CA2243320A1 - Inhibitors of farnesyl-protein transferase - Google Patents

Inhibitors of farnesyl-protein transferase Download PDF

Info

Publication number
CA2243320A1
CA2243320A1 CA 2243320 CA2243320A CA2243320A1 CA 2243320 A1 CA2243320 A1 CA 2243320A1 CA 2243320 CA2243320 CA 2243320 CA 2243320 A CA2243320 A CA 2243320A CA 2243320 A1 CA2243320 A1 CA 2243320A1
Authority
CA
Canada
Prior art keywords
hydrogen
substituted
alkyl
aryl
unsubstituted
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
CA 2243320
Other languages
French (fr)
Inventor
Christopher J. Dinsmore
George D. Hartman
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Merck and Co Inc
Original Assignee
Individual
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from GBGB9605708.8A external-priority patent/GB9605708D0/en
Application filed by Individual filed Critical Individual
Priority claimed from PCT/US1997/001354 external-priority patent/WO1997027852A1/en
Publication of CA2243320A1 publication Critical patent/CA2243320A1/en
Abandoned legal-status Critical Current

Links

Landscapes

  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The present invention is directed to compounds which inhibit farnesyl-protein transferase (FTase) and the farnesylation of the oncogene protein Ras. The invention is further directed to chemotherapeutic compositions containing the compounds of this invention and methods for inhibiting farnesyl-protein transferase and the farnesylation of the oncogene protein Ras.

Description

TITLE OF THE INVE~NTION
INHIBITO~S OF FARNESYL-PROTEIN TRANSFERASE

BACKGROUND OF THE INVENTION
The Ras proteins (Ha-Ras, Ki4a-Ras, Ki4b-Ras and N-Ras) are part of . .~ign~llin~ pathway that links cell surface growth factor receptors to nuclear signals initiating cellular proliferation. Biological and bioche nical studies of Ras action indicate that Ras functions like a G-regulato~ protein. In the inactive state, Ras is bound to GDP. Upon growth fac-or receptor activation Ras is induced to exchange GDP for GTP and u:ldergoes a conformational change. The GTP-bound form of Ras propagates the growth stim~ tory signal until the signal is termin~te~l by the intrinsic GTPase activity of Ras, which returns the protein to i :s inactive GDP bound form (D.R. Lowy and D.M.
Willumsen. Ann. Rev. Biochem. 62:851-891 (1993)). Mutated ras genes (Ha- as, 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 loc~li7e~ to the plasma membrane for both normal anc oncogenic functions. At least 3 post-translational modifications are involved with Ras membrane loc~li7~tion, and all 3 modifications occur at the C-terminus of Ras. The Ras C-terminus contains a ~equence motif termed a "CAAX" or "Cys-Aaal-Aaa2-Xaa"
box (Cys i- cysteine, Aaa is an aliphatic amino acid, the Xaa is any amino acid) (Willumsen et al., Nature 310:583-586 (19~4)). Depending on the speci~lc sequence, this motif serves as a signal sequence for the enzymes f-.rnesyl-protein transferase or geranylgeranyl-protein transferase, which catalyze the alkylation of the cysteine residue of the CAAX mo:if with a Cls or C20 isoprenoid, respectively. (S. Clarke., Ann. Rev. 3iochem. 61:355-386 (1992); W.R. Schafer and J. Rine, Ann. Rev. Genetics 30:209-237 (1992)). The Ras protein is one of several pr~teins 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 l~min.~, and the gAmm~ subunit of transducin. James, et al., J. Biol. Chem. 269, 14182 (1994) have identified a peroxisome associated protein Pxf which S 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 transforrned 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 m~m~ry and salivary carcinomas in ras transgenic mice (N.E. Kohl et al., Nature Medicine, 1:792-797 (1995).
Indirect inhibition of farnesyl-protein transferase in vivo has been demonstrated with lovastatin (Merck & Co., Rahway, NJ) and compactin (Hancock et al., ibid; Casey et al., ibid; Schafer et al., Science 245:379 (1989)). These drugs inhibit HMG-CoA reductase, the rate limiting enzyme for the production of polyisoprenoids including farnesyl pyrophosphate. Parnesyl-protein transferase lltili7es 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 el al., Science, 249:1133-1139 (1990); Manne et al., Proc. Natl. Acad. Sci USA,87:7541-7545(1990)). Inhibitionoffarnesylpyrophosphate biosynthesis by inhibiting HMG-CoA reductase blocks Ras membrane loc~li7~tion in cultured cells. However, direct inhibition of farnesyl-protein transferase would be more specific and attended by fewer side effects thar would occur with the required dose of a general inhibitor of isoprene biosynthesis.
Inhibitors of farnesyl-protein transferase (FPTase) have been descri ~ed in two general classes. The first are analogs of farnesyl 5 diphosphate (FPP), while the second class of inhibitors is related to the protein sub-trates (e.g., Ras) for the enzyme. The peptide derived inhibitors that have been described are generally cysteine cont~inin~;
molecules tlat are related to the CAAX motif that is the signal for protein prenylation. (Schaber et al., ibid; Reiss et. al., ibid; Reiss et al., 0 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); Grahaln, et al., J. Med. Chem., 37, 725 (1994)).
15 In general, leletion of the thiol from a CAAX derivative has been shown to d-amatically reduce the inhibitory potency of the compound.
However, t~e thiol group potentially places limitations on the therapeutic application of FPTase inhibitors with respect to pharmacok netics, pharrnacodynarnics and toxicity. Therefore, a 20 functional replacement for the thiol is desirable.
It has recently been reported that famesyl-protein transferase inhibitors are inhibitors of proliferation of vascular smooth muscle cell and are therefore useful in the prevention and thereapy of arteriosclerosis and diabetic disturbance of blood vessels (JP H7-25 112930).
It has recently been disclosed that certain tricycliccompounds which optionally incorporate a piperidine moiety are inhibitors of FPTase (WO 95/10514, WO 95/10515 and WO 95/10516).
Imidazole-cont~inin~ inhibitors of farnesyl protein transferase have also 30 been disclo ed (WO 95/09001 and EP 0 675 112 A1).
It is, therefore, an object of this invention to develop peptidomir~etic compounds that do not have a thiol moiety, and that will inhibit farnesyl-protein transferase and thus, the post-translational farnesylation of proteins. It is a further object of this invention to WO 97n7852 PCT~US97/01354 develop chemotherapeutic compositions cont~ining the compounds of this invention and methods for producing the compounds of this mventlon.

S SUMMARY OF THE INVENTION
The present invention comprises small molecule peptidomim~o,tic amide-cont~inin~ compounds which inhibit the farnesyl-protein transferase. The instant compounds lack a thiol moiety and thus offer unique advantages in terms of improved pharmacokinetic behavior 10 in ~nim~l~, prevention of thiol-dependent chemical reactions, such as rapid autoxidation and disulfide formation with endogenous thiols, and re~ ce-1 systemic toxicity. Further contained in this invention are chemotherapeutic compositions cont~inin~ these farnesyl transferase inhibitors and methods for their production.
The compounds of this invention are illustrated by the formula I:

( I 6)r ~1 7~ /R3 V - A1 (CR1 a2)nA2(CR 1b2)n ~Wl - (CR22)p--A3--DETAILED DESCRIPTION OF THE INVENTION
The compounds of this invention are useful in the inhibition of farnesyl-protein transferase and the farnesylation of the oncogene protein Ras. In a first embodiment of this invention, the inhibitors of 25 farnesyl-protein transferase are illustrated by the formula I:

( I 6)r ~1 7~ /R3 V - A1(CRta2)nA2(CR1b2)n ~W~!- (CR22~p--A3--y wherein:
R1a, R1b a~d R2 are independently selected from:
a) hydrogen, b) aryl, heterocycle, C3-Clo cycloaLkyl, C2-c6 alkenyl, C2-C6 alkynyl, R8O-, R9s(o)m-~ R8C(O)NR8-, CN, NO2, (R8)2N-C(NR8)-, R8C(O)-, R8OC(O)-, N3, -N(R8)2, or R9OC(o)NR8-, c) Cl-C6 alkyl unsubstituted or substituted by aryl, heterocyclic, C3-C1o cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, R8O-, R9S(o)m-~ R8C(O)NR8-, CN, (R8)2N-C(NR8)-, R8C(O)-, R8OC(O)-, N3, -N(R8)2, or R9OC(o)-NR8-;
R3 and R4 are independently selected from F, Cl, Br, N(R8)2, CF3, N02, (R8)o-, (R9)S(o)m-~ (R8)C(O)NH-, H2N-C(NH)-, (R8)C(O)-, (R8)OC(O)-, N3, CN, CF3(CH2)nO-, (R9)OC(O)NR8-, C]-c2o alkyl, substituted or unsubstituted aryl and substituted or unsubstituted heterocycle;

