CA2207252A1 - Inhibitors of prenyl transferases - Google Patents

Inhibitors of prenyl transferases

Info

Publication number
CA2207252A1
CA2207252A1 CA002207252A CA2207252A CA2207252A1 CA 2207252 A1 CA2207252 A1 CA 2207252A1 CA 002207252 A CA002207252 A CA 002207252A CA 2207252 A CA2207252 A CA 2207252A CA 2207252 A1 CA2207252 A1 CA 2207252A1
Authority
CA
Canada
Prior art keywords
acid
group
amino
compound
ras
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.)
Granted
Application number
CA002207252A
Other languages
French (fr)
Other versions
CA2207252C (en
Inventor
Said Sebti
Andrew Hamilton
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.)
University of Pittsburgh
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 US08/371,682 external-priority patent/US5705686A/en
Priority claimed from US08/451,839 external-priority patent/US5834434A/en
Priority claimed from US08/582,076 external-priority patent/US6011175A/en
Application filed by Individual filed Critical Individual
Publication of CA2207252A1 publication Critical patent/CA2207252A1/en
Application granted granted Critical
Publication of CA2207252C publication Critical patent/CA2207252C/en
Anticipated expiration legal-status Critical
Expired - Lifetime legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C323/00Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups
    • C07C323/23Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups containing thio groups and nitrogen atoms, not being part of nitro or nitroso groups, bound to the same carbon skeleton
    • C07C323/24Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups containing thio groups and nitrogen atoms, not being part of nitro or nitroso groups, bound to the same carbon skeleton having the sulfur atoms of the thio groups bound to acyclic carbon atoms of the carbon skeleton
    • C07C323/25Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups containing thio groups and nitrogen atoms, not being part of nitro or nitroso groups, bound to the same carbon skeleton having the sulfur atoms of the thio groups bound to acyclic carbon atoms of the carbon skeleton the carbon skeleton being acyclic and saturated
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C323/00Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups
    • C07C323/50Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups containing thio groups and carboxyl groups bound to the same carbon skeleton
    • C07C323/51Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups containing thio groups and carboxyl groups bound to the same carbon skeleton having the sulfur atoms of the thio groups bound to acyclic carbon atoms of the carbon skeleton
    • C07C323/57Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups containing thio groups and carboxyl groups bound to the same carbon skeleton having the sulfur atoms of the thio groups bound to acyclic carbon atoms of the carbon skeleton the carbon skeleton being further substituted by nitrogen atoms, not being part of nitro or nitroso groups
    • C07C323/58Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups containing thio groups and carboxyl groups bound to the same carbon skeleton having the sulfur atoms of the thio groups bound to acyclic carbon atoms of the carbon skeleton the carbon skeleton being further substituted by nitrogen atoms, not being part of nitro or nitroso groups with amino groups bound to the carbon skeleton
    • C07C323/59Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups containing thio groups and carboxyl groups bound to the same carbon skeleton having the sulfur atoms of the thio groups bound to acyclic carbon atoms of the carbon skeleton the carbon skeleton being further substituted by nitrogen atoms, not being part of nitro or nitroso groups with amino groups bound to the carbon skeleton with acylated amino groups bound to the carbon skeleton
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C323/00Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups
    • C07C323/50Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups containing thio groups and carboxyl groups bound to the same carbon skeleton
    • C07C323/51Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups containing thio groups and carboxyl groups bound to the same carbon skeleton having the sulfur atoms of the thio groups bound to acyclic carbon atoms of the carbon skeleton
    • C07C323/60Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups containing thio groups and carboxyl groups bound to the same carbon skeleton having the sulfur atoms of the thio groups bound to acyclic carbon atoms of the carbon skeleton with the carbon atom of at least one of the carboxyl groups bound to nitrogen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D207/00Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D207/02Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D207/30Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having two double bonds between ring members or between ring members and non-ring members
    • C07D207/34Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having two double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D215/00Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems
    • C07D215/02Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom
    • C07D215/16Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D215/38Nitrogen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D217/00Heterocyclic compounds containing isoquinoline or hydrogenated isoquinoline ring systems
    • C07D217/22Heterocyclic compounds containing isoquinoline or hydrogenated isoquinoline ring systems with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to carbon atoms of the nitrogen-containing ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D263/00Heterocyclic compounds containing 1,3-oxazole or hydrogenated 1,3-oxazole rings
    • C07D263/02Heterocyclic compounds containing 1,3-oxazole or hydrogenated 1,3-oxazole rings not condensed with other rings
    • C07D263/30Heterocyclic compounds containing 1,3-oxazole or hydrogenated 1,3-oxazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members
    • C07D263/34Heterocyclic compounds containing 1,3-oxazole or hydrogenated 1,3-oxazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D263/48Nitrogen atoms not forming part of a nitro radical
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D277/00Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings
    • C07D277/02Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings not condensed with other rings
    • C07D277/20Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members
    • C07D277/32Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D277/38Nitrogen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D307/00Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom
    • C07D307/02Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom not condensed with other rings
    • C07D307/34Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members
    • C07D307/56Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D307/66Nitrogen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/10Tetrapeptides
    • C07K5/1002Tetrapeptides with the first amino acid being neutral
    • C07K5/1005Tetrapeptides with the first amino acid being neutral and aliphatic
    • C07K5/1013Tetrapeptides with the first amino acid being neutral and aliphatic the side chain containing O or S as heteroatoms, e.g. Cys, Ser
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Abstract

Compounds which inhibit prenyl transferases, particularly farnysyltransferase and geranylgeranyl transferase I, processes for preparing the compounds, pharmaceutical compositions containing the compounds, and methods of use.

Description

CA 022072~2 1997-06-06 WO96/21456 PCT~S961~1559 INHIBITORS OF PRENYL TRANSFERASES

BACKGROUND OF THE INVENTION
l. Field of the Invention The present invention relates to novel peptidomimetics and other compounds which are useful as inhibitors of protein isoprenyl transferases (particularly protein farnesyltransferase and geranylgeranyltransferase) and as anti-cancer drugs, to compositions containing such compounds and to methods of use.
2. Background Information Ras proteins are plasma membrane- associated GTPases that function as relay switches that transduce biological information from extracellular signals to the nucleus (29-31). In normal cells Ras proteins cycle between the GDP-(inactive) and GTP-(active) bound forms to regulate proliferation and differentiation. The mechanism by which extracellular signals, such as epidermal and platelet derived growth factor (EGF
and PDGF), transduce their biological information to the nucleus via Ras proteins has recently been unraveled (29-3l). Binding of the growth factors to tyrosine kinase receptors results in autophosphorylation of various tyrosines which then bind src-homology 2 (SH2) domains of several signaling proteins. One of these, a cytosolic complex of GRB-2 and a ras exchanger (m-SOS-l), is recruited by the tyrosine phosphorylated receptor where mSOS-l catalyzes the exchange of GDP for GTP
on Ras, hence activating it. GTP-bound Ras ; recruits Raf, a serine/threonine kinase, to the plasma membrane where it is activated. Raf triggers a kinase cascade by phosphorylating mitogen-activated protein (MAP) CA 022072~2 l997-06-06 W O96/21456 PCTrUS96/01559 kinase/extracellular-regulated protein kinase (ERK) kinase (MEK) which in turn phosphorylates MAP Kinase on threonine and tyrosine residues.
Activated MAP Kinase translocates to the nucleus where it phosphorylates transcription factors (31). Termination of this growth signal is accomplished by hydrolysis of Ras-GTP to Ras-GDP.
Ras oncogenes are the most frequently identified activated oncogenes in human tumors (1-3). In a large number of human cancers, Ras is GTP-locked because of mutations in amino acids 12, 13, or 61 and the above Ras pathway no longer requires an upstream growth signal and is uninterrupted. As a consequence, enzymes in this pathway such as Raf, MEK and MAP Kinase are constitutively activated.
In addition to its inability to hydrolyze GTP, oncogenic Ras must be plasma membrane-bound to cause malignant transformation (13). Ras is posttranslationally modified by a lipid group, farnesyl, which mediates its association with the plasma membrane (10-14).
Post-translational events leading to membrane association of p21ras have previously been disclosed (10-14). The p21ras proteins are first made as pro-p21ras in the cytosol where they are modified on cysteine 186 of their carboxyl terminal sequence CAlA2X (C = cys~eine, A1 and A2 =
isoleucine, leucine or valine and X = methionine or serine) by the cholesterol biosynthesis intermediate farnesyl pyrophosphate (FPP). This farnesylation reaction is then followed by peptidase removal of the AlA2X tripeptide and carboxymethylation of the re~;n;ng cysteine. The 7 processed p21ras proteins associate with the inner CA 022072~2 1997-06-06 WO96/21456 PCT~S96/01559 surface of the plasma membrane (10-14).
p21Ras farnesyltransferase, the enzyme responsible for catalyzing the transfer of farnesyl, a 15-carbon isoprenoid, from FPP to the cysteine of the CAlA2X carboxyl terminus of p21ras, has been purified to homogeneity from rat brain (15,16). The enzyme is a heterodimer composed of and ~ subunits of molecular weights 49 and 46 kDa, respectively (17). The ~ subunit has been shown to bind p21ras (17). Because p21ras farnesylation and subsequent membrane association are required for p21ras transforming activity (13), it has been proposed that p21ras farnesyltransferase would be a useful anticancer therapy target. Accordingly, an intensive search for inhibitors of the enzyme is underway (18-24, 33-44). Potential inhibitor candidates are CA1A2X
tetrapeptides which have been shown to be farnesylated by p2lras farnesyltransferase and appear to be potent inhibitors of this enzyme in vitro (15,18,21-24). Competition studies have demonstrated that CAlA2X peptides with the greatest inhibitory activity are those where A1 and A2 are hydrophobic peptides with charged or hydrophilic residues in the central positions demonstrating very little inhibitory activity (18,21,23). A
major drawback with the use of peptides as ~ therapeutic agents is their low cellular uptake and their rapid inactivation by proteases.
The research efforts directed towards farnesyltransferase and the inhibition of its activity are further illustrated by the following patents or published patent applications:
., CA 022072~2 l997-06-06 W O96/21456 PCTrUS96/015~9 U.S. 5,141,851 Of the above disclosures, EPA 0520823 A2 discloses compounds which are useful in the inhibition of farnesyl-protein transferase and the farnesylation of the oncogene protein ras. The compounds of EPA
0520823 A2 are illustrated by the formula:
Cys-Xaa1-dXaa2-Xaa3 or pharmaceutically acceptable salts thereof, wherein Cys is a cysteine amino acid;
Xaal is an amino acid in natural L-isomer form;
dXaa2 is an amino acid in unnatural D-isomer form;
and~0 Xaa3 is an amino acid in natural L-isomer form.
The preferred compounds are said to be CV(Dl)S and CV(Df)M, the amino acids being identified by conventional 3 letter and single letter abbreviations as follows:

Cysteine Cys C
Glycine Gly G
Isoleucine Ile Leucine Leu L
Lysine Lys K
Methionine Met M
Phenylalanine Phe F
Serine Ser S
Threonine Thr T
Valine Val V

CA 022072~2 1997-06-06 WO 96/21456 ~CT/US96/01559 EPA 0523873 A1 discloses a modification of the compounds of EPA 0520823 A2 wherein Xaa3 is phenylalanine or p-fluorophenylalanine.
EPA 0461869 describes compounds which inhibit farnesylation of Ras protein of the formula:
Cys-Aaa1-Aaa2-Xaa where Aaa1 and Aaa2 are aliphatic amino acids and Xaa is an a~ino acid. The aliphatic amino acids which are disclosed are Ala, Val, Leu and Ile.
Preferred compounds are those where Aaa1 is Val, Aaa2 is Leu, Ile or Val and Xaa is Ser or Met.
Preferred specific compounds are:
Cys-Val-Leu-Ser Cys-Val-Ile-Met Cys-Val-Val-Met U.S. patent 5,141,851 and WO 91/16340 disclose the purified farnesyl protein transferase and certain peptide inhibitors therefor, including, for example, CVIM, TKCVIM and KKSKTKCVIM.
WO 92/18465 discloses certain farnesyl compounds which inhibit the enzymatic methylation of proteins including ras proteins.
EPA 0456180 A1 is directed to a farnesylprotein transferase assay which can be used to identify substances that block farnesylation of ras oncogene gene products while EPA 0512865 A2 discloses certain cyclic compounds that are useful for lowering cholesterol and inhibiting farnesylprotein transferase.
As will be evident from the foregoing, there is a great deal of research effort directed towards the development of inhibitors of farnesyltransferase. However, there still rem~;n~

CA 022072~2 1997-06-06 W O96/21456 PCTrUS96/01559 a need for improvements in this critically important area.
An enzyme closely related to farnesyltransferase, geranylgeranyltransferase I
(GGTase I), attaches the lipid geranylgeranyl to the cysteine of the CAAX box of proteins where X
is leucine (49,69). FTase and GGTase I are ~/~
heterodimers that share the ~ subunit (61,62).
Cross-linking experiments suggested that both substrates (FPP and Ras CAAX) interact with the submit of FTase (17,63). Although GGTase I
prefers leucine at the X position, its substrate specificity was shown to overlap with that of FTase in vitro (64). Furthermore, GGTase I is also able to transfer farnesyl to a leucine terminating peptide (65).
Although CAAX peptides are potent competitive inhibitors of FTase, rapid degradation and low cellular uptake limit their use as therapeutic agents. The stragegy of the present invention to develop superior compounds for inhibiting FTase and GGTase has been to replace several amino acids in the CAAX motif by peptidemimics. The rationale behind this strategy is based on the existance of a hydrophobic pocket at the enzyme active site that interacts with the hydrophobic "AA" dipeptide of the carboxyl termini CAAX of Ras molecules. In this regard, two very potent inhibitors of FTase (i.e. Cys-3AMBA-Met and Cys-4ABA-Met) were disclosed by us in an earlier U.S. patent application. The peptidomimetic Cys-4A3A-Met incorporates a hydrophobic/aromatic spacer (i.e.
4-aminobenzoic acid) between Cys and Met. The present application discloses several derivatives of Cys-4ABA-Met where positions 2 and 3 of 4-amino CA 022072~2 1997-06-06 WO96/214S6 PCT~S96101559 benzoic acid were modified by several alkyl, and/or aromatic groups, compounds that show great promise of ability to selectivelY antagonize RAS-dependent signaling and to selectively inhibit the growth of human tumors with aberrant Ras function.
Of the four types of Ras proteins (H-, N-, K4A- and R4B-Ras) expre~_ed by mAmm~l lan ~ells, K4B-Ras (also called K-Ras4B) is the most frequently mutated form of Ras in human cancers (l,3). Although several laboratories have demonstrated potent inhibition of oncogenic H-Ras processing and signaling (43,44), this disruption has not been shown with K-Ras4B. Previous studies have targeted H-Ras and not K-Ras4B as a target for the development of inhibitors. One recent report indicates that K-Ras4B can be geranylgeranylated in vitro, but with relatively low efficiency; its Km for GGTase I is 7 times higher than its Km for FTase (67). GGTase I CAAX-based inhibitors that can blockgeranylgeranylaticn processing have not been reported.
Recently, we have shown that a potent inhibitor of FTase disrupts K-Ras4B processing but only at very high concentrations that also inhibited the processing of geranylgeranylated proteins (66). This suggested that K-Ras4B may be geranylgeranylated, and that therefore inhibitors targeted at GGTase I would be effective in disrupting oncogenic K-Ràs4B processing and signalling, and in treatment of cancers which were related to this form of Ras.

