Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
INHIBITORS OF PRENYL-PROTEIN TRANSFERASE
Document Type and Number:
WIPO Patent Application WO/2001/051125
Kind Code:
A1
Abstract:
The present invention is directed to peptidomimetic macrocyclic compounds which inhibit prenyl-protein transferase and the prenylation of the oncogene protein Ras. The invention is further directed to chemotherapeutic compositions containing the compounds of this invention and methods for inhibiting prenyl-protein transferase and the prenylation of the oncogene protein Ras.

Inventors:
DESOLMS S JANE (US)
MACTOUGH SUZANNE C (US)
SHAW ANTHONY W (US)
Application Number:
PCT/US2001/000632
Publication Date:
July 19, 2001
Filing Date:
January 09, 2001
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
MERCK & CO INC (US)
DESOLMS S JANE (US)
MACTOUGH SUZANNE C (US)
SHAW ANTHONY W (US)
International Classes:
A61P9/04; A61P13/12; A61P35/00; C07D498/08; C07D515/08; (IPC1-7): A61P35/00; A61K31/4985; C07D515/18; C07D498/18
Domestic Patent References:
WO2000001701A12000-01-13
Attorney, Agent or Firm:
MERCK & CO., INC. (NJ, US)
Muthard, David A. (INC. 126 EAST LINCOLN AVENUE RAHWAY, NEW JERSEY, US)
MERCK & CO., INC. (NJ, US)
Download PDF:
Claims:
WHAT IS CLAIMED IS :
1. A compound of the formula A : wherein : Rla, Rlb Rlc and Rld are independently selected from : a) hydrogen, b) aryl, heterocycle, C3C10 cycloalkyl, R10O, R11S(O)m, R10C(O)NR10, (R10)2NC(O), CN, NO2, (R10)2NC(NR10), R10C(O), R10OC(O),N(R10)2, or R11 OC(O)NR10, c) unsubstituted or substituted ClC6 alkyl, unsubstituted or substituted C2C6 alkenyl or unsubstituted or substituted C2C6 alkynyl, wherein the substituent on the substituted C1C 6 alkyl, substituted C2C6 alkenyl or substituted C2C6 alkynyl is selected from unsubstituted or substituted aryl, heterocyclic, C3Clo cycloalkyl, C2C6 alkenyl, C2 C6 alkynyl, RlOp, R11S(O)m, R10C(O)NR10, (R10)2NC(O), CN, (R10)2NC(NR10), R10C(O), R10OC(O), N(R10)2, and RlloC (o)NRl0 or two Rias, two Rlbs or two Rl cs, on the same carbon atom may be combined to form (CH2) t ; R2a, R2b, R3a and R3b are independently selected from H ; unsubstituted or substituted Cl8 alkyl, unsubstituted or substituted C28 alkenyl, unsubstituted or substituted C28 alkynyl, unsubstituted or substituted aryl, unsubstituted or substituted heterocycle, wherein the substituted group 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, 2) C36 cycloalkyl, 3) OR6, 4) SR4, S (O) R4, S02R4, 5)NR6R7 15) N3, or 16) F ; or R2a and R3a are attached to the same carbon atom and are combined to form (CH2) u wherein one of the carbon atoms is optionally replaced by a moiety selected from O, S (O) m,NC (O), andN (COR10); and R2a and R3a are optionally attached to the same carbon atom ; R4 is selected from C14 alkyl, C36 cycloalkyl, heterocycle, aryl, unsubstituted or substituted with : a) C14 alkoxy, b) aryl or heterocycle, c) halogen, d) HO, f)SO2R11 g) (R10)2, or h) C14 perfluoroalkyl ; R5, R6 and R7 are independently selected from : 1) hydrogen, 2) RI OC (O), or R10OC (O), and 3) C1C6 alkyl, C2C6 alkenyl, C2C6 alkynyl, C3_6 cycloalkyl, heterocycle, aryl, aroyl, heteroaroyl, arylsulfonyl, heteroarylsulfonyl, unsubstituted or substituted with one or more substituents selected from : a) R100, b) aryl or heterocycle, c) halogen, f) SO2R11 , g) N (R10)2, h) C36 cycloalkyl i) C1C6 perfluoroalkyl, j) (R10)2NC(NR10), k) R10OC(O), R11OC(O)NR10, m) CN, and n) N02 ; or R6 and R7 may be joined in a ring ; and independently, R5 and R7 may be joined in a ring ; R8 is independently selected from : a) hydrogen, b) unsubstituted or substituted aryl, unsubstituted or substituted heterocycle, C3Clo cycloalkyl, C2C6 alkenyl, C2C6 alkynyl, ClC6 perfluoroalkyl, F, Cl, Br, R120, R11S (O) m, RlC (O) NRl0, (R10)2NC(O), R102NC(NR10), CN, N02, R10C(O), R10OC(O), N(R10)2, or R11OC(O)NR10, and c) C IC6 alkyl unsubstituted or substituted by unsubstituted or substituted aryl, unsubstituted or substituted heterocycle, C3C10 cycloalkyl, C2C6 alkenyl, C2C6 alkynyl, CiC6 perfluoroalkyl, F, Cl, Br, R10O, R11S(O)m, R10C(O)NH, (R10)2NC(O), R102N C(NR10), CN, R10C(O), R10OC(O), N(R10)2, or R10OC(O)NH; R9 is selected from : a) hydrogen, b) C2C6 alkenyl, C2C6 alkynyl, perfluoroalkyl, F, Cl, Br, R10O, R11S(O)m, R10C(O)NR10, (R10)2NC(O), R102NC(NR10), CN, NO2, R10C(O), R10OC(O), N(R10)2, or R11OC(O)NR10, and c) ClC6 alkyl unsubstituted or substituted by C1C6 perfluoroalkyl, F, Cl, Br, R10O, R11S(O) m, R10C(O)NR10, (R10)2NC(O), R102N C (KR. 10), CN, RlOC (O), ROC (O),N (RlO) 2, or R11OC(O)NR10; R10 is independently selected from hydrogen, ClC 6 alkyl, benzyl, unsubstituted or substituted aryl and unsubstituted or substituted heterocycle ; RI 1 is independently selected from ClC6 alkyl unsubstituted or substituted aryl and unsubstituted or substituted heterocycle ; R12 is independently selected from hydrogen, C1C6 alkyl, ClC3 perfluoroalkyl, unsubstituted or substituted benzyl, unsubstituted or substituted aryl, unsubstituted or substituted heterocycle, and C1C6 alkyl substituted with unsubstituted or substituted aryl or unsubstituted or substituted heterocycle ; A1 is selected from a bond,C (O),C (O) NR10,NRl OC (o), O,N (R10)), S 2N(R10), N(R10)S(O)2, and S (O) m ; A2 is selected from a bond,C (O),C (O) NR10,NR10C (O), O,N (R10), S (0) 2N(R10), N(R10)S(O)2, NR10C(O)NR10, S(O) m andC (Rlc) 2 ; G1, G2 and G3 are independently selected from H2 and O ; W is heterocycle ; V is selected from : a) heterocycle, and b) aryl ; X is selected from : a bond, C(O), C(O)NR10, NR10C(O), NR10C(O)O, O C(O)NR10, NR10C(O)NR10, O, N(R10), S(O)2N(R10), N(R10)S(O)2 and S (O) m ; Xl is selected from : a bond,C (O),NR1OC (O),N (R10) S (0) 2 and S (O) 2 ; Y is selected from a bond,C (=O),C (O) NR10, C(O)O, orS (=O) m ; Zl is selected from unsubstituted or substituted aryl and unsubstituted or substituted heterocycle, wherein the substituted aryl or substituted heterocycle is substituted with one or more of : 1) C18 alkyl, C28 alkenyl or C28 alkynyl, unsubstituted or substituted with : a) C1 4 alkoxy, b) NR6R7, c) C36 cycloalkyl, d) aryl or heterocycle, e) HO, f)S mR4, g)C (O) NR6R7, or h) Cl4perfluoroalkyl ; 2) substituted or unsubstituted aryl or substituted or unsubstituted heterocycle, 3) halogen, 4) OR6, 5) NR6R7, 6) CN, 7) No2, 8) CF3 ; 9)S mR4, 10) OS(O)2R4, 11)C (O) NR6R7, 12)C (O) OR6, or 13) C3C6 cycloalkyl ; Z2 is selected from a bond, unsubstituted or substituted aryl and unsubstituted or substituted heterocycle, wherein the substituted aryl or substituted heterocycle is substituted with one or more of : 1) C18 alkyl, C28 alkenyl or C28 alkynyl, unsubstituted or substituted with : a) C1 4 alkoxy, b) NR6R7, c) C36 cycloalkyl, d) aryl or heterocycle, e) HO, f)S mR4,R g)C (O) NR6R7, or h) Cl4perfluoroalkyl ; 2) substituted or unsubstituted aryl or substituted or unsubstituted heterocycle, 3) halogen, 4) OR6, 5) NR6R7, 6) CN, 7) N02, 8) CF3, 9)S (0) mR, 10) OS(O) 2R4, 11)C (O) NR6R7, 12)C (O) OR6, or 13) C3C6 cycloalkyl ; m is 0, 1 or 2 ; n is 0, 1, 2, 3 or 4 ; p is independently 0, 1, 2, 3 or 4 ; q is 1 or 2 ; r is 0 to 5 ; s is independently 0, 1, 2 or 3 ; tis2, 3, 4, 5 or 6 ;, and u is 4 or 5 ; or a pharmaceutically acceptable salt or stereoisomer thereof.
2. The compound according to Claim 1 of the formula B : B wherein : Ris, R1b and R1c is independently selected from : a) hydrogen, b) aryl, heterocycle, C3Clo cycloalkyl, R10O,N (R10) 2 or C2C6 alkenyl, and c) ClC6 alkyl unsubstituted or substituted by aryl, heterocycle, C3Cl 0 cycloalkyl, C2C6 alkenyl, R10O, orN (RlO) 2 ; Rld is selected from : a) hydrogen, b) unsubstituted or substituted aryl, unsubstituted or substituted heterocycle, C3C10 cycloalkyl, Rl0O, RlOC (O) NRl0, (R10) 2N C(O) NR10, N(R10)2, or Rl 1 OC (O) NRI 0, c) unsubstituted or substituted C1C 6 alkyl, unsubstituted or substituted C2C6 alkenyl or unsubstituted or substituted C2C6 alkynyl, wherein the substituent on the substituted ClC6 6 alkyl, substituted C2C6 alkenyl or substituted C2C6 alkynyl is selected from unsubstituted or substituted aryl, heterocyclic, C3C10 cycloalkyl, C2C6 alkenyl, C2 C6 alkynyl, ClC6 perfluoroallcyl, halogen, RloO, R4S (O) m, R4S(O)2NR10, R10C(O)NR10, (R10)2NC(O), CN, (R10)2N C(NR10), R10C(O), R10OC(O), N(R10)2, and R11OC(O)NR10; R3a is selected from H and CH3 ; R2a is selected from H ; and C1 5 alkyl, unbranched or branched, unsubstituted or substituted with one or more of : 1) aryl, 2) heterocycle, 3) OR6, 4) SR4, S02R4, or and any two of R2a and R3a are optionally attached to the same carbon atom ; R4 is selected from : C14 alkyl and C3_6 cycloalkyl, unsubstituted or substituted with : a) Cl4 alkoxy, b) halogen, or c) aryl or heterocycle ; R6 and R7 are independently selected from H ; C16 alkyl, C3_6 cycloalkyl, heterocycle, aryl, aroyl, heteroaroyl, arylsulfonyl, heteroarylsulfonyl, unsubstituted or substituted with one or two : a) C14 alkoxy, b) aryl or heterocycle, c) halogen, d) HO, f)S02R" g) N 0) 2, or h) C36 cycloalkyl ; R8 is independently selected from : a) hydrogen, b) unsubstituted or substituted aryl, ClC6 alkyl, C2C6 alkenyl, C2C6 alkynyl, ClC6 perfluoroalkyl, F, Cl, R120, RIOC (O) NRl0, CN, NO2, (R10)2NC(NR10), R10C(O), N(R10)2, or R11OC(O)NR10, and c) C1C6 alkyl substituted by : unsubstituted or substituted aryl, ClC6 perfluoroalkyl, R10O, R10C(O)NR10, (R10)2NC(NR0), R10C(O), N(R10)2, or R11OC(O)NR10; R9a is selected from hydrogen, C1C6 alkyl and ClC6 perfluoroalkyl ; RIO is independently selected from hydrogen, C1C6 alkyl, benzyl and unsubstituted or substituted aryl ; Rl l is independently selected from C 1C6 alkyl and unsubstituted or substituted aryl ; R12 is independently selected from hydrogen, C1C6 alkyl, unsubstituted or substituted benzyl, unsubstituted or substituted aryl, unsubstituted or substituted heterocycle, and ClC6 alkyl substituted with unsubstituted or substituted aryl or unsubstituted or substituted heterocycle ; A1 is selected from a bond,N (R10), S (O) m and O ; G1, G2 and G3 are independently selected from H2 and O ; V is selected from : a) heterocycle selected from pyridinyl, pyridonyl, 2oxopiperidinyl, indolyl, quinolinyl and isoquinolinyl, and b) aryl ; X is selected from a bond,C (=O), NR10C (O),OC (O) orS (=O) m ; Y is selected from a bond,C (=O),C (O) NR1O,C (O) O, orS (=O) m ; Z I is selected from unsubstituted or substituted aryl or unsubstituted or substituted heterocycle, wherein the substituted aryl or substituted heterocycle is independently substituted with one or two of : 1) C18 alkyl, C28 alkenyl or C28 alkynyl, unsubstituted or substituted with : a) C14 alkoxy, b) NR6R7, c) C36 cycloalkyl, d) aryl or heterocycle, e) HO, f)S (0) mR, g)C (O) NR6R7, or h) C14 perfluoroalkyl ; 2) substituted or unsubstituted aryl or substituted or unsubstituted heterocycle, 3) halogen, 4) OR6, 5) NR6R7, 6) CN, 7) NO2, 8) CF3, 9)S mR4, 10)OS (0) 2R4, 11)C (O) NR6R7, 12)C (O) OR6, or 13) C3C6 cycloalkyl ; mis 0, 1 or 2 ; n is 0, 1, 2, 3 or 4 ; pis0, 1, 2, 3 or 4 ; r is 0 to 5 ; and s is independently 0, 1, 2 or 3 ; or a pharmaceutically acceptable salt or stereoisomer thereof.
3. The compound according to Claim 2 of the formula C : wherein : Rl a, Rlb and Rlc is independently selected from : a) hydrogen, b) aryl, heterocycle, C3Clo cycloalkyl, R10O, N (R10) 2 or C2C6 alkenyl, and c) C1C6 alkyl unsubstituted or substituted by aryl, heterocycle, C3C10 cycloalkyl, C2C6 alkenyl, R1OO, orN (R10)2; Rad ils selected from : a) hydrogen, b) aryl, heterocycle, C3Clo cycloalkyl, R10O,N (R10) 2 or C2C6 alkenyl, and c) C1C6 alkyl, C2C6 alkenyl or C2C6 alkynyl, unsubstituted or substituted by aryl, heterocycle, C3C10 cycloalkyl, C2C6 alkenyl, Riot, orN (R10) 2 ; R3a is selected from H and CH3 ; R2a is selected from H ; and C1 5 alkyl, unbranched or branched, unsubstituted or substituted with one or more of : 1) aryl, 2) heterocycle, 3) OR6, 4) SR4, S02R4, or and any two of R2a and R3a are optionally attached to the same carbon atom ; R4 is selected from C14 alkyl and C36 cycloalkyl, unsubstituted or substituted with : a) C14 alkoxy, b) halogen, or c) aryl or heterocycle ; R6 and R7 are independently selected from : a) hydrogen, b) C1C6 alkyl, C2C6 alkenyl, C2C6 alkynyl, R10C(O) or R10OC (O) and c) C 1C6 alkyl substituted by C1C6 perfluoroalkyl, R10O, R10C(O)NR10, (R10)2NC(NR10), R10C(O), R10OC(O), N (Rl0) 2) or Rl lOC (O) NRl0; R8 is independently selected from : a) hydrogen, b) unsubstituted or substituted aryl, C1C6 alkyl, C2C6 alkenyl, C2C6 alkynyl, C1C6 perfluoroalkyl, F, Cl, R120, R1OC (O) NRl0, CN, NO2, (R10)2NC(NR10), R10C(O), N(R10)2, or R11OC(O)NR10, and c) C 1C6 alkyl substituted by unsubstituted or substituted aryl, C1C6 perfluoroalkyl, Rio, RlOC (O) NR10, (R10)2NC(NR10), R10C(O), N(R10)2, or R11OC(O)NR10; R9a is selected from hydrogen, ClC6 alkyl and C1C6 perfluoroalkyl ; RI and R12 are independently selected from hydrogen, C1C6 alkyl, benzyl and unsubstituted or substituted aryl ; RI I is independently selected from C1C6 alkyl and unsubstituted or substituted aryl ; Al is selected from a bond,N (Rl0), S (O) m and O ; X is selected from a bond,C (=O), NR10C (O),OC (O) orS (=O) m ; Y is selected from a bond,C (=O),C (O) NR10,C (O) O, orS (=O) m ; Zl is selected from unsubstituted or substituted aryl or unsubstituted or substituted heterocycle, wherein the substituted aryl or substituted heterocycle is substituted with one or two of : 1) C1 4 alkyl, unsubstituted or substituted with : a) C14 alkoxy, b) NR6R7, c) C36 cycloalkyl, d) aryl or heterocycle, e) HO, f)S (O) mR4, or C(O) 2) aryl or heterocycle, 3) halogen, 4) OR6, 5) NR6R7, 6) CN, 7) NO2, 8) CF3, 9)S (0) mR4, 10)C (O) NR6R7, or 11) C3C6 cycloalkyl ; mis 0, 1 or 2 ; n is 0, 1, 2, 3 or 4 ; p is 0, 1, 2, 3 or 4 ; risOto5 ; and s is independently 0, 1, 2 or 3 ; or a pharmaceutically acceptable salt or stereoisomer thereof.
4. The compound according to Claim 3 of the formula D : wherein : Rlb and R1c is independently selected from : a) hydrogen, b) aryl, heterocycle, C3C10 cycloalkyl, R10O or N(R10)2, and c) C 1C6 alkyl unsubstituted or substituted by aryl, heterocycle, C3C10 cycloalkyl, C2C6 alkenyl, R10O, orN (R10) 2 ; Rld is selected from : a) hydrogen, b) aryl, heterocycle, C3Clp cycloalkyl, R10OorN (Rl0) 2, and c) ClC6 6 alkyl, C2C6 alkenyl or C2C6 alkynyl, unsubstituted or substituted by aryl, heterocycle, C3C10 cycloalkyl, C2C6 alkenyl, R1OO, orN (RlO) 2 ; R3a is selected from H and CH3 ; R2a is selected from H ; and C15 alkyl, unbranched or branched, unsubstituted or substituted with one or more of : 1) aryl, 2) heterocycle, 3) OR6, 4) SR4, S02R4, or and any two of R2 and R3 are optionally attached to the same carbon atom ; R4 is selected from C14 alkyl and C36 cycloalkyl, unsubstituted or substituted with : a) C1 4 alkoxy, b) halogen, or c) aryl or heterocycle ; R6 and R7 are independently selected from H ; C 16 alkyl, C36 cycloalkyl, aryl, aroyl, arylsulfonyl, unsubstituted or substituted with one or two : a) C16 alkoxy, b) aryl, c) halogen, d) HO, SO2R11 , g) N (R10) 2 h) C36 cycloalkyl ; or R8 is independently selected from : a) hydrogen, b) unsubstituted or substituted aryl, C1C6 6 alkyl, C2C6 alkenyl, C2C6 alkynyl, ClC perfluoroalkyl, F, Cl, R120, RlOC (O) NRlO, CN, N02, (R10)2NC(NR10), R10C(O), N(R10)2, or R11OC(O)NR10, and c) C1C6 alkyl substituted by unsubstituted or substituted aryl, C lC6 perfluoroalkyl, R10O, R10C(O)NR10, (R10)2NC(NR10) Rl 0C(O), N(R10)2, or R11OC(O)NR10; R9a is selected from hydrogen, C1C6 alkyl and ClC6 perfluoroalkyl ; RI and Rl are independently selected from hydrogen, C1C6 alkyl, benzyl and unsubstituted or substituted aryl ; Rl I is independently selected from C 1C6 alkyl and unsubstituted or substituted aryl ; Al is selected from a bond,N (R10), S (O) m and O ; X is selected from a bond,C (=O),NRl OC (O),OC (O) orS (=O) m ; Y is selected from a bond, C(=O), C(O)NR10, C(O)O, orS (=O) m ; Zl is selected from unsubstituted or substituted aryl or unsubstituted or substituted heterocycle, wherein the substituted aryl or substituted heterocycle is substituted with one or two of : 1) C18 alkyl, C2 8 alkenyl or C28 alkynyl, unsubstituted or substituted with : a) C14 alkoxy, b) NR6R7, c) C36 cycloalkyl, d) aryl or heterocycle, e) HO, f)S mR4, g)C (O) NR6R7, or h) Cl4 perfluoroalkyl ; 2) substituted or unsubstituted aryl or substituted or unsubstituted heterocycle, 3) halogen, 4) OR6, 5) NR6R7, 6) CN, 7) NO2, 8) CF3, <BR> <BR> <BR> 9)S (0) mR4,<BR> <BR> <BR> <BR> <BR> 10)OS (0) 2R4,<BR> <BR> <BR> <BR> <BR> <BR> 11)C (O) NR6R7, 12)C (O) OR6, or 13) C3C6 cycloalkyl ; m is 0, 1 or 2 ; p is independently 0, 1, 2, 3 or 4 ; r is 0 to 5 ; and s is independently 0, 1, 2 or 3 ; or a pharmaceutically acceptable salt or stereoisomer thereof.
5. The compound according to Claim 3 of the formula E : wherein : Rlb and Rl is independently selected from : a) hydrogen, b) aryl, heterocycle, cycloalkyl, R10O, N(R10)2 or C2C6 alkenyl, and c) C1C6 alkyl unsubstituted or substituted by aryl, heterocycle, cycloalkyl, alkenyl, R100, orN (R10) 2 ; Rld is selected from : a) hydrogen, b) aryl, heterocycle, C3Clo cycloalkyl or C2C6 alkenyl, and c) C1C6 alkyl, C2C6 alkenyl or C2C6 alkynyl, unsubstituted or substituted by aryl, heterocycle, C3Clp cycloalkyl, C2C6 alkenyl, R10O, orN (R10) 2 ; R3a is selected from H and CH3 ; R2a is selected from H ; and C 15 alkyl, unbranched or branched, unsubstituted or substituted with one or more of : 1) aryl, 2) heterocycle, 3) OR6, 4) SR4, S02R4, or and any two of R2a and R3a are optionally attached to the same carbon atom ; R4 is selected from C14 alkyl and C36 cycloalkyl, unsubstituted or substituted with : a) C14 alkoxy, b) fluoro, or c) aryl or heterocycle ; R6 and R7 are independently selected from H ; C1 6 alkyl, C36 cycloalkyl, aryl, aroyl, arylsulfonyl, unsubstituted or substituted with one or two : a) C1 4 alkoxy, b) aryl, c) halogen, d) HO, f) SO2R11 g) N (R10)2, or h) C36 cycloalkyl, R8 is independently selected from : a) hydrogen, b) unsubstituted or substituted aryl, C1C6 alkyl, C2C6 alkenyl, C2C6 alkynyl, C1C6 perfluoroalkyl, F, Cl, R120, ROC (O) NRl0, CN, N02, (R10)2NC(NR10), R10C(O), N(R10)2, or R11OC(O)NR10, and c) C1C6 alkyl substituted by unsubstituted or substituted aryl, C1C6 perfluoroalkyl, R10O, R10CF(O)NR10, (R10)2NC(NR10), R10C(O), N(R10)2, or R11 OC(O)NR10; R9a is selected from hydrogen, C1C6 alkyl and C1C6 perfluoroalkyl ; RiO and Rl are independently selected from hydrogen, C1C6 alkyl, benzyl and unsubstituted or substituted aryl ; R11 is independently selected from CiC6 alkyl and unsubstituted or substituted aryl ; Al is selected from a bond,N (RI 0), S (O) m and O ; X is selected from a bond,C (=O),NR10C (O),OC (O) orS (=O) m ; Y is selected from a bond, C(=O), C(O)NR10, C(O)O, orS (=O) m ; Zl is selected from unsubstituted or substituted aryl or unsubstituted or substituted heterocycle, wherein the substituted aryl or substituted heterocycle is substituted with one or two of : 1) C18 alkyl, C28 alkenyl or C28 alkynyl, unsubstituted or substituted with : a) C14 alkoxy, b) NR6R7, c) C36 cycloalkyl, d) aryl or heterocycle, e) HO, f)S mR4, g)C (O) NR6R7, or h) Cl4 perfluoroalkyl ; 2) substituted or unsubstituted aryl or substituted or unsubstituted heterocycle, 3) halogen, 4) OR6, 5) NR6R7, 6) CN, 7) NO2, 8) CF3, 9)S mR4, <BR> <BR> 10)OS (0) 2R4,<BR> <BR> <BR> <BR> 11)C (O) NR6R7, 12)C (O) OR6, or 13) C3C6 cycloalkyl ; m is 0, 1 or 2 ; p is independently 1, 2, 3 or 4 ; r is 0 to 5 ; and s is independently 0, 1, 2 or 3 ; or a pharmaceutically acceptable salt or stereoisomer thereof.
6. A compound which is selected from : 14amino14(1methyl1Himidazol5yl)8oxa2thia1, 18 diazatetracyclo [16. 2. 2. 1"'. 19vl3] tetracosa3 (24), 4, 6, 9 (23), 10, 12hexaene10 carbonitrile 2, 2dioxide ; 14amino4chloro14(1methyl1Himidazol5yl)20oxo8oxa1, 18 diazatetracyclo [16. 2. 2. 13n7. 19,13] tetracosa3 (24), 4, 6, 9 (23), 10, 12hexaene10 carbonitrile ; 18Bromo8 [5 (lmethyllHimidazolyl)]8methyl3, 7 : 1, 17dimetheno11H10, 14 dioxo12, 15ethano16H2oxa9, 10 : 13, 14tetrahydro [9, 12, 15] triazacycloeicosene 4carbonitrile ; 18Chloro8 [5 (methyllHimidazolyl)]8methyl3, 7 : 1, 17dimetheno11H10, 14 dioxo12, 15ethano16H2oxa9, 10 : 13, 14tetrahydro [9, 12, 15] triazacycloeicosene 4carbonitrile ; or a pharmaceutically acceptable salt or stereoisomer thereof.
7. The compound according to Claim 6 which is 14amino14(1methyllHimidazol5yl)8oxa2thia1, 18 diazatetracyclo [16. 2. 2. 13,7.19,13] tetracosa3 (24), 4, 6, 9 (23), 10, 12hexaene10 carbonitrile 2, 2dioxide or a pharmaceutically acceptable salt or stereoisomer thereof.
8. A pharmaceutical composition comprising a pharmaceutical carrier, and dispersed therein, a therapeutically effective amount of a compound of Claim 1.
9. A pharmaceutical composition comprising a pharmaceutical carrier, and dispersed therein, a therapeutically effective amount of a compound of Claim 3.
10. A pharmaceutical composition comprising a pharmaceutical carrier, and dispersed therein, a therapeutically effective amount of a compound of Claim 6.
11. A method for inhibiting prenylprotein transferase which comprises administering to a mammal in need thereof a therapeutically effective amount of a composition of Claim 8.
12. A method for inhibiting prenylprotein transferase which comprises administering to a mammal in need thereof a therapeutically effective amount of a composition of Claim 9.
13. A method for inhibiting prenylprotein transferase which comprises administering to a mammal in need thereof a therapeutically effective amount of a composition of Claim 10.
14. A method for treating cancer which comprises administering to a mammal in need thereof a therapeutically effective amount of a composition of Claim 8.
15. A method for treating cancer which comprises administering to a mammal in need thereof a therapeutically effective amount of a composition of Claim 9.
16. A method for treating cancer which comprises administering to a mammal in need thereof a therapeutically effective amount of a composition of Claim 10.
17. A method for treating neurofibromin benign proliferative disorder which comprises administering to a mammal in need thereof a therapeutically effective amount of a composition of Claim 8.
18. A method for treating blindness related to retinal vascularization which comprises administering to a mammal in need thereof a therapeutically effective amount of a composition of Claim 8.
19. A method for treating infections from hepatitis delta and related viruses which comprises administering to a mammal in need thereof a therapeutically effective amount of a composition of Claim 8.
20. A method for preventing restenosis which comprises administering to a mammal in need thereof a therapeutically effective amount of a composition of Claim 8.
21. A method for treating polycystic kidney disease which comprises administering to a mammal in need thereof a therapeutically effective amount of a composition of Claim 8.
22. A method of conferring radiation sensitivity on a tumor cell using a therapeutically effective amount of a composition of Claim 8 in combination with radiation therapy.
23. A method of using a therapeutically effective amount of a composition of Claim 8 in combination with an antineoplastic to treat cancer.
24. A method according to Claim 23 wherein the antineoplastic is paclitaxel.
25. A pharmaceutical composition made by combining the compound of Claim 1 and a pharmaceutically acceptable carrier.
26. A process for making a pharmaceutical composition comprising combining a compound of Claim 1 and a pharmaceutically acceptable carrier.
Description:
TITLE OF THE INVENTION INHIBITORS OF PRENYL-PROTEIN TRANSFERASE BACKGROUND OF THE INVENTION The Ras proteins (Ha-Ras, Ki4a-Ras, Ki4b-Ras and N-Ras) are part of a signalling 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. LowyandD. M. Willumsen, Ann. Rev. Bioche7n. 62 : 851-891 (1993)). Mutated ras genes (Ha-ras, Ki4a-ras, Ki4b-ras and N-ras) 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.

