Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
INHIBITORS OF PRENYL-PROTEIN TRANSFERASE
Document Type and Number:
WIPO Patent Application WO/2000/034437
Kind Code:
A2
Abstract:
The present invention is directed to compounds which inhibit prenyl-protein transferase (FTase) 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:
DE SOLMS S JANE (US)
GRAHAM SAMUEL L (US)
SHAW ANTHONY W (US)
CICCARONE TERRENCE M (US)
STOKKER GERALD E (US)
Application Number:
PCT/US1999/029075
Publication Date:
June 15, 2000
Filing Date:
December 07, 1999
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
MERCK & CO INC (US)
SOLMS S JANE DE (US)
GRAHAM SAMUEL L (US)
SHAW ANTHONY W (US)
CICCARONE TERRENCE M (US)
STOKKER GERALD E (US)
International Classes:
C07D233/54; C07D401/12; C07D403/12; C07D413/12; C07D471/04; C07D521/00; (IPC1-7): C12N/
Foreign References:
US5739132A1998-04-14
Other References:
MUELLER ET AL.: 'Properties and reactions of substituted 1,2-thiazetidine 1,1-dioxides: synthesis of N-aminoalkylated 1,2-thiazetidine 1,1-dioxides' ARCH. PHARM. vol. 322, no. 8, 1989, pages 515 - 516, XP002930893
GELB M.H ET AL.: 'Protein prenylation: from discovery to prospects for cancer treatment' CURR. OPIN. CHEM. BIOL. vol. 2, no. 1, February 1998, pages 40 - 48, XP002930894
Attorney, Agent or Firm:
MERCK & CO., INC. (NJ, US)
MERCK & CO., INC. (NJ, US)
Download PDF:
Claims:
WHAT IS CLAIMED IS:
1. A compound of the formula I: wherein: RlaS R1b and RIC are independently selected from: a) hydrogen, b) unsubstituted or substituted aryl, unsubstituted or substituted heterocycle, C3Clo cycloalkyl, C2C6 alkenyl, C2C6 alkynyl, R9S (O) q, CN, N02, RSC (O), R8OC (O), (R8) 2NC (O),N (R15)2, OR 15,N (R8) S (O) qR8 or N3; c) ClC6 alkyl, unsubstituted or substituted by unsubstituted or substituted aryl, unsubstituted or substituted heterocycle, C3C 10 cycloalkyl, C2C6 alkenyl, C2C6 alkynyl, R8O, R9S (O) q, CN, R8C (O), R8OC (O), N (R8) 2, N3, or R8C (O) O; RI is independently selected from: a) H, b) unsubstituted or substituted C1C6 alkyl, c) unsubstituted or substituted aryl, d) unsubstituted or substituted heterocycle, e) (ClC6 alkyl) N (R8) 2, f)R8C (O) R8, g) (ClC6 alkyl) OR8, h)N (R8) 2, i)OR8, j)R8NHC (O) R8, k)R8C (O) N (R8) 2, 1) CF3, m) halo, n)C (O) OR8, o) C2C6 alkynyl, p) C2C6 alkenyl, q) perfluoroalkyl, r) N3, s) N02, t) CN, u) R9S (O) q, v) (C1C6 alkyl)NR8C(O) N (R8) 2, and alkyl)R14;w)(C1C6 R2 is independently selected from: a) hydrogen, b) CN, c) N02, d) halogen, e) aryl, unsubstituted or substituted, f) heteroaryl, unsubstituted or substituted, g) ClC6 alkyl, unsubstituted or substituted, h) OR8, i)N3, j) R9S(O)q, k) R8HC=CH, and 1) R8C=C ; R3 is independently selected from: a) H, b) CN, c) N02, d) halogen, e) C1C6 alkyl, unsubstituted or substituted, f) OR8, g) aryl, unsubstituted or substituted, h) heteroaryl, unsubstituted or substituted, and i) CF3; R5 and R6 are independently selected from: a) H, b) CN, c) N02, d) halogen, e) C1C6 alkyl, unsubstituted or substituted, f)N3, g) R9S (O) q, h)HC=CH2, i) HC=C, j) aryl, unsubstituted or substituted, k) heterocycle, unsubstituted or substituted, 1) CF30, m) CF3CH20, n) cycloalkyl, o) CF3, p) (C1C6 alkyl) N (R8) 2, q)(C1C6 alkyl) OR8, r) OR8, s) N (R8) 2, t)C (O) (C1C6 alkyl), u) (C1C6 alkyl) C (O) R8, v)C (O) O (C1C6 alkyl), w)C (O) N (R8) 2, x) (C1C6 alkyl) NR8C (O) N (R8) 2, y) (C1C6 alkyl) NR8C (O) R8, z)C2C6 alkynyl, aa)C2C6 alkenyl, bb) (ClC6 alkyl) N3, cc)(C1C6 alkyl) NR8S (O)(C1C6 alkyl) NR8S (O) q, and dd)(C1C6 alkyl) NR8 (C1C6(C1C6 alkyl) NR8 (C1C6 alkyl) N (R8) 2; R8 is independently selected from hydrogen, unsubstituted or substituted C 1C6 alkyl, unsubstituted or substituted aralkyl, unsubstituted or substituted heterocycle and unsubstituted or substituted aryl; R9 is independently selected from unsubstituted or substituted C1C6 alkyl, unsubstituted or substituted benzyl and unsubstituted or substituted aryl; R14 is unsubstituted saturated heterocycle; R15 is independently selected from a) hydrogen, unsubstituted or substituted aryl, unsubstituted or substituted heterocycle, C3C10 cycloalkyl, C2C6 alkenyl, C2C6 alkynyl, R9S (0) 2, R8C (O), R80C (O), (R8) 2NC (O), and b) C 1C6 alkyl, unsubstituted or substituted by unsubstituted or substituted aryl, unsubstituted or substituted heterocycle, C3C10 cycloalkyl, C2C6 alkenyl, C2C6 alkynyl, R8O, R9S (O) q, CN, R8C (O), R8OC (O), N (R8) 2, N3, or R8C (O) O; A1 and A2 are independently selected from: a) a bond, b)HC=CH, c)C=C, d) O, e) S (O) q, f)OC(O), g) C (O), h) C (O) O, and A3 is selected from a bond,C (=O), and C3C6 cycloalkyl; M is selected from CH2, NR8, O or S (O) q; Q is selected from S (O) q, CH2, C (=S), C (=NR8) or C (=O); W is selected from: a) heterocycle, and b) aryl; Y is selected from: a) aryl, and b) heterocycle; Z is selected from: a) aryl, b) heterocycle, c) C3C6 cycloalkyl, and d) a bond; m is 0,1,2,3 or 4; n is 0,1,2,3 or 4; pis 0,1,2,3 or 4; q is 0, 1 or 2; ris 3, or 4; sis 0,1,2,3 or 4; t is 0, 1, 2 or 3; x is ; provided that the moiety does not represent a bond; and provided that if attachment of to the rest of the molecule is through a nitrogen ring atom, then A1 is NR8, O, or S (O) q; or a pharmaceutically acceptable salt, an optical isomer or stereoisomer thereof.
2. A compound of the formula A: wherein: Rla Rlb and R I C are independently selected from: a) hydrogen, b) unsubstituted or substituted aryl; unsubstituted or substituted heterocycle, C3C10 cycloalkyl, C2C6 alkenyl, C2C6 alkynyl, R8O, R9S (O) q, CN, N02, R8C (O), R8OC (O), N (R8) 2, (R8) 2NC (O), C (O) N (R8), or N3 ; c) C1C6 alkyl, unsubstituted or substituted by unsubstituted or substituted aryl, unsubstituted or substituted heterocycle, C3C10 cycloalkyl, C2C6 alkenyl, C2C6 alkynyl, R80, R9S (O) q, CN, R8C (O), R80C (O), N (R8) 2, N3, or R8C (O) 0 ; R1 is selected from: a) H, b) unsubstituted or substituted ClC6 alkyl, c) unsubstituted or substituted aryl, d) unsubstituted or substituted heterocycle, e) (C1C6 alkyl) N (R8) 2, f)R8C (O) R8, g) (C1C6 alkyl) OR8, h)N (R8) 2, i)OR8, j)R8NHC (O) R8, k)R8C (O) N (R8) 2, 1) CF3, m) halo, n)C (O) OR8, o) C2C6 alkynyl, p) C2C6 alkenyl, q) perfluoroalkyl, r) N3, s) N02, t) CN, and u) R9S (O) q ; R2 is selected from: a) hydrogen, b) CN, c) N02, d) halogen, e) aryl, unsubstituted or substituted, f) heteroaryl, unsubstituted or substituted, g) CiC6 alkyl, unsubstituted or substituted, h) OR8, i) N3, j) R9S (O) q, k) R8HC=CH, and 1) R8C=C; R3 is selected from: a) H, b) CN, c) N02, d) halogen, e) C1C6 alkyl, unsubstituted or substituted, f) OR8, g) aryl, unsubstituted or substituted, h) heteroaryl, unsubstituted or substituted, and i) CF3; R4 is selected from: a) H, b) =O, or c) =S; R5 is selected from: a) H, b) CN, c) N02, d) halogen, e) C1C6 alkyl, unsubstituted or substituted, f) N3, g) R9S (O) q, h)HC=CH2, i) HC=C, j) aryl, unsubstituted or substituted, k) heterocycle, unsubstituted or substituted, 1) CF30, m) CF3CH20, n) C3Ciocycloalkyl, o) CF3, (ClC6 alkyl)(ClC6 alkyl) N (R8) 2, q) (C1C6 alkyl) OR8, r) OR8, s) N (R8) 2, t)C (O) (ClC6 alkyl), and u) (C1C6 alkyl) C (O) R8; R6 is selected from: a) H, b) C1C6 alkyl, unsubstituted or substituted, c) and d)C (O) (CiC6 alkyl); R8 is independently selected from hydrogen, unsubstituted or substituted C1C6 alkyl, unsubstituted or substituted benzyl, unsubstituted or substituted heterocycle and unsubstituted or substituted aryl; R9 is independently selected from unsubstituted or substituted C lC6 alkyl, unsubstituted or substituted benzyl and unsubstituted or substituted aryl; Al and A2 are independently selected from: a) a bond, b)HC=CH, c)C=C, d) O, e) S (O) q, f) OC (O), g) C (O), h) and A3 is selected from a bond,C (=O), and C3C6 cycloalkyl; M is selected from CH2, NH, O or S; W is selected from: a) hydrogen, b) heterocycle, and c) aryl; Y is selected from: a) aryl, and b) heterocycle; Z is selected from: a) aryl, b) heterocycle, c) C3C6 cycloalkyl, and d) a bond; mis 0,1,2,3 or 4; n is 3 or 4; pis 0,1,2,3 or 4; q is 0,1 or 2; r is 0,1, 2,3, or 4; sis 0,1,2,3 or 4; t is ; or a pharmaceutically acceptable salt, an optical isomer or stereoisomer thereof.
3. The compound according to Claim 2 of formula A: wherein: R1a, R1b and Ric are independently selected from: a) hydrogen, b) unsubstituted or substituted aryl; unsubstituted or substituted heterocycle, C3C10 cycloalkyl, C2C6 alkenyl, C2C6 alkynyl, R8O, R9S (O) q, CN, N02, R8C (O), R8OC (O), N (R8) 2, (R8) 2NC (O), C (O) N (R8), or N3; c) C 1C6 alkyl, unsubstituted or substituted by unsubstituted or substituted aryl, unsubstituted or substituted heterocycle, C3C10 cycloalkyl, C2C6 alkenyl, C2C6 alkynyl, R8O, R9S (O) q, CN, R8C (O), R8OC (O), N (R8) 2, N3, or R8C (O) O; RI is selected from: a) H, b) unsubstituted or substituted C1C alkyl, c) unsubstituted or substituted aryl, d) unsubstituted or substituted heterocycle, e)(C1C6(C1C6 alkyl) N (R8) 2, f) (C1C6 alkyl) C (O) R8, g) (C1C6 alkyl) OR8, h)N (R8) 2, i)OR8, j) (C1C6 alkyl) NHC (O) R8, k) (C1C6 alkyl) C (O) N (R8) 2, 1) CF3, and m) halo; R2 is selected from: a) hydrogen, b) CN, c) N02, d) halogen, e) aryl, unsubstituted or substituted, f) heteroaryl, unsubstituted or substituted, g) ClC6 alkyl, unsubstituted or substituted, h) OR8, i) N3, j) R9S (O) q, k) R8HC=CH, and 1) R8C#C; R3 is selected from: a) H, b) CN, c) N02, d) halogen, e) C1C6 alkyl, unsubstituted or substituted, f) OR8, g) aryl, unsubstituted or substituted, h) heteroaryl, unsubstituted or substituted, and i) CF3; R4 is selected from: a) H, b) =O, or c) =S; R5 is selected from: a) H, b) CN, c) N02, d) halogen, e) C1C6 alkyl, unsubstituted or substituted, f) N3, g) R9S (O) q, h)HC=CH2, i) HC=C, j) aryl, unsubstituted or substituted, k) heterocycle, unsubstituted or substituted, 1) CF30, m) CF3CH20, n) C3C 10 cycloalkyl, o) CF3, p) (C1C6 alkyl) N (R8) 2, alkyl)OR8,q)(C1C6 r) OR8, s) N (R8) 2, t)C (O) (C1C6 alkyl), and u)(C1C6 alkyl)(C1C6 alkyl) C (O) R8; R6 is selected from: a) H, b) C1C6 alkyl, unsubstituted or substituted, c) ORS, and d)C (O) (C1C6 alkyl); R8 is independently selected from hydrogen, unsubstituted or substituted C1C6 alkyl, benzyl and unsubstituted or substituted aryl; R9 is independently selected from unsubstituted or substituted ClC6 alkyl, benzyl and unsubstituted or substituted aryl; A1 is selected from: a) a bond, b)HC=CH, c)C=C, d) O, e) S (O) q, f) OC (O), g) C (O), h) C (O) O, and A2 is selected from: a) a bond, b) O, c) S (O) q, d) C (O), and e) NR8 ; A3 is selected from a bond,C (=O), and C3C6 cycloalkyl; M is selected from CH2 or NH; W is selected from: a) heterocycle, and b) aryl; Y is selected from: a) aryl, and b) heterocycle; Z is selected from: a) aryl, b) heterocycle, and c) a bond; mis 0,1,2,3 or 4; n is 3 or 4; pis 0,1,2,3 or 4; qis 0,1 or 2; ris 3, or 4; sis and tis ; or a pharmaceutically acceptable salt, an optical isomer or stereoisomer thereof.
4. The compound according to Claim 3 of formula B: wherein: Rla and Ric are independently selected from: a) hydrogen, b) unsubstituted or substituted aryl; unsubstituted or substituted heterocycle, C3C1o cycloalkyl, C2C6 alkenyl, C2C6 alkynyl, R8O, R9S (O) q, CN, N02, R8C (O), R80C (O), N (R8) 2, (R8) 2NC (O), C (O) N (R8), or N3; c) ClC6 alkyl, unsubstituted or substituted by unsubstituted or substituted aryl, unsubstituted or substituted heterocycle, C3C10 cycloalkyl, C2C6 alkenyl, C2C6 alkynyl, R80, R9S (O) q, CN, R8C (O), R8OC (O), N (R8) 2, N3, or R8C (O) O; Rl is selected from: a) H, b) unsubstituted or substituted ClC6 alkyl, c) unsubstituted or substituted aryl, d) unsubstituted or substituted heterocycle, e) (ClC6 alkyl) N (R8) 2, (C1C6 alkyl) C (O) R8, g) (C1C6 alkyl) OR8, h)N (R8) 2, i)OR8, j)(C1C6 alkyl)(C1C6 alkyl) NHC (O) R8, k) (C1C6 alkyl) C (O) N (R8) 2, 1) CF3, and m) halo; R2 is selected from: a) hydrogen, b) CN, c) N02, d) halogen, e) aryl, unsubstituted or substituted, f) heteroaryl, unsubstituted or substituted, g) C1C6 alkyl, unsubstituted or substituted, h) OR8, i) N3, j) R9S (O) q, k) R8HC=CH, and l) R8C#C; R3 is selected from: a) H, b) CN, c) N02, d) halogen, e) ClC6 alkyl, unsubstituted or substituted, f) OR8, g) aryl, unsubstituted or substituted, h) heteroaryl, unsubstituted or substituted, and i) CF3; R4 is selected from: a) H, b) =O, or c) =S; R5 is selected from: a) H, b) CN, c) N02, d) halogen, e) C1C6 alkyl, unsubstituted or substituted, f)N3, g) R9S (O) q, h)HC=CH2, i) HC=C, j) aryl, unsubstituted or substituted, k) heterocycle, unsubstituted or substituted, 1) CF30, m) CF3CH20, n) C3C 10 cycloalkyl, o) CF3, alkyl)N(R8)2,p)(C1C6 alkyl)OR8,q)(C1C6 r) OR8, s) N (R8) 2, t)C (O) (ClC6 alkyl), and u) (CIC6 alkyl) C (O) R8; R6 is selected from: a) H, b) C1C6 alkyl, unsubstituted or substituted, c) OR8, and d)C (O) (C1C6 alkyl) ; R8 is independently selected from hydrogen, unsubstituted or substituted C1C6 alkyl, benzyl and unsubstituted or substituted aryl; R9 is independently selected from unsubstituted or substituted C1C6 alkyl, benzyl and unsubstituted or substituted aryl; A1 is selected from: a) a bond, b)HC=CH, c)C#C, d) O, e) S (O) q, f) OC (O), g) C (O), h) C (O) O, and W is selected from: a) heterocycle, and b) aryl; Y is selected from: a) aryl, and b) heterocycle; Z is selected from: a) aryl, and b) heterocycle; mis 0,1,2,3 or 4; nis 0,1,2,3 or 4; pis 3 or 4; qis 0,1 or 2; sis and tis ; or a pharmaceutically acceptable salt, an optical isomer or stereoisomer thereof.
5. The compound according to Claim 1 of formula II: wherein: Ric is independently selected from: a) hydrogen, b) unsubstituted or substituted aryl, unsubstituted or substituted heterocycle, C2C6alkenyl,C2C6alkynyl,cycloalkyl, R9S (O) q, CN, N02, R8C (O), R8OC (O), (R8) 2NC (O),N (R15) 2, OR15,N (R8) S (O) qR8 or N3; c) C 1C6 alkyl, unsubstituted or substituted by unsubstituted or substituted aryl, unsubstituted or substituted heterocycle, C3C10 cycloalkyl, C2C6 alkenyl, C2C6 alkynyl, R8O, R9S (O) q, CN, R8C (O), R8OC (O), N (R8) 2, N3, or RSC (O) O; RI is independently selected from: a) H, b) unsubstituted or substituted C1C6 alkyl, c) unsubstituted or substituted aryl, d) unsubstituted or substituted heterocycle, e) (C1C6 alkyl) N (R8) 2, f) R8C(O)R8, g)(C1C6 alkyl) OR8, h)N (R8) 2, i)OR8, j)R8NHC (O) R8, k)R8C (O) N (R8) 2, 1) CF3, m) halo, n)C (O) OR8, o) C2C6 alkynyl, p) C2C6 alkenyl, q) perfluoroalkyl, r) N3, s) N02, t) CN, u) R9S (O) q, v) (ClC6 alkyl) NR8C (O) N (R8) 2, and w)(C1C6 alkyl) R14;(C1C6 alkyl) R14 R2 is independently selected from: a) hydrogen, b) CN, c) N02, d) halogen, e) aryl, unsubstituted or substituted, f) heteroaryl, unsubstituted or substituted, g) C1C6 alkyl, unsubstituted or substituted, h) OR8, i) N3, j) R9S (O) q, k) R8HC=CH, and l) R8C#C; R3 is independently selected from: a) H, b) CN, c) N02, d) halogen, e) C 1c6 alkyl, unsubstituted or substituted, 0 OR8, g) aryl, unsubstituted or substituted, h) heteroaryl, unsubstituted or substituted, and i) CF3; R5 and R6 are independently selected from: a) H, b) CN, c) N02, d) halogen, e) C1C6 alkyl, unsubstituted or substituted, f) N3, g)R9S(O)q, h)HC=CH2, i) HC=C, j) aryl, unsubstituted or substituted, k) heterocycle, unsubstituted or substituted, 1) CF30, m) CF3CH20, n) C3C 10 cycloalkyl, o) CF3, (C1C6 alkyl)(C1C6 alkyl) N (R8) 2, alkyl)OR8,q)(C1C6 r) OR8, s) N (R8) 2, t)C (O) (C1C6 alkyl), u) (C1C6 alkyl) C (O) R8, v)C (O) O (C1C6 alkyl, w) C(O) N (R8) 2, x) (C1C6 alkyl) NR8C (O) N (R8) 2, y)(C1C6 alkyl) NR8C (O) R8, z)C2C6 alkynyl, aa)C2C6 alkenyl, alkyl)N3,bb)(C1C6 cc)(C1C6 alkyl) NR8S (O)(C1C6 alkyl) NR8S (O) q, and dd)(C1C6(C1C6 alkyl) NR8 (C1C6 alkyl) N (R8) 2; R8 is independently selected from hydrogen, unsubstituted or substituted C1C6 alkyl, unsubstituted or substituted aralkyl, unsubstituted or substituted heterocycle and unsubstituted or substituted aryl; R9 is independently selected from unsubstituted or substituted C IC6 alkyl, unsubstituted or substituted benzyl and unsubstituted or substituted aryl; R14 is unsubstituted saturated heterocycle; R15 is independently selected from a) hydrogen, unsubstituted or substituted aryl, unsubstituted or substituted heterocycle, C3C10 cycloalkyl, C2C6 alkenyl, C2C6 alkynyl, R9S (O) 2, R8C (O), R8OC (O), (R8) 2NC (O), and b) C1C6 alkyl, unsubstituted or substituted by unsubstituted or substituted aryl, unsubstituted or substituted heterocycle, C3C 10 cycloalkyl, C2C6 alkenyl, C2C6 alkynyl, R8O, R9S (O) q, CN, R8C (O), R8OC (O), N (R8) 2, N3, or R8C (O) 0 ; A1 and A2 are independently selected from: a) a bond, b)HC=CH, c)C=C, d) 0, e) S (O) q, OC OC (O), g) C (O), h) C (O) O, and i) NR8 ; A3 is selected from a bond,C (=O), and C3C6 cycloalkyl; M is selected from CH2, NR8, O or S (O) q; Q is selected from S (O) q, CH2, C (=S), C (=NR8) or C (=O); W is a heterocycle; Y is selected from a) aryl, and b) heterocycle; Z is selected from a) aryl, and b) heterocycle; mis 0,1,2,3 or 4; n is 3 or 4; pis 0,1,2,3 or 4; qis 0,1 or 2; ris 3, or 4; sis 0,1,2,3 or 4; tis ; xis 0,1,2 or 3; provided that if attachment of to the rest of the molecule is through a nitrogen ring atom, then A1 is NR8, O, or S (O)q; or a pharmaceutically acceptable salt, an optical isomer or stereoisomer thereof.
6. The compound according to Claim 1 of formula III: wherein: Ria and Ric are independently selected from: a) hydrogen, b) unsubstituted or substituted aryl, unsubstituted or substituted heterocycle, C3C10 cycloalkyl, C2C6 alkenyl, C2C6 alkynyl, R9S (O) q, CN, N02, R8C (O), R8OC (O), (R8) 2NC (O),N (R15)2, OR15,N (R8) S (O) qR8 or N3; c) C1C6 alkyl, unsubstituted or substituted by unsubstituted or substituted aryl, unsubstituted or substituted heterocycle, C3C10 cycloalkyl, C2C6 alkenyl, C2C6 alkynyl, R8O, R9S (O) q, CN, R8C (O), R80C (O), N (R8) 2, N3, or R8C (O) O; RI is independently selected from: a) H, b) unsubstituted or substituted ClC6 alkyl, c) unsubstituted or substituted aryl, d) unsubstituted or substituted heterocycle, e) (C1C6 alkyl) N (R8) 2, f)R8C(O)R8, alkyl)OR8,g)(C1C6 h)N (R8) 2, i)OR8, j)R8NHC (O) R8, k)R8C (O) N (R8) 2, 1) CF3, m) halo, n)C (O) OR8, o) C2C6 alkynyl, alkenyl,p)C2C6 q) perfluoroalkyl, r) N3, s) N02, t) CN, u) R9S (O) q, v) (C1C6 alkyl) NR8C (O) N (R8) 2, and alkyl)R14;w)(C1C6 R2 is independently selected from: a) hydrogen, b) CN, c) N02, d) halogen, e) aryl, unsubstituted or substituted, f) heteroaryl, unsubstituted or substituted, g) C1C6 alkyl, unsubstituted or substituted, h) OR8, i) N3, j) R9S (O) q, k) R8HC=CH, and 1) R8C#C; R3 is independently selected from: a) H, b) CN, c) N02, d) halogen, e) C1C6 alkyl, unsubstituted or substituted, f) OR8, g) aryl, unsubstituted or substituted, h) heteroaryl, unsubstituted or substituted, and i) CF3; R5 and R6 are independently selected from: a) H, b) CN, c) N02, d) halogen, e) C1C6 alkyl, unsubstituted or substituted, f) N3, g) R9S (O) q, h)HC=CH2, i) HC=C, j) aryl, unsubstituted or substituted, k) heterocycle, unsubstituted or substituted, 1) CF30, m) CF3CH20, n) C3C 10 cycloalkyl, o) CF3, p) (C1C6 alkyl) N (R8) 2, alkyl)OR8,q)(C1C6 r) OR8, s) N (R8) 2, t) alkyl), u) (C1C6 alkyl) C (O) R8, v)C (O) O (C1C6 alkyl), w) C(O) N (R8) 2, x)(C1C6 alkyl) NR8C (O)(C1C6 alkyl) NR8C (O) N (R8) 2, alkyl)NR8C(O)R8,y)(C1C6 z)C2C6 alkynyl, aa)C2C6 alkenyl, alkyl)N3,bb)(C1C6 cc) (C1C6 alkyl) NR8S (O) q, and dd) (C1C6 alkyl) NR8 (C1C6 alkyl) N (R8) 2 ; R8 is independently selected from hydrogen, unsubstituted or substituted (C1C6 alkyl, unsubstituted or substituted aralkyl, unsubstituted or substituted heterocycle and unsubstituted or substituted aryl; R9 is independently selected from unsubstituted or substituted (C1C6 alkyl, unsubstituted or substituted benzyl and unsubstituted or substituted aryl; R14 is unsubstituted saturated heterocycle; R15 is independently selected from a) hydrogen, unsubstituted or substituted aryl, unsubstituted or substituted heterocycle, CsCio cycloalkyl, C2C6 alkenyl, C2C6 alkynyl, R9S (O) 2, R8C (O), R8OC (O), (R8) 2NC (O), and b) (C1C6 alkyl, unsubstituted or substituted by unsubstituted or substituted aryl, unsubstituted or substituted heterocycle, (C3C10 cycloalkyl, C2C6 alkenyl, C2C6 alkynyl, R8O, R9S (O) q, CN, R8C (O), R8OC (O), N (R8) 2, N3, or R8C (O) 0 ; Al is selected from: a) a bond, b)HC=CH, c)C=C, d) O, e) S (O) q, f) OC (O), g) C (O), h) C (O) O, and A3 is selected from a bond,C (=O), and C3C6 cycloalkyl; M is selected from CH2, NR8, O or S (O) q; Q is selected from S (O) q, CH2, C (=S), C (=NR8) or C (=O); W is a heterocycle selected from pyrrolidinyl, imidazolyl, pyridyl, triazolyl, pyridonyl, 2oxopiperidinyl, indolyl, quinolinyl, isoquinolinyl, and tetrahydroimidazopyridinyl, Y is selected from: a) aryl, and b) pyridyl; Z is selected from: a) aryl, and b) pyridyl; m is 0,1,2,3 or 4; nis 0,1,2,3 or 4; p is 0,1,2,3 or 4; q is 0, 1 or 2; sis 0,1,2,3 or 4; t is 0,1,2 or 3; x is ; provided that if attachment of to the rest of the molecule is through a nitrogen ring atom, then A1 is NR8, O, or S (O) q; or a pharmaceutically acceptable salt, an optical isomer or stereoisomer thereof.
7. The compound according to Claim 2 of formula C: wherein: Ria and Ric are independently selected from: a) hydrogen, b) unsubstituted or substituted aryl; unsubstituted or substituted heterocycle, C3C10 cycloalkyl, C2C6 alkenyl, C2C6 alkynyl, R8O, R9S (O) q, CN, NO2, R8C(O), R8OC(O), N (R8) 2, (R8) 2NC (O), C (O) N (R8), or N3; c) C IC6 alkyl, unsubstituted or substituted by unsubstituted or substituted aryl, unsubstituted or substituted heterocycle, C3Cio cycloalkyl, C2C6 alkenyl, C2C6 alkynyl, R8O, R9S (O) q, CN, R8C (O), R80C (O), N (R8) 2, N3, or R8C (O) O; RI is selected from: a) H, b) unsubstituted or substituted C IC6 alkyl, c) unsubstituted or substituted aryl, d) unsubstituted or substituted heterocycle, e) (C1C6 alkyl) N (R8) 2, (ClC6 alkyl) C (O) R8, g)(C1C6(C1C6 alkyl) OR8, h)N (R8) 2, i)OR8, j) (C1C6 alkyl) NHC (O) R8, k) (C1C6 alkyl) C (O) N (R8) 2, 1) CF3, and m) halo; R2 is selected from: a) hydrogen, b) CN, c) N02, d) halogen, e) aryl, unsubstituted or substituted, f) heteroaryl, unsubstituted or substituted, g) C1C6 alkyl, unsubstituted or substituted, h) OR8, i) N3, j) R9S (O) q, k) R8HC=CH, and 1) R8C=C; R3 is selected from: a) H, b) CN, c) N02, d) halogen, e) ClC6 alkyl, unsubstituted or substituted, f)OR8, g) aryl, unsubstituted or substituted, h) heteroaryl, unsubstituted or substituted, and i) CF3; R4 is selected from: a) H, b) =O, or c) =S; R5 is selected from: a) H, b) CN, c) N02, d) halogen, e) C 1C6 alkyl, unsubstituted or substituted, f)N3, g) R9S (O) q, h)HC=CH2, i) HC=C, j) aryl, unsubstituted or substituted, k) heterocycle, unsubstituted or substituted, 1) CF30, m) CF3CH20, n) C3C 10 cycloalkyl, o) CF3, p) (C1C6 alkyl) N (R8) 2, alkyl)OR8,q)(C1C6 r) OR8, s) N (R8) 2, t)C (O) (C1C6 alkyl), and u) (C1C6 alkyl) C (O) R8; R6 is selected from: a) H, b) C1C6 alkyl, unsubstituted or substituted, c) and d)C (O) (C1C6 alkyl); R8 is independently selected from hydrogen, unsubstituted or substituted C1C6 alkyl, benzyl and unsubstituted or substituted aryl; R9 is independently selected from unsubstituted or substituted C1C6 alkyl, benzyl and unsubstituted or substituted aryl; Axis selected from: a) a bond, b)HC=CH, c)C=C, d) O, e) S (O) q, f) OC(O), g) C (O), h) C (O) O, and W is selected from: a) heterocycle, and b) aryl; mis 0,1,2,3 or 4; n is 3 or 4; pis 3 or 4; q is 0, 1 or 2; sis and tis ; or a pharmaceutically acceptable salt, an optical isomer or stereoisomer thereof.
8. The compound according to Claim 1 of formula IV: wherein: R1C is selected from: a) hydrogen, b) unsubstituted or substituted aryl, unsubstituted or substituted heterocycle, C3C 10 cycloalkyl, C2C6 alkenyl, C2C6 alkynyl; c) C1C6 alkyl, unsubstituted or substituted by unsubstituted or substituted aryl, unsubstituted or substituted heterocycle, C3C10 cycloalkyl, C2C6 alkenyl, C2C6 alkynyl, R8O, R9S (O) q, CN, R8C (O), R8OC (O), N (R8) 2, N3, or R8C (O) 0 ; R1c" is selected from: a) hydrogen, b) N (R15) 2, and c) OR15; R1 is independently selected from: a) H, b) unsubstituted or substituted C1C6 alkyl, c) unsubstituted or substituted aryl, d) unsubstituted or substituted heterocycle, e) (C1C6 alkyl) N (R8) 2, f) R8C(O)R8, g) (ClC6 alkyl) OR8, h)N (R8) 2, i)OR8, j)R8NHC (O) R8, k)R8C (O) N (R8) 2, 1) CF3, m) halo, n)C (O) OR8, o) C2C6 alkynyl, p) C2C6 alkenyl, q) perfluoroalkyl, r) N3, s) N02, t) CN, u) R9S (O) q, v) (C1C6 alkyl) NRSC (O) N (R8) 2, and w) (CiC6 alkyl) R14; R2 is independently selected from: a) hydrogen, b) CN, c) N02, d) halogen, e) aryl, unsubstituted or substituted, f) heteroaryl, unsubstituted or substituted, g) C 1C6 alkyl, unsubstituted or substituted, h) OR8, i) N3, j) R9S (O) q, k) R8HC=CH, and 1) R8C=C ; R3 is independently selected from: a) H, b) CN, c) N02, d) halogen, e) ClC6 alkyl, unsubstituted or substituted, f)OR8, g) aryl, unsubstituted or substituted, h) heteroaryl, unsubstituted or substituted, and i) CF3; R5 is independently selected from: a) H, b) CN, c) N02, d) halogen, e) C1C6 alkyl, unsubstituted or substituted, f) N3, g) R9S (O) q, h)HC=CH2, i) HCC, j) aryl, unsubstituted or substituted, k) heterocycle, unsubstituted or substituted, 1) CF30, m) CF3CH20, n) C3C 10 cycloalkyl, o) CF3, p) (C1C6 alkyl) N (R8) 2, q) (ClC6 alkyl) OR8, r) OR8, s) N (R8) 2, t) alkyl), u) (C1C6 alkyl) C (O) R8, v)C (O) O (C1C6 alkyl), w) C(O) N (R8) 2, x) (C1C6 alkyl)NR8C(O) N (R8) 2, alkyl)NR8C(O)R8,y)(C1C6 z)C2C6 alkynyl, aa)C2C6 alkenyl, alkyl)N3,bb)(C1C6 cc) (ClC6 alkyl) NR8S (O) q, and dd) (C1C6 alkyl)NR8(C1C6 alkyl) N (R8) 2; R6 is selected from a) H, b) C1C6 alkyl, unsubstituted or substituted, c) aryl, unsubstituted or substituted, d) heterocycle, unsubstituted or substituted, e) C3Ciocycloalkyl, (C1C6 alkyl) N (R8) 2, alkyl)OR8,g)(C1C6 h) alkyl), i) (C1C6 alkyl) C (O) R8, j)C (O) O (ClC6 alkyl), k)C (O) N (R8) 2, 1) (C1C6 alkyl) NR8C (O) N (R8) 2, m) (C1C6 alkyl) NR8C (O) R8, n)C2C6 alkynyl, o)C2C6 alkenyl, p)(C1C6(C1C6 alkyl) N3, q) (C1C6 alkyl) NR8S (O) q, and r) (C1C6 alkyl) NR8 (CiC6 alkyl) N (R8) 2; R8 is independently selected from hydrogen, unsubstituted or substituted C1C6 alkyl, unsubstituted or substituted aralkyl, unsubstituted or substituted heterocycle and unsubstituted or substituted aryl; R9 is independently selected from unsubstituted or substituted C1C6 alkyl, unsubstituted or substituted benzyl and unsubstituted or substituted aryl; R14 is unsubstituted saturated heterocycle; R15 is independently selected from a) hydrogen, unsubstituted or substituted aryl, unsubstituted or substituted heterocycle, C2C6alkenyl,C2C6alkynyl,cycloalkyl, R9S (O) 2, R8C (O), R80C (O), (R8) 2NC (O), and b) C1C6 alkyl, unsubstituted or substituted by unsubstituted or substituted aryl, unsubstituted or substituted heterocycle, C3C10 cycloalkyl, C2C6 alkenyl, C2C6 alkynyl, R8O, R9S (O) q, CN, R8C (O), R80C (O), N (R8) 2, N3, or R8C (O) O; Al is selected from a) O, b) S (O) q, c) C (O), and M is selected from CH2, NR8, O or S (O) q; Q is selected from S (O) q, CH2, C (=S), C (=NR8) or C (=O); W is a heterocycle selected from imidazolyl, triazolyl or pyridyl; Y is selected from phenyl or pyridyl ; Z is selected from phenyl or pyridyl; mis 3 or 4; nis 3 or 4; q is 0,1 or 2; sis 0,1,2,3 or 4; tis ; xis 0,1,2 or 3; provided that if attachment of to the rest of the molecule is through a nitrogen ring atom, then Al is NR8, O, or S (O) q; or a pharmaceutically acceptable salt, an optical isomer or stereoisomer thereof.
9. The compound according to Claim 2 of formula D: R I a and Ric are independently selected from: a) hydrogen, b) unsubstituted or substituted aryl; unsubstituted or substituted heterocycle, CsCio cycloalkyl, C2C6 alkenyl, C2C6 alkynyl, R8O, R9S (O) q, CN, NO2, R8C(O), R8OC(O), N (R8) 2, (R8) 2NC (O), C (O) N (R8), orN3; c) C1C6 alkyl, unsubstituted or substituted by unsubstituted or substituted aryl, unsubstituted or substituted heterocycle, C3C 10 cycloalkyl, C2C6 alkenyl, C2C6 alkynyl, R80, R9S (O) q, CN, R8C (O), R80C (O), N (R8) 2, N3, or R8C (O) 0 ; R1 is selected from: a) H, b) unsubstituted or substituted ClC6 alkyl, c) unsubstituted or substituted aryl, d) unsubstituted or substituted heterocycle, e) (C1C6 alkyl) N (R8) 2, (C1C6 alkyl) C (O) R8, alkyl)OR8,g)(C1C6 h)N (R8) 2, i)OR8, j) (C1C6 alkyl) NHC (O) R8, k) (C1C6 alkyl) C (O) N (R8) 2, I) CF3, and m) halo; R2 is selected from: a) hydrogen, b) CN, c) N02, d) halogen, e) aryl, unsubstituted or substituted, f) heteroaryl, unsubstituted or substituted, g) C 1C6 alkyl, unsubstituted or substituted, h) OR8, i) N3, j) R9S (O) q, k) R8HC=CH, and 1) R8C C; R3 is selected from: a) H, b) CN, c) N02, d) halogen, e) ClC6 alkyl, unsubstituted or substituted, f) OR8, g) aryl, unsubstituted or substituted, h) heteroaryl, unsubstituted or substituted, and i) CF3; R4 is selected from: a) H, or b) =O; R5 is selected from: a) H, b) CN, c) N02, d) halogen, e) C1C6 alkyl, unsubstituted or substituted, f)N3, <BR> <BR> <BR> g) R9S(O)q,<BR> <BR> <BR> <BR> <BR> <BR> <BR> <BR> h) HC=CH2,<BR> <BR> <BR> <BR> <BR> <BR> <BR> i)HC#C, j) aryl, unsubstituted or substituted, k) heterocycle, unsubstituted or substituted, 1) CF30, m) CF3CH20, n) C3Ciocycloalkyl, o) CF3, p)(C1C6 alkyl)(C1C6 alkyl) N (R8) 2, alkyl)OR8,q)(C1C6 r) OR8, s) N (R8) 2, t)C (O) (C 1C6 alkyl), and u)(C1C6 alkyl)(C1C6 alkyl) C (O) R8; R6 is selected from: a) H, b) ClC6 alkyl, unsubstituted or substituted, and c)C (O) (ClC6 alkyl); R8 is independently selected from hydrogen, unsubstituted or substituted ClC6 alkyl, benzyl and unsubstituted or substituted aryl; R9 is independently selected from unsubstituted or substituted C1C6 alkyl, benzyl and unsubstituted or substituted aryl; W is a heterocycle, selected from imidazolyl or pyridyl; mis 0,1,2,3 or 4; n is 0,1,2,3 or 4; pis 3 or 4; q is 0,1 or 2; s is 3 or 4; and tis ; or a pharmaceutically acceptable salt, an optical isomer or stereoisomer thereof.
10. A compound which is selected from the group consisting of : 4imidazol1ylmethyl2 2 (2oxopiperidin1yl)phenoxybenzonitrile 4imidazol1ylmethyl2 3 (2oxopiperidin1yl)phenoxybenzonitrile 4imidazol1ylmethyl2 4 (2oxopiperidin1yl)phenoxybenzonitrile 4imidazol1ylmethyl2[2(3methyl2oxopiperidin1yl)phenoxy]benzonitrile 4Imidazol1ylmethyl22 (2oxopyrrolidin1yl)phenoxybenzonitrile 4imidazolIylmethyl2 (3morpholin4ylphenoxy)benzonitn'le 4imidazol1ylmethyl2 (3piperidin1ylmethylphenoxy)benzonitrile 4imidazol1ylmethyl2[3(2oxoazepan1yl)phenoxy]benzonitrile 2 2 (3, 3dimethyl2oxopiperidin1yl)phenoxy4imidazol1ylmethyl benzonitrile 2 3 (3ethyl1methyl2oxoazepan3yl)phenoxy4imidazol1ylmethyl benzonitrile 2 3 (3 (S)ethyl1methyl2oxoazepan3yl)phenoxy4imidazol1ylmethyl benzonitrile 23 (3 (R)ethyl1methyl2oxoazepan3yl)phenoxy4imidazol1ylmethyl benzonitrile 2 3 (3ethyl1methyl2oxoazepan3yl)phenoxy4 (2methylimidazol1 yl) methylbenzonitrile 23 (3ethyl1methyl2oxoazepan3yl)phenoxy4 (5methylimidazol1 yl)methylbenzonitrile 2 3 (3ethyl1methyl2oxoazepan3yl)phenoxy4 (2, 5dimethylimidazol1 yl) methylbenzonitrile 2[3(3ethyl1methyl2oxoazepan3yl)phenoxy]4[1, 2,4] triazol4ylmethyl benzonitrile 2[3(3ethyl1methyl2oxoazepan3yl)phenoxy]4[1, 2,4] triazol1ylmethyl benzonitrile 4imidazol1ylmethyl2 3 (lmethyl2oxoazepan3yl)phenoxybenzonitrile 4imidazol1ylmethyl2 3 (lmethyl2oxoazocan3yl)phenoxybenzonitrile 4imidazol1ylmethyl2 3 (lmethyl2oxopiperidin3yl)phenoxybenzonitrile 4imidazol1ylmethyl2 3 (1methyl2oxopyrrolidin3yl)phenoxybenzonitrile 4imidazol1ylmethyl2 3 (3ethyl1methyl2oxopiperidin3yl)phenoxy benzonitrile 4imidazol1ylmethyl2 3 (2oxoazepan3yl)phenoxybenzonitrile 2 3 (3hydroxymethyl1methyl2oxoazepan3yl)phenoxy4imidazol1 ylmethylbenzonitrile 2 3 (3cyclopropylmethyl1methyl2oxoazepan3yl)phenoxy4imidazolI ylmethylbenzonitrile 2 4bromo3 (3cyclopropylmethyl1methyl2oxoazepan3yl)phenoxy4 imidazol1ylmethylbenzonitrile 2 3 (3methoxymethyl1methyl2oxoazepan3yl)phenoxy4imidazol1 ylmethylbenzonitrile 2 3 (3ethyl2oxoazepan3yl)phenoxy4imidazol1ylmethylbenzonitrile 2 3 (3ethylazepan3yl)phenoxy4imidazol1ylmethylbenzonitrile 23 (lacetyl3ethylazepan3yl)phenoxy4imidazol1ylmethylbenzonitrile 3 3 (2cyano5imidazollylmethylphenoxy)phenyl3ethylazepane1carboxylic acidtertbutyl ester 4 5 (2aminoethyl)2methylimidazol1ylmethyl2 3 (3ethyl1methyl2oxo azepan3yl)phenoxy]benzonitrile 2 3 (3ethyl1methyl2oxoazepan3yl)phenoxy4 2methyl5 (2morpholin4 ylethyl)imidazol1ylmethylbenzonitrile N[2(3{4cyano3[3(3ethyl1methyl2oxoazepan3yl)phenoxy]benzyl}2 methyl3Himidazol4yl)ethylacetamide 3ethyl3 3 (3imidazol1ylmethylphenoxy)phenyl1methylazepan2one 23 (3ethyl1methyl2oxoazepan3yl)phenoxy4 (3methyl3Himidazol4 ylmethyl)benzonitrile 2 3 (3ethyl1methyl2oxoazepan3yl)phenoxy4 (3Himidazol4ylmethyl) benzonitrile 2 3 (3ethyl1methyl2oxoazepan3yl)phenoxy4 hydroxy (3methyl3H imidazol4yl)methylbenzonitrile 4 amino (3methyl3Himidazol4yl)methyl2 3 (3ethyl1methyl2oxo azepan3yl)phenoxy]benzonitrile 2 3 (3ethyl1methyl2oxoazepan3yl)benzyl4 (3methyl3Himidazole4 carbonyl)benzonitrile 2 3 (3ethyl1methyl2oxoazepan3yl)phenoxy4 (hydroxypyridin3yl methyl)benzonitrile 2 3 (3ethyl1methyl2oxoazepan3yl)phenoxy4pyridin3ylmethyl benzonitrile 2 3 (3ethyl1methyl2oxoazepan3yl)phenoxy4pyridin2ylmethyl benzonitrile 2[3(3ethyl1methyl2oxoazepan3yl)phenoxy]4[1hydroxy1(3methyl3H imidazol4yl)ethyl]benzonitrile 2 3 (3R)ethylImethyl2oxoazepan3yl)phenoxy4 I (R) aminoI (3methyl 3Himidazol4yl)ethylbenzonitrile 2 3 (3S)ethyl Imethyl2oxoazepan3yl)phenoxy4 I (S) amino I (3methyl 3Himidazol4yl)ethylbenzonitrile 2 3 (3S)ethylImethyl2oxoazepan3yl)phenoxy4 I (R) aminoI (3methyl 3Himidazol4yl)ethylbenzonitrile 2[3(3R)ethyl1methyl2oxoazepan3yl)phenoxy]4[1(S)amino1(3methyl 3Himidazol4yl)ethylbenzonitrile 2 33 (R)nbutyl1methyl2oxoazepan3yl)phenoxy4 1 (R) amino1 (3 methyl3Himidazol4yl)ethylbenzonitrile 2 33 (S)nbutyl1methyl2oxoazepan3yl)phenoxy4 1 (S) amino1 (3methyl 3Himidazol4yl)ethylbenzonitrile 2 33 (S)nbutyl1methyl2oxoazepan3yl)phenoxy4 1 (R) amino1 (3methyl 3Himidazol4yl)ethylbenzonitrile 2 33 (R)nbutyl1methyl2oxoazepan3yl)phenoxy4 1 (S) amino1 (3methyl 3Himidazol4yl)ethylbenzonitrile 2[33(R)cyclopropylmethyl1methyl2oxoazepan3yl)phenoxy]4[1(R)amino 1 (3methyl3Himidazol4yl)ethylbenzonitrile 2 33 (S)cyclopropylmethyl1methyl2oxoazepan3yl)phenoxy4 1 (S) amino 1 (3methyl3Himidazol4yl)ethylbenzonitrile 2 33 (R)cyclopropylmethyl1methyl2oxoazepan3yl)phenoxy4 l (S) amino 1 (3methyl3Himidazol4yl)ethylbenzonitrile 2 33 (S)cyclopropylmethyl1methyl2oxoazepan3yl)phenoxy4 1 (R) amino 1 (3methyl3Himidazol4yl)ethylbenzonitrile 2 33 (R)cyclopropylethyl1methyl2oxoazepan3yl)phenoxy4 1 (R) amino 1 (3methyl3 Himidazol4yl)ethylbenzonitrile 2[33(S)cyclopropylethyl1methyl2oxoazepan3yl)phenoxy]4[1(S)amino1 (3methyl3Himidazol4yl)ethylbenzonitrile 2 33 (R)cyclopropylethyl1methyl2oxoazepan3yl)phenoxy4 1 ( S)amino 1 (3methyl3Himidazol4yl)ethylbenzonitrile 2 33 (S)cyclopropylethyl1methyl2oxoazepan3yl)phenoxy4 1 (R) amino 1 (3methyl3Himidazol4yl)ethylbenzonitrile 2 33 (R)propyl1methyl2oxoazepan3yl)phenoxy4 1 (R) amino1 (3 methyl3Himidazol4yl)ethyl]benzonitrile 2 33 (S) propyl1methyl2oxoazepan3yl)phenoxy4 1 ( S) amino1 (3 methyl3Himidazol4yl)ethylbenzonitrile 2 33 (R)propyl1methyl2oxoazepan3yl)phenoxy4 1 ( S) amino1 (3 methyl3Himidazol4yl)ethyl]benzonitrile 2 33 (S)propyl1methyl2oxoazepan3yl)phenoxy4 1 (R)amino1 (3 methyl3Himidazol4yl)ethylbenzonitrile 2 33 (R)methoxymethyl1methyl2oxoazepan3yl)phenoxy4 1 (R) amino1 (3methyl3Himidazol4yl)ethylbenzonitrile 2 33 (S)methoxymethyl1methyl2oxoazepan3yl)phenoxy4 1 ( S)amino 1 (3methyl3Himidazol4yl)ethylbenzonitrile 2 33 (R)methoxymethyl1methyl2oxoazepan3yl)phenoxy4 1 ( S)amino1 (3methyl3Himidazol4yl)ethylbenzonitrile 2 33 (S)methoxymethyl1methyl2oxoazepan3yl)phenoxy4 1 (R) amino1 (3methyl3Himidazol4yl)ethylbenzonitrile 2 33 (R)ethoxymethyl1methyl2oxoazepan3yl)phenoxy4 1 (R) amino1 (3methyl3Himidazol4yl)ethylbenzonitrile 2 33 (S)ethoxymethyl1methyl2oxoazepan3yl)phenoxy4 1 (S) amino1 (3methyl3Himidazol4yl)ethylbenzonitrile 2 33 (R)ethoxymethyl1methyl2oxoazepan3yl)phenoxy4 1 ( S)amino1 (3methyl3Himidazol4yl)ethylbenzonitrile 2 33 (S)ethoxymethyl1methyl2oxoazepan3yl)phenoxy4 1 (R) amino1 (3methyl3Himidazol4yl)ethylbenzonitrile 2 33 (R)cyclopropylmethyl1methyl2oxopiperidin3yl)phenoxy4 1 (R)amino1 (3methyl3Himidazol4yl)ethylbenzonitrile 2 33 (S)cyclopropylmethyllmethyl2oxopiperidin3yl)phenoxy4 I (S)amino1 (3methyl3Himidazol4yl)ethylbenzonitrile 2[33(R)cyclopropylmethyl1methyl2oxopiperidin3yl)phenoxy]4[1(S) aminoI (3methyl3Himidazol4yl)ethylbenzonitrile 2 33 (Scyclopropylmethyl1methyl2oxopiperidin3yl)phenoxy4 l (R) amino 1 (3methyl3Himidazol4yl)ethylbenzonitrile 2 [33 (R)benzyl1methyl2oxopiperidin3yl)phenoxy4 1 (R) amino1 (3 methyl3Himidazol4yl)ethylbenzonitrile 2 33 (S)benzyl1methyl2oxopiperidin3yl)phenoxy4 1 ( S) amino1 (3 methyl3Himidazol4yl)ethylbenzonitrile 2[33(R)benzyl1methyl2oxopiperidin3yl)phenoxy]4[1(S)amino1(3 methyl3Himidazol4yl)ethylbenzonitrile 2 33 (S)benzyl1methyl2oxopiperidin3yl)phenoxy4 1 (R) amino1 (3 methyl3Himidazol4yl)ethylbenzonitrile 2[3methyl1methyl2oxoazepan3yl)phenoxy]4[1amino1(3methyl3H imidazol4yl)ethylbenzonitrile 2 3 (3ethylImethyl2oxoazep an3yl)phenoxy4 1 (3methyl3Himi dazol4 yl)vinylbenzonitrile 2 3 (3ethyl1methyl2oxoazepan3yl)phenoxy4 1 (3methyl3Himidazol4 yl)cyclopropylbenzonitrile 2 3N, Ndimethylaminomethyllmethyl2oxoazepan3yl)phenoxy4 I hydroxy1 (3methyl3Himidazol4yl)ethylbenzonitrile 2[3N,Ndimethylaminomethyl1methyl2oxoazepan3yl)phenoxy]4[1amino 1 (3methyl3Himidazol4yl)ethylbenzonitrile 2 3 (3N, Ndimethylaminomethyllmethyl2oxoazepan3yl)phenoxy4 1 (3 methyl3Himidazol4yl)vinylbenzonitrile 4 5 (2N, Ndimethylaminoethyl)2methylimidazol1ylmethyl2 3 (3Sethyl1 methyl2oxoazepan3yl)phenoxybenzonitrile 4 5 (2N, Ndimethylaminoethyl)2methylimidazol1ylmethyl2 3 (3Rethyl1 methyl2oxoazepan3yl)phenoxybenzonitrile 2 (3 {4cyano3 3 (3Sethyl1methyl2oxoazepan3yl)phenoxybenzyl}2 methyl3Himidazol4yl)ethylurea 2 (3 {4cyano3 3 (3Rethyllmethyl2oxoazepan3yl)phenoxybenzyl}2 methyl3Himidazol4yl)ethylurea 2 3 (3ethyl1methyl2oxoazepan3yl)phenoxy4pyridinNoxide3ylmethyl benzonitrile 2 31methyl2oxopyrrolidin3yl)phenoxy4 lamino1 (3methyl3Himidazol 4yl)ethylbenzonitrile 2 3 ( 1methyl2oxoazepan3yl)phenoxy4 1amino1 (3methyl3Himidazol4 yl)ethylbenzonitrile 2 3 (3hydroxymethyl1methyl2oxoazepan3yl)phenoxy4 1amino1 (3 methyl3Himidazol4yl)ethylbenzonitrile 2 3 (3cyclopropylmethyl1methyl2oxopyrrolidin3yl)phenoxy4 laminol (3methyl3Himidazol4yl)ethylbenzonitrile 2 33 (R)cyclopropylmethyl1methyl2oxopyrrolidin3yl)phenoxy4 1 (R) amino1 (3methyl3Himidazol4yl)ethylbenzonitrile 2 33 (S)cyclopropylmethyl1methyl2oxopyrrolidin3yl)phenoxy4 1 (S) amino1 (3methyl3Himidazol4yl)ethylbenzonitn'le 2 33 (R)cyclopropylmethyl1methyl2oxopyrrolidin3yl)phenoxy4 1 ( S) amino1 (3methyl3Himidazol4yl)ethylbenzonitrile 2 33 (S)cyclopropylmethyl1methyl2oxopyrrolidin3yl)phenoxy4 1 (R) amino1 (3methyl3Himidazol4yl)ethylbenzonitrile 2 33 (R)benzyllmethyl2oxopyrrolidin3yl)phenoxy4 I (R) aminol (3 methyl3Himidazol4yl)ethylbenzonitrile 2 33 (S)benzyllmethyl2oxopyrrolidin3yl)phenoxy4 I ( S)aminol (3 methyl3Himidazol4yl)ethylbenzonitrile 2 33 (R)benzyl1methyl2oxopyrrolidin3yl)phenoxy4 1 ( S)amino1 (3 methyl3Himidazol4yl)ethylbenzonitrile 2 [33 (S)benzyl1methyl2oxopyrrolidin3yl)phenoxy4 [1(R amino1(3 methyl3Himidazol4yl)ethylbenzonitrile 4 1 (4allyl5bromopyridin3yl)1hydroxyethyl2 3 (3ethyl1methyl2oxo azepan3yl)phenoxybenzonitrile 1tertbutyl3 (R) 1 {4cyano3 3 (3Rethyl1methyl2oxoazepan3yl)phenoxy phenyl}1(3methyl3Himidazol4yl)ethyl]urea 1tertbutyl3(S)[1{4cyano3[3(3Sethyl1methyl2oxoazepan3yl)phenoxy] phenyl}1 (3methyl3Himidazol4yl)ethylurea 1tertbutyl3 (R) 1 {4cyano3 3 (3 Sethyl1methyl2oxoazepan3yl)phenoxy phenyl}1(3methyl3Himidazol4yl)ethylurea 1tertbutyl3(S) l {4cyano3 3 (3Rethyllmethyl2oxoazepan3yl)phenoxy phenyl}1 (3methyl3Himidazol4yl)ethylurea l l {4cyano3 (R) 3 (3Rethyllmethyl2oxoazepan3yl)phenoxyphenyl}l (3methyl3Himidazol4yl)ethyl3methylurea 1[1{4cyano3(S)[3(3Sethyl1methyl2oxoazepan3yl)phenoxy]phenyl}1 (3methyl3Himidazol4yl)ethyl3methylurea 1 1 {4cyano3 (R) 3 (3 Sethyl1methyl2oxoazepan3yl)phenoxyphenyl}1 (3methyl3Himidazol4yl)ethyl3methylurea 1 1 {4cyano3 (S) 3 (3Rethyl1methyl2oxoazepan3yl)phenoxyphenyl}1 (3methyl3Himidazol4yl)ethyl3methylurea 1 1 {4cyano3 (R) 3 (3Sethyl1methyl2oxoazepan3yl)phenoxyphenyl}1 (3methyl3Himidazol4yl)ethyl3phenylurea l l {4cyano3 (S) 3 (3Sethyllmethyl2oxoazepan3yl)phenoxypheny !}l (3methyl3Himidazol4yl)ethyl3phenylurea 1 1 {4cyano3 (R) 3 (3Rethyl1methyl2oxoazepan3yl)phenoxyphenyl}1 (3methyl3Himidazol4yl)ethyl3phenylurea 1 I 14cyano3 (R) 3 (3Sethyl 1methyl2oxoazepan3yl)phenoxyphenyllI (3methyl3Himidazol4yl)ethyl3phenylurea 1 {4cyano3 (R) 3 (3Rethyl1methyl2oxoazepan3yl)phenoxyphenyl}1 (3 methyl3Himidazol4yl)ethylurea [1{4cyano3(S)[3(3Sethyl1methyl2oxoazepan3yl)phenoxy]phenyl}1(3 methyl3Himidazol4yl)ethylurea 1 {4cyano3 (R) 3 (3Sethy !lmethyl2oxoazepan3yl)phenoxyphenyl}l (3 methyl3Himidazol4yl)ethylurea [1{4cyano3(S)[3(3Rethyl1methyl2oxoazepan3yl)phenoxy]phenyl}1(3 methyl3Himidazol4yl)ethyl]urea N[1{4cyano3[3(3ethyl1methyl2oxoazepan3yl)phenoxyl]phenyl}1(3 methyl3Himidazol4yl)ethylacetamide 2 3 (3 Sethyllmethyl2oxoazepan3yl)phenoxy4 1methylamino1 (3 methyl3Himidazol4yl)ethylbenzonitrile 2 3 (3Rethyl1methyl2oxoazepan3yl)phenoxy4 1methylamino1 (3 methyl3Himidazol4yl)ethylbenzonitrile 2 3 (3 Sethyl1methyl2oxoazepan3yl)phenoxy4 l (R)methylamino1 (3 methyl3Himidazol4yl)ethylbenzonitrile 2 3 (3 Sethyl1methyl2oxoazepan3yl)phenoxy4 1 (S)methylamino1 (3 methyl3Himidazol4yl)ethylbenzonitrile 2 5 (3 Sethyl1methyl2oxoazepan3yl)2iodophenoxy4imidazol1ylmethyl benzonitrile 2 5 (3Rethyl1methyl2oxoazepan3yl)2iodophenoxy4imidazol1ylmethyl benzonitrile 2 5 (3Sethyl1methyl2oxoazepan3yl)2bromophenoxy4imidazol1 ylmethylbenzonitrile 2 5 (3Rethyl1methyl2oxoazepan3yl)2bromophenoxy4imidazol1 ylmethylbenzonitrile 41amino1 (3methyl3Himidazol4yl)ethyl2 2dimethyl aminomethyl5 (3S ethyl1methyl2oxoazepan3yl)phenoxybenzonitrile 2[3(3ethyl1methyl2oxoazepan3yl)phenoxy]4(4, 5,6,7tetrahydro3H imidazo [4,5] pyridin4yl)benzonitrile 2 3ethyl2oxoazepan3yl)phenoxy4 1 amino1 (3methyl3Himidazol4yl) ethylbenzonitrile 4 laminoI (3methyl3 Himidazol4yl)ethyl213 3ethylI (3hydroxy propyl)2oxoazepan3ylphenoxy}benzonitrile 4[1amino1(3methyl3Himidazol4yl)ethyl]2{3[3ethyl1propyl2oxo azepan3ylphenoxy}benzonitrile 2 {3 1 (2aminoethyl)3ethyl2oxoazepan3ylphenoxy}4 1amino1 (3 methyl3Himidazol4yl)ethylbenzonitrile {2 3 (3 {5 lamino1 (3methyl3Himidazol4yl)ethyl 2cyanophenoxy} phenyl)3ethyl2oxoazepan1ylethyl}urea N {2 3 (3 {5 lamino1 (3methyl3Himidazol4yl)ethyl 2cyanophenoxy} phenyl)3ethyl2oxoazepan1ylethyl}N'methyl urea 4[1(R)amino1(3methyl3Himidazol4yl)ethyl]2{3[1methyl2oxo3(R) (3, 3, 3trifluoropropyl)azepan3ylphenoxy}benzonitrile 4[1(S)amino1(3methyl3Himidazol4yl)ethyl]2{3[1methyl2oxo3(S) (3,3,3trifluoropropyl)azepan3yl]phenoxy}benzonitrile 4 1 (R)amino1(3methyl3Himidazol4yl)ethyl]2{3[1methyl2oxo3 (S) (3,3,3trifluoropropyl)azepan3yl]phenoxy}benzonitrile 4[1(S)amino1(3methyl3Himidazol4yl)ethyl]2{3[1methyl2oxo3(R) (3,3,3trifluoropropyl)azepan3yl]phenoxy}benzonitrile 4 1amino1 (3methyl3Himidazol4yl)ethyl2 {3 1 (2dimethylaminoethyl)3 ethyl2oxoazepan3ylphenoxy}benzonitrile 4 1 (R or S) amino1 (3methyl3Himidazol4yl)ethyl2 {3 1 (2hydroxyethyl) 3ethyl2oxoazepan3ylphenoxy}benzonitrile 4laminol (3methyl3HimidazoI4yl)ethyl2 {3 l (2methoxyethyl)3Rethyl 2oxoazepan3ylphenoxy}benzonitrile 4Imidazol1ylmethyl2 3 (lmethyl7oxoazepan2yl)phenoxybenzonitrile hydrochloride 4 lAmino1 (3methyl3Himidazol4yl)ethyl2 3 (4methyl3oxo2 propylmorpholin2yl)phenoxybenzonitrile 4 1 amino1 (3methyl3Himidazol4yl)ethyl2 3 (morpholine4sulfonyl) phenoxybenzonitrile 4 lamino1 (3methyl3Himidazol4yl)ethyl2 {3 1 (2methoxyethyl)3Rethyl 2oxoazepan3ylphenoxy}benzonitrile; 2 3 (3Rethyllmethyl2oxoazepan3yl)phenoxy4 5 (2morpholin4ylethyl) imidazol1ylmethyl benzonitrile; or a pharmaceutically acceptable salt, an optical isomer or stereoisomer thereof.
11. The compound according to Claim 10 which is 2 {3 1methyl2oxo3 (R or S) (3,3,3trifluoropropyl)azepan3ylphenoxy}4 1 (R or S)amino1 (3methyl3Himidazol4yl)ethylbenzonitrile; or a pharmaceutically acceptable salt, an optical isomer or stereoisomer thereof.
12. The compound according to Claim 10 which is 4 1amino1 (3methyl3Himidazol4yl)ethyl2 {3 1 (2methoxyethyl)3Rethyl 2oxoazepan3ylphenoxy}benzonitrile; or a pharmaceutically acceptable salt, an optical isomer or stereoisomer thereof.
13. The compound according to Claim 10 which is: 2 33 (R or S)benzyl1methyl2oxopyrrolidin3yl)phenoxy4 1 (R or S) amino 1 (3methyl3Himidazol4yl)ethylbenzonitn'le; or a pharmaceutically acceptable salt, an optical isomer or stereoisomer thereof.
14. The compound according to Claim 10 which is: 2[3(3cycloproylmethyl1methyl2oxopiperidin3yl)phenoxy]4[1amino1(3 methyl3Himidazol4yl)ethylbenzonitrile or a pharmaceutically acceptable salt, an optical isomer or stereoisomer thereof.
15. The compound according to Claim 10 which is: 2 3 (3ethyl1methyl2oxoazepan3yl)phenoxy4 lamino1 (3methyl3H imidazol4yl)ethylbenzonitrile or a pharmaceutically acceptable salt, an optical isomer or stereoisomer thereof.
16. The compound according to Claim 10 which is: 2 3 (3Rethyl1methyl2oxoazepan3yl)phenoxy4 5 (2morpholin4ylethyl) imidazol1ylmethyl benzonitrile; or a pharmaceutically acceptable salt, an optical isomer or stereoisomer thereof.
17. The compound according to Claim 10 which is: 2 3 (3ethyl1methyl2oxoazepan3yl)phenoxy4 2methyl5 (2morpholin4 ylethyl)imidazol1ylmethylbenzonitrile; or a pharmaceutically acceptable salt, an optical isomer or stereoisomer thereof.
18. The compound according to Claim 10 which is: 2 3 (lacetyl3ethylazepan3yl)phenoxy4imidazol1ylmethylbenzonitrile; or a pharmaceutically acceptable salt, an optical isomer or stereoisomer thereof.
19. The compound according to Claim 10 which is: 2 3 (3ethyl1methyl2oxoazepan3yl)phenoxy4 1 (3methyl3Himidazol4 yl)cyclopropylbenzonitrile or a pharmaceutically acceptable salt, an optical isomer or stereoisomer thereof.
20. The compound according to Claim 10 which is: 2 (3 {4cyano3 3 (3ethyl1methyl2oxoazepan3yl)phenoxybenzyl}2 methyl3Himidazol4yl)ethylurea or a pharmaceutically acceptable salt, an optical isomer or stereoisomer thereof.
21. The compound according to Claim 10 which is: 2 3 (3ethyl1methyl2oxoazepan3yl)phenoxy4 lmethylamino1 (3methyl 3Himidazol4yl)ethylbenzonitrile or a pharmaceutically acceptable salt, an optical isomer or stereoisomer thereof.
22. The compound according to Claim 10 which is: {2 3 (3 {5 lamino1 (3methyl3Himidazol4yl)ethyl 2cyanophenoxy} phenyl)3ethyl2oxoazepan1ylethyl}urea or a pharmaceutically acceptable salt, an optical isomer or stereoisomer thereof.
23. A pharmaceutical composition comprising a pharmaceutical carrier, and dispersed therein, a therapeutically effective amount of a compound of Claim 1.
24. A pharmaceutical composition comprising a pharmaceutical carrier, and dispersed therein, a therapeutically effective amount of a compound of Claim 2.
25. A pharmaceutical composition comprising a pharmaceutical carrier, and dispersed therein, a therapeutically effective amount of a compound of Claim 4.
26. A pharmaceutical composition comprising a pharmaceutical carrier, and dispersed therein, a therapeutically effective amount of a compound of Claim 10.
27. A method for inhibiting farnesylprotein transferase which comprises administering to a mammal in need thereof a therapeutically effective amount of a composition of Claim 23.
28. A method for inhibiting farnesylprotein transferase which comprises administering to a mammal in need thereof a therapeutically effective amount of a composition of Claim 24.
29. A method for inhibiting farnesylprotein transferase which comprises administering to a mammal in need thereof a therapeutically effective amount of a composition of Claim 25.
30. A method for inhibiting farnesylprotein transferase which comprises administering to a mammal in need thereof a therapeutically effective amount of a composition of Claim 26.
31. A method for treating cancer which comprises administering to a mammal in need thereof a therapeutically effective amount of a composition of Claim 23.
32. A method for treating cancer which comprises administering to a mammal in need thereof a therapeutically effective amount of a composition of Claim 24.
33. A method for treating cancer which comprises administering to a mammal in need thereof a therapeutically effective amount of a composition of Claim 25.
34. A method for treating cancer which comprises administering to a mammal in need thereof a therapeutically effective amount of a composition of Claim 26.
35. A method for treating neurofibromen benign proliferative disorder which comprises administering to a mammal in need thereof a therapeutically effective amount of a composition of Claim 23.
36. 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 23.
37. 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 23.
38. A method for preventing restenosis which comprises administering to a mammal in need thereof a therapeutically effective amount of a composition of Claim 23.
39. 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 23.
40. A pharmaceutical composition made by combining the compound of Claim 1 and a pharmaceutically acceptable carrier.
41. A process for making a pharmaceutical composition comprising combining a compound of Claim 1 and a pharmaceutically acceptable carrier.
42. A pharmaceutical composition made by combining the compound of Claim 2 and a pharmaceutically acceptable carrier.
43. A process for making a pharmaceutical composition comprising combining a compound of Claim 2 and a pharmaceutically acceptable carrier.
44. A method of conferring radiation sensitivity on a tumor cell using a therapeutically effective amount of a composition of Claim 23 in combination with radiation therapy.
45. A method of using a therapeutically effective amount of a composition of Claim 23 in combination with an antineoplastic.
46. A method according to Claim 45 wherein the antineoplastic is paclitaxel.
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. Lowy and D. M. Willumsen, Ann. Rev. Biochem. 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 farnesyl-protein transferase or geranylgeranyl-protein transferase, which catalyze the alkylation of the cysteine residue of the CAAX motif with a C 15 or C20 isoprenoid, respectively.