=

W O96/21456 PCTrUS96/01559 Summary of the Invention In accordance with the present invention there are compounds.of the formula (A-L):

R~

R3~ ~ 2 . R3' 1 ~2 R3~ 1 ~ ,; 2 Rl~L,--~R2 R3' ~ ~ N

l,L1 r~-~/R2 , L, ~ 12 ~"1 ~T=~R2 R3 O~ N (J~ (K) or ~ (L) CA 022072~2 1997-06-06 WO96/21456 PCT~S96101559 wherein Rl' is i) hydrogen;
ii) lower alkyl;
iii) alkenyl;
iv) alkoxy;
v) thioalkoXYL
vi) halo;
vii) haloalkyl;
viii) aryl-L2-, wherein L2 is absent, -CH2-, -CH2CH2-, -CH(CH3)-, -O-, ~S(O)q wherein q is 0, 1, or 2, -N(R')- wherein R' is hydrogen or lower alkyl, or -C(O)- and aryl is selected from the group consisting of phenyl, naphthyl, tetrahydronaphthyl, indanyl and indenyl and the aryl group is unsubstituted or substituted; or ix) heterocyclic-L3- wherein L3 is absent, -CH2-, -CH2CH2-, -CH(CH3)-, -O-, ~S(O)q wherein q is 0, 1 or 2, -N(R')- wherein R' is hydrogen or loweralkyl, or -C(O)- and heterocyclic is a monocyclic heterocyclic wherein the heterocyclic is unsubstituted or substituted with one, two, or three substituents independently selected from the group consisting of loweralkyl, hydroxy, hydroxyalkyl, halo, nitro, oxo (=O), amino, N-protected amino, alkoxy, thioalkoxy and haloalkyl;

Rla is hydrogen or lower alkyl;

R2' is Ri) ~ 12a J~' R

wherein Rl2a is hydrogen, loweralkyl or -C(O)O-Rl3, wherein Rl3 is hydrogen or a carboxy-protecting CA 022072~2 l997-06-06 W O96/21456 PCTrUS96/01559 group and Rl2b is hydrogen or loweralkyl, with the proviso that Rl2a and Rl2b are not both hydrogen, ii) -C(O)NH-CH(Rl4)-C(O)ORl5 wherein Rl4 is a) loweralkyl, b) cycloalkyl, c) cycloalkylalkyl, d) alkoxyalkyl, e) thioalkoxyalkyl, f) hydroxyalkyl, g) aminoalkyl, h) carboxyalkyl, i) alkoxycarbonylalkyl, j) arylalkyl or k) alkylsulfonylalkyl and Rl5 is hydrogen or a carboxy-protecting group or iii) -C(O)-HN ~ p (CH2)1-3 R3' is A

Il Ro~CH2~CH~C~

where A represents O or 2H, and Ro represents SH, NH2, or CxHy~SO2~NH~/ wherein CXHy is a straight chain saturated or unsaturated hydrocarbon, with x being between 1 and 20 and y between 3 and 41, inclusive; and CA 022072~2 1997-06-06 WO96121456 PCT~S96101559 L1 is -NH-;
or pharmaceutically acceptable salts or prodrugs thereof.
An important embodiment of the present invention is based on the finding that a novel group of peptidomimetics as represented by Formula (I) have a high inhibitory pot~ncy against human tumor p21ras farnesyltransferase and inhibit tumor growth of human carcinomas:

C~X (I) where C stands for the cysteine radical, or for the reduced form of the cysteine radical (R-2-amino-3-mercaptopropyl amine); ~ is the radical of a non-peptide aminoalkyl- or amino-substituted phenyl carboxylic acid; and X is the radical of an amino acid, preferably Met. Any other natural or synthetic amino acid can also be used at this position. The invention also includes pharmaceutically acceptable salts and prodrugs of Formula (I).
A particularly preferred compound in this regard is:

HS

O

CA 022072~2 1997-06-06 W O 96/21456 PCTrUS961015~9 In this compound the cysteine radical is in the reduced form and the spacer group is 2-phenyl-4-aminobenzoic acid.
Another preferred compound of the invention is:

3 ~ ~

The compounds of Formula (I) are different from the prior art farnesyltransferase inhibitors in that they do not include separate peptide amino acids A1, A2 as in prior art inhibitors represented by the formula CA1A2X. The present compounds are consequently free from peptidic amide bonds.
It is also to be noted that the present compounds are not farnesylated by the enzyme.
They are, therefore, true inhibitors, not just alternative substrates. This may explain the high inhibitory action of the present compounds relative to their parent compounds which are farnesylated.
A further important feature of the invention is the provision of the compounds of Formula (I) in the form of pro-drugs. Broadly speaking, this is accomplished by functionalizing the terminal end groups (amino, cysteine sulfur and carboxy ~roups) of the compounds with hydrophobic, enzyme-sensitive moieties which serve to increase the plasma membrane permeability and cellular uptake of the compounds and consequently their efficiency in inhibiting tumor cell growth. In addition, - = -CA 022072~2 1997-06-06 WO96/21456 PCT~S96101S59 prodrugs for amino and cysteine sulfur groups can include loweralkycarbonyl, arylcarbony, arylalkylcarbony, alkoxycarbonyl, aryloxycarbonyl, cycloalkylcarbonyk, cycloalkoxycarbonyl, and other groups well known to those skilled in the art.
In this regard, a particularly preferred compound of the invention is the methylester form of FTI-276, which is illustrated in Figure lA.
The above-mentioned pro-drug aspect of the invention is applicable not only to the compounds of the invention but also to prior peptide inhibitors CAlA2X as well as any other peptide with potential for biological uses for the purpose of improving the overall effectiveness of such compounds, as hereinafter described.
A further modification involves the provision of CAlA2X tetrapeptides or C~X peptidomimetics which have been modified by functionalizing the sulfhydryl group of the cysteine C with an alkyl phosphonate substituent, as hereinafter described.
Another important embodiment of the invention contemplates replacing the A1A2X portion of the CAlA2X tetrapeptide inhibitors with a non-amino acid component while retaining the desired farnesyltransferase inhibiting activity. These compounds may be illustrated by Formula (II):

C~ (II) where C is cysteine or reduced cysteine and ~
represents an aryl or heterocyclic substituent such as 3-aminomethyl-biphenyl-3'-carboxylic acid, which does not include a peptide amino acid but corresponds essentially in size with AlA2X, as hereinafter described. The invention also includes pharmaceutically acceptable salts and prodrugs of Formula (II).

CA 022072~2 1997-06-06 W O96/21456 PCTrUS96/01559 The invention also includes compounds in which further substitutions have been made at the cysteine position. These compounds comprise free cysteine thiol and/or terminal amino groups at one end and include a carboxylic acid or carboxylate group at the other end, the carboxylic acid or carboxylate group being separated from the cysteine thiol and/or terminal amino group by a hydrophobic spacer moiety which is free from any linking amido group as in prior CAAX mimetics. As with other compounds of the invention, these compounds are not subject to proteolytic degradation inside cells while retaining the structural features required for FTase inhibition.
The compounds selectively inhibit FTase both in vitro and in vivo and offer a number of other advantages over prior CAAX peptide mimetics.
Compounds of this embodiment may be illustrated by the formula:
C~B (III) where C~ is IAI
Ro -CH2-CH-C-A represents O or 2H, and Ro represents SH, NH2, or CxHy~SO2~NH~, wherein CXHy is a straight chain saturated or unsaturated hydrocarbon, with x being between 1 and 20 and y between 3 and 41, inclusive; and B stands for -NHR, where R is an aryl group. The invention also includes pharmaceutically acceptable salts and prodrugs of Formula (III).
In one preferred embodiment of the invention, R is a biphenyl substituted with one or more -COOH

CA 022072~2 1997-06-06 W O 96/21456 PCTrUS96/015S9 groups and/or lower alkyl, e.g., methyl, as represented by the formula:

H2N~A R4 SH H ~ R3 Rl R2 where Rl and R3 represent H or COOH; R2 represents H, COOH, CH3, or COOCH3; R4 represents H or OCH3;
and A represents 2H or O. This formula represents a series of 4-amino-3'-carboxybiphenyl derivatives which mimic the Val-Ile-Met tripeptide but have restricted conformational flexibility. Reduction of the cysteine amide bond (where A is H,H) provides a completely non-peptidic Ras CA~X
mlmetic.
Preferably, R is a biphenyl group with a -COOH substitution in the 3'- or 4'-position, most preferably the 3~-position, with respect to the NH-aryl group. The -COOH substituent may appear as such or in pharmaceutically acceptable salt or ester form, e.g., as the alkali metal salt or methyl ester.
The features of the invention are illustrated herein by reference to the CAAX tetrapeptide known as CVIM (see EP 0461869 and U.S. Patent 5,141,851) and C-4ABA-M. These compounds are, respectively, Cys-Val-Ile-Met and Cys-4 aminobenzoic acid-Met where Cys is the cysteine radical and Met is the methionine radical.
A preferred non-peptide CAAX mimetic of the invention is reduced cys-4-amino-3'-biphenylcarboxylate identified as 4 in Figure 12, CA 022072~2 1997-06-06 W 096/21456 PCTtUS96tO1559 which is also designated FTI-265. This derivative contains no amide bonds and thus is a true non-peptide mimic of the CAAX tetrapeptide.
The compounds of the invention may be used in the carboxylic acid form or as pharmaceutically acceptable salts or esters thereof. Lower alkyl esters are preferred although other ester forms, e.g., phenyl esters, may also be used.
It is also an object of the present invention to provide a CA~X peptidomimetic that inhibits GGTase I.
Accordingly, it is an object of the present invention to provide a substance and means of disrupting oncogenic K-Ras4B processing and signaling that affects geranylgeranylation and/or farnesylation processing.
It is a further object of the invention to provide a pharmaceutical composition for treating cancer which is responsive to geranylgeranyl transferase inhibitors, such as, but not limited to, pancreatic and colon cancer.
The latter objects are accomplished by replacing the central "AA" of CAAX tetrapeptides by a hydrophobic spacer and incorporating a leucine or isoleucine residue in the C-terminal position to optimize recognition by GGTase I.
Additionally, the cysteine moiety may be replaced by reduced cysteine, or by other functional groups as hereinafter disclosed.
An important embodiment of the present invention is based on the finding that a novel group of peptidomimetics as represented by Formula (IV) have a high inhibitory potency against geranylgeranyl transferase and disrupt oncogenic K-Ras4B processing and signalling:

C~L (IV) CA 022072~2 1997-06-06 WO96121456 PCT~S96/~1559 where C stands for the cysteine radical, or for the reduced form of the cysteine radical (R-2-amino-3-mercaptopropyl amine); ~ is the radical of a non-peptide aminoalkyl- or amino-substituted phenyl carboxylic acid; and L is the radical of leucine or isoleucine. The invention also includes pharmaceutically acceptable salts and prodrugs of the compounds of Formula (IV).
Preferred compounds of this embodiment are derivatives of Cys-4ABA-Leu which are substituted at the 2 and/or 3 positions of the phenyl ring of 4-aminobenzoic acid (4ABA). The substitutions at these positions include, but are not limited to alkyl, alkoxy and aryl (particularly to straight chain or branched groups of l-l0 carbons of the aforementioned) and naphthyl, heterocyclic rings and heteroaromatic rings.
A particularly preferred compound of this aspect of the invention, GGTI-287, is illustrated in Figure 17. In this compound the cysteine radical is in the reduced form and the spacer group is 2-phenyl-4-aminobenzoic acid. Another preferred compound, also shown in Figure 17, is GGTI-297, which contains the spacer group 2-naphthyl-4-aminobenzoic acid. Other spacer groups which will be readily evident as useful are described herein in connection with farnesyl-transferase inhibitors.
A further important feature of the invention is the provision of the compounds of the invention in the form of pro-drugs. By "pro-drug" is meant a compound to which in vivo modification occurs to produce the active compound. Such compounds may, for example, be more readily delivered to their sites of action as pro-drugs. Broadly speaking, the pro-drugs of the instant invention are CA 022072~2 1997-06-06 W O96/21456 PCTrUS96/01559 produced by functionalizing the terminal end groups (amino, cysteine sulfur and carboxy groups) of the compounds with hydrophobic, enzyme-sensitive moieties which serve to increase the plasma membrane permeability and cellular uptake of the compounds and consequently their efficiency in inhibiting tumor cell growth.
In this regard, a particularly preferred compound of the invention is the methylester form of GGTI-287, GGTI-286, also illustrated in Figure 17.