R5 is selected from:
a) hydrogen, b) unsubstituted or substituted aryl, c) unsubstituted or substituted heterocyclic, d) unsubstituted or substituted C3-Clo cycloalkyl, and e) Cl-C6 alkyl substituted with hydrogen or a group selected from unsubstituted or substituted aryl, unsubstituted or substituted heterocyclic, unsubstituted or substituted C3-C10 cycloalkyl, N(R8)2, CF3, N02, (R8)o-, (R9)S(o)m-~ (R8)C(O)NH-, H2N-c(NH)-~ (R8)C(O)-, (R8)OC(O)-, N3, CN (R9)OC(O)NR8-;
s R6 is independently selected from:
a) hydrogen, b) aryl, heterocycle, C3-Clo cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, perfluoroaL~yl, F, Cl, Br, R80-, R9S(o)m-, R8C(O)NR8-, CN, NO2, R82N-C(NR8)-, R8C(O)-, R8OC(O)-, N3, -N(R8)2, or R9OC(o)NR8-, and c) Cl-C6 aLkyl unsubstituted or substituted by aryl, heterocycle, C3-Clo cycloaL~yl, C2-c6 aL~enyl, C2-C6 alkynyl, perfluoroalkyl, F, Cl, Br, R8O-, R9S(o)m-~
R8C(O)NH-, CN, H2N-c(NH)-~ R8C(O)-, R8OC(O)-, N3, -N(R8)2, or R8OC(O)NH-;

R7 is selected from:
a) hydrogen, b) C2-C6 alkenyl, C2-C6 alkynyl, perfluoroalkyl, F, Cl, Br, R8O-, R9S(o)m-~ R8C(O)NR8-, CN, NO2, (R8)2N-C-(NR8)-, R8C(O~-, R8OC(O)-, N3, -N(R8)2, or R9OC(o)NR8-, and c) Cl-C6 alkyl unsubstituted or substituted by perfluoroalkyL
F, Cl, Br, R8O-, R9S(o)m-~ R8C(O)NR8-, CN, (R8)2N-C(NR8)-, R8C(O)-, R8OC(O)-, N3, -N(R8)2, or R9OC(o)NR8-;

R8 is independently selected from hydrogen, Cl-C6 alkyl, benzyl and aryl;

~ R9 is independently selected from Cl-C6 alkyl and aryl;

A l and A2 are independently selected from: a bond, -CH=CH-, -C_C-, CA 02243320 l998-07-l7 -C(O)-? -C(O)NR8-, -NR8C(O)-, O, -N(R8)-, -S(0)2N(R8)-, -N(R8)S(0)2-, or S(O)m;

A3 is selected from: -NR5C(o)- or -C(O)NH-;
s V is selected from:
a) hydrogen, b) heterocycle, c) aryl, d) Cl-C20 aL~yl wherein from O to 4 carbon atoms are replaced with a a heteroatom selected from 0, S, and N, and e) C2-C20 aL~enyl, provided th~t V is not hydrogen if A1 is S(O)m and V is not hydrogen 15 if A1 is a bond, n is O and A2 is S(O)m;

W is a heterocycle;

Y is aryl or heteroaryl;
m is O, 1 or 2;
nis 0, 1, 2, 3 or4;
p is I 0, 1, 2, 3 or 4;
r is O to 5, provided that r is O when V is hydrogen; and tis Oorl;

or the pharmaceutically acceptable salts thereof.

A preferred embodiment of the compounds of this 30 invention are illustrated by the formula Ia:

WO 971278~2 PCTIUS97/01354 ( l 6)r R7a V - A1(CRla2)nA2(CR1b2)~N~_~J ~3 (CR22~--A3 {

wherein:

5 R1a and R1b are independently selected from: hydrogen or C1-C6 alkyl;

R2 is independently selected from:
a) hydrogen, b) aryl, heterocycle, cycloalkyl, R8O-, -N(R~)2 or C2-C6 aLkenyl, c) C 1-C6 alkyl unsubstituted or substituted by aryl, heterocycle, cycloalkyl, alkenyl, R80-, or-N(R8)2;

15 R3 and R4 are independently selected from F, Cl, Br, N(R8)2, CF3, NO2, (R8)o-, (R9)S(O)m-, (R8)C(O)NH-, H2N-C(NH)-, (R8)C(O)-, (R8)OC(O)-, N3, CN, (R9)OC(O)NR8-, Cl-C20 alkyl, substituted or unsubstituted aryl and substituted or unsubstituted heterocycle;

R5 is selected from:
a) hydrogen, and b) C1-C6 alkyl substituted with hydrogen or a group selected from unsubstituted or substituted aryl, unsubstituted or substituted heterocyclic, unsubstituted or substituted C3-C1o cycloalkyl, N(R8)2~ CF3, N02, (R8)o-, WO 97/27852 PCT/~JS97/01354 (R9)S(o)m-~ (R8)C(O)NH-, H2N-C(NH)-, (R8)C(O)-, (R8)OC(O)-, N3, CN (R9)OC(O)NR8-;

R6 is indep~-ndently selected from:
a) hydrogen, b) Cl-c6 alkyl, C2-c6 alkenyl, C2-c6 alkynyl, Cl-c6 perfluoroalkyl, F, Cl, R8O-, R8C(O)NR8-, CN, NO2, (R8)2N-C(NR8)-, R8C(O)-, R8OC(O)-, -N(R8)2, or R9OC(o)NR8-, and c) Cl-C6 alkyl substituted by Cl-C6 perfluoroaL~yl, R80-, R8C(O)NR8-, (R8)2N-C(NR8)-, R8C(O)-, R80C(O)-, -N(R8)2, or R9OC(o)NR8-;

R7a is hydrogen or methyl;
R8 is independently selected from hydrogen, Cl-C6 alkyl, benzyl and aryl;

R9 is independently selected from Cl-C6 aL~yl and aryl;
A l and A2 are independently selected from: a bond, -CH=CH-, -C~C-, -C(O)-, -C(O)NR8-, O, -N(R8)-, or S(O)m;

A3 is selected from: -NR5C(o)- or -C(O)NH-;
V is selected from:
a) hydrogen, b) heterocycle selected from pyrrolidinyl, imidazolyl, pyridinyl, thiazolyl, pyridonyl, 2-oxopiperidinyl, indolyl, quinolinyl, isoquinolinyl, and thienyl, c) aryl, d) Cl-C20 alkyl wherein from 0 to 4 carbon atoms are replaced with a a heteroatom selected from O, S, and N, and WO 97n7852 PCT/US97101354 e) C2-C20 aLkenyl, and provided that V is not hydrogen if A1 is S(O)m and V is not hydrogen if A1 is a bond, n is 0 and A2 is S(O)m;

S mis 0, 1 or 2;
nis 0, 1, 2, 3 or4;
p is 0, 1, 2, 3 or 4; and r is 0 to S, provided that r is 0 when V is hydrogen;

10 or the pharmaceutically acceptable salts thereof.

A second preferred embodiment of the compounds of this invention are illustrated by the formula Ic:

V-A1(CR1az)nA2(cR1b2)n(w~-(cR22)p A3{~

wherein:

Rla and R1b are independently selected from: hydrogen or C1-C6 alkyl;
R2 is independently selected from:
a) hydrogen, b) aryl, heterocycle, cycloalkyl, R80-, -N(R8)2 or C2-C6 alkenyl, c) C1-C6 alkyl unsubstituted or substituted by aryl, heterocycle, cycloalkyl, aLkenyl, R80-, or -N(R8)2;

R3 and R4 are independently selected from F, Cl, Br, N(R8)2~ CF3, N02, (R8)o-, (R9)S(o)m-, (R8)C(O)NH-, H2N-C(NH)-, (R8)C(O)-, (R8)OC(O)-, N3, CN, (R9)OC(O)NR8-, Cl-C20 alkyl, substituted or unsubstituted aryl and substituted or unsubstituted heterocycle;
s R5 is selected from:
a) hydrogen, and b) C1-C6 alkyl substituted with hydrogen or a group selected from unsubstituted or substituted aryl, unsubstituted or substituted heterocyclic, unsubstituted or substituted C3-Clo cycloalkyl, N(R8)2, CF3, N02, (R8)o-, (R9)S(o)m-~ (R8)C(O)NH-, H2N-C(NH)-, (R8)C(O)-, (R8)OC(O)-, N3, CN (R9)OC(O)NR8-;
R6 is independently selected from:
a) hydrogen, b) Cl-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C~-C6 perfluoroalkyl, F, Cl, R80-, R8C(O)NR8-, CN, N02, (R8)2N-C(NR8)-, R8C(O)-, R8OC(O)-, -N(R8)2, or R9OC(o)NR8-, and c) Cl-c6 alkyl substituted by Cl-c6 perfluoroalkyl, R8O-, R8C(O)NR8-, (R8)2N-C(NR8)-, R8C(O)-, R80C(O)-, -N(R8)2, or R9OC(o)NR8-;
R7 is selected from: hydrogen and Cl-C6 aLkyl;

R8 is indep~ndently selected from hydrogen, Cl-c6 alkyl, benzyl and aryl;
R9 is independently selected from Cl-C6 aLkyl and aryl;

Al and A2 are independently selected from: a bond, -CH=CH-, -C-C-, -C(O)-, -C(O)NR8-, O, -N(R8)-, or S(O)m;

WO 97t27852 rCT/US97101354 A3 is selected from: -NRSC(O)- or -C(O)NH-;

V is selected from:
a) hydrogen, b) heterocycle selected from pyrrolidinyl, imidazolyl, pyridinyl, thiazolyl, pyridonyl, 2-oxopiperidinyl, indolyl, quinolinyl, isoquinolinyl, and thienyl, c) aryl, d) Cl-C20 alkyl wherein from 0 to 4 carbon atoms are replaced with a a heteroatom selected from O, S, and N, and e) C2-C20 aLkenyl, and provided that V is not hydrogen if Al is S(O)m and V is not hydrogen 15 if Al is a bond, n is 0 and A2 is S(O)m;

W is a heterocycle selected from pyrrolidinyl, pyridinyl, thiazolyl, pyridonyl, 2-oxopiperidinyl, indolyl, quinolinyl, or isoquinolinyl;

m is 0, 1 or 2;
n is 0, 1, 2, 3 or 4;
p is 0, 1, 2, 3 or 4;
r is 0 to 5, provided that r is 0 when V is hydrogen; and tis l;
or the pharrnaceutically acceptable salts thereof.