Such enzymes may be generally termed prenyl-protein transferases. (S. Clarke., Ann.

Rev. Biochem. 61: 355-386 (1992); W. R. Schafer and J. Rine, Ann. Rev. Genetics 30: 209-237 (1992)). 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 polyiso- prenoids 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. Chem., 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 compounds that will inhibit prenyl-protein transferase and thus, the post-translational isoprenylation 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 non-prodrug compounds which inhibit prenyl-protein transferase. Further contained in this invention are chemo- therapeutic compositions containing these prenyl-protein transferase inhibitors and methods for their production.

The compounds of this invention are illustrated by the formulae I and A:

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

wherein: RiaRib and Ric are independently selected from: a) hydrogen, b) unsubstituted or substituted aryl, unsubstituted or substituted heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, R9S (O) q-, CN, N02, R8C (O)-, R8OC (O)-, (R8) 2NC (O)-,-N (R15) 2, -OR15, -N(R8) S (O) qR8 or N3; c) C1-C6 alkyl, unsubstituted or substituted by unsubstituted or substituted aryl, unsubstituted or substituted heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, R8O-, R9S (O) q-, CN, R8C (O)-, R80C (O)-, N (R8) 2, N3, or R8C (O) O-; R1 is independently selected from: a) H, b) unsubstituted or substituted Cl-C6 alkyl, c) unsubstituted or substituted aryl, d) unsubstituted or substituted heterocycle, e)-(C1-C6 alkyl)(C1-C6 alkyl) N (R8) 2, f)-R8C (O) R8 g)- (C1-C6 alkyl) OR8, h)-N (R8) 2, i)-OR8,

j)-R8NHC (O) R8, k)-RSC (O) N (R8) 2, 1) CF3, m) halo, n)-C (O) OR8, o) C2-C6 alkynyl, p) C2-C6 alkenyl, q) perfluoroalkyl, r) N3, s) N02, t) CN, u) R9S (O) q-, v)-(Cl-C6 alkyl) NR8C (O)(Cl-C6 alkyl) NR8C (O) N (R8) 2, and alkyl)R14;w)-(C1-C6 R2 is independently selected from: a) hydrogen, b) CN, c) N02, d) halogen, e) aryl, unsubstituted or substituted, f) heteroaryl, unsubstituted or substituted, g) Cl-C6 alkyl, unsubstituted or substituted, h) OR8, i) N3, j) R9S(O)q, k) R8HC=CH-, and l) R8C#C-; R3 is independently selected from: a) H, b) CN, c) N02, d) halogen,

e) C 1-C6 alkyl, unsubstituted or substituted, f) OR8, g) aryl, unsubstituted or substituted, h) heteroaryl, unsubstituted or substituted, and i) CF3; R5 and R6 are independently selected from: a) H, b) CN, c) N02, d) halogen, e) C1-C6 alkyl, unsubstituted or substituted, f)N3, g) R9S (O) q, h)-HC=CH2, i) HC=C-, j) aryl, unsubstituted or substituted, k) heterocycle, unsubstituted or substituted, 1) CF30-, m) CF3CH20-, n) C3-C 10 cycloalkyl, o) CF3, alkyl)N(R8)2,p)-(C1-C6 q)- (Cl-C6 alkyl) OR8, r) OR8, s) N (R8) 2, t) alkyl), u) -(C1-C6 alkyl) C (O) R8, v)-C (O) O (C1-C6 alkyl), w)-C (O) N (R8) 2, x) -(C1-C6 alkyl) NR8C (O) N (R8) 2, alkyl)NR8C(O)R8,y)-(C1-C6 z)-C2-C6 alkynyl, aa)-C2-C6 alkenyl,

alkyl)N3,bb)-(C1-C6 cc) -(C1-C6 alkyl) NR8S (O) q, and dd) -(C1-C6 alkyl) NR8 (C1-C6 alkyl) N (R8) 2; R8 is independently selected from hydrogen, unsubstituted or substituted C 1-C6 alkyl, unsubstituted or substituted aralkyl, unsubstituted or substituted heterocycle and unsubstituted or substituted aryl; R9 is independently selected from unsubstituted or substituted C I-C6 alkyl, unsubstituted or substituted benzyl and unsubstituted or substituted aryl; R14 is unsubstituted saturated heterocycle; R15 is independently selected from a) hydrogen, unsubstituted or substituted aryl, unsubstituted or substituted heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, R9S (O) 2-, R8C (O)-, R8OC (O)-, (R8) 2NC (O)-, and b) Cl-C6 alkyl, unsubstituted or substituted by unsubstituted or substituted aryl, unsubstituted or substituted heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, R8O-, R9S (O) q-, CN, R8C (O)-, R80C (O)-, N (R8) 2, N3, or R8C (O) O-; A1 and A2 are independently selected from: a) a bond, b)-HC=CH-, c)-C=C-, d) O, e) S (O) q, OC OC (O), g) C (O), h) C (O) O, and

A3 is selected from a bond,-C (=O)-, and C3-C6 cycloalkyl; M is selected from CH2, NR8, O or S (O) q; Q is selected from S (O) q, CH2, C (=S), C (=NR8) or C (=O); W is selected from: a) heterocycle, and b) aryl; Y is selected from: a) aryl, and b) heterocycle; Z is selected from: a) aryl, b) heterocycle, c) C3-C6 cycloalkyl, and d) a bond; m is 0,1,2,3 or 4; n is 3 or 4; pis 3 or 4; q is 0,1 or 2; ris 0,1,2,3, or 4; s is 0,1,2,3 or 4; tis ; xis ; provided that the moiety does not represent a bond; and provided that if attachment of

to the rest of the molecule is through a nitrogen ring atom, then A1 is NR8, O, or S (O)q; or a pharmaceutically acceptable salt, an optical isomer or stereoisomer thereof.

Another embodiment of the compounds of this invention is illustrated by the formula A:

wherein: Rlav Rlb and R I C are independently selected from: a) hydrogen, b) unsubstituted or substituted aryl; unsubstituted or substituted heterocycle, C3-Clo cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, R8O-, R9S (O) q-CN, NO2, R8C (O)-, R8OC (O)-, N (R8) 2, (R8) 2NC (O)-, C (O) N (R8)-, or N3;

c) Cl-C6 alkyl, unsubstituted or substituted by unsubstituted or substituted aryl, unsubstituted or substituted heterocycle, C3-C10, cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, R8O-, R9S (O) q-, CN, R8C (O)-, R8OC (O)-, N (R8) 2, N3, or R8C (O) O-; RI is selected from: a) H, b) unsubstituted or substituted Cl-C6 alkyl, c) unsubstituted or substituted aryl, d) unsubstituted or substituted heterocycle, e)- (Cl-C6 alkyl) N (R8) 2, f)-R8C(O)R8, g)- (C1-C6 alkyl) OR8, h)-N (R8) 2, i)-OR8, j) -R8NHC(O)R8, k)-R8C (O) N (R8) 2, 1) CF3, m) halo, n)-C (O) OR8, o) C2-C6 alkynyl, p) C2-C6 alkenyl, q) perfluoroalkyl, r) N3, s) N02, t) CN, and u) R9S (O) q- ; R2 is selected from: a) hydrogen, b) CN, c) N02, d) halogen, e) aryl, unsubstituted or substituted,

f) heteroaryl, unsubstituted or substituted, g) C1-C6 alkyl, unsubstituted or substituted, h) ORS, i) N3, j) R9S (O) q, k) R8HC=CH-, and l) R8C#C-; R3 is selected from: a) H, b) CN, c) N02, d) halogen, e) C1-C6 alkyl, unsubstituted or substituted, f) OR8, g) aryl, unsubstituted or substituted, h) heteroaryl, unsubstituted or substituted, and i) CF3; R4 is selected from: a) H, b) =O, or c) =S; R5 is selected from: a) H, b) CN, c) N02, d) halogen, e) C1-C6 alkyl, unsubstituted or substituted, f) N3, g) R9S (O) q, h)-HC=CH2, i) HC#C-,

j) aryl, unsubstituted or substituted, k) heterocycle, unsubstituted or substituted, 1) CF30-, m) CF3CH20-, n) C3-C 10 cycloalkyl, o) CF3, p)- (C1-C6 alkyl) N (R8) 2, q)-(C1-C6 alkyl) OR8, r) OR8, s) N (R8) 2, t)-C (O) (C1-C6 alkyl), and u) -(C1-C6 alkyl) C (O) R8; R6 is selected from: a) H, b) C1-C6 alkyl, unsubstituted or substituted, c) OR8, and d)-C (O) (C1-C6 alkyl); R8 is independently selected from hydrogen, unsubstituted or substituted C1-C6 alkyl, unsubstituted or substituted benzyl, unsubstituted or substituted heterocycle and unsubstituted or substituted aryl; R9 is independently selected from unsubstituted or substituted C1-C6 alkyl, unsubstituted or substituted benzyl and unsubstituted or substituted aryl; AL and A2 are independently selected from: a) a bond, b)-HC=CH-, c)-C=C-, d) O, e) S (O) q, OC(O),

g) C (O), h) C (O) O, and A3 is selected from a bond,-C (=O)-, and C3-C6 cycloalkyl; M is selected from CH2, NH, O or S; W is selected from: a) hydrogen, b) heterocycle, and c) aryl; Y is selected from: a) aryl, and b) heterocycle; Z is selected from: a) aryl, b) heterocycle, c) C3-C6 cycloalkyl, and d) a bond; m is 0,1,2,3 or 4; n is 0,1,2,3 or 4; pis 3 or 4; q is 0,1 or 2; r is 0, 1,2,3, or 4; sis 3 or 4; 0,1,2or3;tis or a pharmaceutically acceptable salt, an optical isomer or stereoisomer thereof.

In a further embodiment, the compounds of the instant invention are illustrated by formula A:

wherein: R1a, R1b and Ric are independently selected from: a) hydrogen, b) unsubstituted or substituted aryl; unsubstituted or substituted heterocycle, C3-Clo cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, R8O-, R9S (O) q-, CN, N02, R8C (O)-, R80C (O)-, N (R8) 2, (R8) 2NC (O)-, C (O) N (R8)-, orN3; c) C1-C6 alkyl, unsubstituted or substituted by unsubstituted or substituted aryl, unsubstituted or substituted heterocycle, C3-Cl0 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, R80-, R9S (O) q-, CN, R8C (O)-, R80C (O)-, N (R8) 2, N3, or R8C (O) 0- ; RI is selected from: a) H, b) unsubstituted or substituted C1-C6 alkyl, c) unsubstituted or substituted aryl, d) unsubstituted or substituted heterocycle, e)- (C I-C6 alkyl) N (R8) 2, f) -(C1-C6 alkyl) C (O) R8, g)-(C1-C6 alkyl) OR8,

h)-N (R8) 2, i)-OR8, j)-(C1-C6 alkyl) NHC (O) R8, k) -(C1-C6 alkyl) C (O) N (R8) 2, 1) CF3, and m) halo; R2 is selected from: a) hydrogen, b) CN, c) N02, d) halogen, e) aryl, unsubstituted or substituted, f) heteroaryl, unsubstituted or substituted, g) C1-C6 alkyl, unsubstituted or substituted, h) OR8, i)N3, j) R9S (O) q, k) R8HC=CH-, and 1) R8C=-C- ; R3 is selected from: a) H, b) CN, c) N02, d) halogen, e) Cl-C6 alkyl, unsubstituted or substituted, f) OR8, g) aryl, unsubstituted or substituted, h) heteroaryl, unsubstituted or substituted, and i) CF3; R4 is selected from: a) H,

b) =0, or c) =S; R5 is selected from: a) H, b) CN, c) N02, d) halogen, e) C1-C6 alkyl, unsubstituted or substituted, f)n3, g) R9S(O)q, h)-HC=CH2, i) HC=-C-, j) aryl, unsubstituted or substituted, k) heterocycle, unsubstituted or substituted, 1) CF30-, m) CF3CH20-, n) C3-C 10 cycloalkyl, o) CF3, p)- (Cl-C6 alkyl) N (R8) 2, alkyl)OR8,q)-(C1-C6 r) OR8, s) N (R8) 2, t)-C (O) (Cl-C6 alkyl), and u) -(C1-C6 alkyl) C (O) R8; R6 is selected from: a) H, b) C1-C6 alkyl, unsubstituted or substituted, C) and d)-C (O) (C1-C6 alkyl); R8 is independently selected from

hydrogen, unsubstituted or substituted Cl-C6 alkyl, benzyl and unsubstituted or substituted aryl; R9 is independently selected from unsubstituted or substituted C1-C6 alkyl, benzyl and unsubstituted or substituted aryl; Al is selected from: a) a bond, b)-HC=CH-, c)-C=C-, d) O, e) S (O) q, f) OC (O), g) C (O), h) C (O) O, and i)NR8; A2 is selected from: a) a bond, b) O, c) S (O) q, d) C (O), and A3 is selected from a bond,-C (=O)-, and C3-C6 cycloalkyl; M is selected from CH2 or NH; W is selected from: a) heterocycle, and b) aryl; Y is selected from:

a) aryl, and b) heterocycle; Z is selected from: a) aryl, b) heterocycle, and c) a bond; mis 0,1,2,3 or 4; n is 3 or 4; pis 3 or 4; q is 0, 1 or 2; ris 3, or 4; s is and 0,1,2or3;tis or a pharmaceutically acceptable salt, an optical isomer or stereoisomer thereof.

Another embodiment of the compounds of this invention is illustrated by formula B: wherein: Rla and R I C are independently selected from: a) hydrogen,

b) unsubstituted or substituted aryl; unsubstituted or substituted heterocycle,C2-C6alkenyl,C2-C6alkynyl,R8O-,cycloalkyl, R9S (O) q-, CN, N02, R8C (O)-, R80C (O)-, N (R8) 2, (R8) 2NC (O)-, C (O) N (R8)-, or N3; c) C1-C6 alkyl, unsubstituted or substituted by unsubstituted or substituted aryl, unsubstituted or substituted heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, R8O-, R9S (O) q-, CN, R8C (O)-, R80C (O)-, N (R8) 2, N3, or R8C (O) 0- ; R1 is selected from: a) H, b) unsubstituted or substituted C l-C6 alkyl, c) unsubstituted or substituted aryl, d) unsubstituted or substituted heterocycle, e) -(C1-C6 alkyl) N (R8) 2, - (C1-C6 alkyl) C (O) R8, g)-(C1-C6 alkyl) OR8, h)-N (R8) 2, i)-OR8, j)- (C I-C6 alkyl) NHC (O) R8, k)-(C1-C6(C1-C6 alkyl) C (O) N (R8) 2, 1) CF3, and m) halo; R2 is selected from: a) hydrogen, b) CN, c) N02, d) halogen, e) aryl, unsubstituted or substituted, f) heteroaryl, unsubstituted or substituted, g) C1-C6 alkyl, unsubstituted or substituted, h) OR8, N3,

j) R9S (O) q, k) R8HC=CH-, and 1) R8C=-C-; R3 is selected from: a) H, b) CN, c) N02, d) halogen, e) C1-C6 alkyl, unsubstituted or substituted, f) OR8, g) aryl, unsubstituted or substituted, h) heteroaryl, unsubstituted or substituted, and i) CF3; R4 is selected from: a) H, b) =0, or c) =S; R5 is selected from: a) H, b) CN, c) N02, d) halogen, e) Cl-C6 alkyl, unsubstituted or substituted, f) N3, g) R9S(O)q, h)-HC=CH2, i) HC_C-, j) aryl, unsubstituted or substituted, k) heterocycle, unsubstituted or substituted, 1) CF30-, m) CF3CH20-,

n) C3-C 10 cycloalkyl, o) CF3, p) -(C1-C6 alkyl) N (R8) 2, alkyl)OR8,q)-(C1-C6 r) OR8, s) N (R8) 2, t)-C (O) (C1-C6 alkyl), and u) -(C1-C6 alkyl) C (O) R8; R6 is selected from: a) H, b) Cl-C6 alkyl, unsubstituted or substituted, c) and d)-C (O) (Cl-C6 alkyl); R8 is independently selected from hydrogen, unsubstituted or substituted C1-C6 alkyl, benzyl and unsubstituted or substituted aryl; R9 is independently selected from unsubstituted or substituted C1-C6 alkyl, benzyl and unsubstituted or substituted aryl; A1 is selected from: a) a bond, b)-HC=CH-, c)-C=C-, d) O, e) S (O) q, 0 OC (O), g) C (O), h) and i) NR8 ;

W is selected from: a) heterocycle, and b) aryl; Y is selected from: a) aryl, and b) heterocycle; Z is selected from: a) aryl, and b) heterocycle; m is 3 or 4; n is 3 or 4; pis 3 or 4; qis 0,1 or 2; sis and tis ; or a pharmaceutically acceptable salt, an optical isomer or stereoisomer thereof.

Another embodiment of the compounds of this invention is illustrated by the formula II:

wherein: Ric is independently selected from: a) hydrogen, b) unsubstituted or substituted aryl, unsubstituted or substituted heterocycle, C3-Clo cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, R9S (O) q-, CN, N02, R8C (O)-, R80C (O)-, (R8) 2NC (O)-,-N (R15) 2, -OR 15,-N (R8) S (O) qR8 or N3; c) Cl-C6 alkyl, unsubstituted or substituted by unsubstituted or substituted aryl, unsubstituted or substituted heterocycle, C3-C 10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, R8O-, R9S (O) q-, CN, R8C (O)-, R80C (O)-, N (R8) 2, N3, or R8C (O) 0- ; R1 is independently selected from: a) H, b) unsubstituted or substituted C1-C6 alkyl, c) unsubstituted or substituted aryl, d) unsubstituted or substituted heterocycle, e) -(C1-C6 alkyl) N (R8) 2, f)-R8C(O)R8, g)-(C1-C6 alkyl) OR8, h)-N (R8) 2, i)-OR8, j)-R8NHC (O) R8, k)-R8C (O) N (R8) 2, 1) CF3, m) halo, n)-C (O) OR8, o) C2-C6 alkynyl, p) C2-C6 alkenyl, q) perfluoroalkyl, r) N3, s) N02, t) CN, u) R9S (O) q-,

v) -(C1-C6 alkyl)NR8C(O) N (R8) 2, and w)- (Ci-C6 alkyl) R14 ; R2 is independently selected from: a) hydrogen, b) CN, c) N02, d) halogen, e) aryl, unsubstituted or substituted, heteroaryl, unsubstituted or substituted, g) C1-C6 alkyl, unsubstituted or substituted, h) OR8, N N3, j) R9S (O) q, k) R8HC=CH-, and l) R8C#C-; R3 is independently selected from: a) H, b) CN, c) N02, d) halogen, e) C1-C6 alkyl, unsubstituted or substituted, f)OR8, g) aryl, unsubstituted or substituted, h) heteroaryl, unsubstituted or substituted, and i) CF3; R5 and R6 are independently selected from: a) H, b) CN, c) N02, d) halogen, e) C1-C6 alkyl, unsubstituted or substituted,

f)N3, g) R9S (O) q, h)-HC=CH2, i) HC=-C-, j) aryl, unsubstituted or substituted, k) heterocycle, unsubstituted or substituted, 1) CF30-, m) CF3CH20-, n) C3-C 10 cycloalkyl, o) CF3, p)-(Cl-C6(Cl-C6 alkyl) N (R8) 2, alkyl)OR8,q)-(C1-C6 r) OR8, s) N (R8) 2, t) alkyl), u) -(C1-C6 alkyl) C (O) R8, v)-C (O) O (Cl-C6 alkyl), w)-C (O) N (R8) 2, x) -(C1-C6 alkyl)NR8C(O) N (R8) 2, y) -(C1-C6 alkyl) NR8C (O) R8, z)-C2-C6 alkynyl, aa)-C2-C6 alkenyl, alkyl)N3,bb)-(C1-C6 alkyl)NR8S(O)q,andcc)-(C1-C6 dd) -(C1-C6 alkyl)NR8(C1-C6 alkyl) N (R8) 2; R8 is independently selected from hydrogen, unsubstituted or substituted Cl-C6 alkyl, unsubstituted or substituted aralkyl, unsubstituted or substituted heterocycle and unsubstituted or substituted aryl; R9 is independently selected from unsubstituted or substituted C1-C6 alkyl, unsubstituted or substituted benzyl and unsubstituted or substituted aryl;

R14 is unsubstituted saturated heterocycle; R15 is independently selected from a) hydrogen, unsubstituted or substituted aryl, unsubstituted or substituted heterocycle, C3-Clo cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, R9S (0) 2-, RSC (O)-, R80C (O)-, (R8) 2NC (O)-, and b) C1-C6 alkyl, unsubstituted or substituted by unsubstituted or substituted aryl, unsubstituted or substituted heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, R8O-, R9S (O) q-, CN, R8C (O)-, R80C (O)-, N (R8) 2, N3, or R8C (O) 0- ; Al and A2 are independently selected from: a) a bond, b)-HC=CH-, c)-C=C-, d) O, e) S (O) q, f) OC(O), g) C (O), h) C (O) O, and A3 is selected from a bond,-C (=O)-, and C3-C6 cycloalkyl; M is selected from CH2, NR8, 0 or S (O) q; Q is selected from S (O) q, CH2, C (=S), C (=NR8) or C (=O); W is a heterocycle; Y is selected from a) aryl, and

b) heterocycle; Z is selected from a) aryl, and b) heterocycle; m is 0,1,2,3 or 4; nis 0,1,2,3 or 4; pis 0,1,2,3 or 4; qis 0,1 or 2 ; ris 0,1,2,3, or 4; sis 3 or 4; t is ; xxis 0, or3;2 provided that if attachment of to the rest of the molecule is through a nitrogen ring atom, then Al is NR8, O, or S (O) q; or a pharmaceutically acceptable salt, an optical isomer or stereoisomer thereof.