The compounds of the invention may be used in the same manner as prior CAAX tetrapeptide inhibitors to inhibit p21ras farnesyltransferase or geranylgeranyl transferase in any host containing these enzymes. This includes both ln vitro and in vivo use. Compounds which inhibit farnesyltransferase, notably human tumor p21ras farnesyltransferase, and consequently inhibit the farnesylation of the oncogene protein Ras, may be used in the treatment of cancer or cancer cells.
It is noted that many human cancers have activated ras and, as typical of such cancers, there may be mentioned colorectal carcinoma, myeloid leukemias, exocrine pancreatic carcinoma and the like.
Likewise, compounds which inhibit geranylgeranyl transferase may be used in the treatment of cancer which is related to K-Ras4B.
The compounds of the invention may be used in pharmaceutical compositions of conventional form suitable for oral, subcutaneous, intravenous, intraperitoneal or intramuscular administration to a mammal or host. This includes, for example, tablets or capsules, sterile solutions or suspensions comprising one or more compounds of the invention with a pharmaceutically acceptable CA 022072~2 1997-06-06 WO96/21456 PCT~S96/01559 carrier and with or without other additives.
Typical carriers for tablet or capsule use include, for example, lactose or corn starch. For oral compositions, aqueous suspensions may be used with conventional suspending agents, flavoring agents and the like.
The amount of inhibitor administered to obtain the desired inhibitory effect will vary but can be readily determined. It is expected that the compounds of the present invention will be administered to humans or other mammals as pharmaceutical or chemotherapeutic agents in dosages of .1 to 1000 mg/kg body weight, preferably 1 to 500 mg/kg body weight and most preferably 10-50 mg/kg body weight. The required dose for a given individual or disease will vary, but can be determined by ordinary skilled practitioners using routine methods. The compounds may be administered via methods well known in the pharmaceutical and medical arts, which include, but are not limited to oral, parenteral, topical, and respiratory (inhalation) routes. Pharmaceutical preparations may contain suitable carriers or diluents. Means of determ;n;ng suitable carriers and diluents are well known in the pharmaceutical arts.
The term "carboxy protecting group~, as used herein, refers to a carboxylic acid protecting ester group employed to block or protect the carboxylic acid functionally while the reactions involving other functional sites of the compound are carried out. Carboxy protecting groups are disclosed in Greene, "Protective Groups in Organic Synthesis", pp. 152-186 (1981), which is hereby incorporated herein by reference. In addition, a carboxy protecting group can be used as a prodrug whereby the carboxy protecting group can be CA 022072~2 1997-06-06 W O 96/21456 PCTrUS96/01559 readily cleaved in vivo, for example by enzymatic hydrolysis, to release the biologically active parent. A comprehensive discussion of the prodrug concept is provided by T. Higuchi and V. Stella in "Prodrugs as Novel Delivery Systems", vol. 14 of the ACS Symposium Series, American Chemical Society (1975), which is hereby incorporated by reference. Such carboxy protecting groups are will known to those skilled in the art, having been extensively used in the protection of carboxyl groups in the penicillin and cephalosporin fields, as described in U.S. Pat.
No. 3,840,556 and 3,719,667, the disclosures of which are hereby incorporated by reference.
Examples of esters useful as prodrugs for compounds containing carboxyl groups can be found on pages 14-21 of "Bioreversible Carriers in Drug Design: Theory and Application", edited by E.B.
Roche, Permagon Press, New York, (1987) which is hereby incorporated by reference. Representative carboxy protecting groups are C1 to C8 loweralkyl (e.g. methyl, ethyl or tertiary butyl and the like); arylalkyl, for example, phenethyl or benzyl and substituted drivatives thereof, for example 5-indanyl and the like; dialkylaminoalkyl (e.g.dimethylaminoethyl and the like); alkanoyloxyalkyl groups such as acetoxymethol, butyryloxymethyl, valeryloxymethyl, isobutyryloxymethyl, isovaleryloxymethyl, 1-(propionyloxy)-1-ethyl, 1-(pivaloyloxyl)-1-ethyl, 1-methyl-1-(propionyloxy)-1-ethyl, pivaloyloxymethyl, propionyloxymethyl and the like; cycloalkanoyloxyalkyl groups such as cyclopropylcarbonyloxymethyl, cyclobutylcarbonyloxymethyl, cyclopentylcarbonyloxymethyl, cyclohexylcarbonyloxymethyl and the like;
aroyloxyalkyl, such as benzoyloxymethyl, CA 022072~2 1997-06-06 WO96/21456 PCT~S96101559 benzoyloxyethyl and the like;
arylalkylcarbonyloxyalkyl, such as benzylcarbonyloxymethyl, 2-benzylcarbonyloxyethyl and the like; alkoxycarbonylalkyl or cycloalkyloxycarbonylalkyl, such as methoxycarbonylmethyl, cyclohexyloxycarbonylmethyl, l-methoxycarbonyl-1-ethyl, and the like; alkoxycarbonyloxyalkyl or cycloalkyloxycarbonylalkyl, such as methoxycarbonyloxymethyl, t-butyloxycarbonyloxymethyl, 1-ethoxycarbonyloxy-1-ethyl, l-cyclohexyloxycarbonyloxy-l-ethyl and the like; aryloxycarbonyloxyalkyl, such as 2-(phenoxycarbonyloxy)ethyl, 2-(5-indanyloxycarbonyloxy)ethyl and the like;
alkoxyalkylcarbonyloxyalkyl, such as 2-(1-methoxy-2-methyl~ropan-2-oyloxy)ethyl and the likei arylalkyloxycarbonyloxyalkyl, such as 2-(benzyloxycarbonyloxy)ethyl and the like;
arylalkenyloxycarbonyloxyalkyl, such as 2-(3-phenylpropen-2-yloxycarbonyloxy)ethyl and the like; alkoxycarbonylaminoalkyl, such as t-butyloxycarbonylaminomethyl and the like;
alkylaminocarbonylaminoalkyl, such as methylaminocarbonylaminomethyl and the like;
alkanoylaminoalkyl, such as acetylaminomethyl and the like; heterocycliccarbonyloxyalkyl, such as 4-methylpiperazinylcarbonyloxymethyl and the like;
dialkylaminocarbonylalkyl, such as dimethylaminocarbonylmethyl, diethylaminocarbonylmethyl and the like; (5-(loweralkyl)-2-oxo-1,3-dioxolen-4-yl)alkyl, such as (5-t-butyl-2-oxo-1,3-dioxolen-4-yl)methyl and the like; and (5-phenyl-2-oxo-1,3-dioxolen-4-yl)alkyl, such as (5-phenyl-2-oxo-1,3-dioxolen-4-yl)methyl and the like.

CA 022072~2 1997-06-06 W O 96/21456 PCTnUS96/01559 Preferred carboxy-protected compounds of the invention are compounds wherein the protected carboxy group is a loweralkyl, cycloalkyl or arylalkyl ester, for example, methyl ester, ethyl ester, propyl ester, isopropyl ester, butyl ester, sec-butyl ester, isobutyl ester, amyl ester, isoamyl ester, octyl ester, cyclohexyl ester, phenylethyl ester and the like or an alkanoyloxyalkyl, cycloalkanoyloxyalkyl, aroyloxyalkyl or an arylalkylcarbonyloxyalkyl ester.
The term "N-protecting group" or "N-protected" as used herein refers to those groups intended to protect the N-terminus of an amino acid or peptide or to protect an amino group against undesirable reactions during synthetic procedures. Commonly used N-protecting groups are disclosed in Greene, "Protective Groups in Organic Synthesis," (John Wiley & Sons, New York (1981)), which is hereby incorporated herein by reference.
The term "alkanoyl" as used herein refers to R29C(O)-O- wherein R29 is a loweralkyl group.
The term "alkanoylaminoalkyl" as used herein refers to a loweralkyl radical to which is appended R7l-NH- wherein R,1 is an alkanoyl group.
The term "alkanoyloxy" as used herein refers to R29C(O)-O- wherein R29 is a loweralkyl group.
The term "alkanoyloxyalkyl" as used herein refers to a loweralkyl radical to which is appended an alkanoyloxy group.
The term "alkenyl" as used herein refers to a straight or branched chain hydrocarbon containing from 2 to 10 carbon atoms and also containing at least one carbon-carbon double bond. Examples of alkenyl include -CH=CH2, -CH2CH=CH2, -C(CH3)=CH2, -CH2CH=CHCH3, and the like.

CA 022072~2 1997-06-06 WO96/21456 PCT~S96101559 The term "alkenylene" as used herein refers to a divalent group derived from a straight or branched chain hydrocarbon containing from 2 to 10 carbon atoms and also containing at least one carbon-carbon double bond. Examples of alkenylene include -CH=CH-, -CH2CH=CH-, -C(CH3)=CH-, -CH2CH=CHCH2-, and the like.
The term "alkoxy" as used herein refers to R30O- wherein R30 is loweralkyl as defined above.
Representative examples of alkoxy groups include methoxy, ethoxy, t-butoxy and the like.
The term "alkoxyalkoxy" as used herein refers to R3lO-R32O- wherein R31 is loweralkyl as defined above and R32 is an alkylene radical.
Representative examples of alkoxyalkoxy groups include methoxymethoxy, ethoxymethoxy, t-butoxymethoxy and the like.
The term "alkoxyalkyl" as used herein refers to an alkoxy group as previously defined appended to an alkyl group as previously defined. Examples of alkoxyalkyl include, but are not limited to, methoxymethyl, methoxyethyl, isopropoxymethyl and the like.
The term "alkoxyalkylcarbonyloxyalkyl" as used herein refers to a loweralkyl radical to which is appended R66-C(O)-O- wherein R66 is an alkoxyalkyl group.
The term "alkoxycarbonyl" as used herein refers to an alkoxy group as previously defined appended to the parent molecular moiety through a carbonyl group. Examples of alkoxycarbonyl include methoxycarbonyl, ethoxycarbonyl, isopropoxycarbonyl and the like.
The term "alkoxycarbonylaklyl" as used herein refers to an alkoxylcarbonyl group as previously defined appended to a loweralkyl radical.

CA 022072~2 l997-06-06 W O 96/21456 PCTrUS96/01559 Examples of alkoxycarbonylaklyl include methoxycarbonylmethyl, 2-ethoxycarbonylethyl and the like.
The term "alkoxycarbonylaminoalkyl" as used herein refers to a loweralkyl radical to which is appended R69-NH- wherein R69 is an alkoxycarbonyl group.
The term "alkoxycarbonyloxyalkyl" as used herein refers to a loweralkyl radical to which is appended R63-O- wherein R63 is an alkoxycarbonyl group.
The term "alkylamino" as used herein refers to R3sNH- wherein R35 is a loweralkyl group, for example, methylamino, ethylamino, butylamino, and the like.
The term "alkylaminoalkyl" as used herein refers a loweralkyl radical to which is appended an alkylamino group.
The term "alkylaminocarbonylaminoalkyl" as used herein refers to a loweralkyl radical to which is appended R70-C(O)-~H- wherein R70 is an alkylamino group.
The term "alkylene" as used herein refers to a divalent group derived from a straight or branched saturated hydrocarbon having from 1 to 10 carbon atoms by the removal of two hydrogen atoms, for example methylene, 1,2-ethylene, 1,1-ethylene, 1,3-propylene, 2,2-dimethylpropylene, and the like.
The term "alkylsulfinyl" as used herein refers to R33S(O)- wherein R33 is a loweralkyl group.
The term "alkylsulfonyl" as used herein refers to R34S(O) 2- wherein R34 is a loweralkyl group.

CA 022072~2 1997-06-06 W096/21456 PCT~S96J01559 The term "alkylsul~onylalkyl" as used herein refers to a loweralkyl radical to which is appended an alkylsulfonyl group.
The term "alkynyl'~ as used herein refers to a straight or branched chain hydrocarbon containing from 2 to 10 carbon atoms and also containing at least one carbon-carbon triple bond. Examples o~
alkynyl include - CeCH, -CH2C_CH, -C~2C_CCH3, and the like.
The term "alkynylene" as used herein refers to a divalent group derived from a straight or branched chain hydrocarbon containing from 2 to 10 carbon atoms and also containing at least one carbon-carbon triple bond. Examples of alkynylene include -CaC-, -CH2C_C-, -CH2C_CCH2, and the like.
The term "amino" as used herein refers to -H2.
The term "aminoalkyl" as used herein refers to a loweralkyl radical to which is appended anamino group.
The term "aroyloxyalkyl" as used herein refers to a loweralkyl radical to which is appended an aroyloxy group (i.e., R61-C(O)O-wherein R61 is an aryl group).
The term "aryl" as used herein refers to a mono-or bicyclic carbocyclic ring system having one or two aromatic rings including, but not limited to, phenyl, naphthyl, tetrahydronaphthyl, indanyl, indenyl and the like. Aryl groups (including bicyclic aryl groups) can be unsubstituted or substituted with one, two or three substituents independently selected from loweralkyl, haloalkyl, alkoxy, thioalkoxy, amino, alkylamino, dialkylamino, hydroxy, halo, mercapto, nitro, cyano, carboxaldehyde, carboxy, alkoxycarbonyl, haloalkyl-C(O)-NH-, haloalkenyl-C(O)-NH- and CA 022072~2 1997-06-06 W O96/21456 PCTrUS96/01559 carboxamide. In addition, substituted aryl groups include tetrafluorophenyl and pentafluorophenyl.
The term ''arylalkenyl'l as used herein refers to an alkenyl radical to which is appended an aryl group.
The term "arylalkenyloxycarbonyloxyalkyl" as used herein refers to a loweralkyl radical to which is appended R68-O-C(O)-O- wherein R68 is an arylalkenyl group.
The term "arylalkyl" as used herein refers to a loweralkyl radical to which is appended an aryl group. Representative arylaikyl groups include benzyl, phenylethyl, hydroxybenzyl, fluorobenzyl, fluorophenylethyl and the like.
The term "arylalkylcarbonyloxyalkyl" as used herein refers to a loweralkyl radical to which is appended an arylalkylcarbonyloxy group (i.e., R62C(O)o-wherein R62 is an arylalkyl group).
The term "arylalkyloxycarbonyloxyalkyl" as used herein refers to a loweralkyl radical to which is appended R67-O-C(O)-O- wherein R67 is an arylalkyl group.
The term "aryloxyalkyl" as used herein refers to a loweralkyl radical to which is appended R6s-O-wherein R65 is an aryl group.
The term "aryloxthioalkoxyalkyl" as used herein refers to a loweralkyl radical to which is appended R75-S- wherein R75 is an aryloxyalkyl group.
The term "aryloxycarbonyalkyl" as used herein refers to a loweralkyl radical to which is appended R65-O-C(O)-O- wherein R65 is an aryl group.
The term "arylsulfonyl" as used herein refers to R36S(O) 2- wherein R36 is an aryl group.
The term "arylsulfonyloxy" as used herein refers to R37S(O) 2~- wherein R37 is an aryl group.

