the intravenous, intramuscular,

intraperitoneal, subcutaneous, rectal and topical routes of administration.

The pharmaceutical compositions containing the active ingredient may be in a form suitable for oral use, for example, as tablets, troches, lozenges, aqueous or oily suspensions, dispersible powders or granules, emulsions, hard or soft capsules, or syrups or elixirs. Compositions intended for oral use may be prepared according to any method known to the art for the manufacture of pharmaceutical compositions and such compositions may contain one or more agents selected from the group consisting of sweetening agents, flavoring agents, coloring agents and preserving agents in order to provide pharmaceutically elegant and palatable preparations.

Tablets contain the active ingredient in admixture with non-toxic pharmaceutically acceptable excipients which are suitable for the manufacture of tablets. These excipients may be for example, inert diluents, such as calcium carbonate, sodium carbonate, lactose, calcium phosphate or sodium phosphate; granulating and disintegrating agents, for example, microcrystalline cellulose, sodium crosscarmellose, corn starch, or alginic acid; binding agents, for example starch, gelatin, polyvinyl-pyrrolidone or acacia, and lubricating agents, for example, magnesium stearate, stearic acid or talc. The tablets may be uncoated or they may be coated by known techniques to mask the unpleasant taste of the drug or delay disintegration and absorption in the gastrointestinal tract and thereby provide a sustained action over a longer period. For example, a water soluble taste masking material such as hydroxypropylmethyl- cellulose or hydroxypropylcellulose, or a time delay material such as ethyl cellulose, cellulose acetate buryrate may be employed.

Formulations for oral use may also be presented as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent, for example, calcium carbonate, calcium phosphate or kaolin, or as soft gelatin capsules wherein the active ingredient is mixed with water soluble carrier such as polyethyleneglycol or an oil medium, for example peanut oil, liquid paraffin, or olive oil.

Aqueous suspensions contain the active material in admixture with excipients suitable for the manufacture of aqueous suspensions. Such excipients are suspending agents, for example sodium carboxymethylcellulose, methylcellulose, hydroxypropyl- methyl-cellulose, sodium alginate, polyvinyl-pyrrolidone, gum tragacanth and gum acacia; dispersing or wetting agents may be a naturally-occurring phosphatide, for example lecithin, or condensation products of an alkylene oxide with fatty acids, for example polyoxyethylene stearate, or condensation products of ethylene oxide with long chain aliphatic alcohols, for example heptadecaethylene-oxycetanol, or condensation products of ethylene oxide with partial esters derived from fatty acids and a hexitol such as polyoxyethylene sorbitol monooleate, or condensation products of ethylene oxide with partial esters derived from fatty acids and hexitol anhydrides, for example polyethylene sorbitan monooleate. The aqueous suspensions may also contain one or more preservatives, for example ethyl, or n-propyl p-hydroxybenzoate, one or more coloring agents, one or more flavoring agents, and one or more sweetening agents, such as sucrose, saccharin or aspartame.

Oily suspensions may be formulated by suspending the active ingredient in a vegetable oil, for example arachis oil, olive oil, sesame oil or coconut oil, or in mineral oil such as liquid paraffin.

The oily suspensions may contain a thickening agent, for example beeswax, hard paraffin or cetyl alcohol. Sweetening agents such as those set forth above, and flavoring agents may be added to provide a palatable oral preparation. These compositions may be preserved by the addition of an anti-oxidant such as butylated hydroxyanisol or alpha-tocopherol.

Dispersible powders and granules suitable for preparation of an aqueous suspension by the addition of water provide the active ingredient in admixture with a dispersing or wetting agent, suspending agent and one or more preservatives.

Suitable dispersing or wetting agents and suspending agents are exemplified by those already mentioned above. Additional

excipients, for example sweetening, flavoring and coloring agents, may also be present. These compositions may be preserved by the addition of an anti-oxidant such as ascorbic acid.

The pharmaceutical compositions of the invention may also be in the form of an oil-in-water emulsions. The oily phase may be a vegetable oil, for example olive oil or arachis oil, or a mineral oil, for example liquid paraffin or mixtures of these. Suitable emulsifying agents may be naturally-occurring phosphatides, for example soy bean lecithin, and esters or partial esters derived from fatty acids and hexitol anhydrides, for example sorbitan monooleate, and condensation products of the said partial esters with ethylene oxide, for example polyoxyethylene sorbitan monooleate. The emulsions may also contain sweetening, flavouring agents, preservatives and antioxidants.

Syrups and elixirs may be formulated with sweetening agents, for example glycerol, propylene glycol, sorbitol or sucrose.

Such formulations may also contain a demulcent, a preservative, flavoring and coloring agents and antioxidant.

The pharmaceutical compositions may be in the form of a sterile injectable aqueous solutions. Among the acceptable vehicles and solvents that may be employed are water, Ringer's solution and isotonic sodium chloride solution.

The sterile injectable preparation may also be a sterile injectable oil-in-water microemulsion where the active ingredient is dissolved in the oily phase. For example, the active ingredient may be first dissolved in a mixture of soybean oil and lecithin. The oil solution then introduced into a water and glycerol mixture and processed to form a microemulation.

The injectable solutions or microemulsions may be introduced into a patient's blood-stream by local bolus injection.

Alternatively, it may be advantageous to administer the solution or microemulsion in such a way as to maintain a constant circulating concentration of the instant compound. In order to maintain such

a constant concentration, a continuous intravenous delivery device may be utilized. An example of such a device is the Deltec CADD- PLUS TM model 5400 intravenous pump.

The pharmaceutical compositions may be in the form of a sterile injectable aqueous or oleagenous suspension for intramuscular and subcutaneous administration. This suspension may be formulated according to the known art using those suitable dispersing or wetting agents and suspending agents which have been mentioned above. The sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally- acceptable diluent or solvent, for example as a solution in 1,3-butane diol. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose any bland fixed oil may be employed including synthetic mono- or diglycerides. In addition, fatty acids such as oleic acid find use in the preparation of inj ectables.

Compounds of Formula A may also be administered in the form of a suppositories for rectal administration of the drug.

These compositions can be prepared by mixing the drug with a suitable non-irritating excipient which is solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum to release the drug. Such materials include cocoa butter, glycerinated gelatin, hydrogenated vegetable oils, mixtures of polyethylene glycols of various molecular weights and fatty acid esters of polyethylene glycol.

For topical use, creams, ointments, jellies, solutions or suspensions, etc., containing the compound of Formula A are employed. (For purposes of this application, topical application shall include mouth washes and gargles.) The compounds for the present invention can be administered in intranasal form via topical use of suitable intranasal vehicles and delivery devices, or via transdermal routes, using those forms of transdermal skin patches well known to those of ordinary skill in the art. To be administered in the form of a transdermal

delivery system, the dosage administration will, of course, be continuous rather than intermittent throughout the dosage regimen.

As used herein, the term "composition" is intended to encompass a product comprising the specified ingredients in the specific amounts, as well as any product which results, directly or indirectly, from combination of the specific ingredients in the specified amounts.

The compounds identified by the instant method may also be co-administered with other well known therapeutic agents that are selected for their particular usefulness against the condition that is being treated. For example, the instant compounds may be useful in combination with known anti-cancer and cytotoxic agents.

Similarly, the instant compounds may be useful in combination with agents that are effective in the treatment and prevention of neuro- fibromatosis, restinosis, polycystic kidney disease, infections of hepatitis delta and related viruses and fungal infections. The instant compounds may also be useful in combination with other inhibitors of parts of the signaling pathway that links cell surface growth factor receptors to nuclear signals initiating cellular proliferation.

The instant compounds may be utilized in combination with farnesyl pyrophosphate competitive inhibitors of the activity of farnesyl-protein transferase or in combination with a compound which has Raf antagonist activity. The instant compounds may also be co-administered with compounds that are selective inhibitors of geranylgeranyl protein transferase or selective inhibitors of farnesyl- protein transferase.

The compounds of the instant invention may also be co-administered with other well known cancer therapeutic agents that are selected for their particular usefulness against the condition that is being treated. Included in such combinations of therapeutic agents are combinations of the instant prenyl-protein transferase inhibitors and an antineoplastic agent. It is also understood that the instant combination of antineoplastic agent and inhibitor of prenyl-protein transferase may be used in conjunction with other methods of

treating cancer and/or tumors, including radiation therapy and surgery.

If formulated as a fixed dose, such combination products employ the combinations of this invention within the dosage range described below and the other pharmaceutically active agent(s) within its approved dosage range. Combinations of the instant invention may alternatively be used sequentially with known pharmaceutically acceptable agent(s) when a multiple combination formulation is inappropriate.

Radiation therapy, including x-rays or gamma rays which are delivered from either an externally applied beam or by implantation of tiny radioactive sources, may also be used in combination with an inhibitor of prenyl-protein transferase alone to treat cancer.

Additionally, compounds of the instant invention may also be useful as radiation sensitizers, as described in WO 97/38697, published on October 23, 1997, and herein incorporated by reference.

The instant compounds may also be useful in combination with other inhibitors of parts of the signaling pathway that links cell surface growth factor receptors to nuclear signals initiating cellular proliferation. Thus, the instant compounds may be utilized in combination with farnesyl pyrophosphate competitive inhibitors of the activity of farnesyl-protein transferase or in combination with a compound which has Raf antagonist activity.

The instant compounds may also be useful in combination with an integrin antagonist for the treatment of cancer, as described in U.S. Ser. No. 09/055,487, filed April 6, 1998, which is incorporated herein by reference.

As used herein the term an integrin antagonist refers to compounds which selectively antagonize, inhibit or counteract binding of a physiological ligand to an integrin(s) that is involved

in the regulation of angiogenisis, or in the growth and invasiveness of tumor cells. In particular, the term refers to compounds which selectively antagonize, inhibit or counteract binding of a physio- logical ligand to the avp3 integrin, which selectively antagonize, inhibit or counteract binding of a physiological ligand to the ccv5 integrin, which antagonize, inhibit or counteract binding of a physiological ligand to both the αv#3 integrin and the ocv 5 integrin, or which antagonize, inhibit or counteract the activity of the particular integrin(s) expressed on capillary endothelial cells. The term also refers to antagonists of the oev 6, αv#8, aipi, oc2 l, α5#1, α#1 and α6#4 integrins. The term also refers to antagonists of any combination of αv#3, αv#5, αv#6, αv#8, α1#1, α2#1, a5pl, α6#1 and α6#4 integrins. The instant compounds may also be useful with other agents that inhibit angiogenisis and thereby inhibit the growth and invasiveness of tumor cells, including, but not limited to angiostatin and endostatin.

When a composition according to this invention is administered into a human subject, the daily dosage will normally be determined by the prescribing physician with the dosage generally varying according to the age, weight, and response of the individual patient, as well as the severity of the patient's symptoms.

In one exemplary application, a suitable amount of a prenyl-protein transferase inhibitor are administered to a mammal undergoing treatment for cancer. Administration occurs in an amount of each type of inhibitor of between about 0.1 mg/kg of body weight to about 60 mg/kg of body weight per day, preferably of between 0.5 mg/kg of body weight to about 40 mg/kg of body weight per day. A particular therapeutic dosage that comprises the instant composition includes from about 0.01mg to about 1000mg of a prenyl-protein transferase inhibitor. Preferably, the dosage comprises from about 1mg to about 1000mg of a prenyl-protein transferase inhibitor.

Examples of an antineoplastic agent include, in

general, microtubule-stabilising agents (such as paclitaxel (also known as Taxol), docetaxel (also known as Taxotere), or their derivatives); alkylating agents, anti-metabolites; epidophyllotoxin; an antineoplastic enzyme; a topoisomerase inhibitor; procarbazine; mitoxantrone; platinum coordination complexes; biological response modifiers and growth inhibitors; hormonal/anti-hormonal therapeutic agents and haematopoietic growth factors.

Example classes of antineoplastic agents include, for example, the anthracycline family of drugs, the vinca drugs, the mitomycins, the bleomycins, the cytotoxic nucleosides, the taxanes, the epothilones, discodermolide, the pteridine family of drugs, diynenes and the podophyllotoxins. Particularly useful members of those classes include, for example, doxorubicin, carminomycin, daunorubicin, aminopterin, methotrexate, methopterin, dichloro- methotrexate, mitomycin C, porfiromycin, 5-fluorouracil, 6-mercaptopurine, gemcitabine, cytosine arabinoside, podophyllotoxin or podo-phyllotoxin derivatives such as etoposide, etoposide phosphate or teniposide, melphalan, vinblastine, vincristine, leurosidine, vindesine, leurosine, paclitaxel and the like.

Other useful antineoplastic agents include estramustine, cisplatin, carboplatin, cyclophosphamide, bleomycin, gemcitibine, ifosamide, melphalan, hexamethyl melamine, thiotepa, cytarabin, idatrexate, trimetrexate, dacarbazine, L-asparaginase, camptothecin, CPT- 11, topotecan, ara-C, bicalutamide, flutamide, leuprolide, pyridobenzoindole derivatives, interferons and interleukins.

Compounds of the instant invention that are identified by the properties described hereinabove include: (a) a compound represented by formula I:

wherein: Rla is selected from: hydrogen or C1-C6 alkyl; Rlb is independently selected from: a) hydrogen, b) aryl, heterocycle, cycloalkyl, R10O-, -N(R10)2 or C2-C6 alkenyl, c) C1-C6 alkyl unsubstituted or substituted by aryl, heterocycle, cycloalkyl, alkenyl, R10O-, or -N(R10)2; R3 and R4 selected from H and CH3 ; R2 is selected from H; unsubstituted or substituted aryl, unsubstituted or substituted heteroaryl, or C1-5 alkyl, unbranched or branched, unsubstituted or substituted with one or more of: 1) aryl, 2) heterocycle, 3) OR6, 4) SR6a, SO2R6a, or and R2 and R3 are optionally attached to the same carbon atom; R6 and R7 are independently selected from: H; C1-4 alkyl, C3-6 cycloalkyl, aryl, heterocycle, unsubstituted or substituted with: a) C 1-4 alkoxy, b) halogen, c) perfluoro-C1-4 alkyl, or

d) aryl or heterocycle; R6a is selected from: C1-4 alkyl or C3-6 cycloalkyl, unsubstituted or substituted with: a) C 1-4 alkoxy, b) halogen, or c) aryl or heterocycle; R8 is independently selected from: a) hydrogen, b) C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, Cl-C6 perfluoroalkyl, F, C1, R10O-, R10C(O)NR10-, CN, NO2, (R10)2N-C(NR10)-, R10C(O)-, -N(R10)2, or R11OC(O)NR10-, and c) C1-C6 alkyl substituted by C1-C6 perfluoroalkyl, R10O-, R10C(O)NR10-, (R10)2N-C(NR10)-, R10C(O)-, -N(R10)2, or R11OC(O)NR10-; R9a is hydrogen or methyl; R10 is independently selected from hydrogen, C1-C6 alkyl, C1-C6 perfluoroalkyl, 2,2,2,-trifluoroethyl, benzyl and aryl; Rll is independently selected from C1-C6 alkyl and aryl; A1 and A2 are independently selected from: a bond, -CH=CH-, -CC-, -C(O)-, -C(O)NR10-, O, -N(R10)-, or S(O)m; V is selected from: a) hydrogen,

b) heterocycle selected from pyrrolidinyl, imidazolyl, pyridinyl, thiazolyl, pyridonyl, 2-oxopiperidinyl, indolyl, quinolinyl, isoquinolinyl, and thienyl, c) aryl, d) Cl -C20 alkyl wherein from 0 to 4 carbon atoms are replaced with a heteroatom selected from 0, S, and N, and e) C2-C20 alkenyl, and provided that V is not hydrogen if A1 is S(O)m and V is not hydrogen if A1 is a bond, n is 0 and A2 is S(O)m; X is -CH2- or Z is selected from: 1) a unsubstituted or substituted group selected from aryl, heteroaryl, arylmethyl, heteroarylmethyl, arylsulfonyl, heteroarylsulfonyl, wherein the substituted group is substituted with one or more of the following: a) C 1-4 alkyl, unsubstituted or substituted with: C1-4 alkoxy, NR6R7, C3-6 cycloalkyl, unsubstituted or substituted aryl, heterocycle, HO, -S(O)mR6a, or -C(O)NR6R7, b) aryl or heterocycle, c) halogen, d) OR6, e) NR6R7, f) CN, g) NO2, h) CF3; i) S(O)mR6a, j) -C(O)NR6R7, or k) C3-C6 cycloalkyl; or 2) unsubstituted C1-C6 alkyl, substituted C1-C6 alkyl, unsubstituted C3-C6 cycloalkyl or substituted C3-C6 cycloalkyl, wherein the substituted C1-C6 alkyl and substituted

C3-C6 cycloalkyl is substituted with one or two of the following: a) C 1-4 alkoxy, b) NR6R7, c) C3-6 cycloalkyl, d) -NR6C(O)R7, e) HO, f) -S(O)mR6a, g) halogen, or h) perfluoroalkyl; m is 0, 1 or 2; n is 0, 1, 2, 3 or 4; p is 0, 1, 2, 3 or 4; and r is 0 to 5, provided that r is 0 when V is hydrogen; provided that the substituent (R8)r- V -A1(CR1a2)nA2(CR1a2)n - is not H; b) the inhibitors of farnesyl-protein transferase are illustrated by the formula II: wherein: Rla is selected from: hydrogen or C1-C6 alkyl; Rlb is independently selected from: a) hydrogen,

b) aryl, heterocycle, cycloalkyl, R10O-, -N(R10)2 or C2-C6 alkenyl, c) C1-C6 alkyl unsubstituted or substituted by unsubstituted or substituted aryl, heterocycle, cycloalkyl, alkenyl, R10O-, or -N(R10)2; R1c is selected from: a) hydrogen, b) unsubstituted or substituted C1-C6 alkyl wherein the substituent on the substituted C1-C6 alkyl is selected from unsubstituted or substituted aryl, heterocyclic, C3-C 10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, R10O-, R11S(O)m-, R10C(O)NR10-, (R10)2N-C(O)-, CN, (R10)2N-C(NR10)-, R10C(O)-, R10OC(O)-, N3, -N(R10)2, and R1 1OC(O)-NR10-, and c) unsubstituted or substituted aryl; R3 and R4 independently selected from H and CH3; R2 is selected from H; OR10; or C1-5 alkyl, unbranched or branched, unsubstituted or substituted with one or more of: 1) aryl, 2) heterocycle, 3) OR6, 4) SR6a, SO2R6a, or and R2, R3 and R4 are optionally attached to the same carbon atom;

R6 and R7 are independently selected from: H; C1-4 alkyl, C3-6 cycloalkyl, aryl, heterocycle, unsubstituted or substituted with: a) C 1-4 alkoxy, b) halogen, or c) aryl or heterocycle; R6a is selected from: C1-4 alkyl or C3-6 cycloalkyl, unsubstituted or substituted with: a) C 1-4 alkoxy, b) halogen, or c) aryl or heterocycle; R8 is independently selected from: a) hydrogen, b) C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, Cl-C6 perfluoroalkyl, F, C1, R10O-, R10C(O)NR10-, CN, NO2, (R1 0)2N-C(NR10)-, R1 0C(O)-, -N(R10)2, or R11OC(O)NR10-, and c) C1-C6 alkyl substituted by C1-C6 perfluoroalkyl, R10O-, R10C(O)NR10-, (R10)2N-C(NR10)-, R10C(O)-, -N(R10)2, or R11OC(O)NR10-; R9a is hydrogen or methyl; R10 is independently selected from hydrogen, C1-C6 alkyl, C1-C6 perfluoroalkyl, 2,2,2-trifluoroethyl, benzyl and aryl; Rll is independently selected from C1-C6 alkyl and aryl; R12 is selected from: H; unsubstituted or substituted C1-8 alkyl, unsubstituted or substituted aryl or unsubstituted or substituted heterocycle, wherein the substituted alkyl, substituted aryl or substituted heterocycle is substituted with one or more of:

1) aryl or heterocycle, unsubstituted or substituted with: a) C1-4 alkyl, b) (CH2) pOR6, c) (CH2)pNR6R7, d) halogen, e) CN, f) aryl or heteroaryl, g) perfluoro-C1-4 alkyl, h) SR6a, S(O)R6a, SO2R6a, 2) C3-6 cycloalkyl, 3) OR6, 4) SR6a, S(O)R6a, or SO2R6a,

15) N3, 16) F, 17) perfluoro-C1-4-alkyl, or 18) C1-6-alkyl; A1 and A2 are independently selected from: a bond, -CH=CH-, -CsC-, -C(O)-, -C(O)NR10-, -NR10C(O)-, O, -N(R10)-, or S(O)m; V is selected from: a) hydrogen, b) heterocycle selected from pyrrolidinyl, imidazolyl, pyridinyl, thiazolyl, pyridonyl, 2-oxopiperidinyl, indolyl, quinolinyl, isoquinolinyl, and thienyl,

c) aryl, d) C1-C20 alkyl wherein from 0 to 4 carbon atoms are replaced with a heteroatom selected from 0, S, and N, and e) C2-C20 alkenyl, and provided that V is not hydrogen if A1 is S(O)m and V is not hydrogen if A1 is a bond, n is 0 and A2 is S(O)m; X is -CH2- or -C(=O)-; X1 is a bond, -C(=O)-, -NR6C(=O)-, -NR6-, -O- or Y is selected from: a) hydrogen, b) R10O-, R11S(O)m-, R10C(O)NR10-, (R10)2N-C(O)-, CN, NO2, (R10)2N-C(NR10)-, R12C(O)-, R10OC(O)-, N3, F, -N(R10)2, or R11 OC(O)NR10-, c) unsubstituted or substituted Cl -C6 alkyl wherein the substituent on the substituted C1-C6 alkyl is selected from unsubstituted or substituted aryl, R10O-, R10C(O)NR10-, (R10)2N-C(O)-, R10C(O)- and R10OC(O)-; Z is an unsubstituted or substituted aryl, wherein the substituted aryl is substituted with one or more of the following: 1) C1-4 alkyl, unsubstituted or substituted with: a) C 1-4 alkoxy, b) NR6R7, c) C3-6 cycloalkyl, d) aryl, substituted aryl or heterocycle, e) HO, f) -S(O)mR6a, or g) -C(O)NR6R7, 2) aryl or heterocycle, 3) halogen, 4) OR6, 5) NR6R7, 6) CN, 7) NO2, 8) CF3; 9) -S(O)mR6a, 10) -C(O)NR6R7, or 11) C3-C6 cycloalkyl; m is 0, 1 or 2; n is 0, 1, 2, 3 or 4; p is 0, 1, 2, 3 or 4; and r is 0 to 5, provided that r is 0 when V is hydrogen; and v is 0, 1 or 2; (c) a compound represented by formula III: wherein: R1 is independently selected from: hydrogen or C1-C6 alkyl; R2 is independently selected from: a) hydrogen, b) substituted or unsubstituted aryl, substituted or unsubstituted heterocycle, C3-C10 cycloalkyl, R10O- or C2-C6 alkenyl, c) C1-C6 alkyl unsubstituted or substituted by aryl, heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, R10O-, or -N(R10)2; R3 is selected from: a) hydrogen,

b) C1-C6 alkyl unsubstituted or substituted by C2-C6 alkenyl, R10O-, R11S(O)m-, R10C(O)NR10-, CN, N3, (R10)2N-C(NR10)-, R10C(O)-, -N(R10)2, or R11OC(O)NR10-, c) substituted or unsubstituted aryl, substituted or unsubstituted heterocycle, C3-C 10 cycloalkyl, C2-C6 alkenyl, fluoro, chloro, R12O-, R11S(O)m-, R10C(O)NR10-, CN, NO2, (R10)2N-C(NR10)-, R10C(O)-, N3, -N(R10)2, or R1 1OC(O)NR10-, and d) C1-C6 alkyl substituted with an unsubstituted or substituted group selected from substituted or unsubstituted aryl, substituted or unsubstituted heterocyclic and C3 -C 10 cycloalkyl; R4 and R5 are independently selected from: a) hydrogen, b) C1-C6 alkyl unsubstituted or substituted by C2-C6 alkenyl, R10O-, R11S(O)m-, R10C(O)NR10-, CN, N3, (R10)2N-C(NR10)-, R10C(O)-, -N(R10)2, or R11 OC(O)NR10-, c) substituted or unsubstituted aryl, substituted or unsubstituted heterocycle, C3 -C 10 cycloalkyl, C2-C6 alkenyl, fluoro, chloro, R100-, R11S(O)m-, R10C(O)NR10-, CN, NO2, (R10)2N-C(NR10)-, R10C(O)-, N3, -N(R10)2, or R11OC(O)NR10-, and d) C1-C6 alkyl substituted with an unsubstituted or substituted group selected from substituted or unsubstituted aryl, substituted or unsubstituted heterocyclic and C3-C10 cycloalkyl; R6 is independently selected from:

a) hydrogen, b) substituted or unsubstituted aryl, substituted or unsubstituted heterocycle, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 perfluoroalkyl, F, C1, R10O-, allyloxy, R10C(O)NR10-, CN, NO2, (R10)2N-C(NR10)-, R10C(O)-, -N(R10)2, (R12)2NC(O)- or R11OC(O)NR10-, and c) C1-C6 alkyl substituted by C1-C6 perfluoroalkyl, R10O-, R10C(O)NR10-, (R10)2N-C(NR10)-, R10C(O)-, -N(R10)2, or R11OC(O)NR10-; R7 is independently selected from a) hydrogen, b) unsubstituted or substituted aryl, c) unsubstituted or substituted heterocycle, d) unsubstituted or substituted cycloalkyl, and e) C1-C6 alkyl substituted with hydrogen or an unsubstituted or substituted group selected from aryl, heterocycle and cycloalkyl; wherein heterocycle is selected from pyrrolidinyl, imidazolyl, pyridinyl, thiazolyl, pyridonyl, indolyl, quinolinyl, isoquinolinyl, and thienyl; R8 is selected from: a) hydrogen, b) C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 perfluoroalkyl, F, C1, R10O-, R10C(O)NR10-, CN, NO2, (R10)2N-C(NR10)-, R10C(O)-, R10OC(O)-, -N(R10)2, or R11OC(O)NR10-, and c) C1-C6 alkyl substituted by C1-C6 perfluoroalkyl, R10O-, R10C(O)NR10-, (R10)2N-C(NR10)-, R10C(O)-, R10OC(O)-, -N(R10)2, or R11OC(O)NR10-;

R9 is selected from: a) hydrogen, b) C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 perfluoroalkyl, F, Cl, R10O-, R11S(O)m-, R10C(O)NR10-, CN, NO2, (R10)2N-C(NR10)-, R10C(O)-, R10OC(O)-, -N(R10)2, or R11OC(O)NR10-, and c) C1-C6 alkyl unsubstituted or substituted by C1-C6 perfluoroalkyl, F, Cl, R10O-, R11S(O)m-, R10C(O)NR10-, CN, (R10)2N-C(NR10)-, R10C(O)-, R10OC(O)-, -N(R10)2, or R11OC(O)NR10-; R10 is independently selected from hydrogen, C1-C6 alkyl, Cl-C6 perfluoroalkyl, 2,2,2-trifluoroethyl, benzyl and aryl; Rll is independently selected from C1-C6 alkyl and aryl; R12 is independently selected from hydrogen, C1-C6 alkyl, C1-C6 alkyl substituted with CO2R10, C1-C6 alkyl substituted with aryl, C1-C6 alkyl substituted with substituted aryl, C1-C6 alkyl substituted with heterocycle, C1-C6 alkyl substituted with substituted heterocycle, aryl and substituted aryl; A1 and A2 are independently selected from: a bond, -CH=CH-, -C#C-, -C(O)-, -C(O)NR7-, -NR7C(O)-, -S(O)2NR7-, -NR7S(O)2-, O, -N(R7)-, or S(O)m; A3 is selected from: a bond, -C(O)NR7-, -NR7C(O)-, -S(O)2NR7-, -NR7S(O)2- or -N(R7)-; A4 is selected from: a bond, 0, -N(R7)- or S; V is selected from: a) hydrogen,

b) heterocycle selected from pyrrolidinyl, imidazolyl, pyridinyl, thiazolyl, pyridonyl, 2-oxopiperidinyl, indolyl, quinolinyl, isoquinolinyl, and thienyl, c) aryl, d) C1-C20 alkyl wherein from 0 to 4 carbon atoms are replaced with a heteroatom selected from 0, S, and N, and e) C2-C20 alkenyl, and provided that V is not hydrogen if A1 is S(O)m and V is not hydrogen if Al is a bond, n is 0 and A2 is S(O)m; Z is independently (R1)2 or O; m is 0, 1 or 2; n is 0, 1, 2, 3 or 4; p is 0, 1, 2, 3 or 4; qis O or 1; and r is 0 to 5, provided that r is 0 when V is hydrogen; d) a compound represented by formula A: wherein: Rla is selected from: hydrogen or C1-C6 alkyl; Rlb is independently selected from: a) hydrogen, b) aryl, heterocycle, cycloalkyl, R10O-, -N(R10)2 or C2-C6 alkenyl,

c) C1-C6 alkyl unsubstituted or substituted by aryl, heterocycle, cycloalkyl, alkenyl, R10O-, or -N(R10)2; R2a, R2b and R3 are independently selected from: a) hydrogen, b) C1-C6 alkyl unsubstituted or substituted by C2-C6 alkenyl, R10O-, R11S(O)m-, R10C(O)NR10-, CN, N3, (R10)2N-C(NR10)-, R10C(O)-, R10OC(O)-, -N(R10)2, or R11OC(O)NR10-, c) unsubstituted or substituted aryl, unsubstituted or substituted heterocycle, unsubstituted or substituted cycloalkyl, alkenyl, R10O-, R11S(O)M-, R10C(O)NR10-, CN, NO2, (R10)2N-C(NR10)-, R10C(O)-, R10OC(O)-, N3, -N(R10)2, halogen or R1 1OC(O)NR10-, and d) C 1 -C6 alkyl substituted with an unsubstituted or substituted group selected from aryl, heterocyclic and C3-C10 cycloalkyl; R4 is R5 is hydrogen; R8 is selected from: a) hydrogen, b) C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, Cl-C6 perfluoroalkyl, F, C1, R100-, R10C(O)NR10-, CN, N02, (R10)2N-C(NR10)-, R10C(O)-, R10OC(O)-, -N(R10)2, or R11OC(O)NR10-, and

c) C1-C6 alkyl substituted by C1-C6 perfluoroalkyl, R100-, R10C(O)NR10-, (R10)2N-C(NR10)-, R10C(O)-, R10OC(O)-, -N(R10)2, or R1 1OC(O)NR10-; R9a is independently selected from C1-C6 alkyl and aryl; R10 is independently selected from hydrogen, Cl -C6 alkyl, Cl -C6 perfluoroalkyl, 2,2,2-trifluoroethyl, benzyl and aryl; Rll is independently selected from C1-C6 alkyl, benzyl and aryl; A1 and A2 are independently selected from: a bond, -CH=CH-, -C#C-, -C(O)-, -C(O)NR8-, -NR8C(O)-, O, -N(R8)-, -S(O)2N(R8)-, -N(R8)S(O)2-, or S(O)m; V is selected from: a) hydrogen, b) heterocycle selected from pyrrolidinyl, imidazolyl, pyridinyl, thiazolyl, pyridonyl, 2-oxopiperidinyl, indolyl, quinolinyl, isoquinolinyl, and thienyl, c) aryl, d) C1-C20 alkyl wherein from 0 to 4 carbon atoms are replaced with a heteroatom selected from 0, S, and N, and e) C2-C20 alkenyl, provided that V is not hydrogen if A1 is S(O)m and V is not hydrogen if A1 is a bond, n is 0 and A2 is S(O)m; Z is H2 or O; m is 0, 1 or 2; n is 0, 1, 2, 3 or 4; p is independently 0, 1, 2, 3 or 4; and r is 0 to 5, provided that r is 0 when V is hydrogen;

or the pharmaceutically acceptable salts thereof.

In a further embodiment of the formula I compounds of this invention, the inhibitors of farnesyl-protein transferase are illustrated by the formula I-a: wherein: Rlb is independently selected from: a) hydrogen, b) aryl, heterocycle, cycloalkyl, R10O-, -N(R10)2 or C2-C6 alkenyl, c) C1-C6 alkyl unsubstituted or substituted by aryl, heterocycle, cycloalkyl, alkenyl, R10O-, or -N(R10)2; R2 is selected from H; unsubstituted or substituted aryl or C1-5 alkyl, unbranched or branched, unsubstituted or substituted with one or more of: 1) aryl, 2) heteroaryl, 3) OR6, or 4) SR6a; R6 and R7 are independently selected from: : C1 4 alkyl, aryl, and heteroaryl, unsubstituted or substituted with: a) C 1-4 alkoxy, b) halogen, c) pefrluoro-C1-4 alkyl, or d) aryl or heteroaryl;

R6a is selected from: C1-4 alkyl, unsubstituted or substituted with: a) C 1-4 alkoxy, or b) aryl or heteroaryl; R8 is independently selected from: a) hydrogen, b) C1-6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, Cl-C6 perfluoroalkyl, F, C1, R10O-, R10C(O)NR10-, CN, NO2, (R10)2N-C(NR10)-, R10C(O)-, -N(R10)2, or Rll 1OC(O)NR10-, and c) C1-C6 alkyl substituted by C1-C6 perfluoroalkyl, R10O-, R10C(O)NR10-, (R10)2N-C(NR10)-, R10C(O)-, -N(R10)2, or R11OC(O)NR10-; R10 is independently selected from hydrogen, C1-C6 alkyl, C1-C6 perfluoroalkyl, 2,2,2-trifluoroethyl, benzyl and aryl; R11 is independently selected from C1-C6 alkyl and aryl; X is -CH2- or Z is an unsubstituted or substituted group selected from aryl, arylmethyl and arylsulfonyl, wherein the substituted group is substituted with one or more of the following: a) C1-4 alkyl, unsubstituted or substituted with: C1-4 alkoxy, NR6R7, C3-6 cycloalkyl, unsubstituted or substituted aryl, heterocycle, HO, -S(O)mR6a, or -C(O)NR6R7, b) aryl or heterocycle, c) halogen, d) OR6,

e) NR6R7, f) CN, g) N02, h) CF3; i) S(O)mR6a, j> -C(O)NR6R7, or k) C3-C6 cycloalkyl; m is 0, 1 or 2; and p is 0, 1, 2, 3 or 4; and r is 0 to 3; or the pharmaceutically acceptable salts thereof; In another embodiment of this invention, the inhibitors of farnesyl-protein transferase are illustrated by the formula II-a: wherein: Rlb is independently selected from: a) hydrogen, b) aryl, heterocycle, cycloalkyl, R100-, -N(R10)2 or C2-C6 alkenyl, c) C1-C6 alkyl unsubstituted or substituted by unsubstituted or substituted aryl, heterocycle, cycloalkyl, alkenyl, R10O-, or -N(R10)2; Rle is selected from: a) hydrogen,

b) unsubstituted or substituted C1-C6 alkyl wherein the substituent on the substituted C1-C6 alkyl is selected from unsubstituted or substituted aryl, heterocyclic, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, R100-, R11S(O)m-, R10C(O)NR10-, (R10)2N-C(O)-, CN, (R10)2N-C(NR10)-, R10C(O)-, R10OC(O)-, N3, -N(R10)2, and R110C(O)-NR10-, and c) unsubstituted or substituted aryl; R6, R7 and R7a are independently selected from: H; C1-4 alkyl, C3-6 cycloalkyl, aryl, heterocycle, unsubstituted or substituted with: a) C 1-4 alkoxy, b) halogen, or c) aryl or heterocycle; R6a is selected from: C1-4 alkyl or C3-6 cycloalkyl, unsubstituted or substituted with: a) C 1-4 alkoxy, b) halogen, or c) aryl or heterocycle; R8 is independently selected from: a) hydrogen, b) C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, Cl-C6 perfluoroalkyl, F, C1, R100-, R10C(O)NR10-, CN, NO2, (R10)2N-C(NR10)-, R10C(O), -N(R10)2, or R11OC(O)NR10-, and c) C1-C6 alkyl substituted by C1-C6 perfluoroalkyl, R10-, R10C(O)NR10-, (R10)2N-C(NR10)-, R10C(O)-, -N(R10)2, or R11OC(O)NR10-;

R10 is independently selected from hydrogen, C1-C6 alkyl, C1-C6 perfluoroalkyl, 2,2,2-trifluoroethyl, benzyl and aryl; R11 is independently selected from C1-C6 alkyl and substituted or unsubstituted aryl; R12 is selected from: H; unsubstituted or substituted C1-8 alkyl, unsubstituted or substituted aryl or unsubstituted or substituted heterocycle, wherein the substituted alkyl, substituted aryl or substituted heterocycle is substituted with one or more of: 1) aryl or heterocycle, unsubstituted or substituted with: a) C1-4 alkyl, b) halogen, c) CN, d) perfluoro-C1-4 alkyl, 2) C3-6 cycloalkyl, 3) OR6, 4) SR6a, S(O)R6a, or So2R6a 7) N3, 8) F, 9) pertluorn-C14-alkyl, or 10) C1-6-alkyl; X1 is a bond, -C(=O)- (=O) or

Y is selected from: a) hydrogen, b) R10O-, R11S(O)m-, R10C(O)NR10-, (R10)2N-C(O)-, CN, NO2, (R10)2N-C(NR10)-, R12C(O)-, R10OC(O)-, N3, F, -N(R10)2, or R11OC(O)NR10-, c) unsubstituted or substituted C1-C6 alkyl wherein the substituent on the substituted C1-C6 alkyl is selected from unsubstituted or substituted aryl, R100-, R10C(O)NR10-, (R10)2N-C(O)-, R10C(O)- and R10OC(O)-; Z is an unsubstituted or substituted aryl, wherein the substituted aryl is substituted with one or more of the following: 1) C1-4 alkyl, unsubstituted or substituted with: a) C1-4 alkoxy, b) NR6R7, c) C3-6 cycloalkyl, d) aryl, substituted aryl or heterocycle, e) HO, f) -S(O)mR6a, or g) -C(O)NR6R7, 2) aryl or heterocycle, 3) halogen, 4) OR6, 5) NR6R7, 6) CN, 7) N02, 8) CF3; 9) -S(O)mR6a, 10) -C(O)NR6R7, or 11) C3-C6 cycloalkyl; m is 0, 1 or 2; p is 1 or 2 ; ris O to 3; and

v is 0, 1 or 2; or a pharmaceutically acceptable salt thereof.