Another embodiment of the compounds of this invention is illustrated by the formula III:

wherein: Rla and Rlc are independently selected from: a) hydrogen, b) unsubstituted or substituted aryl, unsubstituted or substituted heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, R9S (O) q-, CN, N02, R8C (O)-, R80C (O)-, (R8) 2NC (O)-,-N (R15) 2, -OR15,-N (R8) S (O) qR8 or N3; c) Cl-C6 alkyl, unsubstituted or substituted by unsubstituted or substituted aryl, unsubstituted or substituted heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, R8O-, R9S (O) q-, CN, R8C (O)-, R80C (O)-, N (R8) 2, N3, or R8C (O) O-; Rl is independently selected from: a) H, b) unsubstituted or substituted C1-C6 alkyl, c) unsubstituted or substituted aryl, d) unsubstituted or substituted heterocycle, e) -(C1-C6 alkyl) N (R8) 2, f)-R8C (0) R8, g)- (Cl-C6 alkyl) OR8, h)-N (R8) 2, i)-OR8,

j)-R8NHC (O) R8, k)-R8C (O) N (R8) 2, 1) CF3, m) halo, n)-C (O) OR8, o) C2-C6 alkynyl, p) C2-C6 alkenyl, q) perfluoroalkyl, r) N3, s) N02, t) CN, u) R9S (O) q-, v)-(C1-C6 alkyl) NR8C (O)(C1-C6 alkyl) NR8C (O) N (R8) 2, and alkyl)R14;w)-(C1-C6 R2 is independently selected from: a) hydrogen, b) CN, c) N02, d) halogen, e) aryl, unsubstituted or substituted, heteroaryl, unsubstituted or substituted, g) C1-C6 alkyl, unsubstituted or substituted, h) ORS, i) N3, j) R9S (O) q, k) R8HC=CH-, and l) R8C#C-; R3 is independently selected from: a) H, b) CN, c) N02, d) halogen,

e) C1-C6 alkyl, unsubstituted or substituted, f)OR8, g) aryl, unsubstituted or substituted, h) heteroaryl, unsubstituted or substituted, and i) CF3; R5 and R6 are independently selected from: a) H, b) CN, c) N02, d) halogen, e) C I-C6 alkyl, unsubstituted or substituted, f) N3, g) R9S (O) q, h)-HC=CH2, i) HC-C-, j) aryl, unsubstituted or substituted, k) heterocycle, unsubstituted or substituted, 1) CF30-, m) CF3CH20-, n) C3-C 10 cycloalkyl, o) CF3, -(Cl-C6 alkyl)(Cl-C6 alkyl) N (R8) 2, alkyl)OR8,q)-(C1-C6 r) ORS, s) N (R8) 2, t) alkyl), u) -(C1-C6 alkyl) C (O) R8, v)-C (O) O (C1-C6 alkyl), w)-C (O) N (R8) 2, x) -(C1-C6 alkyl)NR8C(O) N (R8) 2, alkyl)NR8C(O)R8,y)-(C1-C6 z)-C2-C6 alkynyl, aa)-C2-C6 alkenyl,

alkyl)N3,bb)-(C1-C6 cc)-(C1-C6(C1-C6 alkyl) NRSS (O) q, and dd) -(C1-C6 alkyl)NR8(C1-C6 alkyl) N (R8) 2; R8 is independently selected from hydrogen, unsubstituted or substituted C I-C6 alkyl, unsubstituted or substituted aralkyl, unsubstituted or substituted heterocycle and unsubstituted or substituted aryl; R9 is independently selected from unsubstituted or substituted C1-C6 alkyl, unsubstituted or substituted benzyl and unsubstituted or substituted aryl; R14 is unsubstituted saturated heterocycle; R15 is independently selected from a) hydrogen, unsubstituted or substituted aryl, unsubstituted or substituted heterocycle, C3-Clo cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, R9S (0) 2-, R8C (O)-, R80C (O)-, (R8) 2NC (O)-, and b) C 1-C6 alkyl, unsubstituted or substituted by unsubstituted or substituted aryl, unsubstituted or substituted heterocycle, C3-C 10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, R80-, R9S (O) q-, CN, R8C (O)-, R80C (O)-, N (R8) 2, N3, or R8C (O) O-; Al is selected from: a) a bond, b)-HC=CH-, c)-C=C-, d) O, e) S (O) q, f)OC(O), g) C (O), h) C (O) O, and i) NR8 ;

A3 is selected from a bond,-C (=O)-, and C3-C6 cycloalkyl; M is selected from CH2, NR8,0 or S (O) q; Q is selected from S (O) q, CH2, C (=S), C (=NR8) or C (=O); W is a heterocycle selected from pyrrolidinyl, imidazolyl, pyridyl, triazolyl, pyridonyl, 2-oxopiperidinyl, indolyl, quinolinyl, isoquinolinyl, and tetrahydroimidazopyridinyl, Y is selected from: a) aryl, and b) pyridyl; Z is selected from: a) aryl, and b) pyridyl; mis 3 or 4; n is 3 or 4; pis 0,1,2,3 or 4; q is 0,1 or 2; sis 0,1,2,3 or 4; tis ; x is ; provided that if attachment of to the rest of the molecule is through a nitrogen ring atom, then Al is NR8, O, or S (O) q;

or a pharmaceutically acceptable salt, an optical isomer or stereoisomer thereof.

Another embodiment of the compounds of this invention is illustrated by the formula C: wherein: Rla and R1c are independently selected from: a) hydrogen, b) unsubstituted or substituted aryl; unsubstituted or substituted heterocycle, C3-CIo cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, R8O-, R9S (O) q-, CN, N02, R8C (O)-, R80C (O)-, N (R8) 2, (R8) 2NC (O)-, C (O) N (R8)-, or N3; c) Cl-C6 alkyl, unsubstituted or substituted by unsubstituted or substituted aryl, unsubstituted or substituted heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, R80-, R9S (O) q-, CN, R8C (O)-, R80C (O)-, N (R8) 2, N3, or R8C (O) 0- ; R1 is selected from: a) H, b) unsubstituted or substituted C 1-C6 alkyl, c) unsubstituted or substituted aryl, d) unsubstituted or substituted heterocycle, e)- (C1-C6 alkyl) N (R8) 2,

0-(c-c6 alkyl)(c-c6 alkyl) C (O) R8, alkyl)OR8,g)-(C1-C6 h)-N (R8) 2, i)-OR8, j) -(C1-C6 alkyl) NHC (O) R8, k) -(C1-C6 alkyl) C (O) N (R8) 2, 1) CF3, and m) halo; R2 is selected from: a) hydrogen, b) CN, c) N02, d) halogen, e) aryl, unsubstituted or substituted, heteroaryl, unsubstituted or substituted, g) C1-C6 alkyl, unsubstituted or substituted, h) OR8, i)N3, j) R9S (O) q, k) R8HC=CH-, and 1) R8C#C-; R3 is selected from: a) H, b) CN, c) N02, d) halogen, e) C1-C6 alkyl, unsubstituted or substituted, f)OR8, g) aryl, unsubstituted or substituted, h) heteroaryl, unsubstituted or substituted, and i) CF3;

R4 is selected from: a) H, b) =0, or c) =S; R5 is selected from: a) H, b) CN, c) N02, d) halogen, e) C 1-C6 alkyl, unsubstituted or substituted, f)N3, g)R9S(O)q, h)-HC=CH2, i) HC=-C-, j) aryl, unsubstituted or substituted, k) heterocycle, unsubstituted or substituted, 1) CF30-, m) CF3CH20-, n) C3-C 10 cycloalkyl, o) CF3, p)-(C1-C6 alkyl)(C1-C6 alkyl) N (R8) 2, alkyl)OR8,q)-(C1-C6 r) OR8, s) N (R8) 2, t)-C (O) (C1-C6 alkyl), and u) -(C1-C6 alkyl) C (O) R8; R6 is selected from: a) H, b) C 1-C6 alkyl, unsubstituted or substituted, c) OR8, and d)-C (O) (C1-C6 alkyl) ;

R8 is independently selected from hydrogen, unsubstituted or substituted C1-C6 alkyl, benzyl and unsubstituted or substituted aryl; R9 is independently selected from unsubstituted or substituted Cl-C6 alkyl, benzyl and unsubstituted or substituted aryl; A1 is selected from: a) a bond, b)-HC=CH-, c)-C=C-, d) O, e) S (O) q, f) (O), g) C (O), h) C (O) O, and W is selected from: a) heterocycle, and b) aryl; mis 0,1,2,3 or 4; n is 0,1,2,3 or 4; p is 3 or 4; q is 0, 1 or 2; s is 3 or 4; and t is ; or a pharmaceutically acceptable salt, an optical isomer or stereoisomer thereof.

Another embodiment of the compounds of this invention is illustrated by formula IV:

wherein: R1c'is selected from: a) hydrogen, b) unsubstituted or substituted aryl, unsubstituted or substituted heterocycle, C3-Clo cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl; c) C1-C6 alkyl, unsubstituted or substituted by unsubstituted or substituted aryl, unsubstituted or substituted heterocycle, C3-C 10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, R8O-, R9S (O) q-, CN, R8C (O)-, R80C (O)-, N (R8) 2, N3, or R8C (O) 0- ; Roc ils selected from: a) hydrogen, b) N (R15) 2, and c) OR15; RI is independently selected from: a) H, b) unsubstituted or substituted C1-C6 alkyl, c) unsubstituted or substituted aryl, d) unsubstituted or substituted heterocycle, C)- (C I-C6 alkyl) N (R8) 2, f)-R8C (O) R8, g)- (C1-C6 alkyl) OR8, h)-N (R8) 2,

i)-OR8, j) -R8NHC(O)R8, k)-R8C (O) N (R8) 2, 1) CF3, m) halo, n)-C (O) OR8, o) C2-C6 alkynyl, p) C2-C6 alkenyl, q) perfluoroalkyl, r) N3, s) N02, t) CN, u) R9S (O) q-, v)-(C1-C6 alkyl) NR8C (O)(C1-C6 alkyl) NR8C (O) N (R8) 2, and alkyl)R14;w)-(C1-C6 R2 is independently selected from: a) hydrogen, b) CN, c) N02, d) halogen, e) aryl, unsubstituted or substituted, heteroaryl, unsubstituted or substituted, g) C1-C6 alkyl, unsubstituted or substituted, h) OR8, i) N3, j) R9S(O)q, k) R8HC=CH-, and l) R8C#C-; R3 is independently selected from: a) H, b) CN, c) N02,

d) halogen, e) C 1-C6 alkyl, unsubstituted or substituted, f)OR8, g) aryl, unsubstituted or substituted, h) heteroaryl, unsubstituted or substituted, and i) CF3; R5 is independently selected from: a) H, b) CN, c) N02, d) halogen, e) C1-C6 alkyl, unsubstituted or substituted, f) N3, g) R9S(O)q, h)-HC=CH2, i) HC--C-, j) aryl, unsubstituted or substituted, k) heterocycle, unsubstituted or substituted, 1) CF30-, m) CF3CH20-, n) cycloalkyl, o) CF3, p) -(C1-C6 alkyl) N (R8) 2, q)- (Cl-C6 alkyl) OR8, r) OR8, s) N (R8) 2, t)-C (O) (CI-C6 alkyl), u) -(C1-C6 alkyl) C (O) R8, v)-C (O) O (C1-C6 alkyl), w)-C (O) N (R8) 2, x) -(C1-C6 alkyl) NR8C (O) N (R8) 2, alkyl)NR8C(O)R8,y)-(C1-C6 z)-C2-C6 alkynyl,

aa)-C2-C6 alkenyl, alkyl)N3,bb)-(C1-C6 cc)-(C1-C6 alkyl) NR8S (O)(C1-C6 alkyl) NR8S (O) q, and dd) -(C1-C6 alkyl) NR8 (CI-C6 alkyl) N (R8) 2; R6 is selected from a) H, b) Cl-C6 alkyl, unsubstituted or substituted, c) aryl, unsubstituted or substituted, d) heterocycle, unsubstituted or substituted, e) C3-Ciocycloalkyl, f)- (Ci-C6alkyl) N (R8) 2, alkyl)OR8,g)-(C1-C6 h)-C (O) (C1-C6 alkyl), i) -(C1-C6 alkyl) C (O) R8, j)-C (O) O (C1-C6 alkyl), k)-C (O) N (R8) 2, 1)- (C1-C6 alkyl) NR8C (O) N (R8) 2, m)-(C1-C6 alkyl) NR8C (O) R8, n)-C2-C6 alkynyl, o)-C2-C6 alkenyl, alkyl)N3,p)-(C1-C6 q)-(C1-C6 alkyl) NR8S (O)(C1-C6 alkyl) NR8S (O) q, and r)-(C1-C6 alkyl) NR8 (Cl-C6 alkyl)(C1-C6 alkyl) NR8 (Cl-C6 alkyl) N (R8) 2; R8 is independently selected from hydrogen, unsubstituted or substituted C1-C6 alkyl, unsubstituted or substituted aralkyl, unsubstituted or substituted heterocycle and unsubstituted or substituted aryl; R9 is independently selected from unsubstituted or substituted C1-C6 alkyl, unsubstituted or substituted benzyl and unsubstituted or substituted aryl;

R14 is unsubstituted saturated heterocycle; R15 is independently selected from a) hydrogen, unsubstituted or substituted aryl, unsubstituted or substituted heterocycle, C3-Clo cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, R9S (0) 2-, R8C (O)-, R80C (O)-, (R8) 2NC (O)-, and b) Cl-C6 alkyl, unsubstituted or substituted by unsubstituted or substituted aryl, unsubstituted or substituted heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, R80-, R9S (O) q-, CN, R8C (O)-, R80C (O)-, N (R8) 2, N3, or R8C (O) 0- ; A1 is selected from a) O, b) S (O) q, c) C (O), and d)NR8; M is selected from CH2, NR8,0 or S (O) q; Q is selected from S (O) q, CH2, C (=S), C (=NR8) or C (=O); W is a heterocycle selected from imidazolyl, triazolyl or pyridyl; Y is selected from phenyl or pyridyl; Z is selected from phenyl or pyridyl; mis 0,1,2,3 or 4; n is 3 or 4; qis 0,1 or 2; sis 3 or 4; tis 0,1,2 or 3; x is 0,1,2 or 3; provided that if attachment of

to the rest of the molecule is through a nitrogen ring atom, then A is NRS Q, or S (O) q; or a pharmaceutically acceptable salt, an optical isomer or stereoisomer thereof.

Another embodiment of the compounds of this invention is illustrated by formulaD: Rla and Ric are independently selected from: a) hydrogen, b) unsubstituted or substituted aryl; unsubstituted or substituted heterocycle, C3-Clo cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, R80-, R9S (O) q-, CN, N02, R8C (O)-, R80C (O)-, N (R8) 2, (R8) 2NC (O)-, C (O) N (R8)-, or N3; 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, R80-, R9S (O) q-, CN, R8C (O)-, R80C (O)-, N (R8) 2, N3, or R8C (O) 0- ;

RI is selected from: a) H, b) unsubstituted or substituted C I-C6 alkyl, c) unsubstituted or substituted aryl, d) unsubstituted or substituted heterocycle, e) -(C1-C6 alkyl) N (R8) 2, - (Cl-C6 alkyl) C (O) R8, g)- (C1-C6 alkyl) OR8, h)-N (R8) 2, i)-OR8, j) -(C1-C6 alkyl) NHC (O) R8, k) -(C1-C6 alkyl) C (O) N (R8) 2, 1) CF3, and m) halo; R2 is selected from: a) hydrogen, b) CN, c) N02, d) halogen, e) aryl, unsubstituted or substituted, heteroaryl, unsubstituted or substituted, g) Cl-C6 alkyl, unsubstituted or substituted, h) OR8, i) N3, j) R9S (O) q, k) R8HC=CH-, and l) R8C#C-; R3 is selected from: a) H, b) CN, c) N02, d) halogen,

e) Cl-C6 alkyl, unsubstituted or substituted, f)OR8, g) aryl, unsubstituted or substituted, h) heteroaryl, unsubstituted or substituted, and i) CF3; R4 is selected from: a) H, or b) =O; R5 is selected from: a) H, b) CN, c) N02, d) halogen, e) C1-C6 alkyl, unsubstituted or substituted, f) N3, g) R9S (O) q, h)-HC=CH2, i) HC=C-, j) aryl, unsubstituted or substituted, k) heterocycle, unsubstituted or substituted, 1) CF30-, m) CF3CH20-, n) cycloalkyl, o) CF3, -(Cl-C6 alkyl) N (R8) 2, alkyl)OR8,q)-(C1-C6 r) OR8, s) N (R8) 2, t)-C (O) (C I-C6 alkyl), and u) -(C1-C6 alkyl) C (O) R8; R6 is selected from:

a) H, b) Cl-C6 alkyl, unsubstituted or substituted, and c)-C (O) (CI-C6 alkyl); R8 is independently selected from hydrogen, unsubstituted or substituted Cl-C6 alkyl, benzyl and unsubstituted or substituted aryl; R9 is independently selected from unsubstituted or substituted C 1-C6 alkyl, benzyl and unsubstituted or substituted aryl; W is a heterocycle, selected from imidazolyl or pyridyl; mis 0,1,2,3 or 4; nis 0,1,2,3 or 4; pis 0,1,2,3 or 4; q is 0,1 or 2; sis 3 or 4; and tis ; or a pharmaceutically acceptable salt, an optical isomer or stereoisomer thereof.

Specific examples of the compounds of the invention are: 4-imidazol-1-ylmethyl-2- 2- (2-oxo-piperidin-1-yl)-phenoxy-benzonitrile 4-imidazol-1-ylmethyl-2- 3- (2-oxo-piperidin-1-yl)-phenoxy-benzonitrile 4-imidazol-1-ylmethyl-2- 4- (2-oxo-piperidin-1-yl)-phenoxy-benzonitrile 4-imidazol-1-ylmethyl-2- 2- (3-methyl-2-oxo-piperidin-1-yl)-phenoxy-benzonitrile 4-Imidazol-I-ylmethyl-2-[2-(2-oxo-pyrrolidin-1-yl)-phenoxy]- benzonitrile

4-imidazol-1-ylmethyl-2- (3-morpholin-4-yl-phenoxy)-benzonitrile 4-imidazol-1-ylmethyl-2- (3-piperidin-1-ylmethyl-phenoxy)-benzonitrile 4-imidazol-1-ylmethyl-2- 3-(2-oxo-azepan-1-yl)-phenoxy-benzonitrile 2- 2- (3, 3-dimethyl-2-oxo-piperidin-1-yl)-phenoxy-4-imidazol-1-ylmeth yl- benzonitrile 2- 3- (3-ethyl-I-methyl-2-oxo-azepan-3-yl)-phenoxy-4-imidazol-I-yl methyl- benzonitrile 2- 3- (3 (S)-ethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-4-imidazol-1-y lmethyl- benzonitrile 2-3- (3 (R)-ethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-4-imidazol-1-y lmethyl- benzonitrile 2- 3- (3-ethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-4- (2-methyl-imidazol-1- yl) methyl-benzonitrile 2-3- (3-ethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-4- (5-methyl-imidazol-1- yl)methyl-benzonitrile 2-3- (3-ethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-4- (2, 5-dimethyl-imidazol-I- yl)methyl-benzonitrile 2-[3-(3-ethyl-1-methyl-2-oxo-azepan-3-yl)phenoxy]-4-[1, 2,4] triazol-4-ylmethyl- benzonitrile 2- 3- (3-ethyl-I-methyl-2-oxo-azepan-3-yl)-phenoxy-4- 1,2,4 triazol-1-ylmethyl- benzonitrile 4-imidazol-1-ylmethyl-2- 3- ( 1-methyl-2-oxo-azepan-3-yl)-phenoxy-benzonitrile 4-imidazol-1-ylmethyl-2- 3-(1-methyl-2-oxo-azocan-3-yl)-phenoxy-benzonitrile

4-imidazol-1-ylmethyl-2- 3- (l-methyl-2-oxo-piperidin-3-yl)-phenoxy-benzonitrile 4-imidazol-1-ylmethyl-2- 3- (l-methyl-2-oxo-pyrrolidin-3-yl)-phenoxy-benzonitrile 4-imidazol-1-ylmethyl-2-[3-(3-ethyl-1-methyl-2-oxo-piperidin -3-yl)-phenoxy]- benzonitrile 4-imidazol-1-ylmethyl-2- 3- (2-oxo-azepan-3-yl)-phenoxy-benzonitrile 2-3- (3-hydroxymethyl-I-methyl-2-oxo-azepan-3-yl)-phenoxy-4-imida zol-1- ylmethyl-benzonitrile 2- 3- (3-cyclopropylmethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-4-i midazol-1- ylmethyl-benzonitrile 2-4-bromo-3- (3-cyclopropylmethyl-l-methyl-2-oxo-azepan-3-yl)-phenoxy-4- imidazol-1-ylmethyl-benzonitrile 2- 3- (3-methoxymethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-4-imida zol-1- ylmethyl-benzonitrile 2- 3- (3-ethyl-2-oxo-azepan-3-yl)-phenoxy-4-imidazol-1-ylmethyl-be nzonitrile 2- 3- (3-ethyl-azepan-3-yl)-phenoxy-4-imidazol-1-ylmethyl-benzonit rile 2- 3- (l-acetyl-3-ethyl-azepan-3-yl)-phenoxy-4-imidazol-1-ylmethyl -benzonitrile 3- 3- (2-cyano-5-imidazol-1-ylmethyl-phenoxy)-phenyl-3-ethyl-azepa ne-1-carboxylic acid-tert-butyl ester 4- 5- (2-amino-ethyl)-2-methyl-imidazol-1-ylmethyl-2- 3- (3-ethyl-1-methyl-2-oxo- azepan-3-yl)-phenoxy-benzonitrile

2- 3- (3-ethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-4- 2-methyl-5- (2-morpholin-4- yl-ethyl)-imidazol-1-ylmethyl-benzonitrile N- 2- (3- {4-cyano-3- 3- (3-ethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-benzyl}-2- methyl-3H-imidazol-4-yl)-ethyl-acetamide 3-ethyl-3- 3- (3-imidazol-1-ylmethyl-phenoxy)-phenyl-I-methyl-azepan-2-one 2- 3- (3-ethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-4- (3-methyl-3-H-imidazol-4- ylmethyl)-benzonitrile 2-[3-(3-ethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy]-4-(3H-imi dazol-4-ylmethyl)- benzonitrile 2- 3- (3-ethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-4- hydroxy- (3-methyl-3-H- imidazol-4-yl)-methyl]-benzonitrile 4- amino- (3-methyl-3-H-imidazol-4-yl)-methyl-2- 3- (3-ethyl-1-methyl-2-oxo- azepan-3-yl)-phenoxy-benzonitrile 2-3- (3-ethyl-1-methyl-2-oxo-azepan-3-yl)-benzyl-4- (3-methyl-3H-imidazole-4- carbonyl)-benzonitrile 2- 3- (3-ethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-4- (hydroxy-pyridin-3-yl- methyl)-benzonitrile 2- 3- (3-ethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-4-pyridin-3-ylm ethyl- benzonitrile 2- 3- (3-ethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-4-pyridin-2-ylm ethyl- benzonitrile 2- 3- (3-ethyl- I-methyl-2-oxo-azepan-3-yl)-phenoxy-4- I-hydroxy-I- (3-methyl-3H- imidazol-4-yl)-ethyl-benzonitrile

2- 3- (3R)-ethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-4- l- (R) amino-1- (3-methyl- 3H-imidazol-4-yl)-ethyl-benzonitrile 2- 3- ( S)-ethyl-l-methyl-2-oxo-azepan-3-yl)-phenoxy-4- l- (S) amino-1- (3-methyl- 3H-imidazol-4-yl)-ethyl-benzonitrile 2- 3- (3S)-ethyl-I-methyl-2-oxo-azepan-3-yl)-phenoxy-4- l- (R) amino-I- (3-methyl- 3H-imidazol-4-yl)-ethyl-benzonitrile 2- 3- (3R)-ethyl-I-methyl-2-oxo-azepan-3-yl)-phenoxy-4- l- (S) amino- I- (3-methyl- 3H-imidazol-4-yl)-ethyl-benzonitrile 2- 3-3 (R)-n-butyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-4- 1- (R) amino-1- (3- methyl-3H-imidazol-4-yl)-ethyl-benzonitrile 2- 3-3 (S)-n-butyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-4- 1- (S) amino-1- (3-methyl- 3H-imidazol-4-yl)-ethyl-benzonitrile 2- 3-3 (S)-n-butyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-4- 1- (R) amino-1- (3-methyl- 3H-imidazol-4-yl)-ethyl-benzonitrile 2- 3-3 (R)-n-butyl-l-methyl-2-oxo-azepan-3-yl)-phenoxy-4- 1- (S) amino-1- (3-methyl- 3H-imidazol-4-yl)-ethyl-benzonitrile 2- 3-3 (R)-cyclopropylmethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-4- 1- (R) amino- 1- (3-methyl-3H-imidazol-4-yl)-ethyl-benzonitrile 2- 3-3 (S)-cyclopropylmethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-4- 1- (S) amino- 1- (3-methyl-3H-imidazol-4-yl)-ethyl-benzonitrile

2- 3-3 (R)-cyclopropylmethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-4- 1- (S) amino- 1- (3-methyl-3H-imidazol-4-yl)-ethyl-benzonitrile 2-[3-3(S)-cyclopropylmethyl-1-methyl-2-oxo-azepan-3-yl)-phen oxy]-4-[1-(R)amino- 1- (3-methyl-3H-imidazol-4-yl)-ethyl-benzonitrile 2- 3-3 (R)-cyclopropylethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-4- 1- (R)- amino- 1- (3-methyl-3H-imidazol-4-yl)-ethyl-benzonitrile 2- 3-3 (S)-cyclopropylethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-4- 1- (S)- amino-1- (3-methyl-3H-imidazol-4-yl)-ethyl-benzonitrile 2-[3-3(R)-cyclopropylethyl-1-methyl-2-oxo-azepan-3-yl)-pheno xy]-4-[1-(S)-amino- 1- (3-methyl-3H-imidazol-4-yl)-ethyl-benzonitrile 2- 3-3 (S)-cyclopropylethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-4- 1- (R)- amino- 1- (3-methyl-3H-imidazol-4-yl)-ethyl-benzonitrile 2- 3-3 (R)-propyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-4- 1- (R)- amino-1- (3- methyl-3H-imidazol-4-yl)-ethyl-benzonitrile 2- 3-3 (S)- propyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-4- 1- ( S) amino-1- (3- methyl-3H-imidazol-4-yl)-ethyl-benzonitrile 2- 3-3 (R)-propyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-4- 1- ( S) amino-1- (3- methyl-3H-imidazol-4-yl)-ethyl-benzonitrile 2- 3-3 (S)-propyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-4- 1- (R)-amino-1- (3- methyl-3H-imidazol-4-yl)-ethyl-benzonitrile

2- 3-3 (R)-methoxymethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-4- 1- (R)- amino-1- (3-methyl-3H-imidazol-4-yl)-ethyl-benzonitrile 2- 3-3 (S)-methoxymethyl-l-methyl-2-oxo-azepan-3-yl)-phenoxy-4- 1- ( S)-amino- 1-(3-methyl-3H-imidazol-4-yl)-ethyl]-benzonitrile 2- 3-3 (R)-methoxymethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-4- 1- (S)-amino-1- (3-methyl-3H-imidazol-4-yl)-ethyl-benzonitrile 2- [3-3 (S)-methoxymethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-4- 1- (R)- amino-1- (3-methyl-3H-imidazol-4-yl)-ethyl-benzonitrile 2- 3-3 (R)-ethoxymethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-4- 1- (R)- amino-1- (3-methyl-3H-imidazol-4-yl)-ethyl-benzonitrile 2- 3-3 (S)-ethoxymethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-4- l- (S)- amino-1- (3-methyl-3H-imidazol-4-yl)-ethyl-benzonitrile 2- 3-3 (R)-ethoxymethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-4- 1- ( S)-amino-l- (3-methyl-3H-imidazol-4-yl)-ethyl-benzonitrile 2- 3-3 (S)-ethoxymethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-4- 1- (R)- amino-1- (3-methyl-3H-imidazol-4-yl)-ethyl-benzonitrile 2- 3-3 (R)-cyclopropylmethyl-1-methyl-2-oxo-piperidin-3-yl)-phenoxy -4- 1- (R)amino-1- (3-methyl-3H-imidazol-4-yl)-ethyl-benzonitrile 2- 3-3 (S)-cyclopropylmethyl-1-methyl-2-oxo-piperidin-3-yl)-phenoxy -4- 1- (S)amino-1- (3-methyl-3H-imidazol-4-yl)-ethyl-benzonitrile

2- 3-3 (R)-cyclopropylmethyl-1-methyl-2-oxo-piperidin-3-yl)-phenoxy -4- 1- (S) amino-I- (3-methyl-3H-imidazol-4-yl)-ethyl-benzonitrile 2-[3-3(S-cyclopropylmethyl-1-methyl-2-oxo-piperidin-3-yl)-ph enoxy]-4-[1-(R)amino- 1- (3-methyl-3H-imidazol-4-yl)-ethyl-benzonitrile 2- 3-3 (R)-benzyl-l-methyl-2-oxo-piperidin-3-yl)-phenoxy-4- 1- (R) amino-1- (3- methyl-3H-imidazol-4-yl)-ethyl-benzonitrile 2- 3-3 (S)-benzyl-l-methyl-2-oxo-piperidin-3-yl)-phenoxy-4- l- ( S) amino-l- (3- methyl-3H-imidazol-4-yl)-ethyl-benzonitrile 2- 3-3 (R)-benzyl-1-methyl-2-oxo-piperidin-3-yl)-phenoxy-4- 1- ( S) amino-l- (3- methyl-3H-imidazol-4-yl)-ethyl-benzonitrile 2- 3-3 (S)-benzyl-1-methyl-2-oxo-piperidin-3-yl)-phenoxy-4- 1- (R) amino-1- (3- methyl-3H-imidazol-4-yl)-ethyl-benzonitrile 2- 3-methyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-4- 1-amino-1- (3-methyl-3H- imidazol-4-yl)-ethyl-benzonitrile 2- 3- (3-ethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-4- 1- (3-methyl-3-H-imidazol-4- yl)-vinyl-benzonitrile 2- 3- (3-ethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-4- 1- (3-methyl-3-H-imidazol-4- yl)-cyclopropyl-benzonitrile 2-[3-N,N-dimethylaminomethyl-1-methyl-2-oxo-azepan-3-yl)-phe noxy]-4-[1- hydroxy-l- (3-methyl-3H-imidazol-4-yl)-ethyl-benzonitrile 2-[3-N,N-dimethylaminomethyl-1-methyl-2-oxo-azepan-3-yl)-phe noxy]-4-[1-amino- 1- (3-methyl-3H-imidazol-4-yl)-ethyl-benzonitrile

2- 3- (3-N, N-dimethylaminomethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-4- 1- (3- methyl-3-H-imidazol-4-yl)-vinyl-benzonitrile 4- 5- (2-N, N-dimethylaminoethyl)-2-methyl-imidazol-1-ylmethyl-2- 3- (3S-ethyl-l- methyl-2-oxo-azepan-3-yl)-phenoxy-benzonitrile 4- 5- (2-N, N-dimethylaminoethyl)-2-methyl-imidazol- I-ylmethyl-2- 3- (3R-ethyl- I- methyl-2-oxo-azepan-3-yl)-phenoxy-benzonitrile 2- (3- {4-cyano-3- 3- (3S-ethyI-l-methyl-2-oxo-azepan-3-yl)-phenoxy-benzyI}-2- methyl-3H-imidazol-4-yl)-ethyl-urea 2- (3- {4-cyano-3- 3- (3R-ethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-benzyl}-2- methyl-3H-imidazol-4-yl)-ethyl-urea 2- 3- (3-ethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-4-pyridin-N-oxi de-3-ylmethyl- benzonitrile 2- 3-1-methyl-2-oxo-pyrrolidin-3-yl)-phenoxy-4- l-amino-1- (3-methyl-3H-imidazol- 4-yl)-ethyl-benzonitrile 2-[3-(1-methyl-2-oxo-azepan-3-yl)-phenoxy]-4-[1-amino-1-(3-m ethyl-3H-imidazol-4- yl)-ethyl-benzonitrile 2- 3- (3-hydroxymethyl-I-methyl-2-oxo-azepan-3-yl)-phenoxy-4- l-amino-1- (3- methyl-3H-imidazol-4-yl)-ethyl-benzonitrile 2- 3- (3-cyclopropylmethyl-1-methyl-2-oxo-pyrrolidin-3-yl)-phenoxy -4- 1-amino-1- (3-methyl-3H-imidazol-4-yl)-ethyl-benzonitrile 2- 3-3 (R)-cyclopropylmethyl-1-methyl-2-oxo-pyrrolidin-3-yl)-phenox y-4- 1- (R)- amino-1- (3-methyl-3H-imidazol-4-yl)-ethyl-benzonitrile

2- 3-3 (S)-cyclopropylmethyl-l-methyl-2-oxo-pyrrolidin-3-yl)-phenox y-4- l- (S)- amino-1- (3-methyl-3H-imidazol-4-yl)-ethyl-benzonitrile 2- 3-3 (R)-cyclopropylmethyl-l-methyl-2-oxo-pyrrolidin-3-yl)-phenox y-4- l- (S)- amino-l- (3-methyl-3H-imidazol-4-yl)-ethyl-benzonitrile 2- 3-3 (S)-cyclopropylmethyl-l-methyl-2-oxo-pyrrolidin-3-yl)-phenox y-4- l- (R) amino-I- (3-methyl-3H-imidazol-4-yl)-ethyl-benzonitrile 2- 3-3 (R)-benzyl-l-methyl-2-oxo-pyrrolidin-3-yl)-phenoxy-4- l- (R)- amino-1- (3- methyl-3H-imidazol-4-yl)-ethyl-benzonitrile 2- 3-3 (S)-benzyl-l-methyl-2-oxo-pyrrolidin-3-yl)-phenoxy-4- 1- ( S)-amino-1- (3- methyl-3H-imidazol-4-yl)-ethyl-benzonitrile 2- 3-3 (R)-benzyl-1-methyl-2-oxo-pyrrolidin-3-yl)-phenoxy-4- 1- ( S)-amino-1- (3- methyl-3H-imidazol-4-yl)-ethyl-benzonitrile 2- 3-3 (S)-benzyl-1-methyl-2-oxo-pyrrolidin-3-yl)-phenoxy-4- 1- (R- amino-1- (3- methyl-3H-imidazol-4-yl)-ethyl-benzonitrile 4- 1- (4-allyl-5-bromo-pyridin-3-yl)-1-hydroxy-ethyl-2- 3- (3-ethyl-1-methyl-2-oxo- azepan-3-yl)-phenoxy]-benzonitrile 1-tert-butyl-3 (R)- 1- {4-cyano-3- 3- (3R-ethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy- phenyl}-1- (3-methyl-3H-imidazol-4-yl)-ethyl-urea 1-tert-butyl-3(S)-[1-{4-cyano-3-[3-(3S-ethyl-1-methyl-2-oxo- azepan-3-yl)-phenoxy]- phenyl}-1-(3-methyl-3H-imidazol-4-yl)-ethyl]-urea 1-tert-butyl-3 (R)- 1- {4-cyano-3- 3- (3 S-ethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy- phenyl}-1- (3-methyl-3H-imidazol-4-yl)-ethyl-urea

1-tert-butyl-3 (S)- 1- {4-cyano-3- 3- (3R-ethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy- phenyl}-I-(3-methyl-3H-imidazol-4-yl)-ethyl]-urea 1- 1- {4-cyano-3 (R)- 3- (3R-ethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-phenyl}-1- (3-methyl-3H-imidazol-4-yl)-ethyl-3-methyl-urea 1- 1- {4-cyano-3 (S)- 3- (3 S-ethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-phenyl}-1- (3-methyl-3H-imidazol-4-yl)-ethyl-3-methyl-urea 1- 1- f 4-cyano-3 (R)- 3- (3 S-ethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-phenyl}-1- (3-methyl-3H-imidazol-4-yl)-ethyl-3-methyl-urea 1- 1- {4-cyano-3 (S)- 3- (3R-ethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-phenyl}-1- (3-methyl-3H-imidazol-4-yl)-ethyl-3-methyl-urea 1- 1- {4-cyano-3 (R)- 3- (3 S-ethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-phenyl}-1- (3-methyl-3H-imidazol-4-yl)-ethyl-3-phenyl-urea 1-[1-{4-cyano-3(S)-[3-(3S-ethyl-1-methyl-2-oxo-azepan-3-yl)- phenoxy]-phenyl}-1- (3-methyl-3H-imidazol-4-yl)-ethyl-3-phenyl-urea l- l- {4-cyano-3 (R)- 3- (3R-ethyl-l-methyl-2-oxo-azepan-3-yl)-phenoxy-phenyl}-l- (3-methyl-3H-imidazol-4-yl)-ethyl-3-phenyl-urea l- l- {4-cyano-3 (R)- 3- (3S-ethyl-l-methyl-2-oxo-azepan-3-yl)-phenoxy-phenyl}-l- (3-methyl-3H-imidazol-4-yl)-ethyl-3-phenyl-urea [1-{4-cyano-3(R)-[3-(3R-ethyl-1-methyl-2-oxo-azepan-3-yl)-ph enoxy]phenyl}-1-(3- methyl-3H-imidazol-4-yl)-ethyl]-urea l- {4-cyano-3 (S)- 3- (3S-ethyl-l-methyl-2-oxo-azepan-3-yl)-phenoxy-phenyl}-l- (3- methyl-3H-imidazol-4-yl)-ethyl-urea

1- {4-cyano-3 (R)- 3- (3S-ethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-phenyl}-1- (3- methyl-3H-imidazol-4-yl)-ethyl-urea 1- {4-cyano-3 (S)- 3- (3R-ethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-phenyl}-1- (3- methyl-3H-imidazol-4-yl)-ethyl-urea N 1- {4-cyano-3- 3- (3-ethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-phenyl}-1- (3- methyl-3H-imidazol-4-yl)-ethyl-acetamide 2- 3- (3S-ethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-4- l-methylamino-1- (3- methyl-3H-imidazol-4-yl)-ethyl-benzonitrile 2- 3- (3R-ethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-4- 1-methylamino-l- (3- methyl-3H-imidazol-4-yl)-ethyl-benzonitrile 2- 3- (3S-ethyl-I-methyl-2-oxo-azepan-3-yl)-phenoxy-4- l (R)-methylamino-I- (3- methyl-3H-imidazol-4-yl)-ethyl-benzonitrile 2- 3- (3 S-ethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-4- 1 (S)-methylamino-1- (3- methyl-3H-imidazol-4-yl)-ethyl-benzonitrile 2- 5- (3 S-ethyl-1-methyl-2-oxo-azepan-3-yl)-2-iodo-phenoxy-4-imidazo l-1-ylmethyl- benzonitrile 2- 5- (3R-ethyl-1-methyl-2-oxo-azepan-3-yl)-2-iodo-phenoxy-4-imida zol-1-ylmethyl- benzonitrile 2- 5- ( S-ethyl-1-methyl-2-oxo-azepan-3-yl)-2-bromo-phenoxy-4-imidaz ol-1- ylmethyl-benzonitrile 2- 5- (3R-ethyl-1-methyl-2-oxo-azepan-3-yl)-2-bromo-phenoxy-4-imid azol-1- ylmethyl-benzonitrile 4-[1-amino-1-(3-methyl-3H-imidazol-4-yl)-ethyl]-2-[2-dimethy laminomethyl-5-(3S- ethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-benzonitrile

2-[3-(3-ethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy]-4-(4,5,6, 7-tetrahydro-3H- imidazo[4,5-] pyridin-4-yl)-benzonitrile 2- 3-ethyl-2-oxo-azepan-3-yl)-phenoxy-4- 1- amino-1- (3-methyl-3H-imidazol-4-yl)- ethyl-benzonitrile 4- 1-amino-1- (3-methyl-3H-imidazol-4-yl)-ethyl-2- {3- 3-ethyl-1- (3-hydroxy- propyl)-2-oxo-azepan-3-yl-phenoxy}-benzonitrile 4- l-amino-l- (3-methyl-3H-imidazol-4-y !)-ethyl-2- {3- 3-ethyl-l-propyl-2-oxo- azepan-3-yl-phenoxy}-benzonitrile 2- {3- 1-(2-amino-ethyl)-3-ethyl-2-oxo-azepan-3-yl-phenoxy}-4-1-ami no-1-(3- methyl-3H-imidazol-4-yl)-ethyl-benzonitrile {2- 3- (3- {5- 1-amino-1- (3-methyl-3H-imidazol-4-yl)-ethyl 2-cyano-phenoxy}- phenyl)-3-ethyl-2-oxo-azepan-l-yl-ethyl}-urea N- {2- 3- (3- {5- 1-amino-1- (3-methyl-3H-imidazol-4-yl)-ethyl 2-cyano-phenoxy}- phenyl)-3-ethyl-2-oxo-azepan-1-yl-ethyl}-N'-methyl urea 4- 1 (R)-amino-1- (3-methyl-3H-imidazol-4-yl)-ethyl-2- {3- 1-methyl-2-oxo-3 (R)- (3,3,3-trifluoro-propyl)-azepan-3-yl]-phenoxy}-benzonitrile 4- 1 (S)-amino-1- (3-methyl-3H-imidazol-4-yl)-ethyl-2- {3- 1-methyl-2-oxo-3 (S)- (3,3,3-trifluoro-propyl)-azepan-3-yl]-phenoxy}-benzonitrile 4- 1 (R)-amino-1- (3-methyl-3H-imidazol-4-yl)-ethyl-2- {3- 1-methyl-2-oxo-3 (S)- (3,3,3-trifluoro-propyl)-azepan-3-yl]-phenoxy}-benzonitrile 4- 1 (S)-amino-1- (3-methyl-3H-imidazol-4-yl)-ethyl-2- {3- 1-methyl-2-oxo-3 (R)- (3,3,3-trifluoro-propyl)-azepan-3-yl]-phenoxy}-benzonitrile 4- l-amino-l- (3-methyl-3H-imidazol-4-yl)-ethyl-2- {3- l- (2-dimethylamino-ethyl)-3- ethyl-2-oxo-azepan-3-yl-phenoxy}-benzonitrile

4- 1- (R or S) amino-1- (3-methyl-3H-imidazol-4-yl)-ethyl-2- {3- 1- (2-hydroxyethyl)- 3-ethyl-2-oxo-azepan-3-yl-phenoxy}-benzonitrile 4- 1-amino-1- (3-methyl-3H-imidazol-4-yl)-ethyl-2- {3- 1- (2-methoxyethyl)-3R-ethyl- 2-oxo-azepan-3-yl-phenoxy}-benzonitrile 4-Imidazol-1-ylmethyl-2- 3- (l-methyl-7-oxo-azepan-2-yl)-phenoxy-benzonitrile hydrochloride 4- 1-Amino-1- (3-methyl-3H-imidazol-4-yl)-ethyl-2- 3- (4-methyl-3-oxo-2- propyl-morpholin-2-yl)-phenoxy-benzonitrile 4- 1- amino-1- (3-methyl-3H-imidazol-4-yl)-ethyl-2- 3- (morpholine-4-sulfonyl)- phenoxy-benzonitrile 4- l-amino-1- (3-methyl-3 H-imidazol-4-yl)-ethyl-2- {3- 1- (2-methoxyethyl)-3R-ethyl- 2-oxo-azepan-3-yl-phenoxy}-benzonitrile; 2- 3- (3R-ethyl-l-methyl-2-oxo-azepan-3-yl)-phenoxy-4- 5- (2-morpholin-4-yl-ethyl)- imidazol-1-ylmethyl benzonitrile; or a pharmaceutically acceptable salt, an optical isomer or stereoisomer thereof.