Ras must be localized to the plasma membrane for both normal and oncogenic functions. At least 3 post-translational modifications 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-Aaal- Aaa2-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 famesyl-protein transferase or geranylgeranyl-protein transferase type I, which catalyze the alkylation of the cysteine residue of the CAAX motif with a C 15 or C20 isoprenoid, respectively. (S. Clare., Ann. Rev. Biochem. 61 : 355-386 (1992) ; W. R. Schafer and J.

Rine, Ann. Rev. Genetics 30 : 209-237 (1992)). The term prenyl-protein transferase may be used to refer generally to farnesyl-protein transferase and geranylgeranyl- protein transferase type I. The Ras protein is one of several proteins that are known to undergo post-translational farnesylation. Other farnesylated proteins include the Ras- related GTP-binding proteins such as Rho, fungal mating factors, the nuclear lamins, and the gamma subunit oftransducin. 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.

Inhibition of farnesyl-protein transferase has been shown to block the growth of 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 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 ras-dependent 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 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 polyisoprenoids including farnesyl pyrophosphate. 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)). Inhibition of farnesyl pyrophosphate biosynthesis by inhibiting HMG-CoA reductase blocks Ras membrane localization in cultured cells. However, direct inhibition of farnesyl- protein transferase would be more specific and attended by fewer side effects than would occur with the required dose of a general inhibitor of isoprene biosynthesis.

Inhibitors of farnesyl-protein transferase (FPTase) have been described in two general classes. The first are analogs of farnesyl diphosphate (FPP), while the second class of inhibitors is related to the 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. Chef., 37, 725 (1994)). In

general, deletion of the thiol from a CAAX derivative has been shown to dramatically reduce the inhibitory potency of the compound. However, the thiol group potentially places limitations on the therapeutic application of FPTase inhibitors with respect to pharmacokinetics, pharmacodynamics and toxicity. Therefore, a functional replacement for the thiol is desirable.

It has recently been reported that farnesyl-protein transferase inhibitors are inhibitors of proliferation of vascular smooth muscle cells and are therefore useful in the prevention and therapy of arteriosclerosis and diabetic disturbance of blood vessels (JP H7-112930).

It has recently been disclosed that certain tricyclic compounds which optionally incorporate a piperidine moiety are inhibitors of FPTase (WO 95/10514, WO 95/10515 and WO 95/10516). Imidazole-containing inhibitors of farnesyl protein transferase have also been disclosed (WO 95/09001 and EP 0 675 112 Al).

It is, therefore, an object of this invention to develop peptidomimetic compounds that do not have a thiol moiety, and that will inhibit prenyl-protein transferase and thus, the post-translational prenylation of proteins. It is a further object of this invention to develop chemotherapeutic compositions containing the compounds of this invention and methods for producing the compounds of this invention.

SUMMARY OF THE INVENTION The present invention comprises peptidomimetic piperazine-containing macrocyclic compounds which inhibit prenyl-protein transferase. Further contained in this invention are chemotherapeutic compositions containing these prenyl-protein transferase inhibitors and methods for their production.

The compounds of this invention are illustrated by the formula A :

A DETAILED DESCRIPTION OF THE INVENTION The compounds of this invention are useful in the inhibition of prenyl- protein transferase and the prenylation of the oncogene protein Ras. In a first embodiment of this invention, the inhibitors of prenyl-protein transferase are illustrated by the formula A :

A wherein : Rla, Rlb, Rlc and Rld are independently selected from : a) hydrogen,

b) aryl, heterocycle, C3-Clo cycloalkyl, R100-, R11S(O)m-, R10C(O)NR10-, (R10)2N-C(O)-, CN, NO2, (R10)2N-C(NR10)-, RlOC (O)-, RlOOC (O)-,-N (Rl0) 2, orRllOC (O) NRl0-, c) unsubstituted or substituted C1-C 6 alkyl, unsubstituted or substituted C2-C6 alkenyl or unsubstituted or substituted C2-C6 alkynyl, wherein the substituent on the substituted C1-C6 alkyl, substituted C2-C6 alkenyl or substituted C2-C6 alkynyl is selected from unsubstituted or substituted aryl, heterocyclic, C3-ClO 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)-, -N(R10)2, and Rl (0)-NRlO-, or two R1as, two Rlbs or two Rocs, on the same carbon atom maybe combined to form -(CH2)t-; R2a, R2b, R3a and R3b are independently selected from H ; unsubstituted or substituted Cl-8 alkyl, unsubstituted or substituted C2-8 alkenyl, unsubstituted or substituted C2-8 alkynyl, unsubstituted or substituted aryl, unsubstituted or substituted heterocycle, wherein the substituted group is substituted with one or more of : 1) aryl or heterocycle, unsubstituted or substituted with : a) C1 4 alkyl, b) (CH2) pOR6, (CH2)pNR6R7, d) halogen, e) CN, 2) C3-6 cycloalkyl, 3) OR6, 4) SR4, S (O) R4, S02R4,

Rua and R3a are attached to the same carbon atom and are combined to form - (CH2) u- wherein one of the carbon atoms is optionally replaced by a moiety selected from O, S (O) m,-NC (O)-, and-N (COR10)-; and R2a and R3a are optionally attached to the same carbon atom ; R4 is selected from C1 4 alkyl, C3-6 cycloalkyl, heterocycle, aryl, unsubstituted or substituted with : a) C1 4 alkoxy, b) aryl or heterocycle, c) halogen, g) N (RlO) 2, or h) C1 4perfluoroalkyl ; R5, R6 and R7 are independently selected from : 1) hydrogen, 2) Rl OC (O)-, or Rl OOC (O)-, and 3) C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3_6 cycloalkyl, heterocycle, aryl, aroyl, heteroaroyl, arylsulfonyl, heteroarylsulfonyl, unsubstituted or substituted with one or more substituents selected from : a) R10O-, b) aryl or heterocycle, c) halogen, R10C(O)NR10-,

f)-SO2R" g) N (R10)2, h) C3-6 cycloalkyl, i) C1-C6 perfluoroalkyl, <BR> <BR> <BR> j) (R10)2N-C(NR10)-,<BR> k) R10OC(O)-, l) R11OC(O)NR10-, m) CN, and n) NO2; or R6 and R7 may be joined in a ring ; and independently, R5 and R7 may be joined in a ring ; R8 is independently selected from : a) hydrogen, b) unsubstituted or substituted aryl, unsubstituted or substituted heterocycle, C3-Clo cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, Cl-C6 perfluoroallcyl, F, Cl, Br, R12O-, R11S(O)m-, R10C(O)NR10-, (R10)2NC(O)-, R102N-C(NR10)-, CN, NO2, R10C (O)-, R10OC(O)-, -N (Rl0) 2, or Rl lOC (O) NRl0-, and c) C 1-C6 alkyl unsubstituted or substituted by unsubstituted or substituted aryl, unsubstituted or substituted heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 perfluoroalkyl, F, Cl, Br, R10O-, R11S(O)m-, R10C(O)NH-, (R10)2NC(O)-, R102N- C(NR10)-, CN, R10C(O)-, R10OC(O)-, -N(R10)2, or R10OC(O)NH-; R9 is selected from : a) hydrogen,

b) C2-C6 alkenyl, C2-C6 alkynyl, perfluoroalkyl, F, Cl, Br, R10O-, R11S(O)m-, R10C(O)NR10-, (R10)2NC(O)-, R102N-C(NR10)-, CN, NO2, R10C(O)-, R10OC(O)-, -N(R10)2, or R11OC(O)NR10-, and c) C 1-C6 alkyl unsubstituted or substituted by C1-C6 perfluoroalkyl, F, Cl, Br, R10O-, R11S(O)m-, R10C(O)NR10-, (R10)2NC(O)-, R102N- C(NR10)-, CN, R10C(O)-, R10OC(O)-, -N(R10)2, or R10 is independently selected from hydrogen, Cl-C6 alkyl, benzyl, unsubstituted or substituted aryl and unsubstituted or substituted heterocycle ; Rl l is independently selected from C 1-C6 alkyl unsubstituted or substituted aryl and unsubstituted or substituted heterocycle ; R12 is independently selected from hydrogen, C1-C6 alkyl, Cl-C3 perfluoroalkyl, unsubstituted or substituted benzyl, unsubstituted or substituted aryl, unsubstituted or substituted heterocycle, and C 1-C6 alkyl substituted with unsubstituted or substituted aryl or unsubstituted or substituted heterocycle ; Al is selected from a bond, -C(O)-, -C(O)NR10-, -NR10C(O)-, O, -N(R10)-, -S 2N(R10)-, -N(R10)S(O)2-, and S(O) m ; A2 is selected from a bond,-C (O)-,-C (O) NR10-, -NR10C(O)-, O, -N(R10)-, -S (0) 2N(R10)-, -N(R10)S(O)2-, -NR10C(O)NR10-, S (O) m and -C(R1c)2-; G1, G2 and G3 are independently selected from H2 and O ; W is heterocycle ; V is selected from : a) heterocycle, and b) aryl ;

X is selected from : a bond, -C(O)-, -C(O)NR10-, -NR10C(O)-, -NR10C(O)-O-, -O- C(O)NR10-, -NR10C(O)NR10-, O, -N(R10)-, -S(O)2N(R10)-, -N(R10)S(O)2- and S (O) m ; Xl is selected from : a bond,-C (O)-, -NR10C (O)-,-N (R10) S (0) 2- and S (O) 2 ; Y is selected from a bond,-C (=O)-,-C (O) NRl0-,-C (O) O-, or-S (=O) m ; zl is selected from unsubstituted or substituted aryl and unsubstituted or substituted heterocycle, wherein the substituted aryl or substituted heterocycle is substituted with one or more of : 1) C1-8 alkyl, C2-8 alkenyl or C2-8 alkynyl, unsubstituted or substituted with : a) C1-4 alkoxy, b) NR6R7, c) C3-6 cycloalkyl, d) aryl or heterocycle, e) HO, f)-S mR4, g)-C (O) NR6R7, or h) C1 4 perfluoroalkyl ; 2) substituted or unsubstituted aryl or substituted or unsubstituted heterocycle, 3) halogen, 4) OR6, 5) NR6R7, 6) CN, 7) NO2, 8) CF3 ; <BR> <BR> <BR> 9)-S (0) mR4,<BR> <BR> <BR> <BR> <BR> <BR> 10)-OS (0) 2R4, 11) -C(O)NR6R7, 12)-C (O) OR6, or 13) C3-C6 cycloalkyl ;

z2 is selected from a bond, unsubstituted or substituted aryl and unsubstituted or substituted heterocycle, wherein the substituted aryl or substituted heterocycle is substituted with one or more of : 1) C1-8 alkyl, C2-8 alkenyl or C2-8 alkynyl, unsubstituted or substituted with : a) C1-4 alkoxy, b) NR6R7, c) C3-6 cycloalkyl, d) aryl or heterocycle, e) HO, f)-S (O) mR4) g)-C (O) NR6R7, or h) C1 4perfluoroalkyl ; 2) substituted or unsubstituted aryl or substituted or unsubstituted heterocycle, 3) halogen, 4) OR6, 5) NR6I 6) CN, 7) NO2, <BR> <BR> <BR> 8) CF3,<BR> <BR> <BR> <BR> <BR> 9)-S (0) mR4, 10) -OS(O)2R4,R 11)-C (O) NR6R7, 12)-C (O) OR6, or 13) C3-C6 cycloalkyl ; m is 0, 1 or 2 ; n is 0, 1, 2, 3 or 4 ; p is independently 0, 1, 2, 3 or 4 ; q is 1 or 2 ; r is 0 to 5 ; s is independently 0, 1, 2 or 3 ;

t is 2, 3, 4, 5 or 6 ; and u is 4 or 5 ; or a pharmaceutically acceptable salt or stereoisomer thereof.

In another embodiment of this invention, the inhibitors of prenyl- protein transferase are illustrated by the formula A : A wherein : Rla, R1b, R1c and R1d are independently selected from : a) hydrogen, b) aryl, heterocycle, C3-C p cycloalkyl, R10-, R1 S (O) m-, R10C(O)NR10-, (R10)2N-C(O)-, CN, NO2, (R10)2N-C(NR10)-, R10C(O)-, R10OC(O)-, -N(R10)2, or R11OC(O)NR10-, c) unsubstituted or substituted Cl-C6 alkyl, unsubstituted or substituted C2-C6 alkenyl or unsubstituted or substituted C2-C6 alkynyl, wherein the substituent on the substituted C1-C6 alkyl, substituted C2-C6 alkenyl or substituted C2-C6 alkynyl is selected from unsubstituted or substituted aryl, heterocyclic, C3-Clp cycloalkyl, C2-C6 alkenyl, C2- C6 alkynyl, RlOO-, R11S(O)m-, R10C(O)NR10-, (R10)2N-C(O)-, CN, (R10)2N-C(NR10)-, R10C(O)-, R10OC(O)-, -N(R10)2, and R1 OC(O)-NR10-; R2a, R2b, R3a and R3b are independently selected from H ; unsubstituted or substituted C1-8 alkyl, unsubstituted or substituted C2 8 alkenyl, unsubstituted or substituted C2-8 alkynyl, unsubstituted or substituted aryl, unsubstituted or substituted heterocycle,

wherein the substituted group 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, 2) C3-6 cycloalkyl, 3) OR6, 4) SR4, S (O) R4, S02R4, 5)-NR6R7

15) N3, or 16) F ; or R2a and R3a are attached to the same C atom and are combined to form- (CH2) u- wherein one of the carbon atoms is optionally replaced by a moiety selected from O, S (O) m,-NC (O)-, and-N (COR10)-; and R2a and R3a are optionally attached to the same carbon atom ; R4 is selected from C1 4 alkyl, C3-6 cycloalkyl, heterocycle, aryl, unsubstituted or substituted with : a) C1 4 alkoxy, b) aryl or heterocycle, c) halogen, d) HO,

- SO2R11 , or g) N (RlO) 2 ; R5, R6 and R7 are independently selected from H ; C1 4 alkyl, C3-6 cycloalkyl, heterocycle, aryl, aroyl, heteroaroyl, arylsulfonyl, heteroarylsulfonyl, unsubstituted or substituted with : a) C1-4 alkoxy, b) aryl or heterocycle, c) halogen, -SO2R11 , or g) (R10)2; or R6 and R7 may be joined in a ring ; and independently, R5 and R7 may be joined in a ring ; R8 is independently selected from : a) hydrogen, b) unsubstituted or substituted aryl, unsubstituted or substituted heterocycle, C3-Clo cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, perfluoroalkyl, F, Cl, Br, R120-, RllS (O) m-, R1C (O) NRl0-,

(R10)2NC(O)-, R102N-C(NR10)-, CN, NO2, R10C(O)-, R10OC(O)-, -N (R10) 2, or Rl 1 OC (O) NRl0-, and c) C1-C6 alkyl unsubstituted or substituted by unsubstituted or substituted aryl, unsubstituted or substituted heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkyl, perfluoroalkyl, F, Cl, Br, R10O-, R11S(O)m-, R10C(O)NH-, (R10)2NC(O)-, R102N-C(NR10)-, CN, R10C(O)-, R10OC(O)-, -N(R10)2, or R10OC(O)NH-; R9 is selected from : a) hydrogen, b) C2-C6 alkenyl, C2-C6 alkynyl, perfluoroalkyl, F, Cl, Br, Rl 0O-, R11S(O)m-, R10C(O)NR10-, (R10)2NC(O)-, R102N-C(NR10)-, CN, N02, RlOC (O)-, RlOOC (O)-,-N (Rl0) 2, or Rl lOC (O) NRl0-, and c) C1-C6 alkyl unsubstituted or substituted by perfluoroalkyl, F, Cl, Br, Rl R11S(O)m-, R10C(O)NR10-, (R10)2NC(O)-, R102N- C (NR_, CN, RlOC (O)-, R10OC (O)-,-N (R10) 2, or R10 is independently selected from hydrogen, Cl-C6 6 alkyl, benzyl, unsubstituted or substituted aryl and unsubstituted or substituted heterocycle ; RI 1 is independently selected from C 1-C6 alkyl unsubstituted or substituted aryl and unsubstituted or substituted heterocycle ; R12 is independently selected from hydrogen, Cl-C6 6 alkyl, Cl-C3 perfluoroalkyl, unsubstituted or substituted benzyl, unsubstituted or substituted aryl, unsubstituted or substituted heterocycle, and C1-C alkyl substituted with unsubstituted or substituted aryl or unsubstituted or substituted heterocycle ; Al is selected from a bond,-C (O)-,-C (O) NR10-,-NR10C (O)-, O,-N (RI 0)-, -S (0) 2N (RlO)-,-N (RlO) S (0) 2-, and S (0) m ; A2 is selected from a bond,-C (O)-,-C (O) NR10-,-NRl OC(O)-, O,-N (R10) -, -S 2N(R10)-, -N(R10)S(O)2-, S(O)m and-C (Rlc) 2- ;

G1, G2 and G3 are independently selected from H2 and O ; W is heterocycle ; V is selected from : a) heterocycle, and b) aryl ; X is selected from : a bond, -C(O)-, -C(O)NR10-, -NR10C(O)-, -NR10C(O)-O-, -O- C(O)NR10-, -NR10C(O)NR10-, O, -N(R10)-, -S(O)2N(R10)-, -N(R10)S(O)2- and S (O) m ; Xl is selected from : a bond,-C (O)-,-NR10C (O)-,-N (R10) S (0) 2- and S (O) 2 ; Y is selected from a bond,-C (=O)-,-C (O) NR10-,-C (O) O-, or-S (=O) m ; Zl is selected from unsubstituted or substituted aryl and unsubstituted or substituted heterocycle, wherein the substituted aryl or substituted heterocycle is substituted with one or more of : 1) C1 4 alkyl, unsubstituted or substituted with : a) C1-4 alkoxy, b) NR6R7, c) C3-6 cycloalkyl, d) aryl or heterocycle, e) HO, f)-S (O) mR4, or ) 2) aryl or heterocycle, 3) halogen, 4) OR6, 5) NR6R7, 6) CN, 7) NO2,

8) CF3, 9)-S mR4, 10)-C (O) NR6R7, or 11) C3-C6cycloallcyl ; Z2 is selected from a bond, unsubstituted or substituted aryl and unsubstituted or substituted heterocycle, wherein the substituted aryl or substituted heterocycle is substituted with one or more of : 1) C1-4 alkyl, unsubstituted or substituted with : a) C 1-4 alkoxy, b) NR6R7, c) C3-6 cycloalkyl, d) aryl or heterocycle, e) HO, f)-S (O) mR4, or ) 2) aryl or heterocycle, 3) halogen, 4) OR6, 5) NR6R7, 6) CN, 7) NO2, 8) CF3, 9)-S (0) mR4, 10)-C (O) NR6R7, or 11) C3-C6 cycloalkyl ; m is 0, 1 or 2 ; nisO, 1, 2, 3 or 4 ; p is independently 0, 1, 2, 3 or 4 ; q is 1 or 2 ; r is 0 to 5 ; s is independently 0, 1, 2 or 3 ; and u is 4 or 5 ;

or a pharmaceutically acceptable salt or stereoisomer thereof.