In a further embodiment of this invention, the inhibitors of farnesyl-protein transferase are illustrated by the formula III-a: wherein: R2 is independently selected from: a) hydrogen, b) aryl, heterocycle, cycloalkyl, R10O-, -N(R10)2 or C2-C6 alkenyl, c) C1-C6 alkyl unsubstituted or substituted by aryl, heterocycle, cycloalkyl, alkenyl, R10O-, or -N(R10)2; R3 is selected from: a) hydrogen, b) C1-C6 alkyl unsubstituted or substituted by C2-C6 alkenyl, R10O-, R11S(O)m-, R10C(O)NR10-, CN, N3, (R10)2N-C(NR10)-, R10C(O)-, -N(R10)2, or R11OC(O)NR10-, c) substituted or unsubstituted aryl, substituted or unsubstituted heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, fluoro, chloro, R12O-, R11S(O)m-, R10C(O)NR10-, CN, NO2, (R1 0)2N-C(NR10)-, R10C(O)-, N3, -N(R10)2, or R11OC(O)NR10-, and

d) C1-C6 alkyl substituted with an unsubstituted or substituted group selected from substituted or unsubstituted aryl, substituted or unsubstituted heterocyclic and C3-C10 cycloalkyl; R4 and R5 are independently selected from: a) hydrogen, b) C1-C6 alkyl unsubstituted or substituted by R10O- or -N(R1 0)2, c) substituted or unsubstituted aryl, substituted or unsubstituted heterocycle, C3-C 10 cycloalkyl, C2-C6 alkenyl, fluoro, chloro, R10O-, R1 1S(O)m-, R10C(O)NR10-, CN, N02, (R10)2N-C(NR10)-, R10C(O)-, N3, -N(R10)2, or R11OC(O)NR10-, and d) C1-C6 alkyl substituted with an unsubstituted or substituted group selected from substituted or unsubstituted aryl, substituted or unsubstituted heterocyclic and C3 -C 10 cycloalkyl; R6 is independently selected from: a) hydrogen, b) substituted or unsubstituted aryl, substituted or unsubstituted heterocycle, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 perfluoroalkyl, F, C1, R100-, allyloxy, R10C(O)NR10-, CN, NO2, (R10)2N-C(NR10)-, R10C(O)-, -N(R10)2, (R12)2NC(O)- or R11OC(O)NR10-, and c) C1-C6 alkyl substituted by C1-C6 perfluoroalkyl, R10O-, R10C(O)NR10-, (R10)2N-C(NR10)-, R10C(O)-, -N(R10)2, or R1 1OC(O)NR10-; R7 is independently selected from

a) hydrogen, b) unsubstituted or substituted aryl, c) unsubstituted or substituted heterocycle, d) unsubstituted or substituted cycloalkyl, and e) C1-C6 alkyl substituted with hydrogen or an unsubstituted or substituted group selected from aryl, heterocycle and cycloalkyl; wherein heterocycle is selected from pyrrolidinyl, imidazolyl, pyridinyl, thiazolyl, pyridonyl, indolyl, quinolinyl, isoquinolinyl, and thienyl; R8 is independently selected from: a) hydrogen, b) C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, Cl-C6 perfluoroalkyl, F, C1, R10O-, R10C(O)NR10-, CN, NO2, (R10)2N-C(NR10)-, R10C(O)-, -N(R10)2, or R11OC(O)NR10-, and c) C1-C6 alkyl substituted by C1-C6 perfluoroalkyl, R10O-, R10C(O)NR10-, (R10)2N-C(NR10)-, R10C(O)-, -N(R10)2, or Rl 1OC(O)NR10-; R10 is independently selected from hydrogen, Cl -C6 alkyl, C1-C6 perfluoroalkyl, 2,2,2-trifluoroethyl, benzyl and aryl; Rll is independently selected from C1-C6 alkyl and aryl; R12 is independently selected from hydrogen, C1-C6 alkyl, C1-C6 alkyl substituted with C02R10, C1-C6 alkyl substituted with aryl, C1-C6 alkyl substituted with substituted aryl, C1-C6 alkyl substituted with heterocycle, C1-C6 alkyl substituted with substituted heterocycle, aryl and substituted aryl; A3 is selected from: a bond, -C(O)NR7-, -NR7C(O)-, -S(O)2NR7-,

-NR7S(O)2- or -N(R7)-; Z is independently H2 or O; m is 0, 1 or 2; and n is 0, 1, 2, 3 or 4; ps is 0, 1, 2, 3 or 4; qis 0 or 1; and r is 0 to 3; or the pharmaceutically acceptable salts thereof.

In a further embodiment of the formula A compounds of this invention, the inhibitors of farnesyl-protein transferase are illustrated by the formula A-i: wherein: Rlb is independently selected from: a) hydrogen, b) aryl, heterocycle, cycloalkyl, R10O-, -N(R10)2 or C2-C6 alkenyl, c) C1-C6 alkyl unsubstituted or substituted by aryl, heterocycle, cycloalkyl, alkenyl, R100-, or -N(R10)2; R2a and R2b are independently selected from: a) hydrogen, b) C1-C6 alkyl unsubstituted or substituted by C2-C6 alkenyl, R100-, R11S(O)m-, R10C(O)NR10-, CN, N3, (R10)2N-C(NR10)-, R10C(O)-, R10OC(O)-, -N(R10)2, or R11OC(O)NR10-,

c) unsubstituted or substituted aryl, unsubstituted or substituted heterocycle, unsubstituted or substituted cycloalkyl, alkenyl, R100-, R11S(O)m-, R10C(O)NR10-, CN, NO2, (R10)2N-C(NR10)-, R10C(O)-, R10OC(O)-, N3, -N(R10)2, halogen or R11OC(O)NR10-, and d) C1-C6 alkyl substituted with an unsubstituted or substituted group selected from aryl, heterocyclic and C3-C 10 cycloalkyl; R4 is R5 is hydrogen; R8 is independently selected from: a) hydrogen, b) C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, Cl-C6 perfluoroalkyl, F, C1, R10O-, R10C(O)NR10-, CN, NO2, (R10)2N-C(NR10)-, R10C(O)-, -N(R10)2, or R110C(O)NR10-, and c) C1-C6 alkyl substituted by C1-C6 perfluoroalkyl, R10O-, R10C(O)NR10-, (R10)2N-C(NR10)-, R10C(O)-, -N(R10)2, or R11OC(O)NR10-; R10 is independently selected from hydrogen, C1-C6 alkyl, substituted or unsubstituted C1-C6 aralkyl and substituted or unsub stituted aryl;

R1 1 is independently selected from C1-C6 alkyl, benzyl and aryl; Z is H2 or O; m is 0, 1 or 2; n is 0, 1, 2, 3 or 4; p is independently 0, 1 or 2; and r is 0 to 5; or the pharmaceutically acceptable salts thereof.

Specific compounds which are inhibitors of prenyl- protein transferases and are therefore useful in the present invention include: 1-[2(R)-Amino-3-mercaptopropyl]-2(S)-[(3-pyridyl)methoxyethy l)]- 4-( 1 -naphthoyl)piperazine 1-[2(R)-Amino-3-mercaptopropyl]-2(S)-(benzyloxymethyl)-4(1- naphthoyl)piperazine 1-[2(R)-Amino-3-mercaptopropyl]-2(S)-(benzyloxymethyl)-4-[7- (2,3-dihydrobenzofuroyl)]piperazine 1-[2(R)-Amino-3-mercaptopropyl]-2(S)-(benzamido)-4-(1- naphthoyl)piperazine 1-[2(R)-Amino-3-mercaptopropyl]-2(S)-[4-(5-dimethylamino-1- naphyhalenesulfonamido)-1-butyl]-4-(1-naphthonyl)piperazine N-[2(S)-(1-(4-Nitrophenylmethyl)-1H-imidazol-5-ylacetyl)amin o- 3 (S)-methylpentyl]-N-1-naphthylmethyl-glycyl-methionine N-[2(S)-(1-(4-Nitrophenylmethyl)-1H-imidazol-5-ylacetyl)amin o- 3 (S)-methylpentyl] -N-l -naphthylmethyl-glycyl-methionine methyl ester

N-[2(S)-([1-(4-cyanobenzyl)-1H-imidazol-5-yl]acetylamino)-3( S)- methylpentyl]-N-( 1 -naphthylmethyl)glycyl-methionine N-[2(S)-([1-(4-cyanobenzyl)-1H-imidazol-5-yl]acetylamino)-3( S)- methylpentyl]-N-(1 -naphthylmethyl)glycyl-methionine methyl ester 2(S)-n-Butyl-4-(1-naphthoyl)-1-[1-(2-naphthylmethyl)imidazol -5- ylmethyl]-piperazine 2(S)-n-Butyl- 1 - [1 -(4-cyanobenzyl)imidazol-5-ylmethyl] -4-( 1 - naphthoyl)piperazine 1-{[1-(4-cyanobenzyl)-1H-imidazol-5-yl]acetyl}-2(S)-n- butyl-4(1-naphthonyl)piperazine 1 -(3-chlorophenyl)-4- [1 -(4-cyanobenzyl)imidazolylmethyl]-2- piperazinone 1-phenyl-4-[1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl]-piperaz in-2-one 1-(3-trifluoromethylphenyl)-4-[1-(4-cyanobenzyl)-1H-imidazol -5- ylmethyl] -piperazin-2-one 1-(3-bromophenyl)-4-[1-(4-cyanobenzyl)-1H-imidazol-5-ylmethy l]- piperazin-2-one 5(S)-(2-[2,2,2-trifluoroethoxy]ethyl)-1-(3-trifluoromethylph enyl)- 4-[1- (4-cyanobenzyl)-4-imidazolylmethyl]-piperazin-2-one 1-(5,6,7,8-tetrahydronaphthyl)-4-[1-(4-cyanobenzyl)-1H-imida zol-5- ylmethyl]-piperazin-2-one 1-(2-methyl-3-chlorophenyl)-4-[1-(4-cyanobenzyl)-4- imidazolylmethyl)] -piperazin-2-one

2(RS)-{[1-(Naphth-2-ylmethyl)-1H-imidazol-5-yl)] acetyl}amino-3-(t- butoxycarbonyl)amino- N-(2-methylbenzyl) propionamide N-{1-(4-Cyanobenzyl)-1H-imidazol-5-ylmethyl}-4(R)-benzyloxy- 2(S)- (N'-acetyl-N'-3 -chlorobenzyl) aminomethylpyrrolidine N- { 1 -(4-Cyanobenzyl)- 1 H-imidazol-5 -ylethyl 1 -4(R)-benzyloxy-2(S)- {N'-acetyl-N'-3-chlorobenzyl}aminomethyl pyrrolidine 1-[1-(4-Cyanobenzyl)-1H-imidazol-5-ylacetyl] pyrrolidin-2(S)- ylmethyl]-(N-2-methylbenzyl)-glycine N'-(3-chlorophenylmethyl) amide 1-[1-(4-Cyanobenzyl)-1H-imidazol-5-ylacetyl] pyrrolidin-2(S)- ylmethyl]-(N-2-methylbenzyl)-glycine N'-methyl-N'-(3- chlorophenylmethyl) amide (S)-2-[(1 (4-Cyanobenzyl)-5-imidazolylmethyl)amino -N- (benzyloxycarbonyl)-N- (3 -chlorobenzyl)-4- (methanesulfonyl)butanamine 1-(3,5-Dichlorobenzenesulfonyl)-3(S)-[N-(1-(4-cyanobenzyl)-1 H- imidazol-5-ylethyl)carbamoyl] piperidine N-{[1-(4-Cyanobenzyl)-1H-imidazol-5-yl]methyl}-4-(3- methylphenyl)-4-hydroxy piperidine, N-{[1-(4-Cyanobenzyl)-1H-imidazol-5-yl]methyl}-4-(3- chlorophenyl)-4 hydroxy piperidine, 4-[1-(4-cyanobenzyl)-5-imidazolylmethyl]-1-(2,3-dimethylphen yl)- piperazine-2,3-dione 1-(2-(3-Trifluoromethoxyphenyl)-pyrid-5-ylmethyl)-5-(4- cyanobenzyl)imidazole

4-(5-[1-(3 -Chloro-phenyl)-2- oxo-l ,2-dihydro-pyridin-4-ylmethyl] - imidazol-1-ylmethyl}-2-methoxy-benzonitrile 3(R)-3-[1-(4-Cyanobenzyl)imidazol-5-yl-ethylamino]-5-phenyl- 1- (2,2,2-trifluoroethyl)-H-benzo[e][1,4] diazepine 3(S)-3-[1-(4-Cyanobenzyl) imidazol-5-yl]-ethylamino]-5-phenyl-1- (2,2,2-trifluoroethyl)-H-benzo[e][1,4] diazepine N-[1-(4-Cyanobenzyl)-1H-imidazol-5-ylacethyl)pyrrolidin-2(S) - ylmethyl]- N-(1-naphthylmethyl)glycyl-methionine N-[1-(4-Cyanobenzyl)-1H-imidazol-5-ylacetyl)pyrrolidin-2(S)- ylmethyl]- N-(1-naphthylmethyl)glycyl-methionine methyl ester N-[1-(1H-Imidazol-4-ylpropionyl)pyrrolidin-2(S)-ylmethyl]- N-(2- methoxybenzyl)glycyl-methionine N-[1-(1H-Imidazol-4-ylpropionyl)pyrrolidin-2(S)-ylmethyl]- N-(2- methoxybenzyl)glycyl-methionine methyl ester 2(S)-(4-Acetamido-1-butyl)-1-[2(R)-amino-3-mercaptopropyl]-4 -(1- naphthoyl)piperazine 2(RS)-{[1-(Naphth-2-ylmethyl)-1H-imidazol-5-yl)] acetyl}amino-3-(t- butoxycarbonyl)amino- N-cyclohexyl-propionamide 1-{2(R,S)-[1-(4-cyanobenzyl)-1H-imidazol-5-yl]propanoyl}-2(S )-n- butyl-4-(1-naphthoyl)piperazine 1-[1-(4-cyanobenzyl)imidazol-5-ylmethyl]-4- (diphenylmethyl)piperazine 1-(Diphenylmethyl)-3(S)-[N-(1-(4-cyanobenzyl)-2-methyl-1H- imidazol- 5 -ylethyl)-N- (ac etyl) aminomethyl] piperidine

N-[1-(1H-Imidazol-4-ylpropionyl)pyrrolidin-2(S)-ylmethyl]- N-(2- chlorobenzyl)glycyl-methionine N-[1-(1H-Imidazol-4-ylpropionyl)pyrrolidin-2(S)-ylmethyl]- N-(2- chlorobenzyl)glycyl-methionine methyl ester 3(R)-3-[1-(4-Cyanobenzyl)imidazol-5-yl-methylamino]-5-phenyl -1- (2,2,2-trifluoroethyl)-H-benzo[e][1,4] diazepine 1-(3-trifluoromethoxyphenyl)-4-[1-(4- cyanobenzyl)imidazolylmethyl]-2-piperazinone 1-(2,5-dimethylphenyl)-4-[1-(4-cyanobenzyl)imidazolylmethyl] -2- piperazinone 1-(3-methylphenyl)-4-[1-(4-cyanobenzyl)imidazolylmethyl]-2- piperazinone 1 -(3 -iodophenyl)-4- [1- (4-cyanobenzyl)imidazolylmethyll -2- piperazinone 1-(3-chlorophenyl)-4-[1-(4-cyano-3- methoxybenzyl)imidazolylmethyl]-2-piperazinone 1-(3-trifluoromethoxyphenyl)-4- [1-(4-cyano-3- methoxybenzyl)imidazolyl methyl]-2-piperazinone 4-[((1-(4-cyanobenzyl)-5-imidazolyl)methyl)amino]benzophenon e 1-(1-{[3-(4-cyano-benzyl)-3H-imidazol-4-yl]-acetyl}-pyrrolid in- 2(S)-ylmethyl)-3(S)-ethyl-pyrrolidine-2(S)-carboxylic acid 3-chloro- benzylamide or the pharmaceutically acceptable salt thereof.

Compounds within the scope of this invention previously described as inhibitors of farnesyl-protein transferase but which have been further identified by the instant assays as inhibitors of prenyl- protein transferases and are therefore useful in the present invention, and methods of synthesis thereof, can be found in the following patents, pending applications and publications, which are herein incorporated by reference: U.S. Pat. No. 5,736,539 (April 7, 1998); WO 95/00497 (January 5, 1995) U.S. Pat. No. 5,652,257 (July 29, 1997); WO 96/10034 (April 4, 1996) WO 96/30343 (October 3, 1996); USSN 08/412,829 filed on March 29, 1995; and USSN 08/470,690 filed on June 6, 1995; and USSN 08/600,728 filed on February 28, 1996; U.S. Pat. No. 5,661,161 (August 26, 1997); U.S. Pat. No. 5,756,528 (May 6, 1998); WO 96/39137 (December 12, 1996); WO 96/37204 (November 28, 1996); USSN 08/449,038 filed on May 24, 1995; USSN 08/648,330 filed on May 15, 1996; WO 97/18813 (May 29, 1997); USSN 08/749,254 filed on November 15, 1996; WO 97/38665 (October 23, 1997); USSN 08/831,308 filed on April 1, 1997; WO 97/36889 (October 9, 1997); USSN 08/823,923 filed on March 25, 1997 ; WO 97/36901 (October 9, 1997); USSN 08/827,483 filed on March 27, 1997 ; WO 97/36879 (October 9, 1997); USSN 08/823,920 filed on March 25, 1997 ; WO 97/36605 (October 9, 1997); USSN 08/823,934 filed on March 25, 1997 ; WO 98/28980, (July 9, 1998); USSN 08/997,171 filed on December 22, 1997; and

USSN 60/014,791 filed on April 3, 1996; USSN 08/831,308, filed on April 4, 1997.

All patents, publications and pending patent applications identified are hereby incorporated by reference.

With respect to the compounds of formulas II-a through II-n the following definitions apply: The term "alkyl" refers to a monovalent alkane (hydrocarbon) derived radical containing from 1 to 15 carbon atoms unless otherwise defined. It may be straight, branched or cyclic.

Preferred straight or branched alkyl groups include methyl, ethyl, propyl, isopropyl, butyl and t-butyl. Preferred cycloalkyl groups include cyclopentyl and cyclohexyl.

When substituted alkyl is present, this refers to a straight, branched or cyclic alkyl group as defined above, substituted with 1-3 groups as defined with respect to each variable.

Heteroalkyl refers to an alkyl group having from 2-15 carbon atoms, and interrupted by from 1-4 heteroatoms selected from 0, S and N.

The term "alkenyl" refers to a hydrocarbon radical straight, branched or cyclic containing from 2 to 15 carbon atoms and at least one carbon to carbon double bond. Preferably one carbon to carbon double bond is present, and up to four non- aromatic (non-resonating) carbon-carbon double bonds may be present. Examples of alkenyl groups include vinyl, allyl, iso- propenyl, pentenyl, hexenyl, heptenyl, cyclopropenyl, cyclobutenyl, cyclopentenyl, cyclohexenyl, l-propenyl, 2-butenyl, 2-methyl-2- butenyl, isoprenyl, farnesyl, geranyl, geranylgeranyl and the like.

Preferred alkenyl groups include ethenyl, propenyl, butenyl and cyclohexenyl. As described above with respect to alkyl, the straight, branched or cyclic portion of the alkenyl group may contain double bonds and may be substituted when a substituted alkenyl group is provided.

The term "alkynyl" refers to a hydrocarbon radical straight, branched or cyclic, containing from 2 to 15 carbon atoms and at least one carbon to carbon triple bond. Up to three carbon- carbon triple bonds may be present. Preferred alkynyl groups include ethynyl, propynyl and butynyl. As described above with respect to alkyl, the straight, branched or cyclic portion of the alkynyl group may contain triple bonds and may be substituted when a substituted alkynyl group is provided.

Aryl refers to aromatic rings e.g., phenyl, substituted phenyl and like groups as well as rings which are fused, e.g., naphthyl and the like. Aryl thus contains at least one ring having at least 6 atoms, with up to two such rings being present, containing up to 10 atoms therein, with alternating (resonating) double bonds between adjacent carbon atoms. The preferred aryl groups are phenyl and naphthyl. Aryl groups may likewise be substituted as defined below. Preferred substituted aryls include phenyl and naphthyl substituted with one or two groups. With regard to the farnesyl transferase inhibitors, "aryl" is intended to include any stable monocyclic, bicyclic or tricyclic carbon rings of up to 7 members in each ring, wherein at least one ring is aromatic.

Examples of aryl groups include phenyl, naphthyl, anthracenyl, biphenyl, tetrahydronaphthyl, indanyl, phenanthrenyl and the like.

The term "heteroaryl" refers to a monocyclic aromatic hydrocarbon group having 5 or 6 ring atoms, or a bicyclic aromatic group having 8 to 10 atoms, containing at least one heteroatom, O, S or N, in which a carbon or nitrogen atom is the point of attachment, and in which one additional carbon atom is optionally replaced by a heteroatom selected from 0 or S, and in which from 1 to 3 additional carbon atoms are optionally replaced by nitrogen heteroatoms. The heteroaryl group is optionally substituted with up to three groups.