The preferred compounds of the instant invention are: 2-{3-[1-methyl-2-oxo-3-(R or S)- (3,3,3-trifluoro-propyl)-azepan-3-yl-phenoxy}-4- l (R or S)-amino-l-(3-methyl-3H-imidazol-4-yl)-ethyl-benzonitrile ; 5 4-[1-amino-1-(3-methyl-3H-imidazol-4-yl)-ethyl]-2-{3-[1-(2-m ethoxyethyl)-3R-ethyl- 2-oxo-azepan-3-yl-phenoxy}-benzonitrile; 2- 3-3 (R or S)-benzyl-l-methyl-2-oxo-pyrrolidin-3-yl)-phenoxy-4- 1- (R or S) amino- 1- (3-methyl-3H-imidazol-4-yl)-ethyl-benzonitrile;

2- 3- (3-cycloproylmethyl-1-methyl-2-oxo-piperidin-3-yl)-phenoxy-4 - 1-amino-1- (3- methyl-3H-imidazol-4-yl)-ethyl-benzonitrile 2- 3- (3-ethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-4- l-amino-l- (3-methyl-3H- imidazol-4-yl)-ethyl-benzonitrile

2- 3- (3R-ethyl-l-methyl-2-oxo-azepan-3-yl)-phenoxy-4- 5- (2-morpholin-4-yl-ethyl)- imidazol-1-ylmethyl benzonitrile; (+) 2- 3- (3-ethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-4-imidazol-1-yl methyl- benzonitrile;

2- 3- (3-cyclopropylmethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-4-i midazol-1- ylmethyl-benzonitrile;

2- 3- (3-ethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-4- 2-methyl-5- (2-morpholin-4- yl-ethyl)-imidazol-1-ylmethyl-benzonitrile; 2- 3- (l-acetyl-3-ethyl-azepan-3-yl)-phenoxy-4-imidazol-l-ylmethyl -benzonitrile;

2- 3- (3-ethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-4- l-hydroxy-l- (3-methyl-3H- imidazol-4-yl)-ethyl-benzonitrile

2-[3-(3-ethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy]-4-[1-(3-m ethyl-3-H-imidazol-4- yl)-cyclopropyl-benzonitrile 2- (3- {4-cyano-3- 3- (3-ethyl-l-methyl-2-oxo-azepan-3-yl)-phenoxy-benzyl}-2- methyl-3H-imidazol-4-yl)-ethyl-urea

2- 3- (3-cyclopropylmethyl-l-methyl-2-oxo-azepan-3-yl)-phenoxy-4- 1-amino-1- (3- methyl-3H-imidazol-4-yl)-ethyl-benzonitrile 2- 3- (3-n-butyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-4- l-amino-l- (3-methyl-3H- imidazol-4-yl)-ethyl-benzonitrile

2-3-(3-ethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-4- 1-methylamino-I-(3-methyl- 3H-imidazol-4-yl)-ethyl]-benzonitrile

2- 3- (3-cyclopropylmethyl-1-methyl-2-oxo-pyrrolidin-3-yl)-phenoxy -4- 1-amino-I- (3-methyl-3H-imidazol-4-yl)-ethyl-benzonitrile

{2- 3- (3- {5- 1-amino-1- (3-methyl-3H-imidazol-4-yl)-ethyl 2-cyano-phenoxy}- phenyl)-3-ethyl-2-oxo-azepan-l-yl-ethyl}-urea or a pharmaceutically acceptable salt, an optical isomer or stereoisomer thereof.

The compounds of the present invention may have asymmetric centers and occur as racemates, racemic mixtures, and as individual diastereomers, with all possible isomers, including optical isomers, being included in the present invention.

When any variable, term or substituent (e. g. aryl, heterocycle, n, Rl a, etc.) occurs more than one time in any formula or generic structure, its definition on each occurrence is independent from the definition 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 1 to 6 carbon atoms, unless otherwise specified;"alkoxy"represents an alkyl group having 1 to 4 carbon atoms, unless otherwise indicated, attached through an oxygen bridge."Halogen"or "halo"as used herein means fluoro, chloro, bromo and iodo.

As used herein,"aryl"is intended to mean any stable monocyclic, bicyclic or tricyclic 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, indanonyl, biphenyl, tetralinyl, tetralonyl, fluorenonyl, phenanthryl, anthryl or acenaphthyl.

As used herein,"aralkyl"is intended to mean an aryl moiety, as defined above, attached through a C1-C6 alkyl linker, where alkyl is defined above.

Examples of aralkyls inlcude, but are not limited to, benzyl, naphthylmethyl and phenylpropyl.

As used herein,"heteroaralkyl"is intended to mean a heteroaryl moiety, as defined below, attached through a C I-C6 alkyl linker, where alkyl is defined above. Examples of heteroaralkyls include, but are not limited to, 2-pyridylmethyl, 2-morpholinylethyl, 2-imidazolylethyl, 2-quinolinylmethyl, 2-imidazolylmethyl and the like.

The term heterocycle or heterocyclic, as used herein, represents a stable 5-to 7-membered monocyclic or stable 8-to 11-membered bicyclic hetero- cyclic 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 hetero- cyclic rings is fused to a benzene ring. The heterocyclic ring may be attached at any heteroatom or carbon atom which results in the creation of a stable structure.

Examples of such heterocyclic elements include, but are not limited to, azepinyl, benzimidazolyl, benzisoxazolyl, benzofurazanyl, benzopyranyl, benzothiopyranyl, benzofuryl, benzothiazolyl, benzothienyl, benzoxazolyl, benzopyrazolyl, chromanyl, cinnolinyl, dibenzofuranyl, dihydrobenzofuryl, dihydrobenzothienyl, dihydrobenzothiopyranyl, dihydrobenzothiopyranyl sulfone, furyl, imidazolidinyl, imidazolinyl, imidazolyl, indolinyl, indolyl, isochromanyl, isoindolinyl, isoquinolinyl, isothiazolidinyl, isothiazolyl, isothiazolidinyl, morpholinyl, naphthyridinyl, oxadiazolyl, 2-oxoazepinyl, 2-oxopiperazinyl, 2-oxopiperdinyl, 2-oxopyrrolidinyl, 2-oxopyridyl, 2-oxoquinolinyl, piperidyl, piperazinyl, pyridyl, pyridinyl-N-oxide, pyrazinyl, pyrazolidinyl, pyrazolyl, pyridazinyl, pyrimidinyl, pyrrolidinyl, pyrrolyl, quinazolinyl, quinolinyl, quinoxalinyl, tetrahydrofuryl, tetrahydroimidazopyridinyl, tetrahydroisoquinolinyl, tetrahydroquinolinyl, thiamorpholinyl, thiamorpholinyl sulfoxide, thiazolyl, thiazolinyl, thienofuryl, thienothienyl, thienyl and triazolyl.

Examples of a"saturated heterocycle"may include, but are not limited to, morpholinyl, piperidinyl, piperazinyl, pyrrolidinyl, thiamorpholinyl, and the like.

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 heteroaryl 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, tetrahydroimidazopyridinyl, tetrahydroisoquinolinyl, tetrahydroquinolinyl, thiazolyl, thienofuryl, thienothienyl, and thienyl.

As used herein, the terms"substituted Cl-C6 alkyl"and"substituted C l-C6 alkoxy"are intended to include the branch or straight-chain alkyl group of the specified number of carbon atoms, wherein the carbon atoms may be substituted with F, Cl, Br, CF3, N3, N02, NH2, oxo,-OH,-O (C1-C6 alkyl), S (0) 0 2, (Cl-C6 alkyl) S (0) 0-2-, (Cl-C6 alkyl) S (0) o 2 (C1-C6 alkyl)-, C3-C1o cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl,-C (O) NH, (C1-C6 alkyl) C (O) NH-, H2NC (O) NH-, H2NC (NH)-, (Cl-C6 alkyl) C (O)-,-O (C1-C6 alkyl) CF3, (C1-C6 alkyl) OC (O)-, (Cl-C6 alkyl) O (C1-C6 alkyl)-, (C1-C6 alkyl) C (0) 2 (Cl-C6 alkyl)-, (Cl-C6 alkyl) OC (O) NH-, aryl, benzyl, heterocycle, aralkyl, heteroaralkyl, halo-aryl, halo-benzyl, halo-heterocycle, cyano-aryl, cyano-benzyl and cyano-heterocycle.

As used herein, the terms"substituted aryl","substituted heteroaryl", "substituted aralkyl", substituted heteroaralkyl","substituted benzyl"and"substituted hetrocycle"are intended to include the cyclic group containing from 1 to 3 substitutents in addition to the point of attachment to the rest of the compound. Such substitutents are preferably selected from the group which includes but is not limited to F, Cl, Br, CF3, NH2, N (Cl-C6 alkyl) 2, N02, CN, N3, Cl-C20 alkyl, Cl-C6 alkoxy,-OH,-O (C 1-C (alkyl), S (0) 0-2, (Cl-C6 alkyl) S (0) o-2-, (Cl-C6 alkyl) S (0) o 2 (CI-C6 alkyl)-, (Cl-c6 alkyl) C (O) NH-, H2NC (O) NH-, H2N-C (NH)-, (C1-C6 alkyl) C (O)-, (C1-C6 alkyl) OC (O)-, (C1-C6 alkyl)O(C1-C6 alkyl)-, (C1-C6 alkyl) C (0) 2 (C1-C6 alkyl)-, (C1-C6 alkyl) OC (O) NH-, aryl, aralkyl, heteroaryl, heteroaralkyl, halo-aryl, halo-aralkyl, halo-heterocycle, halo-heteroaralkyl, cyano-aryl, cyano-aralkyl, cyano-heterocycle and cyano-heteroaralkyl.

Examples of the moiety include, but are not limited to,

Examples of the moiety include, but are not limited to, As used herein for substituent A3, examples of"C3-C6 cycloalkyl"include, but are not limited to:

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 atoms or heteroatoms.

Preferably, Rlc is independently selected from: hydrogen, unsubstituted or substituted C1-C6 alkyl, unsubstituted or substituted aryl, unsubstituted or substituted heterocycle,-OR15, or-N (R15) 2.

Preferably, R1c is selected from N (R15) 2 or OR15.

Preferably, Rl is independently selected from H, halo, unsubstituted or substituted C 1-C6 alkyl,-OR8,-N (R8) 2,- (C1-C6 alkyl) N (R8) 2, unsubstituted or substituted aryl, unsubstituted or substituted heterocycle,- (Cl-C6 alkyl) NR8C (O) N (R8) 2, or (Ci-C6 alkyl) R14. Preferably, RI is not Cl-C6 alkyl substituted with unsubstituted or substituted aryl.

Preferably, R2 is independently selected from H, OR8, CN, unsubstituted or substituted aryl or halogen. Most preferably, R2 is CN.

Preferably, R3 is independently selected from hydrogen, halogen, CN, N02, and unsubstituted or substituted Cl-C6 alkyl.

Preferably, R4 of formulae A-D is selected from H or =0.

Preferably, R5 is independently selected from hydrogen, halogen, unsubstituted or substituted C1-C6 alkyl, unsubstituted or substituted aryl, unsubstituted or substituted heterocycle,- (C 1-C6 alkyl) N (R8) 2, C (O) N (RS) 2 or - (Ci-C6 alkyl) OR8.

Preferably, A1 and A2 are independently selected from: a bond, O, -NR8, C (O) and S (O) q. Most preferably, Al is 0,-NR8, C (O) or S (O) q and A2 is a bond.

Preferably, A3 is selected from a bond, or C3-C6 cycloalKyl.

Preferably, M of formulae 1-111 is selected from CH2 or NR8.

Preferably, Q of formulae I-III is selected from C (=O), CH2, or S (O) m.

Preferably, W is a heterocycle. Most preferably, W is imidazolyl, pyridyl.

Preferably, Y is selected from phenyl or pyridyl. Most preferably, Y is phenyl.

Preferably, Z is selected from aryl, a bond or heteroaryl. Most preferably, Z is phenyl or pyridyl.

Preferably, m, n, p, q, r and s are independently 0,1, or 2.

Preferably, t is 1,2 or 3. Most preferably, t is 2.

Preferably, x of formulae I-III is selected from 0,1 or 2.

Preferably, (C (Rla) 2) nAl (C (Rla) 2) n represents 0,-NR8, C (O), S (O) q or C1-C6 alkyl. More preferably, (C (Rla) 2) nAl (C (Rla) 2) n represents 0,-NR8, C (O) or S (O) q.

Preferably, (C (Rlb) 2) rA2 (C (Rlb) 2) r represents a bond or C1-C6 alkyl.

More preferably, (C (Rlb) 2) rA2 (C (Rlb) 2) r represents a bond.

Preferably, (CH2) nA1 (CH2) n represents 0,-NR8, C (O), S (O) q or C 1-C6 alkyl. More preferably, (CH2) nA1 (CH2) n represents O, -NR8, C (O) or S (O) q.

Preferably, (CH2) rA2 (CH2) r represents a bond or Cl-C6 alkyl. More preferably, (CH2) rA2 (CH2) r represents a bond.

Preferably, the ring system is attached to the rest of the molecule through a non-nitrogen ring atom.

Preferably, the moiety is selected from:

It is intended that the definition of any substituent or variable (e. g., RI a, R3, in, p, etc.) at a particular location in a molecule is independent of its definitions elsewhere in that molecule. Thus,-C (Rla) 2 represents-CH2,-CHCH3, -CHC2H5, 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.

Abbreviations which may be used in the description of the chemistry and in the Examples that follow include: Ac20 Acetic anhydride; AIBN 2,2'-azobisisobutyronitrile Boc t-Butoxycarbonyl; CBz Carbobenzyloxy; DBU 1,8-diazabicyclo [5.4.0] undec-7-ene; DMAP 4-Dimethylaminopyridine; DME 1,2-Dimethoxyethane; DMF Dimethylformamide; DMSO Methyl sulfoxide; DTT Dithiothreitol; EDC 1- (3-dimethylaminopropyl)-3-ethyl-carbodiimide- hydrochloride; EDTA Ethylenediaminetetraacetic acid; Et3N Triethylamine; EtOAc Ethyl acetate; EtOH Ethanol; FAB Fast atom bombardment; HEPES 4- (2-hydroxyethyl)-1-piperazineethanesulfonic acid; HOBT 1-Hydroxybenzotriazole hydrate; HOOBT 3-Hydroxy-1,2,2-benzotriazin-4 (3H)-one; HPLC High-performance liquid chromatography; MCPBA m-Chloroperoxybenzoic acid; MeOH methanol; MsCI Methanesulfonyl chloride;

NaHMDS Sodium bis (trimethylsilyl) amide; NBS N-bromosuccinimide; PMSF a toluenesulfonyl chloride; Py or pyr Pyridine; RPLC Reverse Phase Liquid Chromatography; TFA Trifluoroacetic acid; THF Tetrahydrofuran.

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 Schemes 1 to 18 describe the synthesis of compounds of formula A.

The starting materials can be obtained from commercial sources or they can be obtained using standard transformations (e. g. esterification of the hydroxy acid) from commercially available materials.

In Scheme 1, amino-hydroxybenzoates of type 1 can be converted to the corresponding iodide 2 by treatment with acidic aqueous NaN02 followed by the addition of KI. The phenol may then be alkylated by treatment with a base such as NaH or Cs2C03 in an organic solvent (for example DMF) followed by the addition of an electrophile to yield 3. Reduction of the ester of 3 using, for example, LiBH4 in THF then yields the alcohol 4 which can in turn be treated with Zn (CN) 2 in DMF and a palladium catalyst to give 5. The alcohol of 5 can be converted into a leaving group of 6 in a number of ways. One such procedure involves reaction of the alcohol with a sulfonyl chloride in the presence of an organic base (e. g. triethylamine) in an organic solvent such as dichloromethane. A second method requires the reaction of the alcohol with CBr4 and a phosphine such as triphenyl phosphine in an organic solvent such as dichloromethane. A third method involves reaction of the alcohol with N- bromosuccinimide and dimethyl sulfide in dichloromethane. The reaction of 6 with imidazole in a polar solvent such as DMF then affords compounds of formula 7.

In addition, 6 upon reaction with 4-iodo-1-tritylimidazole in THF with 1,2- dibromoethane, Zn and NiCl2 (PPh3) 2 gives a compound of formula 8. Subsequent methanolysis of 8 yields compounds of formula 9. Treating a compound of formula

8 with a suitably substituted alkyl or aralkyl halide or mesylate, followed by methanolysis yields a compound of formula 10.

An alternative route for the synthesis of compounds of formula 7 is shown in Scheme 2. Iodo-hydroxybenzoic acids of structure 2 may be converted to the corresponding cyano 11. The ester may be reduced by treating 11 with LiBH4 (and THF) to produce the alcohol 12. The primary hydroxyl can be converted into a leaving group by reacting the alcohol with CBr4 and a phosphine, such as triphenyl phosphine (in an organic solvent such as dichloromethane). The reaction of 13 with imidazole affords 14, which can be converted to 7 by treatment with a base, such as Cs2CO3, and a suitably substituted alkyl or aralkyl halide or mesylate.

Scheme 3 illustrates one route for the synthesis of compounds of formula 21. The bromotoluene 15 can be treated with KMn04 to yield the bromo- fluorobenzoic acid 16. The acid 16 can be reduced with BH3 in THF to give the bromobenzene 17. The bromobenzene 17 can be converted to the corresponding cyanobenzene 18 by treating 17 with Zn (CN) 2 and a palladium catalyst. The alcohol of intermediate 18 can be converted into a leaving group by reacting the alcohol with NBS and DMS to produce 19. Treatment of 19 with a nitrogen-containing hetero- cycle yields 20. Compounds of formula 21 can be obtained by treating 20 with Cs2C03 or KF/Alumina and a phenol.

Scheme 4 describes one route for preparing compounds of formulae 27 and 28. Compound 18 is oxidized with pyridine-sulfoxide complex, for example, to aldehyde 22. Treatment of compound 22 with a Grignard reagent RCMgBr provides compound 23. Oxidation of 23 gives ketone 24 which is treated with 4-iodo-1- tritylimidazole and EtMgBr to obtain alcohol 25. Subsequent protection of alcohol 25, quaternization, and removal of the protecting groups provides compound 26.

Compound 26 is converted to compounds of formula 27 by treatment with RbOH and a base such as Cs2CO3 or KF on alumina. Compounds of formula 27 are converted to compounds of formula 28 by treatment with SOC12 followed by ammonia.

Scheme 4A depicts the synthesis of compounds of formula 29, where the nitrogen atom attached to benzylic carbon is substituted, using sulfonyl chloride and a substituted amine to convert compound 27 to compound 29.

Scheme 5 illustrates an alternate way of preparing intermediate 25.

The aldehyde 22 is treated with 4-iodo-1-tritylimidazole and EtMgBr to obtain the

alcohol 30. The alcohol 30 was then converted to the ketone 31 using MnO2.

Intermediate 25 was obtained by treating the ketone 31 with a substituted MgBr.

Scheme 6 illustrates several routes using compounds of formula 32 to prepare various amide, urea, carbamate and sulfonamide derivatives (33-36).

Scheme 7 describes a route for the chiral preparation of compounds of formula 40. Using the commercially available t-butyl sulfinamide and techniques described in the literature (J. A. Ellman et al., J. Org. Chem., 1999, vol. 64, p. 1278- 1284), intermediates of formula 39 are prepared as single enantiomers. Treating 39 with an alcohol and cesium carbonate or KF/alumina yields compounds of formula 40 as single enantiomers.

Another route for the chiral synthesis of intermediate 39, which uses a protected imidazolyl group, is described in Scheme 8.

Schemes 9 and 10 illustrate alternate ways of preparing compounds of formula 37. Scheme 9 uses techniques known in the literature (P. Molina et al., Tetrahedron Letters, vol. 37, N. 52, p. 9353-9356) to prepare intermediate 45.

Scheme 10 illustrates an alternative route to compounds of formula 37.

Schemes 11 and 12 illustrate the synthesis of compounds of formula 49 and 50, using techniques previously described above.

Schemes 13 and 14 illustrate syntheses of compounds of the formulae 53,55, and 56 wherein substituent W is present as a pyridyl moiety. Similar synthetic strategies for preparing compounds that incorporate other heterocyclic moieties for variable W are also well known in the art.

Schemes 15-17 depict the preparation of various lactam phenols using an allyl compound 58 as a common intermediate. In Scheme 15 compound 58 is hydrogenated and deprotected to give compound 60. In Scheme 16 compound 58 is hydroborated to provide compound 61. Ozonolysis and reduction of 58 yields compound 62. Deprotection of 59 and 61 give 63 which can be further elaborated to the amino derivative 65. Scheme 17 illustrates modifications of the amino compound 65 by reductive alkylation, acylation, sulfonylation or carbamylation to provide compounds 66,67,68, and 69, respectively.

Scheme 18 illustrates a method to prepare sultam phenols of formula 75. By treating compound 70 with MgS02 in THF and sulfonyl chloride, compound 71 is obtained. Compound 71 is converted to the substituted sulfonamide 72, which

is then treated with NaH to obtain the compounds of formula 73. Compound 73 is treated with a suitably substituted halide and base to provide intermediate 74.

Intermediate 74 is deprotected to give a sultam phenol of formula 75.

SCHEME 1 CO2R CO2R NaNO2, aq. HCI rua f'Il THF, H20 a Ra OH R OH then KI 2 RbBr, DMF OH ccR r i JL, Zn (CN) 2 a f'Il b LiBH4 a b Pd (Ph3P) 4 3 y THF 4 DMF OH Br 4 Ph3P/ . OH Br Ra-OR R OR T F THF 5 CN 6 CN imidazole DMF Zn, NiC12 (Ph3P) 2 N4R9 I R9 BrCH2CH2Br TH F /N =, Tr RaRb\N J CN Ra <_ORb R or 8 CN 8 CN SCHEME (CONT'D.) dR < , Tr N N N R I Ra b 2) methanol 8 CN 10 CN \ halo, mesylate Methanol 7N fRa-I OR b 9 CN SCHEME 2 C02R C02R //LiBH4 a Ra I OH Ra OH LiBH4 2 11 CN Rg . OHBr N fr OH OH M /ph3p/N "YCBr"\ CBr4 \J 12 CN 13 CN N4R9 N N CS2CO3 a Rbx l, I OR' 14 CN X = halo, OMs 7 CN SCHEME 3 CH3 02H H /KMn04/gH3 Zn (CN) 2 a f R \ I F > Ra \ I > R I > THF Fd cat. Br Br Br 15 16 17 Het 0 Most Heu /PBS Nos DOMS _F wF CN 19 20 20 Het 1) Cs2CO3 N--- or g KF/Alumina v Ra n 2) RbOH ORb CN

SCHEME 4 H H Rc H H f 1 RCMgBr 1 pyr-S03 Ra-R R F F F CN CN CN 22 23 18 tu Ruz N ^ NTr /= J N OH Mono2 Rg Rc R jazz CN EtMgBr 24 T CN 25 Tr I NRg Rg (NI, Rg OU N O N > Ra/1) RdX > Rd Rc Ra-I-A.- 3) OH- CN ! F 26 CN Rg CS2CO3 0 or Ra R > roc d roc KF/alumina Ra- RbOH \ ORb R CON \ ORS CN! 27 28 CN SCHEME 4A SCHEME 5 SCHEME 6 SCHEME 7 rn 0 N Rg O S 4 $\F O 1) RMgX au \ I F Ra (2) HCI F CN 37 CN 38 R9 R9 N% N/ NH2 RbOH'NH2 N N /CS2CO3/ Ra- (01'Ra- I F KF/alumina < OR CN CN 39 40 SCHEME 8 SCHEME 9 SCHEME 10

SCHEME 11 Het/ N Cl Ra-I CON CN RUS Cs CO l con 20 (§s KF/Alumina Q, wM lu HO x R6 s Het (R) s N' N---N) t Q.. M Q, Rai y lX Rus o \/ CN 49 SCHEME 12 9 4 N-J/OH /-RC R X Ra f/ìl R CN ce R I (R5 ) s CS2CO3) t USUUg < ) or ort~t) tKF/Alumina Q. M Xi"\ N \ ou l art RUZ R Rc t /y Q,"r-M rus CN 50 SCHEME 13 Rg Rg H O/ BrMg-OH OH F Ras /F > Ra.. \ CN 22 CN 1) SOC'2 51 2) Zn/HOAc Ru ru or 1) PhOCSCI N-1 DMAP Ra2) nBu3SnH Ra- F 52 CN RbOH Cs2CO3 or KF/alumina Rg v \ OR' CN 53 SCHEME 14 Ru ru OH 1) Mn02 R_ RbOH y N 2) R M Br, OH Cs2C03 N) 9 KF/alumina RS(XF RSfXFCN 54 CN 51 \F/F CN CN 51 Ru ru 1) SOC12 OH NHR -/ R c R c Roc 2) ReNH2 CN CN 55 56 SCHEME 15 SCHEME 16 SCHEME 17 SCHEME 18 1) MgS02, THF r> RO/Br2) SOCI2 RO SOCI2 ru RO n R6NH X I SONR6 ru na x RS R6/ /n Nk 72 t ( X R6 Base O s, Nn 2 73 >/ R O/ 74 73 R6 BBr3 0 soNß > H O<>) tH rus 75

In the above Schemes, it is understood that R independently represents an alkyl or an aryl; Ra independently represents R2 or protected precursors thereof ; Rb independently represents the following moiety:

Rc independently represents R I C or protected precursors thereof ; Rd and R independently represents Rl or protected precursors thereof ; Re and Rf independently represent R8 or protected precursors thereof ; and

X is independently represents a halide.

In order to simplify the structures described in the above schemes, mutiple designations of a substituent (i. e. Ra, Rb, RC, Rd, Re, Rf and Rg) have not been included. However, it is understood that there may be several, independently selected substitutients around each of the rings described hereinabove, as seen in formulae I-III and A-D, hereinabove.

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 60 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 61. 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 farnesyl-protein transferase is further characterized by: b) 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 IC50 for the inhibition of the farnesylation of hDJ protein.

When measuring such IC50s the assays described in Examples 65 and 66 may be utilized.

It is also preferred that the selective inhibitor of farnesyl-protein transferase is further characterized by: c) 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 ICso 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: d) 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 64 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 Example 64, it is preferred that the dual inhibitor compound has an in vitro inhibitory activity (IC50) that is less than about 12mM against K4B-Ras dependent activation of MAP kinases in cells.

The Class II prenyl-protein transferase inhibitor may also be characterized by: a) an ICso (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 ICso for inhibiting the farnesylation 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 II 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 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 ICso (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 II 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 ICso (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 64.

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 farnesylation 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, a component of NF-1 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 medicine, 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, the prenyl-protein transferase inhibitor 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. Ophthalmol. 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.

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 polyethyl- eneglycol 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, flavouring agents, preservatives and antioxidants.

Syrups and elixirs may be formulated with sweetening agents, for example glycerol, propylene glycol, sorbitol or sucrose. Such formulations may also contain a demulcent, a preservative, flavoring and coloring agents and antioxidant.

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

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

The injectable solutions or microemulsions may be introduced into a patient's blood-stream by local bolus injection. Alternatively, it may be advantageous to administer the solution or microemulsion in such a way as to maintain a constant circulating concentration of the instant compound. In order to maintain such a constant concentration, a continuous intravenous delivery device may be utilized.

An example of such a device is the Deltec CADD-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 transdermal skin patches well known to those of ordinary skill in the art. To be administered in the form of a transdermal delivery system, the dosage administration will, of course, be continuous rather than intermittent throughout the dosage regimen. 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 useful- ness against the condition that is being treated. For example, the compounds of the instant invention may also be co-administered with other well known cancer therapeutic agents that are selected for their particular usefulness against the condition that is being treated. Included in such combinations of therapeutic agents are combin- ations of the instant farnesyl-protein transferase inhibitors and an antineoplastic agent.

It is also understood that such a combination of antineoplastic agent and inhibitor of farnesyl-protein transferase may be used in conjunction with other methods of treating cancer and/or tumors, including radiation therapy and surgery.

Examples of an antineoplastic agent include, in general, microtubule- stabilizing agents (such as paclitaxel (also known as Taxol@), docetaxel (also known as Taxotere (W), epothilone A, epothilone B, desoxyepothilone A, desoxyepothilone B or their derivatives); microtubule-disruptor agents ; alkylating agents, anti-metabolites; epidophyllotoxin; an antineoplastic enzyme; a topoisomerase inhibitor; procarbazine; mitoxantrone; platinum coordination complexes; biological response modifiers and growth inhibitors; hormonal/anti-hormonal therapeutic agents and haematopoietic growth factors.

Example classes of antineoplastic agents include, for example, the anthracycline family of drugs, the vinca drugs, the mitomycins, the bleomycins, the cytotoxic nucleosides, the taxanes, the epothilones, discodermolide, the pteridine family of drugs, diynenes and the podophyllotoxins. Particularly useful members of those classes include, for example, doxorubicin, carminomycin, daunorubicin, aminopterin, methotrexate, methopterin, dichloro-methotrexate, mitomycin C, porfiromycin, herceptin, 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, camptothecin, CPT-11, topotecan, ara-C, bicalutamide, flutamide, leuprolide, pyridobenzoindole derivatives, interferons and interleukins.

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

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 farnesyl-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 farnesyl-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, which is incorporated herein by reference.

As used herein the term an integrin antagonist refers to compounds which selectively antagonize, inhibit or counteract binding of a physiological ligand to an integrin (s) that is involved in the regulation of angiogenisis, or in the growth and invasiveness of tumor cells. In particular, the term refers to compounds which selectively antagonize, inhibit or counteract binding of a physiological ligand to the avp3 integrin, which selectively antagonize, inhibit or counteract binding of a physiological ligand to the αvß5 integrin, which antagonize, inhibit or counteract binding of a physiological ligand to both the avß3 integrin and the avß5 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 alßl, a2ßl, a5ßl, a6ßl and a6ß4 integrins. The term also refers to antagonists of any combination of avp3 integrin, avß5, al ß 1, a2ß 1, a5ß 1, a6ß 1 and a6ß4 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 (t) ; 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", Chemistry & 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 II. HO p HO COOH O OH Lactone Open-Acid ICI In HMG-CoA reductase inhibitor's 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 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 4-imidazol-1-ylmethyl-2- 2- (2-oxo-piperidin-1-yl)-phenoxy- benzonitrile 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-H20 bath and acidified slowly with 6N HCl 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. lH NMR (CDC13) 5 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-fluorobenzyl alcohol 4-Bromo-3-fluorobenzoic acid, as described above, (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 (Na2SO4), filtered, and concentrated to give the title compound as an oil which solidified on standing.

1H NMR (CDCl3) b 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-hydroxymethylbenzonitrile 4-Bromo-3-fluorobenzyl alcohol, as described in Step B above, (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 (PPh2) 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 (Na2SO4). Filtration and concentration

to dryness gave the title compound as a white solid after chromatography (silica gel, hexane: EtOAc, 6.5: 3.5). lH NMR (CDC13) 8 7.61 (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 4-Bromomethyl-2-fluoro-benzonitrile N-Bromosuccinimide (6.6 g, 0.037 mol) was dissolved in CH2C12 (150 mL), cooled to 0°C and treated with dimethylsulfide (3.27 mL, 0.0446 mol).

The solution was cooled to-20°C then treated dropwise with a solution of 2-fluoro- 4-hydroxymethylbenzonitrile, as described in Step C above, (3.74 g, 0.0248 mol) in CH2C12 (30 mL). After the addition, the reaction mixture was stirred at 0°C for 2 h then left to warm to ambient temperature overnight. The reaction mixture was added to ice/H20, extracted with EtOAc, the organic layer separated, washed with brine and dried (MgS04). Filtration and concentration to dryness gave the title compound which was purified after chromatography (silica gel, 5-10% EtOAc/hexane).

1H NMR (CDC13) 5 7.61 (dd, 1H, J = 8,8 Hz), 7.26-7.30 (m, 2H), 4.45 (s, 2H).

Step E: Preparation of 2-fluoro-4-imidazol-1-ylmethyl-benzonitrile 4-Bromomethyl-2-fluoro-benzonitrile, as described in Step D above, (3.44g, 16.0 mmol) and imidazole (5.47 g, 80.3 mmol) were dissolved in DMF (40 mL) and stirred at ambient temperature for 2 h. The DMF was removed in vacuo and the residue was partitioned between EtOAc (300 mL) and aqueous saturated NaHC03 solution. The organic layer was separated, washed with NaHC03 solution, H20, brine, and dried (MgS04). Filtration and concentration to dryness gave the title compound after chromatography (silica gel, 1-2% CH30H/CH2CI2).

1H NMR (CDC13) 8 7.62 (dd, 1H, J = 8.5,9.5 Hz), 7.57 (s, 1H), 7.16 (s, 1H), 7.00 (d, 1H, J = 8.5 Hz), 6.94 (d, 1H, J = 9.5 Hz), 6.91 (s, 1H), 5.21 (s, 2H).

Step F : Preparation of 2- (2-oxo-piperidin-1-yl)-phenol To a solution of 2-aminophenol (1.09 g, 0.01 mol), Et3N (4.46 mL, 0.032 mol) and 4-dimethylaminopyridine (0.122 g, 0.001 mol) in CHC13 (20 mL) in an ice-H20 bath was added dropwise 5-bromovaleryl chloride (2.95 mL, 0.022 mol) with stirring. After 2 hr, the reaction mixture was washed with IN HC1 until the aqueous layer was acidic, then washed with H20, aqueous saturated NaHC03

solution, brine, and dried (Na2SO4). Filtration and concentration to dryness gave a yellow oil which solidifed on standing. This bisacylated product was dissolved in DMF (20 mL) and heated at 80°C with cesium carbonate (4.89 g, 0.015 mol) for 3 hr, then partitioned between EtOAc and ice water. The aqueous layer was extracted with EtOAc (3 x), the organics combined, washed with H20, aqueous saturated NaHC03 solution, brine, and dried (Na2SO4). Filtration and concentration to dryness gave a crude product which was treated with 1 N NaOH solution (12 mL, 0.012 mol) in THF (20 mL)-H20 (10 mL) with stirring at ambient temperature for 2 hr. The reaction mixture was neutralized with IN HCI (12 mL, 0.012 mol), concentrated, and extracted with EtOAc (3x), the organics combined, washed with H20, aqueous saturated NaHC03 solution, brine, and dried (Na2SO4). Filtration and concentration to dryness gave the title compound.

Step G : Preparation of 4-imidazol-1-ylmethyl-2-2-(2-oxo-piperidin- 1-yl)-phenoxyl-benzonitrile 2-Fluoro-4-imidazol-1-ylmethyl-benzonitrile (as described in Step E above) (0.080 g, 0.4 mmol), 2- (2-oxo-piperidin-1-yl)-phenol (as described in Step F above) (0.091 g, 0.5 mmol) and cesium carbonate (0.261 g, 0.8 mmol) were combined in DMF (2.0 mL) and heated at 50°C for 18 hr. The reaction mixture was partitioned between EtOAc and a minimum volume of H2O. Additional product was salted out from the aqueous layer with solid NaCl, and extracted into EtOAc. The organic layers were combined, dried (Na2SO4), filtered and concentrated to give the title compound after RP HPLC on a Waters Prep Pak column eluting with a 0.1 % TFA/H20: 0.1% TFA/CH3CN gradient followed by conversion to the free base.

FAB MS 373 (M+1).

Analysis calculated for C22H20N402'0.45 H20: C, 69.43; H, 5.54; N, 14.72; Found: C, 69.41; H, 5.46; N, 14.67.

Using the methods described above, but substituting the requisite phenol for 2- (2-oxo- piperidin-1-yl)-phenol in Step G, the following compounds were prepared: 4-imidazol-1-ylmethyl-2-!-benzonitrile FAB MS 361 (M+1)

4-imidazol-1-ylmethyl-2- (3-piperidin-1-vlmethyl-phenoxy-benzonitrile FAB MS 373 (M+1) 4-imidazol-1-ylmethyl-2- 3- (2-oxo-azepan-1-yl)-phenoxy-benzonitrile Analysis calculated for C23H22N402'0.30 C2H50H'0.50 HCI: C, 62.30; H, 5.61; N, 12.32; Found: C, 62.32; H, 5.82; N, 12.35.

Using the methods described above, but substituting the requisite phenol for 2- (2-oxo- piperidin-1-yl)-phenol in Step G, the following compounds are prepared: 4-imidazol-1-ylmethyl-2-[3-(2-oxo-piperidin-1-yl)-phenoxy]-b enzonitrile 4-imidazol-1-ylmethyl-2-[4-(2-oxo-piperidin-1-yl)-phenoxvl-b enzonitrile EXAMPLE 2 Preparation of 4-imidazol-1-ylmethyl-2- 2- (3-methyl-2-oxo-piperidin-1-yl)-phenoxy- benzonitrile Step A: Preparation of2- (3-methvl-2-oxo-piperidin-1-yl)-phenol Lithium diethylamide (2M solution in THF) (3.92 mL, 7.84 mmol) was added to a solution of 2- (2-oxo-piperidin-1-yl)-phenol (as described in Example 1, Step F) (0.50 g, 2.61 mmol) in THF (5 mL) at-78°C with stirring under Ar. After 30 min, iodomethane (0.488 mL, 7.84 mmol) was added and the reaction left to come to room temperature overnight. The reaction was treated with H20, concentrated to remove the THF, then partitioned between diethyl ether and H20. The organic layer was separated, washed with brine, dried (Na2SO4), filtered, and concentrated to dryness to give the title compound after chromatography (silica gel, 1% CH30H, 29% EtOAc, 70% hexane).

1H NMR (CDC13) 6 7.14-7.23 (m, 3H), 7.52 (dd, 1H, J = 1.5,8 Hz), 6.93-6.98 (m, 1H), 3.70-3.85 (m, 2H), 2.63-2.72 (m, 1H), 2.08-2.17 (m, 1H), 1.89-2.05 (m, 2H), 1.60-1.70 (m, 1H), 1.36 (d, 3H, J = 7 Hz).

Step B : Preparation of 4-imidazol-1-ylmethyl-2- 2- (3-methyl-2-oxo- piperidin-1-yl)-phenoxyl-benzonitrile Following the procedure outlined in Example 1, Step G, but substituting the phenol of Step A for the phenol used in Example 1, Step G, the title compound was prepared.

FAB MS 387 (M+1).

Analysis calculated for C25H26N402 1. 85 HC1 @ 0.35 Et20: C, 61.07; H, 5.75; N, 11.66; Found: C, 60.98; H, 5.93; N, 11.68.

Using the procedures outlined above, but resubmitting 2- (3-methyl-2-oxo-piperidin-1- yl)-phenol to the conditions of Step A to give 2- (3, 3-dimethyl-2-oxo-piperidin-1-yl)- phenol, the following compound was prepared: 2- 2- (3, 3-dimethyl-2-oxo-piperidin-1-yl)-phenoxy-4-imidazol-1-ylmeth yl- benzonitrile FAB MS 401 (M+1).

Analysis calculated for C24H24N402'1.65 HCl-0.70 Et20: C, 62.80; H, 6.42; N, 10.93; Found: C, 62.84; H, 6.10; N, 10.94.

EXAMPLE 3 Preparation of 2- 3- (3-ethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-4-imidazol-1- ylmethyl-benzonitrile hydrochloride 2-Fluoro-4-imidazol-1-ylmethyl-benzonitrile (as described in Example 1, Step E) (0.045 g, 0.222 mmol), 3-ethyl-3- (3-hydroxy-phenyl)-l-methyl-azepan-2- one, commercially available from Maybridge, (0.055 g, 0.222 mmol), KFAl203 (0.136 g) and 18-Crown-6 (0.006 g, 0.023 mmol) were refluxed in CH3CN (2.5 mL) under Ar for 48 hr. The solution was filtered, concentrated in vacuo, and purified using reverse phase chromatography (95/5-5/95 H20/CH3CN with 0.1% TFA, flow = 65 mL/min). The compound was converted to the free base using satd. NaHC03 solution, extracting with CH2C12 (3x), drying (MgS04), filtering and treating with IN HCl ethereal solution to give the title compound.

FAB MS (M+1) = 429 Analysis calculated for C26H28N402'1.05 HCl#0. 05 H20: C, 66.76; H, 6.28; N, 11.98; Found: C, 66.80; H, 6.17; N, 11.69.

Chromatography of racemic 2- 3- (3-ethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy- 4-imidazol-1-ylmethyl-benzonitrile by normal phase LC on a chirasil AD column provided both enantiomer A: FAB MS (M+1) = 429 Analysis calculated for C26H28N402'1.50 HCl-1.25 H20: C, 61.74; H, 6.38; N, 11.08; Found: C, 61.77; H, 6.39; N, 11.25. and enantiomer B: FAB MS (M+1) = 429 Analysis calculated for C26H28N402'1.00 HCl#1.95 H2O : C, 62.44; H, 6.63; N, 11.20; Found: C, 62.43; H, 6.70; N, 11.46.

Using the procedure outlined above, but substituting the appropriate commercially available phenols for the 3-ethyl-3- (3-hydroxy-phenyl)-l-methyl-azepan-2-one, the following compounds were prepared: 4-Imidazol-1-ylmethyl-2-[2-(2-oxo-pyrrolidin-1-yl)-phenoxy]- benzonitrile FAB MS (M+1) = 359 Analysis calculated for C2lHlgN402-1.00 HCl#1. 00 H20: C, 61.08; H, 5.13; N, 13.57; Found: C, 61.05; H, 5. 23; N, 13.28.

Using the procedures above but substituting the appropriate nitrogen-containing heterocycle for imidazole in Example 1, Step E, the following compounds were prepared: 2-[3-(3-ethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy]-4-(2-meth yl-imidazol- 1-yl ! methyl-benzonitrile

FAB MS (M+1) = 443 2- 3- (3-ethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-4- (5-methyl-imidazol- 1-yl)methyl-benzonitrile FAB MS (M+1) = 443 2-[3-(3-ethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy]-4-(2,5-di methyl-imidazol- 1-yl) methyl-benzonitrile FAB MS (M+1) = 457 2-[3-(3-ethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy]-4-[1, 2,4] triazol-4- vlmethyl-benzonitrile FAB MS (M+1) = 430 2-[3-(3-ethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy]-4-[1, 2,4] triazol-1- vlmethyl-benzonitrile FAB MS (M+1) = 430 EXAMPLE 4 Preparation of 4-imidazol-1-ylmethyl-2- 3- ( 1-methyl-2-oxo-azepan-3-yl)- phenoxy-benzonitrile hydrochloride Step : Preparation of 3- (3-methoxy-phenyl)-l-methyl-azepan-2-one To 1.6 M n-BuLi in hexane (31.3 mL, 50 mmol) at 0°C was added a solution of 2,2,6,6-tetramethyl piperidine (8.43 mL, 50 mmol) in anh. THF (100 mL) over 5 min. N-methylcaprolactam (6.41 mL, 50 mmol) in anhydrous THF (20 mL) was added. After stirring the reaction mixture for 10 min, a further portion of 1.6M n-BuLi in hexane (31.3 mL, 50 mmol) was added, the mixture was stirred for 10 min, and 2-chloroanisole (6.34 mL, 50 mmol) was added. After stirring for 30 min under Ar, the reaction was quenched with H20, stirred for 30 min, concentrated in vacuo, diluted with EtOAc and washed with 5N HCI (100 mL) and brine. The organic layer was dried (MgS04), concentrated and purified using Si02 chromatography (10% EtOAc/CH2Cl2) to give the title compound.