CA 022072~2 1997-06-06 W Og6/21456 PCTnUS96/01559 The term "carboxyalkyl" as used herein refers to a loweralkyl radical to which is appended a carboxy (-COOH) group.
The term "carboxaldehyde" as used herein refers to the group -C(O)H.
The term "carboxamide" as used herein refers to the group -C(O)NH2.
The term "cyanoalkyl" as used herein refers to a loweralkyl radical to which is appended a cyano (-CN) group.
The term "cycloalkanoylalkyl" as used herein refers to a loweralkyl radical to which is appended a cycloalkanoyl group (i.e., R60-C(O)-wherein R60 is a cycloalkyl group).
The term "cycloalkanoyloxyalkyl" as used herein refers to a loweralkyl radical to which is ap~ended a cycloalkanoyloxy group (i e=; R6U-C(O)O
wherein R60 is a cycloalkyl group).
The term "cycloalkenyl" as used herein refers to an alicyclic group comprising from 3 to 10 carbon atoms and containing a carbon-carbon double bond including, but not limited to, cyclopentenyl, cyclohexenyl and the like.
The term "cycloalkyl" as used herein refers to an alicyclic group comprising from 3 to 10 carbon atoms including, but not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, norbornyl, adamantyl and the like.
The term "cycloalkylalkyl" as used herein refers to a loweralkyl radical to which is appended a cycloalkyl group. Representative examples of cycloalkylalkyl include cyclopropylmethyl, cyclohexylmethyl, 2-(cyclopropyl)ethyl, adamantylmethyl and the like.
The term "cycloalkyloxycarbonyloxyalkyl" as used herein refers to a loweralkyl radical to CA 022072~2 1997-06-06 W O96/214S6 PCT~US96/01559 which is appended R64-O-C(O)-O- wherein R64 is a cycloalkyl group.
The term "dialkoxyalkyl" as used herein refers to a loweralkyl radical to which is appended two alkoxy groups.
The term "dialkylamino" as used herein refers to R38R39N- wherein R38 and R39 are independently selected from loweralkyl, for example, dimethylamino, diethylamino, methyl propylamino, and the like.
The term "dialkylaminoalkyl" as used herein refers to a loweralkyl radical to which is appended a dialkylamino group.
The term "dialkyaminocarbonylalkyl" as used herein refers to a loweralkyl radical to which is appended R,3-C(O)- wherein R73 is a dialkylamino group.
The term "dioxoalkyl~' as used herein refers to a loweralkyl radical which is substituted with two oxo (=0) groups.
The term "dithioalkoxyalkyl" as used herein refers to a loweralkyl radical to which is appended two thioalkoxy groups.
The term "halogen~ or ~'halo" as used herein refers to I, Br, Cl or F.
The term ~haloalkenyl~ as used herein refers to an alkenyl radical, as defined above, bearing at least one halogen substituent.
The term "haloalkyl" as used herein refers to a lower alkyl radical, as defined above, bearing at least one halogen substituent, for example, chloromethyl, fluoroethyl or trifluoromethyl and the like.
The term "heterocyclic ring" or "heterocyclic~ or "heterocycle" as used herein refers to a 5-, 6- or 7-membered ring containing one, two or three heteroatoms independently CA 022072~2 1997-06-06 W O96/214S6 PCTnUS96/015S9 selected from the group consisting of nitrogen, oxygen and sulfur or a 5-membered ring containing 4 nitrogen atoms; and includes a 5-, 6- or 7-membered ring cont~;n;ng one, two or three nitrogen atoms; one oxygen atom; one sulfur atom;
one nitrogen and one sulfur atom; one nitrogen and one oxygen atom; two oxygen atoms in non-ad]acent positions; one oxygen and one sulfur atom in non-adjacent positions; two sulfur atoms in non-adjacent positions; two sulfur atoms in adjacentpositions and one nitrogen atom; two adjacent nitrogen atoms and one sulfur atom; two non-adjacent nitrogen atoms and one sulfur atom; two non-adjacent nitrogen atoms and one oxygen atom.
The 5-membered ring has 0-2 double bonds and the 6- and 7-membered rings have 0-3 double bonds.
~ The term "heterocyclic" also includes bicyclic, tricyclic and tetracyclic groups in which any of the above heterocyclic rings is fused to one or two rings independently selected from the group consisting of an aryl ring, a cyclohexane ring, a cyclohexene ring, a cyclopentane ring, a cyclopenene ring and another monocyclic heterocyclic ring (for example, indolyl, quinolyl, isoquinolyl, tetrahydroquinolyl, benzofuryl or benzothienyl and the like). Heterocyclics include: pyrrolyl, pyrrolinyl, pyrrolidinyl, pyrazolyl, pyrazolinyl, pyrazolidinyl, imidazolyl, imidazolinyl, imidazolidinyl, pyridyl, piperidinyl, homopiperidinyl, pyrazinyl, piperazinyl, pyrimidinyl, pyridazinyl, oxazolyl, oxazolidinyl, isoxazolyl, isoxazolidinyl, morpholinyl, thiomorpholinyl, thiazolyl, thiazolidinyl, isothiazolyl, isothiazolidinyl, indolyl, quinolinyl, isoquinolinyl, benzimidazolyl, benzothiazolyl, benzoxazolyl, furyl, thienyl, thiazolidinyl, isothiazolyl, CA 022072~2 1997-06-06 WO96121456 PCT~S96/01559 triazolyl, tetrazolyl, oxadiazolyl, thiadiazolyl, pyrimidyl, tetrahydrofuranyl, dihydrofuranyl, tetrahydrothienyl, dihydrothienyl, dihydroindolyl, tetrahydroquinolyl, tetrahydroisoquinolyl, pyranyl, dihydropyranyl, dithiazolyl, benzofuranyl and benzothienyl. Heterocyclics also include bridged bicyclic groups wherein a monocyclic heterocyclic group is bridged by an alkylene group, for example, ~ , and the like.

Heterocyclics also include compounds of the formula ~\ ~ ~ X~
~o'Y
wherein X*.is -CH2-, -CH2O- or -O- and Y* is -C(O)-or -(C(R")2)V- wherein R" is hydrogen or Cl-C4-alkyl and v is 1, 2 or 3 such as 1,3-benzodioxolyl, 1,4-benzodioxanyl and the like.
Heterocyclics can be unsubstituted or substituted with one, two or three substituents independently selected from the group consisting of a) hydroxy, b) -SH, c) halo, d) oxo (=O), e) thioxo (=S), f) amino, g) -NHOH, h) alkylamino, i) dialkylamino, CA 022072~2 1997-06-06 WO96/21456 PCT~S96101559 j) alkoxy, k) alkoxyalkoxy.
1) haloalkyl.
_ m) hydroxyalkyl, n) alkoxyalkyl, o) cycloalkyl, p) cycloalkenyl, q) alkenyl, r) alkynyl, s) aryl, t) arylalkyl, u) -COOH, v) -SO3H, w) loweralkyl, x) alkoxycarbonyl, y) -C (O) NH2 ~
z) -C (S) NH2, aa) -C(=N-OH)NH2, bb) loweralkyl-C(O)-, cc) loweralkyl-C(S)-, dd) formyl, ee) cyano, and ff) nitro.

The term "(heterocyclic)alkyl" as used herein refers to a heterocyclic group as defined above appended to a loweralkyl radical as defined above.
Examples of heterocyclic alkyl include 2-pyridylmethyl, 4-pyridylmethyl, 4-quinolinylmethyl and the like.
The term "heterocycliccarbonyloxyalkyl" as used herein refers to a loweralkyl radical to which is appended R72-C(O)-O- wherein R72 is a heterocyclic group.
The term "hydroxyalkyl" as used herein refers to a loweralkyl radical to which is appended an hydroxy group.

CA 022072~2 1997-06-06 W O 96/21456 PCTrUS96/01559 The term "hydroxythioalkoxy" as used herein refers to R51S- wherein R51 is a hydroxyalkyl group.
The term "loweralkyl" as used herein refers to branched or straight chain alkyl groups comprising one to ten carbon atoms, including methyl, ethyl, propyl, isopropyl, n-butyl, neopentyl and the like.
The term "N-protected alkylaminoalkyl" as used herein refers to an alkylaminoalkyl group wherein the nitrogen is N-protected.
The term "oxoalkyloxy" as used herein refers to an alkoxy radical wherein the loweralkyl moiety is substituted with an oxo (=O) group.
The term "spiroalkyl" as used herein refers to an alkylene diradical, both ends of which are bonded to the same carbon atom of the parent group to form a spirocyclic group.
The term "thioalkoxy" as used herein refers to Rs2S- wherein R52 is loweralkyl. Examples of thioalkoxy include, but are not limited to, methylthio, ethylthio and the like.
The term "thioalkoxyalkyl" as used herein refers to a thioalkoxy group as previously defined appended to a loweralkyl group as previously defined. Examples of thioalkoxyalkyl include thiomethoxymethyl, 2-thiomethoxyethyl and the like.
The present invention also relates to processes for preparing the compounds of formula (l)-(Xll) and to the synthetic intermediates useful in such processes.
In a further aspect of the present invention are disclosed pharmaceutical compositions which comprise a compound of the present invention in combination with a pharmaceutically acceptable carrler .

CA 022072~2 1997-06-06 W096/21456 PCT~S96/01559 In yet another aspect of the present invention are disclosed pharmaceutical compositions which comprise a compound of the present invention in combination with another chemotherapeutic agent and a pharmaceutically acceptable carrier.
In yet another aspect of the present invention is disclosed a method for inhibiting protein isoprenyl transferases (i.e., protein farnesyltransferase and/or geranylgeranyltransferase) in a human or lower m~mm~ 1, comprising administering to the patient a therapeutically effective amount of a compound of the invention.
In yet another aspect of the present invention is disclosed a method for inhibiting post-translational modiflcation of the oncogenic Ras protein by protein farnesyltransferase, protein geranylgeranyltransferase or both.
In yet another aspect of the present invention is disclosed a method for treatment of conditions mediated by farnesylated or geranylgeranylated proteins, for example, treatment of Ras associated tumors in humans and other m~mm~ ls .
In yet another aspect of the present invention is disclosed a method for inhibiting or treating cancer in a hllm~n or lower mammal, comprising administering to the patient a therapeutically effective amount of a compound of the invention alone or in combination with another chemotherapeutic agent.
In yet another aspect of the present invention is disclosed a method for treating or preventing restenosis in a hllm~n or lower mammal, comprising administering to the patient a CA 022072~2 1997-06-06 WO96/21456 PCT~S96/01559 therapeutically effective amount of a compound of the invention.
The compounds of the present invention can be used in the form of pharmaceutically acceptable salts derived from inorganic or organic acids.
These salts include but are not limited to the following: acetate, adipate, alginate, citrate, aspartate, benzoate, benzenesulfonate, bisulfate, butyrate, camphorate, camphorsulfonate, digluconate, cyclopentanepropionate, dodeoylsulfate, ethanesulfonate, glucoheptanoate, glycerophosphate, hemisulfaté, heptanoate, hexanoate, fumarate, hydrochloride, hydrobromide, hydroiodide, 2-hydroxy-ethanesulfonate, lactate, maleate, methanesulfonate, nicotinate, 2-naphthalenesulfonate, oxalate, pamoate, pectinate, persulfate, 3-phenylpropionate, picrate, pivalate, propionate, succinate, tartrate, thiocyanate, p-toluenesulfonate and undecanoate. Also, the basic nitrogen-containing groups can be quaternized with such agents as lQweralky halides (such as methyl, ethyl, propyl, and butyl chloride, bromides, and iodides), dialkyl sulfates like dimethyl, diethyl, dibutyl, and diamyl sulfates, long chain halides such as decyl, lauryl, myristyl and stearyl chlorides, bromides and iodides, aralkyl halides like benzyl and phenethyl bromides, and others.
Water or oil-soluble or disperisble products are thereby obtained.
Examples of acids which may be employed to form pharmaceutically acceptable acid addition sales include such inorganic acids as hydrochloric acid, sulphuric acid and phosphoric acid and such organic acids as oxalic acid, maleic acid, succinic acid and citric acid.
Basic addition salts can be prepared in si tu during the final isolation and purification of the CA 022072~2 1997-06-06 WO96/21456 PCT~S96/01559 compounds of formulas A-L, or separately by reacting the carboxylic acid function with a suitable base such as the hydroxide, carbonate or bicarbonate of a pharmaceutically acceptable metal cation or with ammonia, or an organic primary, secondary or tertiary amine. Such pharmaceutically acceptable salts include, but are not limited to, cations based on the alkali and alkaline earth metals, such as sodium, lithium, potassium, calcium, magnesium, aluminum salts and the likes, as well as nontoxic ammonium, quaternary ammonium, and amine cations, including, but not limited to ammonium, tetramethylammonium, tetraethylammonium, methylamine, dimethylamine, trimethylamine, triethylamine, ethylamine, and the like. Other representative organic amines useful for the formation of base addition salts include diethylamine, ethylenediamine, ethanolamine, diethanolamine, piperazine and the like.
Other features of the invention will also be hereinafter apparent.

Brief Description of the Drawinqs Figure 1: Ras CAAX pepti~m;metics and FTase/GGTase I activities A. Structures of CVIM, FTI-249, FTI-276 and FTI-277. FTI-276 and FTI-277 were synthesized as described in Examples 10 and 11. B. FTase and GGTase I inhibition assays were carried out as described in Example 12 by determining the ability of FTI-276 to inhibit the transfer of farnesyl and geranylgeranyl to recombinant H-Ras-CVLS and H-Ras-CVLL, respectively. The data are representative of at least three different experiments.

CA 022072~2 1997-06-06 W O96/21456 PCTrUS96/01~59 Figure 2: Inhibition of Ras and RaplA Processing A. H-RasF cells were treated with various concentrations of FTI-277, lysed and the lysates immunoblotted with anti-Ras or anti-RaplA
antibodies as described in Example 13. B.
pZIPneo, H-RasF, H-RasGG, Raf and S186 cells were treated with vehicle or FTI-277 (5 ~M), lysed and lysates immunoblotted by anti-Ras antibody. Data is representative of 5 different experiments. The cells were obtained from Dr. Ch~nn;ng Derr University of North Carolina, Chapel Hill, North Carolina.

Figure 3: Effects of FTI-277 on Ras/Raf Association. pZIPneo, H-RasF, H-RasGG and S186 cells were treated with vehicle or FTI-277 (5 ~M), homogenized and the membrane (A) and cytosolic (B) fractions were separated and immunoprecipitated by an anti-Raf antibody. The immunoprecipitates were then separated by SDS-PAGE and immunoblotted with anti-RAS antibody as described in Example 14.
Data is representative of three different experiments.