Heteroaryl thus includes aromatic and partially aromatic groups which contain one or more heteroatoms. Examples of this type are thiophene, purine, imidazopyridine, pyridine, oxazole,

thiazole, oxazine, pyrazole, tetrazole, imidazole, pyridine, pyrimidine, pyrazine and triazine. Examples of partially aromatic groups are tetrahydroimidazo [4,5 -cipyridine, phthalidyl and saccharinyl, as defined below.

With regard to the farnesyl transferase inhibitors, the term heterocycle or heterocyclic, as used herein, represents a stable 5- to 7-membered monocyclic or stable 8- to 1 1-membered bicyclic or stable 11-15 membered tricyclic heterocycle ring which is either saturated or unsaturated, and which consists of carbon atoms and from one to four heteroatoms selected from the group consisting of N, 0, and S, and including any bicyclic group in which any of the above-defined heterocyclic rings is fused to a benzene ring. The heterocyclic ring may be attached at any heteroatom or carbon atom which results in the creation of a stable structure. Examples of such heterocyclic elements include, but are not limited to, azepinyl, benzimidazolyl, benzisoxazolyl, benzofurazanyl, benzopyranyl, benzothiopyranyl, benzofuryl, benzothiazolyl, benzothienyl, benzoxazolyl, chromanyl, cinnolinyl, dihydrobenzofuryl, dihydro- benzothienyl, dihydrobenzothiopyranyl, dihydrobenzothio-pyranyl sulfone, furyl, imidazolidinyl, imidazolinyl, imidazolyl, indolinyl, indolyl, isochromanyl, isoindolinyl, isoquinolinyl, isothiazolidinyl, isothiazolyl, isothiazolidinyl, morpholinyl, naphthyridinyl, oxadiazolyl, 2-oxoazepinyl, 2-oxopiperazinyl, 2-oxopiperidinyl, 2-oxopyrrolidinyl, piperidyl, piperazinyl, pyridyl, pyridyl N-oxide, pyridonyl, pyrazinyl, pyrazolidinyl, pyrazolyl, pyrimidinyl, pyrrolidinyl, pyrrolyl, quinazolinyl, quinolinyl, quinolinyl N-oxide, quinoxalinyl, tetrahydrofuryl, tetrahydroisoquinolinyl, tetrahydro- quinolinyl, thiamorpholinyl, thiamorpholinyl sulfoxide, thiazolyl, thiazolinyl, thienofuryl, thienothienyl, and thienyl. Preferably, heterocycle is selected from imidazolyl, 2-oxopyrrolidinyl, piperidyl, pyridyl and pyrrolidinyl.

With regard to the farnesyl transferase inhibitors, the terms "substituted aryl", "substituted heterocycle" and "substituted cycloalkyl" are intended to include the cyclic group which is substituted with 1 or 2 substituents selected from the group which includes but is not limited to F, C1, Br, CF3, NH2, N(C1-C6 alkyl)2, N02, CN, (C1-C6 alkyl)O-, -OH, (C1-C6 alkyl)S(O)m-, (C1-C6 alkyl)C(O)NH-, H2N-C(NH)-, (C1-C6 alkyl)C(O)-, (Cl -C6 alkyl)OC(O)-, N3,(C1-C6 alkyl)OC(O)NH- and C1-C20 alkyl.

In the present method, amino acids which are disclosed are identified both by conventional 3 letter and single letter abbreviations as indicated below: Alanine Ala A Arginine Arg R Asparagine Asn N Aspartic acid Asp D Asparagine or Aspartic acid Asx B Cysteine Cys C Glutamine Gln Q Glutamic acid Glu E Glutamine or Glutamic acid Glx Z Glycine Gly G Histidine His H Isoleucine Ile I Leucine Leu L Lysine Lys K Methionine Met M Phenylalanine Phe F Proline Pro P Serine Ser S Threonine Thr T Tryptophan Trp W Tyrosine Tyr Y Valine Val V

With respect to the term "CAAX" the letter "A" represents an aliphatic amino acid and is not limited to alanine.

The compounds used in the present method may have asymmetric centers and occur as racemates, racemic mixtures, and as individual diastereomers, with all possible isomers, including optical isomers, being included in the present invention. Unless otherwise specified, named amino acids are understood to have the natural "L" stereoconfiguration When R2 and R3 are combined to form - (CH2)u -, cyclic moieties are formed. Examples of such cyclic moieties include, but are not limited to: In addition, such cyclic moieties may optionally include a heteroatom(s). Examples of such heteroatom-containing cyclic moieties include, but are not limited to: When R6 and R7, R7 and R7, or are combined to form - (CH2)u -, cyclic moieties are formed. Examples of such cyclic moieties include, but are not limited to:

The pharmaceutically acceptable salts of the compounds of this invention include the conventional non-toxic salts of the compounds of this invention as formed, e.g., from non-toxic inorganic or organic acids. For example, such conventional non-toxic salts include those derived from inorganic acids such as hydrochloric, hydrobromic, sulfuric, sulfamic, phosphoric, nitric and the like: and the salts prepared from organic acids such as acetic, propionic, succinic, glycolic, stearic, lactic, malic, tartaric, citric, ascorbic, pamoic, maleic, hydroxymaleic, phenyl-acetic, glutamic, benzoic, salicylic, sulfanilic, 2-acetoxy-benzoic, fumaric, toluenesulfonic, methanesulfonic, ethane disulfonic, oxalic, isethionic, trifluoroacetic and the like.

It is intended that the definition of any substituent or variable (e.g., R10, Z, n, etc.) at a particular location in a molecule be independent of its definitions elsewhere in that molecule. Thus, -N(R10)2 represents -NHH, -NHCH3, -NHC2HS, etc. It is under- stood that substituents and substitution patterns on the compounds of the instant invention can be selected by one of ordinary skill in the art to provide compounds that are chemically stable and that can be readily synthesized by techniques known in the art as well as those methods set forth below.

The pharmaceutically acceptable salts of the compounds of this invention can be synthesized from the compounds of this invention which contain a basic moiety by conventional chemical methods. Generally, the salts are prepared by reacting the free base with stoichiometric amounts or with an excess of the desired salt- forming inorganic or organic acid in a suitable solvent or various combinations of solvents.

The compounds used in the methods of the instant invention are useful in various pharmaceutically acceptable salt forms. The term "pharmaceutically acceptable salt" refers to those salt forms which would be apparent to the pharma-ceutical chemist. i.e., those which are substantially non-toxic and which provide the desired pharmacokinetic properties, palatability, absorption, distribution, metabolism or excretion. Other factors, more practical in nature, which are also important in the selection, are cost of the raw materials, ease of crystallization, yield, stability, hygroscopicity and flowability of the resulting bulk drug. Conveniently, pharmaceutical compositions may be prepared from the active ingredients in combination with pharmaceutically acceptable carriers.

Pharmaceutically acceptable salts include conventional non-toxic salts or quarternary ammonium salts formed, e.g., from non-toxic inorganic or organic acids. Non-toxic salts include those derived from inorganic acids such as hydrochloric, hydrobromic, sulfuric, sulfamic, phosphoric, nitric and the like; and the salts prepared from organic acids such as acetic, propionic, succinic, glycolic, stearic, lactic, malic, tartaric, citric, ascorbic, pamoic, sulfanilic, 2-acetoxybenzoic, fumaric, toluenesulfonic, methanesulfonic, ethane disulfonic, oxalic, isethionic, trifluoroacetic and the like.

The pharmaceutically acceptable salts of the present invention can be synthesized by conventional chemical methods.

Generally, the salts are prepared by reacting the free base or acid with stoichiometric amounts or with an excess of the desired salt- forming inorganic or organic acid or base, in a suitable solvent or solvent combination.

Abbreviations used in the description of the chemistry and in the Examples that follow are: Ac2O Acetic anhydride; Boc t-Butoxycarbonyl; DBU 1, ,8-diazabicyclo[S.4.0]undec-7-ene;

DMAP 4-Dimethylaminopyridine ; DME 1,2-Dimethoxyethane; DMF Dimethylformamide; EDC 1 - (3 -dimethylaminopropyl)- 3 -ethyl-carbodiimide- hydrochloride; HOBT 1 -Hydroxybenzotriazole hydrate; Et3N Triethylamine; EtOAc Ethyl acetate; FAB Fast atom bombardment; HOOBT 3-Hydroxy-1,2,2-benzotriazin-4(3H)-one; HPLC High-performance liquid chromatography; MCPBA m-Chloroperoxybenzoic acid; MsCl Methanesulfonyl chloride; NaHMDS Sodium bis(trimethylsilyl)amide; Py Pyridine; TFA Trifluoroacetic acid; THF Tetrahydrofuran.

The farnesyl transferase inhibitors of formula I can be synthesized in accordance with Schemes 1-11, in addition to other standard manipulations such as ester hydrolysis, cleavage of protecting groups, etc., as may be known in the literature or exemplified in the experimental procedures. Substituents R, Ra and Rb, as shown in the Schemes, represent the substituents R2, R3, R4, and R5; however their point of attachment to the ring is illustrative only and is not meant to be limiting.

These reactions may be employed in a linear sequence to provide the compounds of the invention or they may be used to synthesize fragments which are subsequently joined by the alkylation reactions described in the Schemes.

Synopsis of Schemes 1-11: The requisite intermediates are in some cases commercially available, or can be prepared according to literature procedures, for the most part.

Piperazin-5-ones can be prepared as shown in Scheme 1.

Thus, the protected suitably substituted amino acid IV can be converted to the corresponding aldehyde V by first forming the amide and then reducing it with LAH. Reductive amination of Boc-protected amino aldehydes V gives rise to compound VI. The intermediate VI can be converted to a piperazinone by acylation with chloroacetyl chloride to give VII, followed by base-induced cyclization to VIII. Deprotection, followed by reductive alkylation with a protected imidazole carboxalde- hyde leads to IX, which can be alkylated with an arylmethylhalide to give the imidazolium salt X. Final removal of protecting groups by either solvolysis with a lower alkyl alcohol, such as methanol, or treatment with triethylsilane in methylene chloride in the presence of trifluoroacetic acid gives the final product XI.

The intermediate VIII can be reductively alkylated with a variety of aldehydes, such as XII. The aldehydes can be prepared by standard procedures, such as that described by O. P. Goel, U. Krolls, M. Stier and S. Kesten in Organic Svntheses, 1988, 67, 69-75, from the appropriate amino acid (Scheme 2). The reductive alkylation can be accomplished at pH 5-7 with a variety of reducing agents, such as sodium triacetoxyborohydride or sodium cyanoborohydride in a solvent such as dichloroethane, methanol or dimethylformamide. The product XIII can be deprotected to give the final compounds XIV with trifluoro- acetic acid in methylene chloride. The final product XIV is isolated in the salt form, for example, as a trifluoroacetate, hydrochloride or acetate salt, among others. The product diamine XIV can further be selectively protected to obtain XV, which can subsequently be reductively alkylated with a second aldehyde to obtain XVI. Removal of the protecting group, and conversion to cyclized products such as the dihydroimidazole XVII can be accomplished by literature procedures.

Alternatively, the imidazole acetic acid XVIII can be converted to the acetate XIX by standard procedures, and XIX can be first reacted with an alkyl halide, then treated with refluxing methanol to provide the regiospecifically alkylated imidazole acetic acid ester XX

(Scheme 3). Hydrolysis and reaction with piperazinone VIII in the presence of condensing reagents such as l-(3-dimethylaminopropyl)- 3-ethylcarbodiimide (EDC) leads to acylated products such as XXI.

If the piperazinone VIII is reductively alkylated with an aldehyde which also has a protected hydroxyl group, such as XXII in Scheme 4, the protecting groups can be subsequently removed to unmask the hydroxyl group (Schemes 4, 5). The alcohol can be oxidized under standard conditions to e.g. an aldehyde, which can then be reacted with a variety of organometallic reagents such as Grignard reagents, to obtain secondary alcohols such as XXIV. In addition, the fully deprotected amino alcohol XXV can be reductively alkylated (under conditions described previously) with a variety of aldehydes to obtain secondary amines, such as XXVI (Scheme 5), or tertiary amines.

The Boc protected amino alcohol XXIII can also be utilized to synthesize 2-aziridinylmethylpiperazinones such as XXVII (Scheme 6). Treating XXIII with 1,1 '-sulfonyldiimidazole and sodium hydride in a solvent such as dimethylformamide led to the formation of aziridine XXVII. The aziridine reacted in the presence of a nucleophile, such as a thiol, in the presence of base to yield the ring-opened product XXVIII.

In addition, the piperazinone VIII can be reacted with aldehydes derived from amino acids such as O-alkylated tyrosines, according to standard procedures, to obtain compounds such as XXX (Scheme 7). When R' is an aryl group, XXX can first be hydrogenated to unmask the phenol, and the amine group deprotected with acid to produce XXXI. Alternatively, the amine protecting group in XXX can be removed, and O-alkylated phenolic amines such as XXXII produced.

Scheme 8 illustrates the use of an optionally substituted homoserine lactone XXXIII to prepare a Boc-protected piperazinone XXXVII. Intermediate XXXVII may be deprotected and reductively alkylated or acylated as illustrated in the previous Schemes.

Alternatively, the hydroxyl moiety of intermediate XXXVII may be mesylated and displaced by a suitable nucleophile, such as the sodium salt of ethane thiol, to provide an intermediate XXXVIII. Intermediate

XXXVII may also be oxidized to provide the carboxylic acid on intermediate IXL, which can be utilized form an ester or amide moiety.

N-Aralkyl-piperazin-5-ones can be prepared as shown in Scheme 9. Reductive amination of Boc-protected amino aldehydes V (prepared from III as described previously) gives rise to compound XL.

This is then reacted with bromoacetyl bromide under Schotten-Baumann conditions; ring closure is effected with a base such as sodium hydride in a polar aprotic solvent such as dimethylformamide to give XLI. The carbamate protecting group is removed under acidic conditions such as trifluoroacetic acid in methylene chloride, or hydrogen chloride gas in methanol or ethyl acetate, and the resulting piperazine can then be carried on to final products as described in Schemes 1-7.

The isomeric piperazin-3-ones can be prepared as described in Scheme 10. The imine formed from arylcarboxamides XLII and 2-aminoglycinal diethyl acetal (XLIII) can be reduced under a variety of conditions, including sodium triacetoxyborohydride in dichloroethane, to give the amine XLIV. Amino acids I can be coupled to amines XLIV under standard conditions, and the resulting amide XLV when treated with aqueous acid in tetrahydrofuran can cyclize to the unsaturated XLVI. Catalytic hydrogenation under standard conditions gives the requisite intermediate XLVII, which is elaborated to final products as described in Schemes 1-7.

Amino acids of the general formula IL which have a sidechain not found in natural amino acids may be prepared by the reactions illustrated in Scheme 11 starting with the readily prepared imine XLVIII. SCHEME 1 SCHEME 1 (continued) SCHEME 2 SCHEME 2 (continued) SCHEME 3 CH2C02CH3 1) ArCH2X CH3CN (C 8H 5)3C B r vWe 5) 39 retlux (C2H5)3N . 2)CH3OH,reflux DMF Tr XIX SCHEME 3 (continued) SCHEME 4 SCHEME 5 SCHEME 6 SCHEME 7 R'CH20 pyridine SO3 R'CH20Xo DMSO BocNH CH2OH 20°C BocNH CH0 200C

SCHEME 7 (continued) SCHEME 8 SCHEME 8 (continued) HO sub vi M BocN N-Ar o 1 (COCI)2, Et3N DMSO 1. MsGI,iPr2NEt 2. 2.NaCIO2,t-BuOH /\ 2. NaCI02, t-BuOH 2. NaSEt, DMF 2-Me-2-butene EtS sub HO sub ,X'7 M BocN N-Ar BocN N-Ar H0 80 XXXVIII IXL

SCHEME 9 SCHEME 10

SCHEME 11 1. KOtBu, THF R2 r CO2Et R2X » CO2Et g N H2N Ph 2. 5% aqueous HCI HCI XLVIII Reactions used to generate the compounds of the formula (II) are prepared by employing reactions as shown in the Schemes 16- 37, in addition to other standard manipulations such as ester hydrolysis, cleavage of protecting groups, etc., as may be known in the literature or exemplified in the experimental procedures. Substituents Ra and Rb, as shown in the Schemes, represent the substituents R2, R3, R4, and R5; substituent "sub" represents a suitable substituent on the substituent Z.

The point of attachment of such substituents to a ring is illustrative only and is not meant to be limiting.

These reactions may be employed in a linear sequence to provide the compounds of the invention or they may be used to synthesize fragments which are subsequently joined by the alkylation reactions described in the Schemes.

Synopsis of Schemes 16-37: The requisite intermediates utilized as starting material in the Schemes hereinbelow are in some cases commercially available, or can be prepared according to literature procedures. In Scheme 16, for example, a suitably substituted Boc protected isonipecotate LI may be

deprotonated and then treated with a suitably substituted alkylating group, such as a suitably substituted benzyl bromide, to provide the gem disubstituted intermediate LIII. Deprotection and reduction provides the hydroxymethyl piperidine LIV which can be utilized is synthesis of compounds of the invention or which may be nitrogen- protected and methylated to give the intermediate LV.

As shown in Scheme 17, the protected piperidine intermediate LIII can be deprotected and reductively alkylated with aldehydes such as l-trityl-4-imidazolyl-carboxaldehyde or l-trityl- 4-imidazolylacetaldehyde, to give products such as LVI. The trityl protecting group can be removed from LVI to give LVII, or alternatively, LVI can first be treated with an alkyl halide then subsequently deprotected to give the alkylated imidazole LVIII.

The deprotected intermediate LIII can also be reductively alkylated with a variety of other aldehydes and acids as shown above in Schemes 4-7.

An alternative synthesis of the hydroxymethyl intermediate LIV and utilization of that intermediate in the synthesis of the instant compounds which incorporate the preferred imidazolyl moiety is illustrated in Scheme 18. Scheme 19 illustrates the reductive alkylation of intermediate LIV to provide a 4-cyanobenzylimidazolyl substituted piperidine. The cyano moiety may be selectively hydrolyzed with sodium borate to provide the corresponding amido compound of the instant invention.

Scheme 20 alternative preparation of the methyl ether intermediate LV and the alkylation of LV with a suitably substituted imidazolylmethyl chloride to provide the instant compound. Prepara- tion of the homologous l-(imidazolylethyl)piperidine is is illustrated in Scheme 21.

Specific substitution on the piperidine of the compounds of the instant invention may be accomplished as illustrated in Scheme 22. Thus, metal-halogen exchange coupling of a butynyl moiety to an isonicotinate, followed by hydrogenation, provides the 2-butylpiperidine

intermediate that can then undergo the reactions previously described to provide the compound of the instant invention.

Incorporation of a 4-amido moiety for LV is illustrated in Scheme 23.

Scheme 24 illustrates the synthesis of the instant compounds wherein the moiety Z is attached directly to the piperidine ring. Thus the piperidone LIX is treated with a suitably substituted phenyl Grignard reagent to provide the gem disubstituted piperidine LX. Deprotection provides the key intermediate LXI. Intermediate LXI may be acetylated as described above to provide the instant compound LXII (Scheme 25).

As illustrated in Scheme 26, the protected piperidine LX may be dehydrated and then hydroborated to provide the 3- hydroxypiperidine LXIII. This compound may be deprotected and further derivatized to provide compounds of the instant invention (as shown in Scheme 27) or the hydroxyl group may be alkylated, as shown in Scheme 26, prior to deprotection and further manipulation.

The dehydration product may also be catalytically reduced to provide the des-hydroxy intermediate LXV, as shown in Scheme 28, which can be processed via the reactions illustrated in the previous Schemes.

Schemes 29 and 30 illustrate further chemical manipula- tions of the 4-carboxylic acid functionality to provide instant compounds wherein the substituent Y is an acetylamine or sulfonamide moiety.

Scheme 31 illustrates incorporation of a nitrile moiety in the 4-position of the piperidine of the compounds of formula II. Thus, the hydroxyl moiety of a suitably substituted 4-hydroxypiperidine is substituted with nitrile to provide intermediate LXVI, which can undergo reactions previously described in Schemes 17-21.