In a more preferred embodiment of this invention, the inhibitors of farnesyl-protein transferase are illustrated by the formula 30 Ic:

)~N
(\CR2 )--A3~l~

R6 Ic R4 wherein:

Rlb is independently selected from:
a) hydrogen, b) aryl, heterocycle, cycloalkyl, R8O-, -N(R8)2 or C2-C6 alkenyl, c) Cl-C6 aLkyl unsubstituted or substituted by aryl, heterocycle, cycloaLkyl, alkenyl, R80-, or-N(R8)2;
R2 are independently selected from: hydrogen or Cl-C6 aLkyl;

R3 and R4 are independently selected from F, Cl, Br, N(R8)2, CF3, N02, (R8)o-, (R9)S(o)m-~ (R8)C(O)NH-, H2N-C(NH)-, (R8)C(O)-, (R8)OC(O)-, N3, CN, (R9)OC(O)NR8-, Cl-C20 alkyl, substituted or unsubstituted aryl and substituted or unsubstituted heterocycle;

R5 is selected from:
a) hydrogen, and b) Cl-C6 alkyl substituted with hydrogen or a group selected from unsubstituted or substituted aryl, unsubstituted or substituted heterocyclic, unsubstituted or substituted C3-Clo cycloalkyl, N(R8)2~ CF3, N02, (R8)o-, (R9)S(o)m-~ (R8)C(O)NH-, H2N-C(NH~-, (R8)C(O)-, (R8)0C(O)-, N3, CN (R9)OC(o)NR8-;

WO 97n7852 PCT/US97/01354 R6 is independently selected from:
a) hydrogen, b) Cl-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, Cl-C6 perfluoroalkyl, F, Cl, R8O-, R8C(O)NR8-, CN, NO2, (R8)2N-C(NR8)-, R8C(O)-, R8OC(O)-, -N(R8)2, or R9OC(o)NR8-, and c) Cl-C6 aLkyl substituted by Cl-c6 perfluoroalkyl, R80-, R8C(O)NR8-, (R8)2N-C(NR8)-, R8C(O)-, R80C(O)-, -N(R8)2, or R9OC(o)NR8-;

R8 is independently selected from hydrogen, Cl-C6 aLkyl, benzyl and aryl;
15 R9 is independently selected from Cl-C6 aLkyl and aryl;
A3 is selected from: -NRSC(O)- or -C(O)NH-;

mis 0,lor2;and 20 pis 0, 1, 2, 3 or4;
or the pharmaceutically acceptable salts thereof.

In a second more preferred embodiment of this invention, 25 the inhibitors of farnesyl-protein transferase are illustrated by the formula Id:

N \J R3 (CR22)p A3 c~4 NC Id n whereln:

Rlb is independently selected from:
a) hydrogen, b) aryl, heterocycle, cycloalkyl, R8O-, -N(R8)2 or C2-C6 alkenyl, c) Cl-C6 alkyl unsubstituted or substituted by aryl, heterocycle, cycloaL~yl, alkenyl, R80-, or-N(R8)2;

10 R2 are independently selected from: hydrogen or Cl-C6 alkyl;

R3 and R4 are independently selected from F, Cl, Br, N(R8)2, CF3, N02, (R8)o-, (R9)S(o)m-~ (R8)C(O)NH-, H2N-C(NH)-, (R8)C(O)-, (R8)OC(O)-, N3, CN, (R9)OC(O)NR8-, Cl -C20 alkyl, substituted or unsubstituted aryl and substituted or unsubstituted heterocycle;

RS is selected from:
a) hydrogen, and b) Cl-C6 alkyl substituted with hydrogen or a group selected from unsubstituted or substituted aryl, unsubstituted or substituted heterocyclic, unsubstituted or substituted C3-Clo cycloaL~yl, N(R8)2, CF3, N02, (R8)o-, (R9)S(o)m-~ (R8)C(O)NH-, H2N-c(NH)-~ (R8)C(O)-, (R8)OC(O)-, N3, CN (R9)OC(O)NR8-;

R8 is independently selected from hydrogen, Cl-c6 alkyl, benzyl and aryl;

R9 is independently selected from Cl-C6 alkyl and aryl;

A3 is selected from: -NR5C(o)- or -C(O)NH-;

mis 0,lor2;and pis 0, 1, 2, 3 or4;

5 or the pharmaceutically acceptable salts thereof.

Specific examples of the compounds of the invention are:

N-(3-chlorophenyl)-3- [ 1 -(4-cyanobenzyl)-5-imidazolyl]propionamide 10 hydrochloride (1) Cl N

or the pharmaceutically acceptable salts 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 (e.g. aryl, heterocycle, Rla, R2 etc.) occurs more than one time in any 20 constituent, its definition on each occurence is independent at every other occurence. Also, combinations of substituents/or variables are permissible only if such combinations result in stable compounds.
As used herein, "alkyl" is intended to include both branched and straight-chain saturated aliphatic hydrocarbon groups having the 25 specified number of carbon atoms; "alkoxy" represents an alkyl group of indicated number of carbon atoms attached through an oxygen WO 97t27852 PCT/US97/013~i4 bridge. "Halogen" or "halo" as used herein means fluoro, chloro, bromo and iodo.
As used herein, "aryl" is intended to mean any stable monocyclic or bicyclic carbon ring of up to 7 members in each ring, S wherein at least one ring is aromatic. Examples of such aryl elements include phenyl, naphthyl, tetrahydronaphthyl, indanyl, biphenyl, phenanthryl, anthryl or acenaphthyl.
The term heterocycle or heterocyclic, as used herein, represents a stable S- to 7-membered monocyclic or stable 8- to 11-10 membered bicyclic heterocyclic ring which is either saturated orn~tllrated, and which consists of carbon atoms and from one to four heteroatoms selected from the group consisting of N, O, and S, and including any bicyclic group in which any of the above-defined heterocyclic rings is fused to a benzene ring. The heterocyclic ring may lS be attached at any heteroatom or carbon atom which results in the creation of a stable structure. Examples of such heterocyclic elements include, but are not limited to, azepinyl, benzimidazolyl, benzisoxazolyl, benzofurazanyl, benzopyranyl, benzothiopyranyl, benzofuryl, benzothiazolyl, benzothienyl, benzoxazolyl, chromanyl, cinnolinyl, 20 dihydrobenzofuryl, dihydrobenzothienyl, dihydrobenzothiopyranyl, dihydrobenzothiopyranyl sulfone, furyl, imidazolidinyl, imidazolinyl, imidazolyl, indolinyl, indolyl, isochromanyl, isoindolinyl, isoquinolinyl, isothiazolidinyl, isothiazolyl, isothiazolidinyl, morpholinyl, naphthyridinyl, oxadiazolyl, 2-oxoazepinyl, 2-oxopiperazinyl, 2-25 oxopiperdinyl, 2-oxopyrrolidinyl, piperidyl, piperazinyl, pyridyl, pyrazinyl, pyrazolidinyl, pyrazolyl, pyridazinyl, pyrimidinyl, pyrrolidinyl, pyrrolyl, quinazolinyl, quinolinyl, quinoxalinyl, tetrahydrofuryl, tetrahydroisoquinolinyl, tetrahydroquinolinyl, thiamorpholinyl, thiamorpholinyl sulfoxide, thiazolyl, thiazolinyl, 30 thienofuryl, thienothienyl, and thienyl.
As used herein, "heteroaryl" is intended to mean any stable - monocyclic or bicyclic carbon ring of up to 7 members in each ring, wherein at least one ring is aromatic and wherein from one to four carbon atoms are replaced by heteroatoms selected from the group consisting of N, O, and S. Examples of such heterocyclic elements include, but are not limited to, benzimidazolyl, benzisoxazolyl, benzofurazanyl, benzopyranyl, benzothiopyranyl, benzofuryl, benzothiazolyl, benzothienyl, benzoxazolyl, chromanyl, cinnolinyl, dihydrobenzofuryl, dihydrobenzothienyl, dihydrobenzothiopyranyl, dihydrobenzothiopyranyl sulfone, furyl, imi(~7.01yl, indolinyl, indolyl, isochromanyl, isoindolinyl, isoquinolinyl, isothiazolyl, naphthyridinyl, ox~ 7olyl, pyridyl, pyrazinyl, pyrazolyl, pyridazinyl, pyrimidinyl, pyrrolyl, ~uinazolinyl, quinolinyl, quinoxalinyl, tetrahydroisoquinolinyl, tetrahydroquinolinyl, thiazolyl, thienofuryl, thienothienyl, and thienyl.
As used herein, the terms "substituted aryl", "substituted heterocycle" and "substit~lte~ cycloaLkyl" are intended to include the cyclic group which is substituted with 1 or 2 substitutents selected from the group which includes but is not limited to F, Cl, Br, CF3, NH2, N(Cl-C6 allcyl)2, N02, CN, (Cl-C6 alkyl)O-, -OH, (Cl-C6 aLkyl)S(O)m-, (Cl-C6 alkyl)C(O)NH-, H2N-C(NH)-, (Cl-C6 aLkyl)C(O)-, (Cl-C6 alkyl)OC(O)-, N3,(Cl-C6 alkyl)OC(O)NH- and C1-C20 alkyl.
Lines drawn into the ring systems from substituents (such as from R3, R4 etc.) indicate that the bond may be attached to any of the substitutable ring carbon atoms.
The pharm~t~elltically acceptable salts of the compounds of this invention include the conventional non-toxic salts of the compounds of this invention as fo~ned, 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, glutarnic, benzoic, salicylic, sulfanilic, 2-acetoxy-benzoic, fumaric, toluenesulfonic, methanesulfonic, ethane disulfonic, oxalic, isethionic, trifluoroacetic and the like.