Step B : Preparation of 3- (3-hydroxv-phenyl)-1-methyl-azepan-2-one 3- (3-Methoxy-phenyl)-1-methyl-azepan-2-one (as described in Step A above) (1.067 g, 4.58 mmol) was refluxed in 48% HBr (5 mL) for Ih. The reaction was diluted with EtOAc, washed with H20, brine, dried (MgS04), and concentrated to give the title compound.

Step C : Preparation of 4-imidazol-1-ylmethyl-2- 3- (1-methyl-2-oxo-azepan- 3-yl)-phenoxy-benzonitrile hydrochloride 2-Fluoro-4-imidazol-1-ylmethyl-benzonitrile (as described in Example 1, Step E) (0.100 g, 0.497 mmol), 3- (3-hydroxy-phenyl)-l-methyl-azepan-2-one (0.141 g, 0.646 mmol), KF*Al203 (0.141 g) and 18-Crown-6 (0.014 g, 0.053 mmol) were refluxed in CH3CN (8 mL) under Ar for 48 h. The solution was filtered, concentrated in vacuo, and purified using reverse phase chromatography (95/5-5/95 H20/CH3CN with 0.1% TFA, flow = 65 mL/min). The compound was converted to its free base using satd. NaHC03 solution, extracted with CH2C12 (3x), dried (MgS04), filtered and treated with 1N HCl ethereal solution to give the title compound.

FAB MS (M+1) = 401.

Analysis calculated for C24H24N402'1.00 HCl#0. 65 H2O 0.20 CH2C12: C, 62.55; H, 5.58; N, 12.06; Found: C, 62.86; H, 5.61; N, 11.67.

Using the procedures outlined above but substituting the appropriate N-methylated lactam for N-methylcaprolactam, the following compounds were prepared: 4-imidazol-1-ylmethyl-2-r3- (l-methyl-2-oxo-azocan-3-yl)-phenoxy-benzonitrile FAB MS (M+1) = 415 Analysis calculated for C25H26N4O2#1.85HCl#0. 35 (C2H5) 20: C, 62.42; H, 6.22; N, 11.03; Found: C, 62.42; H, 6.15; N, 11.01.

4-imidazol-1-ylmethyl-2-[3-(1-methyl-2-oxo-piperidin-3-yl )-phenoxy]-benzonitrile FAB MS (M+1) = 387 4-imidazol-1-ylmethyl-2-[3-(1-methyl-2-oxo-pyrrolidin-3-yl)- phenoxy]-benzonitrile

FAB MS (M+1) = 373 Quenching the reaction in Step A with ethyl iodide instead of H20, provided: 4-imidazol-1-ylmethyl-2- 3- (3-ethyl-1-methyl-2-oxo-piperidin-3-yl)- phenoxy-benzonitrile FAB MS (M+1) = 415 EXAMPLE 5 Preparation of 4-Imidazol-1-ylmethyl-2- 3- (2-oxo-azepan-3-yl)-phenoxy- benzonitrile hydrochloride Step A: Preparation of 1-trimethylsilanyl-azepan-2-one Caprolactam, commercially available from Maybridge, (10.0 g, 88.0 mmol), Et3N (14.8 mL, 106 mmol) and chlorotrimethylsilane (13.5 mL, 106 mmol) were dissolved in toluene (60 mL) and refluxed under Ar for 3h. The reaction was filtered and concentrated to give the title compound.

Step B: Preparation of 3- (3-methoxy-phenyl)-azepan-2-one To 1.6 M n-BuLi in hexane (31.3 mL, 50 mmol) at 0°C was added a solution of 2,2,6,6-tetramethyl piperidine (8.43 mL, 50 mmol) in anhydrous THF (100 mL) over 5 min. 1-Trimethylsilanyl-azepan-2-one (as described in Step A above) (9.26 g, 50 mmol) in anhydrous THF (20 mL) was added. After stirring the reaction mixture for 10 min a further portion of 1.6M n-BuLi in hexane (31.3 mL, 50 mmol) was added, the mixture stirred for 10 min, and 2-chloroanisole (6.34 mL, 50 mmol) was added. After stirring for 30 min under Ar, the reaction was quenched with H20, stirred for 30 min, concentrated in vacuo, diluted with EtOAc, washed with 5N HCI (100 mL) and brine. The organic layer was dried (MgS04), concentrated and purified using Si02 chromatography (0.5% MeOH/CH2Cl2) to give the title compound.

Step C: Preparation of 3- (3-hydroxy-phenyl)-azepan-2-one A 1M solution of BBr3 (1.0 mL, 1.00 mmol) in CH2C12 was added to 3- (3-methoxy-phenyl)-azepan-2-one (as described in Step B above) (0.200 g, 0.912 mmol) dissolved in anhydrous CH2C12 (5 mL) at-78°C. After 30 hr of stirring at RT,

the reaction was quenched with H20, extracted with CH2CI2 (3X), dried (MgS04), concentrated and purified using reverse phase chromatography (95/5-5/95 H20/CH3CN with 0.1% TFA, flow = 65 mL/min) to give the title compound.

Step D : Preparation of 4-imidazol-1-ylmethyl-2- 3- (2-oxo-azepan-3-yl)- phenoxy-benzonitrile hydrochloride 2-Fluoro-4-imidazol-1-ylmethyl-benzonitrile (as described in Example 1, Step E) (0.039 g, 0.194 mmol), 3- (3-hydroxy-phenyl)-azepan-2-one (as described in Step C above) (0.040 g, 0.646 mmol), KF*Al203 (0.048 g) and 18-Crown-6 (0.004 g, 0.015 mmol) were refluxed in CH3CN (6 mL) under Ar for 72 h. The solution was filtered, concentrated in vacuo, and purified using reverse phase chromatography (95/5-5/95 H20/CH3CN with 0.1% TFA, flow = 65 mL/min). The compound was converted to its free base using satd. NaHC03 solution, extracted with CH2CI2 (3x), dried (MgS04), filtered and treated with IN HCl ethereal solution to give the title compound.

FAB MS (M+1) = 387.

Analysis calculated for C23H22N402'1.00 HCl-0.40 CH2C12: C, H, 5.25; N, 12.26; Found: C, 61.47; H, 5.21; N, 11.94.

EXAMPLE 6 Preparation of 2- 3- (3-hydroxymethyl-l-methyl-2-oxo-azepan-3-yl)-phenoxy-4- imidazol-1-ylmethyl-benzonitrile hydrochloride Step A : Preparation of 3-methoxymethyl-3-(3-methoxy-phenyl)-1-methyl- azepan-2-one To 1.6 M n-BuLi in hexane (3.38 mL, 5.42 mmol) at 0°C was added a solution of 2,2,6,6-tetramethyl piperidine (0.91 mL, 5.42 mmol) in anh. THF (10 mL) over 5 min. A solution of 3- (3-methoxy-phenyl)-1-methyl-azepan-2-one (as described in Example 4, Step A) (0.632 g, 2.71 mmol) in anhydrous THF (10 mL) was added and stirred 0°C for 1 h. lodomethyl methyl ether (0.275 mL, 3.25 mmol) was added.

After stirring for 30 min, the reaction was quenched with H20, concentrated in vacuo,

diluted with EtOAc, washed with 5N HCI (100 mL), water and brine. The organic layer was dried (MgS04) and concentrated to give the title compound.

Step B : Preparation of 3-hydroxymethyl-3- (3-hydroxy-phenyl)-1-methyl- azepan-2-one A 1M solution of BBr3 (1.48 mL, 1.48 mmol) in CH2C12 was added to 3-methoxymethyl-3- (3-methoxy-phenyl)-l-methyl-azepan-2-one (as described in Step A above) (0.391 g, 1.35 mmol) dissolved in anh. CH2C12 (5 mL) at-78°C. After 24h of stirring at RT, the reaction was quenched with H20, extracted with CH2CI2 (3X), dried (MgS04), concentrated and purified using Si02 chromatography (1-1.5% MeOH/CH2Cl2with HOAc) to give the title compound.

Step C: Preparation of 2- 3- (3-hydroxymethyl-l-methyl-2-oxo-azepan-3-yl)- phenoxy-4-imidazol-1-ylmethyl-benzonitrile hydrochloride 2-Fluoro-4-imidazol-1-ylmethyl-benzonitrile (as described in Example 1, Step E) (0.089 g, 0.441 mmol), 3-hydroxymethyl-3-(3-hydroxy-phenyl)-1-methyl- azepan-2-one (0.100 g, 0.401 mmol), KFAl203 (0.120 g) and 18-crown-6 (0.10 g, 0.038 mmol) were refluxed in CH3CN (5 mL) under Ar for 24h. The solution was filtered, concentrated in vacuo, and purified using reverse phase chromatography (95/5-5/95 H20/CH3CN with 0.1 % TFA, flow = 65 mL/min). The compound was converted to its free base using satd. NaHC03 solution, extracted with CH2CI2 (3x), dried (MgSO4), filtered and treated with IN HCl ethereal solution to give the title compound. FAB MS (M+1) = 431.

Analysis calculated for C25H26N403'1.00 Hui-1.25 H2O#0.15 CH2Cl2 : C, 60.14; H, 5.98; N, 11.16; Found: C, 60.15 ; H, 5.96; N, 11.01.

Using the procedures above but substituting cyclopropylmethyl bromide for iodomethyl methyl ether in Step A, the following compound was prepared: 2-[3-(3-Cyclopropylmethyl-1-methyl-2-oxo-azepan-3-yl)-phenox y]-4- imidazol-1-ylmethyl-benzonitrile hydrochloride FAB MS (M+1) = 455.

Analysis calculated for C28H3oN403-1.25 HCl#0. 55 H20: C, 65.93; H, 6.39; N, 10.99;

Found: C, 65.95; H, 6.39; N, 10.67.

Chromatographic isolation of a brominated byproduct from the demethylation with BBr3 in Step B gave the following: 2-[4-Bromo-3-(3-cyclopropylmethyl-1-methyl-2-oxo-azepan-3-yl )-phenoxy]- 4-imidazol-1-ylmethyl-benzonitrile hydrochloride FAB MS (M+1) =533.

Analysis calculated for C28H2gBrN403 1.90 CF3CO2H 1 85 H20: C, 48.75; H, 4.45; N, 7.15; Found: C, 48.76; H, 4.43; N, 7.39.

EXAMPLE 7 Preparation of 2- 3- (3-methoxymethyl-l-methyl-2-oxo-azepan-3-yl)-phenoxy-4- imidazol-1-vlmethyl-benzonitrile hydrochloride Step A : Preparation of 3-methoxymethyl-3- (3-hydroxy-phenyl)-1-methyl- azepan-2-one 3-Methoxymethyl-3- (3-methoxy-phenyl)-l-methyl-azepan-2-one (as described in Example 6, Step A) (0.391 g, 1.48 mmol) and LiI (0.500 g, 3.73 mol) were heated in collidine (3.5 mL) at 200°C for 10 h. The reaction mixture was cooled, diluted with EtOAc, washed with IN HCl (2X), H20, brine, dried (MgS04) and concentrated to give the title compound.

Step B : Preparation of 2- 3- (3-methoxymethyl-1-methyl-2-oxo-azepan-3-yl)- phenoxvl-4-imidazol-1-ylmethyl-benzonitrile hydrochloride 2-Fluoro-4-imidazol-1-ylmethyl-benzonitrile (as described in Example 1, Step E) (0.098 g, 0.488 mmol), 3-methoxymethyl-3- (3-hydroxy-phenyl)-l-methyl- azepan-2-one (as described in Step A above) (0.129 g, 0.488 mmol), KFAl203 (0.155 g) and 18-Crown-6 (0.13 g, 0.049 mmol) were refluxed in CH3CN (10 mL) under Ar for 72h. The solution concentrated in vacuo, diluted with EtOAc, washed with satd. NaHC03 solution, H20, brine, dried (MgS04) and purified using Si02 chromatography (1-2% MeOH/CH2C12 with NH40H). The compound treated with IN HCl ethereal solution to give the title compound. FAB MS (M+1) =445.

Analysis calculated for C26H28N403'1.40 HCl 1.05 H20: C, 60.69; H, 6.17; N, 10.89; Found: C, 60.68; H, 6.16; N, 10.66.

EXAMPLE 8 Preparation of 2- 3- (3-ethyl-2-oxo-azepan-3-yl)-phenoxy-4-imidazol-1-ylmethyl- benzonitrile Step A : Preparation of 3-ethyl-3- (3-methoxy-phenyl)-azepan-2-one To 1.6 M n-BuLi in hexane (31.3 mL, 50 mmol) at 0°C was added a solution of 2,2,6,6-tetramethyl piperidine (8.43 mL, 50 mmol) in anhydrous THF (100 mL) over 5 min. 1-Trimethylsilanyl-azepan-2-one (as described in Example 5, Step A) (9.26 g, 50 mmol) in anhydrous THF (20 mL) was added. After stirring the reaction mixture for 10 min, a further portion of 1.6M n-BuLi in hexane (31.3 mL, 50 mmol) was added, the mixture stirred for 10 min, and 2-chloroanisole (6.34 mL, 50 mmol) was added. After stirring for 30 min under Ar, ethyl iodide (3.90 mL, 48.7 mmol) was added, the reaction mixture was stirred for 30 min, quenched with H20, concentrated in vacuo, diluted with EtOAc, and washed with 5N HCl (100 mL) and brine. The organic layer was dried (MgS04), concentrated and purified using SiO2 chromatography (10-15% EtOAc/CH2Cl2) to give the title compound.

Step B: Preparation of 3-ethyl-3- (3-hydroxy-phenyl)-azepan-2-one A 1M solution of BBr3 (7.1 mL, 7.1 mmol) in CH2Cl2 was added to 3-ethyl-3- (3-methoxy-phenyl)-azepan-2-one (as described in Step A above) (0.800 g, 0.323 mmol) dissolved in anhydrous CH2C12 (5 mL) at-78°C. After 48h of stirring at room temperature, the reaction was quenched with H20, extracted with CH2C12 (3X), dried (MgS04), concentrated and purified using reverse phase chromatography (95/5- 5/95 H20/CH3CN with 0.1% TFA, flow = 65 mL/min) to give the title compound.

Step C : Preparation of 2- 3- (3-ethyl-2-oxo-azepan-3-yl)-phenoxy-4-imidazol- 1-ylmethyl-benzonitrile 2-Fluoro-4-imidazol-1-ylmethyl-benzonitrile (as described in Example 1, Step E) (0.065 g, 0.325 mmol), 3-ethyl-3- (3-hydroxy-phenyl)-azepan-2-one (as

described in Step B above) (0.076 g, 0.325 mmol), KF-A1203 (0. 091 g) and 18- Crown-6 (0.008 g, 0.030 mmol) were refluxed in CH3CN (8 mL) under Ar for 72h.

The solution was filtered, concentrated in vacuo, and purified using Si02 chromatography (0.5-3.0% MeOH/CH2Cl2). The compound was treated with IN HCl ethereal solution to give the title compound.

FAB MS (M+1) = 415.

Analysis calculated for C25H26N402'1.00 HCl-0.10 H2O 0.40 CH2C12: C, 62.67; H, 5.80; N, 11.51; Found: C, 62.72; H, 5.58; N, 11.36.

EXAMPLE 9 Preparation of 2- 3- (3-ethyl-azepan-3-yl)-phenoxy-4-imidazol-1-ylmethyl- benzonitrile bishydrochloride Step A: Preparation of 3-ethyl-3- (3-methoxy-phenvl)-azepane 3-Ethyl-3- (3-methoxy-phenyl)-azepan-2-one (as described in Example 8, Step A) (1.05 g, 4.24 mmol) in anhydrous THF (4.2 mL) was added to a solution of LiAlH4 (0.193 g, 5.09 mmol) in anhydrous THF (4.2 mL) and refluxed for 24h. The solution was cooled, quenched with H20 (0.18 mL), 15% NaOH (0.18 mL), H20 (0.54 mL), filtered and concentrated. The residue was purified using reverse phase chromatography (95/5-5/95 H20/CH3CN with 0.1% TFA, flow = 65 mL/min) and converted to its free base using saturated NaHC03 solution, extracted with CH2Cl2 (3x) and dried (MgS04) to give the title compound.

Step B : Preparation of 3- (3-ethyl-azepan-3-vl)-phenol A 1M solution of BBr3 (2.56 mL, 2.56 mmol) in CH2C12 was added to 3-ethyl-3- (3-methoxy-phenyl)-azepane (as described in Example 8, Step A) (0.300 g, 0.1.28 mmol) dissolved in anhydrous CH2Cl2 (10 mL) at-78°C. After 20h of stirring at room temperature, the reaction was quenched with sat. NaHC03 solution, extracted with CH2Cl2 (3X), dried (MgS04) and concentrated to give the title compound.

Step C : Preparation of 2- 3- (3-ethyl-azepan-3-yl)-phenoxy-4-imidazol- 1-ylmethyl-benzonitrilebishydrochloride

2-Fluoro-4-imidazol-1-ylmethyl-benzonitrile (as described in Example 1, Step E) (0.113 g, 0.560 mmol), 3- (3-ethyl-azepan-3-yl)-phenol (as described in Step B above) (0.123 g, 0.560 mmol), KF*Al203 (0.148 g) and 18-Crown-6 (0.012 g, 0.038 mmol) were refluxed in CH3CN (5 mL) under Ar for 24h. The solution was filtered, concentrated in vacuo, and purified using reverse phase chromatography (95/5-5/95 H20/CH3CN with 0.1 % TFA, flow = 65 mL/min). The compound was converted to its free base using saturated NaHC03 solution, extracted with CH2C12 (3x), dried (MgS04), filtered and treated with IN HCl ethereal solution to give the title compound. FAB MS (M+1) = 401.

Analysis calculated for C2sH28N401 2.00 HCl#0. 10 H2O 0.45 CH2C12: C, 59.53; H, 6.11; N, 10.91; Found: C, 59.55; H, 6.13; N, 10.59.

EXAMPLE 10 Preparation of 2- 3- (1-acetyl-3-ethyl-azepan-3-yl)-phenoxy-4-imidazol-1-ylmethyl - benzonitrile hydrochloride 2- 3- (3-Ethyl-azepan-3-yl)-phenoxy-4-imidazol-1-ylmethyl- benzonitrile bistrifluoroacetate (as described in Example 9) (0.116 g, 0.184 mmol), Et3N (0.103 mL, 0.736 mmol) and acetyl chloride (0.020 mL, 0.276 mmol) were dissolved in anhydrous. CH2C12 (5 mL). After lh, the reaction was concentrated in vacuo, and purified using reverse phase chromatography (95/5-5/95 H20/CH3CN with 0.1 % TFA, flow = 65 mL/min). The compound was converted to its free base using saturated NaHC03 solution, extracted with CH2C12 (3x), dried (MgSO4), filtered and treated with IN CL ethereal solution to give the title compound.

FAB MS (M+1) = 443.

Analysis calculated for C27H30N4o2 1.00 HCl#0. 35 H2O#0. 40 CH2C12: C, 63.37; H, 6.31; N, 10.79; Found: C, 63.44; H, 6.42; N, 10.39. acylated2-[3-(3-ethyl-azepan-3-yl)-phenoxy]-4-imidazol-1-ylm ethyl-benzonitrilewas with Boc2O to give 3- 3- (2-cyano-5-imidazol-1-ylmethyl-phenoxy)-phenyl-3-ethyl- azepane-1-carboxylic acid-tert-butyl ester FAB MS (M+1) = 501.

Analysis calculated for C30H36N4o3 0.40 H20:

C, 70.95; H, 7.30; N, 11.03; Found: C, 70.92; H, 7.00; N, 10.73.

EXAMPLE 11 Preparation of 4- 5- (2-amino-ethyl)-2-methyl-imidazol-1-ylmethyl-2- 3- (3-ethyl- 1-methyl-2-oxo-azepan-3-yl)-phenoxy-benzonitrile dihydrochloride Step A : Preparation of {2- 3- (4-cyano-3-fluoro-benzyl)-2-methyl-3H-imidazol- 4-yl-ethyl-carbamic acid tert-butyl ester To a solution of NG-pivaloyloxymethyl-Na-phthaloyl-histamine (J. C.

Emmett, F. H. Holloway, and J. L. Turner, J. Chem. Soc., Perkin Trans. 1,1341, (1979)) (4.59 g, 0.0124 mmol) in acetonitrile (40 mL) was added 2-fluoro-4-imidazol- 1-ylmethyl-benzonitrile (as described in Example 1, Step E) (2.8 g, 0.013 mmol) and the mixture was heated to reflux for 18 hr. A white solid precipitate formed which after cooling to 0°C was collected by filtration to obtain the quaternary salt. This intermediate was dissolved in EtOH (100 mL), hydrazine (1.46 mL, 0.046 mmol) was added, and the mixture was heated at reflux for 4 hr. A white precipitate was observed and the reaction was cooled to 25°C. Dimethylphthalate (11.4 mL, 0.0699 mmol) was added and the mixture was again refluxed for 18 hr. After cooling to 25°C the precipitate was removed by filtration and washed with EtOAc. The filtrate was evaporated in vacuo and the residue was dissolved in THF (125 mL) and H20 (25 mL). To this solution was added solid Na2CO3 (4.0 g, 0.0377 mol) and BOC20 (4.47 g, 0.020 mol) and the reaction was stirred for 18 hr. The THF was removed in vacuo and the mixture was partitioned with EtOAc and saturated NaHC03. The EtOAc layer was washed with brine, dried with MgSO4, and evaporated in vacuo to obtain the title product after chromatography (silica gel, CH2Cl2: MeOH: NH40H/ 97: 3: 0.3).

Step B : Preparation of 2- (3- {4-cyano-3- 3- (3-ethyl-1-methyl-2-oxo-azepan- 3-yl)-phenoxy-benzyl}-2-methyl-3H-imidazol-4-yl)-ethyl-carba mic acid tert-butyl ester Following the procedure outlined in Example 2, but using {2- 3- (4- cyano-3-fluoro-benzyl)-2-methyl-3H-imidazol-4-yl-ethyl}-carb amic acid tert-butyl ester (as described in Step A above) (0.60 g, 1.67 mmol) and 3-ethyl-3- (3-hydroxy-

phenyl)-1-methyl-azepan-2-one (0.41 g, 1.67 mmol), the title compound was obtained after chromatography (CH2Cl2: CH30H: NH40H, 98: 2: 0.2) Step C: Preparation of 4- 5- (2-amino-ethyl)-2-methyl-imidazol-1-ylmethyl- 2- 3- (3-ethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-benzonitrile dihydrochloride To a solution of 2- (3- {4-cyano-3- 3- (3-ethyl-1-methyl-2-oxo-azepan- 3-yl)-phenoxy-benzyl}-2-methyl-3H-imidazol-4-yl)-ethyl-carba mic acid tert-butyl ester (as described in Step B above) (0.2 g, 0.41 mmol) in CH2C12 (6.0 mL) was added TFA (3.0 mL) and the solution was stirred for 0.5 hr. The solvents were removed in vacuo and the crude product was purified by preparative HPLC.

Conversion to the HCl salt yielded the title compound.

FAB mass spectrum m/e 486 (m+1).

Analysis calculated for C29H35N502'2.2 HCI: C, 61.55; H, 6.63; N, 12.38; Found: C, 61.56; H, 6.45; N, 11.83.

EXAMPLE 12 Preparation of 2- 3- (3-ethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-4- 2-methyl-5- (2-morpholin-4-yl-ethyl !-imidazol-1-ylmethyl-benzonitrile To a solution of 4- 5- (2-amino-ethyl)-2-methyl-imidazol-1-ylmethyl- 2- 3- (3-ethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-benzonitrile dihydrochloride (as described in Example 11) (0.25 g, 0.456 mmol) in acetonitrile (35.0 mL) and triethyl- amine (1.8 mL) was added 2-bromoethyl ether (0.133 mL, 1.06 mmol) and the mixture was refluxed for 48 hr. The solvents were removed in vacuo to obtain the title compound after purification by preparative HPLC.

FAB mass spectrum m/e 556 (m+1).

Analysis calculated for C33H4lN503-0. 8 H20: C, 69.51; H, 7.53; N, 12.28; Found: C, H, N, 12.13.

EXAMPLE 13

Preparation of N- 2- (3- {4-cyano-3- 3- (3-ethyl-1-methyl-2-oxo-azepan-3-yl)- phenoxyl-benzyl $-2-methyl-3H-imidazol-4-yl !-ethyl-acetamide To a solution of 4- 5- (2-amino-ethyl)-2-methyl-imidazol-1-ylmethyl- 2- 3- (3-ethyl-l-methyl-2-oxo-azepan-3-yl)-phenoxy-benzonitn*le dlhydrochloride (as described in Example 11) (0.06 g, 0.108 mmol) in CH2C12 (3.0 mL) was added triethylamine (0.06 mL, 0.43 mmol) and acetyl chloride (0.010 mL, 0.14 mmol).

After stirring for 2 hr, the solvents were removed in vacuo and the residue was partitioned with EtOAc and saturated NaHC03. The EtOAc was dried with brine and MgS04 and evaporated in vacuo to obtain the title compound.

FAB mass spectrum m/e 528 (m+1).

EXAMPLE 14 Preparation of 3-ethyl-3- 3- (3-imidazol-1-ylmethyl-phenoxy)-phenyl-1-methyl- azepan-2-one hydrochloride Step A: Preparation of 3- 3- (3-ethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy- benzaldehyde 3-Ethyl-3- (3-hydroxy-phenyl)-1-methyl-azepan-2-one (as described in Example 8, Step B) (0.10 g, 0.4 mmol), 3-formylphenyl-boronic acid (0.12 g, 0.8 mmol), cupric acetate (0.073 g, 0.4 mmol) and powdered 4A molecular sieves (0.12 g) were combined under Ar, diluted with anhydrous CH2C12 (2 mL) and pyridine (0.162 mL, 2.0 mmol) and stirred at ambient temperature for 48 hrs. The reaction mixture was filtered, and the filtrated concentrated to dryness and partitioned between EtOAc and H20. The aqueous layer was washed with EtOAc, the organics combined, washed with H20,10% aqueous NaOH solution, H20, brine, and dried (Na2SO4).

Filtration and concentration to dryness gave the title compound as a white solid after trituration with diethyl ether/hexane.

Step B: Preparation of 3-ethyl-3- 3-hydroxymethyl-phenoxy)-phenyl-1- methyl-azepan-2-one 3- 3- (3-Ethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy- benzaldehyde (as described in Step A above) (0.096 g, 0.273 mmol) in absolute ethanol (2 mL) was treated with sodium borohydride (0.010 g, 0.273 mmol) with stirring at ambient temperature. After 2 hrs the reaction was quenched with H20, concentrated to

remove the EtOH, and the residue partitioned between EtOAc and H20. The aqueous layer was washed with EtOAc, the organics combined, washed with brine, and dried (Na2SO4). Filtration and concentration to dryness gave the title compound as a colorless oil which solidified on standing.

Step C : Preparation of 3-ethyl-3- 3- (3-imidazol-1-ylmethyl-phenoxy)-phenyl- 1-methyl-azepan-2-onehydrochloride 3-ethyl-3- 3-hydroxymethyl-phenoxy)-phenyl-1-methyl-azepan-2-one (as described in Step B above) (0.090 g, 0.26 mmol) and N, N'-carbonyldiimidazole (0.176 g, 1.04 mmol) and imidazole (0.027 g, 0.4 mmol) were dissolved in acetonitrile (10 mL) and heated at 80°C. for 48 hrs. The reaction mixture was concentrated to dryness and purified on a Waters RP HPLC Prep Pak eluting with 0.1% TFA/H20: 0. 1%TFA/CH3CN to give the title compound after conversion to the hydrochloride salt.

FAB mass spectrum m/e 404 (m+1).

Analysis calculated for C2sH2gN302 1.0 HCI * 0.85 H20: C, 65.94; H, 7.02; N, 9.23; Found: C, 66.18; H, 6.95; N, 8.83.

EXAMPLE 15 Preparation of 2- 3- (3-ethyl-l-methyl-2-oxo-azepan-3-yl)-phenoxy-4- (3-methyl- 3-H-imidazol-4-ylmethvl)-benzonitrile hydrochloride Step A : Preparation of 2-fluoro-4- (1-trityl-1H-imidazol-4-ylmethyl)- benzonitrile Zinc (0.301 g, 4.63 mmol) and dibromoethane (0.040 mL, 0.463 mmol) were stirred in anhydrous THF (2 mL) under Ar for 1.5 h. A solution of 4- bromomethyl-2-fluoro-benzonitrile (as described in Example 1, Step D) (0.546 g, 2.55 mmol) in anhydrous THF (1 mL) was added dropwise and stirred for 1 h. A mixture of 4-iodo-1-trityl-1H-imidazole (0.808 g, 1.85 mmol) and NiCl2 (PPh3) 2 (0.120 g, 0.183 mmol) were added together to the reaction. After stirring overnight under Ar, the reaction was quenched with NH4Cl (9 mL), stirred for 2 h, extracted with CH2Cl2

(3X), dried (MgS04), concentrated and purified using Si02 chromatography (0.5% MeOH/CH2Cl2 w/NH40H) to give the title compound.

Step B : Preparation of 2- 3- (3-ethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy- 4-(1-tritvl-lH-imidazol-4-Ylmethyl)-benzonitrile 2-Fluoro-4- (1-trityl-1H-imidazol-4-ylmethyl)-benzonitrile (as described in Step A above) (0.348 g, 0.784 mmol), 3-ethyl-3- (3-hydroxy-phenyl)-l- methyl-azepan-2-one (as described in Example 8, Step B) (0.194 mg, 0.784 mmol), KFAl203 (0. 232 g) and 18-Crown-6 (0.020 g, 0.076 mmol) were refluxed in CH3CN (15 mL) under Ar for 48h. The solution was concentrated in vacuo, diluted with EtOAc, washed with IN NaOH solution, water, brine, dried (MgS04), concentrated and purified using Si02 chromatography (1.0% MeOH/CH2Cl2) to give the title compound.

Step C : Preparation of 2- 3- (3-ethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy- 4- (3-methyl-3-H-imidazol-4-ylmethyl)-benzonitrile hydrochloride 2- 3- (3-Ethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-4- (l-trityl-1 H- imidazol-4-ylmethyl)-benzonitrile (as described in Step B above) (0.100 g, 0.149 mmol) and dimethyl sulfate (0.014 mL, 0.149 mmol) were dissolved in EtOAc (10 mL) and heated at 60°C for 18 h under Ar. The reaction was concentrated in vacuo, triturated with Et20, dissolved in MeOH (15 mL) and refluxed for Ih. The solution was concentrated in vacuo and purified using reverse phase chromatography (95/5-5/95 H20/CH3CN with 0.1% TFA, flow = 65 mL/min). The compound was converted to its free base using saturated NaHC03 solution, extracted with CH2C12 (3x), dried (MgS04), filtered and treated with IN HCl ethereal solution to give the title compound.

FAB MS (M+1) = 443 EXAMPLE 16 Preparation of 2- 3- (3-ethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-4- (3H- imidazol-4-ylmethyl)-benzonitrile hydrochloride 2- 3- (3-ethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-4- (l-trityl-1 H- imidazol-4-ylmethyl)-benzonitrile (as described in Example 15, Step B) (0.074 g,

0.110 mmol) and triethylsilane (0.140 mL, 0.828 mmol) were stirred in CH2C12 (5 mL) and TFA (2.5 mL) for 1 h. The solution was concentrated in vacuo and purified using reverse phase chromatography (95/5-5/95 H20/CH3CN with 0.1% TFA, flow = 65 mL/min). The compound was converted to its free base using saturated NaHC03 solution, extracted with CH2C12 (3x), dried (MgS04), filtered and treated with IN HC1 ethereal solution to give the title compound.

FAB MS (M+1) = 429 EXAMPLE 17 Preparation of 2- 3- (3-ethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-4- hydroxy- (3-methyl-3-H-imidazol-4-yl)-methyll-benzonitrile Step A: Preparation of 2-Fluoro-4-formylbenzonitrile 2-Fluoro-4-hydroxymethylbenzonitrile (as described in Example 1, Step C) (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 (Na2SO4). Filtration and concentration gave the title compound after purification by chromatography (silica gel, hexane: EtOAc, 3: 1).

1H NMR (CDC13) 8 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 B : Preparation of 2-fluoro-4-hydroxy-(1-trityl-lH-imidazol-4-yl)- methyll-benzonitrile To a solution of 4-iodo-1-trityl-lH-imidazole (5.00 g, 11.5 mmol) in anhydrous CH2C12 (30 mL) was added a 3.0M solution of ethylmagnesium bromide (6.58 mL, 19.7 mmol) with stirring under Ar. After 3h, the reaction mixture was cooled to-78°C and a solution of 2-fluoro-4-formyl-benzonitrile (as described in Step A above) (1.70g, 11.5 mmol) dissolved in CH2C12 (20 mL) was added dropwise. The reaction was allowed to warm to room temperature over 2h, quenched with saturated

NH4C1 solution, diluted with satd. NaHC03 solution to pH=8.5, and extracted with CH2C12 (3X). The combined organic layers were dried (MgS04), concentrated and purified using Si02 chromatography (0-1 % MeOH/CH2Cl2) to yield the title compound.

Step C : Preparation of acetic acid (4-cyano-3-fluoro-phenyl)- (1-trityl-lH- imidazol-4-vl)-methyl ester 2-Fluoro-4- hydroxy- (1-trityl-lH-imidazol-4-yl)-methyl-benzonitrile (as described in Step B above) (4.05 g, 8.81 mmol), pyridine (2.14 mL, 26.4 mmol), and acetic anhydride (12.5 mL, 132 mmol) were stirred in anhydrous DMF (60 mL) for 3h under Ar. The reaction was concentrated in vacuo, diluted with EtOAc (250 mL), washed with H20 (2X), brine, dried (MgS04) and concentrated to give the title compound.

Step D : Preparation of acetic acid (4-cyano-3-fluoro-phenyl)- (3-methyl-3H imidazol-4-vl)-methyl ester Acetic acid (4-cyano-3-fluoro-phenyl)- (1-trityl-1H-imidazol-4-yl)- methyl ester (as described in Step C above) (4.60 g, 9.17 mmol) and dimethyl sulfate (0.83 mL, 8.81 mmol) were dissolved in EtOAc (20 mL) and heated at 60°C overnight under Ar. The reaction was concentrated in vacuo, diluted with MeOH (30 mL), and refluxed for lh. Concentrated in vacuo and purified using Si02 chromatography (0.5-4% MeOH/CH2C12 with NH40H) to give the title compound.

Step E: Preparation of 2-fluoro-4- hydroxy- (3-methyl-3H-imidazol-4-yl)- methyl-benzonitrile Acetic acid (4-cyano-3-fluoro-phenyl)- (3-methyl-3H imidazol-4-yl)- methyl ester (as described in Step C above) (1.26 g, 4.59 mmol) and NaOH (5.5 mL, 5.5 mmol) were dissolved in THF (15 mL) and H20 (25 mL). After Ih, the reaction was diluted with satd. NaHC03 solution, extracted with CH2CI2 (3X), dried (MgS04) and concentrated to give the title compound.

Step F : Preparation of 2- 3- (3-ethyl-l-methyl-2-oxo-azepan-3-yl)-phenoxy- 4- hydroxy- (3-methyl-3-H-imidazol-4-vl)-methyll-benzonitrile

2-Fluoro-4- hydroxy- (3-methyl-3H-imidazol-4-yl)-methyl- benzonitrile (as described in Step E above) (0.162 g, 0.700 mmol), 3-ethyl-3- (3- hydroxy-phenyl)-1-methyl-azepan-2-one (0.173 g, 0.700 mmol) and KF-A1203 (0.208 g) and 18-Crown-6 (0.017 g, 0.064 mmol) were dissolved in anhydrous CH3CN (7 mL) and refluxed under Ar for 24 h. The reaction was filtered, concentrated and purified using Si02 chromatography (1-3% MeOH/CH2C12).

FAB MS (M+1) = 459.

EXAMPLE 18 Preparation of 4- amino- (3-methyl-3-H-imidazol-4-yl)-methyl-2- 3- (3-ethyl- 1-methyl-2-oxo-azepan-3-yl)-phenoxy-benzonitrile 2- 3- (3-ethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-4- hydroxy- (3- methyl-3-H-imidazol-4-yl)-methyl-benzonitrile (as described in Example 17) (0.093 g, 0.202 mmol) was dissolved in SOC12 (5 mL) and stirred at room temperature for 2h under Ar. The solution was concentrated in vacuo and azeotroped with CH2Cl2 (3X).

The solid was dissolved in CHC13 (20 mL) and cooled to-78°C. NH3 (g) was bubbled through the solution and stirred for 16 h while warming to room temperature under Ar. The solution was concentrated in vacuo and purified using reverse phase chromatography (95/5-5/95 H20/CH3CN with 0.1% TFA, flow = 65 mL/min). The compound was converted to its free base using saturated NaHC03 solution, extracted with CH2Cl2 (3x), dried (MgS04), filtered and concentrated to give the title compound. FAB MS (M+1) = 458.

Analysis calculated for C27H31N502'0. 35 H20: C, 69.90; H, 6.89; N, 15.10; Found: C, 69.95; H, 7.08; N, 14.72.

EXAMPLE 19 Preparation of 2- 3- (3-ethyl-1-methyl-2-oxo-azepan-3-yl)-benzyl-4- (3-methyl- 3H-imidazole-4-carbonyl)-benzonitrile trifluoroacetate Step A: Preparation of 2-fluoro-4- amino- (3-methyl-3H-imidazol-4-yl)- methyll-benzonitrile

2-Fluoro-4- hydroxy- (3-methyl-3H-imidazol-4-yl)-methyl- benzonitrile (0.542 g, 2.31 mmol) was dissolved in SOC12 (15 mL) and stite at room temperature for 2h under Ar2. The solution was concentrated in vacuum and azeotroped with CH2C12 (3X). The solid was dissolved in CHC13 (30 mL) and cooled to-78°C. NH3 (g) was bubbled through the solution and stirred for 16 h while warming to room temperature under Ar. After concentration to dryness and chromatography (silica gel, 1-2% CH30H/CH2C12 with NH3), the title compound was obtained.

Step B: Preparation of 2- 3- (3-ethyl-1-methyl-2-oxo-azepan-3-yl)-benzyl- 4- (3-methvl-3H-imidazole-4-carbonyl)-benzonitrile trifluoroacetate 2-Fluoro-4- amino- (3-methyl-3H-imidazol-4-yl)-methyl-benzonitrile (as described in Step A above) (0.012 g, 0.052 mmol), 3-ethyl-3- (3-hydroxy-phenyl)- 1-methyl-azepan-2-one (0.014 g, 0.056 mmol), KF-A1203 (0.020 g) and 18-Crown-6 (0.001 g) were dissolved in anhydrous CH3CN (2 mL) and refluxed under Ar for 24 h. The reaction was filtered, concentrated and purified using RP LC on a VYDAC column eluting with 0.1% TFA/CH3CN to give the title compound.

FAB MS (M+1) = 457.

EXAMPLE 20 Preparation of 2- 3- (3-ethyl-l-methyl-2-oxo-azepan-3-yl)-phenoxy-4- (hydroxy- pyridin-3-yl-methvl)-benzonitrile Step A: Preparation of 4-Bromo-3-fluorobenzaldehvde To a well-stirred mixture of 4-bromo-3-fluorobenzyl alcohol (as described in Example 1, Step B) (10.25 g, 0.05 mol), TEMPO (0.781 g, 0.005 mol) and tetrabutylammonium fluoride (1.39 g, 0.005 mol) in CH2C12 (200 mL) and a solution of0. 5M NaHC02/0. 05M K2C03 (200 mL) was added N-chlorosuccinimide (9.35 g, 0.07 mol). After 6 hrs, the layers were separated, the aqueous layer back- washed with CH2C12 (2 x 50 mL), the organics combined and dried (Na2SO4). The solution was filtered, concentrated to half its volume, then chromatographed (silica gel, CH2Cl2) to give the title compound.

1H NMR (CDCl3) 6 9.96 (s, 1H), 7.78 (dd, 1H, J = 2,8 Hz), 7.62 (dd, 1H, J = 2,8 Hz), 7.56 (dd, 1H, J = 2,8 Hz).

Step B: Preparation of (4-bromo-3-fluoro-phenyl)-pyridin-3-yl-methanol To a solution of 3-bromopyridine (0.986 mL, 10.23 mmol) in diethyl ether (20 mL) at-78°C was added n-butyl lithium (1.6M/hexane, 7.0 mL, 11.25 mmol) dropwise and the mixture was stirred for 0.5 hr. A turbid solution of magnesium bromide, preformed with magnesium (0.372 g, 15.3 mmol) and dibromoethane (1.3 mL, 15.3 mmol) in THF (20 mL) at-78°C for 20 min., was added. The resulting mixture was stirred at-78°C for 15 min. and then at 0°C for 20 min. To this mixture was added 4-bromo-3-fluorobenzaldehyde (as described in Step A above) (1.6 g, 7.88 mmol) in THF (10 mL) at 0°C and the reaction was stirred for 15 min. at °C and at 25°C for 2 hr. The reaction was cooled to 0°C and quenched with saturated NH4Cl. The mixture was partitioned with EtOAc and the organic layer was washed with water, brine and dried over MgS04. The solvents were removed in vacuo to obtain the title compound after chromatography (silica gel, CH2Cl2: MeOH: NH40H 98: 2: 0.2).

Step C: Preparation of 2-fluoro-4- (hydroxy-pyridin-3-yl-methyl)-benzonitrile Using the procedure described in Example 1, Step C, the title compound was prepared starting with (4-bromo-3-fluoro-phenyl)-pyridin-3-yl- methanol (1.0 g, 3.54 mmol).

Step D : Preparation of 2- 3- (3-ethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy- 4- (hydroxy-pYridin-3-yl-methyl)-benzonitrile Using the procedure described in Example 2, the crude title compound was prepared starting with 2-fluoro-4- (hydroxy-pyridin-3-yl-methyl)-benzonitrile (as described in Step C above) (0.075 g, 0.329 mmol) and 3-ethyl-3- (3-hydroxy-phenyl)- 1-methyl-azepan-2-one (0.082 g, 0.329 mmol). Purification by preparative HPLC and conversion to the free base yielded the title compound.

FAB mass spectrum m/e 456 (m+1).

Analysis calculated for C2gH29N303 0.55 H20: C, 72.25; H, 6.52; N, 9.03; Found: C, 72.23; H, 6.44; N, 8.71.

EXAMPLE 21 Preparation of 2- 3- (3-ethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-4-pyridin- 3-ylmethyl-benzonitrile hydrochloride Step A : Preparation of thiocarbonic acid 0- (4-cyano-3-fluoro-phenyl)- pyridin-3-yl-methyl ester O-phenvl ester To a solution of 2-fluoro-4- (hydroxy-pyridin-3-yl-methyl)-benzonitrile (as described in Example 20, Step C) (0.4 g, 1.75 mmol) in CH2C12 (25.0 mL) at 0°C was added phenyl thionochloroformate (0.388 mL, 2.8 mmol) and DMAP (0.471 g, 3.86 mmol). The solution was stirred at 25°C for 3hr. The reaction was partitioned with saturated NaHC03 and CH2Cl2. The CH2Cl2 layer was washed with brine, dried with MgS04, and evaporated in vacuo to obtain the title compound after chromatography (silica gel, CH2Cl2: MeOH 99: 1).

Step B: Preparation of 2-fluoro-4-pyridin-3-vlmethyl-benzonitrile To a solution of thiocarbonic acid 0- (4-cyano-3-fluoro-phenyl)- pyridin-3-yl-methyl ester O-phenyl ester (as described in Step A above) (0.5 g, 1.37 mmol) in benzene (2.0 mL) was added AIBN (0.18 g, 1.1 mmol) and tri-n- butyltinhydride (1.09 mL, 4.0 mmol) and the solution was heated to refluxed for 2 hr.