In a third embodiment of this invention, the inhibitors of prenyl-protein transferase are illustrated by the formula A : A wherein : Ria is selected from hydrogen and C1-C alkyl ; Rlb and Rlc are independently selected from : a) hydrogen, b) aryl, heterocycle, C3-C10 cycloalkyl, R10O-or-N (R10) 2, and c) unsubstituted or substituted C1-C6 alkyl wherein the substituent on the substituted C1-C6 alkyl is selected from unsubstituted or substituted aryl, heterocycle, C3-Clo cycloalkyl, C2-C6 alkenyl, Ro-and -N(R10)2; Rld is selected from : a) hydrogen,

b) unsubstituted or substituted aryl, unsubstituted or substituted heterocycle, C3-Clo cycloalkyl, R100-, RlOC (O) NR10-, (R10)2N- C (o) NRlO-,-N (RlO) 2, orRllOC (0) NRlO-, c) unsubstituted or substituted C1-C6 alkyl, unsubstituted or substituted C2-C6 alkenyl or unsubstituted or substituted C2-C6 alkynyl, wherein the substituent on the substituted C1-C6 6 alkyl, substituted C2-C6 alkenyl or substituted C2-C6 alkynyl is selected from unsubstituted or substituted aryl, heterocyclic, C3-C10 cycloalkyl, C2-C6 alkenyl, C2- C6 alkynyl, Cl-C6 perfluoroalkyl, halogen, R100-, R4S (O) m-, R4S(O)2NR10-, R10C(O)NR10-, (R10)2N-C(O)-, CN, (R10)2N- C(NR10)-, R10C(O)-, R10OC(O)-, -N(R10)2, and R11OC(O)-NR10-; R2b, R3a and R3b are independently selected from H and CH3 ; R2a is independently selected from H ; or C1-5 alkyl, unbranched or branched, unsubstituted or substituted with one or more of : 1) aryl, 2) heterocycle, 3) OR6, 4) SR4, S02R4, or and R2a and R3a are optionally attached to the same carbon atom ; R4 is selected from C1-4 alkyl and C3-6 cycloalkyl, unsubstituted or substituted with : a) C 1-4 alkoxy, b) halogen, or c) aryl or heterocycle ;

R6 and R7 are independently selected from H ; C1-4 alkyl, C3-6 cycloalkyl, aryl and heterocycle, unsubstituted or substituted with : a) C 1-4 alkoxy, b) halogen, or c) aryl or heterocycle ; R8 is independently selected from : a) hydrogen, b) unsubstituted or substituted aryl, unsubstituted or substituted heterocycle, Cl-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 perfluoroalkyl, F, Cl, R12O-, R10C(O)NR10-, CN, N02, (R10) 2N- C(NR10)-, R10C(O)-, -N(R10)2, or R11OC(O)NR10-, and c) C1-C6 alkyl substituted by : unsubstituted or substituted aryl, unsubstituted or substituted heterocycle, C1-C6 perfluoroalkyl, R10O-, R10C(O)NR10-, (R10)2N-C(NR10)-, R10C(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-, Rl 1S(O)m-, R10C(O)NR10-, CN, NO2, (R10)2N-C(NR10)-, RlOC (O)-,-N (R10) 2, or RllOC (O) NR10-, and c) C 1-C6 alkyl unsubstituted or substituted by C1-C6 perfluoroalkyl, F, Cl, R100-, Ri lS (O) m-, RlOC (O) NRl0-, CN, (Rlo) 2N-C (NR10)-, R10C(O)-, -N(R10)2, or R11OC(O)NR10-; R10 is independently selected from hydrogen, C1-C6 6 alkyl, benzyl, unsubstituted or substituted aryl and unsubstituted or substituted heterocycle ; RI l is independently selected from C1-C6 alkyl, unsubstituted or substituted aryl and unsubstituted or substituted heterocycle ; Ru 2 ils independently selected from hydrogen, C1-C6 alkyl, Cl-C3 perfluoroalkyl, unsubstituted or substituted benzyl, unsubstituted or substituted aryl, unsubstituted or

substituted heterocycle, and Cl-C6 alkyl substituted with unsubstituted or substituted aryl or unsubstituted or substituted heterocycle ; Al is selected from a bond,-C (O)-,-C (O) NR10-,-NR10C (O)-, O,-N (R10) -, -S 2N(R10)-, -N(R10)S(O)2-, and S(O) m ; A2 is selected from a bond,-C (O)-,-C (O) NR10-,-NR10C (O)-, O,-N (RI 0)-, -SO) 2N (RlO)-,-NN(Rl0 S (0) 2-, ( ) m and-C (Rlc) 2- ; G1, G2 and G3 are independently selected from H2 and O ; V is selected from : a) heterocycle selected from pyridinyl, pyridonyl, 2-oxopiperidinyl, indolyl, quinolinyl and isoquinolinyl, and b) aryl ; W is a heterocycle selected from imidazolyl, pyridinyl and triazolyl ; X is selected from : a bond, -C(O)-, -C(O)NR10-, -NR10C(O)-, -NR10C(O)-O-, -O- C (O)NR10-, -NR10C(O)NR10-, O, -N(R10)-, -S(O)2N(R10)-, -N(R10)S(O)2- and S(O)m; Xl is selected from : a bond,-C (O)-,-NR10C (O)-,-N (R10) S (0) 2- and S (O) 2 ; Y is selected from a bond,-C (=O)-,-C (O) NRl0-,-C (O) O-, or-S (=O) m ; zl is selected from unsubstituted or substituted aryl or unsubstituted or substituted heterocycle, wherein the substituted aryl or substituted heterocycle is independently substituted with one or two of : 1) C1 4 alkyl, unsubstituted or substituted with : a) C1-4 alkoxy, b) NR6R7, c) C3-6 cycloalkyl, d) aryl or heterocycle,

e) HO, f)-S (O) mR4 or ) -C(O) 2) aryl or heterocycle, 3) halogen, 4) OR6, 5) NR6R7, 6) CN, 7) N02, 8) CF3 ; 9)-S mR4, 10)-C (O) NR6R7, or 11) C3-C6 cycloalkyl ; z2 is selected from a bond, unsubstituted or substituted aryl and unsubstituted or substituted heterocycle, wherein the substituted aryl or substituted heterocycle is substituted independently with one or two of: 1) C1 4 alkyl, unsubstituted or substituted with : a) C1-4 alkoxy, b) NR6R7, c) C3-6 cycloalkyl, d) aryl or heterocycle, e) HO, f)-S (O) mR4, or ) -C(O) 2) aryl or heterocycle, 3) halogen, 4) OR6, 5) NR6R7, 6) CN, 7) N02, 8) CF3, 9)-S (0) mR4, 10)-C (O) NR6R7, or

11) C3-C6 cycloalkyl ; m is 0, 1 or 2 ; nisO, 1, 2, 3 or 4 ; pis0, 1, 2, 3 or 4 ; qis 1 or2 ; r is 0 to 5 ; s is independently 0, 1, 2 or 3 ; and uis4or5 ; or a pharmaceutically acceptable salt or stereoisomer thereof.

In a fourth embodiment of this invention, the inhibitors of prenyl- protein transferase are illustrated by the formula B : B wherein : Rla, Rlb and Rlc is independently selected from : a) hydrogen, b) aryl, heterocycle, C3-Clo cycloalkyl, R100-,-N (R10) 2 or C2-C6 alkenyl, and

c) C 1-C6 alkyl unsubstituted or substituted by aryl, heterocycle, C3-Cl0 cycloalkyl, C2-C6 alkenyl, R10O-, or-N (R10) 2 ; Rld is selected from : a) hydrogen, b) unsubstituted or substituted aryl, unsubstituted or substituted heterocycle, C3-Clo cycloalkyl, R1OO-, R10C (O) NRl0-, (R10) 2N- C(O) NR10-, -N(R10)2, or R11OC(O)NR10-, c) unsubstituted or substituted C1-C6 alkyl, unsubstituted or substituted C2-C6 alkenyl or unsubstituted or substituted C2-C6 alkynyl, wherein the substituent on the substituted C1-C6 alkyl, substituted C2-C6 alkenyl or substituted C2-C6 alkynyl is selected from unsubstituted or substituted aryl, heterocyclic, C3-Clo cycloalkyl, C2-C6 alkenyl, C2- C6 alkynyl, C1-C6 perfluoroalkyl, halogen, R100-, R4S (O) m-, R4S (0) 2NR10-, RlOC (O) NR10-, (Rlo) 2N-C (O)-, CN, (RlO) 2N- C(NR10)-, R10C(O)-, R10OC(O)-, -N(R10)2, and R11 OC(O)-NR10-; R3a is selected from H and CH3 ; R2a is selected from H ; and C1 5 alkyl, unbranched or branched, unsubstituted or substituted with one or more of : 1) aryl, 2) heterocycle, 3) OR6, 4) SR4, S02R4, or and any two of R2a and R3a are optionally attached to the same carbon atom ;

R4 is selected from : C1-4 alkyl and C3-6 cycloalkyl, unsubstituted or substituted with : a) C1-4 alkoxy, b) halogen, or c) aryl or heterocycle ; R6 and R7 are independently selected from H ; Cl-6 alkyl, C3_6 cycloalkyl, heterocycle, aryl, aroyl, heteroaroyl, arylsulfonyl, heteroarylsulfonyl, unsubstituted or substituted with one or two : a) Cl-4 alkoxy, b) aryl or heterocycle, c) halogen, d) HO, -SO2R11 g) N (R10)2, 2, or h) C3-6 cycloalkyl ; R8 is independently selected from : a) hydrogen, b) unsubstituted or substituted aryl, Cl-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, Cl-C6 perfluoroalkyl, F, Cl, R120-, RlOC (O) NR10-, CN, N02, (R10) 2N-C (NR10)-, RlOC -N(R10)2, or R11OC(O)NR10-, and c) C1-C6 alkyl substituted by : unsubstituted or substituted aryl, Cl-C6 perfluoroalkyl, R10-, RIOC (O) NRlO-, (R10) 2N-C (NRl0) R10C(O)-, -N(R10)2, or R11OC(O)NR10-; R9a is selected from hydrogen, C1-C6 alkyl and C1-C6 perfluoroalkyl ;

R10 is independently selected from hydrogen, Cl-C6 alkyl, benzyl and unsubstituted or substituted aryl ; Rl I is independently selected from Cl-C6 alkyl and unsubstituted or substituted aryl ; R12 is independently selected from hydrogen, Cl-C6 alkyl, unsubstituted or substituted benzyl, unsubstituted or substituted aryl, unsubstituted or substituted heterocycle, and C1-C6 alkyl substituted with unsubstituted or substituted aryl or unsubstituted or substituted heterocycle ; A1 is selected from a bond, -N(R10)-, S (O) m and O ; G1, G2 and G3 are independently selected from H2 and O ; V is selected from : a) heterocycle selected from pyridinyl, pyridonyl, 2-oxopiperidinyl, indolyl, quinolinyl and isoquinolinyl, and b) aryl ; X is selected from a bond,-C (=O)-,-NR10C (O)-,-OC (O)- or-S (=O) m ; Y is selected from a bond,-C (=O)-,-C (O) NR10-, -C(O)O-, or-S (=O) m ; Zl is selected from unsubstituted or substituted aryl or unsubstituted or substituted heterocycle, wherein the substituted aryl or substituted heterocycle is independently substituted with one or two of : 1) C1-8 alkyl, C2 g alkenyl or C2-8 alkynyl, unsubstituted or substituted with : a) C1-4 alkoxy, b) NR6R7, c) C3-6 cycloalkyl, d) aryl or heterocycle, e) HO, f)-S mR4,

) $-C(O)NR6R7, or h) C1-4 perfluoroalkyl ; 2) substituted or unsubstituted aryl or substituted or unsubstituted heterocycle, 3) halogen, 4) OR6, 5) NR6R7, 6) CN, 7) N02, 8) CF3, <BR> <BR> 9)-S (0) mR4,<BR> <BR> <BR> <BR> <BR> 10)-OS (0) 2R4,<BR> <BR> <BR> <BR> <BR> <BR> 11)-C (O) NR6R7, 12)-C (O) OR6, or 13) C3-C6 cycloalkyl ; m is 0, 1 or 2 ; n is 0, 1, 2, 3 or 4 ; pis0, 1, 2, 3 or 4 ; r is 0 to 5 ; and s is independently 0, 1, 2 or 3 ; or a pharmaceutically acceptable salt or stereoisomer thereof.

A preferred embodiment of the compounds of this invention is illustrated by the formula C-1 :

C-1 wherein : R1a, R1b and R1c is independently selected from : a) hydrogen, b) aryl, heterocycle, C3-Clo cycloalkyl, R10O-,-N (R10) 2 or C2-C6 alkenyl, and c) C1-C6 alkyl unsubstituted or substituted by aryl, heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, R10O-, or-N (R10)2 ; Rld is selected from : a) hydrogen, b) unsubstituted or substituted aryl, unsubstituted or substituted heterocycle, C3-C10 cycloalkyl, R100-, RlOC (O) NRlo-, (R10)N- C (O) -N(R10)2, or R11OC(O)NR10-, c) unsubstituted or substituted C1-C6 alkyl, unsubstituted or substituted C2-C6 alkenyl or unsubstituted or substituted C2-C6 alkynyl, wherein the substituent on the substituted Cl-C6 6 alkyl, substituted C2-C6 alkenyl or substituted C2-C6 alkynyl is selected from unsubstituted or substituted aryl, heterocyclic, C3-C10 cycloalkyl, C2-C6 alkenyl, C2- C6 alkynyl, Cl-C6 perfluoroalkyl, halogen, R1OO-, R4S (O) m-,

R4S(O)2NR10-, R10C(O)NR10-, (R10)2N-C(O)-, CN, (R10)2N- C(NR10)-, R10C(O)-, R10OC(O)-, -N(R10)2, and R11OC(O)-NR10-; R3a is selected from H and CH3 ; R2a is selected from H ; and C 1-5 alkyl, unbranched or branched, unsubstituted or substituted with one or more of : 1) aryl, 2) heterocycle, 3) OR6, 4) SR4, SO2R4, or and any two of R2a and R3a are optionally attached to the same carbon atom ; R4 is selected from C1-4 alkyl and C3_6 cycloalkyl, unsubstituted or substituted with : a) C1 4 alkoxy, b) halogen, or c) aryl or heterocycle ; R6 and R7 are independently selected from H ; C1-6 alkyl, C3-6 cycloalkyl, heterocycle, aryl, aroyl, heteroaroyl, arylsulfonyl, heteroarylsulfonyl, unsubstituted or substituted with one or two : a) C1-4 alkoxy, b) aryl or heterocycle, c) halogen, d) HO,

f)-S02R" g) N (RlO) 2, or h) C3-6 cycloalkyl ; R8 is independently selected from : a) hydrogen, b) unsubstituted or substituted aryl, C1-C6 6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, Cl-C6 perfluoroalkyl, F, Cl, R120-, RIOC (O) NRlO-, CN, NO2 (R10 2N-C 10)-, Rl0C (O)-,-N (R10) 2, or RllOC (O) NRlo-, and c) C1-C6 alkyl substituted by unsubstituted or substituted aryl, Cl-C6 perfluoroalkyl, R10O-, R10C(O)NR10-, (R10)2N-C(NR10)-, R10C(O)-, -N(R10)2, or R11OC(O)NR10-; R9a is selected from hydrogen, C1-C6 alkyl and C1-C6 perfluoroalkyl ; Rl is independently selected from hydrogen, C1-C6 alkyl, benzyl and unsubstituted or substituted aryl ; Rl 1 is independently selected from C1-C alkyl and unsubstituted or substituted aryl ; R12 is independently selected from hydrogen, C1-C6 6 alkyl, unsubstituted or substituted benzyl, unsubstituted or substituted aryl, unsubstituted or substituted heterocycle, and C1-C6 alkyl substituted with unsubstituted or substituted aryl or unsubstituted or substituted heterocycle ; Al is selected from a bond,-N (RlO)-, S (O) m and O ; V is phenyl or pyridyl ;

X is selected from a bond,-C (=O)-, -NR10C(O)-, -OC(O)- or -S(=O)m ; Y is selected from a bond,-C (=O)-,-C (O) NRlO-,-C (O) O-, or-S (=O) m ; Zl is selected from unsubstituted or substituted aryl or unsubstituted or substituted heterocycle, wherein the substituted aryl or substituted heterocycle is substituted with one or two of : 1) C1-8 alkyl, C2-8 alkenyl or C2 8 alkynyl, unsubstituted or substituted with : a) C1-4 alkoxy, ) c) C3-6 cycloalkyl, d) aryl or heterocycle, e) HO, f)-S mR4, g)-C (O) NR6R7, or h) C1-4 perfluoroalkyl ; 2) substituted or unsubstituted aryl or substituted or unsubstituted heterocycle, 3) halogen, 4) OR6, 5) NR6R7, 6) CN, 7) NO2, 8) CF3, <BR> <BR> 9)-S (o) mR4,<BR> <BR> <BR> <BR> 10)-OS (0) 2R4,<BR> <BR> <BR> <BR> 11)-C (O) NR6R7, 12)-C (O) OR6, or 13) C3-C6 cycloalkyl ; m is 0, 1 or 2 ; n is 0, 1, 2, 3 or 4 ; p is independently 0, 1, 2, 3 or 4 ;

r is 0 to 5 ; and s is independently 0, 1, 2 or 3 ; or a pharmaceutically acceptable salt or stereoisomer thereof.

Another preferred embodiment of the compounds of this invention is illustrated by the formula C : wherein : R1a, Rlb and Rlc is independently selected from : a) hydrogen, b) aryl, heterocycle, C3-Clp cycloalkyl, R100-,-N (Rl) 2 or C2-C6 alkenyl, and c) C 1-C6 alkyl unsubstituted or substituted by aryl, heterocycle, C3-Cl0 cycloalkyl, C2-C6 alkenyl, R10O-, or-N (R10) 2 ; Rld is selected from : a) hydrogen, b) aryl, heterocycle, C3-C10 cycloalkyl, R10O-, -N(R10)2 or C2-C6 alkenyl, and

c) C1-C6 alkyl, C2-C6 alkenyl or C2-C6 alkynyl, unsubstituted or substituted by aryl, heterocycle, C3-Clo cycloalkyl, C2-C6 alkenyl, R10O-, or-N (R10)2 ; R3a is selected from H and CH3 ; R2a is selected from H ; and C1-5 alkyl, unbranched or branched, unsubstituted or substituted with one or more of : 1) aryl, 2) heterocycle, 3) OR6, 4) SR4, S02R4, or and any two of R2a and R3a are optionally attached to the same carbon atom ; R4 is selected from C1-4 alkyl and C3-6 cycloalkyl, unsubstituted or substituted with : a) C1-4 alkoxy, b) halogen, or c) aryl or heterocycle ; R6 and R7 are independently selected from : a) hydrogen, b) Cl-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, R10C(O)- or R10OC (O)- and c) C1-C6 alkyl substituted by Cl-C6 perfluoroalkyl, R100-, R10C(O)NR10-, (R10)2N-C(NR10)-, R10C(O)-, R10OC(O)-, -N (R10)2, or Rl l OC (O) NR10-; R8 is independently selected from :

a) hydrogen, b) unsubstituted or substituted aryl, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 perfluoroalkyl, F,#Cl, R12O-, R10C(O)NR10-, CN, N02, (R10) 2N-C(NR10)-, R10C(O)-, -N(R10)2, or R11OC(O)NR10-, and c) C1-C6 alkyl substituted by unsubstituted or substituted aryl, Cl-C6 perfluoroalkyl, R100-, RIOC (O) NR10-, (R10)2N-C(NR10)- R10C(O)-, -N(R10)2, or R11OC(O)NR10-; R9a is selected from hydrogen, C1-C6 alkyl and C1-C6 perfluoroalkyl ; R10 and R12 are independently selected from hydrogen, C1-C6 alkyl, benzyl and unsubstituted or substituted aryl ; Ru1 ils independently selected from C1-C6 alkyl and unsubstituted or substituted aryl ; A1 is selected from a bond,-N (RlO)-, S (O) m and O ; X is selected from a bond,-C (=O)-,-NR10C (O)-,-OC (O)- or -S(=O)m ; Y is selected from a bond,-C (=O)-,-C (O) NRlO-,-C (O) O-, or-S (=O) m ; zl is selected from unsubstituted or substituted aryl or unsubstituted or substituted heterocycle, wherein the substituted aryl or substituted heterocycle is substituted with one or two of : 1) C1-4 alkyl, unsubstituted or substituted with : a) C1-4 alkoxy, b) NR6R7, c) C3_6 cycloalkyl, d) aryl or heterocycle, e) HO, f)-S (O) mR4, or ) 2) aryl or heterocycle,

3) halogen, 4) OR6, 5) NR6R7, 6) CN, 7) NO2, 8) CF3, 9)-S (0) mR4, 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 ; r is 0 to 5 ; and s is independently 0, 1, 2 or 3 ; or a pharmaceutically acceptable salt or stereoisomer thereof.