Scheme 32 illustrates the preparation of several pyridyl intermediates that may be utilized with the piperidine intermediates such as compound LI in Scheme 16 to provide the instant compounds.

Scheme 33 shows a generalized reaction sequence which utilizes such pyridyl intermediates.

Compounds of the instant invention wherein X1 is a carbonyl moiety may be prepared as shown in Scheme 34. Inter- mediate LXVII may undergo subsequent reactions as illustrated in Schemes 17-21 to provide the instant compounds. Preparation of the instant compounds wherein X1 is sulfur in its various oxidation states is shown in Scheme 35. Intermediates LXVIII-LXXI may undergo the previously described reactions to provide the instant compounds.

Scheme 36 illustrated preparation of compounds of the formula A wherein Y is hydrogen. Thus, suitably substituted isonipecotic acid may be treated with N,O-dimethylhydroxylamine and the intermediate LXXII reacted with a suitably substituted phenyl Grignard reagent to provide intermediate LXXIII. That intermediate may undergo the reactions previously described in Schemes 17-21 and may be further modified by reduction of the phenyl ketone to provide the alcohol LXXIV.

Compounds of the instant invention wherein X1 is an amine moiety may be prepared as shown in Scheme 37. Thus the N-protected 4-piperidinone may be reacted with a suitably substituted aniline in the presence of trimethylsilylcyanide to provide the 4-cyano- 4-aminopiperidine LXXV. Intermediate LXXV may then be converted in sequence to the corresponding amide LXXVI, ester LXXVII and alcohol LXXVIII. Intermediates LXXVI-LXXVIII can be deprotected and can then undergo the reactions previously described in Schemes 17-21 to provide the compounds of the instant invention.

SCHEME 16 SCHEME 17 SCHEME 18 SCHEME 19 SCHEME 20 SCHEME 21 SCHEME 22 SCHEME 23

SCHEME 24 SCHEME 25 SCHEME 26 0 1. POCI3, pyridine 0 2. BH3, H202 NaOH sub LX SCHEME 27 SCHEME 28 0 OH 1. POCI3, pyridlne 9 2. >> 2. H2, Pd/C LX \ç~ sub O # N sub HCl # O # # LXV <BR> <BR> Ar# CH2C<BR> <BR> <BR> <BR> N#<BR> <BR> # # sub #<BR> <BR> <BR> <BR> <BR> H-N#-# N EDC'HCI HOBt DMF <BR> <BR> <BR> <BR> <BR> <BR> Ar#<BR> <BR> <BR> # O # # sub<BR> <BR> <BR> <BR> <BR> N # -N - # SCHEME 29 (Boc)20 Ra ° (Boc) 20 N [OH Aq NaOH \ HN\ Rb 0 0 1. Na HMDS Benzyl alcohol Ra benzyl 1. Na HMDS EDC, DMAP Ob, N \ < 0 N 0 Rb a, 0 0 Pd on C RM½ R on \ sub O Rb a HO ONy\ O N \ sub 0 Rb SCHEME 29 (continued) SCHEME 30 SCHEME 31 SCHEME 32 SCHEME 32 (continued) SCHEME 33 SCHEME 34 SCHEME 35 NaHMDS Lyo N&'WlaCO2R - R = CH3, CH3CH2 1 1} sub Ra R1 CO2R N LiAIH4 sub Ra S NX OH KH, CH31 NOH KH, CH3l LXVIII sub Ra °S X NalO4 LXIX ¼ sub Ra ° rl) FOCH3 > N) t m Oxone t° sA l} LXX sub Ra Q rl- OCH, y½ ½m½) LXXI sub SCHEME 36 SCHEME 37

Compounds of this invention of formula (III) are prepared by employing the reactions shown in the following Reaction Schemes 38-51, in addition to other standard manipulations such as ester hydrolysis, cleavage of protecting groups, etc., as may be known in the literature or exemplified in the experimental procedures. Some key bond-forming and peptide modifying reactions are: Reaction A Amide bond formation and protecting group cleavage using standard solution or solid phase methodologies.

Reaction B Preparation of a reduced peptide subunit by reductive alkylation of an amine by an aldehyde using sodium cyanoborohydride or other reducing agents.

Reaction C Alkylation of a reduced peptide subunit with an alkyl or aralkyl halide or, alternatively, reductive alkylation of a reduced peptide subunit with an aldehyde using sodium cyanoborohydride or other reducing agents.

Reaction D Peptide bond formation and protecting group cleavage using standard solution or solid phase methodologies.

Reaction E Preparation of a reduced subunit by borane reduction of the amide moiety.

These reactions may be employed in a linear sequence to provide the compounds of the invention or they may be used to synthesize fragments which are subsequently joined by the alkylation reactions described in the Reaction Schemes and in Reaction Schemes 43-51 hereinbelow.

REACTION SCHEME 38 Reaction A. Coupling of residues to form an amide bond REACTION SCHEME 39 Reaction B. Preparation of reduced peptide subunits by reductive alkylation

REACTION SCHEME 40 Reaction C. Alkvlation/reductive alkylation of reduced peptide subunits O RAHO R7XL, base 0 NN J4OR or H OR RB O II RICH, NaCNBH3 REACTION SCHEME 41 Reaction D. Coupling of residues to form an amide bond

REACTION SCHEME 42 Reaction E. Preparation of reduced dipeptides from peptides where RA and RB are R2, R3 or R5 as previously defined; RC and RD are R7 or R12 ; XL is a leaving group, e.g., Br-, I- or MsO-; and Ry is defined such that R7 is generated by the reductive alkylation process.

In addition to the reactions described in Reaction Schemes 26-30, other reactions used to generate the compounds of formula (III) of this invention are shown in the Reaction Schemes 43-51. All of the substituents shown in the Reaction Schemes, represent the same substi- tuents as defined hereinabove. The substituent "Ar" in the Reaction Schemes represents a carbocyclic or heterocyclic, substituted or unsubstituted aromatic ring.

These reactions may be employed in a linear sequence to provide the compounds of the invention or they may be used to synthesize fragments which are subsequently joined by the alkylation reactions described in the Reaction Schemes. The sequential order whereby substituents are incorporated into the compounds is often not critical and thus the order of reactions described in the Reaction Schemes are illustrative only and are not limiting.

Synopsis of Reaction Schemes 43-51: The requisite intermediates are in some cases commercially available, or can be readily prepared according to known literature

procedures, including those described in Reaction Schemes 38-42 hereinabove.

Reaction Scheme 43 illustrates incorporation of the cyclic amine moiety, such as a reduced prolyl moiety, into the compounds of the formula III of the instant invention. Reduction of the azide LXXXI provides the amine LXXXII, which may be mono- or di-substituted using techniques described above. As an example, incorporation of a naphthylmethyl group and an acetyl group is illustrated.

As shown in Reaction Scheme 44, direct attachment of a aromatic ring to a substituted amine such as LXXXIII is accomplished by coupling with a triarylbismuth reagent, such as tris(3-chlorophenyl) bismuth.

Reaction Scheme 45 illustrates the use of protecting groups to prepare compounds of the instant invention wherein the cyclic amine contains an alkoxy moiety. The hydroxy moiety of key intermediate LXXXIVa may be further converted to a fluoro or phenoxy moiety, as shown in Reaction Scheme 46. Intermediates LXXXV and LXXXVI may then be further elaborated to provide the instant compounds.

Reaction Scheme 474 illustrates syntheses of instant compounds wherein the variable -(CR42)qA3(CR52)nR6 is a suitably substituted a-hydroxybenzyl moiety. Thus the protected intermediate aldehyde is treated with a suitably substituted phenyl Grignard reagent to provide the enantiomeric mixture LXXXVII. Treatment of the mixture with 2-picolinyl chloride allows chromatographic resolution of compounds LXXXVIII and IXC. Removal of the picolinoyl group followed by deprotection provides the optically pure intermediate XC which can be further processed as described hereinabove to yield the instant compounds.

Syntheses of imidazole-containing intermediates useful in synthesis of instant compounds wherein the variable p is 0 or 1 and Z is H2 are shown in Reaction Scheme 48 and 49. Thus the mesylate XCI can be utilized to alkylate a suitably substituted amine or cyclic amine, while aldehyde XCII can be used to similarly reductively alkylate such an amine.

Reaction Scheme 50 illustrates the syntheses of imidazole-containing intermediates wherein the attachment point of the -(CR22)p-C(Z)- moiety to W (imidazolyl) is through an imidazole ring nitrogen. Reaction Scheme 51 illustrates the synthesis of an intermediate wherein an R2 substituent is a methyl.

REACTION SCHEME 43

REACTION SCHEME 43 (continued) REACTION SCHEME 44 Dy0yN 0 Cu(OAc)2 t gt NEt3, Bi (aryl) 3 N CH3 H REACTION SCHEME 45 REACTION SCHEME 45 (continued) < OR 0 I. HCl NaH, RX HCI -- 0 O II.EDC Coupling or N'K reductive amination or alkylation R' REACTION SCHEME 46 OH F DAST 0N 0 N o o 0y LXXXIII LXXXIV LXXXI I I J, W R R t eN nol t eNX Ph,, DEAD, e )rN 0 LXXXI II R LXXXV X LXXXIII R R REACTION SCHEME 47 OTBS 2-Picolinoyl Chloride N NEt3 OHO R LXXXVII REACTION SCHEME 47 (continued) OH Coupling/alkylation N etc HO R CN REACTION SCHEME 48 REACTION SCHEME 49 CH2OAc 1) ArCH2X CH3CN NC reflux, Ac20'2 2) CH30H, reflux pyridine I Tr REACTION SCHEME 50

REACTION SCHEME 51 The prenyl transferase inhibitors of formula (A) can be synthesized in accordance with Reaction Scheme below, in addition to other standard manipulations such as ester hydrolysis, cleavage of protecting groups, etc., as may be known in the literature or exemplified in the experimental procedures. Some key reactions utilized to form the aminodiphenyl moiety of the instant compounds are shown.

The reactions may be employed in a linear sequence to provide the compounds of the invention or they may be used to synthesize fragments which are subsequently joined by the alkylation reactions described in the Reaction Scheme.

A method of forming the benzophenone intermediates, illustrated in Reaction Scheme 52, is a Stille reaction with an aryl stannane. Such amine intermediates may then be reacted as illustrated hereinabove with a variety of aldehydes and esters/acids.

REACTION SCHEME 52

EXAMPLES Examples provided are intended to assist in a further understanding of the invention. Particular materials employed, species and conditions are intended to be further illustrative of the invention and not limitative of the reasonable scope thereof.

The standard workup referred to in the examples refers to solvent extraction and washing the organic solution with 10% citric acid, 10% sodium bicarbonate and brine as appropriate. Solutions were dried over sodium sulfate and evaporated in vacuo on a rotary evaporator.

EXAMPLES 1 1-(3-Chlorophenyl)-4-[1-(4-cyanobenzyl)imidazolylmethyl]-2- piperazinone dihvdrochloride (Compound 1) Step A: Preparation of 1 -triphenylmethyl-4-(hydroxymethyl)- imidazole To a solution of 4-(hydroxymethyl)imidazole hydrochloride (35.0 g, 260 mmol) in 250 mL of dry DMF at room temperature was added triethylamine (90.6 mL, 650 mmol). A white solid precipitated from the solution. Chlorotriphenylmethane (76.1 g, 273 mmol) in 500 mL of DMF was added dropwise. The reaction mixture was stirred for 20 hours, poured over ice, filtered, and washed with ice water. The resulting product was slurried with cold dioxane, filtered, and dried in vacuo to provide the titled product as a white solid which was sufficiently pure for use in the next step.

Step B: Preparation of 1-triphenylmethyl-4-(acetoxymethyl)- imidazole Alcohol from Step A (260 mmol, prepared above) was suspended in 500 mL of pyridine. Acetic anhydride (74 mL, 780 mmol) was added dropwise, and the reaction was stirred for

48 hours during which it became homogeneous. The solution was poured into 2 L of EtOAc, washed with water (3 x 1 L), 5% aq. HC1 soln. (2 x 1 L), sat. aq. NaHCO3, and brine, then dried (Na2SO4), filtered, and concentrated in vacuo to provide the crude product.

The acetate was isolated as a white powder which was sufficiently pure for use in the next reaction.

Step C: Preparation of 1-(4-cyanobenzyl)-5-(acetoxymethyl)- imidazole hvdrobromide A solution of the product from Step B (85.8 g, 225 mmol) and a-bromo-p-tolunitrile (50.1 g, 232 mmol) in 500 mL of EtOAc was stirred at 600C for 20 hours, during which a pale yellow precipitate formed. The reaction was cooled to room temperature and filtered to provide the solid imidazolium bromide salt. The filtrate was concentrated in vacuo to a volume 200 mL, reheated at 60"C for two hours, cooled to room temperature, and filtered again.

The filtrate was concentrated in vacuo to a volume 100 mL, reheated at 60"C for another two hours, cooled to room temperature, and concentrated in vacuo to provide a pale yellow solid. All of the solid material was combined, dissolved in 500 mL of methanol, and warmed to 600C. After two hours, the solution was reconcentrated in vacuo to provide a white solid which was triturated with hexane to remove soluble materials. Removal of residual solvents in vacuo provided the titled product hydrobromide as a white solid which was used in the next step without further purification.

Step D: Preparation of 1-(4-cyanobenzyl)-5-(hydroxymethyl)- imidazole To a solution of the acetate from Step C (50.4 g, 150 mmol) in 1.5 L of 3:1 THF/water at OOC was added lithium hydroxide monohydrate (18.9 g, 450 mmol). After one hour, the reaction was concentrated in vacuo, diluted with EtOAc (3 L), and washed with water, sat. aq. NaHCO3 and brine. The solution was then dried (Na2SO4), filtered, and concentrated in vacuo to provide

the crude product as a pale yellow fluffy solid which was sufficiently pure for use in the next step without further purification.

Step E: Preparation of 1-(4-cyanobenzyl)-5- imidazolecarboxaldehvde To a solution of the alcohol from Step D (21.5 g, 101 mmol) in 500 mL of DMSO at room temperature was added triethylamine (56 mL, 402 mmol), then SO3-pyridine complex (40.5 g, 254 mmol). After 45 minutes, the reaction was poured into 2.5 L of EtOAc, washed with water (4 x 1 L) and brine, dried (Na2SO4), filtered, and concentrated in vacuo to provide the aldehyde as a white powder which was sufficiently pure for use in the next step without further purification.

Step F: Preparation of N-(3-chlorophenyl)ethylenediamine hvdrochloride To a solution of 3-chloroaniline (30.0 mL, 284 mmol) in 500 mL of dichloromethane at OOC was added dropwise a solution of 4 N HC1 in 1,4-dioxane (80 mL, 320 mmol HC1). The solution was warmed to room temperature, then concentrated to dryness in vacuo to provide a white powder. A mixture of this powder with 2-oxazolidinone (24.6 g, 282 mmol) was heated under nitrogen atmosphere at 1600C for 10 hours, during which the solids melted, and gas evolution was observed. The reaction was allowed to cool, forming the crude diamine hydrochloride salt as a pale brown solid.

Step G: Preparation of N-(tert-butoxycarbonyl)-N'-(3- chlorophenyl)ethylenediamine The amine hydrochloride from Step F (ca. 282 mmol, crude material prepared above) was taken up in 500 mL of THF and 500 mL of sat. aq. NaHCO3 soln., cooled to OOC, and di-tert- butylpyrocarbonate (61.6 g, 282 mmol) was added. After 30 h, the reaction was poured into EtOAc, washed with water and brine, dried (Na2S04), filtered, and concentrated in vacuo to provide the titled

carbamate as a brown oil which was used in the next step without further purification.

Step H: Preparation of N-[2-(tert-butoxycarbamoyl)ethyl]-N-(3- chlorophenyl)-2-chloroacetamide A solution of the product from Step G (77 g, ca. 282 mmol) and triethylamine (67 mL, 480 mmol) in 500 mL of CH2C12 was cooled to OOC. Chloroacetyl chloride (25.5 mL, 320 mmol) was added dropwise, and the reaction was maintained at OOC with stirring. After 3 h, another portion of chloroacetyl chloride (3.0 mL) was added dropwise. After 30 min, the reaction was poured into EtOAc (2 L) and washed with water, sat. aq. NH4C1 soln, sat. aq. NaHCO3 soln., and brine. The solution was dried (Na2SO4), filtered, and concentrated in vacuo to provide the chloroacetamide as a brown oil which was used in the next step without further purification.

Step I: Preparation of 4-(tert-butoxycarbonyl)-1-(3- chlorophenyl)-2-piperazinone To a solution of the chloroacetamide from Step H (ca.

282 mmol) in 700 mL of dry DMF was added K2CO3 (88 g, 0.64 mol). The solution was heated in an oil bath at 70-750C for 20 hrs., cooled to room temperature, and concentrated in vacuo to remove ca. 500 mL of DMF. The remaining material was poured into 33% EtOAc/hexane, washed with water and brine, dried (Na2SO4), filtered, and concentrated in vacuo to provide the product as a brown oil. This material was purified by silica gel chromatography (25- 50% EtOAc/hexane) to yield pure product, along with a sample of product (ca. 65% pure by HPLC) containing a less polar impurity.

Step J: Preparation of l-(3-chloroDhenvl)-2-iDerazinone Through a solution of Boc-protected piperazinone from Step 1(17.19 g, 55.4 mmol) in 500 mL of EtOAc at -780C was bubbled anhydrous HC1 gas. The saturated solution was warmed to

0°C, and stirred for 12 hours. Nitrogen gas was bubbled through the reaction to remove excess HC1, and the mixture was warmed to room temperature. The solution was concentrated in vacuo to provide the hydrochloride as a white powder. This material was taken up in 300 mL of CH2C12 and treated with dilute aqueous NaHCO3 solution.

The aqueous phase was extracted with CH2C12 (8 x 300 mL) until tlc analysis indicated complete extraction. The combined organic mixture was dried (Na2SO4), filtered, and concentrated in vacuo to provide the titled free amine as a pale brown oil.

Step K: Preparation of 1-(3-chlorophenyl)-4-[1-(4- cyanob enzyl)imi daz olylmethyll -2-piperazinone dihvdrochloride To a solution of the amine from Step J (55.4 mmol, prepared above) in 200 mL of 1,2-dichloroethane at 0°C was added 4Å powdered molecular sieves (10 g), followed by sodium triacetoxy- borohydride (17.7 g, 83.3 mmol). The imidazole carboxaldehyde from Step E of Example 1(11.9 g, 56.4 mmol) was added, and the reaction was stirred at 00C. After 26 hours, the reaction was poured into EtOAc, washed with dilute aq. NaHCO3, and the aqueous layer was back-extracted with EtOAc. The combined organics were washed with brine, dried (Na2SO4), filtered, and concentrated in vacuo.

The resulting product was taken up in 500 mL of 5:1 benzene:CH2C12, and propylamine (20 mL) was added. The mixture was stirred for 12 hours, then concentrated in vacuo to afford a pale yellow foam.

This material was purified by silica gel chromatography (2-7% MeOH/CH2C12), and the resultant white foam was taken up in CH2C12 and treated with 2.1 equivalents of 1 M HCVether solution. After concentrated in vacuo, the product dihydrochloride was isolated as a white powder.

Examples 2-5 (Table 1) were prepared using the above protocol, which describes the synthesis of the structurally related compound

1- (3-chlorophenyl)-4- [1- (4-cyanobenzyl)-imidazolylmethyl]-2- piperazinone dihydrochloride. In Step F, the appropriately substituted aniline was used in place of 3-chloroaniline.

Table 1: 1-Aryl-4-[1-(4-cyanobenzyl)imidazolylmethyl]-2- piperazinones

FAB mass spectrum CHN Example X (M+1) Analvsis 2 3-OCF3 456 C23H20 F3N5O2#2.0HCl#0.60H2O calcd; C, 51.24; H, 4.34; N, 12.99. found; C, 51.31; H, 4.33; N, 12.94.