It is intended that the definition of any substituent or variable (e.g., R1a, n, etc.) at a particular location in a molecule be independent of its definitions elsewhere in that molecule. Thus, -N(R8)2 represents -NHH, -NHCH3, -NHc2H5t etc. It is understood 5 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.
Preferably, R1a, R1b and R2 are independently selected from: hydrogen, -N(R8)2, R8C(O)NR8- or Cl-C6 alkyl unsubstituted or substituted by -N(R8)2, R8O- or R8C(O)NR8-.
Preferably, R3 and R4 are independently selected from:
hydrogen, perfluoroalkyl, F, Cl, Br, R80-, R9s(o)m-~ CN, N02, 1~ R82N-C(NR8)-, R8C(O)-, R8OC(O)-, N3, -N(R8)~ or R9OC(o)NR8-and Cl-C6 alkyl.
Preferably, R5 is hydrogen or Cl-C6 aL~yl substituted with hydrogen, R9S(O)m-, CF3- or an unsubstituted or substituted aryl group.
Preferably, R6 is selected from: hydrogen, perfluoroalkyl, F, Cl, Br, R8O-, R9S(o)m-~ CN, NO2, R82N-c(NR8)-~ R8C(O)-, R8OC(O)-, N3, -N(R8)2, or R9OC(o)NR8- and C1-C6 alkyl.
Preferably, R7 is hydrogen or methyl. Most preferably, R7 is hydrogen.
Preferably, R8 is selected from H, C 1-C6 alkyl and benzyl.
Preferably, A1 and A2 are independently selected from: a bond, -C(O)NR8-, -NR8C(O)-, O, -N(R8)-, -S(0)2N(R8)- and-N(R8)S(0)2- .
Preferably, V is selected from hydrogen, heterocycle and aryl. More preferably, V is phenyl.
Preferably, Y is selected from phenyl, pyridyl, furanyl and thienyl. More preferably, Y is phenyl.
Preferably, n and r are independently 0, 1, or 2.
Preferably p is 1, 2 or 3.

Preferably t is 1.
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.
S Generally, the salts are l~le~ared either by ion exchange chromatography or by reacting the free base with stoichiometric amounts or with an excess of the desired salt-forming inorganic or organic acid in a suitable solvent or various combinations of solvents.
Reactions used to generate the compounds of this invention 10 are prepared by employing reactions as shown in Schemes 1-12, in addition to other standard manipulations such as ester hydrolysis, cleavage of protecting groups, etc., as may be known in the literature or exempli~led in the e~lJe~ ental procedures. Substituents R and R CH2-, as shown in the Schemes, represent the substituents R8, R9 and 15 others, depending on the compound of the instant invention that is being synthesized.
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 20 reactions described in the Schemes.

Synopsis of Schemes 1 - 12:
The requisite intermediates are in some cases commercially available, or can be prepared according to literature procedures, for the 25 most part. Scheme 1 illustrates the synthesis of one of the preferred embodiments of the inet~nt invention, wherein the variable W is present as a imidazolyl moiety that is substituted with a suitably substituted benzyl group. Substituted protected imidazole alkanols II can be prepared by methods known in the art. such as those described by F.
30 Schneider, Z. Physiol. Chem., 3:206-210 (1961) and C.P. Stewart, Biochem. Journal, 17:130-133(1923). Intermediate II can then be oxidized to the imidazole alkanoic acid by methods known in the art.
Some imidazole alkanoic acids containing alkyl substituents on the carbon atoms of the imidazole ring can be prepared by hydrolysis of the corresponding nitriles, which can be prepared by methods known in the art (see for example: ~. Med. Chem., 19:923 (1976)).
Benzylation and deprotection of the imidazole aL~anoate ester provides intermediate IV which can be hydrolyzed to the acid V
5 and coupled to a suitably substituted analine VI to provide the instant compound VII.
Schemes 2-5 illustrate syntheses of other suitably substituted alkanoate esters useful in the syntheses of the instant compounds wherein the variable W is present as a pyridyl moiety.
10 Similar synthetic strategies for preparing ester intermediates that incorporate other heterocyclic moieties for variable W are also well known in the art.
The suitably substituted aniline V can be reacted with a variety of other carboxylic acids, such as VIII, as illustrated in Scheme 15 6. The product IX can be deprotected to give the instant compound X.
The product X is isolated in the salt form, for example, as a trifluoroacetate, hydrochloride or acetate salt, among others. The product (li~ine X can further be selectively protected to obtain XI, which can subsequently be reductively aLkylated with a suitable 20 aldehyde, such as XII, to obtain XIII. Removal of the protecting group, and conversion to cyclized products such as the dihydroimidazole XIV
can be accomplished by literature procedures.
If the aniline V is coupled to an acid which also has a protected hydroxyl group, such as XV in Scheme 7, the protecting 25 groups can be subsequently removed to llnm~lc the hydroxyl group (Schemes 7 and 8). The primary alcohol XVI can be oxidized under standard conditions to e.g. an aldehyde, which can then be reacted with a variety of organometallic reagents such as Grignard reagents, to obtain secorldary alcohols such as XVIII. In addition, the fully 30 deprotected amino alcohol XIX can be reductively alkylated (under conditions described previously) with a variety of aldehydes to obtain secondary amines, such as XXX (Scheme 8), or tertiary amines.
The Boc protected amino alcohol XVIa can also be utilized to synthesi~ aziridines such as XXI (Scheme 9). Treating XVIa with 1,1'-sulfonyldiimidazole and sodium hydride in a solvent such as dimethylformamide leads to the formation of aziridine XXI. The aziridine can be reacted in the presence of a nucleophile, such as a thiol, in the presence of base to yield the ring-opened product XXII, which 5 can be deprotected to provide the instant compound.
In addition, the aniline V can be reacted with acids derived from amino acids such as O-alkylated tyrosines, according to standard procedures, to obtain compounds such as XXVI. When R' is an aryl group, XXVI can first be hydrogenated to llnm~k the phenol, and the 10 amine group deprotected with acid to produce XXVII. Alternatively, the amine protecting group in XXVI can be removed, and O-alkylated phenolic arnines such as XXVIII produced.
Schemes 11-12 illustrate the preparation of compounds of the instant invention wherein the orientation of the A3 amide moiety is 15 reversed relative to the compounds of Schemes 1-10. Thus the aLkanol II may be protected and converted to the corresponding amine XXX via the azide, as shown in Scheme 11. Alternatively, if the al)plo~,iately substituted protected amine, such as a protected histamine XXXI, is available, that reagent may be ring alkylated to provide the intermediate 20 amine XXXII.
As shown in Scheme 12, an amine such as intermediate XXX may be reacted with a suitably subsituted benzoic acid, such as compound XXXIII, to provide the instant compound XXXIV. The carbamate nitrogen of XXXIV may be subsequently alkylated as 25 described previously in Scheme 1.