In another embodiment of this invention, the inhibitors of prenyl- protein transferase are illustrated by the formula D :

wherein : Rlb and Rlc is independently selected from : a) hydrogen, b) aryl, heterocycle, C3-C10 cycloalkyl, R10O- or -N(R10)2, and c) C 1-C6 alkyl unsubstituted or substituted by aryl, heterocycle, C3-Cl0 cycloalkyl, C2-C6 alkenyl, R10O-, or-N (R10) 2 ; Rld is selected from : a) hydrogen, b) aryl, heterocycle, C3-Clp cycloalkyl, Rl0-or-N (Rl ) 2, and c) Cl-C6 alkyl, C2-C6 alkenyl or C2-C6 alkynyl, unsubstituted or substituted by aryl, heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, R10O-, or-N (R10) 2 ; R3a is selected from H and CH3 ; R2a is selected from H ; and C1-5 alkyl, unbranched or branched, unsubstituted or substituted with one or more of : 1) aryl, 2) heterocycle, 3) OR6, 4) SR4, S02R4, or and any two of R2 and R3 are optionally attached to the same carbon atom ; R4 is selected from C1-4 alkyl and C3_6 cycloalkyl, unsubstituted or substituted with : a) C1 4 alkoxy, b) halogen, or

c) aryl or heterocycle ; R6 and R7 are independently selected from H ; Cl-6 alkyl, C3_6 cycloalkyl, aryl, aroyl, arylsulfonyl, unsubstituted or substituted with one or two : a) C1-4 alkoxy, b) aryl, c) halogen, d) HO, f) -SO2R11 g) N (R10) 2 h) C3-6 cycloalkyl ; or R8 is independently selected from : a) hydrogen, b) unsubstituted or substituted aryl, C1-C6 6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 perfluoroalkyl, F, Cl, R120-, RIOC (O) NRlO-, CN, N02, (R10) 2N-C (NR10)-, R10C(O)-, -N(R10)2, or R11OC(O)NR10-, and c) C1-C6 alkyl substituted by unsubstituted or substituted aryl, Cl-C6 perfluoroalkyl, R10O-, R10C(O)NR10-, (R10) 2N-C (NR10)-, Rl0C )-,-N (Rl0) 2, or RllOC (O) NR10 ; R9a is selected from hydrogen, C1-C6 alkyl and C1-C6 perfluoroalkyl ; R10 and R12 are independently selected from hydrogen, C1-C6 6 alkyl, benzyl and unsubstituted or substituted aryl ; Rl 1 is independently selected from Cl-C6 alkyl and unsubstituted or substituted aryl ; Al is selected from a bond,-N (RlO)-, S (O) m and O ;

X is selected from a bond,-C (=O)-,-NR10C (O)-,-OC (O)- or-S (=O) m ; Y is selected from a bond,-C (=O)-,-C (O) NR10-,-C (O) O-, or-S (=O) m ; Zl is selected from unsubstituted or substituted aryl or unsubstituted or substituted heterocycle, wherein the substituted aryl or substituted heterocycle is substituted with one or two of : 1) C1-8 alkyl, C2-8 alkenyl or C2-8 alkynyl, unsubstituted or substituted with : a) C1-4 alkoxy, b) NR6R7, c) C3-6 cycloalkyl, d) aryl or heterocycle, e) HO, f)-S (0) mR ) -C(O)NR6R7, OR h) C1-4 perfluoroalkyl ; 2) substituted or unsubstituted aryl or substituted or unsubstituted heterocycle, 3) halogen, 5) OR6, 6) CN, 7) N02, <BR> <BR> <BR> 8) CF3,<BR> <BR> <BR> <BR> <BR> 9)-S (0) mR4, 10) -OS(O)2R4, 11)-C (O) NR6R7, 12)-C (O) OR6, or 13) C3-C6 cycloalkyl ; m is 0, 1 or 2 ; p is independently 0, 1, 2, 3 or 4 ;

ris0to 5 ; and s is independently 0, 1, 2 or 3 ; or a pharmaceutically acceptable salt or stereoisomer thereof.

In a further preferred embodiment of this invention, the inhibitors of prenyl-protein transferase are illustrated by the formula E :

wherein : Rlb and Rlc is independently selected from : a) hydrogen, b) aryl, heterocycle, cycloalkyl, R10O-, -N(R10)2 or C2-C6 alkenyl, and c) C1-C6 alkyl unsubstituted or substituted by aryl, heterocycle, cycloalkyl, alkenyl, R100-, or-N (RlO) 2 ; Rld is selected from : a) hydrogen, b) aryl, heterocycle, C3-Clo cycloalkyl or C2-C6 alkenyl, and

c) Cl-C6 alkyl, C2-C6 alkenyl or C2-C6 alkynyl, unsubstituted or substituted by aryl, heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, R10O-, or -N(R10) 2 ; R3a is selected from H and CH3 ; R2a is selected from H ;

and C1 5 alkyl, unbranched or branched, unsubstituted or substituted with one or more of : 1) aryl, 2) heterocycle, 3) OR6, 4) SR4, S02R4, or and any two of R2a and R3a are optionally attached to the same carbon atom ; R4 is selected from C1-4 alkyl and C3_6 cycloalkyl, unsubstituted or substituted with : a) C 1-4 alkoxy, b) fluoro, or c) aryl or heterocycle ; R6 and R7 are independently selected from H ; C1-6 alkyl, C3-6 cycloalkyl, aryl, aroyl, arylsulfonyl, unsubstituted or substituted with one or two : a) Cl-4 alkoxy, b) aryl, c) halogen, d) HO,

f)-S02R" g) N (RlO) 2, or h) C3-6 cycloalkyl, R8 is independently selected from : a) hydrogen, b) unsubstituted or substituted aryl, Cl-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 perfluortoalkyl, F, Cl, R120-, R1OC (O) NRl0-, CN, N02, (R10)2N-C(NR10)-, R10C(O)-, -N(R10)2, or R11OC(O)NR10-, and c) C1-C6 alkyl substituted by unsubstituted or substituted aryl, C1-C6 perfluoroalkyl, R10O-, R10C(O)NR10-, (R10)2N-C(NR10)-, R10C(O)-, -N(R10)2, or R11OC(O)NR10-; R9a is selected from hydrogen, C1-C6 alkyl and C1-C6 perfluoroalkyl ; RI and R12 are independently selected from hydrogen, C1-C6 alkyl, benzyl and unsubstituted or substituted aryl ; RI l is independently selected from C 1-C6 alkyl and unsubstituted or substituted aryl ; Al is selected from a bond,-N (R10)-, S (O) m and O ; X is selected from a bond,-C (=O)-, -NR10C(O)-, -OC(O)- or -S(=O)m ; Y is selected from a bond,-C (=O)-,-C (O) NR10-,-C (O) O-, or-S (=O) m ; Zl is selected from unsubstituted or substituted aryl or unsubstituted or substituted heterocycle, wherein the substituted aryl or substituted heterocycle is substituted with one or two of :

1) C1-8 alkyl, C2-8 alkenyl or C2-8 alkynyl, unsubstituted or substituted with : a) C1-4 alkoxy, c) C3_6 cycloalkyl, d) aryl or heterocycle, e) HO, -S(O)mR4, g)-C (O) NR6R7, or h) C1-4 perfluoroalkyl ; 2) substituted or unsubstituted aryl or substituted or unsubstituted heterocycle, 3) halogen, 4) OR6, 5) NR6R7, 6) CN, 7) NO2, 8) CF3, -S(O)mR4, 10)-OS (0) 2R4, 11)-C (O) NR6R7, 12)-C (O) OR6, or 13) C3-C6 cycloalkyl ; m is 0, 1 or 2 ; p is independently 1, 2, 3 or 4 ; r is 0 to 5 ; and s is independently 0, 1, 2 or 3 ; or a pharmaceutically acceptable salt or stereoisomer thereof.

The following compounds are specific examples of the compounds of the instant invention :

14-amino-14- (1-methyl-lH-imidazol-5-yl)-8-oxa-2-thia-1, 18- diazatetracyclo [16. 2. 2. 137. 19,13]tetracosa-3 (24), 4, 6, 9 (23), 10, 12-hexaene-10- carbonitrile 2, 2-dioxide ; 14-amino-4-chloro-14-(1-methyl-1H-imidazol-5-yl)-20-oxo-8-ox a-1, 18- diazatetracyclo [16. 2. 2. 137. 1'] tetracosa-3 (24), 4, 6, 9 (23), 10, 12-hexaene-10- carbonitrile ; 18-Bromo-8- [5- (1-methyl-1H-imidazolyl)]-8-methyl-3, 7 : 1, 17-dimetheno-1 lH-10, 14- dioxo-12, 15-ethano-16H-2-oxa-9, 10 : 13, 14-tetrahydro- [9, 12, 15] triaza-cycloeicosene- 4-carbonitrile ; 18-Chloro-8- [5- (l-methyl-lH-imidazolyl)]-8-methyl-3, 7 : 1, 17-dimetheno-1 lH-10, 14- dioxo-12, 15-ethano-16H-2-oxa-9, 10 : 13, 14-tetrahydro- [9, 12, 15] triaza-cycloeicosene- 4-carbonitrile ; or a pharmaceutically acceptable salt or stereoisomer thereof.

A specific example of the compounds of the instant invention is : 14-amino-14- (l-methyl-lH-imidazol-5-yl)-8-oxa-2-thia-1, 18- diazatetracyclo [16. 2. 2. 13,7.19,13]tetracosa-3 (24), 4, 6, 9 (23), 10, 12-hexaene-10- carbonitrile 2, 2-dioxide or a pharmaceutically acceptable salt or stereoisomer thereof.

The compounds of the present invention may have asymmetric centers, chiral axes and chiral planes, and occur as racemates, racemic mixtures, and as individual diastereomers, with all possible isomers, including optical isomers, being included in the present invention. (See E. L. Eliel and S. H. Wilen Stereochemistry of Carbon Compounds (John Wiley and Sons, New York 1994), in particular pages 1119-1190) When any variable (e. g. aryl, heterocycle, Rl a, R6 etc.) occurs more than one time in any constituent, its definition on each occurrence is independent at every other occurrence. Also, combinations of substituents/or variables are permissible only if such combinations result in stable compounds.

As used herein,"alkyl"is intended to include both branched and straight-chain saturated aliphatic hydrocarbon groups having the specified number of carbon atoms ;"alkoxy"represents an alkyl group of indicated number of carbon atoms attached through an oxygen bridge."Halogen"or"halo"as used herein means fluoro, chloro, bromo and iodo.

Preferably, alkenyl is C2-C6 alkenyl.

Preferably, alkynyl is C2-C6 alkynyl.

As used herein,"cycloalkyl"is intended to include cyclic saturated aliphatic hydrocarbon groups having the specified number of carbon atoms.

Preferably, cycloalkyl is C3-Clp cycloalkyl. Examples of such cycloalkyl elements include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl and cycloheptyl.

As used herein,"aryl"is intended to mean any stable monocyclic or bicyclic carbon ring of up to 7 members in each ring, wherein at least one ring is aromatic. Examples of such aryl elements include phenyl, naphthyl, tetrahydronaphthyl, indanyl, biphenyl, phenanthryl, anthryl or acenaphthyl.

The term heterocycle or heterocyclic, as used herein, represents a stable 5-to 7-membered monocyclic or stable 8-to 11-membered bicyclic heterocyclic 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, O, 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.

The term heterocycle or heterocyclic includes heteroaryl moieties. Examples of such heterocyclic elements include, but are not limited to, azepinyl, benzimidazolyl, benzisoxazolyl, benzofurazanyl, benzopyranyl, benzothiopyranyl, benzofuryl,

benzothiazolyl, benzothienyl, benzoxazolyl, chromanyl, cinnolinyl, dihydrobenzofuryl, dihydrobenzothienyl, dihydrobenzothiopyranyl, dihydrobenzothiopyranyl sulfone, 1, 3-dioxolanyl, furyl, imidazolidinyl, imidazolinyl, imidazolyl, indolinyl, indolyl, isoclmomanyl, isoindolinyl, isoquinolinyl, isothiazolidinyl, isothiazolyl, isothiazolidinyl, morpholinyl, naphthyridinyl, oxadiazolyl, 2-oxoazepinyl, oxazolyl, 2-oxopiperazinyl, 2-oxopiperdinyl, 2- oxopyrrolidinyl, piperidyl, piperazinyl, pyridyl, pyrazinyl, pyrazolidinyl, pyrazolyl, pyridazinyl, pyrimidinyl, pyrrolidinyl, pyrrolyl, quinazolinyl, quinolinyl, quinoxalinyl, tetrahydrofuryl, tetrahydroisoquinolinyl, tetrahydroquinolinyl, thiamorpholinyl, thiamorpholinyl sulfoxide, thiazolyl, thiazolinyl, thienofuryl, thienothienyl, and thienyl. An embodiment of the examples of such heterocyclic elements include, but are not limited to, azepinyl, benzimidazolyl, benzisoxazolyl, benzofurazanyl, benzopyranyl, benzothiopyranyl, benzofuryl, benzothiazolyl, benzothienyl, benzoxazolyl, chromanyl, cinnolinyl, dihydrobenzofuryl, dihydrobenzothienyl, dihydrobenzothiopyranyl, dihydrobenzothiopyranyl sulfone, furyl, imidazolidinyl, imidazolinyl, imidazolyl, indolinyl, indolyl, isochromanyl, isoindolinyl, isoquinolinyl, isothiazolidinyl, isothiazolyl, isothiazolidinyl, morpholinyl, naphthyridinyl, oxadiazolyl, 2-oxoazepinyl, oxazolyl, 2-oxopiperazinyl, 2-oxopiperdinyl, 2- oxopyrrolidinyl, piperidyl, piperazinyl, pyridyl, 2-pyridinonyl, pyrazinyl, pyrazolidinyl, pyrazolyl, pyridazinyl, pyrimidinyl, pyrrolidinyl, pyrrolyl, quinazolinyl, quinolinyl, quinoxalinyl, tetrahydrofuryl, tetrahydroisoquinolinyl, tetrahydroquinolinyl, thiamorpholinyl, thiamorpholinyl sulfoxide, thiazolyl, thiazolinyl, thienofuryl, thienothienyl, thienyl and triazolyl.

As used herein,"heteroaryl"is intended to mean any stable monocyclic or bicyclic carbon ring of up to 7 members in each ring, wherein at least one ring is aromatic and wherein from one to four carbon atoms are replaced by heteroatoms selected from the group consisting of N, O, and S. Examples of such heterocyclic elements include, but are not limited to, benzimidazolyl, benzisoxazolyl, benzofurazanyl, benzopyranyl, benzothiopyranyl, benzofuryl, benzothiazolyl, benzothienyl, benzoxazolyl, chromanyl, cinnolinyl, dihydrobenzofuryl, dihydrobenzothienyl, dihydrobenzothiopyranyl, dihydrobenzothiopyranyl sulfone,. furyl, imidazolyl, indolinyl, indolyl, isochromanyl, isoindolinyl, isoquinolinyl, isothiazolyl, naphthyridinyl, oxadiazolyl, pyridyl, pyrazinyl, pyrazolyl, pyridazinyl, pyrimidinyl, pyrrolyl, quinazolinyl, quinolinyl, quinoxalinyl, tetrahydroisoquinolinyl, tetrahydroquinolinyl, thiazolyl, thienofuryl, thienothienyl, thienyl and triazolyl.

As used herein, unless otherwise specifically defined, substituted alkyl, substituted cycloalkyl, substituted aroyl, substituted aryl, substituted heteroaroyl, substituted heteroaryl, substituted arylsulfonyl, substituted heteroaryl- sulfonyl and substituted heterocycle include moieties containing from 1 to 3 substituents in addition to the point of attachment to the rest of the compound.

Preferably, such substituents are selected from the group which includes but is not limited to F, Cl, Br, CF3, NH2, N (C1-C6 alkyl) 2, N02, CN, (C I-C6 alkyl) O-, (aryl) O-,-OH, (Cl-C6 alkyl) S (O) m, (C1-C6 alkyl) C (O) NH-, H2N-C (NH)-, (C1-C6 alkyl) C (O)-, (C1-C6 alkyl) OC (O)-, (C1-C6 alkyl) OC (O) NH-, phenyl, pyridyl, imidazolyl, oxazolyl, isoxazolyl, thiazolyl, thienyl, furyl, isothiazolyl and C1-C20 alkyl.

As used herein in the definition of R2 and R3, the term"the substituted group"intended to mean a substituted Cl-8 alkyl, substituted C2-8 alkenyl, substituted C2-8 alkynyl, substituted aryl or substituted heterocycle from which the substituent (s) R2 and R3 are selected.

Preferably, as used herein in the definition of R6 and R7, the substituted CI-6 alkyl, substituted C2 6 alkenyl, substituted C2 6 alkynyl, substituted C3 6 cycloalkyl, substituted aroyl, substituted aryl, substituted heteroaroyl, substituted arylsulfonyl, substituted heteroarylsulfonyl and substituted heterocycle, include moieties containing from 1 to 3 substituents in addition to the point of attachment to the rest of the compound.

The moiety formed when, in the definition of Rla, Rlb, Rlc and Rld, two Rlasn two Rlbs, two Rlcs or two Rlds, on the same carbon atom are combined to form- (CH2) t- is illustrated by the following : 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 :

The moiety formed when, in the definition of R5, R6 and R7, R6 and R7 or R5 and R7 are joined to form a ring, is illustrated by, but not limited to, the following :

Lines drawn into the ring systems from substituents (such as from R2, R3, R4 etc.) indicate that the indicated bond may be attached to any of the substitutable ring carbon or nitrogen atoms.

Preferably, Rla and Rlb are independently selected from : hydrogen, -N (RI 0) 2, R10C (O) NRl- or unsubstituted or substituted C1-C6 alkyl wherein the substituent on the substituted Cl-C6 alkyl is selected from unsubstituted or substituted phenyl,-N (R10)2, R10O- and R10C(O)NR10-.

Preferably, Ric is independently selected from : hydrogen, or unsubstituted or substituted C1-C6 alkyl wherein the substituent on the substituted

C 1-C6 alkyl is selected from unsubstituted or substituted phenyl,-N (R10)2, R10O- and Rloc (o) NRl0- Preferably, Rld is selected from : a) hydrogen, b) substituted or unsubstituted aryl, substituted or unsubstituted heterocycle, C3-C6 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, R10O-, -N(R10)2, R10C(O)NR10-, (R10)2N-C(O)-, or R10OC (O)-, and c) unsubstituted or substituted C1-C6 alkyl wherein the substituent on the substituted Cl-C6 alkyl is selected from substituted or unsubstituted aryl, substituted or unsubstituted heterocycle, halo, Cl-C6 perfluoroalkyl, C3-C6 (cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, R10O-, R4S(O)m-, R10C(O)NR10-, (R10)2N-C(O)-, CN, (R10)2N-C(NR10)-, R4S(O)2NR10-, -S(O)2N(R10)2, R10C(O)-, R10OC(O)-, -N(R10)2, and R11 OC(O)-NR10-.

Preferably, R2a is selected from H, and an unsubstituted or substituted group, the group selected from C1-8 alkyl, C2-8 alkenyl and C2 8 alkynyl ; wherein the substituted group 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, 2) C3-6 cycloalkyl, 3) OR6, 4) SR4, S (O) R4, S02R4, 5) -NR6R7

15) N3, or 16) F.

Preferably, R2b, R3a and R3b are independently selected from : hydrogen and C1-C6 alkyl.

Preferably, R4 is unsubstituted or substituted C1-C6 alkyl, unsubstituted or substituted aryl and unsubstituted or substituted cycloalkyl.

Preferably, R5, R6 and R7 is selected from : hydrogen, unsubstituted or substituted Cl-C6 alkyl, unsubstituted or substituted aryl and unsubstituted or substituted cycloalkyl.

Preferably, R9 is hydrogen or methyl.

Preferably, R10 is selected from H, Cl-C6 alkyl and benzyl.

Preferably, Al and A2 are independently selected from : a bond, -C(O)NR10-, -NR10C(O)-, O, -N(R10)-, -S(O)2N(R10)-, S(=O) m and -N (RlO) S (0) 2-.

Preferably, G1 is O or H2. Preferably, G2 and G3 are hydrogen.

Preferably, V is selected from heteroaryl and aryl. More preferably, V is phenyl or pyridyl.

Preferably, X and Y are independently selected from : a bond,- (S=O)m, -C(O)NR10-, -NR10C(O)-, -S(O) 2N (RlO)-,-N (RlO) S (0) 2-. and-C (=0)-.

Preferably, Zl and z2 are independently selected from unsubstituted or substituted phenyl, unsubstituted or substituted naphthyl, unsubstituted or substituted pyridyl, unsubstituted or substituted furanyl and unsubstituted or substituted thienyl.

More preferably, Z I is selected from unsubstituted or substituted phenyl and unsubstituted or substituted naphthyl. More preferably, Z2 is selected from a bond and unsubstituted or substituted phenyl.

Preferably, W is selected from imidazolyl, oxazolyl, pyridyl, and triazolyl. More preferably, W is selected from imidazolyl and pyridyl.

Preferably, n is 0, 1, or 2.

Preferably, r is 1 or 2.

Preferably p is 1, 2 or 3.

Preferably s is 0 or 1.

Preferably, the moiety is selected from :

wherein R9a is selected from hydrogen or methyl.

It is intended that the definition of any substituent or variable (e. g., Rla, R9, n, etc.) at a particular location in a molecule be independent of its definitions elsewhere in that molecule. Thus,-N (RlO) 2 represents-NHH,-NHCH3,-NHC2H5, etc. It is understood 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, from readily available starting materials.

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, phenylacetic, glutamic, benzoic, salicylic, sulfanilic, 2-acetoxy-benzoic, fumaric, toluenesulfonic, methanesulfonic, ethane disulfonic, oxalic, isethionic, trifluoroacetic and the like.

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 either by ion exchange chromatography or 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.

Reactions used to generate the compounds of this invention are prepared by employing reactions as shown in the Schemes 1-18, 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, Rb and Rsub, as shown in the Schemes, represent the substituents R2, R3, R4, and R5, and substituents on Zl and z2 ; 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 1-18 : The requisite intermediates are in some cases commercially available, or can be prepared according to literature procedures. In Scheme 1, for example, the general synthesis of macrocyclic compounds of the instant invention containing suitably substituted piperazines. Preparation of the substituted piperazine intermediate is essentially that described by J. S. Kiely and S. R. Priebe in Organic Preparations and Proceedings Int., 1990, 22, 761-768. Boc-protected amino acids I, available commercially or by procedures known to those skilled in the art, can be coupled to N-benzyl amino acid esters using a variety of dehydrating agents such as DCC (dicyclohexycarbodiimide) or EDC-HCI (1-ethyl-3- (3- dimethylaminopropyl) carbodiimide hydrochloride) in a solvent such as methylene chloride, chloroform, dichloroethane, or in dimethylformamide. The product II is then deprotected with acid, for example hydrogen chloride in chloroform or ethyl acetate, or trifluoroacetic acid in methylene chloride, and cyclized under weakly basic conditions to give the diketopiperazine m. Reduction of III with lithium aluminum hydride in refluxing ether gives the piperazine IV, which may then be deprotected by catalytic reduction to provide intermediate V. Intermediate V may then be reductively alkylated with intermediate aldehyde VII, prepared from a suitably substituted halohydroxytoluene VI in several steps as illustrated. Cesium carbonate mediated intramolecular cyclization of intermediate VIII provides compound IX of the instant invention. This cyclization reaction depends on the presence of an electron withdrawing moiety (such as nitro, cyano, and the like) either ortho or para to the fluorine atom. The cyclization product can then be deprotected to provide the compound of the instant invention.

Scheme 2 illustrates the synthesis of a compound of the instant invention wherein"W"is the preferred imidazolyl moiety. Thus, trityl protected iodoimidazolyl is reacted with the aldehyde X to provide the alcohol XI. Compound XI can be manipulated to provide the substituted imidazolyl ketone XII. The ketone XII may be further functionalized to intermediates XIII and XIV, or, as generally illustrated in Scheme I, elaborated to the instant compound XVI.