The solvents were removed in vacuo to obtain the title compound after chromatography (silica gel, CH2Cl2: MeOH 99: 1).

Step C : Preparation of 2- 3- (3-ethyl-1-methyl-2-oxo-azepan-3-yl) phenoxy]- 4-pyridin-3-ylmethyl-benzonitrile hydrochloride Using the procedure described in Example 2, the crude title compound was prepared starting with 2-fluoro-4-pyridin-3-yl-methyl-benzonitrile (as described in Step B above) (0.075 g, 0.354 mmol) and 3-ethyl-3- (3-hydroxy-phenyl)-l-methyl- azepan-2-one (0.087 g, 0.354 mmol). Purification by preparative HPLC and conversion to the HCl salt yielded the title compound.

FAB mass spectrum m/e 440 (m+1).

Analysis calculated for C28H29N302'1. 05 H20'1.25 HCI: C, 66.71; H, 6.47; N, 8.34; Found: C, 67.02; H, 6.47; N, 7.95.

EXAMPLE 22 Preparation of 2- 3- (3-ethyl-l-methyl-2-oxo-azepan-3-yl)-phenoxy-4-pyridin- 2-ylmethyl-benzonitrile hydrochloride Step A: Preparation of (4-bromo-3-fluoro-phenyl)-pyridin-2-yl-methanol To a solution of n-butyllithium (2.5 M/hexane, 0.98 mL, 2.46 mmol) in THF (7 mL) at-78°C was added 2-bromopyridine (0.235 mL, 2.46 mmol) dropwise.

The mixture was stirred at-78°C for 1 hr. This solution was added via cannula to a solution of 4-bromo-3-fluorobenzaldehyde (as described in Example 20, Step A) (0.5 g, 2.46 mmol) in THF (5 mL) at-78°C. The resulting solution was stirred at-78°C for 15 min. and at 25°C for 2 hr. The reaction was cooled to 0°C and quenched with saturated NH4Cl. The mixture was partitioned with EtOAc and the organic layer was washed with water, brine and dried over MgS04. The solvents were removed in vacuo to obtain the title compound after chromatography (silica gel, CH2Cl2: MeOH 99: 1).

Step B: Preparation of 2-fluoro-4- (hvdroxy-pyridin-2-yl-methyl)-benzonitrile Using the procedure described in Example 1, Step C, the title compound was prepared starting with (4-bromo-3-fluoro-phenyl)-pyridin-2-yl- methanol (0.28 g, 0.993 mmol). The product was purified and isolated by pouring the reaction into H20 and partitioning with EtOAc. The EtOAc was washed with 1M HCl which now contains the product. The HCl layer was adjusted to pH = 9 with solid Na2CO3 and extracted twice with EtOAc. The EtOAc layers were combined and washed with brine, dried with MgS04 and evaporated in vacuo to obtain the title compound.

Step C: Preparation of 2- (4-bromo-3-fluoro-phenyl)-chloro-methyl-pvridine To a solution of 2-fluoro-4- (hydroxy-pyridin-2-yl-methyl)-benzonitrile (as described in Step B above) (0.27 g, 0.957 mmol) in CHC13 (15 mL) was added thionyl chloride (0.104 mL, 1.44 mmol) and the solution was refluxed for 1 hr. The reaction was cooled and evaporated in vacuo. The residue was partitioned with EtOAc and 50% Na2CO3. The EtOAc was washed with H20, brine, dried with MgS04 and evaporated in vacuo to obtain the title compound which was used without further purification.

Step D: Preparation of 2- (4-bromo-3-fluoro-benzyl)-pyridine To a solution of 2-[(4-bromo-3-fluoro-phenyl)-chloro-methyl]-pyrdine (as described in Step C above) (0.29 g, 0.965 mmol) in HOAc (10 mL) was added zinc powder (0.315 g, 4.82 mmol) and the mixture was refluxed for 1 hr. The HOAc was removed in vacuo and the residue was partitioned with EtOAc and saturated NaHC03. The EtOAc was washed with H20, brine, dried with MgS04, and evaporated in vacuo to obtain the title compound which was used without further purification.

Step E: Preparation of 2-fluoro-4-pyridin-2-ylmethyl-benzonitrile Using the procedure described in Example 1, Step C, the title compound was prepared starting with (4-bromo-3-fluoro-phenyl)-pyridin-3-yl- methanol (0.27 g).

Step F : Preparation of 2- 3- (3-ethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy- 4-pvridin-2-vlmethyl-benzonitrile hydrochloride Using the procedure described in Example 2, the title compound was prepared starting with compound 2-fluoro-4-pyridin-2-ylmethyl-benzonitrile (as described in Step E above) (0.16 g, 0.754 mmol) and 3-ethyl-3- (3-hydroxy-phenyl)-l- methyl-azepan-2-one (0.186 g, 0.754 mmol).

FAB mass spectrum m/e 440 (m+1).

Analysis calculated for C28H29 N302 0.85 H2O 1.15 HCI: C, 67.69; H, 6.46; N, 8.46; Found: C, 67.64; H, 6.47; N, 8.09.

EXAMPLE 23 Preparation of 2- 3- (3-ethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-4- 1-hydroxy- 1- (3-methvl-3H-imidazol-4-yl)-eth1-benzonitrile 2-3- (3-Ethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-4- (3-methyl-

3H-imidazole-4-carbonyl)-benzonitrile (Example 19) (0.216 g, 0.473 mmol) was dissolved in anhydrous THF (10 mL) and a 3.0 M solution of MeMgBr (1.10 mL, 3.30 mmol) was added and stirred at RT. The reaction was quenched with NH4Cl after Ih, concentrated, diluted with EtOAc, washed with satd. NaHC03 solution, water, brine, dried (MgS04), concentrated and purified using Si02 chromatography (1-3 % MeOH/CH2Cl2 w/NH40H) to give the title compound. FT/ICR MS (M+1) = 473.

EXAMPLE 24 Preparation of 2- 3- (3-ethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-4- 1-amino- 1-(3-methyl-3H-imidazol-4-yl)-ethyl]-benzonitrile 2- 3- (3-ethyl-I-methyl-2-oxo-azepan-3-yl)-phenoxy-4- l-hydroxy-I- (3-methyl-3H-imidazol-4-yl)-ethyl-benzonitrile (as described in Example 23) (0.099 g, 0.209 mmol) was dissolved in SOC12 (5 mL) and stirred at RT for 2h. The solution was concentrated in vacuo and azeotroped with anhydrous CH2CI2 (3X). The solid was dissolved in CHC13 (5 mL) and cooled to-78°C. NH3 (g) was bubbled through the solution and stirred for 2h while warming to RT under Ar. The solution was concentrated in vacuo and purified using reverse phase chromatography (95/5-5/95 H2O/CH3CN with 0.1 % TFA, flow = 65 mL/min). The compound was converted to its free base using saturated NaHC03 solution, extracted with CH2C12 (3x), dried (MgS04), filtered and treated with IN HCI ethereal solution to give the title compound. FAB MS (M+1) = 472.

Analysis calculated for C2gH33Ns02'0.35 EtOAc: C, 70.28; H, 7.18; N, 13.94; Found: C, 70.37; H, 7.29; N, 13.88.

EXAMPLE 24A Preparation of 2- 3- (3R-ethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-4- 1- (R or S) (+) amino-1-(3-methyl-3H-imidazol-4-yl)-ethyl-benzonitrile Step A: Preparation of (+) and (-)-3-ethyl-3- (3-methoxy-phenyl)- 1-methyl-azepan-2-one

Racemic 3-ethyl-3- (3-methoxy-phenyl)-l-methyl-azepan-2-one was prepared as described in Example 5, but substituting N-methylcaprolactam for caprolactam in Step A and quenching the reaction of Step B with ethyl iodide instead of water. This procedure is described in the literature (A. C. White et al., Eur. J Med.

Chem. Chimica Therapeutica, 15 [4], 375-385 (1980)). The racemic product was separated on an HPLC chiracel AD column eluting with 90: 10 hexane: ethanol to give R (+)-3-ethyl-3- (3-methoxy-phenyl)-l-methyl-azepan-2-one [a] D +59.8° (c =l, CCl4) and S (-)-3-ethyl-3- (3-methoxy-phenyl)-l-methyl-azepan-2-one [a] D-60.8° (c = 1, CC14).

Step B : Preparation of S (-) 3-ethyl-3- (3-hydroxy-phenyl)-1-methyl- azepan- 2-one 1 M BBr3 in CH2CI2 (77.7 mL, 0.0777 mol) was added to a solution of S (-)-3-Ethyl-3- (3-methoxy-phenyl)-1-methyl-azepan-2-one (as described above in Step A) (6.04 g, 0.026 mol) in CH2CI2 (400 mL) cooled to 0°C in an ice-H2O bath.

After 3.5 hours, the reaction was added to a saturated NaHC03 solution and the layers separated. The aqueous layer was washed with CH2CI2 (3 x 50 mL). The organic layers were combined, washed with brine and dried with Na2SO4. Filtration and concentration to dryness gave the title compound.

[a] D-43.0° (c=l, EtOH) R- (+)-3-ethyl-3- (3-hydroxy-phenyl)-1-methyl-azepan-2-one was prepared in a like manner using the enantiomer obtained in Step A. a D +46. 9° (c = l, EtOH) Step C: Preparation of 2-Fluoro-4- (3-methyl-3H-imidazole-4-carbonyl)- benzonitrile 2-Fluoro-4- hydroxy- (3-methyl-3H-imidazol-4-yl)-methyl- benzonitrile (as described in Example 17, Step E) (0.655 g, 2.83 mmol) and Mn02 (1.23 g, 14.2 mmol) were stirred in CH2CI2 (50 mL) and CH3CN (5 mL) for 3 h. The solution was filtered and concentrated to yield the title compound.

Step D: Preparation of 2-fluoro-4- 1-hydroxy-1- (3-methyl-3H-imidazol-4-yl)- ethyl-benzonitrile 2-Fluoro-4- (3-methyl-3H-imidazole-4-carbonyl)-benzonitn'le (as described above in Step C) (0.603 g, 2.63 mmol) was dissolved in anhydrous THF (30

mL). A solution of 3. OM MeMgBr in diethyl ether (2.55 mL, 7.65 mmol) was added and stirred for 15 min. The reaction was quenched with NH4CI solution, diluted with CH2C12 and saturated NaHC03 solution and seperated. The aqueous layer was back extracted with CH2CI2 (3X), the combined organic layers dried (MgS04) and concentrated to give the title compound.

Step E : Preparation of 2-fluoro-4- 1-amino-1- (3-methyl-3H-imidazol-4-yl)- ethyl-benzonitrile 2-Fluoro-4- I-hydroxy-1- (3-methyl-3H-imidazol-4-yl)-ethyl- benzonitrile (as described above in Step D) (1.88 g, 0.0078 mol) was added to SOCI ? (25 mL) in an ice-H20 bath, and stirred at RT for 45 min. The solution was concentrated in vacuo and azeotroped with anhydrous CH2CI2 (3X). The solid was dissolved in CH2Cl2 (20 mL) and cooled to-78°C. NH3 (g) was bubbled through the solution to double the volume, then the solution was stirred for 2 hours while warming to RT under Ar. The solution was concentrated in vacuo, and partitioned between H2O and CH2Cl2. The aqueous layer was back extracted with CH2CI2.. The organic layers were combined, and dried with MgS04. Filtration and concentration gave the title compound.

Step F: Preparation of (+) and (-)-2-fluoro-4- 1-amino-1- (3-methyl- 3H-imidazol-4-yl)-ethyll-benzonitrile Racemic 2-fluoro-4- 1-amino-1- (3-methyl-3H-imidazol-4-yl)-ethyl- benzonitrile (as described above in Step E) was separated on an HPLC chiracel OD column eluting with 60: 40 ethanol: hexane to give (+)-2-fluoro-4- 1-amino-1- (3- methyl-3H-imidazol-4-yl)-ethyl-benzonitrile [a] D + 99.6° (c =l, EtOH) and (-)-2- fluoro-4- 1-amino-1- (3-methyl-3H-imidazol-4-yl)-ethyl-benzonitrile [a] D-111.6° (c = 1, EtOH).

Step G : Preparation of a diastereomer of 2- 3- (3-ethyl-1-methyl-2-oxo-azepan- 3-yl)-phenoxy]-4- 1-amino-1- (3-methyl-3H-imidazol-4-yl)-ethyl- benzonitrile Following the methods described in Example 24, (3R)-Ethyl-3- (3- hydroxy-phenyl)-1-methyl-azepan-2-one and (+)-2-fluoro-4- 1-amino-1- (3-methyl- 3H-imidazol-4-yl)-ethyl-benzonitrile (as described above in Step F) were reacted to give 2- 3- (3R)-ethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-4- 1- (R or S) (+) amino-

1- (3-methyl-3H-imidazol-4-yl)-ethyl-benzonitrile.

FAB MS (M+1) = 472.

Analysis calculated for H2O: C, 69.58; H, 7.15; N, 14.49; Found: C, 69. 57; H, 6.90; N, 14.23.

EXAMPLE 24B Following the methods described in Example 24A, (3R)-Ethyl-3- (3- hydroxy-phenyl)-1-methyl-azepan-2-one and (-)-2-fluoro-4-[1-amino-1-(3-methyl-3H- imidazol-4-yl)-ethyl]-benzonitrile were reacted to give 2-[3-(3R)-ethyl-1-methyl-2- oxo-azepan-3-yl)-phenoxy-4- 1- (R or S) (-) amino-1- (3-methyl-3H-imidazol-4-yl)- ethyl-benzonitrile.

FAB MS (M+1) = 472.

Analysis calculated for C28H33N5O2: C, 71.31; H, 7.05; N, 14.85; Found: C, 71. 36; H, 6.95; N, 14.58.

EXAMPLE 24C Following the methods described in Example 24A, (3S)-Ethyl-3- (3- hydroxy-phenyl)-1-methyl-azepan-2-one and (+)-2-fluoro-4- 1-amino-1- (3-methyl- 3H-imidazol-4-yl)-ethyl-benzonitrile were reacted to give 2-[3-(3S)-ethyl-1-methyl- 2-oxo-azepan-3-yl)-phenoxy-4- 1- (R or S) (+) amino-1- (3-methyl-3H-imidazol-4-yl)- ethyl-benzonitrile.

FAB MS (M+1) = 472.

Analysis calculated for C28H33N502 0 20 H2O#0. 25 CH2CI2: C, 68.34; H, 6.88; N, 14.11; Found: C, 68.49; H, 7.00; N, 13.72.

EXAMPLE 24D Following the methods described in Example 24A, (3S)-Ethyl-3- (3- hydroxy-phenyl)-1-methyl-azepan-2-one and (-)-2-fluoro-4- [1-amino-1-(3-methyl-3H- imidazol-4-yl)-ethyl]-benzonitrile were reacted to give 2-[3-(3S)-ethyl-1-methyl-2-

oxo-azepan-3-yl)-phenoxy-4- 1- (R or S) (-) amino-1- (3-methyl-3H-imidazol-4-yl)- ethyl]-benzonitrile.

FAB MS (M+1) = 472.

Analysis calculated for C28H33Ns02'0.85 H20: C, 69.06; H, 7.18; N, 14.38; Found: C, 68.72; H, 6.92; N, 14.77.

EXAMPLE 24E Preparation of (-)-2-fluoro-4- [1-amino-1-(3-methyl-3H-imidazol-4-yl)-ethyl]- benzonitrile The following is an alternate way of preparing (-)-2-fluoro-4- 1-amino- 1- (3-methyl-3H-imidazol-4-yl)-ethyl-benzonitrile: Step A : Preparation of N-[(4-cyano-3-fluoro-phenyl)-(3-methyl-3H- imidazol-4-yl)-methylene-2-methylpropanesulfinamide 2-Fluoro-4- (3-methyl-3H-imidazole-4-carbonyl)-benzonitrile (as described in Example 24A, Step C) (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/CH2CI2) to give the title compound.

Step B: Preparation of N- 1-(4-cyano-3-fluoro-phenyl)-1-(3-methyl-3H- imidazol-4-yl)-ethyl-2-methylpropanesulfinamide N-[(4-cyano-3-fluoro-phenyl)-(3-methyl-3H-imidazol-4-yl)- methylene-2-methylpropanesulfinamide (as described above in Step B) (1.50 g, 4.51 mmol) was dissolved in anhydrous THF (30 mL) at 0°C to which a 3.0M solution of MeMgBr (4.50 mL, 13.5 mmol) in Et20 was added. The reaction was quenched with aq. NH4Cl solution, diluted with saturated NaHC03 solution and extracted with CH2Cl2 (3X). The combined organic layers were dried (MgS04), filtered, concentrated, and recrystallized from 95% EtOAc/Hexane to give the title compound.

Step C : Preparation of (-)-4- 1-amino-1- (3-methyl-3H-imidazol-4-yl)- ethyll-2-fluoro-benzonitrile bishydrochloride A cold methanolic HCI solution (50 mL) was added to N 1- (4-cyano- 3-fluoro-phenyl)-1- (3-methyl-3H-imidazol-4-yl)-ethyl- 2-methylpropanesulfinamide (as described above in Step B) (0.880 g, 2.51 mmol) dissolved in MeOH (50 mL) and stirred for lh at RT. After concentration and trituration with EtOAc the title compound was obtained as a bis HCI 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 A, (+)-2- fluoro-4- 1-amino-I- (3-methyl-3H-imidazol-4-yl)-ethyl-benzonitrile was obtained.

EXAMPLE 25 Preparation of the diastereomers of 2- 3-3 (R or S)-n-butyl-1-methyl-2-oxo-azepan-3- yl)-phenoxyl-4-l- (R or S) amino-I- (3-methyl-3H-imidazol-4-yl)-ethyll-benzonitrile Using the methods described in Example 24A, but substituting racemic 3-n-butyl-3- (3-methoxy-phenyl)-1-methyl-azepan-2-one (prepared as described in Example 6, Step A, but using n-butyl bromide in place of iodomethyl methylether), the following compounds were prepared, wherein the" (-)" and" (+)" represent the optical rotation of the starting intermediates that are combined to obtain the compounds below: 2- 3-3 (R or S) (+)-n-butyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-4- 1- (R or S) (+) amino-1- yl)-ethyl-benzonitrile FAB MS (M+1) = 500 2- 3-3 (R or S) (+)-n-butyl-l-methyl-2-oxo-azepan-3-yl)-phenoxy-4- I- (R or S) (-) amino-1- (3-methyl-3H-imidazol-4-yl)-ethyll-benzonitrile FABMS (M+1) =500 Analysis calculated for C3oH39N502'2.80 CF3CO2H: C, 52.21; H, 4.90; N, 8.55; Found: C, 52.19; H, 4.69; N, 8.34.

2- 3-3 (R or S) (-)-n-butyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-4- 1- (R or S) (-) amino-1- (3-methyl-3H-imidazol-4-yl)-ethylJ-benzonitrile

FAB MS (M+1) = 500 Analysis calculated for C3oH39N502-2. 50 CF3CO2H-1.0 H2O : C, 52.36; H, 5.21; N, 8.73; Found: C, 52.35; H, 4.82; N, 8.45.

2- 3-3 (R or S) (-)-n-butyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-4- 1- (R or S) (+) amino-1- (3-methyl-3H-imidazol-4-yl)-ethyll-benzonitrile FAB MS (M+1) = 500 EXAMPLE 25A Preparation of the diastereomers of 2-{3-[1-methyl-2-oxo-3-(R or S)- (3,3,3-trifluoro- propyl)-azepan-3-yl-phenoxy}-4- 1 (R or S)-amino-1- (3-methyl-3H-imidazol-4- yl)-ethyll-benzonitrile Using the methods described in Example 24A, but substituting racemic (3,3, 3-trifluoro-propyl)-3-(3-methoxy-phenyl)-1-methyl-azepan-2-o ne (prepared as described in Example 6, Step A, but using 3,3,3-trifluoro-propyl bromide in place of iodomethyl methylether), the following compounds were prepared, wherein the" (-)" and" (+)" represent the optical rotation of the starting intermediates that are combined to obtain the compounds below: 2- {3- l-methyl-2-oxo-3- (RorS) (+)- (3,3,3-trifluoro-propyl)-azepan-3-yl- phenoxy}-4-[1 (R or S) (+)-amino-1- (3-methyl-3H-imidazol-4-yl)-ethyl-benzonitrile FAB MS (M+1) = 540 2- {3- 1-methyl-2-oxo-3- (R or S) (-)- (3,3,3-trifluoro-propyl)-azepan-3-yl- phenoxyT-4-1 (R or S)(-)-amino-1-(3-methyl-3H-imidazol-4-yl)-ethyl]- benzonitrile FAB MS (M+1) = 540 2- {3- 1-methyl-2-oxo-3- (R or S) (+)- (3,3,3-trifluoro-propyl)-azepan-3-yl- phenoxy}-4- 1 (R or S) (-)-amino-1- (3-methyl-3H-imidazol-4-yl)-ethvll-benzonitrile FAB MS (M+1) = 540 2- {3- l-methyl-2-oxo-3- (RorS) (-)- (3,3,3-trifluoro-propyl)-azepan-3-yl- phenoxy}-4- 1 (R or S) (+)-amino-1- (3-methyl-3H-imidazol-4-yl)-ethyl-benzonitrile

FAB MS (M+1) = 540 EXAMPLE 26 Preparation of the diastereomers of 2- 3-3 (R or S)-cyclopropylmethyl-1-methyl- 2-oxo-azepan-3-yl)-phenoxy-4- 1- (R or S) amino-1- (3-methyl-3H-imidazol-4- yl)-ethyl]-benzonitrile Using the methods described in Example 24A, but substituting racemic as3-cyclopropylmethyl-3-(3-methoxy-phenyl)-1-methyl-azepan-2 -one(prepared described in Example 6, Step A, but using cyclopropylmethyl bromide in place of iodomethyl methylether), the following compounds were prepared, wherein the" (-)" and" (+)" represent the optical rotation of the starting intermediates that are combined to obtain the compounds below: 2- 3-3 (R or S) (-)-cyclopropylmethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-4- 1- (R or S) (+) amino-1- (3-methyl-3H-imidazol-4-yl)-ethyll-benzonitrile FAB MS (M+1) = 498 HRMS measured 498. 2866; theoretical 498.2863 2- 3-3 (R or S) (+)-cyclopropylmethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-4- 1- (R or S) (+) amino-1- (3-methyl-3H-imidazol-4-yl)-ethyl-benzonitrile FAB MS (M+1) = 498 HRMS measured 498.2851; theoretical 498. 2863 2- 3-3 (R or S) (-)-cyclopropylmethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy- 4- 1- (R or S) (+ amino-1- (3-methvl-3H-imidazol-4-yl)-ethyl-benzonitrile FAB MS (M+1) = 498 HRMS measured 498.2845; theoretical 498.2863 2- 3-3 (R or S) (+)-cyclopropylmethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-4- [1- (R or S) (-) amino-1- (3-methvl-3H-imidazol-4-vl)-ethyl-benzonitrile FAB MS (M+1) = 498 Analysis calculated for C30H35N5O2#0. 35 H20: C, H, 7.14; N, 13.90; Found: C, 71.37; H, 7.39; N, 14.28.

EXAMPLE 26A Preparation of the diastereomers of 2- 3-3 (R or S)-cyclopropylethyl-1-methyl-2-oxo- azepan-3-yl)-phenoxy-4- 1- (R or S) amino-1-(3-methyl-3H-imidazol-4-yl)-ethyl]- benzonitrile Using the methods described in Example 24A, but substituting racemic 3-cyclopropylethyl-3- (3-methoxy-phenyl)-l-methyl-azepan-2-one (prepared as described in Example 6, Step A, but using cyclopropylethyl bromide in place of iodomethyl methylether), the following compounds were prepared, wherein the" (-)" and" (+)" represent the optical rotation of the starting intermediates that are combined to obtain the compounds below : 2- 3-3 (R or S) (-)-cyclopropylethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-4- 1- (R or S) (+) amino-1- (3-methyl-3H-imidazol-4-yl)-ethyll-benzonitrile FAB MS (M+1) = 512 2- 3-3 (R or S) (+)-cyclopropylethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-4- 1- (R or S) (+) amino-1- (3-methyl-3H-imidazol-4-yl)-ethyll-benzonitrile FAB MS (M+1) = 512 2- 3-3 (R or S) (-)-cyclopropylethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-4- 1- (R or S) (+) amino-1- (3-methvl-3H-imidazol-4-yl)-ethy-benzonitrile FAB MS (M+1) =512 2- 3-3 (R or S) (+)-cyclopropylethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-4-1 -(R or S) (-) amino-1- (3-methvl-3H-imidazol-4-yl)-ethvll-benzonitrile FABFABMS (M+1) 512 EXAMPLE 26B

Preparation of the diastereomers of 2- 3-3 (R or S)-propyl-1-methyl-2-oxo-azepan-3- S)amino-1-(3-methyl-3H-imidazol-4-yl)-ethyl]-benzonitrileyl) -phenoxy]-4-[1-(Ror Using the methods described in Example 24A, but substituting racemic 3-propyl-3- (3-methoxy-phenyl)-1-methyl-azepan-2-one (prepared as described in Example 6, Step A, but using propyl bromide in place of iodomethyl methylether), the following compounds were prepared, wherein the" (-)" and" (+)" represent the optical rotation of the starting intermediates that are combined to obtain the compounds below: 2- 3-3 (R or S) (-)-propyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-4- 1- (R or S) (+) amino-1- (3-methyl-3H-imidazol-4-Yl)-ethl-benzonitrile FAB MS (M+1) = 486 2- 3-3 (R or S) (+)- propyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-4- 1- (R or S) (+) amino-1- (3-methyl-3H-imidazol-4-yl)-ethyl-benzonitrile FAB MS (M+1) = 486 2- 3-3 (R or S) (-)- propyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-4- 1- (R or S) (+) amino-1- (3-methvl-3H-imidazol-4-yl)-ethyll-benzonitrile FAB MS (M+1) = 486 2- 3-3 (R or S) (+)- propyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-4- 1- (R or S) (-) amino-1- (3-methyl-3H-imidazol-4-vl)-ethyl-benzonitrile FAB MS (M+1) = 486 EXAMPLE 26C Preparation of the diastereomers of 2- 3-3 (R or S)-methoxymethyl-1-methyl-2-oxo- azepan-3-yl)-phenoxy-4- 1- (R or S) amino-1- (3-methyl-3H-imidazol-4-yl)-ethyl- benzonitrile

Using the methods described in Example 24A, but substituting racemic 3-methoxymethyl-3- (3-methoxy-phenyl)-l-methyl-azepan-2-one (Example 6, Step A), the following compounds were prepared, wherein the" (-)" and" (+)" represent the optical rotation of the starting intermediates that are combined to obtain the compounds below: 2- 3-3 (R or S) (-)-methoxymethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-4- 1- (R or S) (+) amino-1-(3-methyl-3H-imidazol-4-yl)-ethyl-benzonitrile FAB MS (M+1) = 488 2- 3-3 (R or S) (+)-methoxymethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-4- 1- (R or S) (+) amino-1- (3-methyl-3H-imidazol-4-yl)-ethyl-benzonitrile FAB MS (M+1) = 488 2- 3-3 (R or S) (-)-methoxymethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-4- 1- (R or S) (+) amino-1- (3-methyl-3H-imidazol-4-yl)-ethyl-benzonitrile FAB MS (M+1) = 488 2- 3-3 (R or S) (+)-methoxymethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-4- 1- (R or S)(-)amino-1-(3-methyl-3H-imidazol-4-yl)-ethyl]-benzonitrile FAB MS (M+1) = 488 EXAMPLE 26D Preparation of the diastereomers of 2- 3-3 (R or S)-ethoxymethyl-1-methyl-2-oxo- azepan-3-yl)-phenoxy-4- 1- (R or S) amino-1- (3-methyl-3H-imidazol-4-yl)-ethyl- benzonitrile Using the methods described in Example 24A, but substituting racemic 3-cyclopropylethyl-3- (3-methoxy-phenyl)-l-methyl-azepan-2-one (prepared as described in Example 6, Step A, but using bromomethyl ethylether in place of

iodomethyl methylether), the following compounds were prepared, wherein the"(-)" and" (+)" represent the optical rotation of the starting intermediates that are combined to obtain the compounds below: 2- 3-3 (R or S) (-)-ethoxymethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-4- 1- (R or S) (+) amino-1-(3-methyl-3H-imidazol-4-yl)-ethyl-benzonitrile FAB MS (M+1) = 502 2- 3-3 (R or S) (+)-ethoxymethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-4- 1- (R or S) (+) amino-1- (3-methyl-3H-imidazol-4-yl)-ethvl-benzonitrile FAB MS (M+1) = 502 2- 3-3 (R or S) (-)-ethoxymethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-4- 1- (R or S)(+)amino-1-(3-methyl-3H-imidazol-4-yl)-ethyl]-benzonitrile FAB MS (M+1) = 502 2- 3-3 (R or S) (+)-ethoxymethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-4- 1- (R or S) (-) amino-1- (3-methyl-3H-imidazol-4-yl)-ethyl-benzonitrile FAB MS (M+1) = 502 EXAMPLE 27 Preparation of the diastereomers of 2- 3-3 (R or S)-cycloproylmethyl-1-methyl-2-oxo- piperidin-3-yl)-phenoxy-4- 1- (R or S) amino-1- (3-methyl-3H-imidazol-4-yl)-ethyl- benzonitrile Using the methods described in Example 24A, but substituting racemic 3-cyclopropylmethyl-3- (3-methoxy-phenyl)-1-methyl-piperidin-2-one (prepared using techniques described in Example 4, Step A, and in Example 6, Step A, but using N-methyl-2-piperidinone instead of N-methylcaprolactam in Example 4 and cyclo- propylmethyl bromide in place of iodomethyl methylether in Example 6), the following compounds were prepared, wherein the" (-)" and" (+)" represent the optical rotation of the starting intermediates that are combined to obtain the compounds below:

2- 3-3 (R or S) (-)-cyclopropylmethyl-1-methyl-2-oxo-piperidin-3-yl)-phenoxy -4- 1- (R or S) (+) amino-1- (3-methvl-3H-imidazol-4-yl)-ethyl-benzonitrile FAB MS (M+1) = 484 2- 3-3 (R or S) (+)-cyclopropylmethyl-l-methyl-2-oxo-piperidin-3-yl)-phenoxy -4- 1- (R or S) (+) amino-1- (3-methyl-3H-imidazol-4-yl)-ethyl-benzonitrile FAB MS (M+1) = 484 2- 3-3 (R or S) (-)-cyclopropylmethyl-l-methyl-2-oxo-piperidin-3-yl)-phenoxy -4- 1- (R or S) (-) amino-1- (3-methyl-3H-imidazol-4-yl)-ethyll-benzonitrile FAB MS (M+1) = 484 2- 3-3 (R or S) (+)-cyclopropylmethyl-l-methyl-2-oxo-piperidin-3-yl)-phenoxy -4- 1- (R or S) (-) amino-1- (3-methyl-3H-imidazol-4-yl)-ethvl-benzonitrile FAB MS (M+1) = 484 EXAMPLE 27A Preparation of the diastereomers of 2- 3-3 (R or S)-benzyl-1-methyl-2-oxo-piperidin- 3-yl)-phenoxy-4- 1- (R or S) amino-1- (3-methyl-3H-imidazol-4-yl)-ethyl- benzonitrile Using the methods described in Example 24A, but substituting racemic 3-benzyl-3- (3-methoxy-phenyl)-1-methyl-piperidin-2-one (prepared using techniques described in Example 4, Step A, and in Example 6, Step A, but using N-methyl-2- piperidinone instead of N-methylcaprolactam in Example 4 and benzyl bromide in place of iodomethyl methylether in Example 6), the following compounds were prepared, wherein the" (-)" and" (+)" represent the optical rotation of the starting intermediates that are combined to obtain the compounds below: 2- 3-3 (R or S) (-)-benzyl-1-methyl-2-oxo-piperidin-3-yl)-phenoxy-4- 1- (R or S) (+) amino-1-(3-methvl-3H-imidazol-4-yl)-ethyll-benzonitrile (M+1)=520FABMS

2- 3-3 (R or S) (+)-benzyl-1-methyl-2-oxo-piperidin-3-yl)-phenoxy-4- 1- (R or S) (+) amino-1-!-ethyl-benzonitrile FAB MS (M+1) = 520 2- 3-3 (R or S) (-)-benzyl-1-methyl-2-oxo-piperidin-3-yl)-phenoxy-4- 1- (R or S) (-) amino-1- (3-methyl-3H-imidazol-4-vl)-ethyl-benzonitrile FAB MS (M+1) = 520 2- 3-3 (R or S) (+)-benzyl-1-methyl-2-oxo-piperidin-3-yl)-phenoxy-4- 1- (R or S) (-) amino-1- (3-methyl-3H-imidazol-4-yl)-ethyl-benzonitrile FAB MS (M+1) = 520 EXAMPLE 28 Preparation amino-2-[3-methyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy]-4-[1- 1-(3-methyl-3H-imidazol-4-Yl)-ethyll-benzonitrile Using the methods described in Example 24A, but substituting racemic 3-methyl-3- (3-hydroxy-phenyl)-1-methyl-azepan-2-one (prepared as described in Example 4, Step A and in Example 6, Step A, but using methyl iodide in place of iodomethyl methylether), and racemic 2-fluoro-4-[1-amino-1-(3-methyl-3H-imidazol- 4-yl)-ethyl]-benzonitrile (Example 24A, Step F), the following compound was prepared: 2-3-methyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-4- 1-amino-1- (3-methyl-3H- imidazol-4-yl)-ethyll-benzonitrile FAB MS (M+1) = 458 EXAMPLE 29 Preparation of 2- 3- (3-ethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-4- 1- (3- methyl-3-H-imidazol-4-yl)-vinyl]-benzonitrile Step A: Preparation of 2-fluoro-4-formylbenzonitrile

2-Fluoro-4-hydroxymethylbenzonitrile (as described in Example 9, Step C) (10 g, 0,066 mol) and triethylamine (32.3 mL, 0.231 mol) were dissolved in CH2CI2 (100 mL)-DMSO (20 mL) at < 5°C with stirring and treated dropwise with a solution of pyridineSO3 complex (31.5 g, 0.198 mol) in DMSO (70 mL). The reaction mixture was maintained at a temperature <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 CH2CI2 and H20. The organic layer was separated, washed well with H20, brine, and dried (Na2SO4). Filtration and concentration gave the title compound after purification by chromatography (silica gel, hexane: EtOAc, 3: 1).

1H NMR (CDCl3) 8 10.06 (d, 1H, J = 2 Hz), 7.86 (dd, 1H, J = 5,8 Hz), 7.798 (dd, 1H, 1,8Hz),7.728(dd,1H,J=1,8Hz).J= Step B: Preparation of 2-fluoro-4- hydroxy- (1-trityl-1H-imidazol-4-yl)- methyl-benzonitrile To a solution of 4-iodo-1-trityl-1H-imidazole (5.00 g, 11.5 mmol) in anhydrous CH2Cl2 (30 mL) was added a 3.0M solution of ethylmagnesium bromide (6.58 mL, 19.7 mmol) with stirring under Ar. After 3h, the reaction mixture was cooled to-78°C and a solution of 2-fluoro-4-formyl-benzonitrile (as described in Step A above) (1.70g, 11.5 mmol) dissolved in CH2CI2 (20 mL) was added dropwise.

The reaction was allowed to warm to RT over 2h, quenched with saturated NH4Cl solution, diluted with satd. NaHC03 solution to pH=8.5, and extracted with CH2CI2 (3X). The combined organic layers were dried (MgSO4), concentrated and purified using Si02 chromatography (0-1% MeOH/CH2Cl2) to yield the title compound.

Step C : Preparation of acetic acid (4-cyano-3-fluoro-phenyl)- (I-trityl-lH- imidazol-4-yl)-methyl ester 2-Fluoro-4-[hydroxy-(1-trityl-lH-imidazol-4-yl)-methyl]-benz onitrile (as described in Step B above) (4.05 g, 8.81 mmol), pyridine (2.14 mL, 26.4 mmol), and acetic anhydride (12.5 mL, 132 mmol) were stirred in anhydrous DMF (60 mL) for 3h under Ar. The reaction was concentrated in vacuo, diluted with EtOAc (250 mL), washed with H20 (2X), brine, dried (MgSO4) and concentrated to give the title compound.

Step D: Preparation of acetic acid (4-cyano-3-fluoro-phenyl)- (3-methyl-3H- imidazol-4yl)-methvl ester _ Acetic acid (4-cyano-3-fluoro-phenyl)-(1-trityl-lH-imidazol-4-yl)- methyl ester (as described in Step C above) (4.60 g, 9.17 mmol) and dimethyl sulfate (0.83 mL, 8.81 mmol) were dissolved in EtOAc (20 mL) and heated at 60°C overnight under Ar. The reaction was concentrated in vacuo, diluted with MeOH (30 mL), and refluxed for I hr, concentrated in vacuo and purified using Si02 chromatography (0.5-4% MeOH/CH2Cl2 with NH40H) to give the title compound.

Step E: Preparation of 2-fluoro-4- hydroxy- (3-methyl-3H-imidazol- 4-vl)-methvl-benzonitrile Acetic (4-cyano-3-fluoro-phenyl)-(3-methyl-3H-imidazol-4-yl)- methyl ester (as described in Step D above) (1.26 g, 4.59 mmol) and NaOH (5.5 mL, 5.5 mmol) were dissolved in THF (15 mL) and H20 (25 mL). After Ih, the reaction was diluted with satd. NaHC03 solution, extracted with CH2CI2 (3X), dried (MgSO4) and concentrated to give the title compound.

Step 2-fluoro-4-[1-(3-methyl-3-H-imidazol-4-yl)-Preparation vinyl-benzonitrile Thionyl chloride (15 mL) was added to a suspension of 2-fluoro-4- hydroxy- (3-methyl-3H-imidazol-4-yl)-methyl-benzonitrile (as described in Step E above) (0.268 g) in CHCl3 (10 mL)-benzene (30 mL) and the reaction mixture was heated at reflux for 2 hr. The reaction mixture was concentrated to dryness, dissolved in CH2C12, and re-concentrated to give the title compound.

FAB MS (M+1) 228.

1H NMR (CDCl3) # 7.81 (s, IH), 7.15-7.23 (m, 1H), 7.11 (s, 1H), 5.85 (s, 1H), 5.63 (s, 1H), 3.40 (s, 3H).

Step of2-[3-(3-ethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy]-Prepara tion 4-[1-(3-methyl-3-H-imidazol-4-yl)-vinyl]-benzonitrile Using the procedure described in Example 3, the title compound was prepared starting with 2-fluoro-4- 1- (3-methyl-3-H-imidazol-4-yl)-vinyl-benzonitrile (as described in Step F above) and 3-ethyl-3-(3-hydroxy-phenyl)-1-methyl-azepan-2- one.

FAB mass spectrum m/e 455 (m+1).

Analysis calculated for C28H30 N402 0.70 H2O 1.30 HCl : C, 65.35; H, 6.41; N, 10.89; Found: C, 65.36; H, 6.41; N, 10.53.

EXAMPLE 30 Preparation of 2- 3- (3-ethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-4- 1- (3-methyl- 3-H-imidazol-4-vl)-cvclopropvll-benzonitrile Step A : Preparation of 2-fluoro-4- 1- (3-methyl-3-H-imidazol-4-yl)- cyclopropyll-benzonitrile To a suspension of sodium hydride (60% dispersion) (0.040 g, 1.05 mmol) in dry DMF (2 mL) at ambient temperature under Ar was added trimethyl- sulfoxonium iodide (0.230 g, 1.05 mmol) in one portion with stirring. After 5 min, H2 evolution ceased, and after 15 min, a solution of 2-fluoro-4- 1- (3-methyl-3-H- imidazol-4-yl)-vinyl]-benzonitrile (as described above in Example 29, Step F) (0.240 g, 1.0 mmol) in DMF (4 mL) was added and the mixture was stirred at room temperature for 18 hr. The reaction mixture was partitioned between diethyl ether and ice water, the aqueous layer washed with ether, the organic layers combined, washed with brine, and dried (Na2SO4). Filtration and concentration to dryness gave the title compound after purification by RPLC on a Waters PrepPak column with a H20/CH3CN gradient.

FAB MS (M+1) 242.

Step B: Preparation of 2- 3- (3-ethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy- 4- 1-(3-methYl-3-H-imidazol-4-yl !-cyclopropyll-benzonitrile Using the procedure described in Example 3, the title compound was prepared starting with 2-fluoro-4-[1-(3-methyl-3-H-imidazol-4-yl)-cyclopropyl]- benzonitrile (as described in Step A above) (0.050 g, 0.21 mmol) and 3-ethyl-3- (3- hydroxy-phenyl)-1-methyl-azepan-2-one (0.078 g, 0.21 mmol).

FAB mass spectrum m/e 469 (M+1).

Analysis calculated for C29H32N402'1.00 H20: C, H, 7.04; N, 11.51; Found: C, 71.35; H, 6.72; N, 11.49.

EXAMPLE 31 Preparation of 2- 3-NN-dimethylaminomethyl-l-methyl-2-oxo-azepan-3-yl)- phenoxyl-4- 1-hydroxv- (3-methvl-3H-imidazol-4-yl)-ethyl-benzonitrile Step A : Preparation of3-bromomethyl-3- (3-hydroxy-phenyl)-l-methyl- azepan-2-one 3-Hydroxymethyl-3- (3-hydroxy-phenyl)-1-methyl-azepan-2-one (as described in Example 6, Step B) (0.60 g, 2.40 mmol), triphenylphosphine (0.94 g, 3.60 mmol), and carbon tetrabromide (1.19 g, 3.60 mmol) were dissolved in CH2C12 (15 mL), stirred under Ar for 16h, then the solution was concentrated in vacuo and purified using SiO2 chromatography (0.5% MeOH/CH2CI2) to give the title compound.

Step B : Preparation of 3-N, N-dimethylaminomethyl-3- (3-hydroxy-phenyl)- 1-methyl-azepan-2-one 3-Bromomethyl-3- (3-hydroxy-phenyl)-l-methyl-azepan-2-one (as described above in Step A) (0.263 g, 0.842 mmol) was dissolved in DMF (5 mL) in a sealed vessel. The solution was cooled to-78°C and saturated with dimethyl amine gas. The reaction vessel was sealed and heated at 80°C for 16h. The solution was concentrated to give the title compound.

Step C : Preparation of 2- 3-N, N-dimethylaminomethyl-1-methyl-2-oxo- azepan-3-yl)-phenoxy]-4- l-hydroxy-l- (3-methyl-3H-imidazol-4-yl)- ethyll-benzonitrile 2-Fluoro-4- 1-hydroxy-1- (3-methyl-3H-imidazol-4-yl)-ethyl- benzonitrile (as described in Example 24A, Step D) and 3-N, N-dimethylaminomethyl- 3- (3-hydroxy-phenyl)-l-methyl-azepan-2-one (as described above in Step B) were reacted as described in EXAMPLE 17, Step F, to give the title compound as its trifluoroacetate salt after purification by RPLC.

FAB MS (M+1) = 502 HRMS measured 502.2824; theoretical 502.2812 EXAMPLE 32

Preparation of 2- 3-N, N-dimethylaminomethyl-1-methyl-2-oxo-azepan-3- yl)-phenoxy-4- 1-amino-l- (3-methyl-3H-imidazol-4-yl)-ethyl'-benzonitrile Following the procedure described in Example 24A but using 2- 3- N, N-dimethylaminomethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-4- 1-hydroxy-1- (3-methyl-3H-imidazol-4-yl)-ethyl-benzonitrile (as described in Example 31) as the starting material, the title compound was prepared.

FAB mass spectrum m/e 501 (M+1).

Analysis calculated for C29H36N602'0. 05 H20'0.55 CH2Cl2: C, 64.73; H, 6.84; N, 15.33; Found: C, 64.79; H, 6.50; N, 15.13.

EXAMPLE 33 Preparation of 2- 3- (3-N, N-dimethylaminomethyl-1-methyl-2-oxo-azepan- 3-yl)-phenoxy]-4-[1-(3-methyl-3-H-imidazol-4-yl)-vinyl]-benz onitrile From the methods described in Example 32, the title compound was obtained as a byproduct.