WO 971278~i2 PCT/US971013~;4 (CR 2)p+1-~H TrCI Et3N Tr~N~/(CR 2)p+1-~H
N DMF ~N

oxidation Tr~N~/(CR 2)p-C02H (siMe3)cHN2 ~=N

Br 1. ~ N ~/ (CR 2)p-Tr (CR Z)p-co2cH3 F~ N HBr 2. MeOH IV

~ (C R 1 b2)p-CO2H

N~S
LiOH
THF. H20 /=~
R6J~y V

SCHEME 1 (continued) CRlb2)p-c02H EDC. Host N~ Et3N, DMF

R6~ H2N~

R3 Vi N~(CRlb2)p~ ~R4 CA 02243320 l998-07-l7 WO 97127852 PCTtUS97/01354 CH3 1) HNO2,Br2 ,~CO2CH3 ~ 2) KMnO4 H2N N~ 3) MeOH,H+ Br N

~/\ MgCI R 3,CO2CH3 Zncl2~Nicl2(ph3p)2 N

[~,CO2CH3 ~\ MgCI [~3 ,CO2CH3 Zn, CuCN

Br~CO2CH3 ~/\MgCI

N ZnC12. NiC12(Ph3P)2 ~,CO2CH3 N

WO 97t27852 PCTtUS97/01354 Br~1. LDA, CO2 Br~

N2. MeOH, H+ N

¢~
ZnCI2, Nicl2(ph3p)2 N

co2CH3 ~3~ 1. LDA, CO2 ~Br 2. (CH3)3SiCHN2 R6 ~\ Br R6 ~

~\ CO2CH3 Zn, NiC12(Ph3P)2 N~

Prot1 NH ~ Vlll H2N~ Prot2NH CH2CO2H
R3 R4 EDC, HOBt Et3N, DMF

~deprotection Prot1NH ~ ¦ 4 lX NHProt2 NH2 (~ ~ Prot3X

o R4 CH2CI2 ,~CHO
H ~R l~l Xll Prot3NH~ 14 NaBH(OAc)3 O R Et3N, CICH2CH2CI

- 2g -SCHEME 6 (continued) ~ deprotection BocNH

~ NH
~/ Xlll ~/
N~~ NC
~= NH AgCN

~/

/~0 R4 N~,N~
~ XIV

EDC, HOBt H2N~3 Et3N, DMF

\ 4 BnO

Prot1 NH CH2CO2H
V

XV

BnOy~ ~ R3 20% Pd(OH)2 H2 r NHProt1 CH3CO2H

~/
CICOCOCI
~o R4 DMSO CH2C12 NHProt1 (C2H5)3N
XVI

SCHEME 7 (continued~
H R3 1. R'MgX
~ ~ ~ (C2H5)20 ~ 2.deprotection HNHProt1 XVII

H~~o R-~
R' NH2 XVIII

WO 97127852 PCT/USg7/01354 SCH~ME 8 HO ~ ~ R3 deprotection NHProt~

XVI

~ R'CHO

NH2 ~ 4 CICH~CH2CI

XIX

HO
NH
R'CH2 XX

CA 02243320 l998-07-l7 R4 H /= N

N~R3 ~2 ~ NaH, DMF 0~C
NHBoc XVla ~ R'SH

<~0 R CH30H
Boc XXI

N

NHBoc XXII

H~ ~3 THF-H20 , ~3 2) CH2N2, EtOAc H2N C02H BocNH C02CH3 XXIII XXIV

R'CH20 R'CH2X ,~

Cs2CO3 BocNH CO2CH3 DMF
XXV

R'CH20 LiOH ~b~

BocNH C02H

SCHEME 10 (continued) ~ N~ R3 EDC, HOBt NHBoc ~
CICH2CH2CI \ HCI
XXVI \\ETOAc 1 ) 20% Pd(OH)2 / R4 CH30H, CH3CO2H / ~ R
2) HCI, EtOAc / ~ >~

R'CH20 R4\ ~ R3 N H2 HO ~ H

XXVII

WO 97n78s2 PCT/US97/01354 SCHE~aE 1 1 prot1~ ~(CR1b2)p-OH AC20 py Prot~ (CR 2)p-OAc ~-Br N~/(CR1b2)p-OAc EtOAc ~=< THF, H20 2. N-~Je,l~r~tect R6~

) -OH (CR1b ) -N
~N 1. MsCI, NEt3 ~N
~=~ 2. BU4N-N3 R6~ XXIX R6 (CR1 b2) -N H
H2, Pd/C N~
R

XXX

SCHEME 1 1 (continued) rBr Prot~ ~_ R6~ Prot~ NHProt2 \~ N~ NHProt2 N~+
H EtOAc ~=<
R6~ Br~
XXXI

N~_ deprotection ~N NH2 6~
R

XXXII

CA 02243320 l998-07-l7 W O 97/27852 PCTrUS97/01354 (CRlb2)p-NH

Et3N
R6/~
XXX

(CR1b ) -N~ R5-X
N NaH
n-Bu4NI, DMF

O~R4 (CR1b2) -N
\~N R~
,~
'~--R6 XXXV

The instant compounds are useful as pharmaceutical agents for m~rnm~l~, especially for hllm~n~. These compounds may be ~lmini.~tered to patients for use in the treatment of cancer. Examples of the type of cancer which may be treated with the compounds of this 5 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 forrnation (i.e., neurofibromen (NF-l), neu, scr, 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 angiogenisis, thereby affecting th¢ growth of tumors (J. Rak et al. Cancer Research, 55:4575-4580 (1995)). Such anti-angiogenisis properties of the instant compounds may also be useful in the treatment of certain forms of blindness 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 ~llmini~tration of an effective amount of the compounds of the invention to a m~mm~l in need of such treatment. For example, a component of NF-l 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 transhlmin~l 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 compounds of this invention may be ~(lmini~tered to m~mm~l~, preferably hllm~n~, either alone or, preferably, in 5 combination with pharmaceutically acceptable carriers or diluents, optionally with known adjuvants, such as alum, in a pharmaceutical composition, according to standard pharmaceutical practice. The compounds can be ~1mini.~tered orally or parenterally, including the intravenous, intrarnuscular, intraperitoneal, subcutaneous, rectal and 10 topical routes of ~rlmini~tration.
For oral use of a chemotherapeutic compound according to this invention, the selected compound may be ~-lmini.~tered, for example, in the form of tablets or capsules, or as an aqueous solution or suspension. In the case of tablets for oral use, carriers which are 15 commonly used include lactose and corn starch, and lubricating agents, such as magnesium stearate, are commonly added. For oral ~lministration in capsule form, useful diluents include lactose and dried corn starch. When aqueous suspensions are required for oral use, the active ingredient is combined with emulsifying and suspending agents.
20 If desired, certain sweetening and/or flavoring agents may be added.
For intramuscular, intraperitoneal, subcutaneous and intravenous use, sterile solutions of the active ingredient are usually prepared, and the pH of the solutions should be suitably adjusted and buffered. For intravenous use, the total concentration of solutes should be controlled 25 in order to render the preparation isotonic.
The present invention also encompasses a pharmaceutical composition useful in the treatment of cancer, comprising the ~rlministration of a therapeutically effective amount of the compounds of this invention, with or without pharmaceutically acceptable carriers 30 or diluents. Suitable compositions of this invention include aqueous solutions comprising compounds of this invention and pharmacolo-gically acceptable carriers, e.g., saline, at a pH level, e.g., 7.4. The solutions may be introduced into a patient's intramuscular blood-stream by local bolus injection.

When a compound according to this invention is ~lmini~tered into a human subject, the daily dosage will norrnally be determined by the prescribing physician with the dosa~e generally varying according to the age, weight, and response of the individual S patient, as well as the severity of the patient's symptoms.
In one exemplary application, a suitable amount of compound is ~lmini.ctered to a m~mmAl undergoing treatment for cancer. ~ nini~tration occurs in an amount between about 0.1 mg/kg of body weight to about 60 mg/l~g of body weight per day, preferably of between O.S mg/kg of body weight to about 40 mg/kg of body weight per day.
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 ~PTase to farnesylate the substrate, the chemical content of the assay mixtures may be determined by well known immunological, 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 cont~ining 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 cont~ining an unknown arnount of farnesyl-protein transferase, an excess amount of a known substrate of FPTase (for example a tetrapeptide having a cysteine at the arnine 5 terminus) and farnesyl pyrophosphate are incubated for an a~l,ro~fiate 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 subst~nti~lly smaller than the concentration of enzyme in the assay vessel) required to inhibit the 10 enzymatic activity of the sample by 50% is approximately e~ual to half of the concentration of the enzyme in that particular sample.

EXAMPLES

Examples provided are intended to assist in a further underst~n-ling 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.

N-(3-chlorophenyl)-3-[1 -(4-cyanobenzyl)-5-imidazolyl]propionamide hydrochloride (1 ) 25 Step 1: Preparation of methyl 3-(4-imidazolyl)-2-propenoate (2) Through a suspension of urocanic acid (5.0 g) in methanol at 0 ~C was bubbled HCl gas until the solution was saturated. The reaction was stirred for one hour, then concentrated in vacuo to dryness to provide the crude product 2 which was used in the next step without 30 further purification.

Step 2: Preparation of methyl 3-(4-imidazolyl)propionate (3) To a solution of ester 2 (5.3 g) in methanol at room temperature was added 10% p~ flium on carbon (20 mg) under a blanket of argon.. A balloon of hydrogen was applied, and the reaction was stirred for one hour. The solution was ~lltered, and then concentrated in vacuo to dryness to provide the crude product 3 which was sufficiently pure for use in the next step.

Step 3: Preparation of methyl 3-(1 -triphenylmethyl-4-imicl~7olyl)-propionate (4) To a solution of ester 3 (1.77 g) in 10 mL of dry DMF at room temperature was added triethylamine (3.25 mL). A white solid precipitated from the solution. Chlorotriphenylmethane (2.65 g) in 12 mL of DMP was added dropwise. The reaction mixture was stirred for 20 hours, then poured into 40% EtOAc/~exane solution and washed with two portions of water. The organic layer was extracted with sat.
aq. NaHCO3 solution and brine, then dried (Na2SO4), filtered, and lS concentrated in vacuo to provide the crude product 4 which was sufficiently pure for use in the next step.