Figure 4: Effects of FTI-277 on Ras Nucleotide B; n~; ng and Raf ~inase Activity A: H-RasF cells were treated with vehicle or FTI-277, lysed and the lysates immunoprecipitated with anti-Ras antibody. The GTP and GDP were then released from Ras and separated by TLC as described in Example 15. B: pZIPneo and H-RasF
cells were treated with vehicle or FTI-277, lysed and cells lysates immunoprecipitated with an anti-Raf antibody. Raf kinase was assayed by using a 19-mer autophosphorylation peptide as substrate as described in Example 16. Data are representative of three different experiments.

CA 022072~2 1997-06-06 WO96/214S6 PCT~S961015S9 Figure 5: Effect of FTI-277 on Oncogenic Activation of MAPR
A: H-RasF cells were treated with various concentrations of FTI-277, cells lysed and lysates run on SDS-PAGE and ;mml~noblotted with anti-MAPK
antibody. B: pZIPneo, H-RasF, H-RasGG, Raf, and Sl86 cells were treated with vehicle of FTI-277 (5 ~M), lysed and cells lysates processed as for A.
Data are representative of two different experiments.

Figure 6. FTI-276 inhibits selectively Ras processing and oncogenic Ra~ activation of MAP
R;n~e. NIH 3T3 cells transfected with empty vector (pZIPneo), oncogenic (GTP-locked) farnesylated Ras (RasF), geranylgeranylated Ras (RasGG) or a transforming mutant of human Raf-l were obtained from ~h~nn;ng Der and Adrienne Cox (University of North Carolina, Chapel Hill, NC, USA) (26,27). The cells were plated in DMEM/l0 CS (Dubelco's Modified Eagles Medium, l0~ calf serum) on day one and treated with vehicle or FTI-270 (20 ~M) on days 2 and 3. The cells were then harvested on day 4 and lysed in lysis buffer (30 mM HEPES, pH 7.5, l~ TX-l00, l0~ glycerol, l0 mM
NaCl, 5 mM MgCl2, 25 mM NaF, l mM EGTA, 2 mM
Na3VO4, l0 ~g/ml Trypsin inhibitor, 25 ~g/ml leupeptin, l0 ~g/ml aprotinin, 2 mM PMSF). The lysate (35 ~g) was electrophoresed on 15~ SDS-PAGE, transferred to nitrocellulose membranes and immunoblotted simultaneously with anti-Ras antibody Yl3-238 (isolated from hybridomas purchased from ATCC, Rockville, MD) and an Anti-MAP kinase (erk2) antibody (UBI, Lake Placid, NY) as described previously (l7, 22).

CA 022072~2 l997-06-06 W O 96/21456 PCTrUS96/01559 Figure 7. Antitumor efficacy of FTI-276 against h~ n lung carc;no~-~. Calu-1 (Panel A) and NCI-H810 cells (Panel B) were purchased from ATCC and grown in McCoy's 5A medium in 10~ FBS (Fetal Bovine Serum) and RPMI 1640 in 10~ FBS, respectively. The cells were harvested, resuspended in PBS and injected s.c. into the right and left flank of 8 week old female nude mice (107 cells/flank). Nude mice (Harlan Sprague Dawley, Indianapolis, Indiana) were maintained in accordance with the Institutional Animal Care and Use Committee (IACUC) procedures and guidelines.
On day 32 after s.c. implantation of tumors, animals were dosed i.p. with 0.2 ml once daily for 36 days. Control animals (filled circles) received a saline vehicle whereas treated animals (open triangles) were injected with FTI-276 (50 mg/kg). The tumor volumes were determined by measuring the length (l) and the width (w) and calculating the volume (V = (l) X (w)2/2). Data are presented as the average volume of eight tumors in each group for each cell line.
Statistical significance between control and treated groups were evaluated by using student t test (~P<0.05).

Figure 8. Antitumor Efficacy of FTI-276 and FTI-277 in Human Lung Carcinoma (Calu-1) Cells.
Experimental procedure was the same as described in Figure 7.

Figure 9. Inhibition of Tumor Growth in Ras transformed cells by FTI-276 and FTI-277. Ras-transformed NIH 3T3 cells were implanted subcutaneously into nude mice, and daily intraperitoneal injections with FTI-276 and FTI-CA 022072~2 1997-06-06 W096121456 PCT~S96/01559 277 (50 mg/kg) were started when the tumors reached 50 mm3.

Figure 10. Inhibition of Tumor Growth in Raf transfor~ed cells by FTI-276 and FTI-277. Raf-transformed NIH 3T3 cells were implanted subcutaneously into nude mice, and daily intraperitoneal injections with FTI-276 and FTI-277 (50 mg/kg) were started when the tumors reached 50 mm3.

Figure 11. Dose response: Antitumor efficacy and Ras processing correlations. A. Antitumor efficacy was carried out as described in Fig. 3 except that animals were randomly assigned to four groups each of 4 mice each (2 tumors per mouse).
Saline treated groups (circles); FTI-276 treated groups: 10 mg/kg (squares), 50 mg/kg (upward triangles), 100 mg/kg (downward triangles). B.
Ras processing was carried out 5 hours after the last treatement on day 17. Tumors were extracted from the animals, tissumized, and lysed in lysis buffer as described in Fig. 1. ~ysates (25 ~g) were electrophoresed on a 12.5~ SDS-PAGE and immunoblotted with anti-Ras antibody Y13-238 as described previously. The blots were then reprobed with anti-RaplA antibody (Santa Cruze Biotechnologies, Santa Cruz, California).

Figure 12. Structures of CVIM, C-4ABA-M, reduced C-4ABA-M, FTI-265 (4), FTI-271 (5), and FTI-261 (8).

Figure 13. Energy-m;n;m;zed structural conformations for CVIM and farnesyltransferase inhibitor FTI-265.

, CA 022072~2 1997-06-06 W O96/21456 PCTrUS96/01559 Figure~ 14A and B. C~r~rison of FTase and GGTase I inhibition by FTI-265 and FTI-271.

Figure 15. Silica gel T~C relating to Ras CAAX
peptide and peptidomimetic farnesylation.

Figure 16. Ras and RaplA processing in cells using a compound according to the invention.

Figure 17. CAAX peptidomimetic structures.
Structures of FTI-276/277, GGTI-287/286, and GGTI-297.

Figure 18. Disruption of H-Ras and RaplA
processing. NIH 3T3 cells that overexpress oncogenic H-Ras were treated with various concentrations of FTI-277 (0-50 ~M) or GGTI-286 (0-30 ~M). The cells were lysed and the lysates were electrophoresed on SDS-PAGE and imml7nohlotted with either anti-Ras or anti-RaplA antibodies as described in Example 3. U and P designate unprocessed and processed forms of the proteins.
Data are representative of three independent experiments.

Figure 19. Disruption of R-Ras4B processing.
NIH 3T3 cells that overexpress oncogenic K-Ras4B
were treated with FTI-277 or GGTI-286 (0-30 ~M).
The cells were lysed and the lysates were electrophoresed on SDS-PAGE and immunoblotted with anti-Ras antibodies as described in Example 3. U
and P designate unprocessed and processed forms of Ras. The data are representative of three independent experiments.

Figure 20. Inhibition of oncogenic acti~ation of MAP Rinase. NIH 3T3 cells that overexpress CA 022072~2 1997-06-06 WO96/21456 PCT~S96/01559 either oncogenic H-Ras or K-Ras4B were treated with either FTI-277 or GGTI-286 (0-30 ~M). The cells were lysed and the lysates were electrophoresed on SDS-PAGE and immunoblotted with an anti-MAP kinase antibody. P-MARK designates hyperphosphorylated MAP kinase. The data are representative of three independent experiments.

Figure 21. Inhibition of FTaRe and GGTase I
Activity by GGTI-297.

Figure 22. Antitumor Efficacy of GGTI-286 in K-Ras4B.
Description of Preferred Embodiments For ease of reference, the following abbreviations may be used in the present specification:
FTase: farnesyltransferasei GGTase: geranylgeranyltransferase SDS-PAGE: sodium dodecyl sulfate polyacrilamide gel electrophoresis PBS: phosphate-buffered saline;
CAAX: tetrapeptide where C is cysteine, A is an aliphatic amino acid and X is an amino acid DTT: dithiothreitol DOC: deoxycholate BSA: bovine serum albumin GGTase I: geranylgeranyl transferase I;
PAGE: polyacrylamide gel electrophoresis;
MAPK: mitogen activated protein kinase;
FTI: farnesyltransferase inhibitor;
GGTI: geranylgeranyltransferase inhibitor;

CA 022072~2 1997-06-06 W O 96121456 PCTrUS96/01559 PMSF: phenylmethylsulfonyl fluoride.

I. Farnesyltransferase Inhibitors of the type C~X
The peptidomimetics of Formula (I), one of the preferred embodiments of the invention, may be made using procedures which are conventional in the art. Preferably ~ is 2-phenyl-4-Am;nohenzoic acid although constrained derivatives such as tetrahydroisoquinoline-7-carboxylic acid, 2-aminomethyl pyridine-6-carboxylic acid or other heterocyclic derivatives, may also be used.
Compounds in which ~ is an aminomethylbenzoic acid (particularly 3-aminomethylbenzoic acid) are disclosed in U.S. Patent application No.
08/062,287, which is hereby incorporated herein by reference. The acid component of ~ is conveniently reacted with cysteine so that the amino group of ~ and the cysteine carboxyl group react to form an amido group, other reactive substituents in the reactants being suitably protected against undesired reaction. In the case of the reduced-cysteine series of compounds, the amino group of ~ is reacted with a suitably protected cysteinal. The amino acid represented by X, preferably Met, is then reacted through its amino group with the deprotected and activated carboxyl group of spacer compound ~. Following deprotection by removal of other protecting groups, the compound of Formula (I) is obtained.
As an alternative, ~ may first be reacted with the X amino acid followed by reaction with the cysteine or cysteinal component using conventional reaction conditions.
The invention also includes the pharmaceutically acceptable salts of the compounds of Formula (I). These may be obtained by reacting the free base or acid with the appropriate amount CA 022072~2 1997-06-06 W O96/21456 PCT~US96101559 of inorganic or organic acid or base, e.g. an alkali metal hydroxide or carbonate, such as sodium hydroxide, an organic amine, e.g.
trimethylamine or the like. Acid salts include the reaction products obtained with, for example, toluene sulfonic acid, acetic acid, propionic acid or the like as conventionally used in the art.
The compounds of the invention may be used to inhibit p21ras farnesyltransferase in any host cont~;n;ng the same. This includes both in vitro and in vivo use. Because the compounds inhibit farnesyltransferase, notably human tumor p2lras farnesyltransferase, and consequently inhibit the farnesylation of the oncogene protein ras, they may be used in the treatment of cancer or cancer cells. It is noted that many human cancers have activated ras and, as typical of such cancers, there may be mentioned colorectal carcinoma, myeloid leukemias, exocrine pancreatic carcinoma and the like.
The compounds of the invention may be used in pharmaceutical compositions of conventional form suitable for oral, subcutaneous, intravenous, intraperitoneal or intramuscular administration to a m~mm~l or host. This inciudes, for example, tablets or capsules, sterile solutions or suspensions comprising one or more compounds of the invention with a pharmaceutically acceptable carrier and with or without other additives.
Typical carriers for tablet or capsule use include, for example, lactose or corn starch. For oral compositions, aqueous suspensions may be used with conventional suspending agents, flavoring agents and the like.
The amount of inhibitor administered to obtain the desired inhibitory effect will vary but can be readily determined. For human use, daily CA 022072~2 1997-06-06 W O96/21456 PCT~US96/01~59 dosages are dependent on the circumstances, e.g.
age and weight. However, daily dosages of from 0.1 to 20 mg per kg body weight may be mentioned for purposes of illustration.
The various aspects of the invention are further described by reference to the following examples. These examples illustrate, among other things, the preparation of the present peptidomimetics and compounds compared therewith.

In the invention, the ~ component is, in general, any non-peptide amino acid combination or other hydrophobic spacer element that produces a compound which mimics the structure and conformation of CVIM or like tetrapeptides CAlA2X.
A variety of hydrophobic spacers have been used as the ~ component according to this aspect of the invention. This includes, for example, 3-aminobenzoic acid, 4-aminobenzoic acid and 5-aminopentanoic acid as well as heterocyclic carboxylic acids such as tetrahydroiso-quinoline-7-carboxylic acid, 2-aminomethyl pyridine-6-carboxylic acid or the like as mentioned earlier, as replacements for the ~ component of the Formula (I) compounds. Thus, in a broad sense, the peptidomimetics of the invention include variants for Formula (I) where ~ stands for the radical of a non-peptide aminoalkyl or amino-substituted aliphatic or aromatic carboxylic acid or a heterocyclic monocarboxylic acid, for example, 3-aminobenzoic acid (3-ABA), 4-aminobenzoic acid (4-ABA) or 5-aminopentanoic acid (5-APA).
Other suitable ~ substituents which may be mentioned include those obtained by using aminomethyl- or aminocarboxylic acid derivatives of other cyclic hydrophobic compounds such as furan, quinoline, pyrrole, oxazole, imidazole, CA 022072~2 1997-06-06 WO 96121456 PCT~S96101559 pyridine and thiazole. Generally speaking, therefore, the ~ substituent may be derived from any hydrophobic, non-peptidic aminoalkyl- or amino-substituted aliphatic, aromatic or ~ 5 heterocyclic monocarboxylic acid.
According to still another feature of this ~ embodiment of the invention, other effective inhibitors for farnesyltransferase may be provided by incorporating a negatively charged residue onto the compounds of Formula (I). This feature of the invention is based on a consideration of the transition state of the farnesylation reaction and the recognition that the functional enzyme complex must involve a farnesyl pyrophosphate binding site close to the peptide binding region. Compounds representative of this embodiment include peptides prepared with a phosphonate residue linked at different distances to the cysteine sulfur. These derivatives have been prepared by reaction of N-Cbz-cysteine with ethyl 2-chloroethylphosphonate followed by condensation with the C-terminal methionine adduct of 4-aminobenzoic acid (or N-deprotected VIM methyl ester). Deprotection of the phosphonate, carboxylate and amino protecting groups gives analogs (5) and (6), respectively, which contain elements of the tetrapeptide and farnesyl pyrophosphate residues and hence are able to interact with binding groups in both recognition sites in p21ras farnesyltransferase:

O ~p~

3+ ~ ~ ~~ O~

CA 022072~2 1997-06-06 W O 96/21456 PCTrUS96/01559 o ~ '~~

~ H O ~ H O (6) H3N~N~N~ .~o-~ ~ H O
S~

The above described phosphonates as contemplated herein can be structurally represented as follows:

~1-C-~-X

where C, X, ~ and ~ are as previously described and ~l is a phosphonate group joined to cysteine through the cysteine sulphur atom.
As indicated earlier, an important further feature of the invention is the modification of the compounds of the invention, as well as the tetrapeptide p21ras farnesyl transferase inhibitors of the formula CA1A2X, to provide pro-drugs. This involves forming lipophilic enzyme-sensitive derivatives from the compounds byappropriately functionalizing the terminal groups.
For example, the terminal amino groups and the cysteine sulfur can be reacted with benzyl chloroformate to provide carbobenzyloxy ester end groups while the terminal carboxy group at the other end of the compound is converted to an alkyl or aryl ester, e.g. the methyl ester. Other CA 022072~2 1997-06-06 WO96121456 PCT~S96/01559 examples include alkyl esters from l to lO carbons in length, activated esters such as cyanomethyl or trifluoromethyl, cholesterol, cholate or carbohydrate derivatives. The term "lipophilic'~, when used in this context, is meant to include, inter alia, methoxycarbonyl and other long chain or carbamate groups. Examples of such groups are well known to the ordinarily skilled practitioner.
Derivatization of the prior peptides CAlA2X
and the peptidomimetics described herein with lipophilic or hydrophobic, enzyme-sensitive moieties increases the plasma membrane permeability and cellular uptake of the compounds and consequently their efficiency in inhibiting tumor cell growth.
While the carbobenzyloxy derivatives have been referred to as one way o~ functionalizing the peptides and peptidomimetics to improve efficiency, it will be appreciated that a variety of other groups may also be used for the purposes noted. Typical alternatives include cholesterolyl, aryl or aralkyl such as benzyl, phenylethyl, phenylpropyl or naphthyl, or alkyl, typically methyl or other alkyl of, for example, up to 8 carbon atoms or more. It is contemplated that such functional groups would be attached to the cysteine sulfur and the terminal amino and carboxy groups.

CA 022072~2 1997-06-06 PCTrUS96101559 Using C-ABA-M as representative of the present compounds, the functionalized pro-drug embodiment of the invention may be structurally illustrated as follows:

S
O O BBM-CAB

N ~ ~ O

o In the above described BBM-compounds, the "BBM" used in the formulas represents a shorthand reference to the bis-(carboxybenzyloxy)methyl esters of C~M and CVIM.
The functionalized derivatives of the phosphonates described earlier herein are also useful cell growth inhibitors. Correspondingly, the "BMMM" designation used with compounds refers to the carboxy benzyloxy substitution and the three methyl groups in the methylated phosphoric and carboxylic acid end groups.
As noted, the purpose of the functional groups added to the parent compounds is to improve entry of the compounds into tumor cells. Once in the cells, the functional groups are removed to liberate the active compound ~o function in its inhibitory capacity.
As will be recognized by those in the art, the functionalized pro-drugs of the invention can be prepared using conventional and well-known procedures for esterifying amino, SH and CA 022072~2 1997-06-06 W O96/214S6 PCTrUS96/01559 carboxylic acid ~roups. Hence, detail~ of such procedures are not essential for the preparation of the present pro-drugs.

~YNl~SIS OF FTI-232 A. N-BOC-4-~minobenzoic acid 4-amino-benzoic acid (lOg, 72.9 mmol) was placed into a mixture of dioxane (145.8 ml) and 0.5M NaOH (145.8 ml). The solution was cooled to 0~C and di-t-butyl dicarbonate (23.87 g, 109.5 mmol) was added. The reaction mixture was allowed to warm to room temperature and stirred overnight.
The next day, the dioxane was removed, the residue was made acidic and extracted into ethyl acetate.
The ethyl acetate fractions were combined and washed with lN HCl to remove any unreacted starting material. The solution was dried over Na2SO4and the sol~ent was removed in vacuo. The crude material was recrystallized from ethyl acetate/hexanes to yield 12.2 g (70.6~) of pure product. mp 189-190~C; lH NMR (CD30D) 1.52 (9H, s), 7.49 (2H, d, J=8.6 Hz), 7.91 (2H, d, J=8.6 Hz), 9.28 (lH, s); 13C NMR (CD30D) 28.59, 81.29, 118.54, 125.30, 131.81, 145.70, 155.00, 169.80;
anal. calc. for Cl2Hl5NO4, C: 60.76, H: 6.37, N:
5.90; found, C: 60.52, H: 6.43, N: 5.83; HRMS
calc. for C12Hl5NO4, 237.0961, found, 237.1001.

B. N-BOC-4-~m;nohenzoyl methionine methyl e~ter Into a dried, nitrogen filled flask was placed N-BOC-4-~m;nohenzoic acid (8.77 g, 36.97 mmol) in dry CH2Cl2 (148 ml) along with methionine methyl ester hydrochloride (8.12 g, 40.66 mmol).
This solution was cooled in an ice bath and triethylamine (6.7 ml), EDCI (7.80 g, 40.66 mmol) and hydroxybenzotriazole (HOBT, 5.50 g, 40.66 CA 022072~2 1997-06-06 W O 96/21456 PCTrUS96/01559 mmol) were added. The mixture was stirred overnight, diluted with more CH2Cl2 and was extracted 3 times each with lM HCl, lM NaHCO3 and water. The CH2C12 was dried over MgSO4and the solvent was removed in vacuo. The solid was recrystallized from ethyl acetate/ hexanes to yield 9.72 g (71.3~) of pure product. mp 184-185~C; lH NMR (CDC13) 1.53 (9H, s), 2.06-2.18 (4H, m), 2.23-2.33 (lH, m), 2.59 (2H, t, J=7.6 Hz), 3.80 (3H, s)~ 4.92 (lH, m), 7.45 (2H, d, J=8.7 Hz), 7.77 (2H, d, J=8.7 Hz); 13C NMR (CDCl3) 15.59, 28.34, 30.15, 31.64, 52.10, 52.73, 81.20, 117.73, 127.8, 128.33, 141.88, 152.33, 166.50, 172.75;
anal. calc. for Cl8H26N2OsS, C: 56.53, H: 6.85, N:
7.29; found, C: 56.47, H: 6.86, N: 7.29; m/ z (EI) 382 (M).

C. HCl-4-~m;nohenzoyl methionine methyl ester N-BOC-4-aminobenzoyl methionine methyl ester (3.53 g, 9.59 mmol) was placed into CH2Cl2 (30-35 ml) and to it was added 3M HCl/ Bt2O (38.4 ml).
After standing a white precipitate formed. After 2 hours the solution was decanted, and the crystals were collected by centrifugation. The crystals were then washed several times with fresh ether and dried overnight on the vacuum pump.
Meanwhile, the filtrate was left to stand overnight to allow additional product to precipitate. The second fraction was washed with ether and dried overnight on the vacuum pump. The total yield of pure fully deprotected material was 2.87 g (93.9~) yield. mp 158-164~C; lH NMR (CDCl3) 2.10 (3H, s), 2.12-2.29 (lH, m), 2.52-2.71 (lH, m), 2.59 (2H, t, J=7.6 Hz), 3.75 (3H, s), 4.79 (lH, m), 7.02 (2H, d, J=8.6 Hz), 7.55 (2H, d, J=8.6 Hz); 13C NMR (CDC13) 15.23, 31.43, 31.53, 52.91, 52.43, 124.35, 130.56, 135.31, 135.76, CA 022072~2 1997-06-06 W O 96/21456 PCTrUS96/01559 168.95, 173.87; HRMS calc. for Cl3H18N2O3S, 282.1038, found 282.1009.

D. N-BOC-S-trityl-cysteine-4-~m;n~henzoyl methionine methyl ester N-BOC-S-tri~yl-Cys (2.86 g, 6.54 mmol) and triethylamine (1.2 ml) were placed into a dried, N2 filled flask containing dry THF (104 ml). This was cooled to -10~C using an ice/ salt bath and isobutyl chloroformate (0.9 ml), IBCF, was added.
The solution was stirred at -10~C for 40 minutes and HCl-4-aminobenzoyl methionine methyl ester (2.08 g, 6.54 mmol) in dry CH2Cl2 (34.1 ml) with triethylamine (1.2 ml, 1.3 eq) was added. The solution warmed to room temperature and was stirred overnight under N2. The solvent was then removed in vacuo and the residue was taken up in CH2Cl2 and extracted several times each with lM
HCl, H2O and brine (saturated NaCl). The organic layer was dried over Na2SO4 and the solvent was removed in vacuo. The pale yellow foam was then chromatographed on silica gel using a 2:1 hexanes, ethyl acetate elution mixture to yield 2.62 g (54.9~) of pure product. mp 110-111~C; [~]25D=-8.0~ (c=1, CH30H); lH NMR (CDCl3) 1.44 (9H,s), 2.11-2.18 (4H, m), 2.22-2.34 (lH,m), 2.59 (2H, t, J=7.4 Hz), 2.66-2.83 ( 2H, m), 3.80 (3H, s), 3.98 (lH, m), 4.84 (lH, m), 4.92 (lH, m), 6.96 (lH, d, J=7.7 Hz), 7.23-7.33 (9H, m), 7.43-7.46 (6H, m), 7.51 (2H, d, J=8.5 Hz), 7.74 (2H, d, J=8.5 Hz), 8.51 (lH, s); 13C NMR (CDCl3) 15.53, 28.34, 30.72, 30.89, 33.60, 52.23, 52.88, 54.95, 60.50, 67.13, 80.64, 118.81, 119.31, 126.94, 128.07, 128.30, 129.53, 141.06, 144.38, 156.31, 167.02, 170.13, 174.49; anal calc for C40H44N3O6S2-H2O, C: 64.50, H:
6.22, N: 5.64; found C: 64.14 H: 6.19, N: 5.56.

CA 022072~2 1997-06-06 W096/21456 PCT~S96101559 E. HCl-cysteine-4-~inobenzoyl methionine methyl ester N-BOC-S-trityl-cysteine-4-~m;nohenzoyl methionine methyl ester (1 g, 1.37 mmol) was placed into a flask and taken up in CH30H (13.7 ml). To this solution was added a solution of mercuric chloride (0.75 g, 2.74 mmol) in CH30H
(13.7 ml). Upon addition of the mercuric chloride, a white precipitate began to form. The mixture was heated on a steam bath at 65~C for 35 minutes and then it was cooled and the precipitate was filtered and washed sparingly with cold CH30H.
After drying for several minutes on the filter, the solid was placed into a 50 ml 3-neck flask fitted with a gas inlet and outlet. Approximately 20-30 ml of CH30H was added and H2S gas was bubbled through the heterogeneous solution for 30 minutes.
Upon addition of the gas, the white solution turned orange and then black. The solution was centrifuged and the clear, colorless liquid was dried to give a white foam. This solid was placed on the vacuum pump for a short period and then was taken up in CH2Cl2 (10 ml) and the product was precipitated with a 3-4M HCl/ Et20 solution. The precipitate was collected by centrifugation and was washed with ether until pH was neutral. After drying under vacuum overnight, 0.38 g (66.5~) of product was obtained that was >95~ pure by HPLC.
mp foamed 141-143~C, decomp 195~C; [~]25D=+30 (c=l, H20); lH NMR (CD30D) 2.09 (3H, s), 2.14-2.26 (lH,m), 2.51-2.67 (3H, m), 3.05 (lH, dd, J=14.8 Hz, 7.3 Hz), 3.17 (lH, dd, J=14.8 Hz, 4.8 Hz), 3.74 (3H,s), 4.17 (lH, J=7.3 Hz, 4.8 Hz ), 4.75-4.81 (lH, m), 7.74 (2H, d, J=8.6 Hz), 7.87 (2H, d, J=8.6 Hz), 8.67 (lH, d, J=8.4 Hz); 13C NMR (CD30D) 15.23, 26.38, 31.43, 31.56, 52.88, 53.30, 56.92, 120.46, 129.58, 130.75, 142.33, 166.91, 169.66, CA 022072~2 l997-06-06 W O 96/21456 PCTnUS96/01559 174.06; anal calc for Cl6H24ClN3O4S2, C: 45.55 H:
5.73, N: 9.96; found C: 45.31, H: 5.84, N: 9.79.

F. HCl-cysteine-4-~m;nnhenzoyl methionine FTI-232 HCl-cysteine-4-aminobenzoyl methionine methyl ester (0.51 g, 0.7 mmol) was taken up in THF (4.1 ml) and to this solution was added 0.5 M LioH (2.9 ml) at 0~C. The heterogeneous solution was stirred at 0~C for 35-40 minutes and then the THF
was removed in vacuo. The residue was taken up in CH2Cl2 and was washed three times with lM HCl followed by brine. The organic solution was dried over Na2SO4 and the solvent was removed in ~acuo.
The pale yellow solid was taken up in 3 ml of CH2Cl2 and the product was precipitated with 3-4 M
HCl/ Et2O. The solid was collected by centri~ugation, washed several times with ether until the ether washings were neutral and the process repeated until the HPLC appeared pure. A
final yield of 78.6 mg (27.5~) of pure product was obtained. mp sub 157~C, decomp 211~C; [~]25D=+10~
(c=0.8, H2O); 1H NMR (CD30D) 2.09 (3H, s), 2.17-2.32 (lH,m), 2.53-2.66 (3H, m), 3.06 (lH, dd, J=14.6 Hz, 7.2 Hz), 3.19 (lH, dd, J=14.6 Hz, 4.6 Hz), 4.21 (lH, dd, J=7.23 Hz, 4.63 Hz), 4.73-4.78 (lH, m), 7.75 (2H, d, J=8.1 Hz), 7.87 (2H, d, J=8.1 Hz); 13C NMR (CD30D) 15.23, 26.33, 31.58, 31.86, 53.24, 56.98, 120.48, 129.59, 131.10, 142.26, 166.89, 169.66, 175.29; anal calc for C15H22ClN3O4S2, C: 44.16, H: 5.44, N: 10.30; found C:
45.45, H: 5.62, N: 10.03; m/ z (FAB) for free amine, 371 (M+ 1).