3 2,5-(CH3)2 400 C24H25N5O#2.00HCl#0.65H2O calcd; C, 59.54; H, 5.89; N, 14.47 found; C, 59.54; H, 5.95; N, 14. 12.

4 3-CH3 386 C23H23N5O#2.0HCl#0.80H2O calcd; C, 58.43; H, 5.67; N, 14.81. found; C, 58.67; H, 6.00; N, 14.23.

5 3-I 498 C22H20N5OI#2.25HCl#0.90H2O calcd; C, 44.36; H, 4.07; N, 11.76. found; C, 44.37; H, 4.06; N, 11. 42.

EXAMPLE 6 1 - (3-chlorophenyl)-4- [1 -(4-cyano-3-methoxybenzyl)imidazolylmeth 2-piperazinone dihvdrochloride Step A: Preparation of Methvl 4-Amino-3-hvdroxvbenzoate Through a solution of 4-amino-3-hydroxybenzoic acid (75 g, 0.49 mol) in 2.0 L of dry methanol at room temperature was bubbled anhydrous HC1 gas until the solution was saturated. The solution was stirred for 48 hours, then concentrated in vacuo. The product was partitioned between EtOAc and saturated aq. NaHCO3 solution, and the organic layer was washed with brine, dried (Na2SO4), and concentrated in vacuo to provide the titled compound (79 g, 96% yield).

Step B: Preparation of Methyl 3-Hydroxy-4-iodobenzoate A cloudy, dark solution of the product from Step A (79 g, 0.47 mol), 3N HC1 (750 mL), and THF (250 mL) was cooled to OOC. A solution of NaNO2 (35.9 g, 0.52 mol) in 115 mL of water was added over ca. 5 minutes, and the solution was stirred for another 25 minutes. A solution of potassium iodide (312 g, 1.88 mol) in 235 mL of water was added all at once, and the reaction was stirred for an additional 15 minutes. The mixture was poured into EtOAc, shaken, and the layers were separated. The organic phase was washed with water and brine, dried (Na2SO4), and concentrated in vacuo to provide the crude product (148 g). Purification by column chromatography through silica gel (0%-50% EtOAc/hexane) provided the titled product (96 g, 73% yield).

Step C: Preparation of Methvl 4-Cyano-3-hydroxybenzoate A mixture of the iodide product from Step B (101 g, 0.36 mol) and zinc(II)cyanide (30 g, 0.25 mol) in 400 mL of dry DMF was degassed by bubbling argon through the solution for 20 minutes. Tetrakis(triphenylphosphine)palladium (8.5 g, 7.2 mmol) was added, and the solution was heated to 800C for 4 hours. The

solution was cooled to room temperature, then stirred for an additional 36 hours. The reaction was poured into EtOAc/water, and the organic layer was washed with brine (4x), dried (Na2SO4), and concentrated in vacuo to provide the crude product. Purification by column chromatography through silica gel (30%-50% EtOAc/ hexane) provided the titled product (48.8 g, 76% yield).

Step D: Preparation of Methvl 4-Cyano-3-methoxvbenzoate Sodium hydride (9 g, 0.24 mol as 60% wt. disp. mineral oil) was added to a solution of the phenol from Step C (36.1 g, 204 mmol) in 400 mL of dry DMF at room temperature. Iodomethane was added (14 mL. 0.22 mol) was added, and the reaction was stirred for 2 hours. The mixture was poured into EtOAc/water, and the organic layer was washed with water and brine (4x), dried (Na2SO4), and concentrated in vacuo to provide the titled product (37.6 g, 96% yield).

Step E: Preparation of 4-Cvano-3 -methoxybenzyl Alcohol To a solution of the ester from Step D (48.8 g, 255 mmol) in 400 mL of dry THF under argon at room temperature was added lithium borohydride (255 mL, 510 mmol, 2M THF) over 5 minutes. After 1.5 hours, the reaction was warmed to reflux for 0.5 hours, then cooled to room temperature. The solution was poured into EtOAc/lN HC1 soln. [CAUTION], and the layers were separated. The organic layer was washed with water, sat Na2CO3 soln. and brine (4x), dried (Na2SO4), and concentrated in vacuo to provide the titled product (36.3 g, 87% yield).

Step F: Preparation of 4-Cvano-3-methoxybenzyl Bromide A solution of the alcohol from Step E (35.5 g, 218 mmol) in 500 mL of dry THF was cooled to 0°C.

Triphenylphosphine was added (85.7 g, 327 mmol), followed by carbontetrabromide (108.5 g, 327 mmol). The reaction was stirred at 0°C for 30 minutes, then at room temperature for 21 hours. Silica

gel was added (ca. 300 g), and the suspension was concentrated in vacuo. The resulting solid was loaded onto a silica gel chromato- graphy column. Purification by flash chromatography (30%-50% EtOAc/hexane) provided the titled product (42 g, 85% yield).

Step G: Preparation of 1 - (4-cyano-3 -methoxybenzyl)-5 - (acetoxymethyl)-imidazole hydrobromide The titled product was prepared by reacting the bromide from Step F (21.7 g, 96 mmol) with the imidazole product from Step B of Example 1(34.9 g, 91 mmol) using the procedure outlined in Step C of Example 1. The crude product was triturated with hexane to provide the titled product hydrobromide (19.43 g, 88% yield).

Step H: Preparation of 1 - (4-cyano-3 -methoxybenzyl)-5 - (hydroxvmethyl)-imidazole The titled product was prepared by hydrolysis of the acetate from Step G (19.43 g, 68.1 mmol) using the procedure outlined in Step D of Example 1. The crude titled product was isolated in modest yield (11 g, 66% yield). Concentration of the aqueous extracts provided solid material (ca. 100 g) which contained a significant quantity of the titled product, as judged by'H NMR spectroscopy.

Step I: Preparation of 1-(4-cyano-3-methoxybenzyl)-5- imidazolecarboxaldehvde The titled product was prepared by oxidizing the alcohol from Step H (11 g, 45 mmol) using the procedure outlined in Step E of Example 1. The titled aldehyde was isolated as a white powder (7.4 g, 68% yield) which was sufficiently pure for use in the next step without further purification.

Step J: Preparation of 1- (3 -chlorophenyl)-4- [1 -(4-cyano-3 - methoxybenzyl)imidazolylmethyl]-2-piperazinone dihydrochloride

The titled product was prepared by reductive alkylation of the aldehyde from Step I (859 mg, 3.56 mmol) and the amine (hydrochloride) from Step K of Example 1 (800 mg, 3.24 mmol) using the procedure outlined in Step H of Example 1. Purification by flash column chromatography through silica gel (50%-75% acetone CH2C12) and conversion of the resulting white foam to its dihydrochloride salt provided the titled product as a white powder (743 mg, 45% yield). FAB ms (m+l) 437.

Anal. Calc. for C23H23CIN5O2#2.0HCl#0.35CH2Cl2: C, 51.97; H, 4.80; N, 12.98.

Found: C, 52. 11 ; H, 4.80; N, 12.21.

EXAMPLE 7 1-(3-trifluoromethoxyphenyl)-4-[1-(4-cyano-3- methoxvbenzvl)imidazolvl methyll-2-piperazinone dihydrochloride 1-(3-trifluoromethoxy-phenyl)-2-piperazinone hydro- chloride was prepared from 3-trifluoromethoxyaniline using Steps F-J of Example 1. This amine (1.75 g, 5.93 mmol) was coupled to the aldehyde from Step I of Example 6 (1.57 g, 6.52 mmol) using the procedure outlined in Step H of Example 1. Purification by flash column chromatography through silica gel (60%-100% acetone CH2C12) and conversion of the resulting white foam to its dihydrochloride salt provided the titled product as a white powder (1.947 g, 59% yield). FAB ms (m+1) 486.

Anal. Calc. for C24H23F3N5O3#2.0HCl#0.60H2O: C, 50.64; H, 4.46; N, 12.30.

Found: C, 50.69; H, 4.52; N, 12.13.

EXAMPLE 8 <BR> <BR> <BR> <BR> <BR> <BR> 4- [((1 -(4-cyanobenzyl)-5-imidazolyl)methyl)ami hydrochloride The titled product was prepared by reductive alkylation

of the aldehyde from Step E of Example 1 (124 mg, 0.588 mmol) and 4-aminobenzophenone (116 mg, 0.588 mmol) using the procedure outlined in Step K of Example 1. Purification by flash column chromatography through silica gel (2-6% MeOH/CH2Cl2) and conversion to the hydrochloride salt provided the titled product as a white solid (126 mg, 50% yield). FAB ms (m+1) 393.11.

Anal. Calc. for C25H20N5O#1.40HCl#0.40H2O: C, 66.62; H, 4.96; N, 12.43.

Found: C, 66.73; H, 4.94; N, 12.46.

EXAMPLE 9 N-{1-(4-Cyanobenzyl)-1H-imidazol-5-ylethyl}-4(R)-benzyloxy-2 (S)- {N'-acetyl-N'-3-chlorobenzyl}aminomethylpyrolidine Step a: 4(R)-Hydroxyproline methyl ester A suspension of 4(R)-hydroxyproline (35.12g, 267.8 mmol) in methanol (500ml) was saturated with gaseous hydrochloric acid. The resulting solution was allowed to stand for 16 hrs and the solvent evaporated in vacuo to afford the title compound as a white solid.

1H NMR CD30D 6 4.60 (2H, m), 3.86(3H, s), 3.48(1 H, dd, J=3. 6 and 12.0Hz), 3.23(1H, d, J=12.0Hz), 2.43(1H, m) and 2.21(1H, m) ppm.

Step B: N-t-Butoxycarbonyl-4(R)-hydroxyproline methyl ester To a solution of 4(R)-hydroxyproline methyl ester (53.5g, 268mmol), and triethylamine (75ml, 540mmol), in CH2C12 (500ml), at 0°C, was added a solution of di-t-butyl dicarbonate (58.48, 268mmol), in CH2C12 (75ml). The resulting mixture was stirred for 48hrs at room temperature. The solution was washed with 10% aqueous citric acid solution, saturated NaHCO3 solution, dried (Na2SO4) and the solvent evaporated in vacuo. The title

compound was obtained as a yellow oil and used in the next step without further purification.

1H NMR CD30D 6 4.40-4.30 (2H, m), 3.75(3H, m), 3.60-3.40(2H, m), 2.30(1H, m), 2.05(1H, m) and 1.55-1.40(9H, m) ppm.

Step C: N-t-Butoxycarbonyl-4(R)-t-butyldimethylsilyloxy proline methyl ester To a solution of N-t-butoxycarbonyl-4(R)-hydroxy proline methyl ester (65.87g, 268mmol), and triethylamine (41ml, 294mmol), in CH2C12 (536m1), at OOC, was added a solution of t-butyldimethyl silylchloide (42 .49g, 282mmol), in CH2C12 (86ml).

The resulting mixture was stirred for 16hrs at room temperature.

The solution was washed with 10% aqueous citric acid solution, saturated NaHCO3 solution, dried (Na2SO4) and the solvent evaporated in vacuo. The title compound was obtained as a yellow oil and used in the next step without furthur purification.

1H NMR CD30D 5 4.60-4.40 (2H, m), 3.75(3H, m), 3.60-3.20(2H, m), 2.30-1.90(2H, m), 1.45-1.40(9H, m), 0.90-0.85(9H, m), 0. 10- 0.00(6H, m) ppm.

Step D: N-t-Butoxycarbonyl-4(R)-t-butyldimethylsilyloxy-2(S )- hydroxymethvlpyrrolidine A solution of N-t-butoxycarbonyl-4-(R)-t- butyldimethylsilyloxy proline methyl ester (86.65g, 241mmol), in THF (150ml), was added over 90 minutes to a solution of lithium aluminum hydride (247ml of a 1M solution in THF, 247mmol), under argon, so that the temperature did not exceed 12°C. Stirring was continued for 50 mins and then EtOAc (500ml) was added cautiously, followed by sodium sulphate decahydrate (34g), and the resulting mixture stirred for 16 hrs at room temperature.

Anhydrous Na2SO4 (34g) was added and the mixture stirred an additional 30 min and then filtered. The solids were washed with EtOAc (800ml), the filtrates combined and the solvent evaporated in

vacuo. The title compound was obtained as a colorless oil and used in the next step without further purification.

Step E: N-t-Butoxycarbonyl-4(R)-t-butyldimethylsilyloxy-2(S )- methanesulfonyloxymethylpyrrolidine To a solution of N-t-butoxycarbonyl-4(R)-t- butyldimethylsilyloxy-2 (S)-hydroxymethylpyrrolidine (50.0g, 150.8mmol) and triethylamine (42.0ml, 300 mmol) in CH2Cl2(1 1) was added methane sulfonyl chloride (12.4ml, 160mmol) over a period of 5 minutes and stirring was continued for 1 hour. The solvent was evaporated in vacuo diluted with EtOAc (800mL) and washed sequentially with aqueous citric acid and NaHCO3. The organic extracts were dried (Na2SO4), evaporated in vacuo and the residue purified by chromatography (SiO2, 15% EtOAc in hexanes).

The title compound was obtained as a pale yellow solid FAB Mass spectrum, m/z = 410(M+1).

1H NMR CDCl3 6 4.60-4.00 (4H, m), 3.60-3.30(2H, m), 2.98(3H, s), 2.05-2.00(2H, m), 1.48-1.42(9H, m),0.90-0.80(9H, m), 0. 10- 0.00(6H, m) ppm.

Step F: Preparation of N-t-Butoxycarbonyl-4(R)-t- butyldimethylsilyloxy-2(S)-azidomethylpyrrolidine In a flask protected by a safety screen, a solution of N-t-butoxycarbonyl-4(S )-t-butyldimethylsilyloxy-2(S )-methane- sulfonyloxy methyl pyrrolidine(10.40g, 25.39mmol) and tetrabutyl- ammonium azide (8.1 8g, 28.7mmol) in toluene (250ml) was stirred at 80°C for Shr. The reaction was cooled to room temperature and diluted with EtOAc (250ml), washed with water and brine and dried (Na2SO4). The solvent was evaporated in vacuo to afford the title compound as a yellow oil which was used in the next step without furthur purification.

1H NMR CDCl3 6 4.60-3.20 (6H, m), 2.05-1.90(2H, m), 1.47(9H, s), 0.87(9H, s) and 0.10-0.00(6H, m) ppm.

Step G: Preparation of N-t-Butoxycarbonyl-4(R)-t- butyldimethvlsilyloxy-2(S )-aminomethylpyrrolidine A solution of N-t-butoxycarbonyl-4(R)-t-butyldimethyl- silyloxy-2(S)-azidomethylpyrrolidine (9. 06g, 25.3 9mmol) in EtOAc (120ml) was purged with argon and 10% palladium on carbon (1.05 g) added. The flask was evacuated and stirred under an atmosphere of hydrogen (49 psi) for 1 6hrs. The hydrogen was replaced by argon, the catalyst removed by filtration and the solvent evaporated in vacuo. The residue was chromatographed (SiO2, 2.5 to 5% saturated NH40H in acetonitrile, gradient elution), to afford the title compound as an oil.

1H NMR(CDCl3, 400 MHz) 5 4.40-2.60 (6H, s), 2.05-1.80(2H, m), 1.46(9H, s), 1.36(2H, s), 0.87(9H, s), 0.10-0.00(6H, m)ppm.

Step H: Preparation of N-t-B utoxycarbonyl-4(R)-t- butyldimethylsilyloxy-2(S)-{N'-3- chlorobenzyl}aminomethylpyrrolidine To a slurry of 3-chlorobenzaldehyde (1 .2ml, 10.6mmol), crushed 3A molecular sieves (9.5g) and the amine from step G (3.50g, 10.6mmol) in methanol (150 ml) was added sodium cyanoborohydride (11.0ml of a 1M solution in THF, 11. 0mmol) at room temperature. The pH was adjusted to 7 by the addition of glacial acetic acid (0.68ml, 12mmol) and the reaction was stirred for 16 hrs. The reaction was filtered and the filtrate evaporated in vacuo.

The residue was partitioned between EtOAc and saturated NaHCO3 solution and the organic extract washed with brine, dried (Na2SO4), and the solvent evaporated in vacuo. The residue was purified by chromatography (SiO2, 2.5% MeOH in CH2C12) to provide the title compound as an oil.

1HNMR(CDCl3, 400 MHz) 5 7.40-7.10(4H, m), 4.36(1H, s), 4.15- 3.90(2H, m), 3.90-3.30(2H, m), 2.85-2.60(2H, m), 2.05-1.90(2H, m), 1.44(9H, s), 0.87(9H, s) and 0.06(6H, m) ppm.

Step I: Preparation of N-t-Butoxycarbonyl-4(R)-t- butyldimethylsilyloxy-2(S)-{N'-3-chlorobenzyl-N'- acetic - aminomethylpyrrolidine To a solution of N-t-butoxycarbonyl-4(R)-t- butyldimethylsilyloxy-2(S)-{N'-3-chlorobenzyl}-aminomethyl pyrrolidine (3.80g, 8.35 mmol) in CH2C12 (85ml) and triethylamine (2.40ml, 17.0 mmol) at OOC was added acetyl chloride (0.60ml, 8.44 mmol). The reaction was stirred at room temperature for lhr, diluted with water and extracted with CH2Cl2. The extracts were washed with brine, dried (Na2SO4) and the solvent evaporated in vacuo. The residue was purified by chromatography (SiO2, 10 to 25% EtOAc in CH2C12, gradient elution).

1HNMR (CDCl3, 400 MHz) # 7.40-7.00(4H, m), 5.10-3,00(8H, m), 2.20-1.70(5H, m), 1.50-1.30(9H, m), 0.87(9H, s) and 0.06(6H, m) ppm.

Step J: Preparation of N-t-Butoxycarbonyl-4(R)-hydroxy-2(S)- {N' -3-chlorobenzyl-N'-acetyl}-aminomethylpyrrolidine To a solution of N-t-butoxycarbonyl-4(R)-t- butyldimethylsilyloxy -2(S)-( N'-3-chlorobenzyl-N'-acetyl aminomethylpyrrolidine (4.02g, 8.09 mmol) in THF (80ml) at OOC was added tetrabutylammonium fluoride (9.00ml of a 1M solution in THF, 9.00mmol). the reasction was stirred at 0°C for lhr and then at room temperature for 30min. The reaction was quenched by the addition of a saturated NH4C1 solution (50ml), dilution with EtOAc.

The organic extracts were washed with brine, dried (Na2SO4) and the solvent evaporated in vacuo. The residue purified by chromatography (SiO2, 3 to 5% MeOH in CH2C12, gradient elution) to afford the title compound as a foam.

1HNMR (CDC13, 400 MHz) 5 7.40-7.00(4H, m), 5.00-4,00(4H, m), 4.00-3.10(4H, m), 2.30-1.60(5H, m) and 1.50-1.30(9H, m) ppm.

Step K: N-t-Butoxycarbonyl-4(R)-benzyloxyoxy-2(S N'- acetyl-N'-3-chlorobenzyl}aminomethylpyrrolidine

To a solution of N-t-Butoxycarbonyl-4(S)-hydroxy - 2(S)-(N'-acetyl-N' 3-chlorobenzyl) aminomethylpyrrolidine (701 mg, 1.83 mmol) in DMF (9ml) at OOC was added sodium hydride (110mg of a 60% dispersion in mineral oil, 2.75mmol). After 15 min benzyl bromide (0.435m1, 3.66mmol), was added and the reaction stirred at room temperature for 16 hrs. The reaction was quenched with saturated NaHCO3 solution (2ml) and extracted with ethyl acetate.

The organic extract was washed with brine and dried (Na2SO4), and the solvent evaporated in vacuo. The residue was purified by chromatography (SiO2, 25 to 50% EtOAc in CH2C12, gradient elution) to afford the title compound as a foam.