Step 4: Preparation of methyl 3-[1-(4-cyanobenzyl)-5-imidazolyllpropionate (5) A solution of 4 (4.3 g) and a-bromo-p-tolunitrile (2.03 g) in 30 mL of EtOAc was stirred at 65 ~C for 20 hours, during which a pale yellow precipitate formed. The reaction was cooled to room temperature, concentrated in v~cuo, and dissolved in 30 mL of methanol. The resulting solution was warrned to 60 ~C for one hour, then reconcentrated in vacuo. The solid material was triturated to remove soluble materials, then taken up in CH2Cl2/50% aq. NaHCO3 soln. The aqueous layer was extracted three times with CH2Cl2 and concentrated in vacuo to provide the crude product. Purification by silica gel chromatography (5% MeOH/CH2Cl2) gave the titled compound as a white waxy solid.

Step S: Preparation of 3-[1-(4-cyanobenzyl)-5-imidazolyl]-propionic acid hydrochloride (6) To a solution of the methyl ester 5 (1.10 g) in 10 mL of THF and S mL of water was added lithium hydroxide monohydrate (172 mg). After two hours, the reaction was cooled to 0 ~C, and a 1 M
solution of HCl/ether (8.2 mL) was added. The solution was 5 concentrated and dried in vacuo next to P2Os to provide the product 6 as a white solid.

Step 6: Preparation of N-(3-chlorophenyl)-3-[1-(4-cyanobenzyl)-5-imidazolyllpropion~ le hydrochloride (1) To a 0 ~C solution of acid 6 (141 mg) in 2 mL of dry DMP
was added triethyl~mine ~0.089 mL), and 3-chloro~rliline (0.046 mL).
1-Hydroxybenzotriazole hydrate (80 mg) was added, followed by 1-ethyl-3-(3-dimethylaminopropyl)carbodiimide hydrochloride (106 mg).
After six hours, the reaction was poured into EtOAc, washed with sat.
aq. NaHCO3 solution and brine, then dried (Na2SO4), filtered, and concentrated in vacuo to provide the crude product. Puri~lcation by silica gel chromatography (2-6% MeOH/CH2Cl2) gave a white foam which was taken up in CH2Cl2, treated with 1 M HCVether solution, and concentrated in vacuo. The product hydrochloride 1 was isolated as a white powder.

FAB mass spectrum m/e 365 (M+1).
Analysis calculated for C20Hl7ClN4O ~ 1.00 HCl ~ 0.50 H2O:
C, 58.55; H, 4.67; N, 13.65;
Found: C, 58.61; H, 4.53; N, 13.47.

In vitro inhibition of ras farnesyl transferase Assays offarnesyl-protein transferase. Partially purified bovine FPTase and Ras peptides (Ras-CVLS, Ras-CVIM and Ras-CAIL) were prepared as described by Schaber et al., J. Biol. Chem. 265: 14701-14704 (1990), Pompliano, et al., Biochemistry 31:3800 (1992) and Gibbs et al., PNAS U.S.A. 86:6630-6634 (1989), respectively. Bovine FPTase was assayed in a volume of 100 ,ul cont~inin~ 100 mM N-(2-hydroxy ethyl) piperazine-N'-(2-ethane sulfonic acid) (HEPES), pH 7.4, S mM MgC12, 5 mM dithiothreitol (DTT), 100 mM [3H]-farnesyl diphosphate ([3H]-FPP; 740 CBq/mmol, New England Nuclear), 650 S nM Ras-CVLS and 10 ~g/ml FPTase at 31 ~C for 60 min. Reactions were initiated with FPTase and stopped with 1 ml of 1.0 M HCL in ethanol. Precipitates were collected onto filter-mats using a TomTec Mach II cell harvestor, washed with 100~o ethanol, dried and counted in an LKB ,B-plate counter. The assay was linear with respect to both 10 substrates, PPTase levels and time; less than 10% of the [3H]-FPP was utilized during the reaction period. Purified compounds were dissolved in 100% dimethyl sulfoxide (DMSO) and were diluted 20-fold into the assay. Percentage inhibition is measured by the amount of incorporation of radioactivity in the presence of the test compound 15 when compared to the amount of incorporation in the absence of the test compound.
Human FPTase was prepared as described by Omer et al., Biochemistry 32:5167-5176 (1993). Human FPTase activity was assayed as described above with the exception that 0.1% (w/v) 20 polyethylene glycol 20,000, 10 ,UM ZnCl2 and 100 nM Ras-CVIM were added to the reaction mixture. Reactions were performed for 30 min., stopped with 100 ~ul of 30% (v/v) trichloroacetic acid (TCA) in ethanol and processed as described above for the bovine enzyme.
The compound of the instant invention described in 25 Example 1 was tested for inhibitory activity against human FPTase by the assay described above and was found to have IC50 of < 10 ~M.

30 In vivo ras farnesylation assay The cell line used in this assay is a v-ras line derived from either Ratl or NIH3T3 cells, which expressed viral Ha-ras p21. The assay is performed essentially as described in DeClue, J.E. et ah, 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. l~o). After 4 hours at 37~C, the cells are labelled in 3 ml methionine-free DMEM supple-meted with 10% regular DMEM, 2% fetal bovine serum and 400 S mCi[35S]methionine (lO00 Ci/mmol). After an additional 20 hours, the cells are lysed in 1 ml lysis buffer (1% NP40120 mM HEPES, pH 7.5/5 mM MgC12/lmM DTT/lO mg/ml aprotinen/2 mg/ml leupeptin/2 mg/ml antipain/0.5 mM PMS~) and the lysates cleared by centrifugation at 100,000 x g for 45 min. Aliquots of lysates cont~ininE e~ual numbers 10 of acid-precipitable counts are bought to l ml with IP buffer (lysis buffer lacking DTT) and immunoprecipitated with the ras-specific monoclonal antibody Yl3-259 (Furth, M.E. et al., J. Virol. 43:294-304, (1982)). Following a 2 hour antibody incubation at 4~C, 200 ml of a 25% suspension of protein A-Sepharose coated with rabbit anti rat IgG
15 is added for 45 min. The immunoprecipitates are washed four times with IP buffer (20 nM HEPES, pH 7.5/1 mM EDTAt1% Triton X-100Ø5% deoxycholate/0.1%/SDS/0.1 M NaCl) boiled in SDS-PAGE
sample buffer and loaded on 13% acryl~mi~1e gels. When the dye front reached the bottom, the gel is fixed, soaked in Fnli~htening, dried and 20 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.

In vivo growth inhibition assav To detennine the biological conse~uences of FPTase inhibition, the effect of the compounds of the instant invention on the anchorage-independent growth of Ratl cells transformed with either a 30 v-ras, v-raf, or v-mos oncogene is tested. Cells transformed by v-Raf and v-Mos maybe included in the analysis to evaluate the specificity of instant compounds for Ras-induced cell transformation.
Rat 1 cells transformed with either v-ras, v-raf, or v-mos are seeded at a density of 1 x 104 cells per plate (35 mm in diameter) in CA 02243320 l998-07-l7 a 0.3% top agarose layer in medium A (Dulbecco's modified Eagle's medium supplemented with 10% fetal bovine serum) over a bottom agarose layer (0.6%). Both layers contain 0.1% methanol or an appropriate concentration of the instant compound (dissolved in 5 methanol at 1000 times the final concentration used in the assay). The cells are fed twice weekly with 0.5 ml of medium A cont~ining 0.1%
methanol or the concentration of the instant compound.
Photomicrographs are taken 16 days after the cultures are seeded and comparisons are made.

Claims (23)

WHAT IS CLAIMED IS:
1. A compound which inhibits farnesyl-protein transferase of the formula I:

wherein:
R1a, R1b and R2 are independently selected from:
a) hydrogen, b) aryl, heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, R8O-, R9S(O)m-, R8C(O)NR8-,CN, NO2, (R8)2N-C(NR8)-, R8C(O)-, R8OC(O)-, N3, -N(R8)2, or R9OC(O)NR8-, c) C1-C6 alkyl unsubstituted or substituted by aryl, heterocyclic, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, R8O-, R9S(O)m-, R8C(O)NR8-, CN, (R8)2N-C(NR8)-, R8C(O)-, R8OC(O)-, N3, -N(R8)2, or R9OC(O)-NR8-;
R3 and R4 are independently selected from F, C1, Br, N(R8)2, CF3, NO2, (R8)O-, (R9)S(O)m-, (R8)C(O)NH-, H2N-C(NH)-, (R8)C(O)-, (R8)OC(O)-, N3, CN, CF3(CH2)nO-, (R9)OC(O)NR8-, C1-C20 alkyl, substituted or unsubstituted aryl and substituted or unsubstituted heterocycle;
R5 is selected from:
a) hydrogen, b) unsubstituted or substituted aryl, c) unsubstituted or substituted heterocyclic, d) unsubstituted or substituted C3-C10 cycloalkyl, and e) C1-C6 alkyl substituted with hydrogen or a group selected from unsubstituted or substituted aryl, unsubstituted or substituted heterocyclic, unsubstituted or substituted C3-C10 cycloalkyl, N(R8)2, CF3, NO2, (R8)O-, (R9)S(O)m-, (R8)C(O)NH-, H2N-C(NH)-, (R8)C(O)-, (R8)OC(O)-, N3, CN (R9)OC(O)NR8-;