CA 022072~2 1997-06-06 W O 96/21456 PCTrUS96/01559 ~iYNl~;SIS OE FTI-260 A. N-BOC-4-amino-3-methylbenzoic acid 4-amino-3-methylbenzoic acid (S g, 33.1 mmol) was reacted according to the same procedure as N-BOC-4-aminobenzoic acid. The orange-brown solid was recrystallized from ethyl acetate and h~x~nes to yield 4.99 g (60~) of tan prismatic crystals.
mp 180-182~C; lH NMR (CD30D) 1.51 (9H, s), 2.27 (3H, s), 7.66 (lH, d, J=8.1 Hz), 7.79-7.82 (2H, m), 8.32 (lH, s); 13C NMR (CD30D) 17.98, 28.62, 81.47, 123.12, 127.05, 129.14, 130.65, 132.99, 142.45, 155.33, 168.70; anal calc for Cl3Hl7NO4, C:
62.15, H: 6.82, N: 5.58; found C: 62.07, H: 6.86, N: 5.46; m/ z (EI) 251; HRMS calc. for Cl3Hl7NO4, 251.1158; found, 251.1153.

B. N-BOC-4-amino-3-methylbenzoyl methionine methyl ester N-BOC-4-amino-3-methylbenzoic acid (2.00 g, 7.96 mmol) was reacted with methionine methyl ester hydrochloride (1.75 g, 8.76 mmol), EDCI
(1.68 g, 8.76 mmol), HOBT (1.18 g, 8.76 mmol) and Et3N (1.4 ml) in dry CH2Cl2 (31.8 ml) according to the procedure described ~or N-BOC-4-aminobenzoyl methionine methyl ester in Example 1. The crude material was recrystallized from ethyl acetate and hexanes to yield 2.61 g (85.7~) of pure product.
mp 163-165~C; lH NMR (CDCl3) 1.54 (9H,s), 2.06-2.18 (4H, m), 2.23-2.34 (4H, m), 2.59 (2H, t, J=6.8 Hz), 3.80 (3H, s), 4.92 (lH, m), 6.45 (lH, s), 6.88 (lH, d, J=7.5 Hz), 7.63 (lH, d, J=8.6 Hz), 7.66 (lH, s), 8.05 (lH, d, J=8.6); 13C NMR (CDCl3) 15.47, 17.61, 28.22, 30.03, 31.55, 51.93, 52.57, 81.04, 118.73, 125.62, 127.66, 129.54, 139.89, 152.34, 166.58, 172.66.

CA 022072~2 1997-06-06 W O96/21456 PCTrUS96101559 C. HCl-4-~mino-3-methylbenzoyl methionine methyl ester N-BOC-4-amino-3-methylbenzoyl methionine methyl ester (0.99 g, 2.59 mmol) was dissolved in CH2Cl2 (15-20 ml) and precipitated with 3M HCl/
Et2O (20.7 ml). 0.83 g (96.6~) of pale orange precipitate was obtained after drying overnight on the vacuum pump. mp 157-159~C; lH NMR (CD30D) 2.04 (3H,s), 2.11-2.25 (lH, m), 2.47 (3H, s), 2.52-2.68 (3H. m), 3.74 (3~, s), 4.75-4.80 (lH, m), 7.48 (lH, d, J=8.2 Hz), 7.81 (2H, d, J=8.2 Hz), 7.87 (lH, s); 13C NMR (CD30D) 15.23, 17.28, 31.43, 31.51, 52.91, 53.37, 124.41, 127.85, 131.99, 133.63, 134.14, 135.65, 169.05, 173.84; anal.
calc. for Cl~H2lN2O3S, C: 50.52, H: 6.36, N: 8.42;
found C: 50.71, H: 6.40, N: 8.34.

D. N-BOC-S-trityl-cysteine-4-amino-3-methylbenzoyl methionine methyl ester N-BOC-S-trityl-cysteine (0.55 g, 1.25 mmol) in dry THF (25 ml) was reacted with Et3N (0.19 ml), IBCF (0.16 ml, 1.25 mmol) at -10 ~C as described above. HCl-4-amino-3-methylbenzoyl methionine methyl ester (0.42 g, 1.25 mmol) in dry CH2Cl2 (6.5 ml) with Et3N (0.26 ml) was added at -10~C and the reaction mixture was allowed to stir overnight under nitrogen. Workup was carried out as described above and the crude material was chromatographed on silica gel using a 2:1 mixture of hexanes and ethyl acetate as an elution mixture to give 0.12 g (13.9~) of pure product. mp 83-85~C; [~]25D=-14.0~ (c=l, CH30H); lH NMR (CDCl3) 1.44 (9H,s), 2.10-2.17 (4H, m), 2.22-2.32 (4H, m), 2.61 (2H, t, J=6.57 Hz), 2.68-2.70 (lH, m), 2.85-2.90 (lH. m), 3.79 (3H,s), 3.93-4.08 (lH, s), 4.84-4.88 ( lH, m), 4.90-4.95 (lH, m), 6.95 (lH, d, J=7.00 Hz), 7.20-7.33 (9H,m), 7.39 (lH, d, J=6.96 Hz), CA 022072~2 1997-06-06 W O96/21456 PCTrUS96/01559 7.44-7.47 (6H,m), 7.59 (lH, d, J=8.46 Hz), 7.65 ( lH, s), 8.12 ( lH,d, J=8.22 Hz), 8.31 (lH,s); 13C
NMR (CDCl3) 15.39 17.55, 27.70, 28.17, 30.00, 31.43, 31.41, 51.90, 52.51, 59.95, 67.30. 80.74, 84.54, 120.74, 125.33, 126.70, 126.83, 127.89, 128.00, 129.40, 138.92, 144.22, 166.50, 166.89, 168.87, 172.56.

E . TFA- cy~teine-4-~mino-3-methylbenzoyl methionine FTI-260 N-BOC-S-trityl-cysteine-4-amino-3-methylbenzoyl methionine methyl ester (0.27 g, 0.37 mmol) in THF (2.1 ml) was deprotected with 0.5M LiOH (2.9 ml) over 1.5 h at room temperature.
The solvent was removed in vacuo and the residue was taken up in CH2Cl2and extracted 3 times with lN HCl followed by extraction with brine. The organic solution was dried over Na2SO4 and the solvent was removed in vacuo to give 0.19 g (73.5 ~) of the free acid. The free acid was then taken up in CH2Cl2 (1.4 ml) and Et3SiH (0.04 ml) was added followed by trifluoroacetic acid, TFA (1.4 ml).
The reaction mixture was stirred at room temperature for 1 hour. The TFA was removed and the residue was dissolved in H2O and extracted with Et2O until all of the trityl derivative was removed. The water was lyophilized and a crude HPLC showed that the material was impure and contained diastereomers. The product was purified on the preparative HPLC using 0.1~ TFA in water and acetonitrile elution mixture to give 2 diastereomers and only the major component (determined according to the major compound in the HPLC trace) was characterized. mp sub 112~C, foamed 158-163~C, decomp 196-197~C; [~]25D=+12.7~
(c=0.6 H2O), [~]25D=+ 21.0~ (c=l H2O); lH NMR (CD30D) 2.09-2.17 (4H, m), 2.19-2.30 (lH, m), 2.36 (3H, CA 022072~2 1997-06-06 W O96/214S6 PCTrUS96/01559 s), 2.57-2.65 (2H, m), 3.08 (lH, dd, J=14.6 Hz, 6.9 Hz), 3.19 (lH, dd, J=14.6 Hz, 5.2 Hz), 4.25 ( lH, dd, J=6.9, 5.2 Hz), 4.70-4.75 (lH, m), 7.64 (lH, d, J=8.4 Hz), 7.69-7.73 (lH, m), 7.77 (lH, s); 13C NMR (CD30D) 15.23, 18.28, 26.54, 31.58, 32.06, 53.53, 56.66, 125.54, 125.77, 126.74, 131.04, 133.24, 139.26, 167.53, 169.70, 175.59.

A. N-BOC-4-amino-3-methoxybenzoic acid 4-amino-3-methoxybenzoic acid (1 g, 5.98 mmol) was reacted with di-t-butyl dicarbonate (1.96 g, 6.58 mmol) in dioxane (12 ml) and 0.5 M
NaOH (12 ml) according to the same procedure as N-BOC-4-aminobenzoic acid. 1.50 g (93.7~) o~ tan crystals were obtained after recrystallization from ethyl acetate and hexanes. mp 176-178~C; lH
NMR (CD30D) 1.52 (9H, s), 3.92 (3H, s), 7.56 (lH, s), 7.62 (lH, d, J=8.4 Hz), 7.96 (lH, s), 8.03 (lH, d, J=8.4 Hz); 13C NMR (CD30D) 28.53, 56.35, 81.78, 112.01, 118.58, 124.20, 125.76, 133.84, 149.04, 154.20, 169.60; HRMS calc. for Cl3Hl7NO5, 267.1107; found, 267.1103.

B. N-BOC-4-amino-3-methoxybenzoyl methionine methyl ester N-BOC-4-amino-3-methoxybenzoic acid (0.35 g, 1.31 mmol) was reacted with methionine methyl ester hydrochloride (0.9 g, 1.43 mmol) using EDCI
as in N-BOC-4-aminobenzoyl methionine methyl ester. After recrystallization from ethyl acetate and hexanes, 0.36 g (57.2 ~) of pure product was obtained. mp 163-165~C; lH NMR (CDCl3) 1.53 (9H, s), 2.09-2.18 ~4H, m), 2.23-2.35 (lH, m), 2.60 (2H, t, J=6.9 Hz), 3.80 (3H, s), 3.93 (3H, s), 4.92 (lH, br s), 6.93 (lH, d, J=7.6 Hz), 7.25 (lH, CA 022072~2 1997-06-06 WO96/214S6 PCT~S96/01559 m), 7.31 (lH, d, J=10.2 Hz), 7.44(lH, s), 8.15(lH, d, J=8.5 Hz); 13C NMR (CDCl3) 15.47, 28.23, 30.09, 31.48, 52.06, 52.54, 55.81, 80.82, 98.06, 109.38, 116.66, 119.31, 131.52, 147.23, 152.31, 166.57, 172.58; m/ z (FAB) 413 (M+1).

C. HCl-4-amino-3-methoxybenzoyl methionine methyl ester N-BOC-4-amino-3-methoxybenzoyl methionine methyl ester (0.71 g, 1.79 mmol) was taken up in CH2Cl2 (4 ml) and precipitated with 3-4M HCl/ Et2O
(12 ml). The precipitate was washed as usual with Et2O and dried overnight under vacuum to result in 0.55 g (88.3~) of reddish material. mp 176-177~C;
lH NMR (CD30D) 2.08 (3H, s), 2.21 (2H, m), 2.61 (2H, m), 3.74 (3H, s), 4.02 (3H, s), 4.79 (lH, m), 7.50 (lH, d, J=8.2 Hz), 7.57 (lH, d, J=4.1 Hz) 7.67 (lH, s); 13C NMR (CD30D) 15.26, 31.34, 31.42, 52.95, 53.38, 57.12, 112.29, 121.43, 124.57, 124.77, 136.15, 153.67, 168.79, 173.81.

D. N-BOC-S-trityl-cysteine-4-amino-3-methoxybenzoyl methionine methyl ester N-BOC-S-trityl-cysteine (0.76 g, 1.74 mmol) in dry THF (27.5 ml) was reacted with Et3N (0.24 ml), IBCF (0.23 ml, 1.74 mmol) at -10~C as described above. HCl-4-amino-3-methoxybenzoyl methionine methyl ester (0.55 g, 1.58 mmol) in dry CH2Cl2 ( 8.7 ml) with Et3N (0.30 ml) was added to the mixture and was allowed to stir overnight under nitrogen. It was worked up as described for N-BOC-S-trityl-cysteine-4-aminobenzoyl methionine methyl ester in Example 1, and the crude material was chromatographed on silica gel using a 2:1 mixture of hexanes and ethyl acetate to give 0.18 g (15.2~) of pure product. lH NMR (CDCl3) 1.45 (9H, s), 2.05-2.33 (5H, m), 2.57-2.65 (2H, m), CA 022072~2 1997-06-06 WO96/21456 PCT~S96/01559 2.68-2.72 (lH, m), 2.75-2.96 (lH, m), 3.78 (3H, s), 3.84 (3H, s), 4.90-5.00 (lH, m), 5.03-5.18 (lH, m), 7.17-7.48 (17H, m), 8.30-8.38 (lH, m), 8.65 (lH, br s).

E. TFA Cysteine~4-amino-3-methoxybenzoyl methionine FTI-261 N-BOC-S-trityl-cysteine-4-amino-3-methoxybenzoyl methionine methyl ester (0.18 g, 0.24 mmol) was deprotected with LiOH at room temperature as described above to give the free acid. The free acid was then further deprotected in CH2Cl2 (1.2 ml) with Et3SiH (0.04 ml, 0.24 mmol) and TFA (1.2 ml). The product was worked up as described for HCl-cysteine-4-aminobenzoyl methionine in Example 1, and HPLC revealed that the product was impure. The crude material was then purified on the HPLC using 0.1~ TFA in water and acetonitrile as eluting solvents to result in two pure samples that were expected to be diastereomers. The major component (determined according to the major compound in the HPLC trace) was characterized as follows. mp sub 109~C, decomp 191-193~C; [~]25D=-30.oo (c=l, H2O), [~]2~D=+l9-0~ (c=l, H2O); lH NMR (CD30D) 2.10 (3H, s), 2.12-2.18 (lH, m), 2.20-2.32 (lH, m), 2.53-2.71 (2H, m), 3.00 (lH, dd, J=14.6, 7.5), 3.15 (lH, dd, J=14.58, 4.8), 4.77 (lH, dd, J=7.5, 4.8), 7.50 (lH, d, J=8.4 Hz), 7.56 (lH, s), 8.23 (lH, d, J=8.4 Hz); 13C NMR (CD30D) 15.20, 26.65, 31.60, 31.76, 53.27, 56.58, 56.76, 111.04, 121.08, 122.14, 130.85, 131.85, 150.88, 167.21, 169.61, 175.36; m/ z (FAB) for free amine, 402 (M+l).