The olefin of intermediate XV may alernatively be converted to the carboxylic acid in intermediate XVII, as shown in Scheme 3. The carboxylic acid may then be reacted with a suitably protected and substituted piperazine such as IV to

provide intermediate XVIII. Deprotection followed by cesium carbonate mediated cyclization provides the instant compound XX.

Scheme 4 illustrates the synthesis of instant compounds wherein the piperazine nitrogen and the"Y"group form an amide bond. Thus, the protected piperazine XXI is coupled to a naphthoic acid having a suitably positioned benzyloxy moiety. Consecutive removal of the Boc and benzyl protecting groups provided intermediate XXII, which may be reductively alkylated with a suitably substituted aldehyde XXm to give intermediate XXIV. Intramolecular cyclization takes place as previously described using the cesium carbonate conditions to provide instant compound XXV after deprotection.

Scheme 5 illustrates the preparation of instant compounds which incorporate a piperazinone moiety in the macrocyclic ring. Thus the suitably substituted benzyloxybenzyl mesylate XXVI is reacted with a 4-protected 2- piperazinone XXVII to provide the l-benzyl-2-piperazinone intermediate XXVIII.

Intermediate XXVIII is doubly deprotected to provide intermediate XXIX. Reductive N-alkylation of intermediate XXIX with a suitably substituted aldehyde XXIIII provides intermediate XXX, which can undergo intramolecular cyclization under the cesium carbonate conditions and deprotection to give compound XXXI of the instant invention.

Synthesis of compounds of the invention characterized by direct attachment of an aryl moiety to the piperazinone moiety and incorporation of a third aromatic carbocyclic moiety into the macrocycle is illustrated in Scheme 6. A benzyloxyphenoxyaniline XXX1V, prepared in three steps from a suitably substituted 2-benzyloxyphenol XXXII and a suitably substituted nitrochlorobenzene XXXIII, is reacted with chloroacetyl chloride to provide intermediate XXXV. Intermediate XXXV is reacted with a suitably substituted ethanolamine and the resulting amido alcohol cyclized to form the piperazinone moiety of intermediate XXXVI.

Intermediate XXXVI is reductively alkylated as described in Scheme 2 to provide intermediate XXXVII. Deprotection, followed by intramolecular cyclization, then deprotection of the tertiary alcohol provides compound XXXVDI of the instant invention.

Treatment of the hydroxy intermediate VII with thionyl chloride, followed by reaction of the in situ generated chloro intermediate with the piperazinone moiety IXL provides the instant compound XL after the standard deprotection and cyclization sequence, as illustrated in Scheme 7.

Scheme 8 illustrates an alternative synthesis of such"directly attached" compounds which enable incorporation of a substituent at the 3-position of the piperazinone moiety.

Scheme 9 illustrates the synthetic strategy that is employed when the R8 substituent is not an electronic withdrawing moiety either ortho or para to the fluorine atom. In the absence of the electron withdrawing moiety, the intramolecular cyclization can be accomplished via an Ullmann reaction. Thus, a suitably substituted iodo benzaldehyde XLI may be employed in place of intermediate II. Incorporation of a"W"moiety, in this instance a pyridyl group, followed by the previously described elaboration provides the intermediate XLII. Intramolecular cyclization may then be affected under Ullmann conditions, followed by deprotection to provide the instant compound XLE. The tertiary hydroxy moiety of compound XLDI may be removed by boron trifluoroetherate/triethylsilane reduction as illustrated.

Illustrative examples of the preparation of compounds of the instant invention that incorporate a 2, 5-diketopiperazine moiety and a 2, 3-diketopiperazine moiety are shown in Scheme 10 and Scheme 11 respectively.

Scheme 12 illustrates the instant compound wherein the piperazinone nitrogen forms an amide bond with moiety"X". Thus, the piperazinone having a suitable 5-position substituent is treated with chloroacetyl chloride to provide intermediate XLIV. Intermediate XLIV is then reacted with the previously described amine XIV to provide intermediate XLV, which may undergo the previously described cyclization to afford instant compound XLVI.

Scheme 13 illustrates an alternative synthetic method of forming the piperazinone ring in the macrocycle.

Scheme 14 illustrates preparation of an intermediate piperazinone compound XLVII having a substituent at the 3-position that is derived from the starting protected amino acid I.

Incorporation of a spirocyclic moiety (for example, when R2 and R3 are combined to form a ring) is illustrated in Scheme 15.

Scheme 16 illustrates the use of an optionally substituted homoserine lactone XLVHI to prepare a Boc-protected piperazinone IL. Intermediate IL may be deprotected and reductively alkylated or acylated as illustrated in the previous Schemes. Alternatively, the hydroxyl moiety of intermediate IL may be mesylated and displaced by a suitable nucleophile, such as the sodium salt of ethane thiol, to provide an intermediate L. Intermediate IL may also be oxidized to provide the

carboxylic acid on intermediate LI, which can be utilized form an ester or amide moiety.

Amino acids of the general formula LII which have a sidechain not found in natural amino acids may be prepared by the reactions illustrated in Scheme 17 starting with the readily prepared imine LE.

Incorporation of a sulfur containing moiety for Al in the instant compounds is illustrated in Scheme 18. Thus chloroacetyl chloride is reacted with the disulfide protected aniline LIV to provide, after the previously described formation of the piperazinone ring, the intermediate LV. Intermediate LV may be reacted with the preiously described aldehyde to provide intermediate LVI. The sulfide moiety is liberated with dithiothrietol to provide the mercaptan, which undergoes cyclization under cesium carbonate conditions to provide the instant compound LVII after deprotection. The sulfur may be oxidized to either the sulfone or sulfoxide LVE.

As shown in Scheme 19, the ketone moiety of intermediate XII may alternatively be converted to the protected imine LIX, which may react with allyl Grignard to provide, after oxidation, aldehyde intermediate LX. Intermediate LX may then be reacted with a piperazine, such as V, to eventually provide the instant compound LXI.

SCHEME 1 OBn a R b O R Rsub OH CH2NHCHC02C2H5 H O DCC, CH2C12 OBn f j] r-) SUb Rsub O ra /0 HCI, CH2C12 N C02C2H5 2) NaHC03 H 0 ! ! R Ra 0 Ra LAH HAN N HN N THF, reflux /\ b OBn b Rub OBn ib OBn I sub III IV R Rsub Ra 10% Pd/C HN NX H2 CH30H Rb OH _ _/ V Rsub V RUS SCHEME 1 (continued) OH H O R9 W pyr. S03 MgBr or I/ R9 or halo Mn02 R8 halo lui Vu R9 R9 mono2 I\ I\ halo halo R8 R8 SCHEME 1 (continued) 9 R W allyl-MgBr W OH I\ \ R 8 R8 0-H H 9 1) Os04, H20, W OH H OH protect dioxane I\ 2) Na104, H20 F F R8 R' Oprot H ¢X° F R8 vil SCHEME 1 (continued) R9 R9 Oprot Ra w H 0 tk A R8 F v R sub OH pS sub Vt) NaBH (OAc) 3 R9 Oprot Ru ups I R9 Oprot R\ F VI I I = R8 Rsub RS R' Cs2CO3 DMSO heat R 9 Oprot R a w S y XRsUb Rsub R O R IX SCHEME 1 (continued) deprotect R9 OH Ra a N N sub "--< b R8 R- 0 R8 SCHEME 2 SCHEME 2 (continued) N N N y g N / R R F F D8 D8 XXV N 1) OsO4, H20, OH H tO H H__ po R9 2) Na104, H20 F R8 N Oprot H N R8 R9 // IF R' SCHEME 2 (continued N Oprot R a Rua N R + HN N R I \ R OH Va R8 NaBH (OAc) 3 C ! CHCHC ! N, Oprot" Cl2CH2CH2CI2 N Oprot Rua N N N R9 rS : OH pu R8 Cs2CO3 DMSO heat N Oprot R a \ ---N N R n DO ro SCHEME 2 (continued) deprotect OOH Ra / 0/ R8 RUZ R R$ XVI SCHEME 3 N N N allyl-MgBr N OH / R9 I R9 8 R8 R8 R'R' OBn Na104, KMn04 / R9 F R K2CO3 F R 8 OBn 0 0 OH Xm ° OH R F F 08 R XV ! ! SCHEME 3 (continued SCHEME 3 (continued) SCHEME 4 SCHEME 4 (continued) N Oprot H ra HCI'N N Razz R + o OH //F Rb Ra XXI I XXIII Na (OAc) 3BH 4A mol. sieves OCHzCHzC ! N Oprot CICH2CH2CI N Oprot Ra N 0 oh N N XXIV R8 Cs2CO3 DMSO heat na N ZON rob Rb Oprot N R 8 RUZ SCHEME 4 (continued) deprotect Ra N Ra 0 ZON Rab OU R 1 8 Rob S R$ XXV XXV SCHEME 5 SCHEME 5 (continued) Rua N Oprot + 0 H-N N O b (y-OH R r J XXIX I sub. XXIX R$ XXI I I N Oprot Ra \ Na Ac0 B Na (AcO) 3BH r--N N-\ Na 3 -N N R9 4A MS, CICH2CH2CI R 'p zou 3N sub XXX Rsub Ra Ra CS2C03 N-. DMSO (0. 1 M) Rsub 0 R Oprot rus R8 SCHEME 5 (continued) Ra deprotect N Rsub /\ d r-o (/N3\ Rsub OH \\ \/18 R8 Rus I SCHEME 6 nSUb nSUb sub sub 02N<\/4 °2 <\/4 Cs2C03 1. SnCl2, EtOH HO Cl XXXII 0 DMSO 2. H2S04 Bn0 Rsub'Rs \, Bn0 _) Rsub Rsub I sub' ,, Rs Rs Ra R Rsub Rsub a H2N Ra , (,-/OH CICH2COCI I' 0 12N aq NaHC03 i-PrOAc BnOe'2 EtOAc BnO _/ xxxiv Rs XXXV Rs Ra Da H Rsub . /Rsub HCI HN N HN HN di-tert-butyl- azodicarboxylate O O O Buzz EtOAc Bn0 \, Bn0 _ ; XXVI I ub Rsub psub'Rb' SCHEME 6 (continued) N Oprot 1. Na (AcO) 3BH N X HL 4Å MS XXXVI + 0 CICH2CH2CI R9 R9 HCI t F 2. HCI F R8 N-Oprot Ra sub N N- Pd/C, H2 Zozo R 0 THF, TFA X VII pS Rsub' N Oprot Ra Rsub CS2CO3 /N N \/Cs2C03 R9 \ \p p DMSO (0. 1 M F /\ HO \--/-nsub' SCHEME 6 (continued) a Ra sub /R N N N deprotect N Oprot Oprot R 9 1 Rsub R Ra Rsub N N N Or rus Zou sub' JCXV I I I R8 X>O (VI I I SCHEME 7 SCHEME 8 SCHEME 8 (continued SCHEME 8 (continued) SCHEME 9 \ H O R9 9 i \ , N Br R OH MnO N n-BuLi I R8 I R XLI I R8 R9 allyi-MgBr R OH N 5) \1 R8 R8 R 9- R8 R R 1) OsO4, H20, \/OH H OH protect dioxane zozo 2) Na104, H20 I R8 9 Oprot H N 0 I R8 SCHEME 9 (continued R9 Oprot N H N + han pu I V R8 NaBH (OAc) 3 CCHCnC ! /Oprot 9 Oprot N N N--\ zou OH OL <Y-oH I XLII R8 1. NaH, pyridine 2. CuBrSMe2, reflux /Oprot f~/-S N N \--/N 0 O R8 SCHEME 9 (continued deprotect R9 (OH O N N N -4 U R' 8 R XLIII BF3-Et2O Et3SiH v R 9 1 0 N 1-4 N \--/N /O R8 SCHEME 10 SCHEME 10 (continued) R2 R'OBn H OBn \ H HCI H2N ° Rsub/SXF ZIP F R8 N Oprot 0 R2 O. R NaBH (OAc) 3 N N OBn t3\ F XRsub Rsub R8 N Oprot 0 R2 / N N OH /-- w 9 THF, TFA F R4 0 ub F sub 8 Rs o r\ R SCHEME 10 (continued) O R2 CS2CO3 N N DMSO (0. 1 M) jazz R9 Oprot \/O Rsub R8 O 0 R 2 deprotect //-N N N AX 4 R O N Rsub R8 I _J R' SCHEME 11 0 R CH3NHOCH3 HCI oh O N Jf EDC. HCI, HOBT DMF, Et3N, pH 7 O Ra II LAH, Et20 N (CH3) OCH3 O N H o BnO CH2NH2 0 R Rsub O Ra sub II R : H NaBH OAc H CICH2CH2CI pH 6 0 Ra 0 zozo V NHCH2 H3C0 H O 7 \ sub Et3N BnO 0 H3CO2CA HCI OBn HCI Ra N OBn EtOAc EtOAc/\J HCI H2 N < nSUb Rsub SCHEME 11 (continued) 0 H3CO2CX N Oprot Ra N Bn N H I o HC/HsN \ R"r Rsub v R8 N Oprot Ra NaBH (OAc) 3 N X \ /| wN N < OBn _/ I OBn Et3N, CICH2CHzCl J F Rsub RU N Oprot Ra / Pd/C, H2 N N OH 9 THF, TFA R 0 0 F sub Ra R SCHEME 11 (continued) Ra CS2CO3 N N DMSO (0-IM) N /\ 9 Oprot \/O Rsub R8 Ra CS2CO3 vN Ns DMSO (0. 1 M) 0 0 Rsub 18 R SCHEME 12 /O Ra CH3NHOCH3 HCI ON OH EDC. HCI, HOBT DMF, Et3N, pH 7 O Ra ON N (CH3) OCH3 LAH, Et20 H o BnO CH2NH2 O Ra JJ ! LJ >00 H NaBH OAc 3 H CICH2CH2CI pH 6 O Ra > ~ NHCH 1) BrCH2COBr 0 N EtOAc, H20, NaHC03 H 2) NaH, THF, DMF BnO

SCHEME 12 (continued SCHEME 12 (continued) SCHEME 13 Oprot N Oprot borane OH 0 N ruz R F R R8 Oprot N Oprot OMs \N Ns MsCI, NaN3, DMF XN3 I\ R I\ R8 R8 R8 R8 Rsub Han N Oprot CI O / H2, Pd/C N NH2 PO R9 i-Pr2NEt DMF F R8 SCHEME 13 (continued N Oprot H Rsub N N N oH H R NaBH3CN F MESH R 8 HO HO Oprot p Rsub N N N HO R t-Bu02CN=NC02t-Bu N Oprot 4O Rsub __<<, (n-C4Hg) 3P, THF N HO R8 SCHEME 13 CONT'D) 0 CS2CO3 Z--N,-, A DMSO \ heat \ \\ Oprot i heat, il J O i \/tw v 0 R8 \,-N deprotect \\ X O H N Ruz R SCHEME 14 BnO CHO + NH2CH2CH (OC2H5) 2 O Ra OH O N BnO CH2NHCH2CH (OC2H5) 2 H o EDC. HCI, HOBT DMF, Et3N, pH 7 I O Ra 6N HCI >00 THF THE H 0 R'0 Ra 10%Pd/C 0 \ SN Nn CH30H 0 OBn Ra O 0, -N N O XLVII OH SCHEME 15 SCHEME 15 (continued) SCHEME 16 sub. sub sub 1. Boc20, i-Pr2EtN >0 Ouzo H2N 2. DIBAL BocHN OH XLVIII sub I sub' C OH sub Rub OBI OBn N<\4 NaBH (OAc) 3 BocNH CICH2CH2CI Bn0 ) BnO BnO- HO sub HO sub ci Rs CI Rsub CI EtOAc/H20 NaHC03 ci 0 0 O Bon0 HO sub.. Bon N Rub CS2C3 BocN N DMF O O IL Bon0 _ I Rube SCHEME 16 (continued) HO sub Rsub / BocN N O O BnO J IL sub' Il ) L Rb' 1. (COCI) 2, Et3N 1. MsCI, iPr2NEt DMSO 2. Na02, t-BuOH 2. NaSEt, DMF/\ 2-Me-2-butene NaH2PO4 EtS sub HO sub sub Rsub 0 Rsub -BocN N BocN N \-io 0 O O L Bn0 Bn0 bu Rsub SCHEME 17 1. KOtBu, THF R2 cl Et -C02Et -N _ H2N Ph 2. 5% aqueous HCI HCI Lull R2 1. Boc20, NaHC03 )-C02H BocHN 2. LiAIH4, Et2O Lll U !

SCHEME 18 NH2'NH2 ¢40 EtSH ¢i CICH2COCI /+SH/9\S-SEt aq NaHCo3 Rsub Rsub EtOAc LIV sub Ra Ra OH Han S-SEt / HN HN CI O S-SEt Ra 0 S-SEt Ra di-tert-butyl-Rsub azodicarboxylate , HCt-HN N-<\ Bu3P, EtOAc HCI-HN\ N- O S-SEt LV SCHEME 18 (continued Ra 1. Na (AcO) 3BH Rsub N-) 4AMS HCI HN<NX +/ (4 +0 CICH2CH2 2. HCI 0 S-SET F R8 N Oprot Ra Rub i N DTT S-S Et I YF R8 LVI N Oprot Ra Rsub N /N N Cs2C03 R-SH DMF F R8 SCHEME 18 (continued) Ra Rsub r\T H/ N N deprotect N s -nu Oprot a Ra Rsub /- N Zur MCPBA NU IN oroxone k9 ho 9H HO \\ o LVII RU Ra Rab /R N N Nn \ O N N 0 9 is 1. or 2 9 ho , R8 LVIII LVt ! ! SCHEME 19

N t-butyl-S (O)-NH2 N NS (O)-t-butyl N"K N R9 Ti (lV) (OEt) 4 R9 F F R LIX LIX N N allyl-MgBr ß ys NHS (O)-t-butyl 1) OsO4 H20, N < d) oxane N / R 2) Na104, H20 F R8 N NHS (O)-t-butyl N H / R9 H 0 F , Ra LX SCHEME 19 (continued) NHS (O)-t-butyl R9 ¢ O if R 9 0 S F LX NaBH (OAc) 3 +, Ra CICH2CH2CI HAN N Rb OH V Rsub /N NHS (O)-t-butyl N Ra CS2CO3 N N Oh R _ Rsub r) SUb SCHEME 19 (continued)

N HS (O)-t-butyl Ra N \ \-N N-\ "L R /Rsub HCI N O R$ N sub N NH2Ra sub N N N ub 9 Rs /O 0 LXI

In a preferred embodiment of the instant invention the compounds of the invention are selective inhibitors of farnesyl-protein transferase. 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 5, is at least 100 times greater than the in vitro activity of the same compound against geranylgeranyl-protein transferase-type I in the assay described in Example 6. Preferably, a selective compound exhibits at least 1000 times greater activity against one of the enzymatic activities when comparing geranylgeranyl-protein transferase-type I inhibition and farnesyl-protein transferase inhibition.

It is also preferred that the selective inhibitor of famesyl-protein transferase is further characterized by : a) an ICso (a measure of in vitro inhibitory activity) for inhibition of the prenylation of newly synthesized K-Ras protein more than about 100-fold higher than the ECso for the inhibition of the farnesylation of hDJ protein.

When measuring such ICsos and ECSps the assays described in Example 10 may be utilized.

It is also preferred that the selective inhibitor of farnesyl-protein transferase is further characterized by : b) an ICso (a measurement of in vitro inhibitory activity) for inhibition of K4B- Ras dependent activation of MAP kinases in cells at least 100-fold greater than the ECso for inhibition of the farnesylation of the protein hDJ in cells.

It is also preferred that the selective inhibitor of farnesyl-protein transferase is further characterized by : c) an IC50 (a measurement of in vitro inhibitory activity) against H-Ras dependent activation of MAP kinases in cells at least 1000 fold lower than the inhibitory activity (IC50) against H-ras-CVLL (SEQ. ID. NO. : 1) dependent activation of MAP kinases in cells.

When measuring Ras dependent activation of MAP kinases in cells the assays described in Example 9 may be utilized.

In another preferred embodiment of the instant invention the compounds of the invention are dual inhibitors of farnesyl-protein transferase and geranylgeranyl-protein transferase type I. Such a dual inhibitor may be termed a Class II prenyl-protein transferase inhibitor and will exhibit certain characteristics when assessed in in vitro assays, which are dependent on the type of assay employed.

In a SEAP assay, such as described in Examples 9, it is preferred that the dual inhibitor compound has an in vitro inhibitory activity (IC50) that is less than about 121lu against K4B-Ras dependent activation of MAP kinases in cells.

The Class II prenyl-protein transferase inhibitor may also be characterized by : a) an IC50 (a measurement of in vitro inhibitory activity) for inhibiting K4B-Ras dependent activation of MAP kinases in cells between 0. 1 and 100 times the IC50 for inhibiting the famesylation of the protein hDJ in cells ; and b) an ICso (a measurement of in vitro inhibitory activity) for inhibiting 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.

The Class H prenyl-protein transferase inhibitor may also be characterized by : a) an IC50 (a measurement of in vitro inhibitory activity) 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 ; and b) an IC50 (a measurement of in vitro inhibitory activity) 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.

The Class I prenyl-protein transferase inhibitor may also be characterized by : a) an ICso (a measurement of in vitro inhibitory activity) against H-Ras dependent activation of MAP kinases in cells greater than 10-fold lower but less than 2, 500 fold lower than the inhibitory activity (IC50) against H-ras- CVLL (SEQ. ID. NO. : 1) dependent activation of MAP kinases in cells ; and b) an IC50 (a measurement of in vitro inhibitory activity) 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.

A method for measuring the activity of the inhibitors of prenyl-protein transferase, as well as the instant combination compositions, utilized in the instant methods against Ras dependent activation of MAP kinases in cells is described in Example 9.

In yet another embodiment, a compound of the instant invention may be a more potent inhibitor of geranylgeranyl-protein transferase-type I than it is an inhibitor of farnesyl-protein transferase.

The instant compounds are useful as pharmaceutical agents for mammals, especially for humans. These compounds may be administered to patients for use in the treatment of cancer. Examples of the type of cancer which may be treated with the compounds of this invention include, but are not limited to, colorectal carcinoma, exocrine pancreatic carcinoma, myeloid leukemias and neurological tumors. Such tumors may arise by mutations in the ras genes themselves, mutations in the proteins that can regulate Ras activity (i. e., neurofibromin (NF-1), neu, src, abl, lck, fyn) or by other mechanisms.

The compounds of the instant invention inhibit farnesyl-protein transferase and the faniesylation of the oncogene protein Ras. The instant compounds may also inhibit tumor angiogenesis, 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 compounds of this invention are 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 compounds of the invention 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 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 compounds of the instant invention are also useful in the prevention of restenosis after percutaneous transluminal coronary angioplasty by inhibiting neointimal formation (C. Indolfi et al. Nature medici7e, 1 : 541-545 (1995).

The instant compounds 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 be useful for the treatment of fungal infections.

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 the instant invention may also be useful in the prevention and treatment of endometriosis, uterine fibroids, dysfunctional uterine bleeding and endometrial hyperplasia.

In such methods of prevention and treatment as described herein, the prenyl-protein transferase inhibitors of the instant invention 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 prenyl-protein transferase inhibitor may be useful in further combination with drugs known to supress the activity of the ovaries and slow the growth of the endometrial tissue. Such drugs include but are not limited to oral contraceptives, progestins, danazol and GnRH (gonadotropin-releasing hormone) agonists.

Administration of the prenyl-protein transferase inhibitor may also be combined with surgical treatment of endometriosis (such as surgical removal of misplaced endometrial tissue) where appropriate.