Step L: 4(S)-Benzyloxy-2(S)- ( N'-acetyl-N'-3-chlorobenzyl )- aminomethvlpyrrolidine hvdrochloride A solution of the product from step K (0.834g, 1.76 mmol) in EtOAc (25 ml) at OOC was saturated with gaseous hydrogen chloride. The resulting solution was allowed to stand at room temperature for 30min. The solvent was evaporated in vacuo to afford the title compound as a white solid.

Step M: Preparation of 1H-Imidazole-4- acetic acid methyl ester hydrochloride.

A solution of 1H-imidazole-4-acetic acid hydrochloride (4.00g, 24.6 mmol) in methanol (100 ml) was saturated with gaseous hydrogen chloride. The resulting solution was allowed to stand at room temperature (RT) for 18hr. The solvent was evaporated in vacuo to afford the title compound as a white solid.

1H NMR(CDC13, 400 MHz) 6 8.85(1H, s),7.45(1H, s), 3.89(2H, s) and 3.75(3H, s) ppm.

Step N: Preparation of 1-(Triphenylmethyl)-lH-imidazol-4- ylacetic acid methyl ester.

To a solution of the product from Step M (24.85g, 0. 141mol) in dimethyl formamide (DMF) (115ml) was added triethylamine (57.2 ml, 0.412mol) and triphenylmethyl bromide

(S5.3g, 0. 171mol) and the suspension was stirred for 24hr. After this time, the reaction mixture was diluted with ethyl acetate (EtOAc) (1 1) and water (350 ml). The organic phase was washed with sat. aq.

NaHCO3 (350 ml), dried (Na2SO4) and evaporated in vacuo. The residue was purified by flash chromatography (SiO2, 0-100% ethyl acetate in hexanes; gradient elution) to provide the title compound as a white solid.

1H NMR (CDC13, 400 MHz) 6 7.35(1H, s), 7.31(9H, m), 7.22(6H, m), 6.76(1 H, s), 3.68(3H, s) and 3.60(2H, s) ppm.

Step O: Preparation of [1-(4-cyanobenzyl)-lH-imidazol-5- yllacetic acid methyl ester.

To a solution of the product from Step N (8.00g, 20.9mmol) in acetonitrile (70 ml) was added bromo-p-tolunitrile (4. lOg, 20.92 mmol) and heated at 550C for 3 hr. After this time, the reaction was cooled to room temperature and the resulting imidazolium salt (white precipitate) was collected by filtration.

The filtrate was heated at 550C for 18hr. The reaction mixture was cooled to room temperature and evaporated in vacuo. To the residue was added EtOAc (70 ml) and the resulting white precipitate collected by filtration. The precipitated imidazolium salts were combined, suspended in methanol (100 ml) and heated to reflux for 30min. After this time, the solvent was removed in vacuo, the resulting residue was suspended in EtOAc (75ml) and the solid isolated by filtration and washed (EtOAc). The solid was treated with sat aq NaHCO3 (300ml) and CH2C12 (300ml) and stirred at room temperature for 2 hr. The organic layer was separated, dried (MgSO4) and evaporated in vacuo to afford the title compound as a white solid: 1HNMR(CDCl3, 400 MHz) 6 7.65(1 H, d, J=8Hz), 7.53(1 H, s), 7.15(1H, d, J=8Hz), 7.04(1H, s), 5.24(2H, s), 3.62(3H, s) and 3.45(2H, s) ppm.

Step P: Preparation of (l-(4-Cyanobenzyl)-lH-imidazol-5-yl)- ethanol

To a stirred solution of the ester from step 0, (1.50g, 5.88mmol), in methanol (20ml) at OOC, was added sodium borohydride (1.0g, 26.3mmol) portionwise over 5 minutes. The reaction was stirred at OOC for 1 hr and then at room temperature for an additional 1 hr. The reaction was quenched by the addition of sat.NH4Cl solution and the methanol was evaporated in vacuo.. The residue was partitioned between EtOAc and sat NaHCO3 solution and the organic extracts dried (MgSO4), and evaporated in vacuo.. The residue was purified by chromatography (SiO2, 4 to 10% methanol in methylene chloride, gradient elution) to afford the title compound as a solid.

1H NMR CDCl3 6 7.64(2H, d, J=8.2Hz), 7.57(1H, s), 7.11(2H, d, J=8.2Hz), 6.97(1 H, s), 5.23(2H, s), 3.79(2H, t, J=6.2Hz) and 2.66(2H, t, J=6.2Hz) ppm.

Step O: 1-(4-Cyanobenzyl)-imidazol-5-yl-thylmethaneoulfonate A solution of (1-(4-Cyanobenzyl)-1H-imidazol-5-yl)- ethanol (0.500 g, 2.20 mmol) in methylene chloride (6.0 ml) at OOC was treated with Hunig's base (0.460ml, 2.64mmol) and methane sulfonyl chloride (0.204ml, 2.64mmol). After 2 hrs, the reaction was quenched by addition of saturated NaHCO3 solution (50ml) and the mixture was extracted with methylene chloride (50ml), dried (MgSO4) and the solvent evaporated in vacuo. The title compound was used without furthur purification.

1H NMR CDCl3 5 7.69 (1H, s) 7.66(2H, d, J=8.2Hz), 7.15 (2H, d, J=8.2Hz, 7.04 (1H, s), 5.24(2H, s), 4.31(2H, t, J=6.7Hz), 2.96(3H, s), and 2.88(2H, t, J=6.6Hz)ppm.

Step R: N{1-(4-Cyanobenzyl)-1H-imidazol-5-ylethyl}-4-(R)- benzyloxyoxy-2-(S)-{N'-acetyl-N'-3- chlorobenzyl}aminomethylpyrrolidine A mixture of 4(R)-benzyloxy-2(S)- {N'-acetyl-N'-3- chlorobenzyl-aminomethyl}pyrrolidine (199mg, 0.486mmol), the mesylate from step Q (140mg, 0.458mmol), potassium carbonate

(165mg, 1.19mmol), and sodium iodide (289mg, 1.93mmol) in DMF (1.5ml), were heated at 550C for 16 hrs. The cooled mixture was diluted with EtOAc, washed with NaHCO3 solution and brine, dried (Na2SO4) and the solvent evaporated in vacuo. The residue was purified by preparative HPLC (C-18, 95:5 to 5:95 water in acetonitrile containing 0.1% TFA, gradient elution). The title compound was obtained as a white solid after lyophillisation.

Anal. calc'd for C34H36N502Cl 3.00 TFA, 0.85 H2O: C, 51.14; H, 4.37, N, 7.45.

Found: C, 51.15; H, 4.42; N, 6.86.

FAB HRMS exact mass calc'd for C34H37NsO2Cl 582.263579(MH+), Found: 582.263900.

EXAMPLE 10 In vitro inhibition Transferase Assays. Isoprenyl-protein transferase activity assays were carried out at 300C unless noted otherwise. A typical reaction contained (in a final volume of 50 RL) : [3H]farnesyl diphosphate or [3H]geranylgeranyl diphosphate, Ras protein, 50 mM HEPES, pH 7.5, 5 mM MgC12, 5 mM dithiothreitol, 10 uM ZnCl2, 0.1% polyethyleneglycol (PEG) (15,000-20,000 mw) and isoprenyl-protein transferase. A modulating anion such as 10mM glycerol phosphate or 5mM ATP may also be added to the assay medium.

The FPTase employed in the assay was prepared by recombinant expression as described in Omer, C.A., Kral, A.M., Diehl, R.E., Prendergast, G.C., Powers, S., Allen, C.M., Gibbs, J.B. and Kohl, N.E. (1993) Biochemistry 32:5167-5176. The geranylgeranyl-protein transferase-type I employed in the assay was prepared as described in U.S. Pat. No. 5,470,832, incorporated by reference. After thermally pre-equilibrating the assay mixture in the absence of enzyme, reactions were initiated by the addition of isoprenyl-protein transferase and stopped at timed intervals (typically 15 min) by the addition of 1 M HC1

in ethanol (1 mL). The quenched reactions were allowed to stand for 15 m (to complete the precipitation process). After adding 2 mL of 100% ethanol, the reactions were vacuum-filtered through Whatman GF/C filters. Filters were washed four times with 2 mL aliquots of 100% ethanol, mixed with scintillation fluid (10 mL) and then counted in a Beckman LS3801 scintillation counter.

For inhibition studies, assays were run as described above, except inhibitors were prepared as concentrated solutions in 100% dimethyl sulfoxide and then diluted 20-fold into the enzyme assay mixture. Substrate concentrations for inhibitor IC50 determinations were as follows: FTase, 650 nM Ras-CVLS (SEQ.ID.NO.: 2), 100 nM farnesyl diphosphate; GGPTase-I, 500 nM Ras-CAIL (SEQ.ID.NO.: 3), 100 nM geranylgeranyl diphosphate.

EXAMPLE 11 Modified In vitro GGTase inhibition assay The modified geranylgeranyl-protein transferase inhibition assay is carried out at room temperature. A typical reaction contains (in a final volume of 50 CLL): [3H]geranylgeranyl diphosphate, biotinylated Ras peptide, 50 mM HEPES, pH 7.5, a modulating anion (for example 10 mM glycerophosphate or 5mM ATP), 7 mM MgCl2, 10 µM ZnCl2, 0. 1% PEG (15,000-20,000 mw), 2 mM dithiothreitol, and geranylgeranyl-protein transferase type I (GGTase-I). The GGTase- type I enzyme employed in the assay is prepared as described in U. S.

Pat. No. 5,470,832, incorporated by reference. The Ras peptide is derived from the K4B-Ras protein and has the following sequence: biotinyl-GKKKKKKSKTKCVIM (single amino acid code) (SEQ.ID.NO.: 13). Reactions are initiated by the addition of GGTase and stopped at timed intervals (typically 15 min) by the addition of 200 , uL of a 3 mg/mL suspension of streptavidin SPA beads (Scintillation Proximity Assay beads, Amersham) in 0.2 M sodium phosphate, pH 4, containing 50 mM EDTA, and 0.5% BSA. The quenched reactions are allowed to stand for 2 hours before analysis on a Packard TopCount scintillation counter.

For inhibition studies, assays are run as described above, except inhibitors are prepared as concentrated solutions in 100% dimethyl sulfoxide and then diluted 25-fold into the enzyme assay mixture. For inhibition studies with slow-binding inhibitors, GGTase and inhibitors are preincubated for one hour and reactions are initiated by the addition of peptide substrate, following methodology described by J.F. Morrison, C.T. Walsh, Adv. Enzymol. & Related Areas Mol.

Biol., 61 201-301 (1988). IC5o values are determined with Ras peptide near KM concentrations. Enzyme and substrate concentrations for inhibitor IC50 determinations are as follows: 75 pM GGTase-I, 1.6 RM Ras peptide, 100 nM geranylgeranyl diphosphate.

Alternatively, enzymologic Ki values for inhibition of GGPTase-I can be determined using the methodology described by I. H.

Segel ("Enzyme Kinetics", pages 342-345; Wiley and Sons, New York, N.Y. (1975) and references cited therein).

EXAMPLE 13 Cell-basedin vitro ras prenylation assay The cell lines used in this assay consist of either Ratl or NIH3T3 cells transformed by either viral H-ras; an N-ras chimeric gene in which the C-terminal hypervariable region of v-H-ras was substituted with the corresponding region from the N-ras gene; or ras-CVLL (SEQ.ID.NO.: 1), a viral-H-ras mutant in which the C-terminal exon encodes leucine instead of serine, making the encoded protein a substrate for geranylgeranylation by GGPTase I. The assay can also be performed using cell lines transformed with human H-ras, N-ras or Ki4B-ras. The assay is performed essentially as described in DeClue, J.E. et al., Cancer Research 51:712-717, (1991). Cells in 10 cm dishes at 50-75% confluency are treated with the test compound(s) (final concentration of solvent, methanol or dimethyl sulfoxide, is 0.1%). After 4 hours at 37"C, the cells are labeled in 3 ml methionine-free DMEM supple- mented with 10% regular DMEM, 2% fetal bovine serum, 400 ltCi[35S]methionine (1000 Ci/mmol) and test compound(s). Cells treated with lovastatin, a compound that blocks Ras processing in

cells by inhibiting the rate-limiting step in the isoprenoid biosynthetic pathway (Hancock, J.F. et al. Cell, 57:1167 (1989); DeClue, J.E. et al. Cancer Res., 51:712 (1991); Sinensky, M. et al. J. Biol. Chem., 265:19937 (1990)), serve as a positive control in this assay. After an additional 20 hours, the cells are lysed in 1 ml lysis buffer (1% NP40/20 mM HEPES, pH 7.5/5 mM MgC12/lmM DTT/10 mg/ml aprotinen/2 mg/ml leupeptin/2 mg/ml antipain/0.5 mM PMSF) and the lysates cleared by centrifugation at 100,000 x g for 45 min. Alternatively, four hours after the addition of the labeling media, the media is removed, the cells washed, and 3 ml of media containing the same or a different test compound added. Following an additional 16 hour incubation, the lysis is carried out as above. Aliquots of lysates containing equal numbers of acid-precipitable counts are bought to 1 ml with IP buffer (lysis buffer lacking DTT) and immunoprecipitated with the ras-specific monoclonal antibody Y13-259 (Furth, M.E. et al., J. Virol. 43:294-304, (1982)).

Following a 2 hour antibody incubation at 40C, 200 ul of a 25% suspen- sion of protein A-Sepharose coated with rabbit anti rat IgG is added for 45 min. The immunoprecipitates are washed four times with IP buffer (20 nM HEPES, pH 7.5/1 mM EDTA/1% Triton X-100.0.5% deoxycholate/0. 1 %/SDS/0. 1 M NaCl) boiled in SDS-PAGE sample buffer and loaded on 13% acrylamide gels. When the dye front reached the bottom, the gel is fixed, soaked in Enlightening, dried and auto- radiographed. The intensities of the bands corresponding to prenylated and nonprenylated Ras proteins are compared to determine the percent inhibition of prenyl transfer to protein.

EXAMPLE 14 Construction of SEAP reporter plasmid pDSE100 The SEAP reporter plasmid, pDSE100 was constructed by ligating a restriction fragment containing the SEAP coding sequence into the plasmid pCMV-RE-AKI. The SEAP gene is derived from the plasmid pSEAP2-Basic (Clontech, Palo Alto, CA). The plasmid pCMV-RE-AKI was constructed by Deborah Jones (Merck) and contains

5 sequential copies of the 'dyad symmetry response element' cloned upstream of a 'CAT-TATA' sequence derived from the cytomegalovirus immediate early promoter. The plasmid also contains a bovine growth hormone poly-A sequence.

The plasmid, pDSE100 was constructed as follows. A restriction fragment encoding the SEAP coding sequence was cut out of the plasmid pSEAP2-Basic using the restriction enzymes EcoR1 and HpaI. The ends of the linear DNA fragments were filled in with the Klenow fragment of E. coli DNA Polymerase I. The 'blunt ended' DNA containing the SEAP gene was isolated by electrophoresing the digest in an agarose gel and cutting out the 1694 base pair fragment. The vector plasmid pCMV-RE-AKI was linearized with the restriction enzyme Bgl-II and the ends filled in with Klenow DNA Polymerase I. The SEAP DNA fragment was blunt end ligated into the pCMV-RE-AKI vector and the ligation products were transformed into DHS-alpha E. coli cells (Gibco-BRL). Transformants were screened for the proper insert and then mapped for restriction fragment orientation. Properly oriented recombinant constructs were sequenced across the cloning junctions to verify the correct sequence. The resulting plasmid contains the SEAP coding sequence downstream of the DSE and CAT-TATA promoter elements and upstream of the BGH poly-A sequence.

Alternative Construction of SEAP reporter plasmid pDSE101 The SEAP repotrer plasmid, pDSE101 is also constructed by ligating a restriction fragment containing the SEAP coding sequence into the plasmid pCMV-RE-AKI. The SEAP gene is derived from plasmid pGEM7zf(-)/SEAP.

The plasmid pDSE101 was constructed as follows: A restriction fragment containing part of the SEAP gene coding sequence was cut out of the plasmid pGEM7zf(-)/SEAP using the restriction enzymes Apa I and KpnI. The ends of the linear DNA fragments were chewed back with the Klenow fragment of E. coli DNA Polymerase I. The "blunt ended" DNA containing the truncated SEAP gene was isolated by electrophoresing the digest in an agarose gel and

cutting out the 1910 base pair fragment. This 1910 base pair fragment was ligated into the plasmid pCMV-RE-AKI which had been cut with Bgl-II and filled in with E. coli Klenow fragment DNA polymerase.

Recombinant plasmids were screened for insert orientation and sequenced through the ligated junctions. The plasmid pCMV-RE-AKI is derived from plasmid pCMVIE-AKI-DHFR (Whang , Y., Silberklang, M., Morgan, A., Munshi, S., Lenny, A.B., Ellis, R.W., and Kieff, E.

(1987) J. Virol., 61, 1796-1807) by removing an EcoRI fragment containing the DHFR and Neomycin markers. Five copies of the fos promoter serum response element were inserted as described previously (Jones, R.E., Defeo-Jones, D., McAvoy, E.M., Vuocolo, G.A., Wegrzyn, R.J., Haskell, K.M. and Oliff, A. (1991) Oncogene, 6, 745-751) to create plasmid pCMV-RE-AKI.

The plasmid pGEM7zf(-)/SEAP was constructed as follows. The SEAP gene was PCRed, in two segments from a human placenta cDNA library (Clontech) using the following oligos.

Sense strand N-terminal SEAP : 5' GAGAGGGAATTCGGGCCCTTCCTGCAT GCTGCTGCTGCTGCTGCTGCTGGGC 3' (SEQ.ID.NO.:4) Antisense strand N-terminal SEAP : 5' GAGAGAGCTCGAGGTTAACCCGGGT GCGCGGCGTCGGTGGT 3' (SEQ.ID.NO.:5) Sense strand C-terminal SEAP: 5' GAGAGAGTCTAGAGTTAACCCGTGGTCC CCGCGTTGCTTCCT 3' (SEQ.ID.NO.:6) Antisense strand C-terminal SEAP: 5' GAAGAGGAAGCTTGGTACCGCCACTG GGCTGTAGGTGGTGGCT 3' (SEQ.ID.NO.:7)

The N-terminal oligos (SEQ.ID.NO.: 4 and SEQ.ID.NO.: 5) were used to generate a 1560 bp N-terminal PCR product that contained EcoRI and HpaI restriction sites at the ends. The Antisense N-terminal oligo (SEQ.ID.NO.: 5) introduces an internal translation STOP codon within the SEAP gene along with the HpaI site. The C-terminal oligos (SEQ.ID.NO.: 6 and SEQ.ID.NO.: 7) were used to amplify a 412 bp C-terminal PCR product containing HpaI and HindIII restriction sites.

The sense strand C-terminal oligo (SEQ.ID.NO.: 6) introduces the internal STOP codon as well as the HpaI site. Next, the N-terminal amplicon was digested with EcoRI and HpaI while the C-terminal amplicon was digested with HpaI and HindIII. The two fragments comprising each end of the SEAP gene were isolated by electro- phoresing the digest in an agarose gel and isolating the 1560 and 412 base pair fragments. These two fragments were then co-ligated into the vector pGEM7zf(-) (Promega) which had been restriction digested with EcoRI and HindIII and isolated on an agarose gel. The resulting clone, pGEM7zf(-)/SEAP contains the coding sequence for the SEAP gene from amino acids.

Construction of a constitutively expressing SEAP plasmid pCMV-SEAP-A An expression plasmid constitutively expressing the SEAP protein was created by placing the sequence encoding a truncated SEAP gene downstream of the cytomegalovirus (CMV) IE-1 promoter. The expression plasmid also includes the CMV intron A region 5' to the SEAP gene as well as the 3, untranslated region of the bovine growth hormone gene 3' to the SEAP gene.