R6 is independently selected from:
a) hydrogen, b) aryl, heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, perfluoroalkyl, F, Cl, Br, R8O-, R9S(O)m-, R8C(O)NR8-, CN, NO2, R82N-C(NR8)-, R8C(O)-, R8OC(O)-, N3, -N(R8)2, or R9OC(O)NR8-, and c) C1-C6 alkyl unsubstituted or substituted by aryl, heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, perfluoroalkyl, F, Cl, Br, R8O-, R9S(O)m-, R8C(O)NH-, CN, H2N-C(NH)-, R8C(O)-, R8OC(O)-, N3, -N(R8)2, or R8OC(O)NH-;

R7 is selected from:
a) hydrogen, b) C2-C6 alkenyl, C2-C6 alkynyl, perfluoroalkyl, F, Cl, Br, R8O-, R9S(O)m-, R8C(O)NR8-, CN, NO2, (R8)2N-C-(NR8)-, R8C(O)-, R8OC(O)-, N3, -N(R8)2, or R9OC(O)NR8-, and c) C1-C6 alkyl unsubstituted or substituted by perfluoroalkl, F, Cl, Br, R8O-, R9S(O)m-, R8C(O)NR8-, CN, (R8)2N-C(NR8)-, R8C(O)-, R8OC(O)-, N3, -N(R8)2, or R9OC(O)NR8-;
R8 is independently selected from hydrogen, C1-C6 alkyl, benzyl and aryl;

R9 is independently selected from C1-C6 alkyl and aryl;

A1 and A2 are independently selected from: a bond, -CH=CH-, -C=C-, -C(O)-, -C(O)NR8-, -NR8C(O)-, O, -N(R8)-, -S(O)2N(R8)-, -N(R8)S(O)2-, or S(O)m;
A3 is selected from: -NR5C(O)- or -C(O)NH-;

V is selected from:
a) hydrogen, b) heterocycle, c) aryl, d) C1-C20 alkyl wherein from 0 to 4 carbon atoms are replaced with a a heteroatom selected from O, S, and N, and e) C2-C20 alkenyl, provided that V is not hydrogen if A1 is S(O)m and V is not hydrogen if A1 is a bond, n is 0 and A2 is S(O)m;
W is a heterocycle;
Y is aryl or heteroaryl;

m is 0, 1 or2;
n is 0, 1, 2, 3 or 4;
p is 0, 1, 2, 3 or 4;
r is 0 to 5, provided that r is 0 when V is hydrogen; and t is 0 or 1;
or an optical isomer or pharmaceutically acceptable salt thereof.
2. A compound which inhibits farnesyl-protein transferase of the formula Ia:

wherein:

R1a and R1b are independently selected from: hydrogen or C1-C6 alkyl;

R2 is independently selected from:
a) hydrogen, b) aryl, heterocycle, cycloalkyl, R8O-, -N(R8)2 or C2-C6 alkenyl, c) C1-C6 alkyl unsubstituted or substituted by aryl, heterocycle, cycloalkyl, alkenyl, R8O-, or-N(R8)2;
R3 and R4 are independently selected from F, Cl, Br, N(R8)2, CF3, NO2, (R8)O-, (R9)S(O)m-, (R8)C(O)NH-, H2N-C(NH)-, (R8)C(O)-, (R8)OC(O)-, N3, CN, (R9)OC(O)NR8-, C1-C20 alkyl, substituted or unsubstituted aryl and substituted or unsubstituted heterocycle;
R5 is selected from:
a) hydrogen, and b) C1-C6 alkyl substituted with hydrogen or a group selected from unsubstituted or substituted aryl, unsubstituted or substituted heterocyclic, unsubstituted or substituted C3-C10 cycloalkyl, N(R8)2, CF3, NO2, (R8)O-, (R9)S(O)m-, (R8)C(O)NH-, H2N-C(NH)-, (R8)C(O)-, (R8)OC(O)-, N3, CN (R9)OC(O)NR8-;

R6 is independently selected from:
a) hydrogen, b) C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 perfluoroalkyl, F, Cl, R8O-, R8C(O)NR8-, CN, NO2, (R8)2N-C(NR8)-, R8C(O)-, R8OC(O)-, -N(R8)2, or R9OC(O)NR8-, and c) C1-C6 alkyl substituted by C1-C6 perfluoroalkyl, R8O-, R8C(O)NR8-, (R8)2N-C(NR8)-, R8C(O)-, R8OC(O)-, -N(R8)2, or R9OC(o)NR8-;

R7a is hydrogen or methyl;
R8 is independently selected from hydrogen, C1-C6 alkyl, benzyl and aryl;
R9 is independently selected from C1-C6 alkyl and aryl;

A1 and A2 are independently selected from: a bond, -CH=CH-, -C~C-, -C(O)-, -C(O)NR8-, O, -N(R8)-, or S(O)m;
A3 is selected from: -NRSC(O)- or -C(O)NH-;

V is selected from:
a) hydrogen, b) heterocycle selected from pyrrolidinyl, imidazolyl, pyridinyl, thiazolyl, pyridonyl, 2-oxopiperidinyl, indolyl, quinolinyl, isoquinolinyl, and thienyl, c) aryl, d) C1-C20 alkyl wherein from 0 to 4 carbon atoms are replaced with a a heteroatom selected from O, S, and N, and e) C2-C20 alkenyl, and provided that V is not hydrogen if Al is S(O)m and V is not hydrogen if A 1 is a bond, n is 0 and A2 is S(O)m;

m is 0, 1 or 2;
n is 0, 1, 2, 3 or 4;
p is 0, 1, 2, 3 or 4; and r is 0 to 5, provided that r is 0 when V is hydrogen;

or an optical isomer or pharmaceutically acceptable salt thereof.
3. A compound which inhibits farnesyl-protein transferase of the formula Ib:

wherein:

R1a and R1b are independently selected from: hydrogen or C1-C6 alkyl;

R2 is independently selected from:
a) hydrogen, b) aryl, heterocycle, cycloalkyl, R8O-, -N(R8)2 or C2-C6 alkenyl, c) C1-C6 alkyl unsubstituted or substituted by aryl, heterocycle, cycloalkyl, alkenyl, R8O-, or-N(R8)2;

R3 and R4 are independently selected from F, Cl, Br, N(R8)2, CF3, NO2, (R8)O-, (R9)S(O)m-, (R8)C(O)NH-, H2N-C(NH)-, (R8)C(O)-, (R8)OC(O)-, N3, CN, (R9)OC(O)NR8-, C1-C20 alkyl, substituted or unsubstituted aryl and substituted or unsubstituted heterocycle;
R5 is selected from:
a) hydrogen, and b) C1-C6 alkyl substituted with hydrogen or a group selected from unsubstituted or substituted aryl, unsubstituted or substituted heterocyclic, unsubstituted or substituted C3-C10 cycloalkyl, N(R8)2, CF3, NO2, (R8)O-, (R9)S(O)m-, (R8)C(O)NH-, H2N-C(NH)-, (R8)C(O)-, (R8)OC(O)-, N3, CN (R9)OC(O)NR8-;

R6 is independently selected from:
a) hydrogen, b) C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 perfluoroalkyl, F, Cl, R8O-, R8C(O)NR8-, CN, NO2, (R8)2N-C(NR8)-, R8C(O)-, R8OC(O)-, -N(R8)2, or R9OC(O)NR8-, and c) C1-C6 alkyl substituted by C1-C6 perfluoroalkyl, R8O-, R8C(O)NR8-, (R8)2N-C(NR8)-, R8C(O)-, R8OC(O)-, -N(R8)2, or R9OC(O)NR8-;

R7 is selected from: hydrogen and C1-C6 alkyl;
R8 is independently selected from hydrogen, C1-C6 alkyl, benzyl and aryl;
R9 is independently selected from C1-C6 alkyl and aryl;
A1 and A2 are independently selected from: a bond, -CH=CH-, -C~C-, -C(O)-, -C(O)NR8-, O, -N(R8)-, or S(O)m;

A3 is selected from: -NR5C(O)- or -C(O)NH-;

V is selected from:
a) hydrogen, b) heterocycle selected from pyrrolidinyl, imidazolyl, pyridinyl, thiazolyl, pyridonyl, 2-oxopiperidinyl, indolyl, quinolinyl, isoquinolinyl, and thienyl, c) aryl, d) C1-C20 alkyl wherein from 0 to 4 carbon atoms are replaced with a a heteroatom selected from O, S, and N, and e) C2-C20 alkenyl, and provided that V is not hydrogen if A1 is S(O)m and V is not hydrogen if A1 is a bond, n is O and A2 is S(O)m;