The instant compounds may also be useful as inhibitors of corneal inflammation. These compounds may improve the treatment of corneal opacity which results from cauterization-induced corneal inflammation. The instant compounds may also be useful in reducing corneal edema and neovascularization. (K. Sonoda et al., Invest. OpAthalmol. Vis. Sci., 1998, vol. 39, p 2245-2251).

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.

Additionally, the compounds of the instant invention may be administered to a mammal in need thereof using a gel extrusion mechanism (GEM) device, such as that described in USSN 60/144, 643, filed on July 20, 1999, which is hereby incorporated by reference.

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 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 hydroxypropyl-methylcellulose or hydroxypropyl- cellulose, 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, hydroxypropylmethyl-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, flavoring 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-PLUSTM 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 injectables.

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 transdennal 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. Compounds of the present invention may also be delivered as a suppository employing bases such as cocoa butter, glycerinated gelatin, hydrogenated vegetable oils, mixtures of polyethylene glycols of various molecular weights and fatty acid esters of polyethylene glycol.

When a compound 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, sex and response of the individual patient, as well as the severity of the patient's symptoms.

In one exemplary application, a suitable amount of compound is administered to a mammal undergoing treatment for cancer. Administration occurs in an amount 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.

The compounds of the instant invention 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 compounds f 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 such a 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. It is further

understood that any of the therapeutic agents described herein may also be used in combination with a compound of the instant invention and an antineoplastic agent.

Examples of an antineoplastic agent include, in general, microtubule- stabilizing agents such as paclitaxel (also known as Taxol@), docetaxel (also known as Taxotere (g)), epothilone A, epothilone B, desoxyepothilone A, desoxyepothilone B or their derivatives) ; microtubule-disruptor agents ; alkylating agents, for example, nitrogen mustards, ethyleneimine compounds, alkyl sulfonate and other compounds with an alkylating action such as nitrosoureas, cisplatin, and dacarbazine ; anti- metabolites, for example, folic acid, purine or pyrimidine antagonists ; epidophyllotoxin ; an antineoplastic enzyme ; a topoisomerase inhibitor ; procarbazine ; mitoxantrone ; platinum coordination complexes ; biological response modifiers and growth inhibitors ; mitotic inhibitors, for example, vinca alkaloids and derivatives of podophyllotoxin ; cytotoxic antibiotics ; hormonal/anti-hormonal therapeutic agents, haematopoietic growth factors and antibodies (such as trastuzumab, also known as Herceptin'rm).

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, tamoxifen, ifosamide, melphalan, hexamethyl melamine, thiotepa, cytarabin, idatrexate, trimetrexate, dacarbazine, L-asparaginase, dactinomycin, mechlorethamine (nitrogen mustard), streptozocin, cyclophosphamide, carmustine (BCNU), lomustine (CCNU), procarbazine, mitomycin, cytarabine, etoposide, methotrexate, bleomycin, chlorambucil, camptothecin, CPT-11, topotecan, ara-C, bicalutamide, flutamide, leuprolide, pyridobenzoindole derivatives, interferons and interleukins. Particular examples of antineoplastic, or chemotherapeutic, agents are described, for example, by D. J. Stewart in"Nausea and Vomiting : Recent Research and Clinical Advances", Eds. J. Kucharczyk, et al., CRC Press Inc., Boca Raton, Florida, USA (1991), pages

177-203, especially page 188. See also, R. J. Gralla, et al., Cancer Treatment Reports, 68 (1), 163-172 (1984).

The preferred class of antineoplastic agents is the taxanes and the preferred antineoplastic agent is paclitaxel.

The compounds of the instant invention may also be co-administered with antisense oligonucleotides which are specifically hybridizable with RNA or DNA deriving from human ras gene. Such antisense oligonucleotides are described in U. S. Pat. No. 5, 576, 208 and PCT Publ. No. WO 99/22772. The instant compounds are particularly useful when co-administered with the antisense oligonucleotide comprising the amino acid sequence of SEQ. ID. NO : 2 of U. S. Pat. No. 5, 576, 208.

Certain compounds of the instant invention may exhibit very low plasma concentrations and significant inter-individual variation in the plasma levels of the compound. It is believed that very low plasma concentrations and high intersubject variability achieved following administration of certain prenyl-protein transferase inhibitors to mammals may be due to extensive metabolism by cytochrome P450 enzymes prior to entry of drug into the systemic circulation. Prenyl-protein transferase inhibitors may be metabolized by cytochrome P450 enzyme systems, such as CYP3A4, CYP2D6, CYP2C9, CYP2C19 or other cytochrome P450 isoform. If a compound of the instant invention demonstrates an affinity for one or more of the cytochrome P450 enzyme systems, another compound with a higher affinity for the P450 enzyme (s) involved in metabolism should be administered concomitantly.

Examples of compounds that have a comparatively very high affinity for CYP3A4, CYP2D6, CYP2C9, CYP2C19 or other P450 isoform include, but are not limited to, piperonyl butoxide, troleandomycin, erythromycin, proadifen, isoniazid, allylisopropylacetamide, ethinylestradiol, chloramphenicol, 2-ethynylnaphthalene and the like. Such a high affinity compound, when employed in combination with a compound of formula A, may reduce the inter-individual variation and increase the plasma concentration of a compound of formula A to a level having substantial therapeutic activity by inhibiting the metabolism of the compound of formula A.

Additionally, inhibiting the metabolism of a compound of the instant invention prolongs the pharmacokinetic half-life, and thus the pharmacodynamic effect, of the compound.

A compound of the present invention may be employed in conjunction with antiemetic agents to treat nausea or emesis, including acute, delayed, late-phase, and anticipatory emesis, which may result from the use of a compound of the present

invention, alone or with radiation therapy. For the prevention or treatment of emesis a compound of the present invention may be used in conjunction with other anti-emetic agents, especially neurokinin-I receptor antagonists, 5HT3 receptor antagonists, such as ondansetron, granisetron, tropisetron, and zatisetron, GABAB receptor agonists, such as baclofen, or a corticosteroid such as Decadron (dexamethasone), Kenalog, Aristocort, Nasalide, Preferid, Benecorten or others such as disclosed in U. S. Patent Nos. 2, 789, 118, 2, 990, 401, 3, 048, 581, 3, 126, 375, 3, 929, 768, 3, 996, 359, 3, 928, 326 and 3, 749, 712. For the treatment or prevention of emesis, conjunctive therapy with a neurokinin-1 receptor antagonist, a 5HT3 receptor antagonist and a corticosteroid is preferred.

Neurokinin-1 receptor antagonists of use in conjunction with the compounds of the present invention are fully described, for example, in U. S. Patent Nos. 5, 162, 339, 5, 232, 929, 5, 242, 930, 5, 373, 003, 5, 387, 595, 5, 459, 270, 5, 494, 926, 5, 496, 833, 5, 637, 699, 5, 719, 147 ; European Patent Publication Nos. EP 0 360 390, 0 394 989, 0 428 434, 0 429 366, 0 430 771, 0 436 334, 0 443 132, 0 482 539, 0 498 069, 0 499 313, 0 512 901, 0 512 902, 0 514 273, 0 514 274, 0 514 275, 0 514 276, 0 515 681, 0 517 589, 0 520 555, 0 522 808, 0 528 495, 0 532 456, 0 533 280, 0 536 817, 0 545 478, 0 558 156, 0 577 394, 0 585 913, 0 590 152, 0 599 538, 0 610 793, 0 634 402, 0 686 629, 0 693 489, 0 694 535, 0 699 655, 0 699 674, 0 707 006, 0 708 101, 0 709 375, 0 709 376, 0 714 891, 0 723 959, 0 733 632 and 0 776 893 ; PCT International Patent Publication Nos. WO 90/05525, 90/05729, 91/09844, 91/18899, 92/01688, 92/06079, 92/12151, 92/15585, 92/17449, 92/20661, 92/20676, 92/21677, 92/22569, 93/00330, 93/00331, 93/01159, 93/01165, 93/01169, 93/01170, 93/06099, 93/09116, 93/10073, 93/14084, 93/14113, 93/18023, 93/19064, 93/21155, 93/21181, 93/23380, 93/24465, 94/00440, 94/01402, 94/02461, 94/02595, 94/03429, 94/03445, 94/04494, 94/04496, 94/05625, 94/07843, 94/08997, 94/10165, 94/10167, 94/10168, 94/10170, 94/11368, 94/13639, 94/13663, 94/14767, 94/15903, 94/19320, 94/19323, 94/20500, 94/26735, 94/26740, 94/29309, 95/02595, 95/04040, 95/04042, 95/06645, 95/07886, 95/07908, 95/08549, 95/11880, 95/14017, 95/15311, 95/16679, 95/17382, 95/18124, 95/18129, 95/19344, 95/20575, 95/21819, 95/22525, 95/23798, 95/26338, 95/28418, 95/30674, 95/30687, 95/33744, 96/05181, 96/05193, 96/05203, 96/06094, 96/07649, 96/10562, 96/16939, 96/18643, 96/20197, 96/21661, 96/29304, 96/29317, 96/29326, 96/29328, 96/31214, 96/32385, 96/37489, 97/01553, 97/01554, 97/03066, 97/08144, 97/14671, 97/17362, 97/18206, 97/19084, 97/19942 and 97/21702 ; and in British Patent Publication Nos. 2 266 529, 2 268 931, 2 269 170,

2 269 590, 2 271 774, 2 292 144, 2 293 168, 2 293 169, and 2 302 689. The preparation of such compounds is fully described in the aforementioned patents and publications.

A particularly preferred neurokinin-1 receptor antagonist for use in conjunction with the compounds of the present invention is 2- (R)- (1- (R)- (3, 5- bis (trifluoromethyl) phenyl) ethoxy)-3- (S)- (4-fluorophenyl)-4- (3- (5-oxo-lH, 4H-1, 2, 4- triazolo) methyl) morpholine, or a pharmaceutically acceptable salt thereof, which is described in U. S. Patent No. 5, 719, 147.

For the treatment of cancer, it may be desirable to employ a compound of the present invention in conjunction with another pharmacologically active agent (s). A compound of the present invention and the other pharmacologic- ally active agent (s) may be administered to a patient simultaneously, sequentially or in combination. For example, the present compound may employed directly in combination with the other active agent (s), or it may be administered prior, concurrent or subsequent to the administration of the other active agent (s). In general, the currently available dosage forms of the known therapeutic agents for use in such combinations will be suitable.

For example, a compound of the present invention may be presented together with another therapeutic agent in a combined preparation, such as with an antiemetic agent for simultaneous, separate, or sequential use in the relief of emesis associated with employing a compound of the present invention and radiation therapy.

Such combined preparations may be, for example, in the form of a twin pack. A preferred combination comprises a compound of the present invention with antiemetic agents, as described above.

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 the instant 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 co- administered with compounds that are selective inhibitors of geranylgeranyl protein transferase.

In particular, if the compound of the instant invention is a selective inhibitor of famesyl-protein transferase, co-administration with a compound (s) that is a selective inhibitor of geranylgeranyl protein transferase may provide an improved therapeutic effect.

In particular, the compounds disclosed in the following patents and publications may be useful as farnesyl pyrophosphate-competitive inhibitor component of the instant composition : U. S. Ser. Nos. 08/254, 228 and 08/435, 047.

Those patents and publications are incorporated herein by reference.

In practicing methods of this invention, which comprise administering, simultaneously or sequentially or in any order, two or more of a protein substrate- competitive inhibitor and a farnesyl pyrophosphate-competitive inhibitor, such administration can be orally or parenterally, including intravenous, intramuscular, intraperitoneal, subcutaneous, rectal and topical routes of administration. It is preferred that such administration be orally. It is more preferred that such administration be orally and simultaneously. When the protein substrate-competitive inhibitor and farnesyl pyrophosphate-competitive inhibitor are administered sequentially, the administration of each can be by the same method or by different methods.

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, and WO 98/44797, published on October 15, 1998, which are 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 physiological ligand to the ocvß3 integrin, which selectively antagonize, inhibit or counteract binding of a physiological ligand to the avp5 integrin, which antagonize, inhibit or counteract binding of a physiological ligand to both the avp3 integrin and the (xvß5 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 alpl, a2pl, a5pl, a6pl and oe6ß4 integrins. The term also refers to antagonists of any combination of avß3 integrin, avp5 integrin, α1ß1, α2ß1, α5ß1, α6ß1 and α6ß4 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.

The instant compounds may also be useful in combination with an inhibitor of 3-hydroxy-3-methylglutaryl-CoA reductase (HMG-CoA reductase) for the treatment of cancer. Compounds which have inhibitory activity for HMG-CoA reductase can be readily identified by using assays well-known in the art. For example, see the assays described or cited in U. S. Patent 4, 231, 938 at col. 6, and WO 84/02131 at pp. 30-33. The terms"HMG-CoA reductase inhibitor"and "inhibitor of HMG-CoA reductase"have the same meaning when used herein.

Examples of HMG-CoA reductase inhibitors that may be used include but are not limited to lovastatin (MEVACOR@ ; see US Patent No. 4, 231, 938 ; 4, 294, 926 ; 4, 319, 039), simvastatin (ZOCOR ; see US Patent No. 4, 444, 784 ; 4, 820, 850 ; 4, 916, 239), pravastatin (PRAVACHOL ; see US Patent Nos. 4, 346, 227 ; 4, 537, 859 ; 4, 410, 629 ; 5, 030, 447 and 5, 180, 589), fluvastatin (LESCOL ; see US Patent Nos. 5, 354, 772 ; 4, 911, 165 ; 4, 929, 437 ; 5, 189, 164 ; 5, 118, 853 ; 5, 290, 946 ; 5, 356, 896), atorvastatin (LIPITOR ; see US Patent Nos. 5, 273, 995 ; 4, 681, 893 ; 5, 489, 691 ; 5, 342, 952) and cerivastatin (also known as rivastatin and BAYCHOL ; see US Patent No. 5, 177, 080). The structural formulas of these and additional HMG- CoA reductase inhibitors that may be used in the instant methods are described at page 87 of M. Yalpani,"Cholesterol Lowering Drugs", CZ2entistry & Industry, pp. 85- 89 (5 February 1996) and US Patent Nos. 4, 782, 084 and 4, 885, 314. The term HMG- CoA reductase inhibitor as used herein includes all pharmaceutically acceptable lactone and open-acid forms (i. e., where the lactone ring is opened to form the free acid) as well as salt and ester forms of compounds which have HMG-CoA reductase inhibitory activity, and therefor the use of such salts, esters, open-acid and lactone forms is included within the scope of this invention. An illustration of the lactone portion and its corresponding open-acid form is shown below as structures I and H. HO p HO COOH ***COH Lactone Open-Acid 1 11

In HMG-CoA reductase inhibitors where an open-acid form can exist, salt and ester forms may preferably be formed from the open-acid, and all such forms are included within the meaning of the term"HMG-CoA reductase inhibitor"as used herein.

Preferably, the HMG-CoA reductase inhibitor is selected from lovastatin and simvastatin, and most preferably simvastatin. Herein, the term"pharmaceutically acceptable salts"with respect to the HMG-CoA reductase inhibitor shall mean non- toxic salts of the compounds employed in this invention which are generally prepared by reacting the free acid with a suitable organic or inorganic base, particularly those formed from cations such as sodium, potassium, aluminum, calcium, lithium, magnesium, zinc and tetramethylammonium, as well as those salts formed from amines such as ammonia, ethylenediamine, N-methylglucamine, lysine, arginine, ornithine, choline, N, N'-dibenzylethylenediamine, chloroprocaine, diethanolamine, procaine, N-benzylphenethylamine, 1-p-chlorobenzyl-2-pyrrolidine-1'-yl- methylbenzimidazole, diethylamine, piperazine, and tris (hydroxymethyl) aminomethane. Further examples of salt forms of HMG-CoA reductase inhibitors may include, but are not limited to, acetate, benzenesulfonate, benzoate, bicarbonate, bisulfate, bitartrate, borate, bromide, calcium edetate, camsylate, carbonate, chloride, clavulanate, citrate, dihydrochloride, edetate, edisylate, estolate, esylate, fumarate, gluceptate, gluconate, glutamate, glycollylarsanilate, hexylresorcinate, hydrabamine, hydrobromide, hydrochloride, hydroxynapthoate, iodide, isothionate, lactate, lactobionate, laurate, malate, maleate, mandelate, mesylate, methylsulfate, mucate, napsylate, nitrate, oleate, oxalate, pamaote, palmitate, panthothenate, phosphate/diphosphate, polygalacturonate, salicylate, stearate, subacetate, succinate, tannate, tartrate, teoclate, tosylate, triethiodide, and valerate.

Ester derivatives of the described HMG-CoA reductase inhibitor compounds may act as prodrugs which, when absorbed into the bloodstream of a

warm-blooded animal, may cleave in such a manner as to release the drug form and permit the drug to afford improved therapeutic efficacy.

Similarly, the instant compounds may be useful in combination with agents that are effective in the treatment and prevention of NF-1, restenosis, polycystic kidney disease, infections of hepatitis delta and related viruses and fungal infections.

If formulated as a fixed dose, such combination products employ the combinations of this invention within the dosage range described above 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.

The instant compounds may also be useful in combination with prodrugs of antineoplastic agents. In particular, the instant compounds may be co-administered either concurrently or sequentially with a conjugate (termed a "PSA conjugate") which comprises an oligopeptide, that is selectively cleaved by enzymatically active prostate specific antigen (PSA), and an antineoplastic agent.

Such co-administration will be particularly useful in the treatment of prostate cancer or other cancers which are characterized by the presence of enzymatically active PSA in the immediate surrounding cancer cells, which is secreted by the cancer cells.

Compounds which are PSA conjugates and are therefore useful in such a co-administration, and methods of synthesis thereof, can be found in the following patents, pending patent applications and publications which are herein incorporated by reference : U. S. Patent No. 5, 599, 686, granted on Feb. 4, 1997 ; WO 96/00503 (January 11, 1996) ; USSN 08/404, 833, filed on March 15, 1995 ; USSN 08/468, 161, filed on June 6, 1995 ; U. S. Patent No. 5, 866, 679, granted on February 2, 1999 ; WO 98/10651 (March 19, 1998) ; USSN 08/926, 412, filed on September 9, 1997 ; WO 98/18493 (May 7, 1998) ; USSN 08/950, 805, filed on October 14, 1997 ;

WO 99/02175 (January 21, 1999) ; USSN 09/112, 656, filed on July 9, 1998 ; and WO 99/28345 (June 10, 1999) ; USSN 09/193, 365, filed on November 17, 1998.

Compounds which are described as prodrugs wherein the active therapeutic agent is released by the action of enzymatically active PSA and therefore may be useful in such a co-administration, and methods of synthesis thereof, can be found in the following patents, pending patent applications and publications, which are herein incorporated by reference : WO 98/52966 (November 26, 1998).

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

The compounds of the instant invention are also useful as a component in an assay to rapidly determine the presence and quantity of farnesyl- protein transferase (FPTase) in a composition. Thus the composition to be tested may be divided and the two portions contacted with mixtures which comprise a known substrate of FPTase (for example a tetrapeptide having a cysteine at the amine terminus) and farnesyl pyrophosphate and, in one of the mixtures, a compound of the instant invention. After the assay mixtures are incubated for an sufficient period of time, well known in the art, to allow the FPTase to farnesylate the substrate, the chemical content of the assay mixtures may be determined by well known immuno- logical, radiochemical or chromatographic techniques. Because the compounds of the instant invention are selective inhibitors of FPTase, absence or quantitative reduction of the amount of substrate in the assay mixture without the compound of the instant invention relative to the presence of the unchanged substrate in the assay containing the instant compound is indicative of the presence of FPTase in the composition to be tested.

It would be readily apparent to one of ordinary skill in the art that such an assay as described above would be useful in identifying tissue samples which contain farnesyl-protein transferase and quantitating the enzyme. Thus, potent inhibitor compounds of the instant invention may be used in an active site titration assay to determine the quantity of enzyme in the sample. A series of samples composed of aliquots of a tissue extract containing an unknown amount of farnesyl- protein transferase, an excess amount of a known substrate of FPTase (for example a tetrapeptide having a cysteine at the amine terminus) and farnesyl pyrophosphate are incubated for an appropriate period of time in the presence of varying concentrations

of a compound of the instant invention. The concentration of a sufficiently potent inhibitor (i. e., one that has a Ki substantially smaller than the concentration of enzyme in the assay vessel) required to inhibit the enzymatic activity of the sample by 50% is approximately equal to half of the concentration of the enzyme in that particular sample.

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.

EXAMPLE 1 Preparation of 14-amino-14- (1-methyl-lH-imidazol-5-yl)-8-oxa-2-thia-1, 18- diazatetracyclo [16. 2. 2. 137. 19 l3] tetracosa-3 (24), 4, 6, 9 (23), 10, 12-hexaene-10- carbonitrile 2, 2-dioxide Step A : Preparation of 4-Bromo-3-fluorobenzoic acid 4-Bromo-3-fluorotoluene (40. 0 g, 0. 212 mol) was heated at 90°C in H20 (200 mL)-pyridine (200 mL) with mechanical stirring under Ar. Potassium permanganate (KMn04) (67 g, 0. 424 mol) was added portionwise over 3 h. After 4 h, an HPLC of a filtered sample indicated 50 % conversion to the acid. An additional 30 g of KMn04 was added and heating continued overnight. HPLC indicated 81 % conversion. Further KMn04 was added portionwise with reaction monitoring by

HPLC until > 95% conversion was obtained. The reaction mixture was filtered through Celite, the filter pad washed with H20, aq NaOH and EtOH. The filtrate was concentrated to a small volume, then partitioned between 3N NaOH solution and diethyl ether. The aqueous basic layer was separated, cooled in an ice-H2O bath and acidified slowly with 6N HC1 solution to precipitate the white solid product. This was collected by suction filtration and dried at 40°C. in a vacuum oven overnight to give the title compound. mp 190-192°C.

1H NMR (CDC13) 8 7. 83 (dd, 1H, J = 2, 9 Hz), 7. 78 (dd, 1H, J = 2, 8 Hz), 7. 67-7. 71 (m, 1H).

Step B : Preparation of 4-bromo-3-fluorobenzvl alcohol 4-Bromo-3-fluorobenzoic acid (40. 8 g, 0. 187 mol) was dissolved in THF (250 ml) with magnetic stirring under Ar in an ice-H20 bath. The cloudy solution was treated dropwise with borane-THF complex (1 M) (374 mL, 0. 374 mol) over a 1 h period maintaining the internal temperature at < 10°C. The reaction mixture was left to warm to ambient temperature overnight, then cooled in an ice- H20 bath and treated dropwise with H20 (150 mL). The THF was removed on a rotary evaporator, and the residue partitioned between EtOAc and H20. The aqueous layer was extracted with EtOAc (3 x 100 mL), the organic layers combined, washed with brine, and dried (Na2S04), filtered, and concentrated to give the title compound as an oil which solidified on standing.

1H NMR (CDC13) 8 7. 52 (t, 1H, J = 8 Hz), 7. 16 (d, 1H, J = 9 Hz), 7. 02 (d, 1H, J = 8 Hz), 4. 67 (s, 2H), 1. 47 (br s, 1H).