FAB MS (M+1) = 484 HRMS measured484.2730; theoretical 484.2707 EXAMPLE 34 Preparation of 4- 5- (2-N, N-dimethylaminoethyl)-2-methyl-imidazol-1-ylmethyl-2- 3- (3 (S)-ethyl-1-methyl-2-oxo-azepan-3-vl)-phenoxy-benzonitrile bis trifluoroacetate Step A : Preparation of 4- 5- (2-amino-ethyl)-2-methyl-imidazol-1-ylmethyl- 2-fluoro-benzonitrile dihydrochloride A solution of {2- 3- (4-cyano-3-fluoro-benzyl)-2-methyl-3H-imidazol- 4-yl-ethyl}-carbamic acid tert-butyl ester (as described in Example 11, Step A) (1.0 g, 2.9 mmol) in EtOAc (30 mL) was cooled to-20°C and saturated with HCI gas. The cooling bath was removed and the reaction was stirred for 2 hr. The solvent was removed in vacuo to obtain the title compound.

Step B: Preparation of 4- 5- (2- (N, N-dimethylaminoethyl)-2-methyl- imidazol-1-vlmethyl-2-fluoro-benzonitrile To a solution of 4- 5- (2-amino-ethyl)-2-methyl-imidazol-1-ylmethyl- 2-fluoro-benzonitrile dihydrochloride (as described above in Step A) (0.5 g, 1.65 mmol) in MeOH (5 mL) at pH = 4 was added paraformaldehyde (300 mg) and NaCNBH3 (0.311 g, 4.95 mmol). The reaction mixture was stirred for 2 h then additional NaCNBH3 (0.311 g) was added. After 18 h, more NaCNBH3 was added and stirring was continued for 24 h. The MeOH was removed in vacuo and the residue was partitioned between EtOAc and saturated NaHC03. The organic layer was washed with brine and dried (MgS04). Filtration and removal of EtOAc in vacuo gave the title compound as an oil.

Step C: Preparation of 4- 5- (2-N, N-dimethylaminoethyl)-2-methyl- imidazol-1-ylmethyl-2- 3- (3 (S)-ethyl-1-methyl-2-oxo-azepan- 3-yl)-phenoxyl-benzonitrile bis trifluoroacetate To a solution of 4- 5- (2- (N, N-dimethylaminoethyl)-2-methyl-imidazol- 1-ylmethyl-2-fluoro-benzonitrile (as described above in Step B) (0.54 g, 1.86 mmol) in DMF (5 mL) was added 3 (S)-ethyl-3- (3-hydroxy-phenyl)-l-methyl-azepan-2-one (0.80 g, 0.323 mmol) and Cs2C03 (0.96 g, 2.95 mmol). The mixture was heated for 42 hr at 60°C, then partitioned with EtOAc and saturated NaHC03. The organic layer was separated, washed with H2O, brine and dried (MgS04). Filtration and removal of the EtOAc in vacuo the title compound as its bis trifluoroacetate salt after purification by preparative HPLC eluting with 95: 5 to 5: 95 gradient of 0. 1 % TFA/H20: 0. 1% TFA/CH3CN.

FAB mass spectrum m/e 514 (m+1).

EXAMPLE 35 Preparation of 2- (3- {4-cyano-3- 3- (3S-ethyl-1-methyl-2-oxo-azepan-3-yl)- phenoxv-benzyl}-2-methyl-3H-imidazol-4-yl)-ethyl-urea Step A: Preparation of {2- 3- (4-cyano-3-fluoro-benzyl)-2-methyl-3H- imidazol-4-YI-ethylT-tert-butyl urea

To a solution of 4- 5- (2-amino-ethyl)-2-methyl-imidazol-1-ylmethyl- 2-fluoro-benzonitrile dihydrochloride (as described in Example 34, Step A) (0.3 g, 0.97 mmol) in DMF (5.0 mL) was added NEt3 (0.41 mL, 2.97 mmol) and t-butyl- isocyanate (0.125 mL, 1.07 mmol). After 1/2 hr the DMF was removed in vacuo and the residue partitioned between EtOAc and saturated NaHC03. The organic layer was separated, washed with brine and dried (MgS04). Filtration and concentration in vacuo gave the title compound as an oil.

Step B : Preparation of {2- 3- (4-cyano-3-fluoro-benzyl)-2-methyl-3H- imidazol-4-yl-ethvl}-urea {2- 3- (4-cyano-3-fluoro-benzyl)-2-methyl-3H-imidazol-4-yl-ethyl}- tert-butyl urea (as described above in Step A) (0.41 g) was dissolved in TFA/CH2CI2 3: 1 (5.0 mL) and stirred at ambient temperature for 72 hr. The solvents were removed in vacuo to obtain the title compound.

Step C : Preparation of 2- (3- {4-cyano-3- 3- (3S-ethyl-1-methyl-2-oxo- azepan-3-yl)-phenoxy-benzyl}-2-methyl-3H-imidazol-4-yl)- ethyl-urea To a solution of {2- 3- (4-cyano-3-fluoro-benzyl)-2-methyl-3H- imidazol-4-yl-ethyl}-urea (as described above in Step B) (0.15 g, 0.52 mmol) in DMF (5 mL) was added 3 (S)-ethyl-3- (3-hydroxy-phenyl)-l-methyl-azepan-2-one (0.80 g, 0.323 mmol) and Cs2CO3 (0.96 g, 2.95 mmol). The mixture was heated for 18 hr at 60°C. The solution was partitioned with EtOAc and saturated NaHC03.

The organic layer was washed with H2O, brine and dried (MgS04). Filtration and concentration to dryness gave the title compound after purification by preparative RP HPLC.

FAB mass spectrum m/e 529 (M+1).

Analysis calculated for C30H35N6O3#0. 10 H2O 0.4 CH2Cl2: C, 64.68; H, 6.61; N, 14.89; Found: C, 64.67; H, 6.45; N, 14.36.

EXAMPLE 36 Preparation of 2- 3- (3-ethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-4- pyridin-N-oxide-3-ylmethvl-benzonitrile

To a solution of 2- 3- (3-ethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy- 4-pyridin-3-ylmethyl-benzonitrile (as described in Example 21) (0.06 g, 0.137 mmol) in CH2Cl2 (5.0 mL) was added MCPBA (0.07 g, 0.41 mmol) and the reaction was stirred for 1 hr. The reaction mixture was partitioned between EtOAc and saturated NaHC03. The organic layer was washed with brine and dried (MgS04). Filtration and concentration to dryness gave the title compound after purification by preparative RP HPLC.

FAB mass spectrum m/e 456 (M+1).

Analysis calculated for C2gH29N303 0.30 H2O 1 4 CH2C12: C, 60.89; H, 5.63; N, 7.25; Found: C, 60.90; H, 5.68; N, 6.55.

EXAMPLE 37 Preparation of 2- 3- (1-methyl-2-oxo-pyrrolidin-3-yl)-phenoxy-4- 1-amino-1- (3- methyl-3H-imidazol-4-yl)-ethyll-benzonitrile Using the methods described in Example 24A, but substituting racemic 3- (3-methoxy-phenyl)-l-methyl-pyrrolidin-2-one (prepared as described in Example 4, Step A, but using N-methyl-2-pyrrolidinone instead of N-methylcaprolactam), the title compound was prepared.

FAB mass spectrum m/e 416 (M+1).

Similarly, using 3- (3-hydroxy-phenyl)-l-methyl-azepan-2-one (Example 4, Step B) in the methods described in Example 24A, the following compound was prepared: 2-[3-(1-methyl-2-oxo-azepan-3-yl)-phenoxy]-4-[1-amino-1-(3-m ethyl-3H-imidazol-4- yl)-ethyl-benzonitrile HRMS theoretical: 444.2393, measured: 444.2399 Analysis calculated for C26H31N502'0. 25 H20'1.15 CF3CO2H: C, 60.89; H, 5.63; N, 7.25.

Found: C, 60.90; H, 5.68; N, 6.55.

Using 3-hydroxymethyl-3- (3-hydroxy-phenyl)-1-methyl-azepan-2-one (Example 6, Step B) in the methods described in Example 24A, the following compound was prepared: 2- 3- (3-hydroxymethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-4- 1-amino-1- (3- methyl-3H-imidazol-4-yl)-ethyl-benzonitrile HRMS theoretical: 474.2500, measured: 474.2498: Analysis calculated for C27H33N503-0.10 H2O 0.40 CF3CO2H: C, 50.99; H, 4.52; N, 9.35.

Found: C, 51.00; H, 4.50; N, 9.39.

EXAMPLE 38 Preparation of2- 3- (3-cyclopropylmethyl-l-methyl-2-oxo-pyrrolidin-3-yl)-phenoxy - 4- 1-amino-1- (3-methyl-3H-imidazol-4-yl)-ethyll-benzonitrile Using the methods described in Example 24A, but substituting racemic 3- (3-methoxy-phenyl)-l-methyl-pyrrolidin-2-one (prepared using techniques described in Example 4, Step A, and in Example 6, Step A, but using N-methyl-2- pyrrolidinone instead of N-methylcaprolactam in Example 4 and using cyclopropylmethyl bromide in place of iodomethyl methylether in Example 6), the title compound was prepared.

FAB mass spectrum m/e 470 (M+1).

Analysis calculated for C28H3 lN502-1. 00 H20-2.5 CF3CO2H: C, 51.29; H, 4.63; N, 9.06; Found: C, H, 4.42; N, 8.93.

EXAMPLE 38A Preparation of the diastereomers of 2- 3-3 (R or S)-cyclopropyl-1-methyl-2-oxo- pyrrolidin-3-yl)-phenoxy-4- 1- (R or S) amino-1- (3-methyl-3H-imidazol-4-yl)-ethyl- benzonitrile Using the methods described in Example 24A, but substituting racemic 3-cycloproylmethyl-3- (3-methoxy-phenyl)-1-methyl-pyrrolidin-2-one (prepared using techniques described in Example 4, Step A, and in Example 6, Step A, but using

N-methyl-2-pyrrolidinone instead of N-methylcaprolactam in Example 4 and cyclopropylmethyl bromide in place of iodomethyl methylether in Example 6), the following compounds were prepared, wherein the" (-)" and" (+)" represent the optical rotation of the starting intermediates that are combined to obtain the compounds below: 2-[3-3(Ror S)(-)-cyclopropylmethyl-1-methyl-2-oxo-pyrrolidin-3-yl)-phen oxy]-4-[1- (R or S) (+) amino-1- (3-methyl-3H-imidazol-4-yl)-ethl-benzonitrile FAB MS (M+1) = 470 2- 3-3 (R or S) (+)-cyclopropylmethyl-1-methyl-2-oxo-pyrrolidin-3-yl)-phenox y-4- 1- (R or S) (+) amino-1- (3-methyl-3H-imidazol-4-yl)-ethyll-benzonitrile FAB MS (M+1) = 470 2- 3-3 (R or S) (-)-cyclopropylmethyl-1-methyl-2-oxo-pyrrolidin-3-yl)-phenox y-4- 1- (R or S) (-) amino-1-!-ethyl-benzonitrile FAB MS (M+1) = 470 2- 3-3 (R or S) (+)-cyclopropylmethyl-1-methyl-2-oxo-pyrrolidin-3-yl)-phenox y-4- 1- (R or S) (-) amino-1- (3-methyl-3H-imidazol-4-yl)-ethvl-benzonitrile FAB MS (M+1) = 470 EXAMPLE 38B Preparation of the diastereomers of 2- 3-3 (R or S)-benzyl-1-methyl-2-oxo-pyrrolidin- 3-yl)-phenoxy-4- 1- (R or S) amino-1- (3-methyl-3H-imidazol-4-yl)-ethyl- benzonitrile Using the methods described in Example 24A, but substituting racemic 3-benzyl-3- (3-methoxy-phenyl)-l-methyl-pyrrolidin-2-one (prepared using techniques described in Example 4, Step A, and in Example 6, Step A, but using N-methyl-2- pyrrolidinone instead of N-methylcaprolactam in Example 4 and benzyl bromide in

place of iodomethyl methylether in Example 6), the following compounds were prepared, wherein the" (-)" and" (+)" represent the optical rotation of the starting intermediates that are combined to obtain the compounds below: 2- 3-3 (R or S) (-)-benzyl-1-methyl-2-oxo-pyrrolidin-3-yl)-phenoxy-4- 1- (R or S) (+) amino-1- (3-methyl-3H-imidazol-4-yl)-ethyl-benzonitrile FAB MS (M+1) = 506 2- [3-3 (R or S) (+)-benzyl-l-methyl-2-oxo-pyrrolidin-3-yl)-phenoxy-4- l- (R or S) (+) amino-1- (3-methyl-3H-imidazol-4-vl)-ethyll-benzonitrile FAB MS (M+1) = 506 2- 3-3 (R or S) (-)-benzyl-1-methyl-2-oxo-pyrrolidin-3-yl)-phenoxy-4- 1- (R or S) (-) amino-1- (3-methyl-3H-imidazol-4-yl)-ethyl-benzonitrile FAB MS (M+l) = 506 2- 3-3 (R or S) (+)-benzyl-1-methyl-2-oxo-pyrrolidin-3-yl)-phenoxy-4- 1- (R or S) (-) amino-1- (3-methyl-3H-imidazol-4-yl)-ethyll-benzonitrile FAB MS (M+1) = 506 EXAMPLE 39 Preparation of 4- 1- (4-allyl-5-bromo-pyridin-3-yl)-1-hydroxy-ethyl-2- 3- (3- ethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy]-benzonitriletrifl uoroacetate Step A: Preparation of 4-allyl-3n5-dibromo-pyridine A THF (100 mL) solution of 3, 5-dibromopyridine (5 g, 21.1 mmol) at-78°C was added via cannula to a solution of LDA (22.2 mmol) in THF (700 mL) at-78°C keeping the temperature <60°C. After stirring for 10 min. at-78°C, allyl bromide (2.56 mL, 29.5 mmol) was added via syringe and the reaction mixture was stirred for 2 hr at-78°C. Saturated NH4Cl (100 mL) was added slowly to quench the reaction. The organic layer was separated, washed with brine and dried (MgSO4).

Filtration and concentration in vacuo gave the title compound as an oil which was purified by chromatography on silica gel with hexane: ethyl acetate 9: 1.

Step B : Preparation of 4- (4-allyl-5-bromo-pyridin-3-yl)-hydroxy-methyl- 2-fluoro-benzonitrile To a solution of 4-allyl-3, 5-dibromo-pyridine (as described above in Step A) (2.35 g, 8.48 mmol) in THF (100 mL) at-100°C was added n-BuLi (8.48 mmol), and the mixture was stirred for 5 min. 2-Fluoro-4-formyl-benzonitrile (as described in Example 17, Step A) (1.39 g, 9.33 mmol) in THF (5 mL) was added dropwise via syringe. The reaction mixture was stirred at-78°C for 1 hr, quenched with saturated NH4C1 (50 mL), and partitioned between EtOAc and saturated NaHC03. The organic layer was washed with brine and dried (MgS04). Filtration and concentration to dryness gave the title compound which was purified by chromatography on silica gel with hexane: ethyl acetate 7: 3-5: 5.

Step C: Preparation of 4- (4-allyl-5-bromo-pyridine-3-carbonyl)-2- fluoro-benzonitrile To a solution of 4- (4-allyl-5-bromo-pyridin-3-yl)-hydroxy-methyl-2- fluoro-benzonitrile (as described above in Step B) (0.42 g, 1.21 mmol) in CH2CI2 (50 mL) was added Mn02 (1.58 g, 18.1 mmol). After 6 hr the reaction was filtered and the CH2CI2 was removed in vacuo to obtain the title compound.

Step D : Preparation of 4- 1- (4-allyl-5-bromo-pyridin-3-yl)-1-hydroxy- ethyl-2-fluoro-benzonitrile To a solution of 4- (4-allyl-5-bromo-pyridine-3-carbonyl)-2-fluoro- benzonitrile (as described above in Step C) (0.39 g, 1.13 mmol) in THF (20 mL) at 0°C was added methylmagnesium bromide (1.13 mmol, 3M) and the reaction was monitored by TLC. Three more portions (1.13 mmol) of CH3MgBr were added until the reaction was complete. The reaction was quenched with saturated NH4CI (20 mL), then partitioned between EtOAc and saturated NaHC03. The organic layer was washed with brine and dried (MgS04). Filtration and concentration to dryness gave the title compound after purification by chromatography on silica eluting with hexane: ethyl acetate 7: 3.

Step E: Preparation of 4- 1- (4-allyl-5-bromo-pyridin-3-yl)-1-hydroxy-ethyl-

2- 3- (3-ethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-benzonitrile trifluoroacetate To a solution of 4- 1- (4-allyl-5-bromo-pyridin-3-yl)-1-hydroxy-ethyl- 2-fluoro-benzonitrile (as described above in Step D) (0.1 g, 0.277 mmol) in DMF (3 mL) was added 3 (S)-ethyl-3- (3-hydroxy-phenyl)-1-methyl-azepan-2-one (0.068 g, 0.277 mmol) and Cs2C03 (0.25 g, 0.831 mmol) and the mixture was heated at 60°C for 16 hr. The DMF was removed in vacuo and the residue was partition with EtOAc and saturated NaHC03. The organic layer was separated, washed with brine and dried (MgS04). Filtration and concentration to dryness gave the title compound as the trifluoroacetate salt after purification by RP HPLC.

FAB mass spectrum m/e 703 (M+1).

EXAMPLE 40 Preparation of 1-tert-butyl-3 (R or S (+))- l- {4-cyano-3- 3- (3S-ethyl-1-methyl- 2-oxo-azepan-3-yl)-phenoxy-phenyl}-1- (3-methyl-3H-imidazol-4-yl)-ethyl- urea trifluoracetate Step A : Preparation of 1-tert-butyl-3 (R or S (+))- 1- (4-cyano-3-fluoro-phenyl)- 1-(3-methyl-3H-imidazol-4-yl)-ethyl]-urea trifluoroacetate To a solution of (+)-2-fluoro-4- 1-amino-1- (3-methyl-3H-imidazol-4- yl)-ethyl]-benzonitrile (as described in Example 24A, Step F) (0.1 g, 0.41 mmol) in DMF (2 mL) was added NEt3 (0.17 mL, 1.23 mmol) and t-butylisocyanate (0.102 mL, 0.9 mmol) and the reaction was stirred 18 hr. The DMF was removed in vacuo to obtain the title compound after purification by prep RP HPLC.

Step B : Preparation of 1-tert-butyl-3 (R or S (+))- 1- {4-cyano-3- 3- (3S-ethyl- 1-methyl-2-oxo-azepan-3-yl)-phenoxy-phenyl}-1- (3-methyl-3H- imidazol-4-yl)-ethyll-urea trifluoracetate Following the methods described in Example 39, Step E but using 1- tert-butyl-3 (R or S (+))- 1- (4-cyano-3-fluoro-phenyl)-1- (3-methyl-3H-imidazol-4-yl)- ethyl-urea trifluoroacetate (as described above in Step A) (0.026 g, 0.057 mmol) and 3 (S)-ethyl-3- (3-hydroxy-phenyl)-l-methyl-azepan-2-one (as described in Example 24A, Step B) (0.014 g, 0.057 mmol), the title compound was prepared.

FAB mass spectrum m/e 571 (M+1).

Analysis calculated for C33H42N603'1.75 CF3CO2H 0.25 H20: C, 56.58; H, 5.76; N, 10.85; Found: C, 56.58; H, 5.77; N, 10.62.

EXAMPLE 41 Preparation of 1-tert-butyl-3 (R or S (-))-[1-{4-cyano-3-[3-(3S-ethyl-1-methyl- 2-oxo-azepan-3-yl)-phenoxy-phenyl}-1-(3-methyl-3H-imidazol-4 -yl)-ethyl]- urea trifluoracetate To a solution of 2- 3- (3S)-ethyl-1-methyl-2-oxo-azepan-3-yl)- phenoxy-4- 1- (R or S) (-) amino-I- (3-methyl-3H-imidazol-4-yl)-ethyl-benzonitrile (as described in Example 24D) (0.040 g, 0.086 mmol) in DMF (2 mL) was added NEt3 (0.048 mL, 0.34 mmol) and t-butylisocyanate (0.039 mL, 0.34 mmol) and the reaction was stirred and heated at 80°C for 72 hr. The DMF was removed in vacuo to obtain the title compound after purification by prep RP HPLC to obtain the title compound (0.023 g).

FAB mass spectrum m/e 571 (M+1).

Analysis calculated for C33H42N603'1.95 CF3CO2H 0.10 H20: C, 55.75; H, 5.60; N, 10.57; Found: C, 55.74; H, 5.37; N, 10.63.

Using the same procedure but substituting appropriate isocyanate for t-butyl isocyanate the following compounds were prepared: 1- 1- {4-cyano-3 (R or S (-))- 3- (3S-ethyl-l-methyl-2-oxo-azepan-3-yl)- phenoxy-phenyl}-1- (3-methyl-3H-imidazol-4-yl)-ethvll-3-methvl-urea FAB mass spectrum m/e 529 (M+1).

HRMS measured 529.2915; theoretical 529.2921 S(-))-[3-(3S-ethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy]-1-[1 -{4-cyano-3(Ror phenyl $-1-(3-methyl-3H-imidazol-4-yl !-ethyl]-3-phenvl-urea FAB mass spectrum m/e 529 (M+1).

HRMS measured 591.3080; theoretical 591.3078

EXAMPLE 42 Preparation of 1- {4-cyano-3 (R or S (+))- 3- (3 S-ethyl-1-methyl-2-oxo-azepan- 3-yl)-phenoxy]-phenyl}-1-(3-methyl-3H-imidazol-4-yl)-ethyl]- ureatrifluoroacetate 1-tert-Butyl-3 (R or S (+))- 1- {4-cyano-3- 3- (3S-ethyl-1-methyl- 2-oxo-azepan-3-yl)-phenoxy-phenyl}-1-(3-methyl-3H-imidazol-4 -yl)-ethyl]-urea trifluoracetate (as described in Example 40) (0.02 g) was dissolved in TFA (6.0 mL) and CH2Cl2 (3.0 mL) and stirred for 18 hr. The solvents were removed in vacuo and the residue was lyophylized to obtain the title compound.

FAB mass spectrum m/e 515 (M+1).

Analysis calculated for C29H34N603'2.5 CF3CO2H 1.65 H20: C, 49.23; H, 4.84; N, 10.13; Found: C, 49.17; H, 4.44; N, 10.45.

Using the procedure outlined above but substituting 1-tert-Butyl-3 (R or S (-))- 1- {4- cyano-3- 3- ( S-ethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-phenyl}-1- (3-methyl- 3H-imidazol-4-yl)-ethyl-urea trifluoracetate (as described in Example 41), the following compound was prepared: S(-))-[3-(3S-ethyl-1-methyl-2-oxo-azepen-3-yl)-phenoxy]-[1-{ 4-cyano-3(Ror phenyl}3-methvl-3H-imidazol-4-yl)-ethyll-urea trifluoroacetate FAB mass spectrum m/e 515 (M+1).

EXAMPLE 43 Preparation ofN-1- {4-cyano-3- 3- (3S-ethyl-1-methyl-2-oxo-azepan-3-yl)- phenoxy-phenyl}-1- (3-methyl-3H-imidazol-4-yl)-ethvl-acetamide trifluoroacetate 4- 1- (R or S (-) amino-l- (3-methyl-3H-imidazol-4-yl)-ethyl- 2- 3- (3S-ethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-benzonitrile (as described in Example 24G (0.020 g, 0.042 mmol), Et3N (0.009 mL, 0.063 mmol) and acetyl chloride (0.004 mL, 0.051 mmol) were dissolved in DMF (0.30 mL) and stirred at RT for 16h. The reaction was quenched with H20 (0.10 mL), purified using reverse phase chromatography (95/5 to 5/95 H2O/CH3CN w/0.1 % TFA, flow = 15 mL/min) and lyophilized to give the title compound.

FAB mass spectrum m/e 514 (M+1).

HRMS measured 514.2819; theoretical 514.2818 EXAMPLE 44 Preparation of 2- 3- (3 S-ethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-4- 1- methylamino-1- (3-methyl-3H-imidazol-4-yl)-ethyl-benzonitrile Step A : Preparation of 2-fluoro-4- 1-methylamino-1- (3-methyl-3H imidazol-4-yl)-ethyll-benzonitrile 2-Fluoro-4- 1-hydroxy-1- (3-methyl-3H-imidazol-4-yl)-ethyl- benzonitrile (as described in Example 24A, Step D) (0.400 g, 1.65 mmol) was added to SOC12 (5 mL) in an ice-H2O bath, and stirred at RT for 45 min. The solution was concentrated in vacuo and azeotroped with anhydrous CH2Cl2 (3X). The solid was dissolved in CH2C12 (5 mL) and added via syringe to a-78°C solution of CH3NH2 (1), then the solution was stirred for 2h while warming to RT under Ar. The solution was concentrated in vacuo, partitioned between aq. NaHC03 and CH2Cl2, the aqueous layer was back extracted with CHOC12, and the organic layers were combined and dried (MgS04). Filtration, concentration and chromatographic purification using Si02 (1.0 % MeOH/CH2Cl2 with NH40H) gave the title compound.

Step B: Preparation of 2- 3- (3S-ethyl-1-methyl-2-oxo-azepan-3-yl)- phenoxy-4- 1-methylamino-1- (3-methyl-3H-imidazol-4-yl)- ethyl-benzonitrile 2-Fluoro-4- 1-methylamino-1- (3-methyl-3H-imidazol-4-yl)-ethyl- benzonitrile (as described above in Step A) (0.147 g, 0.569 mmol), 3-ethyl-3- (3- hydroxy-phenyl)-l-methyl-azepan-2-one (Example 24A, Step B) (0.140 g, 0.569 mmol), KF-A1203 (0.168 g) and 18-Crown-6 (0.014 g, 0.052 mmol) were refluxed in CH3CN (10 mL) under Ar for 24 h. The solution was filtered, concentrated in vacuo, and purified using Si02 chromatography (0-1.5 % MeOH/CH2Cl2 with NH40H) to give the title compound.

FTMS (M+1) = 486 Analysis calculated for C29H35N502'0.35 H20: C, 70.80; H, 7.32; N, 14.24; Found: C, 70.46; H, 6.92; N, 14.37.

The individual diastereomers were separated by chiral HPLC on a ChirakPak AD column to give: S)-methylamino-1-2-[3-(3S-ethyl-1-methyl-2-oxo-azepan-3-yl)- phenoxy]-4-[1(Ror (3-methyl-3H-imidazol-4-yl)-ethyll-benzonitrile HRMS theoretical: 486.2869, measured: 486.2851: Analysis calculated for C29H35N5O2#0.85H2O : C, 69.53; H, 7.38; N, 13.98.

Found: C, 69.50; H, 7.05; N, 13.84.

2-[3-(3S-ethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy]-4-[1 (R or S)-methylamino-1- (3-methyl-3H-imidazol-4-yl)-ethyl-benzonitrile HRMS theoretical: 486.2869, measured: 486.2863: Analysis calculated for C2gH35N502* 1 65 H20: C, 67.58; H, 7.49; N, 13.59.

Found: C, 67.57; H, 7.12; N, 13.31.

EXAMPLE 45 Preparation of 2- 5- (3S-ethyl-1-methyl-2-oxo-azepan-3-yl)-2-iodo-phenoxy- 4-imidazol-1-ylmethyl-benzonitrile Step A : Preparation of 3S-ethyl-3-(3-hydroxy-4-iodo-phenyl)-1-methyl-azepan- 2-one To a solution of 3S-ethyl-3- (3-hydroxy-phenyl)-1-methyl-azepan-2-one (as described in Example 24A, Step B) (0.10 g, 0.404 mmol) in DMF (2 mL) was added a solution of N-iodosuccinimide in DMF (2 mL) with stirring under Ar at ambient temperature. After stirring for 16 hr the reaction mixture was partitioned between CH2C12 and H20, the organic layer separated, washed with H20, aqueous saturated NaHC03 solution, brine, and dried (Na2SO4). Filtration and concentration to dryness gave the title compound as a white solid after chromatography on silica gel

eluting with EtOAc: hexane, 1: 3 to 1: 2.1D-NOE NMR confirmed the position of the iodo substituent.

Step B : Preparation of 2- 5- (3S-ethyl-1-methyl-2-oxo-azepan-3-yl)-2- iodo-phenoxy-4-imidazol-1-ylmethyl-benzonitrile 3S-Ethyl-3- (3-hydroxy-4-iodo-phenyl)-1-methyl-azepan-2-one (as described above in Step A) (0.118 g, 0.316 mmol), 2-fluoro-4-imidazol-1-ylmethyl- benzonitrile (as described in Example 1, Step E) (0.64 g, 0.316 mmol), KFAl203 (0.100 g) and 18-Crown-6 (0.010 g) were refluxed in CH3CN (10 mL) under Ar for 24 h. The solution was filtered, concentrated in vacuo, and purified using Si02 chromatography (1-2 % MeOH/CH2Cl2 with NH40H) followed by preparative RP HPLC to give the title compound.

FTMS (M+1) = 555 Using the methods outlined above but substituting N-bromosuccinimide for N- iodosuccinimide in Step A and 2-fluoro-4- 1- (-) amino-1- (3-methyl-3H-imidazol- 4-yl)-ethyl]-benzonitrile (as described in Example 24A, Step F) for 2-fluoro-4- imidazol-1-ylmethyl-benzonitrile in Step B, the following compound was prepared: 2-[5-(3S-ethyl-1-methyl-2-oxo-azepan-3-yl)-2-bromo-phenoxy]- 4-imidazol-1- ylmethyl-benzonitrile FTMS (M+1) = 552 Analysis calculated for C28H32Ns02Br* 2.4 HCI: C, 52.71; H, 5.43; N, 10.98; Found: C, 52.62; H, 5.44; N, 11.01.

EXAMPLE 46 Preparation of 4- 1-amino-1- (3-methyl-3H-imidazol-4-yl)-ethyl-2- 2- dimethylaminomethyl-5- (3 S-ethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy- benzonitrile Step A: Preparation of 3- (4-dimethylaminomethyl-3-hydroxy-phenyl)-3S- ethyl-1-methvl-azepan-2-one

Formaldehyde (38 % in H20) (0.084 mL, 0.0011 mol) was added to a mixture of 3S-ethyl-3- (3-hydroxy-phenyl)-1-methyl-azepan-2-one (as described in Example 24A, Step B) (0.25 g, 0.001 mol) and aqueous dimethylamine (40% in H20) (0.45 mL, 0.002 mol) in abs EtOH (3 mL) and the mixture was heated at reflux for 3 hr then concentrated to dryness to give the title compound.

PreparationStepB: of 4-[1-amino-1-(3-methyl-3H-imidazol-4-yl)-ethyl]-2- [2-dimethylaminomethyl-5-(3S-ethyl-1-methyl-2-oxo-azepan-3-y l)- phenoxyl-benzonitrile Using the methods described in Example 45, Step B, but substituting 3- (4-dimethylaminomethyl-3-hydroxy-phenyl)-3 S-ethyl-1-methyl-azepan-2-one and 2-fluoro-4- 1-amino-1- (3-methyl-3H-imidazol-4-yl)-ethyl-benzonitrile (as described in Example 24A, Step F) the title compound was prepared.

FTMS (M+1) = 529 Analysis calculated for C3lH40N602: C, 70.43 ; H, 7.63; N, 15.90; Found: C, 70.37; H, 7.61; N, 15.48.

EXAMPLE 47 Preparation of 2- 3- (3-ethyl-l-methyl-2-oxo-azepan-3-yl)-phenoxy-4- (4,5,6,7- tetrahydro-3H-imidazo 4, 5- pvridin-4-yl)-benzonitrile Step A : Preparation of2-fluoro-4- (4,5,6,7-tetrahydro-3H-imidazo [4,5- clpvt'idin-4-yl)-benzonitrile 2-Fluoro-4-formyl-benzonitrile (as described in Example 29, Step A) (0.268 g, 1.79 mmol) and histamine (0.200 g, 1.79 mmol) were heated neat at 120°C for 1.5h. The molten liquid was cooled to give the title compound.

Step B : Preparation of 4- (4-cyano-3-fluoro-phenyl)-6, 7-dihydro-4H-imidazo di-tert-butylester[4.5-c]pyridine-3,5-dicarboxylicacid 2-Fluoro-4- (4,5,6,7-tetrahydro-3H-imidazo [4,5-c] pyndin-4-yl)- benzonitrile (as described above in Step A) (0.277 g, 1.143 mmol), Boc2O (1. 10 g, 5.03 mmol), Et3N (0.70 mL, 5.03 mmol) were dissolved in CH2Cl2 (20 mL) and

stirred at RT for 3 days. The reaction was concentrated and purified using Si02 chromatography (1% MeOH/CH2Cl2) to give the title compound.

Step C : Preparation of 2- 3- (3-ethyl-1-methyl-2-oxo-azepan-3-yl)- phenoxy-4- (4,5,6,7-tetrahydro-3H-imidazo [4,5-] pyridin-4-yl)- benzonitrile 4- (4-Cyano-3-fluoro-phenyl)-6, 7-dihydro-4H-imidazo [4,5-c] pyridine- 3,5-dicarboxylic acid di-tert-butyl ester (as described above in Step B) (0.120 g, 0.270 mmol), 3-ethyl-3- (3-hydroxy-phenyl)-l-methyl-azepan-2-one (0.067 g, 0.270 mmol), KFAl203 (0.080 g) and 18-Crown-6 (0.007 g) were refluxed in CH3CN (5 mL) under Ar for 24 h. The solution was filtered and concentrated in vacuo. The residue was dissolved in CH2Cl2 (10 mL) and TFA (5 mL) and stirred at RT for 1/2 h. The solution was concentrated in vacuo, purified using reverse phase chromatography (95/5 to 5/95 H2O/CH3CN w/0.1% TFA, flow = 65 mL/min), lyophilized, and then further separated into two diastereomers using a Chiralpak AS column.

Diastereomer A: HRMS Measured = 470.2562, Theoretical = 470.2550 Diastereomer B: HRMS Measured = 470.2570, Theoretical = 470.2550 EXAMPLE 48 Preparation of 2- 3-ethyl-2-oxo-azepan-3-yl)-phenoxy-4- 1- amino-1- (3-methyl- 3H-imidazol-4-yl)-ethyll-benzonitrile Using the methods described in Example 24A, but substituting racemic 3-ethyl-3- (3-hydroxy-phenyl)-azepan-2-one (as described in Example 8, Step B) and racemic 2-fluoro-4- l-amino-1- (3-methyl-3H-imidazol-4-yl)-ethyl-benzonitrile (as described in Example 24A, Step F), the following compound was prepared: 2- 3-ethyl-2-oxo-azepan-3-yl)-phenoxy-4- 1-amino-1- (3-methyl-3H-imidazol-4- yl)-ethyll-benzonitrile FAB MS (M+1) = 458 Analysis calculated for C27H3NsO2 * 0.65 H20:

C, 69.10; H, 6.94; N, 14.92; Found: C, 69.08; H, 6.96; N, 13.86.

EXAMPLE 49 Preparation of 4- 1-amino-1- (3-methyl-3H-imidazol-4-yl)-ethyl-2- {3- 3-ethyl-1- (3-hydroxy-propyl)-2-oxo-azepan-3-yl-phenoxy}-benzonitrile ditrifluoroacetate Step A: Preparation of 1-allyl-3-ethyl-3-(3-methoxy-phenyl)-azepan-2-one To a solution of 3-ethyl-3- (3-methoxy-phenyl)-azepan-2-one (as described in Example 8, Step A) (2.62 g, 10.6 mmol) in DMF (20 mL) at 0°C was added NaH (0.47 g, 11.6 mmol) and the mixture was stirred for 20 min at 0°C and 20 min at 25°C. Allyl bromide (1.1 mL, 12.7 mmol) was added. After 5 hr, additional NaH (0.12 g, 3 mmol) and allyl bromide (0.3 mL, 3.5 mmol) were added and stirring was continued for 16 hr. The reaction was quenched with H20 and the solvents removed in vacuo. The residue was partitioned between EtOAc and saturated NaHC03 solution. The organic layer was separated, washed with brine and dried (MgS04). Filtration and concentration gave the title compound after chromatography on silica gel with hexane and ethyl acetate 9: 1.

Step B: Preparation of 3-ethyl-1- (3-hydroxy-propyl)-3- (3-methoxy-phenyl)- azepan-2-one To a solution of 1-allyl-3-ethyl-3- (3-methoxy-phenyl)-azepan-2-one (as described above in Step A) (0.83 g, 2.89 mmol) in THF (20 mL) at 0°C was added BH3-THF (1M, 2.85 mL, 2.89 mmol) with stirring. After 1 hr at 25°C the reaction mixture was cooled to 0°C and EtOH (5 mL), 1M NaOH (5 mL), and 30% H202 (10 mL) were added. The reaction mixture was stirred for 1 hr at 25°C. The solvents were removed in vacuo and the residue was partitioned between EtOAc and saturated NaHC03 solution. The organic layer was washed with brine and dried (MgS04), the solvents removed in vacuo and the crude product chromatographed on silica gel with hexane and ethyl acetate 1: 1 to obtain the title compound.

Step C : Preparation of 3-ethyl-3- (3-hydroxy-phenyl)-1- (3-hydroxy- propyl)-azepan-2-one

To a solution of 3-ethyl-1- (3-hydroxy-propyl)-3- (3-methoxy-phenyl)- azepan-2-one (as described above in Step B) (0.12 g, 0.393 mmol) in CH2Cl2 (20 mL) at-78°C was added BBr3 (1M, 0.786 mL, 0.786 mmol). The reaction mixture was stirred for 3 hr at 25°C then quenched with H2O and extracted with EtOAc. The organic layer was separated, washed with brine and dried (MgS04). Filtration and concentration to dryness gave the title compound after purification by preparative RP HPLC.

Step D : Preparation of 4- 1-amino-1- (3-methyl-3H-imidazol-4-yl)-ethyl-2- {3- 3-ethyl-1- (3-hydroxy-propyl)-2-oxo-azepan-3-yl-phenoxy}- benzonitrile ditrifluoroacetate Using the procedure described in Example 47, Step C, 3-ethyl-3- (3- hydroxyphenyl)-1- (3-hydroxy-propyl)-azepan-2-one (as described above in Step C) (0.036 g, 0.124 mmol) and 2-fluoro-4- l-amino-1- (3-methyl-3H-imidazol-4-yl)- ethyl]-benzonitrile (as described in Example 24A, Step F) (0.030 g, 0.124 mmol) were reacted to obtain the title compound after purification by RP HPLC.

FAB MS; M+1 =516.

Analysis calculated for C30H37N503 2.5 CF3CO2H 2.15 H2O: C, 50.07; H, 5.26; N, 8.34; Found: C, 50.07; H, 4.91; N, 8.49.

EXAMPLE 50 Preparation of 4-[1-amino-1-(3-methyl-3H-imidazol-4-yl)-ethyl]-2-{3-[3- ethyl-1-propyl-2-oxo-azepan-3-vl-phenoxy}-benzonitrile ditrifluoroacetate Step A: Preparation of 1-propyl-3-ethyl-3-(3-methoxy-phenyl)-azepan-2-one Hydrogenation of 1-allyl-3-ethyl-3- (3-methoxy-phenyl)-azepan-2-one (as described in Example 49, Step A) in a Parr apparatus in EtOH with 10% Pd/C provided the title compound after filtration and concentration.

Step B: Preparation of 1-propyl-3-ethyl-3-(3-hydroxy-phenyl)-azepan-2-one Using the method described in Example 49, Step C, the title compound was prepared.

Step C : Preparation of 4- 1-amino-I- (3-methyl-3H-imidazol-4-yl)-ethyl- 2- {3- 3-ethyl-l-propyl-2-oxo-azepan-3-yl-phenoxy}-benzonitrile ditrifluoroacetate Using the procedure described in Example 47, Step C, 3-ethyl-3- (3- hydroxy-phenyl)-1-propyl-azepan-2-one (as described above in Step B) (0.036 g, 0.124 mmol) and 2-fluoro-4- 1-amino-1- (3-methyl-3H-imidazol-4-yl)-ethyl- benzonitrile (as described in Example 24A, Step F) (0.030 g, 0.124 mmol) were reacted to obtain the title compound after purification by RP HPLC.

FAB MS; M+1 = 500.

Analysis calculated for C30H37NsO3 2.5 CF3CO2H 0.1 H20: C, 53.44; H, 5.09; N, 8.91; Found: C, 53.46; H, 4.77; N, 8.70.

EXAMPLE 51 Preparation of 2- {3- 1- (2-amino-ethyl)-3-ethyl-2-oxo-azepan-3-yl-phenoxy}-4- 11-amino-1-(3-methvl-3H-imidazol-4-yl)-ethyl]-benzonitrile tris-trifluoroacetate Step A : Preparation of 3-ethyl-1- (2-hydroxy-ethyl)-3- (3-methoxy- phenyl)-azepan-2-one A solution of 1-allyl-3-ethyl-3- (3-methoxy-phenyl)-azepan-2-one (as described in Example 49, Step A) (0.9 g, 3.1 mmol) in MeOH (20 mL) at-78°C was saturated with 03 for 20 min. The solution was warmed to-40°C for 10 min then NaBH4 (0.36 g, 9.4 mmol) was added and the reaction mixture was stirred at 25°C for 1 hr. The MeOH was removed in vacuo, and the residue was partitioned between EtOAc and saturated NaHC03 solution. The organic layer was washed with brine and dried (MgS04). Filtration and concentration to dryness gave the title compound.

Step B : Preparation of 1- (2-azido-ethyl)-3-ethyl-3- (3-methoxy-phenyl)- azepan-2-one To a solution of 3-ethyl-l- (2-hydroxy-ethyl)-3- (3-methoxyphenyl)- azepan-2-one (as described above in Step A) (0.6 g, 2.05 mmol) in CH2Cl2 (15 mL) at 0°C was added NEt3 (0.85 mL, 6.15 mmol) and methanesulfonyl chloride (0.19 mL, 2.45 mmol). The mixture was stirred for 30 min at 25°C then NaN3 (0.27 g, 4.1

mmol) in DMF (3 mL) was added. The reaction was stirred for 18 hr then partitioned between EtOAc and saturated NaHC03 solution. The organic layer was washed with brine and dried (MgS04) Filtration and concentration to dryness gave the title compound.

FAB MS; M+1 = 317.

Step C : Preparation of 1- (2-amino-ethyl)-3-ethyl-3- (3-methoxy-phenyl)- azepan-2-one hydrochloride To a solution of 1- (2-azido-ethyl)-3-ethyl-3- (3-methoxy-phenyl)- azepan-2-one (as described above in Step B) (0.56 g, 1.94 mmol) in MeOH (20 mL) and 10% HCl (5 mL) was added 10% Pd/C (0.1 g) under argon. The mixture was placed under 1 atm of H2 and stirred for 2 hr. Filtration and concentration to dryness gave the title compound.

Step D: Preparation of 1- (2-amino-ethyl)-3-ethyl-3- (3-hydroxy-phenyl)- azepan-2-one trifluoroacetate To a solution of 1- (2-amino-ethyl)-3-ethyl-3- (3-methoxy-phenyl)- azepan-2-one hydrochloride (as described above in Step C) (0.1 g, 0.306 mmol) in CH2Cl2 (10 mL) at-78°C was added BBr3 (1M, 0.60 mL, 0.60 mmol). After stirring for 2 hr at 25°C the reaction mixture was quenched with H20, the solvents removed in vacuo and the residue purified by prep RP HPLC to obtain the title compound.

Step E: Preparation of 2- {3- 1- (2-amino-ethyl)-3-ethyl-2-oxo-azepan-3- yl-phenoxy}-4- 1-amino-1- (3-methyl-3H-imidazol-4-yl)-ethyl- benzonitrile tri-trifluoroacetate Using the procedure described in Example 47, Step C, 1- (2-amino- ethyl)-3-ethyl-3- (3-hydroxy-phenyl)-azepan-2-one trifluoroacetate (as described above in Step D) (0.070 g, 0.18 mmol) and 2-fluoro-4- 1- (-) amino-1- (3-methyl-3H imidazol-4-yl)-ethyl]-benzonitrile (as described in Example 24A, step F) (0.044 g, 0.18 mmol) were reacted to obtain the title compound.

FAB MS; M+1 = 501.

Analysis calculated for C29H36N602-3. 45 CF3CO2H: C, 48.23; H, 4.45; N, 9.40; Found: C, 48.59; H, 4.05; N, 9.28.