The plasmid pCMVIE-AKI-DHFR (Whang , Silberklang, M., Morgan, A., Munshi, S., Lenny, A.B., Ellis, R.W., and Kieff, E. (1987) J. Virol., 61:1796-1807) containing the CMV immediate early promoter was cut with EcoRI generating two fragments. The vector fragment was isolated by agarose electrophoresis and religated. The resulting plasmid is named pCMV-AKI. Next, the cytomegalovirus intron A nucleotide sequence was inserted downstream

of the CMV IE1 promter in pCMV-AKI. The intron A sequence was isolated from a genomic clone bank and subcloned into pBR322 to generate plasmid p16T-286. The intron A sequence was mutated at nucleotide 1856 (nucleotide numbering as in Chapman, B.S., Thayer, R.M., Vincent, K.A. and Haigwood, N.L., Nuc.Acids Res. 19, 3979- 3986) to remove a SacI restriction site using site directed mutagenesis.

The mutated intron A sequence was PCRed from the plasmid p 1 6T-287 using the following oligos.

Sense strand: 5' GGCAGAGCTCGTTTAGTGAACCGTCAG 3' (SEQ.ID.NO.: 8) Antisense strand: 5' GAGAGATCTCAAGGACGGTGACTGCAG 3' (SEQ.ID.NO.: 9) These two oligos generate a 991 base pair fragment with a SacI site incorporated by the sense oligo and a Bgl-II fragment incorporated by the antisense oligo. The PCR fragment is trimmed with SacI and Bgl-II and isolated on an agarose gel. The vector pCMV-AKI is cut with SacI and Bgl-II and the larger vector fragment isolated by agarose gel electrophoresis. The two gel isolated fragments are ligated at their respective SacI and Bgl-II sites to create plasmid pCMV-AKI- InA.

The DNA sequence encoding the truncated SEAP gene is inserted into the pCMV-AKI-InA plasmid at the Bgl-II site of the vector. The SEAP gene is cut out of plasmid pGEM7zf(-)/SEAP (described above) using EcoRI and HindIII. The fragment is filled in with Klenow DNA polymerase and the 1970 base pair fragment isolated from the vector fragment by agarose gel electrophoresis. The pCMV- AKI-InA vector is prepared by digesting with Bgl-II and filling in the ends with Klenow DNA polymerase. The final construct is generated by blunt end ligating the SEAP fragment into the pCMV-AKI-InA vector.

Transformants were screened for the proper insert and then mapped for restriction fragment orientation. Properly oriented recombinant

constructs were sequenced across the cloning junctions to verify the correct sequence. The resulting plasmid, named pCMV-SEAP-A (deposited in the ATCC under Budapest Treaty on August 27, 1998, and designated ATCC), contains a modified SEAP sequence downstream of the cytomegalovirus immediately early promoter IE-1 and intron A sequence and upstream of the bovine growth hormone poly-A sequence.

The plasmid expresses SEAP in a constitutive manner when transfected into mammalian cells.

Alternative construction of a constitutively expressing SEAP plasmid pCMV-SEAP-B An expression plasmid constitutively expressing the SEAP protein can be created by placing the sequence encoding a truncated SEAP gene downstream of the cytomegalovirus (CMV) IE-1 promoter and upstream of the 3' unstranslated region of the bovine growth hormone gene.

The plasmid pCMVIE-AKI-DHFR (Whang , Silberklang, M., Morgan, A., Munshi, S., Lenny, A.B., Ellis, R.W., and Kieff, E. (1987) J. Virol., 61:1796-1807) containing the CMV immediate early promoter and bovine growth hormone poly-A sequence can be cut with EcoRI generating two fragments. The vector fragment can be isolated by agarose electrophoresis and religated. The resulting plasmid is named pCMV-AKI. The DNA sequence encoding the truncated SEAP gene can be inserted into the pCMV-AKI plasmid at a unique Bgl-II in the vector. The SEAP gene is cut out of plasmid pGEMzf(-)/SEAP (described above) using EcoRI and HindIII. The fragments are filled in with Klenow DNA polymerase and the 1970 base pair fragment is isolated from the vector fragment by agarose gel electrophoresis. The pCMV-AKI vector is prepared by digesting with Bgl-II and filling in the ends with Klenow DNA polymerase. The final construct is generated by blunt end ligating the SEAP fragment into the vector and transforming the ligation reaction into E. coli DHSa cells.

Transformants can then be screened for the proper insert and mapped for restriction fragment orientation. Properly oriented recombinant constructs would be sequenced across the cloning junctions to verify

the correct sequence. The resulting plasmid, named pCMV-SEAP-B contains a modified SEAP sequence downstream of the cytomegalovirus immediate early promoter, IE1, and upstream of a bovine growth hormone poly-A sequence. The plasmid would express SEAP in a constitutive nammer when transfected into mammalian cells.

Cloning of a Myristylated viral-H-ras expression plasmid pSMS600 A DNA fragment containing viral-H-ras can be PCRed from plasmid "HB-11 (deposited in the ATCC under Budapest Treaty on August 27, 1997, and designated ATCC 209,218) using the following oligos.

Sense strand: S, TCTCCTCGAGGCCACCATGGGGAGTAGCAAGAGCAAGCCTAA <BR> <BR> <BR> <BR> GGACCCCAGCCAGCGCCGGATGACAGAATACAAGCTTGTGGTG G 3'. (SEQ.ID.NO.: 10) Antisense: 5' CACATCTAGATCAGGACAGCACAGACTTGCAGC 3'.

(SEQ.ID.NO.: 11) A sequence encoding the first 15 aminoacids of the v-src gene, containing a myristylation site, is incorporated into the sense strand oligo. The sense strand oligo also optimizes the 'Kozak' translation initiation sequence immediately 5' to the ATG start site. To prevent prenylation at the viral-ras C-terminus, cysteine 186 would be mutated to a serine by substituting a G residue for a C residue in the C-terminal antisense oligo. The PCR primer oligos introduce an XhoI site at the 5' end and a XbaI site at the 3'end. The XhoI-XbaI fragment can be ligated into the mammalian expression plasmid pCI (Promega) cut with XhoI and XbaI. This results in a plasmid, pSMS600, in which the recombinant myr-viral-H-ras gene is constitutively transcribed from the CMV promoter of the pCI vector.

Cloning of a viral-H-ras-CVLL expression plasmid pSMS601

A viral-H-ras clone with a C-terminal sequence encoding the amino acids CVLL can be cloned from the plasmid "HB-11" by PCR using the following oligos.

Sense strand: S, TCTCCTCGAGGCCACCATGACAGAATACAAGCTTGTGGTGG- 3' (SEQ.ID.NO.: 12) Antisense strand: 5' CACTCTAGACTGGTGTCAGAGCAGCACACACTTGCAGC-3' (SEQ.ID.NO.: 13) The sense strand oligo optimizes the 'Kozak' sequence and adds an XhoI site. The antisense strand mutates serine 189 to leucine and adds an XbaI site. The PCR fragment can be trimmed with XhoI and XbaI and ligated into the XhoI-XbaI cut vector pCI (Promega).

This results in a plasmid, pSMS601, in which the mutated viral-H-ras- CVLL gene is constitutively transcribed from the CMV promoter of the pCI vector.

Cloning of cellular-H-ras-Leu61 expression plasmid pSMS620 The human cellular-H-ras gene can be PCRed from a human cerebral cortex cDNA library (Clontech) using the following oligonucleotide primers.

Sense strand: <BR> <BR> 5'-GAGAGAATTCGCCACCATGACGGAATATAAGCTGGTGG-3' <BR> <BR> <BR> <BR> <BR> <BR> (SEQ.ID.NO.:14) Antisense strand: 5' -GAGAGTCGACGCGTCAGGAGAGCACACACTTGC-3' (SEQ.ID.NO.: 15) The primers will amplify a c-H-Ras encoding DNA fragment with the primers contributing an optimized 'Kozak' translation start sequence, an EcoRI site at the N-terminus and a Sal I site at the

C-terminal end. After trimming the ends of the PCR product with EcoRI and Sal I, the c-H-ras fragment can be ligated ligated into an EcoRI -Sal I cut mutagenesis vector pAlter- 1 (Promega). Mutation of glutamine-61 to a leucine can be accomplished using the manufacturer's protocols and the following oligonucleotide: 5'-CCGCCGGCCTGGAGGAGTACAG-3' (SEQ.ID.NO.:16) After selection and sequencing for the correct nucleotide substitution, the mutated c-H-ras-Leu6 1 can be excised from the pAlter- 1 vector, using EcoRI and Sal I, and be directly ligated into the vector pCI (Promega) which has been digested with EcoRI and Sal I. The new recombinant plasmid, pSMS620, will constitutively transcribe c-H-ras- Leu61 from the CMV promoter of the pCI vector.

Cloning of a c-N-ras-Val-12 expression plasmid pSMS630 The human c-N-ras gene can be PCRed from a human cerebral cortex cDNA library (Clontech) using the following oligonucleotide primers.

Sense strand: 5'-GAGAGAATTCGCCACCATGACTGAGTACAAACTGGTGG-3' <BR> <BR> <BR> <BR> <BR> (SEQ.ID.NO.:17) Antisense strand: 5'-GAGAGTCGACTTGTTACATCACCACACATGGC-3' (SEQ.ID.NO.: 18) The primers will amplify a c-N-Ras encoding DNA fragment with the primers contributing an optimized 'Kozak' translation start sequence, an EcoRI site at the N-terminus and a Sal I site at the C-terminal end. After trimming the ends of the PCR product with EcoRI and Sal I, the c-N-ras fragment can be ligated into an EcoRI -Sal I cut mutagenesis vector pAlter-l (Promega). Mutation of glycine- 12 to a valine can be accomplished using the manufacturer's protocols and the following oligonucleotide:

5'-GTTGGAGCAGTTGGTGTTGGG-3' (SEQ.ID.NO.:19) After selection and sequencing for the correct nucleotide substitution, the mutated c-N-ras-Val-12 can be excised from the pAlter- 1 vector, using EcoRI and Sal I, and be directly ligated into the vector pCI (Promega) which has been digested with EcoRI and Sal I. The new recombinant plasmid, pSMS 630, will constitutively transcribe c-N-ras- Val-12 from the CMV promoter of the pCI vector.

Cloning of a c-K4B-ras-Val-12 expression plasmid pSMS640 The human c-K4B-ras gene can be PCRed from a human cerebral cortex cDNA library (Clontech) using the following oligo- nucleotide primers.

Sense strand: 5'-GAGAGGTACCGCCACCATGACTGAATATAAACTTGTGG-3' (SEQ.ID.NO.: 20) Antisense strand: 5' -CTCTGTCGACGTATTTACATAATTACACACTTTGTC-3' (SEQ.ID.NO.: 21) The primers will amplify a c-K4B-Ras encoding DNA fragment with the primers contributing an optimized 'Kozak' translation start sequence, a KpnI site at the N-terminus and a Sal I site at the C-terminal end. After trimming the ends of the PCR product with Kpn I and Sal I, the c-K4B-ras fragment can be ligated into a KpnI -Sal I cut mutagenesis vector pAlter-l (Promega). Mutation of cysteine-12 to a valine can be accomplished using the manufacturer's protocols and the following oligonucleotide: 5' -GTAGTTGGAGCTGTTGGCGTAGGC-3 ' (SEQ.ID.NO.: 22) After selection and sequencing for the correct nucleotide substitution, the mutated c-K4B-ras-Val-12 can be excised from the pAlter-l vector, using KpnI and Sal I, and be directly ligated into the

vector pCI (Promega) which has been digested with KpnI and Sal I.

The new recombinant plasmid will constitutively transcribe c-K4B- ras-Val-12 from the CMV promoter of the pCI vector.

Cloning of c-K-ras4A-Val-12 expression plasmid pSMS650 The human c-K4A-ras gene can be PCRed from a human cerebral cortex cDNA library (Clontech) using the following oligo- nucleotide primers.

Sense strand: 5' -GAGAGGTACCGCCACCATGACTGAATATAAACTTGTGG-3' (SEQ.ID.NO.: 23) Antisense strand: 5- <BR> <BR> <BR> <BR> CTCTGTCGACAGATTACATTATAATGCATTTTTTAATTTTCACA C-3' (SEQ.ID.NO.: 24) The primers will amplify a c-K4A-Ras encoding DNA fragment with the primers contributing an optimized 'Kozak' translation start sequence, a KpnI site at the N-terminus and a Sal I stite at the C-terminal end. After trimming the ends of the PCR product with Kpn I and Sal I, the c-K-ras4A fragment can be ligated into a KpnI -Sal I cut mutagenesis vector pAlter-l (Promega). Mutation of cysteine-12 to a valine can be accomplished using the manufacturer's protocols and the following oligonucleotide: 5' -GTAGTTGGAGCTGTTGGCGTAGGC-3' (SEQ ID .NO.: 25) After selection and sequencing for the correct nucleotide substitution, the mutated c-K4A-ras-Val-12 can be excised from the pAlter-l vector, using KpnI and Sal I, and be directly ligated into the vector pCI (Promega) which has been digested with KpnI and Sal I.

The new recombinant plasmid, pSMS650, will constitutively transcribe c-K4A-ras-Val-12 from the CMV promoter of the pCI vector.

SEAP assay

Human C33A cells (human epitheial carcenoma - ATTC collection) are seeded in 10cm tissue culture plates in DMEM + 10% fetal calf serum + 1X Pen/Strep + 1X glutamine + 1X NEAA. Cells are grown at 370C in a 5% CO2 atmosphere until they reach 50-80% of confluency.

The transient transfection is performed by the CaPO4 method (Sambrook et al., 1989). Thus, expression plasmids for H-ras, N-ras, K-ras, Myr-ras or H-ras-CVLL are co-precipitated with the DSE-SEAP reporter construct. (A ras expression plasmid is not included when the cell is transfected with the pCMV-SEAP plasmid.) For 10cm plates 600CL1 u1 of CaCi2 -DNA solution is added dropwise while vortexing to 600R1 of 2X HBS buffer to give 1.2ml of precipitate solution (see recipes below). This is allowed to sit at room temperature for 20 to 30 minutes. While the precipitate is forming, the media on the C33A cells is replaced with DMEM (minus phenol red; Gibco cat.

No. 31053-028)+ 0.5% charcoal stripped calf serum + 1X (Pen/Strep, Glutamine and nonessential aminoacids). The CaPO4-DNA precipitate is added dropwise to the cells and the plate rocked gently to distribute.

DNA uptake is allowed to proceed for 5-6 hrs at 370C under a 5% CO2 atmosphere.

Following the DNA incubation period, the cells are washed with PBS and trypsinized with lml of 0.05% trypsin. The 1 ml of trypsinized cells is diluted into 10ml of phenol red free DMEM + 0.2% charcoal stripped calf serum + 1X (Pen/Strep, Glutamine and NEAA).

Transfected cells are plated in a 96 well microtiter plate (100R1/well) to which drug, diluted in media, has already been added in a volume of 100µl. The final volume per well is 200R1 with each drug concentration repeated in triplicate over a range of half-log steps.

Incubation of cells and drugs is for 36 hrs at 370 under CO2. At the end of the incubation period, cells are examined micro- scopically for evidence of cell distress. Next, 100µl of media containing the secreted alkaline phosphatase is removed from each well and trans-

ferred to a microtube array for heat treatment at 650C for 1 hr to inactivate endogenous alkaline phosphatases (but not the heat stable secreted phosphatase).

The heat treated media is assayed for alkaline phosphatase by a luminescence assay using the luminescence reagent CSPD (Tropix, Bedford, Mass.). A volume of 50 1 media is combined with 200 µl of CSPD cocktail and incubated for 60 minutes at room temperature. Luminesence is monitored using an ML2200 microplate luminometer (Dynatech). Luminescence reflects the level of activation of the fos reporter construct stimulated by the transiently expressed protein.

DNA-CaPO4 precipitate for 10cm. plate of cells Ras expression plasmid (1µg/µl) 10R1 DSE-SEAP Plasmid (1µg/µl) 2µl Sheared Calf Thymus DNA (1 g/l) 8µl 2M CaCl2 74µl dH2O 506R1 2X HBS Buffer 280mM NaCl 10mM KC1 1.5mM Na2HPO4 2H2O 12mM dextrose 50mM HEPES Final pH = 7.05 Luminesence Buffer (26ml) Assay Buffer 20ml Emerald ReagentTM (Tropix) 2.5ml 100mM homoarginine 2.5ml CSPD Reagent# (Tropix) 1.0ml Assav Buffer

Add 0.05M Na2CO3 to 0.05M NaHCO3 to obtain pH 9.5.

Make lmM in MgC12 EXAMPLE 14 Alternate Expression Plasmids Cloning of a Myristylated c-H-ras expression plasmid pSMS621 A myristylated c-H-ras-Leu-61 expression plasmid can be cloned by PCR from plasmid pSMS620 (above) using the following primers: Sense strand: S' TCTCGAATTCGCCACCATGGGGAGTAGCAAGAGCAAGCCTAA <BR> <BR> <BR> <BR> GGACCCCAGCCAGCGCCGGATGACGGAATATAAGCTGGTGG 3, (SEQ.ID.NO.: 26).

Antisense strand: 5' -GAGAGTCGACGCGTCAGGAGAGCACAGACTTGC-3' (SEQ.ID.NO.: 27) A sequence encoding the first 15 aminoacids of the v-src gene, containing a myristylation site, is incorporated into the \sense strand oligo. The sense strand oligo also optimizes the 'Kozak' translation initiation sequence immediately 5' to the ATG start site.To prevent prenylation at the v-Ras C-terminus, cysteine 186 would be mutated to a serine by substituting a G residue for a C residue in the C-terminal antisense oligo. The PCR primer oligos introduce an EcoRI site at the 5' end and a Sal I site at the 3'end. The EcoRI-Sal I fragment can be ligated into the mammalian expression plasmid pCI (Promega) cut with EcoRI and Sal I. This results in a plasmid, pSMS621 in which the recombinant myr-c-H-ras-Ser-186 gene is constitutively transcribed from the CMV promoter of the pCI vector.

Cloning of a c-H-ras-CVLL expression plasmid pSMS622

A c-H-ras clone with a C-terminal sequence encoding the amino acids CVLL can be cloned from the plasmid pSMS620 (above) by PCR using the following oligos.

Sense strand: 5'-GAGAGAATTCGCCACCATGACGGAATATAAGCTGGTGG-3' (SEQ.ID.NO.: 28) Antisense strand: 5'-GAGAGTCGACGCGTCAGAGGAGCACACACTTGC-3' (SEQ.ID.NO.: 29) The sense strand oligo optimizes the 'Kozak' sequence and adds an EcoRI site. The antisense strand mutates serine 189 to leucine and adds a Sal I site. The PCR fragment can be trimmed with EcoRI and Sal I and ligated into the EcoRI-Sal I cut vector pCI (Promega).

This results in a plasmid, pSMS622 in which the mutated c-H-ras- Leu61-CVLL gene is constitutively transcribed from the CMV promoter of the pCI vector.

EXAMPLE 15 In vivo tumor growth inhibition assay (nude mouse) In vivo efficacy as an inhibitor of the growth of cancer cells may be confirmed by several protocols well known in the art.

Examples of such in vivo efficacy studies are described by N. E. Kohl et al. (Nature Medicine, 1:792-797 (1995)) and N. E. Kohl et al. (Proc.

Nat. Acad. Sci. U.S.A., 91:9141-9145 (1994)).

Rodent fibroblasts transformed with oncogenically mutated human Ha-ras or Ki-ras (106 cells/animal in 1 ml of DMEM salts) are injected subcutaneously into the left flank of 8-12 week old female nude mice (Harlan) on day 0. The mice in each oncogene group are randomly assigned to a vehicle, compound or combination treatment group. Animals are dosed subcutaneously starting on day 1 and daily for the duration of the experiment. Alternatively, the farnesyl-protein transferase inhibitor may be administered by a continuous infusion pump. Compound, compound combination or vehicle is delivered in a total volume of 0.1 ml. Tumors are excised and weighed when all of the vehicle-treated animals exhibited lesions of 0.5 - 1.0 cm in diameter, typically 11-15 days after the cells were injected. The average weight of the tumors in each treatment group for each cell line is calculated.