Step C : Preparation of 2-fluoro-4-hydroxvmethylbenzonitrile 4-Bromo-3-fluorobenzyl alcohol (20 g, 0. 097 mol) was dissolved in DMF (100 mL) then placed under high vacuum for 15 min. The solution was then purged with Ar for 15 min. While purging continued, zinc cyanide (8 g, 0. 068 mol) and the catalyst, Pd [(PPh3)] 4, (5. 63 g, 0. 0049 mol) were added. The reaction mixture was heated at 95°C. under Ar for 18 h, then cooled to ambient temperature and added to H20. The mixture was extracted with EtOAc, then washed with 1M HCI, H20, brine, and dried (Na2S04). Filtration and concentration to dryness gave the title compound as a white solid after chromatography (silica gel, hexane : EtOAc, 6. 5 : 3. 5). lHNMR (CDC13) 67. 61 (t, 1H, J = 8 Hz), 7. 23-7. 29 (m, 2H), 4. 80 (d, 2H,

J = 6 Hz), 1. 93 (t, 1H, J = 6Hz).

Step D : Preparation of 2-Fluoro-4-formylbenzonitrile 2-Fluoro-4-hydroxymethylbenzonitrile (10 g, 0, 066 mol) and triethylamine (32. 3 mL, 0. 231 mol) were dissolved in CH2C12 (100 mL)-DMSO (20 mL) at < 5°C with stirring and treated dropwise with a solution of pyridine-S03 complex (31. 5 g, 0. 198 mol) in DMSO (70 mL) maintaining the reaction mixture temperature at <10°C. The reaction mixture was stirred at 5°C for 1 hr after the addition, then at 20°C for 1 hr, then partitioned between CH2C12 and H20. The organic layer was separated, washed well with H20, brine, and dried (Na2S04).

Filtration and concentration gave the title compound after purification by chromatography (silica gel, hexane : EtOAc, 3 : 1).

1H NMR (CDC13) 6 10. 06 (d, 1H, J = 2 Hz), 7. 86 (dd, 1H, J = 5, 8 Hz), 7. 798 (dd, 1H, J = 1, 8 Hz), 7. 728 (dd, 1H, J = 1, 8 Hz).

Step E : Preparation of 2-Fluoro-4-[hydroxy-(3-methyl-3H-imidazol-4-yl)- methyl-benzonitrile 1-Methylimidazole (15. 88 mL, 0. 199 mol), dissolved in anhydrous THF (500 mL) in flame-dried glassware under Ar, was cooled to-78°C and treated with n-butyl lithium (1. 6M in hexane) (124 mL, 0. 199 mol) via syringe. After stirring for 1 hr chlorotriethylsilane (33. 4 mL, 0. 199 mol) was added and the reaction mixture was left to warm to ambient temperature overnight. The THF was removed in vacuum with gentle warming, and the residue was redissolved in dry THF (500 mL), cooled to -78°C, and treated with sec-butyl lithium (1. 3M in cyclohexane) (153 mL, 0. 199 mol) dropwise. After 1 hr this solution was cannulated into a solution of 2-fluoro-4- fbrmylbenzonitrile (27 g, 0. 181 mol) in THF (200 mL). After 15 min the cooling bath was removed, the mixture was stirred for 2 hr at ambient temperature, then was quenched with saturated NH4C1 solution. After 15 min 10% HC1 was added to pH = 3. After 0. 5 hr the THF was removed in vacuo, the mixture was made basic with solid Na2C03 and extracted with EtOAc (3 x 200 mL). The organics were combined, washed with 10% HC1 (3x), the aqueous acidic layers combined, made basic with solid Na2C03, extracted with EtOAc (3x), the organics combined, washed with brine, and dried (MgS04). Filtration and concentration to dryness gave the title compound.

Step F : Preparation of 2-Fluoro-4- (3-methyl-3H-imidazole-4-carbonyl)- benzonitrile 2-Fluoro-4- [hydroxy- (3-methyl-3H-imidazol-4-yl)-methyl]- benzonitrile (0. 655 g, 2. 83 mmol) and Mn02 (1. 23 g, 14. 2 mmol) were stirred in CH2C12 (50 mL) and CH3CN (5 mL) for 3 h. The solution was filtered and concentrated to yield the title compound.

Step G : Preparation of N-[(4-cyano-3-fluoro-phenyl)-(3-methyl-3H- imidazol-4-yl)-methvlenel-2-methypropanesulfinamide 2-Fluoro-4- (3-methyl-3H-imidazole-4-carbonyl)-benzonitrile (2. 56 g, 11. 2 mmol), titanium (IV) ethoxide (7. 02 mL, 33. 5 mmol) and commercially available (R)- (+)-2-methyl-2-propanesulfinamide (1. 35 g, 11. 17 mmol) were dissolved in anhydrous THF (100 mL) and heated at 75°C for 7 days. The solution was cooled, diluted with brine (100 mL), filtered through a celite pad and washed generously with EtOAc and H20. The filtrate was separated, dried (MgS04), and purified using Si02 chromatography (0-3% MeOH/CH2Cl2) to give the title compound.

Step H : Preparation of N- [4- f [tert-butyl (dimethyl) silylgoxy}-1-(4-cyano-3- fluorophenyl)-l- (1-methyl-1H-imidazol-5-yl) butyl]-2-methyylpropane- 2-sulfinamide Magnesium (0. 634 g, 24. 06 mmol) was flame dried in a 50 mL RB flask equipped with addition funnel and magnetic stirrer under N2. When the flask had cooled, anhydrous THF (6 mL), a pinch of iodine, and a THF solution of Rieke magnesium (1 mL) were added, followed by a small portion of (3-bromopropoxy)- tert-butyl (dimethyl) silane (6. 05 g, 24. 06 mmol) in THF (10 mL) with slight warming to initiate the reaction. The remainder of the bromide solution was added dropwise over 15 min. After 0. 5 h this Grignard solution was added to a solution of N [ (4- <BR> <BR> <BR> cyano-3-fluoro-phenyl)- (3-methyl-3H-imidazol-4-yl)-methylene]-2-methylpropane sulfinamide (1. 0 g, 3. 01 mmol) in THF (3 mL) with cooling in an ice-H20 bath.

Grignard solution was added until the starting imine was gone by hplc. After 2 h, the reaction mixture was quenched with H20, partitioned between CH2C12 and aqueous saturated NaHCO3 solution. The aqueous layer was washed with CH2C12 (2x), the organic layers combined and dried (MgS04), filtered and concentrated to give the

crude product. Purification on an ISCO Combiflash eluting with 1-3% MeOH/CH2Cl2 w/NH40H gave the title compound. MS (M+1).

Step I : Preparation of N- [1- (4-cyano-3-fluorophenyl)-4-hydroxy-1- (l-methyl- lH-imidazol-5-vl ! butyl]-2-methylpropane-2-sulfinamide TFA (1 mL) was added to a solution of N- [4-f [tert- butyl (dimethyl) silyl] oxy}-1- (4-cyano-3-fluorophenyl)-1- (1-methyl-lH-imidazol-5- yl) butyl]-2-metliylpropane-2-sulfinamide (0. 111 g, 0. 219 mmol) in CH3CN () : H20 (20 mL) with stirring at ambient temperature. After 0. 5 h aqueous saturated NaHC03 solution was added, the CH3CN was removed in vacuo, and the residue was chromatographed on a Chiralcel AD column eluting with 90/10, hexane/EtOH to give the two diastereomers of the title compound. MS (M+1) 393.

Step J : Preparation ofN- [l- (4-cyano-3-fluorophenyl)-4-oxo-1- (1-methyl-lH- imidazol-5-vl) butyl]-2-methylpropane-2-sulfinamide Sulfur trioxide pyridine complex (0. 775 g, 4. 866 mmol) in DMSO (5 mL) was added to a solution of (N- [1- (4-cyano-3-fluorophenyl)-4-hydroxy-1- (1- methyl-lH-imidazol-5-yl) butyl]-2-methylpropane-2-sulfinamide (0. 382 g, 0. 973 mmol) and Et3N (1. 36 mL, 0. 985 mmol) in CH2C12 (15 mL) at 0°C. in an ice-H2O bath, then left to come to room temperature overnight. The reaction mixture was partitioned between CH2C12 and aqueous saturated NaHCO3 solution, the aqueous layer washed with CH2C12 (3x), the organic layers combined and dried (MgS04), filtered and concentrated to give the crude product. Purification on an ISCO Combiflash eluting with 2-6 % MeOH/CH2Clz gave the title compound. MS (M+1) 391.

Step K : Preparation of 4- (3-methoxy-benzenesulfonyl)-piperazine-1-carboxylic acid test-butyl ester 1-tert-Butyloxypiperazine (4. 99 g, 26. 81 mmol) was added to a solution of 3-methoxy-benzenesulfonyl chloride (5. 54 g, 26. 81 mmol) and Et3N (4. 48 mL, 32. 17 mmol) in CH2C12 (50 mL) with stirring at 0°C. in an ice-H20 bath. After 15 min. the reaction mixture was added to H2O, the layers separated, the aqueous layer washed with CH2C12 (2x), the organic layers combined, washed with brine, and dried (Na2SO4). Filtration and concentration in vacuo to give the title compound.

Step L : Preparation of 4- (3-hydroxy-benzenesulfonyl)-piperazine-1-carboxylic acid tert-butyl ester 4- (3-Methoxy-benzenesulfonyl)-piperazine-l-carboxylic acid tert-butyl ester (8. 21 g, 0. 023 mol) and sodium ethanethiolate (3. 9 g, 0. 046 mol) were heated in DMF (50 mL) at reflux under Ar for 1 h, then concentrated in vacuo to remove a portion of the DMF. The residue was added to ice-H20 and extracted with EtOAc (3x). The aqueous basic layer was acidified with IN HCI and extracted with EtOAc (4 x 100mL). The organic layers were combined, washed with H2O (2x), brine, and dried (Na2S04). Filtration and concentration in vacuo gave the title compound. 1H NMR (CDC13) 5 7. 40 (t, 1H, J = 8 Hz), 7. 27-7. 29. (m, 1H), 7. 22 (d, 1H, J = 2 Hz), 7. 08 (dd, 1H, J = 2, 5 Hz), 6. 21 (br s, 1H), 3. 51 (t, 2H, J = 5 Hz), 2. 98 (t, 2H, J = 5 Hz), 1. 42 (s, 9H).

Step M : Preparation of 3-(piperazin-1-ylsulfonyl ! phenol hydrochloride HCI gas was bubbled into a solution of 4- (3-hydroxy- benzenesulfonyl)-piperazine-l-carboxylic acid tert-butyl ester (2. 3 g, 9. 5 mmol) in EtOAc (100 mL) at-20°C. with stirring for 10 min. The solution was stoppered, stirred for 1 h, purged with Ar for 15 min, then concentrated in vacuo to the title compound as a white solid. 1H NMR (d6-DMSO) 8 8. 97, br s, 1H), 7. 50 (t, 1H, J = 8 Hz), 7. 12-7. 19 (m, 2H), 3. 175 (d, 2H, J = 5 Hz), 3. 12 (t, 2H, J = 5 Hz).

Step N : Preparation of N-[1-(4-cyano-3-fluorophenyl)-4-{4-[(3- hydroxyphenyl) sulfonyl] piperazin-1-yl}-1- (1-methyl-lH-imidazol-5- yl) butvl]-2-methvlpropane-2-sulfinamide 3- (Piperazin-1-ylsulfonyl) phenol hydrochloride (0. 065 g, 0. 231 mmol) was dissolved in MeOH (5 mL), neutralized with Et3N (0. 032 mL, 0. 231 mmol), then treated with N- [1- (4-cyano-3-fluorophenyl)-4-oxo-1- (1-methyl-lH-imidazol-5- yl) butyl]-2-methylpropane-2-sulfinamide (0. 086 g, 0. 220 mmol). The pH of the solution was adjusted to 5 with HOAc, then NaCNBH3 (0. 021 g, 0. 330 mmol) was added, and the pH was adjusted again to 5. After stirring at ambient temperature for 18 h, the reaction mixture was concentrated in vacuo, partitioned between CH2C12 and aqueous saturated NaHCO3 solution, the aqueous layer washed with CH2CI2 (2x), the organic layers combined and dried (MgS04), filtered and concentrated to give the title product. MS (M+1) 617.

Step 0-Preparation of 14-amino-14-(1-methyl-lH-imidazol-5-yl)-8-oxa-2-thia- 1, 18-diazatetracyclo [16. 2. 2. 13,7.19,13]tetracosa-3 (24), 4, 6, 9 (23), 10, 12- hexaene-I0-carbonitrile 2, 2-dioxide N-[1-(4-cyano-3-fluorophenyl)-4-{4-[(3-hydroxyphenyl)sulfony l] piperazin-1-yl}-1- (1-methyl-lH-imidazol-5-yl) butyl]-2-methylpropane-2-sulfinamide (0. 115 g, 0. 186 mmol) was dissolved in DMF (19 mL) and treated with Cs2CO3 (0. 304 g, 0. 93 mmol). After 24 h at 80°C, the reaction mixture was concentrated in vacuo, partitioned between CH2Clz and H20, the aqueous layer washed with CH2C12 (2x), the organic layers combined, washed with brine, and dried (MgS04). Filtration and concentration in vacuo gave the protected product which was dissolved in MeOH (10 mL) and treated with 4M HC1 in dioxane (5 mL) with stirring. After 15 min. the reaction mixture was concentrated in vacuo, dissolved in H2O : 0. 1% TFA (3 mL), and purified by RP LC on a Delta PrepPak eluting with 95 : 5 to 5 : 95 H20 (0. 1%TFA) : CH3CN(0.1%TFA) and lyophilized to give the product as the TFA salt of a mixture of enantiomers. This product was chromatographed on a Chiralcel OD column eluting with 0. 1% DEA : hexane/ EtOH, 60/40 to give the title compound as a single enantiomer. MS (M+1) 493.

EXAMPLE 2 Preparation of 14-amino-4-chloro-14-(1-methyl-lH-imidazol-5-yl)-20-oXo-8-ox a- 1, 18-diazatetracyclo [16. 2. 2. 13, 1'] tetracosa-3 (24), 4, 6, 9 (23), 10, 12-hexaene-10- carbonitrile trifluoroacetate Step A : Preparation of test-butyl (4-chloro-3-methylphenoxv) diphenylsilane

4-Chloro-3-methylphenol (33. 22 g, 0. 233 mol), tert-butyldiphenylsilyl chloride (62. 89 g, 0. 233 mol), and imidazole (47. 59 g, 0. 699 mol) in DMF (200 mL) were heated at 70°C. for 18 h. The reaction mixture was partitioned between EtOAc/hexane and H2O, the organic layer separated, washed with H2O, brine, and dried (An2SO4). Filtration and concentration to dryness gave the title compound which was used without purification. <BR> <BR> <P>Step B : Preparation of tert-butyl (3-bromomethyl-4-<BR> chlorophenoxv) diphenylsilane tert-Butyl (4-chloro-3-methylphenoxy) diphenylsilane (88. 69 g, 0. 233 mol), N-bromosuccinimide (49. 8 g, 0. 279 mol), and AIBN (3. 82 g, 0. 023 mol) were heated at reflux in CC14 (500 mL) for 7 h. The reaction mixture was concentrated in vacuo, dissolved in 10% EtOAc/hexane, the succinimide filtered off, and the filtrate concentrated in vacuo to give a mixture of the mono-and di-bromides.

Chromatography (Si02) (100% hexane) gave the title compound.

Step C : Preparation of tet-butyl 4- (5- f [tet-butyl (diphenyl) silyl] oxy}-2- chlorobenzyl)-3-oxopiperazine-1-carboxylate Sodium hydride (60% dispersion in mineral oil) (3. 50 g, 0. 0874 mmol) was washed with hexane, suspended in DMF (100 mL) at 0°C. with stirring under Ar, then treated with tert-Butyl-3-oxopiperazine-l-carboxylate (15. 0 g, 0. 728 mol). After 1 h, tert-butyl (3-bromomethyl-4-chlorophenoxy) diphenylsilane (33. 34 g, 0. 0728 mol) in DMF (50 mL) was added dropwise, and the reaction mixture was left to warm to room temperature overnight. The reaction mixture partitioned between EtOAc/hexane (50/50) and H20, the aqueous layer washed with EtOAc/hexane, the organics combined, washed with H2O, brine, and dried (Na2S04). Filtration and concentration to dryness gave the title compound which was used without purification.

Step D : Preparation of 1- (5-1 [tert-butyl (diphenyl) silyloxy}-2- chlorobenzyl) piperazin-2-one HC1 gas was bubbled into a solution of tei-t-butyl 4- (5- { [tert- butyl (diphenyl) silyl] oxy}-2-chlorobenzyl)-3-oxopiperazine-1-carboxylate (0. 20 g, 0. 345 mmol) in EtOAc (20 mL) at 0°C. with stirring for 10 min. The solution was stoppered, stirred for 0. 5 h, purged with Ar for 15 min, then concentrated

in vacuo. The residue was neutralized with aqueous saturated NaHCO3 solution, extracted with CH2Cl2 (3x), the organics combined, dried (MgS04), filtered, and concentrated in vacuo to give the title compound. MS (M+1) 479.

Step E : Preparation of 4-[1-amino-4-[4-(5-{[tert-butyl (diphenyl) silyl] oxy}-2- chlorobenzyl)-3-oxopiperazin-1-yl]-1- (1-methyl-lH-imidazol-5-<BR> yl) butyl]-2-fluorobenzonikile 1-(5-{[tert-Butyl (diphenyl) silyloxy}-2-chlorob enzyl) piperazin-2-one (0. 052 g, 0. 108 mmol) and N- [1- (4-cyano-3-fluorophenyl)-4-oxo-1- (1-methyl-1H imidazol-5-yl) butyl]-2-methylpropane-2-sulfinamide (Example 1, Step J) (0. 028 g, 0. 0. 072 mmol) were dissolved in MeOH (5 mL), the pH of the solution adjusted to 5 with HOAc, then NaCNBH3 (0. 0068 g, 0. 108 mmol) was added. After stirring at ambient temperature for 18 h, the reaction mixture was concentrated in vacuo, partitioned between CH2C12 and aqueous saturated NaHC03 solution, the aqueous layer washed with CH2C12 (2x), the organic layers combined and dried (MgS04).

Filtration and concentration in vacuo gave the protected product which was dissolved in MeOH, treated with 4M HC1 in dioxane (2 mL), stirred for 1. 5 h, concentrated in vacuo, partitioned between CH2Cl2 and aqueous saturated NaHCO3 solution, the aqueous layer washed with CH2C12 (2x), the organic layers combined and dried (MgS04). Filtration and concentration in vacuo, gave the title compound after chromatography on the ISCO Combiflash eluting with 1-5% MeOH/CH2Cl2 with NH40H. MS (M+1) 749.

Step F : Preparation of 14-amino-4-chloro-14- (1-methyl-lH-imidazol-5-yl)-20- oxo-8-oxa-1, 18-diazatetracyclo [16. 2. 2. 13n7. 19¢13] tetracosa- 3 (24), 4, 6, 9 (23 !, 10* 12-hexaene-10-carbonikile trifluoroacetate 4- [l-Amino-4- [4- (5-1 [tert-butyl (diphenyl) silyl] oxy}-2-chlorobenzyl)- 3-oxopiperazin-1-yl]-l- (l-methyl-lH-imidazol-5-yl) butyl]-2-fluorobenzonitrile (0. 033 g, 0. 044 mmol) and KF on alumina (0. 032 g, 0. 22 mmol) in CH3CN (5 mL) were heated at reflux under N2 for 18 h. The reaction mixture was concentrated in vacuo, then purified by RP LC on a Gilson eluting with 95 : 5 to 5 : 95 H20 (0. 1%TFA) : CH3CN (0. 1%TFA) and lyophilized to give the title compound. HR MS : theoretical : 491. 1957 ; measured : 491. 1951.

EXAMPLE 3 Preparation of 18-Bromo-8- [5- (1-methyl-1H-imidazolyl)]-8-methyl- 3, 7 : 1, 17-dimetheno-llH-10, 14-dioxo-12, 15-ethano-16H-2-oxa-9, 10 : 13, 14- tetrahvdro- [9, 12, 15] triaza-cvcloeicosene-4-carbonitrile Step A : Preparation ofN [1- (4-cyano-3-fluoro-phenyl)-1- (3-methyl-3H <BR> <BR> <BR> imidazol-4-yl)-ethyl]-2-methylpropanesulfinamide<BR> <BR> <BR> <BR> <BR> N [ (4-Cyano-3-fluoro-phenyl)- (3-methyl-3H-imidazol-4-yl)- methylene]-2-methylpropanesulfinamide (Example 1, Step G) (1. 50 g, 4. 51 mmol) was dissolved in anhydrous THF (30 mL) at 0°C and treated with a 3. 0M solution of MeMgBr (4. 50 mL, 13. 5 mmol) in Et20. After 15 min the reaction was quenched with aq. NH4Cl solution, diluted with saturated NaHCO3 solution and extracted with CH2C12 (3X). The combined organic layers were dried (MgSO4), filtered, concentrated, and recrystallized from 95% EtOAc/Hexane to give the title compound.

Step B : Preparation of (-)-4- [1-amino-1- (3-methyl-3H-imidazol-4-yl)- ethyl]-2-fluoro-benzonitrile bishydrochloride A cold methanolic HC1 solution (50 mL) was added to N [1- (4-cyano- <BR> <BR> <BR> 3-fluoro-phenyl)-1- (3-methyl-3H-imidazol-4-yl)-ethyl]-2-methylpropanesulfinamid e (0. 880 g, 2. 51 mmol) dissolved in MeOH (50 mL) and stirred for Ih at RT. After concentration and trituration with EtOAc the title compound was obtained as a bis HC1 salt as confirmed by chiral HPLC.

Using the procedure described above, but substituting (S)- (-)-2-methyl- 2-propanesulfinamide for (R)- (+)-2-methyl-2-propanesulfinamide in Step G, (+)-4- [1- amino-1- (3-methyl-3H-imidazol-4-yl)-ethyl]-2-fluoro-benzonitrile was obtained.

Step C : Preparation of4- [1-chloroacetylamino-1- (3-methyl-3H-imidazol-4-yl)- ethyl]-2-fluoro-benzonitrile A solution of 4- [1-amino-1- (3-methyl-3H-imidazol-4-yl)-ethyl]-2- fluoro-benzonitrile and Et3N in CH2C12 is treated with chloroacetyl chloride dropwise with stirring under Ar at 0°C. After 1 hr the reaction was partitioned between EtOAc and H2O, the organic layer separated, dried, filtered and concentrated to give the title compound.

Step D : Preparation of 4-Bromo-l- [methanesulfbnvloxy]-3-methylbenzene To a solution of 4-bromo-3-methylphenol (10. 2 g, 54. 5 mmol) in 100 mL of dichloromethane at 0 °C was added triethylamine (15. 2 mL, 109 mmol), followed by methanesulfonyl chloride (6. 33 mL, 81. 8 mmol). The reaction was stirred overnight, allowing it to warm to room temperature. The solution was poured into EtOAc, washed with water, saturated NH4Cl solution, saturated aq. NaHC03 and brine, dried (Na2S04), filtered, and concentrated in vacuo. The resulting product was isolated as a white solid which required no further purification.

Step F : Preparation of 4-Bromo-3-bromomethyl-1- [methanesulfonyloxvlbenzene To a solution of the product from Step K (10. 38 g, 39. 2 mmol) and N- bromosuccinimide (8. 61 g, 48. 4 mmol) in 80 mL of carbon tetrachloride was added 2, 2'-azobisisobutyronitrile (0. 89 g, 5. 4 mmol), and the reaction was heated at reflux overnight under argon. The solution was cooled to room temperature, concentrated in vacuo, slurried with 30% EtOAc/hexane solution, and filtered. The filtrate was washed with saturated aq. NaHC03 and brine, dried (Na2SO4), filtered, and concentrated in vacuo to provide a 2 : 1 mixture of the titled product and a tribromide as a yellow oil.