EXAMPLE 52 Preparation of {2- 3- (3- {5- 1-amino-1- (3-methyl-3H-imidazol-4-yl)-ethyl 2-cyano- phenoxyS-phenyl)-3-ethyl-2-oxo-azepan-1-yl3-ethyl $-urea ditrifluoroacetate Step A: Preparation of 1-tert-butyl-3- {2- 3-ethyl-3- (3-methoxy-phenyl)- 2-oxo-azepan-1-yl-ethyl}-urea To a solution of 1- (2-amino-ethyl)-3-ethyl-3- (3-methoxy-phenyl)- azepan-2-one hydrochloride (as described in Example 51, Step C) (0. 27 g, 0.826 mmol) in DMF (5.0 mL) was added NEt3 (0.35 mL, 2.48 mmol) and t-butylisocyanate (0.29 mL, 2.48 mmol) and the solution was stirred for 18 hr. The DMF was removed in vacuo, and the residue partitioned between EtOAc and H20. The organic layer was washed with brine and dried (MgS04), and the solvents removed in vacuo to obtain the title compound.

Step B : Preparation of 12- 3-ethyl-3- (3-methoxy-phenyl)-2-oxo-azepan- l-yll-ethyl}-urea To a solution of 1-tert-butyl-3- {2- 3-ethyl-3- (3-methoxy-phenyl)-2- oxo-azepan-1-yl-ethyl}-urea (as described above in Step A) (0.35 g, 0.23 mmol) in CH2Cl2 (2.0 mL) was added TFA (6.0 mL) and the solution was stirred for 72 hr. The solvents were removed in vacuo to obtain the title compound.

Step C : Preparation of {2- 3-ethyl-3- (3-hydroxy-phenyl)-2-oxo-azepan- 1-yl-ethyl}-urea To a solution of {2- 3-ethyl-3- (3-methoxy-phenyl)-2-oxo-azepan-1-yl- ethyl}-urea (as described above in Step B) (0.14 g, 0.421 mmol) in CH2Cl2 (10 mL) at -78°C was added BBr3 (1M, 0.842 mL, 0.842 mmol). After stirring for 1 hr at 25°C the reaction was quenched with H2O and partitioned with EtOAc and 10% HCI. The organic layer was washed with brine and dried (MgS04), and the solvent removed in vacuo to obtain the title compound.

Step D : Preparation of {2- 3- (3- {5- 1-amino-1- (3-methyl-311-imidazol-4-yl)- ethyl]2-cyano-phenoxy}-phenyl)-3-ethyl-2-oxo-azepan-1-yl-eth yl}- urea ditrifluoroacetate

Using the procedure described in Example 47, Step C, {2- 3-ethyl-3- (3-hydroxy-phenyl)-2-oxo-azepan-1-yl-ethyl}-urea (as described above in Step C) (0.070 g, 0. 23 mmol) and 2-fluoro-4- 1- (-) amino-1- (3-methyl-3H-imidazol-4-yl)- ethyl-benzonitrile (as described in Example 24A, Step F) (0.058 g, 0.23 mmol) were reacted to prepare the title compound.

FAB MS; M+1 = 544.

Analysis calculated for C3oH37N703'2.5 CF3CO2H 0.9 H2O: C, 49.81; H, 4.81; N, 11.62; Found: C, 49.81; H, 4.55; N, 11.47.

In a similar manner, using methylisocyanate in place of t-butylisocyanate in Step A, the following compound was prepared: N-{2- 3- (3- {5- 1-amino-1- (3-methyl-3H-imidazol-4-yl)-ethyl 2-cyano-phenoxy}- phenyl)-3-ethvl-2-oxo-azepan-1-yll-ethyl}-N'-methyl urea ditrifluoroacetate FAB MS; M+1 = 558.

Analysis calculated for C3lH4lN703-2.5 CF3C02H'0.4 H20: C, 50. 87; H, 5.02; N, 11.54; Found: C, 50.87; H, 4.73; N, 11.33.

EXAMPLE 53 Preparation of 4- 1-amino-1- (3-methyl-3H-imidazol-4-yl)-ethyl-2- {3- 1- (2-dimethylamino-ethyl)-3-ethyl-2-oxo-azepan-3-yl-phenoxy}-b enzonitrile tri-trifluoroacetate Step A : Preparation of 1- (2-dimethylamino-ethyl)-3-ethyl-3- (3-methoxy- phenyl)-azepan-2-one trifluoroacetate To a solution of 1- (2-amino-ethyl)-3-ethyl-3- (3-methoxy-phenyl)- azepan-2-one hydrochloride (as described in Example 51, Step C) (0.2 g, 0.612 mmol) in MeOH (7.0 mL) was added paraformaldehyde (0.1 g) and NaCNBH3 (0.115 g, 1.84 mmol) and the solution was stirred for 18 hr. The MeOH was removed in vacuo and the residue partitioned between EtOAc and saturated NaHC03 solution.

The organic layer was washed with brine and dried (MgS04), and the solvents removed in vacuo to obtain the title compound after purification by preparative RP HPLC.

Step B : Preparation of 1- (2-dimethylamino-ethyl)-3-ethyl-3- (3- hydroxy-phenyl)-azepan-2-one trifluoroacetate To a solution of 1- (2-dimethylamino-ethyl)-3-ethyl-3- (3-methoxy- phenyl)-azepan-2-one trifluoroacetate (as described above in Step A) (0.1 g, 0.23 mmol) in CH2C12 (10 mL) at-78°C was added BBr3 (1M, 0.46 mL, 0.46 mmol).

After stirring for 2 hr at 25°C the reaction mixture was quenched with H20, the solvents removed in vacuo, and the residue purified by prep RP HPLC to obtain the title compound.

Step C: Preparation of 4- l-amino-1- (3-methyl-3H-imidazol-4-yl)- ethyl-2- {3- 1- (2-dimethylamino-ethyl)-3-ethyl-2-oxo-azepan- 3-yll-phenoxy}-benzonitrile tri-trifluoroacetate Using the procedure described in Example 47, Step C, 1- (2- dimethylamino-ethyl)-3-ethyl-3- (3-hydroxy-phenyl)-azepan-2-one trifluoroacetate (as described above in Step B) (0.060 g, 0.154 mmol) and 2-fluoro-4- 1- (-) amino-1- (3- methyl-3H-imidazol-4-yl)-ethyl-benzonitrile (as described in Example 24A, Step F) (0.040 g, 0.16 mmol) were reacted to obtain the title after purification by preparative RP HPLC.

FAB MS; M+1 = 529.

Analysis calculated for C3lH40N602-3. 70 CF3CO2H: C, 48.52; H, 4.63; N, 8.84; Found: C, 48.55; H, 4.26; N, 8.67.

EXAMPLE 54 Preparation of 4- 1- (R or S) amino-1- (3-methyl-3H-imidazol- 4-yl)-ethyl-2- {3- 1- (2-hydroxyethyl)-3-ethyl-2-oxo-azepan-3-yl-phenoxy}- benzonitrile tris trifluoroacetate Step A: Preparation of 3-ethyl-3- (3-benzyloxv-phenyl)-azepan-2-one

To a solution of 3-ethyl-3- (3-hydroxy-phenyl)-azepan-2-one (Example 8, Step B) (1.28 g, 0.0055 mol) in DMF (50 mL) was added Cs2C03 (1 79 g, 0.055 mmol) and benzyl bromide (0.653 mL, 0.0055 mmol) at ambient temperature. After 1.5 h the reaction was concentrated to remove most of the DMF and the residue was partitioned between EtOAc and H2O. The aqueous layer was washed with EtOAc, the organics combined, washed with brine, and dried (MgS04).

Filtration and concentration to dryness gave the title compound.

Step B: Preparation of 1-allyl-3-ethyl-3- (3-benzyloxy-phenyl)-azepan- 2-one To a solution of 3-ethyl-3- (3-benzyloxy-phenyl)-azepan-2-one, as described above in Step A, (1.65 g, 5.1 mmol) in DMF (25 mL) at 0°C was added NaH (0.265 g, 6.63 mmol) and the mixture was stirred for 20 min at 0°C and 20 min at 25°C. Allyl bromide (0.66 mL, 7.65 mmol) was added. After 5 hr, additional NaH (0.13 g, 3 mmol) and allyl bromide (0.33 mL, 3.5 mmol) were added and stirring was continued for 16 hr. The reaction was quenched with H20 and the solvents removed in vacuo. The residue was partitioned between EtOAc and saturated NaHC03 solution. The organic layer was separated, washed with brine and dried (MgS04).

Filtration and concentration gave the title compound after chromatography on silica gel with hexane and ethyl acetate 9: 1.

Step C: Preparation of 3-ethyl-1- (2-hydroxy-ethyl)-3- (3-benzyloxy- phenyl)-azepan-2-one A solution of 1-allyl-3-ethyl-3- (3-benzyloxy-phenyl)-azepan-2-one, as described in Step B above, (0.87 g, 2.4 mmol) in MeOH (20 mL) at-78°C was saturated with 03 for 25 min. The solution was warmed to-40°C for 10 min then NaBH4 (0.363 g, 9.6 mmol) was added and the reaction mixture was stirred at 25°C for 1 hr. The MeOH was removed in vacuo, and the residue was partitioned between EtOAc and saturated NaHC03 solution. The organic layer was washed with brine and dried (MgS04). Filtration and concentration to dryness gave the title compound after chromatography on silica gel eluting with hexane/EtOAc, 1/1.

Step D: Preparation of 3-ethyl-1- (2-hydroxy-ethyl)-3- (3-hydroxy- phenyl)-azepan-2-one

To a solution of 3-ethyl-1- (2-hydroxy-ethyl)-3- (3-benzyloxy-phenyl)- azepan-2-one, as described above in Step C, (0.270 g, 0.735 mmol) in MeOH (15 mL) was added 10% Pd on C (0.06 g), and the mixture was hydrogenated under a H2 balloon at ambient temperature for 16 h. The reaction mixture was filtered, and the filtrate concentrated to dryness to give the title compound.

Step E: Preparation of 4- 1- (R or S) amino-l- (3-methyl-3H-imidazol-4-yl)- ethyl-2- {3- l- (2-hydroxyethyl)-3-ethyl-2-oxo-azepan-3-yl-phenoxy}- benzonitrile tris trifluoroacetate Using the procedure described in Example 47, Step C, 3-ethyl-1- (2- hydroxy-ethyl)-3- (3-hydroxy-phenyl)-azepan-2-one (0.060 g, 0.216 mmol) and 2- fluoro-4- l- (-) amino-1- (3-methyl-3H-imidazol-4-yl)-ethyl-benzonitrile (as described in Example 24A, Step F) (0.053 g, 0.216 mmol) were reacted to prepare the title compound.

FAB MS; M+1 = 502 Analysis calculated for C29H35N503-2.5 CF3C02H 0.1 H20: C, 51.79; H, 4.82; N, 8.88; Found: C, 51.78; H, 4.68; N, 8.77.

EXAMPLE 55 Preparation of 4- 1-amino-1- (3-methyl-3H-imidazol-4-yl)-ethyl-2- {3- 1- (2- methoxyethyl)-3R-ethyl-2-oxo-azepan-3-yl-phenoxy}-benzonitri le tris trifluoroacetate Step A: Preparation of 3-ethyl-1- (2-methoxy-ethyl)-3- (3-benzyloxy-phenyl)- azepan-2-one Sodium hydride (0.017 g, 0.425 mmol) was added to a solution of 3-ethyl-1- (2-hydroxy-ethyl)-3- (3-benzyloxyphenyl)-azepan-2-one, as described in Example 54, Step C, (0.12 g, 0.327 mmol) in DMF (5 mL) at 0°C with stirring. After 20 min CH3I (0.041 mL, 0.653 mmol) was added and stirring was continued at room temperature for 16 h. The reaction was quenched with H20 and the solvents removed in vacuo. The residue was partitioned between EtOAc and saturated NaHC03

solution. The organic layer was separated, washed with brine and dried (MgS04).

Filtration and concentration gave the title compound.

Step B: Preparation of 3-ethyl-1- (2-methoxyethyl)-3- (3-hydroxy- phenyl)-azepan-2-one To a solution of 3-ethyl-1- (2-methoxy-ethyl)-3- (3-benzyloxy-phenyl)- azepan-2-one, as described above in Step A, (0.120 g, 0.412 mmol) in MeOH (20 mL) was added 10% Pd on C (0.025 g), and the mixture was hydrogenated under a H2 balloon at ambient temperature for 48 h. The reaction mixture was filtered, and the filtrate concentrated to dryness to give the title compound.

Step C: Preparation of 4- 1-amino-1- (3-methyl-3H-imidazol-4-yl)-ethyl- 2- {3- 1- (2-methoxyethyl)-3-ethyl-2-oxo-azepan-3-yl-phenoxy}- benzonitrile tris trifluoroacetate Using the procedure described in Example 47, Step C, 3-ethyl-1- (2- methoxy-ethyl)-3- (3-hydroxy-phenyl)-azepan-2-one, as described above in Step B, (0.090 g, 0.309 mmol) and 2-fluoro-4- 1- (-) amino-1- (3-methyl-3H-imidazol-4-yl)- ethyl]-benzonitrile (as described in Example 24A, Step F) (0.075 g, 0.309 mmol) were reacted to prepare the title compound.

FAB MS; M+1 = 516 Analysis calculated for C3oH37N503'2.4 CF3CO2H: C, 52.95; H, 5.03; N, 8.87; Found: C, 53.08; H, 4.65; N, 8.79.

EXAMPLE 56 Preparation of 4-Imidazol-1-ylmethyl-2- 3- ( 1-methyl-7-oxo-azepan-2-yl)-phenoxy- benzonitrile hydrochloride Step A: Preparation of 2- (3-methoxyphenyl)-cyclohexanone To 1.6 M n-BuLi in hexane (12.5 mL, 20 mmol) at 0°C was added a solution of 2,2,6,6-tetramethyl piperidine (3.4 mL, 20 mmol) in anhydrous THF (40 mL) over 5 min. Cyclohexanone (2.1 mL, 20 mmol) was added dropwise. After stirring the reaction mixture for 10 min, a further portion of 1.6M n-BuLi in hexane

(12.5 mL, 20 mmol) was added, the mixture stirred for 10 min, and 2-chloroanisole (2.5 mL, 20 mmol) was added. After stirring for 30 min under Ar, the reaction mixture was quenched with H20 (300 mL) and extracted with EtOAc (150 mL).

The organic layer was separated, washed with 2N HC1 (100 mL), H20 (200 mL) and brine, then dried (MgS04). Filtration and concentration gave the title compound after Si02 chromatography (CHC13: MeOH, 200: 1).

Step B: Preparation of 2- (3-methoxyphenyl)-cyclohexanone oxime 2- (3-Methoxyphenyl)-cyclohexanone, as described in Step A above, (0.61 g, 3 mmol), hydroxylamine hydrochloride (0.40 g, 6 mmol) and sodium acetate (0.80,6.2 mmol) were combined in EtOH (4 mL): H20 (2 mL) and heated at reflux for 3 h. The reaction mixture was concentrated, then partitioned between EtOAc (100 mL) and H20 (100 mL), the organic layer separated, washed with H20, brine, and dried (MgS04). Filtration and concentration to dryness gave the title compound.

Step C: Preparation of 7- (3-methoxyphenyl)-azepan-2-one 2- (3-Methoxyphenyl)-cyclohexanone oxime, as described above in Step B, (0.62 g, 2.8 mmol) and polyphosphoric acid (20 g) were combined and heated at 115-120°C with stirring for 20 min. The dark mixture was poured into ice (100 g) with stirring, diluted with H20 (200 mL), then extracted with CHC13 (5 x 100 mL), the organic layers combined, washed with brine, dried (MgS04), filtered, and concentrated to dryness to give the title compound after Si02 chromatography (CHC13: MeOH, 100: 1 to 20: 1).

FAB MS (M +1) 220 Step D: Preparation of 7-(1-methyl-3-methoxaphenyl)-azepan-2-one 7- (3-Methoxyphenyl)-azepan-2-one, as described above in Step C, (0.13 g, 0.6 mmol) and NaH (60% dispersion in mineral oil) (0.03 g, 0.75 mmol) were dissolved in THF (5 mL) under Ar at ambient temperature. After 0.25 hours, iodomethane (0.064 mL, 1.0 mmol) was added and stirring was continued for 20 h.

The reaction mixture was partitioned between EtOAc (100 mL)-H20 (100 mL), the organic layer separated, washed with H20, dried (MgS04), filtered and concentrated to give the title compound after Si02 chromatography (CHC13: MeOH, 100: 1 to 50: 1).

FAB MS (M +1) 234

Step E: Preparation of 7- (l-methyl-3-hydroxyphenyl)-azepan-2-one BBr3 (1M in CH2Cl2) (1 mL, 1 mmol) was added dropwise to a solution of 7- (1-methyl-3-methoxyphenyl)-azepan-2-one, as described in Step D above, (0.06 g, 0.26 mmol) in CHCl3 (1 mL) at 0°C with stirring. The ice bath was removed and stirring was continued for 1 h. The reaction mixture was partitioned between EtOAc (100 mL) - H2O (100 mL), the organic layer separated, washed with H20, dried (MgS04), filtered and concentrated to give the title compound after Si02 chromatography (CHCl3: MeOH, 10: 1).

FAB MS (M +1) 219 Step F: Preparation of 4-hnidazol-1-ylmethyl-2- 3- (I-methyl-7-oxo-azepan-2- vl)-phenoxy-benzonitrile 7- (1-Methyl-3-hydroxyphenyl)-azepan-2-one, as described above in Step E, (0.055 g, 0.25 mmol), 2-fluoro-4-imidazol-1-ylmethyl-benzonitrile, as described in Example 1, Step E, (0.055 g, 0.27 mmol) and Cs2CO3 (0.490 g, 0.75 mmol) were dissolved in DMSO (4 mL) and stirred at 50°C under Ar for 3 h. The reaction mixture was cooled and partitioned between EtOAc (120 mL) and H20 (150 mL). The organic layer was separated, washed with H20 (2 x 100 mL), dried (MgS04). Filtration, concentration and RPLC (eluting with a 0.1% TFA/H20: 0.1% TFA/CH3CN gradient, 95: 5 to 5: 95) gave the title compound after conversion to the hydrochloride salt.

HRMS theoretical: 401.1972, measured: 401.1980.

Analysis calculated for C24H24N402-0.55 HCl-0.25 C2HsOH: C, 62.81; H, 5.82; N, 11. 96; Found: C, 62.83; H, 5.56; N, 11.77.

EXAMPLE 57 Preparation of 4- 1-Amino-1- (3-methyl-3H-imidazol-4-yl)-ethyl-2- 3- (4-methyl-3- oxo-2-propyl-morpholin-2-yl)-phenoxyl-benzonitrile Step A: Preparation of 4-Methyl-morpholine-2,3-dione Following the procedure of G. Drefahl, M. Hartmann and A. Skurk, Chem. Ber. 1966,99, diethyl oxalate (1.81 mL, 0.0133 mol) and N-methylamino-

ethanol (1 g, 0.0133 mol) were dissolved in toluene (50 mL) and heated at reflux for 5 h. The reaction mixture was concentrated to dryness, triturated with ether, and the residue chromatographed (Si02,0-5% MeOH in CH2CI2) to give the title compound.

1H NMR (CDC13) 6 4.53 (t, 2H), 3.70 (t, 2H), 3.18 (s, 3H).

Step B: Preparation 2-Hydroxy-2- (3-methoxy-phenyl)-4-methyl-morpholin-3- one Following the procedure described by S. Murahashi et al., Bull. Chem.

Soc. Japan, 69,2079-2090 (1996), 3-bromoanisole (0.633 mL, 5 mmol) in dry THF (2 mL) was added dropwise to a mixture of Mg (0.121 g, 5 mmol) and a few crystals of I2 in THF (2 mL) at ambient temperature under Ar. After stirring for 1 h, this solution was added dropwise to a solution of 4-methyl-morpholine-2,3-dione (0.15 g, 1.16 mmol) in CH ? C12 (2 mL) under Ar at 0°C. and the reaction mixture stirred for 1 h at 0°C then left to warm to room temperature for 1 h. The reaction was quenched with aqueous ammonium chloride solution and partitioned between CH2CI2 and H20. The aqueous layer was washed with CH2CI2, the organic layers combined, dried (MgS04), filtered and concentrated to dryness to give the title compound after chromatography ((SiO2, 1% MeOH in CH2C12). FAB MS (M +1) 238 Step C: Preparation of 2-Allyl-2- (-3-methoxy-phenyl)-4-methyl-morpholine-3- one Following the procedure described by S. Pansare et al., J. Org. Chem.

1998,63,4120-4124,2-hydroxy-2- (3-methoxy-phenyl)-4-methyl-morpholin-3-one, as described in Step C above, (0.133 g, 0.56 mmol) was dissolved in anhydrous CH2C12 (10 mL) under Ar, treated with allyltrimethylsilane (0.534 mL, 3.36 mmol), the mixture cooled to-23°C, then treated with TiC14 (0.37 mL, 3.36 mmol). After 15 min the reaction mixture was quenched slowly with saturated aqueous NH4Cl solution, left to warm to room temperature, and partitioned between CH2C12 and H20. The aqueous layer was washed with CH2C12, the organic layers combined, dried (Na2SO4), filtered and concentrated to dryness to give the title compound after chromatography (Si02,0-1% MeOH in CH2C12).

FAB MS (M +1) 262.

Step D: Preparation of 2- (-3-methoxy-phenyl)-4-methyl-2-propyl-morpholine- 3-one 2-Allyl-2- (-3-methoxy-phenyl)-4-methyl-morpholine-3-one, as described above in Step C, (0.400 g, 1.53 mmol) was dissolved in MeOH (20 mL), the solution purged with Ar, treated with 10% Pd/C (0.060 g), and hydrogenated under a H2 balloon. After 2 h the reaction mixture was filtered through a celite pad and concentrated to dryness to give the title compound.

Step E: Preparation of 2- (-3-hydroxy-phenyl)-4-methyl-2-propyl-morpholine- 3-one BBr3 (1M in CH2Cl2) (0.341 mL, 0.341 mmol) was added dropwise to a solution of 2- (-3-methoxy-phenyl)-4-methyl-2-propyl-morpholine-3-one, as described above in Step D, (0.06 g, 0. 227 mmol) in CH2CI2 (3 mL) at 0°C with stirring. The ice bath was removed and stirring was continued for 16 h. The reaction mixture was quenched with H20, concentrated to dryness, then dissolved in MeOH and purified by RP preparative HPLC on a PrepPak (95/5 to 5/95 H2O/CH3CN w/ 0.1% TFA, flow = 65 mL/min) to give the title compound. FAB MS (M +1) 250 Step F: Preparation of 4- 1-Amino-1- (3-methyl-3H-imidazol-4-yl)-ethyl-2- 3- (4-methyl-3-oxo-2-propyl-morpholin-2-yl)-phenoxy-benzonitril e bistrifluoroacetate Using the procedure described in Example 47, Step C, 2- (-3- hydroxy-phenyl)-4-methyl-2-propyl-morpholine-3-one, as described above in Step E, (0.026 g, 0.104 mmol) and 2-fluoro-4- 1- (-) amino-1- (3-methyl-3H-imidazol-4-yl)- ethyl-benzonitrile (as described in Example 24A, Step F) (0.026 g, 0.104 mmol) were reacted to prepare the title compound which was purified by RP preparative HPLC on a PrepPak (95/5 to 5/95 H20/CH3CN w/0.1% TFA, flow = 65 mL/min).

HRMS Theoretical = 474.2500, Measured = 474.2484, Analysis calculated for C29H33F3N50s'2.5 CF3C02H'0.75 H20: C, 49.77; H, 4.57; N, 9.07; Found: C, 49.76; H, 4.23; N, 9.22.

EXAMPLE 58

Preparation of 4- 1- amino-1- (3-methyl-3H-imidazol-4-yl)-ethyl-2- 3- (morpholine-4- sulfonyl)-phenoxyl-benzonitrile Step A: Preparation of 3-Methoxy benzenesulfonyl morpholine 3-Methoxybenzenesulfonyl chloride (0. 20 g, 0.968 mmol) in acetone (2 mL) at 0°C was treated with excess morpholine (0.253 mL, 2.9 mmol) with stirring. After 0.5 hr the reaction mixture was partitioned between CH2Cl2 (1 mL) and IN HCI, the organic layer separated, washed with brine, dried (Na2SO4), filtered and concentrated to dryness to give the title compound.

Step B: Preparation of 3-Hydroxybenzenesulfonylmorpholine Boron tribromide in CH2C12 (1 M) (3.84 mL, 3.84 mmol) was added to a solution of 3-methoxy benzenesulfonyl morpholine (0.3297 g, 1.28 mmol) in anhydrous CH2C12 (25 mL) at 0°C under N2. After stirring overnight the reaction was partitioned between CH2C12 and aqueous saturated NaHC03 solution, the organic layer separated, washed with H20, brine, and dried (Na2SO4). Filtration and concentration to dryness gave the title compound.

Step C : Preparation of 4- 1- amino-1- (3-methyl-3H-imidazol-4-yl)-ethyl-2- 3- (morpholine-4-sulfonyl)-phenoxy-benzonitrile Using the methods described in Example 24A, but substituting 3- hydroxybenzenesulfonylmorpholine (0.015 g, 0.061 mmol) and 2-fluoro-4- 1-amino- 1- (3-methyl-3H-imidazol-4-yl)-ethyl-benzonitrile (as described in Example 24A, Step F), the title compound was prepared.

FAB MS (M+1) = 468 EXAMPLE 59

Preparation of 2- 3- (3R-ethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-4- 5- (2- morpholin-4-yl-ethyl)-imidazol-1-ylmethyl benzonitrile Step A : Preparation of {2- 3- (4-cyano-3-fluoro-benzyl)-3H-imidazol- 4-yl-ethyl}-carbamic acid tert-butyl ester To a solution of Nr-pivaloyloxymethyl-Na-phthaloyl- histamine (J. C. Emmett, F. H. Holloway, and J. L. Turner, J. Chem. Soc., Perkin Trans. 1,1341, (1979)) (4.59 g, 0.0124 mmol) in acetonitrile (40 mL) was added 4- bromomethyl-2-fluoro-benzonitrile (as described in Example 1, Step D) (2.8 g, 0.013 mmol) and the mixture was heated to reflux for 18 hr. A white solid precipitate formed which after cooling to 0°C was collected by filtration to obtain the quaternary salt. This intermediate was dissolved in EtOH (100 mL), hydrazine (1.46 mL, 0.046 mmol) was added, and the mixture was heated at reflux for 4 hr. A white precipitate was observed and the reaction was cooled to 25°C. Dimethylphthalate (11.4 mL, 0.0699 mmol) was added and the mixture was again refluxed for 18 hr. After cooling to 25°C the precipitate was removed by filtration and washed with EtOAc. The filtrate was evaporated in vacuo and the residue was dissolved in THF (125 mL) and H20 (25 mL). To this solution was added solid Na2C03 (4.0 g, 0.0377 mmol) and BOC20 (4.47 g, 0.020 mmol) and the reaction was stirred for 18 hr. The THF was removed in vacuo and the mixture was partitioned with EtOAc and saturated NaHC03. The EtOAc layer was washed with brine, dried with MgS04, and evaporated in vacuo to obtain the title product after chromatography (silica gel, CH2Cl2: MeOH: NH40H/ 97: 3: 0.3.

Step B : Preparation of 4- 5- (2-amino-ethyl)-imidazol-1-ylmethyl-2-fluoro- benzonitrile dihydrochloride A solution of {2- 3- (4-cyano-3-fluoro-benzyl)-3H-imidazol-4-yl- ethyl}-carbamic acid tert-butyl ester (1.0 g, 0.0029 mmol) in EtOAc (30 mL) was cooled to-20°C and saturated with HCl gas. The cooling bath was removed and the reaction was stirred for 2 hr. The solvent was removed in vacuo to obtain the title compound.

Step C : Preparation of 2-fluoro-4- 5- (2-morpholin-4-yl-ethyl)-imidazol-l- ylmethyl-benzonitrile

To a solution of 4- 5- (2-amino-ethyl)-imidazol-1-ylmethyl-2-fluoro- benzonitrile dihydrochloride (0.92 g, 0.0029 mmol) in acetonitrile (150 mL) and triethylamine (3.2 mL) was added 2-bromoethyl ether (0.839 mL, 0.0067 mmol) and the mixture was refluxed for 48 hr. The solvents were removed in vacuo and the residue was dissolved in EtOAc which was washed twice with 1M HCl (100 mL).

The HCl layers were combined and adjusted to pH = 9 with solid Na2CO3 and extraxcted 3 times with EtOAc. The EtOAc layers were combined and dried with brine and MgS04. Removal of the EtOAc in vacuo yielded the title compound.

Step D: Preparation of 2- 3- (3R-ethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy- 4- 5- (2-morpholin-4-yl-ethyl)-imidazol-1-vlmethyl benzonitrile Using the methods described in Example 24A, but substituting 3R- ethyl-3- (3-hydroxy-phenyl)-azepan-2-one (as described in Example 8, Step B) and 2- fluoro-4- 5- (2-morpholin-4-yl-ethyl)-imidazol-1-ylmethyl-benzonitrile (as described above in Step C), the following compound was prepared: 2- 3- (3R-ethyl-1-methyl-2-oxo-azepan-3-yl)-phenoxy-4- 5- (2-morpholin-4-yl- ethyl) imidazol-1-ylmethyl benzonitrile FAB MS (M+1) = 542 EXAMPLE 60 In vitro inhibition of ras farnesvl 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 3H farnesyl diphosphate, Ras protein, 50 mM HEPES, pH 7.5, 5 mM MgC12,5 mM dithiothreitol, 10 iM 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 HCl 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 described in the above Examples 1-59 were tested for inhibitory activity against human FPTase by the assay described above and were found to have IC50 of < 30 gM.

EXAMPLE 61 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 iL): 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 MgCl2,10 pM 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.: 2). Reactions are initiated by the addition of GGTase and stopped at timed intervals (typically 15 min) by the addition of 200 LL 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. ICso values are determined with Ras peptide near KM concentrations. Enzyme and substrate concentrations for inhibitor

ICso determinations are as follows: 75 pM GGTase-I, 1.6 tM Ras peptide, 100 RM geranylgeranyl diphosphate.

The compounds of the instant invention described in the above Examples 1-59 are tested for inhibitory activity against human GGTase type I by the assay described above.

EXAMPLE 62 Cell-based in vitro ras famesylation 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 pCi [35S] methionine (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 immunoprecipitated with the ras-specific monoclonal antibody Y 13-259 (Furth, M. E. et al., J. Virol. 43: 294-304, (1982)). Following a 2 hour antibody incubation at 4°C, 200 u. L of a 25% suspension of protein A-Sepharose coated with rabbit anti rat IgG is added for 45 min. The immunoprecipitates are washed four times with IP buffer (20 nM HEPES, pH 7.5/1 mM EDTA/1% Triton X-100.0.5% deoxycholate/0.1%/SDS/ 0.1 M 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 farnesylated and nonfarnesylated ras proteins are compared to determine the percent inhibition of farnesyl transfer to protein.

EXAMPLE 63

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 Rail 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 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 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 concentration of the instant compound. Photomicrographs are taken 16 days after the cultures are seeded and comparisons are made.

EXAMPLE 64 Construction of SEAP reporter plasmid pDSE 100 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 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-II 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 pDSElOl was constructed as follows: A restriction fragment containing part of the SEAP gene coding sequence was cut out of the plasmid pGEM7zf (-)/SEAP using the restriction enzymes Apa I and KpnI. The ends of the linear DNA fragments were chewed back with the Klenow fragment of E. coli DNA Polymerase I. The"blunt ended"DNA containing the truncated SEAP gene was isolated by electrophoresing the digest in an agarose gel and cutting out the 1910 base pair fragment. This 1910 base pair fragment was ligated into the plasmid pCMV-RE-AKI which had been cut with Bgl-11 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.: 3)

Antisense strand N-terminal SEAP: 5'GAGAGAGCTCGAGGTTAACCCGGGT GCGCGGCGTCGGTGGT 3' (SEQ. ID. NO.: 4) Sense strand C-terminal SEAP: 5'GAGAGAGTCTAGAGTTAACCCGTGGTCC CCGCGTTGCTTCCT 3' (SEQ. ID. NO.: 5) Antisense strand C-terminal SEAP: 5'GAAGAGGAAGCTTGGTACCGCCACTG GGCTGTAGGTGGTGGCT 3' (SEQ. ID. NO.: 6) The N-terminal oligos (SEQ. ID. NO.: 4 and SEQ. ID. NO.: 5) were used to generate a 1560 bp N-terminal PCR product that contained EcoRI and HpaI restriction sites at the ends. The Antisense N-terminal oligo (SEQ. ID. NO.: 4) introduces an internal translation STOP codon within the SEAP gene along with the HpaI site. The C- terminal oligos (SEQ. ID. NO.: 5 and SEQ. ID. NO.: 6) were used to amplify a 412 bp C-terminal PCR product containing HpaI and HindIII restriction sites. The sense strand C-terminal oligo (SEQ. ID. NO.: 5) introduces the internal STOP codon as well as the HpaI site. Next, the N-terminal amplicon was digested with EcoRI and HpaI while the C-terminal amplicon was digested with HpaI and HindIII. The two fragments comprising each end of the SEAP gene were isolated by electrophoresing the digest in an agarose gel and isolating the 1560 and 412 base pair fragments.

These two fragments were then co-ligated into the vector pGEM7zf (-) (Promega) which had been restriction digested with EcoRI and HindIII and isolated on an agarose gel. The resulting clone, pGEM7zf (-)/SEAP contains the coding sequence for the SEAP gene from amino acids.

Construction of a constitutively expressing SEAP plasmid pCMV-SEAP 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 i to the SEAP gene.

The plasmid pCMVIE-AKI-DHFR (Whang et al, 1987) 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.: 7) Antisense strand: 5'GAGAGATCTCAAGGACGGTGACTGCAG 3' (SEQ. ID. NO.: 8) These two oligos generate a 991 base pair fragment with a SacI site incorporated by the sense oligo and a Bgl-II fragment incorporated by the antisense oligo. The PCR fragment is trimmed with SacI and Bgl-II and isolated on an agarose gel. The vector pCMV-AKI is cut with SacI and Bgl-II and the larger vector fragment isolated by agarose gel electrophoresis. The two gel isolated fragments are ligated at their respective SacI and Bgl-11 sites to create plasmid pCMV-AKI-InA The DNA sequence encoding the truncated SEAP gene is inserted into the pCMV-AKI-InA plasmid at the Bgl-II site of the vector. The SEAP gene is cut out of plasmid pGEM7zf (-)/SEAP (described above) using EcoRI and HindIII. The fragment is filled in with Klenow DNA polymerase and the 1970 base pair fragment isolated from the vector fragment by agarose gel electrophoresis. The pCMV-AKI- InA vector is prepared by digesting with Bgl-II and filling in the ends with Klenow DNA polymerase. The final construct is generated by blunt end ligating the SEAP fragment into the pCMV-AKI-InA vector. Transformants were screened for the proper insert and then mapped for restriction fragment orientation. Properly oriented recombinant constructs were sequenced across the cloning junctions to verify the correct sequence. The resulting plasmid, named pCMV-SEAP, 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.

Cloning of a Myristylated viral-H-ras expression plasmid A DNA fragment containing viral-H-ras can be PCRed from plasmid "H-1" (Ellis R. et al. J. Virol. 36,408,1980) or"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.: 9) Antisense: 5'CACATCTAGATCAGGACAGCACAGACTTGCAGC 3'.

(SEQ. ID. NO.: 10) 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 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 A viral-H-ras clone with a C-terminal sequence encoding the amino acids CVLL can be cloned from the plasmid"H-1" (Ellis R. et al., J. Virol. 36,408, 1980) or"HB-11 (deposited in the ATCC under Budapest Treaty on August 27,1997, and designated ATCC 209,218) by PCR using the following oligos.

Sense strand:

5'TCTCCTCGAGGCCACCATGACAGAATACAAGCTTGTGGTGG-3' (SEQ. ID. NO.: 11) Antisense strand: 5'CACTCTAGACTGGTGTCAGAGCAGCACACACTTGCAGC-3' (SEQ. ID. NO.: 12) 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 in which the mutated viral- H-ras-CVLL gene is constitutively transcribed from the CMV promoter of the pCI vector.

Cloning of c-H-ras-Leu61 expression plasmid The human c-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.: 13) Antisense strand: 5'-GAGAGTCGACGCGTCAGGAGAGCACACACTTGC-3' (SEQ. ID. NO.: 14) 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 stite 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.: 15)

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 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 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.: 16) Antisense strand: 5'-GAGAGTCGACTTGTTACATCACCACACATGGC-3' (SEQ. ID. NO.: 17) 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 stite 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.: 18) 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 will constitutively transcribe c-N-ras-Val-12 from the CMV promoter of the pCI vector.

Cloning of a c-K-ras-Val-12 expression plasmid

The human c-K-ras gene can be PCRed from a human cerebral cortex cDNA library (Clontech) using the following oligonucleotide primers.

Sense strand: 5'-GAGAGGTACCGCCACCATGACTGAATATAAACTTGTGG-3' (SEQ. ID. NO.: 19) Antisense strand: 5'-CTCTGTCGACGTATTTACATAATTACACACTTTGTC-3' (SEQ. ID. NO.: 20) The primers will amplify a c-K-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-K-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.: 21) After selection and sequencing for the correct nucleotide substitution, the mutated c-K-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 1. The new recombinant plasmid will constitutively transcribe c-K-ras-Val-12 from the CMV promoter of the pCI vector.

SEAP assay Human C33A cells (human epitheial carcenoma-ATTC collection) are seeded in 10cm tissue culture plates in DMEM + 10% fetal calf serum + 1X Pen/Strep + 1X glutamine + 1X NEAA. Cells are grown at 37°C in a 5% C02 atmosphere until they reach 50-80% of confluency.

The transient transfection is performed by the CaP04 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. For 10cm

plates 600ml of CaCl2-DNA solution is added dropwise while vortexing to 600ml of 2X HBS buffer to give 1.2ml of precipitate solution (see recipes below). This is allowed to sit at room temperature for 20 to 30 minutes. While the precipitate is forming, the media on the C33A cells is replaced with DMEM (minus phenol red; Gibco cat. # 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 1 Oml of phenol red free DMEM + 0.2% charcoal stripped calf serum + 1 X (Pen/Strep, Glutamine and NEAA). Transfected cells are plated in a 96 well microtiter plate (l00ml/well) to which drug, diluted in media, has already been added in a volume of 100ml. The final volume per well is 200ml with each drug concentration repeated in triplicate over a range of half-log steps.

Incubation of cells and test compound is for 36 hrs at 37°C under CO2.

At the end of the incubation period, cells are examined microscopically for evidence of cell distress. Next, 100 ml of 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 CSPDt (Tropix, Bedford, Mass.). A volume of 50 ml media is combined with 200 ml 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 1 Ocm. plate of cells Ras expression plasmid (lmg/ml) lOml DSE-SEAP Plasmid (lmg/ml) 2ml Sheared Calf Thymus DNA (lmg/ml) 8ml 2M CaCl2 74ml dH20 506ml

2X HBS Buffer 280mM NaCl lOmM KCl 1.5mMNa2HP042H20 12mM dextrose 50mM HEPES Final pH = 7.05 Luminesence Buffer (26ml) Assay Buffer 20ml Emerald ReagentTM (Tropix) 2.5ml 100mM homoarginine 2.5ml CSPD Reagent (B) (Tropix) 1.0ml Assay Buffer Add 0.05M Na2CO3 to 0.05M NaHC03 to obtain pH 9.5.

Make 1mM in MgCl2 EXAMPLE 65 The processing assays employed in this example and in Example 66 modifications of that described by DeClue et al [Cancer Research 51,712-717,1991].

K4B-Ras processing inhibition assay PSN-1 (human pancreatic carcinoma) 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 glutamine (Gibco), 2% fetal bovine serum, respectively) containing the desired concentration of test compound, lovastatin or solvent alone. Cells treated with lovastatin (5-10 1M), 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 pCi/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 tempera- ture), and lysed by vortexing in 1 ml of lysis buffer (1% Nonidet P-40,20 mM HEPES, pH 7.5,150 mM NaCI, 1 mM EDTA, 0.5% deoxycholate, 0.1% SDS, 1 mM DTT, 10 llg/ml AEBSF, 10 pg/ml aprotinin, 2 pg/ml leupeptin and 2 pg/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 appropri- ate volume of lysate is brought to 1 ml with lysis buffer lacking DTT and 8 pg 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 ml 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 NaCI, 10 mM Tris pH 7.4) with 2 mg 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% CO2.

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 IC50 values are generated using 4-parameter curve fits in SigmaPlot software.

EXAMPLE 66 K4B-Ras processing inhibition assay PSN-1 (human pancreatic carcinoma) cells are used for analysis of protein processing. Subconfluent cells in 150 mm dishes are fed with 20 ml of media (RPMI supplemented with 15% fetal bovine serum) containing the desired concentration of prenyl-protein transferase inhibitor or solvent alone. Cells treated with lovastatin (5-10 I1M), 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%.

The cells are incubated at 37°C for 24 hours, the media is then removed and the cells are washed twice with cold PBS. The cells are scraped into 2 ml of cold PBS, collected by centrifugation (10,000 x g for 5 min at 4°C) and frozen at-70°C. Cells are lysed by thawing and addition of lysis buffer (50 mM HEPES, pH 7.2,50 mM NaCl, 1% CHAPS, 0.7 pg/ml aprotinin, 0.7 llg/ml leupeptin 300 pg/ml pefabloc, and 0.3 mM EDTA). The lysate is then centrifuged at 100,000 x g for 60 min at 4°C and the supernatant saved. The supernatant may be subjected to SDS- PAGE, HPLC analysis, and/or chemical cleavage techniques.

The lysate is applied to a HiTrap-SP (Pharmacia Biotech) column in buffer A (50 mM HEPES pH 7.2) and resolved by gradient in buffer A plus 1 M NaCl. Peak fractions containing Ki4B-Ras are pooled, diluted with an equal volume of water and immunoprecipitated with the pan Ras monoclonal antibody, Y13-259 linked to agarose. The protein/antibody mixture is incubated at 4°C for 12 hours.

The immune complex is washed 3 times with PBS, followed by 3 times with water.

The Ras is eluted from the beads by either high pH conditions (pH>10) or by heating at 95°C for 5 minutes, after which the beads are pelleted by brief centrifugation. The supernatant may be subjected to SDS-PAGE, HPLC analysis, and/or chemical cleavage techniques.

EXAMPLE 67 Rapl processing inhibition assay Protocol A: Cells are labeled, incubated and lysed as described in Example 65.

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 ml 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 mg 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 Rapl 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 10 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 Ix 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 37ocC 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 0.1-1001lu.

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 data point, a lOmM stock of the compound is needed).

2gL of each 1000x compound stock is diluted into 1mI media to produce a 2X stock of compound. A vehicle control solution (2pL 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 uL SDS-PAGE sample buffer (Novex) containing 5% 2-mercaptoethanol 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, 20gel 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), 0.5M Tris-HCl pH8.0 and 50mM MgC12.

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 ul of each sample is loaded onto the gel. The gel is run at 15mA for about 3.5 hours. It is important to run the gel far enough so that there will be adequate separation between 21kd (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 Rap 1 a antibody (Santa Cruz Biochemical SC 1482) 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 2ml 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 Rap 1 a Minimum Inhibitory Concentration is determined from the lowest concentration of compound that produces a detectable Rapl a Western signal. The Rap 1 a antibody used recognizes only unprenylated/unprocessed Rap 1 a, so that the

precence of a detectable Rap 1 a Western signal is indicative of inhibition of Rap 1 a prenylation.

Protocol C: This protocol allows the determination of an ECso for inhibition of processing of Rapl a. The assay is run as described in Protocol B with the following modifications. 20 1ll 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 Rap 1 a are detected by blotting with a polyclonal antibody (Rap 1/Krev-1 Ab# 121; Santa Cruz Research Products #sc-65), followed by an alkaline phosphatase- conjugated anti-rabbit IgG antibody. The percentage of unprenylated Rap 1 a relative to the total amount of Rap 1 a is determined by peak integration using Imagequant" software (Molecular Dynamics). Unprenylated Ray 1 a is distinguished from prenylated protein by virtue of the greater apparent molecular weight of the prenylated protein.

Dose-response curves and ECso values are generated using 4-parameter curve fits in SigmaPlot software.

EXAMPLE 68 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 injecte subcutaneously into the left flank of 8-12 week old female nude mice (Harlan) on day 0. The mice in each oncogene group are randomly assigned to a vehicle or compound treatment group. Animals are dosed subcutaneously starting on day 1 and daily for the duration of the experiment. Alternatively, the prenyl-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 injecte. The average weight of the tumors in each treatment group for each cell line is calculated.