W is a heterocycle selected from pyrrolidinyl, pyridinyl, thiazolyl, pyridonyl, 2-oxopiperidinyl, indolyl, quinolinyl, or isoquinolinyl;

m is 0, 1 or 2;
n is 0, 1, 2, 3 or 4;
p is 0, 1, 2, 3 or 4;
r is 0 to 5, provided that r is 0 when V is hydrogen; and t is 1;
or an optical isomer or pharmaceutically acceptable salt thereof.
4. The compound according to Claim 1 of the formula Ic:

wherein:

R1b is independently selected from:
a) hydrogen, b) aryl, heterocycle, cycloalkyl, R8O-, -N(R8)2 or C2-C6 alkenyl, c) C1-C6 alkyl unsubstituted or substituted by aryl, heterocycle, cycloalkyl, alkenyl, R8O-, or-N(R8)2;

R2 are independently selected from: hydrogen or C1-C6 alkyl;
R3 and R4 are independently selected from F, Cl, Br, N(R8)2, CF3, NO2, (R8)O-, (R9)S(O)m-, (R8)C(O)NH-, H2N-C(NH)-, (R8)C(O)-, (R8)OC(O)-, N3, CN, (R9)OC(O)NR8-, C1-C20 alkyl, substituted or unsubstituted aryl and substituted or unsubstituted heterocycle;
R5 is selected from:
a) hydrogen, and b) C1-C6 alkyl substituted with hydrogen or a group selected from unsubstituted or substituted aryl, unsubstituted or substituted heterocyclic, unsubstituted or substituted C3-C10 cycloalkyl, N(R8)2, CF3, NO2, (R8)O-, (R9)S(O)m-, (R8)C(O)NH-, H2N-C(NH)-, (R8)C(O)-, (R8)OC(O)-, N3, CN (R9)OC(O)NR8-;

R6 is independently selected from:
a) hydrogen, b) C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 perfluoroalkyl, F, Cl, R8O-, R8C(O)NR8-, CN, NO2, (R8)2N-C(NR8)-, R8C(O)-, R8OC(O)-, -N(R8)2, or R9OC(O)NR8-, and c) C1-C6 alkyl substituted by C1-C6 perfluoroalkyl, R8O-, R8C(O)NR8-, (R8)2N-C(NR8)-, R8C(O)-, R8OC(O)-, -N(R8)2, or R9OC(O)NR8-;
R8 is independently selected from hydrogen, C1-C6 alkyl, benzyl and aryl;
R9 is independently selected from C1-C6 alkyl and aryl;
A3 is selected from: -NR5C(O)- or -C(O)NH-;

m is 0,1 or 2;and p is 0, 1, 2, 3 or 4;
or an optical isomer or pharmaceutically acceptable salt thereof.
5. The compound according to Claim 1 of the formula Id:

wherein:

R1b is independently selected from:
a) hydrogen, b) aryl, heterocycle, cycloalkyl, R8O-, -N(R8)2 or C2-C6 alkenyl, c) C1-C6 alkyl unsubstituted or substituted by aryl, heterocycle, cycloalkyl, alkenyl, R8O-, or-N(R8)2;

R2 are independently selected from: hydrogen or C1-C6 alkyl;
R3 and R4 are independently selected from F, Cl, Br, N(R8)2, CF3, NO2, (R8)O-, (R9)S(O)m-, (R8)C(O)NH-, H2N-C(NH)-, (R8)C(O)-, (R8)OC(O)-, N3, CN, (R9)OC(O)NR8-, C1-C20 alkyl, substituted or unsubstituted aryl and substituted or unsubstituted heterocycle;
R5 is selected from:
a) hydrogen, and b) C1-C6 alkyl substituted with hydrogen or a group selected from unsubstituted or substituted aryl, unsubstituted or substituted heterocyclic, unsubstituted or substituted C3-C10 cycloalkyl, N(R8)2, CF3, NO2, (R8)O-, (R9)S(O)m-m, (R8)C(O)NH-, H2N-C(NH)-, (R8)C(O)-, (R8)OC(O)-, N3, CN (R9)OC(O)NR8-;
R8 is independently selected from hydrogen, C1-C6 alkyl, benzyl and aryl;
R9 is independently selected from C1-C6 alkyl and aryl;
A3 is selected from: -NR5C(O)- or -C(O)NH-;

m is 0, 1 or 2; and p is 0, 1, 2, 3 or 4;

or an optical isomer or pharmaceutically acceptable salt thereof.
6. A compound which inhibits farnesyl-protein transferase which is:

N-(3-chlorophenyl)-3-[1-(4-cyanobenzyl)-5-imidazolyl]propionamide hydrochloride (1) or a pharmaceutically acceptable salt thereof.
7. A pharmaceutical composition comprising a pharmaceutical carrier, and dispersed therein, a therapeutically effective amount of a compound of Claim 1.
8. A pharmaceutical composition comprising a pharmaceutical carrier, and dispersed therein, a therapeutically effective amount of a compound of Claim 2.
9. A pharmaceutical composition comprising a pharmaceutical carrier, and dispersed therein, a therapeutically effective amount of a compound of Claim 3.
10. A pharmaceutical composition comprising a pharmaceutical carrier, and dispersed therein, a therapeutically effective amount of a compound of Claim 6.
11. A method for inhibiting farnesyl-protein transferase which comprises administering to a mammal in need thereof a therapeutically effective amount of a composition of Claim 7.
12. A method for inhibiting farnesyl-protein transferase which comprises administering to a mammal in need thereof a therapeutically effective amount of a composition of Claim 8.
13. A method for inhibiting farnesyl-protein transferase which comprises administering to a mammal in need thereof a therapeutically effective amount of a composition of Claim 9.
14. A method for inhibiting farnesyl-protein transferase which comprises administering to a mammal in need thereof a therapeutically effective amount of a composition of Claim 10.
15. A method for treating cancer which comprises administering to a mammal in need thereof a therapeutically effective amount of a composition of Claim 7.
16. A method for treating cancer which comprises administering to a mammal in need thereof a therapeutically effective amount of a composition of Claim 8.
17. A method for treating cancer which comprises administering to a mammal in need thereof a therapeutically effective amount of a composition of Claim 9.
18. A method for treating cancer which comprises administering to a mammal in need thereof a therapeutically effective amount of a composition of Claim 10.
19. 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 7.
20. 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 7.
21. 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 7.
22. A method for preventing restenosis which comprises administering to a mammal in need thereof a therapeutically effective amount of a composition of Claim 7.
23. 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 7.
CA 2243320 1996-01-30 1997-01-27 Inhibitors of farnesyl-protein transferase Abandoned CA2243320A1 (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US1079896P 1996-01-30 1996-01-30
US60/010,798 1996-01-30
GBGB9605708.8A GB9605708D0 (en) 1996-03-19 1996-03-19 Inhibitors of farnesyl-protein transferase
GB9605708.8 1996-03-19
PCT/US1997/001354 WO1997027852A1 (en) 1996-01-30 1997-01-27 Inhibitors of farnesyl-protein transferase

Publications (1)

Publication Number Publication Date
CA2243320A1 true CA2243320A1 (en) 1997-08-07

Family

ID=29424171

Family Applications (1)

Application Number Title Priority Date Filing Date
CA 2243320 Abandoned CA2243320A1 (en) 1996-01-30 1997-01-27 Inhibitors of farnesyl-protein transferase

Country Status (1)

Country Link
CA (1) CA2243320A1 (en)

Similar Documents

Publication Publication Date Title
AU704087B2 (en) Inhibitors of farnesyl-protein transferase
US6066738A (en) Inhibitors of farnesyl-protein transferase
US5968965A (en) Inhibitors of farnesyl-protein transferase
AU716338B2 (en) Inhibitors of farnesyl-protein transferase
US5869682A (en) Inhibitors of farnesyl-protein transferase
US5925651A (en) Inhibitors of farnesyl-protein transferase
AU712504B2 (en) Inhibitors of farnesyl-protein transferase
US5780492A (en) Inhibitors of farnesyl-protein transferase
CA2250353A1 (en) Inhibitors of farnesyl-protein transferase
AU715667B2 (en) Inhibitors of farnesyl-protein transferase
US5624936A (en) Inhibitors of farnesyl-protein transferase
CA2249559A1 (en) Inhibitors of farnesyl-protein transferase
AU717298B2 (en) Inhibitors of farnesyl-protein transferase
AU707416B2 (en) Inhibitors of farnesyl-protein transferase
WO1996034010A2 (en) Inhibitors of farnesyl-protein transferase
AU716153B2 (en) Inhibitors of farnesyl-protein transferase
US5627202A (en) Inhibitors of farnesyl-protein transferase
AU703988B2 (en) Inhibitors of farnesyl-protein transferase
EP0900081A1 (en) Inhibitors of farnesyl-protein transferase
CA2243320A1 (en) Inhibitors of farnesyl-protein transferase
CA2243272A1 (en) Inhibitors of farnesyl-protein transferase
WO1996031525A2 (en) Inhibitors of farnesyl-protein transferase
CA2243849A1 (en) Inhibitors of farnesyl-protein transferase
CA2216654A1 (en) Inhibitors of farnesyl-protein transferase
CA2216532A1 (en) Inhibitors of farnesyl-protein transferase

Legal Events

Date Code Title Description
FZDE Dead