Step G : Preparation of 4-tert-butoxycarbonyl-2-piperazinone To a solution of 4-benzyloxycarbonyl-2-piperazinone (9. 44 g, 40. 3 mmol) and di-tert-butyldicarbonate (8. 80 g, 40. 3 mmol) in 100 mL of ethanol was added 10% palladium on carbon (1. 5 g). The solution was stirred at room temperature under an atmosphere of hydrogen for 3 days, then purged with argon. The mixture was filtered through celite, the filter pad was washed with ethanol/THF, and the filtrate was concentrated in vacuo to produce the titled product as a white solid.

Step H : Preparation of 1-[2-Bromo-5-((methanesulfonyl) oxy) benzyl]-4-tert- butoxycarbonyl-2-piperazinone Sodium hydride (0. 89 g, 22. 2 mmol, 60% mineral oil dispersion) was triturated with hexane. The flask was charged with 30 mL of dimethylformamide and cooled to 0°C. The product from Step M was added (3. 65 g, 18. 2 mmol), and the reaction was stirred for 15 minutes at 0°C. A solution of the crude product from Step

L (14. 7 g, ca. 24 mmol) in 40 mL of dimethylformamide was added slowly, and the reaction was allowed to warm to room temperature. After 24 hours, the solution was concentrated in vacuo, and partitioned between EtOAc and saturated NaHC03 solution. The aqueous phase was extracted with EtOAc, and the combined organics were washed with saturated NaHC03 soln and brine, dried (Na2S04), filtered, and concentrated in vacuo. The resulting product was purified by silica gel chromatography (50-75% EtOAc/hexane) to provide the titled product as a yellow solid.

Step I : Preparation of 1-[2-Bromo-5-((methanesulfonyl) oxy) benzyl]-2- piperazinone hydrochloride Through a solution of the product from Step N (7. 71 g, 16. 6 mmol) in 100 mL of ethyl acetate at 0°C was bubbled anhydrous HC1 gas for 10 minutes. After 30 minutes, the solution was concentrated in vacuo to provide the titled salt as a white foam which was used in the next reaction without further purification.

Step J : Preparation of Methanesulfonic acid 4-bromo-3- (4- { [1- (4-cyano-3- fluoro-phenyl)-1- (3-methyl-3H-imidazol-4-yl)-ethylcarbamoyl]- methyl}-2-oxo-piperazin-1-ylmethyl !-phenyl ester 1-[2-Bromo-5-((methanesulfonyl) oxy) benzyl]-2-piperazinone hydrochloride (Step O) and K2C03 are combined in DMF with stirring under Ar at 0°C. and treated with (+)-4- [1- (chloroacetylamino-1- (3-methyl-3H-imidazol-4-yl)- ethyl]-2-fluoro-benzonitrile (Step J) in DMF. After 12 h the reaction mixture was partitioned between EtOAc and H20, the organic layer separated, washed with dilute NaHC03 solution, H2O, brine, and dried (MgS04). Filtration and concentration to dryness gives the title compound.

Step K : Preparation of 18-Bromo-8- [5- (l-methyl-lH-imidazolyl)]-8-methyl- 3, 7 : 1, 17-dimetheno-11H-10, 14-dioxo-12, 15-ethano-16H-2-oxa- 9. 10 : 13, 14-tetrahydro- [9. 12, 15] triaza-cvcloeicosene-4-carbonitrile A solution of methanesulfonic acid 4-bromo-3-(4-{[1-(4-cyano-3- fluoro-phenyl)-1- (3-methyl-3H-imidazol-4-yl)-ethylcarbamoyl]-methyl}-2-oxo- piperazin-l-ylmethyl)-phenyl ester in DMSO is added dropwise to a mixture of Cs2CO3 (0. 750 g, 0. 750 mmol) in DMSO over a 2 hr period at 70°C. under Ar. The reaction mixture was stirred at 70°C. for 20 hr. The cooled mixture was partitioned between EtOAc and H2O, the aqueous layer washed with EtOAc, the organics combined, washed with H20, brine, and dried (Na2S04). Filtration and concentration to dryness gives the title compound.

EXAMPLE 4 Preparation of 18-Chloro-8- [5- (l-methyl-lH-imidazolyl)]-8-methyl-3, 7 : 1, 17- dimetheno-11H-10, 14-dioxo-12, 15-ethano-16H-2-oxa-9, 10 : 13, 14-tetrahydro- zu 12, 15] triaza-cycloeicosene-4-carbonitrile The titled compound is prepared using the procedures described in Example 3, except that in Step D, 4-chloro-3-methylphenol was substituted for 4-bromo-3- methylphenol.

EXAMPLE 5 In vitro inhibition of ras farnesyl transferase Transferase Assays. Isoprenyl-protein transferase activity assays are carried out at 30°C unless noted otherwise. A typical reaction contains (in a final volume of 50 L) : [3H] farnesyl diphosphate, Ras protein, 50 mM HEPES, pH 7. 5, 5 mM MgCl2, 5 mM dithiothreitol, 10 M ZnCl2, 0. 1% polyethyleneglycol (PEG) (15, 000-20, 000 mw) and isoprenyl-protein transferase. The FPTase employed in the assay is 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. After thermally pre-equilibrating the assay mixture in the absence of enzyme, reactions are 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 are allowed to stand for 15 m (to complete the precipitation process). After adding 2 mL of 100% ethanol, the reactions are vacuum-filtered through Whatman GF/C filters. Filters are 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 are run as described above, except inhibitors are prepared as concentrated solutions in 100% dimethyl sulfoxide and then diluted 20-fold into the enzyme assay mixture. Substrate concentrations for inhibitor

IC50 determinations are as follows : FTase, 650 nM Ras-CVLS (SEQ. ID. NO. : 1), 100 nM farnesyl diphosphate.

The compounds of the instant invention are tested for inhibitory activity against human FPTase by the assay described above.

EXAMPLE 6 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 , ut) : [3H] geranylgeranyl diphosphate, biotinylated Ras peptide, 50 mM HEPES, pH 7. 5, a modulating anion (for example 10 mM glycerophosphate or 5mM ATP), 5 mM MgC12, 10 aM ZnCl2, 0. 1% PEG (15, 000-20, 000 mw), 2 mM dithiothreitol, and geranylgeranyl-protein transferase type I (GGTase). 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. : 3). Reactions are initiated by the addition of GGTase and stopped at timed intervals (typically 15 min) by the addition of 200 pL 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. IC50 values are determined with Ras peptide near KM concentrations. Enzyme and substrate concentrations for inhibitor ICso determinations are as follows : 75 pM GGTase-1, 1. 6 1M Ras peptide, 100 nM geranylgeranyl diphosphate.

The compounds of the instant invention are tested for inhibitory activity against human GGTase-type I by the assay described above.

EXAMPLE 7 Cell-based in vitro ras farnesylation assay The cell line used in this assay is a v-ras line derived from either Ratl or NIH3T3 cells, which expressed viral Ha-ras p21. 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 (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 and 400 Ci [35S] methionin (1000 Ci/mmol). After an additional 20 hours, the cells are lysed in 1 ml lysis buffer (1% NP40/20 mM HEPES, pH 7. 5/5 mM MgCl2/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. Aliquots of lysates containing equal numbers of acid- precipitable counts are bought to 1 ml with IP buffer (lysis buffer lacking DTT) and immuno-precipitated 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 4°C, 200 tl of a 25% suspension of protein A-Sepharose coated with rabbit anti rat IgG is added for 45 min. The immuno-precipitates 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 NaCI) 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 autoradiographed. The intensities of the bands corresponding to fanzesylated and nonfarnesylated ras proteins are compared to determine the percent inhibition of famesyl transfer to protein.

EXAMPLE 8 Cell-based in vitro growth inhibition assay To determine the biological consequences of FPTase inhibition, the effect of the compounds of the instant invention on the anchorage-independent growth of Ratl cells transformed with either a v-ras, v-raf, or v-mos oncogene is tested. Cells transformed by v-Raf and v-Mos maybe included in the analysis to evaluate the specificity of instant compounds for Ras-induced cell transformation.

Rat 1 cells transformed with either v-ras, v-raf, or v-mos are seeded

at a density of 1 x 104 cells per plate (35 mm in diameter) in a 0. 3% top agarose layer in medium A (Dulbecco's modified Eagle's medium supplemented with 10% fetal bovine serum) over a bottom agarose layer (0. 6%). Both layers contain 0. 1% methanol or an appropriate concentration of the instant compound (dissolved in methanol at 1000 times the final concentration used in the assay). The cells are fed twice weekly with 0. 5 ml of medium A containing 0. 1 % methanol or the concentra- tion of the instant compound. Photomicrographs are taken 16 days after the cultures are seeded and comparisons are made.

EXAMPLE 9 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 EcoRl 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-ll and the ends filled in with Klenow DNA Polymerase 1. The SEAP DNA fragment was blunt end ligated into the pCMV-RE-AKI vector and the ligation products were transformed into DH5-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 1. 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-tenninal 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 Hpal 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 Hindi 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 Hindi. 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 Hindi 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, Y., 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 pl6T-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 pl6T-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 Sad site incorporated by the sense oligo and a Bgl-II fragment incorporated by the antisense oligo. The PCR fragment is trimmed with Sad 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-bA.

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 Hindou. 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) E-1 promoter and upstream of the 3'unstranslated region of the bovine growth hormone gene.

The plasmid pCMVIE-AKI-DHFR (Whang, Y., Silberklang, M., Morgan, A., Munshi, S., Lelmy, 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 Hindis. 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 DH5a 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 : 5'TCTCCTCGAGGCCACCATGGGGAGTAGCAAGAGCAAGCCTAAGGACCC CAGCCAGCGCCGGATGACAGAATACAAGCTTGTGGTGG 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 : 5'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 : 5'-GAGAGAATTCGCCACCATGACGGAATATAAGCTGGTGG-3' (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-Leu61 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 1. 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' (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-1 (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, pSMS630, 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-1 (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-1 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'-CTCTGTCGACAGATTACATTATAATGCATTTTTTAATTTTCACAC-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-1 (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-1 vector, using KpnI

and Sal I, and be directly ligated into the vector pCI (Promega) which has been digested with Kpnl and Sal 1. 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 1 Ocm tissue culture plates in DMEM + 10% fetal calf serum + 1X Pen/Strep + 1X glutamine + 1X NEAA. Cells are grown at 37°C in a 5% C02 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 10 cm plates 600 1ll of CaC12-DNA solution is added dropwise while vortexing to 600 1ll of 2X HBS buffer to give 1. 2 ml 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 CaP04-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 37°C under a 5% C02 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 10 ml 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 (100 pl/well) to which drug, diluted in media, has already been added in a volume of 100 Ill The final volume per well is 200 u. l with each drug concentration repeated in triplicate over a range of half-log steps.

Incubation of cells and drugs is for 36 hrs at 37'under C02. At the end of the incubation period, cells are examined micro-scopically for evidence of cell distress. Next, 100 pu ouf media containing the secreted alkaline phosphatase is removed from each well and transferred to a microtube array for heat treatment at 65°C 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 Ill 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-CaP04 precipitate for 10cm. plate of cells Ras expression plasmid (1 µg/µ l) 10 µl DSE-SEAP Plasmid (1 µg/µl) 2 µl Sheared Calf Thymus DNA (1 pg/lll) 8 zizi 2M CaCl2 74 il dH20 506 all 2X HBS Buffer 280mM NaCl lOmM KCl 1. 5mM Na2HP04 2H20 12mM dextrose 50mM HEPES Final pH = 7. 05 Luminesence Buffer (26ml) Assay Buffer 20ml Emerald Reagent (Tropix) 2. 5ml 100mM homoarginine 2. 5ml CSPD Reagent (Tropix) 1. 0ml Assay Buffer Add 0. 05M Na2C03 to 0. 05M NaHCO3 to obtain pH 9. 5.

Make 1mM in MgCl2

EXAMPLE 10 The processing assays employed are modifications of that described by DeClue et al rCancer Research 51, 712-717, 19911.

K4B-Ras processing inhibition assay PSN-1 (human pancreatic carcinoma) or viral-K4B-ras-transformed Ratl cells are used for analysis of protein processing. Subconfluent cells in 100 mm dishes are fed with 3. 5 ml of media (methionine-free RPMI supplemented with 2% fetal bovine serum or cysteine-free/methionine-free DMEM supplemented with 0. 035 ml of 200 mM glutamin (Gibco), 2% fetal bovine serum, respectively) containing the desired concentration of test compound, lovastatin or solvent alone. Cells treated with lovastatin (5-10 RM), a compound that blocks Ras processing in cells by inhibiting a rate-limiting step in the isoprenoid biosynthetic pathway, serve as a positive control. Test compounds are prepared as 1000x concentrated solutions in DMSO to yield a final solvent concentration of 0. 1%. Following incubation at 37°C for two hours 204, uCi/ml [35S] Pro-Mix (Amersham, cell labeling grade) is added.

After introducing the label amino acid mixture, the cells are incubated at 37°C for an additional period of time (typically 6 to 24 hours). The media is then removed and the cells are washed once with cold PBS. The cells are scraped into 1 ml of cold PBS, collected by centrifugation (10, 000 x g for 10 sec at room temperature), and lysed by vortexing in 1 ml of lysis buffer (1 % Nonidet P-40, 20 mM HEPES, pH 7. 5, 150 mM NaCl, 1 mM EDTA, 0. 5% deoxycholate, 0. 1% SDS, 1 mM DTT, 10, ug/ml AEBSF, 10 ug/ml aprotinin, 2 ig/ml leupeptin and 2 g/ml antipain).

The lysate is then centrifuged at 15, 000 x g for 10 min at 4°C and the supernatant saved.

For immunoprecipitation of Ki4B-Ras, samples of lysate supernatant containing equal amounts of protein are utilized. Protein concentration is determined by the bradford method utilizing bovine serum albumin as a standard. The appropriate volume of lysate is brought to 1 ml with lysis buffer lacking DTT and 8 Ag of the pan Ras monoclonal antibody, Y13-259, added. The protein/antibody mixture is incubated on ice at 4°C for 24 hours. The immune complex is collected on pansorbin (Calbiochem) coated with rabbit antiserum to rat IgG (Cappel) by tumbling at 4°C for 45 minutes. The pellet is washed 3 times with 1 ml of lysis buffer lacking

DTT and protease inhibitors and resuspended in 100 ill elution buffer (10 mM Tris pH 7. 4, 1% SDS). The Ras is eluted from the beads by heating at 95°C for 5 minutes, after which the beads are pelleted by brief centrifugation (15, 000 x g for 30 sec. at room temperature).

The supernatant is added to 1 ml of Dilution Buffer 0. 1% Triton X- 100, 5 mM EDTA, 50 mM NaCl, 10 mM Tris pH 7. 4) with 2 llg Kirsten-ras specific monoclonal antibody, c-K-ras Ab-1 (Calbiochem). The second protein/antibody mixture is incubated on ice at 4°C for 1-2 hours. The immune complex is collected on pansorbin (Calbiochem) coated with rabbit antiserum to rat IgG (Cappel) by tumbling at 4°C for 45 minutes. The pellet is washed 3 times with 1 ml of lysis buffer lacking DTT and protease inhibitors and resuspended in Laemmli sample buffer. The Ras is eluted from the beads by heating at 95°C for 5 minutes, after which the beads are pelleted by brief centrifugation. The supernatant is subjected to SDS-PAGE on a 12% acrylamide gel (bis-acrylamide : acrylamide, 1 : 100), and the Ras visualized by fluorography. hDJ processing inhibition assay PSN-1 cells are seeded in 24-well assay plates. For each compound to be tested, the cells are treated with a minimum of seven concentrations in half-log steps. The final solvent (DMSO) concentration is 0. 1%. A vehicle-only control is included on each assay plate. The cells are treated for 24 hours at 37°C/5% C02.

The growth media is then aspirated and the samples are washed with PBS. The cells are lysed with SDS-PAGE sample buffer containing 5% 2-mercaptoethanol and heated to 95°C for 5 minutes. After cooling on ice for 10 minutes, a mixture of nucleases is added to reduce viscosity of the samples.

The plates are incubated on ice for another 10 minutes. The samples are loaded onto pre-cast 8% acrylamide gels and electrophoresed at 15 mA/gel for 3-4 hours. The samples are then transferred from the gels to PVDF membranes by Western blotting.

The membranes are blocked for at least 1 hour in buffer containing 2% nonfat dry milk. The membranes are then treated with a monoclonal antibody to hDJ- 2 (Neomarkers Cat. # MS-225), washed, and treated with an alkaline phosphatase- conjugated secondary antibody. The membranes are then treated with a fluorescent detection reagent and scanned on a phosphorimager.

For each sample, the percent of total signal corresponding to the unprenylated species of hDJ (the slower-migrating species) is calculated by densitometry. Dose-response curves and EC50 values are generated using 4- parameter curve fits in SigmaPlot software.

EXAMPLE 11 Rapl processing inhibition assay Protocol A : Cells are labeled, incubated and lysed as described in Example 10.

For immunoprecipitation of Rapl, samples of lysate supernatant containing equal amounts of protein are utilized. Protein concentration is determined by the bradford method utilizing bovine serum albumin as a standard. The appropriate volume of lysate is brought to 1 ml with lysis buffer lacking DTT and 2 ag of the Rapl antibody, Rapl/Krevl (121) (Santa Cruz Biotech), is added. The protein/antibody mixture is incubated on ice at 4°C for 1 hour. The immune complex is collected on pansorbin (Calbiochem) by tumbling at 4°C for 45 minutes. The pellet is washed 3 times with 1 ml of lysis buffer lacking DTT and protease inhibitors and resuspended in 100 u. l elution buffer (10 mM Tris pH 7. 4, 1% SDS). The Rapl is eluted from the beads by heating at 95°C for 5 minutes, after which the beads are pelleted by brief centrifugation (15, 000 x g for 30 sec. at room temperature).

The supernatant is added to 1 ml of Dilution Buffer (0. 1% Triton X- 100, 5 mM EDTA, 50 mM NaCl, 10 mM Tris pH 7. 4) with 2 u. g Rapl antibody, Rapl/Krevl (121) (Santa Cruz Biotech). The second protein/antibody mixture is incubated on ice at 4°C for 1-2 hours. The immune complex is collected on pansorbin (Calbiochem) by tumbling at 4°C for 45 minutes. The pellet is washed 3 times with 1 ml of lysis buffer lacking DTT and protease inhibitors and resuspended in Laemmli sample buffer. The Rapl is eluted from the beads by heating at 95°C for 5 minutes, after which the beads are pelleted by brief centrifugation. The supernatant is subjected to SDS-PAGE on a 12% acrylamide gel (bis-acrylamide : acrylamide, 1 : 100), and the Rap 1 visualized by fluorography.

Protocol B :

PSN-1 cells are passaged every 3-4 days in 10cm plates, splitting near- confluent plates 1 : 20 and 1 : 40. The day before the assay is set up, 5x 106 cells are plated on 15cm plates to ensure the same stage of confluency in each assay. The media for these cells is RPM1 1640 (Gibco), with 15% fetal bovine serum and lx Pen/Strep antibiotic mix. The day of the assay, cells are collected from the 15cm plates by trypsinization and diluted to 400, 000 cells/ml in media. 0. 5ml of these diluted cells are added to each well of 24-well plates, for a final cell number of 200, 000 per well. The cells are then grown at 37 C overnight.

The compounds to be assayed are diluted in DMSO in 1/2-log dilutions. The range of final concentrations to be assayed is generally 0. 1-100 uM.

Four concentrations per compound is typical. The compounds are diluted so that each concentration is 1000x of the final concentration (i. e., for a 10 u. M data point, a 10 mM stock of the compound is needed).

2 aL of each 1000x compound stock is diluted into 1 ml media to produce a 2X stock of compound. A vehicle control solution (2 pLL DMSO to lml media), is utilized. 0. 5 ml of the 2X stocks of compound are added to the cells.

After 24 hours, the media is aspirated from the assay plates. Each well is rinsed with lml PBS, and the PBS is aspirated. 180 gel SDS-PAGE sample buffer (Novex) containing 5% 2-mercapto-ethanol is added to each well. The plates are heated to 100°C for 5 minutes using a heat block containing an adapter for assay plates. The plates are placed on ice. After 10 minutes, 20 FL of an RNAse/DNase mix is added per well. This mix is lmg/ml DNaseI (Worthington Enzymes), 0. 25 mg/ml Rnase A (Worthington Enzymes), 0. 5 M Tris-HCl pH 8. 0 and 50 mM MgCl2.

The plate is left on ice for 10 minutes. Samples are then either loaded on the gel, or stored at-70°C until use.

Each assay plate (usually 3 compounds, each in 4-point titrations, plus controls) requires one 15-well 14% Novex gel. 25 al of each sample is loaded onto the gel. The gel is run at 15 mA for about 3. 5 hours. It is important to run the gel far enough so that there will be adequate separation between 21 kd (Rapl) and 29kd (Rab6).

The gels are then transferred to Novex pre-cut PVDF membranes for 1. 5 hours at 30V (constant voltage). Immediately after transferring, the membranes are blocked overnight in 20ml Western blocking buffer (2% nonfat dry milk in Western wash buffer (PBS + 0. 1% Tween-20). If blocked over the weekend, 0. 02% sodium azide is added. The membranes are blocked at 4°C with slow rocking.

The blocking solution is discarded and 20ml fresh blocking solution containing the anti Rapla antibody (Santa Cruz Biochemical SC1482) at 1 : 1000 (diluted in Western blocking buffer) and the anti Rab6 antibody (Santa Cruz Biochemical SC310) at 1 : 5000 (diluted in Western blocking buffer) are added. The membranes are incubated at room temperature for 1 hour with mild rocking. The blocking solution is then discarded and the membrane is washed 3 times with Western wash buffer for 15 minutes per wash. 20ml blocking solution containing 1 : 1000 (diluted in Western blocking buffer) each of two alkaline phosphatase conjugated antibodies (Alkaline phosphatase conjugated Anti-goat IgG and Alkaline phosphatase conjugated anti-rabbit IgG [Santa Cruz Biochemical]) is then added. The membrane is incubated for one hour and washed 3x as above.

About 2 ml per gel of the Amersham ECF detection reagent is placed on an overhead transparency (ECF) and the PVDF membranes are placed face-down onto the detection reagent. This is incubated for one minute, then the membrane is placed onto a fresh transparency sheet.

The developed transparency sheet is scanned on a phosphorimager and the Rapla Minimum Inhibitory Concentration is determined from the lowest concentration of compound that produces a detectable Rapla Western signal. The Rapla antibody used recognizes only unprenylated/unprocessed Rap la, so that the precence of a detectable Rapla Western signal is indicative of inhibition of Rapla prenylation.

Protocol C : This protocol allows the determination of an ECso for inhibition of processing of Rapla. The assay is run as described in Protocol B with the following modifications. 20 ul of sample is run on pre-cast 10-20% gradient acrylamide mini gels (Novex Inc.) at 15 mA/gel for 2. 5-3 hours. Prenylated and unprenylated forms of Rapla are detected by blotting with a polyclonal antibody (Rapl/Krev-1 Ab#121 ; Santa Cruz Research Products #sc-65), followed by an alkaline phosphatase- conjugated anti-rabbit IgG antibody. The percentage of unprenylated Rapla relative to the total amount of Rap 1 a is determined by peak integration using ImagequantTM software (Molecular Dynamics). Unprenylated Rapla is distinguished from prenylated protein by virtue of the greater apparent molecular weight of the prenylated protein. Dose-response curves and EC5Q values are generated using 4-parameter curve fits in SigmaPlot software.

EXAMPLE 12 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 suboutaneously 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 or compound 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 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.