CA 022072~2 1997-06-06 W O96/21456 PCTrUS96/015~9 A. 4-nitro-2-phenyltoluene 2-bromo-4-nitrotoluene (2.16 g, 10.00 mmol) and phenyl boric acid (1.46 g, 12.00 mmol) were dissolved into anhydrous DMF (25 ml) under nitrogen. To this mixture was added Pd(Ph3P) 4 (0.58 g, 5~). The mixture was heated at 100 ~C
overnight. The solution was poured onto lN HCl and extracted with Et2O. The crude material was chromatographed on silica gel using hexanes as an eluent. After recrystallization from ethanol, 1.23 g (57.6~) of pale orange needles were obtained. mp 69 - 71~C; lH NMR (CDCl3) 2.36 (3H, s), 7.29-7.40 (2H, m), 7.41-7.49 (5H,m), 8.07-8.10 (2H, m); 13C NMR (CDC13) 20.68, 121.96, 124.51, 127.78, 128.41, 128.83, 131.06, 139.44, 142.97, 143.48, 146.05; anal calc. for Cl3H11NO2, C:73.26, H:5.20, N:6.57; found, C:73.10, H:5.12, N:6.50; m/
z (EI) 213; HRMS calc. for Cl3Hl~NO2, 213.0790i found, 213.0793.

B. 4-nitro-2-phenylbenzoic acid 4-nitro-2-phenyltoluene (0.50 g, 2.34 mmol) was dissolved in water (4.6 ml) and pyridine (2.3 ml). The mixture was heated to re~lux and KMnO4 (1.85 g, 11.70 mmol) was added. The reaction mixture was heated overnight and the solution was filtered and washed several times with boiling water. The aqueous solution was made acidic and the product was extracted into ethyl acetate. The ethyl acetate was dried over Na2SO4 and the solvent removed in vacuo to result in 0.37 g (67.9~) of pure yellow product. mp 174-176~C; lH NMR (CD30D) 7.38-7.48 (SH, m), 7.96 (lH, d, J=8.5 Hz), 8.21 (lH, d, J=2.3 Hz), 8.28 (lH, dd, J=8.48, 2.37); 13C
NMR (CD30D) 122.95, 126.09, 129.27, 129.42, 129.49, CA 022072~2 1997-06-06 WO96/21456 PCT~S96101559 131.56, 139.26, 140.42, 144.41, 150.17, 170.52; m/
z (EI) 243 (M).

_ C. 4-nitro-2-phenylbenzoyl methionine methyl e~ter 4-nitro-2-phenylbenzoic acid (0.30 g, 1.23 mmol), methionine methyl ester hydrochloride salt (0.27 g, 1.35 mm~l), EDCI (0.26 g, 1.35 mmol), HOBT ( 0.18 g, 1.35 mmol) and Et3N (0.19 ml) in dry CH2Cl2 (4.9 ml) were reacted according to the above procedure and worked up as described for N-BOC-4-aminobenzoyl methionine methyl ester in Example 1.
After recrystallization from ethyl acetate and hexanes, 0.41 g (85.5~) of pure product was isolated. mp 98-101~C; lH NMR (CDCl3) 1.62-1.73 (lH, m), 1.79-1.88 (lH, m), 1.91 (3H, s), 1.99 (2H, t, J=7.2 Hz), 3.59 (3H, s), 4.53 (lH, m), 6.45 (lH, d, J=7.8 Hz), 7.33-7.40 (5H, m), 7.67 (lH, d, J=8.3 Hz), 8.07-8.12 (2H, m); 13C NMR
(CDCl3) 14.92, 29.11, 30.67, 51.51, 52.29, 121.86, 124.74, 128.27, 128.60, 128.69, 129.52, 137.50, 140.56, 141.02, 148.09, 167.23, 171.23i m/ z (FAB), 389 (M+l).

D. 4-amino-2-phenylbenzoyl methionine methyl ester 4-nitro-2-phenylbenzoyl methionine methyl ester (0.35 g, 0.90 mmol) was taken up in ethyl acetate (9.0 ml). To this mixture was added SnCl2-2H2O (1.02 g, 4.50 mmol) and the reaction was heated under nitrogen at reflux for lh. The mixture was poured onto ice, the solution was made basic using NaHCO3 and the product was extracted into ethyl acetate several times (7-8). The ethyl acetate fractions were combined washed with brine and dried over Na2SO4 and the solvent was removed in vacuo to give 0.24 g (73.4~) of yellow solid.

CA 022072~2 l997-06-06 W O96/21456 PCTrUS96/01559 lH NMR (CDCl3) 1.58-1.70 (lH, m), 1.80-1.92 (lH, m), 1.98 ~3H, s), 2.06 (2H, t, J=7.7 Hz), 3.62 (3H, s), 4.00 (2H, br s), 4.56-4.63 (lH, m), 5.84 (lH, d, J=7.7 Hz), 6.50 (lH, s), 6.61 (lH, d, J=8.4 Hz), 7.29-7.42 (5H, m), 7.58 (lH, d, J=8.3 Hz); 13C NMR (CDC13) 15.02, 29.25, 31.25, 51.57, 52.15, 113.27, 115.88, 123.52, 127.56, 128.37, 128.44, 130.92, 140.66, 141.44, 148.53, 168.58, 171.91.

E. N-BOC-S-trityl-cysteine-4-amino-2-phenylbenzoyl methionine methyl ester N-BOC-S-trityl-cysteine (0.31 g, 0.66 mmol) in dry THF (11 ml) was reacted with Et3N (0.10 ml), IBCF (0.09 ml, 0.73 mmol) at -10 ~C as described for N-BOC-S-trityl-cysteine-4-aminobenzoyl methionine methyl ester in Example 1. 4-amino-2-phenylbenzoyl methionine methyl ester (0.24g, 0.66 mmol) in dry CH2Cl2 (3.5 ml) was added and the mixture was allowed to stir overnight under nitrogen. It was worked up as described as further described for N-BOC-S-trityl-cysteine-4-aminobenzoyl methionine methyl ester in Example 1, and after drying the crude material was chromatographed on silica gel using a 2:1 mixture of hexanes and ethyl acetate to give 84.70 mg (16.0~) of pure product. mp 100-103~C; lH NMR
(CDCl3) 1.41 (9H,s), 1.61-1.78 (lH, m), 1.84-1.95 (lH, m), 2.00 (3H, s), 2.05 (2H, t, J=7.6 Hz), 2.63 (lH, dd, J=12.7 Hz, 6.9 Hz), 2.72 (lH, dd, J=12.7 Hz,~5.51 Hz), 3.64 (3H, s), 4.02 (lH, br s), 4.58-4.63 (lH, m), 4.90 (lH, d, J=7.4 Hz), 6.10 (lH, d, J=6.6 Hz), 7.18-7.30 (lOH, m), 7.37-7.44 (llH, m), 7.50 (lH, s), 7.58 (lH, d, J=8.2 Hz), 8.69 (lH, s); 13C NMR (CDCl3) 15.21, 28.20, 29.38, 31.24, 33.00, 51.77, 52.35, 54.15, 67.30, 80.85, 118.18, 120.86, 126.88, 127.90, 128.03, CA 022072~2 l997-06-06 WO 96/21456 PCTIUS96/015~9 128.56, 128.66, 129.44, 129.79, 130.14, 156.00, 168.52, 169.11, 171.85.

F. TFA Cysteine-4-amino-2-phenylbenzoyl methionine FTI-272 N-BOC-S-trityl-cysteine-4-amino-2-phenylbenzoyl methionine methyl ester (84.70 mg, 0.11 mmol) of was taken up in THF (0.62 ml) and to this was added 0.5 M LiOH (0.32 ml) at 0 ~C. The mixture was stirred at 0 ~C for 35 minutes. The solvent was removed in vacuo using a cold water bath on the rotovap. The residue was worked up as described for Hc]--cysteine-4-~minobenzoyl methionine in Example 1, and 60 mg of the free acid was obtained. This was then dissolved into CH2Cl2 (0.8 ml) and Et3SiH (0.01 ml) was added followed by TFA (0.8 ml). The reaction mixture was stirred at room temperature for 1 h and worked up as described for TFA-cysteine-4-amino-3-methylbenzoyl methionine in Example 2. After lyophilization, 0.0387 g (84.0~) was obtained.
HPLC revealed that no epimerization had occurred, however the material was purified on the HPLC to eliminate baseline impurities. mp 150-154~C;
[~]25D=+2l.5~ (c=0.7, H20/ CH30H); lH NMR (CD30D) 1.62-1.79 (lH, m), 2.00-2.10 (5H, m), 2.16-2.18 (lH, m), 3.03 (lH, dd, J=14.7 Hz, 7.3 Hz), 3.15 (lH, dd, J=14.7 Hz, 4.8 Hz), 4.46 (lH, br s), 7.37-7.41 (5H, m), 7.52- 7.55 (lH, m), 7.65-7.67 (2H, m); 13C NMR (CD30D) 15.03, 26.35, 31.78, 32.79, 57.01, 119.40, 122.35, 128.95, 129.62, 129.71, 130.15, 133.49, 140.50, 141.36, 142.53, 167.05, 167.76, 172.51; anal. calc. for H26F3N3O6S2, C: 49-20, H: 4.67, N: 7.48; found, C:
49.14 H: 4.71, N: 7.42.

CA 022072~2 1997-06-06 W O96/21456 PCTrUS96/015S9 HCl cysteine-4-amino-2-phenylbenzoyl methionine methyl ester FTI274 N-BOC-S-trityl-cysteine-4-amino-2-phenylbenzoyl methionine methyl ester (0.06 g, 0.075 mmol) was dissolved into methanol (2 ml) and to it was added HgCl2 (0.04 g) in methanol (1 ml).
The reaction was carried out as described above to yield 15.7 mg of slightly impure compound by HPLC.
mp 130-132~C; lH NMR (CD30D) 1.72-1.84 (lH, m), 1.90-2.24 (6H, m), 3.05 (lH, dd, J=14.6 Hz, 8.5 Hz), 3.19 (lH, dd, J=14.6 Hz, 3.6 Hz), 3.69 (3H, s), 4.22 (lH, dd, J=.5 Hz, 3.6 Hz), 4.48-4.53 (lH, m), 7.33-7.43 (5H, m), 7.51 (lH, d, J=8.9 Hz), 7.70-7.72 (2H, m); 13C NMR (CD30D) 15.04, 26.36, 30.88, 31.36, 52.85, 53.05, 56.93, 119.42, 122.38, 128.88, 129.55, 129.73, 130.05, 133.17, 140.55, 141.32, 142.52, 166.92, 172.61, 173.58; anal.
calc. for C24H29ClN3O6S2-2H2O, C: 51.20, H: 5.86, N:
8.14; found, C: 51.23 H: 5.60, N: 8.22.

A. 2-bromo-4-nitrobenzoic acid 2-bromo-4-nitrotoluene (5.00 g, 23.14 mmol) was dissolved into pyridine ( 23 ml) and water (46 ml). The heterogeneous mixture was heated to 60 ~C and KMnO4 (18.29 g, 115.7 mmol) was added carefully. The mixture was then heated under reflux overnight. The reaction mixture was filtered and washed with boiling water. The solution was then made acidic and extracted into ethyl acetate, dried over Na2SO4, and the solvent was removed in vacuo. A crude NMR revealed remaining starting material so the solid was taken up in NaOH and washed with hexanes. The aqueous phase was made acidic and the product was CA 022072~2 1997-06-06 W O96/21456 PCTrUS96/01559 extracted into ethyl acetate. The ethyl acetate fractions were combined and dried over Na2SO4 and the solvent was removed in vacuo to yield 3.72 g (65.4~). mp 158-160~C; lH NMR (CD30D) 7.81 (lH, d, J=8.5 Hz), 8.08 (lH, d, J=8.5 Hz), 8.30 (lH, s);
3C NMR (CD30D) 121.96, 122.75, 129.36, 132.24, 139.52, 149.54, 167.75; anal. calc. for C7H4BrNO4 +
0.1 ethyl acetate, C: 34.88, H: 1.90, N: 5.50;
found, C: 34.68, H: 1.86, N: 5.82.

B. 3,5-dimethylphenyl boronic acid Mg turnings (1.44 g, 59.43 mmol) were covered with dry THF (18.8 ml) in a dried, N2 filled flask fitted with an addition funnel and reflux condenser. To this was added 5-bromo-m-xylene (10 g, 54.03 mmol) in THF (15 ml) after initiation of the Grignard reaction. The addition was carried out over several minutes and the reaction mixture was heated at reflux for 1-2 h until most of the Mg had reacted. The reaction mixture was then cooled and transferred to an addition funnel fitted to a N2 fi:Lled flask containing triisopropyl borate (24.9 ml) at -70 ~C. The dropwise addition was carried out over several minutes and the mixture warmed to room temperature and stirred overnight. The grey solution was poured onto 2 M
HCl and immediately turned yellow. The solution was extracted into Et2O and the Et2O fractions were combined, dried over MgSO4 and the solvent was removed in vacuo to yield 2.41 g (29.7~). mp 249-251~C; lH NMR (CDCl3) 2.44 (6H, s), 7.23 (lH, s), 7.84 (2H,'s); 13C NMR (CD30D) 21.36, 133.28, 134.39, 137.48.

C. 4-nitro-2-(3,5-dimethylphenyl)benzoic acid 2-bromo-4-nitrobenzoic acid (0.50 g, 2.03 mmol) and 3,5-dimethylphenyl boronic acid (0.34 g, CA 022072~2 1997-06-06 W O96/21456 PCTrUS96/015~9 2.23 mmol) were dissolved into anhydrous DMF
(dimethylformamide) (25 ml) under nitrogen. To this mixture was added Cs2CO3 (1.66 g, 5.08 mmol) followed by Pd(Ph3P) 4 (O .12 g, 5~). The mixture was heated at 100~C overnight. The solution was poured onto lN HCl and extracted into Et2O. It was dried over MgSO~ and the solvent was removed in vacuo. The crude material was chromatographed on silica gel using a 9:1 mixture of hexanes and ethyl acetate to yield 0.34 g (61.7~) of pure product. lH NMR (CDC13) 2.36 (6H, s), 6.99 (2H, s), 7.07 (lH, s), 8.03 (lH, d, J=9.0 Hz), 8.23-8.25 (2H, m); 13C NMR (CDCl3) 21.28, 121.68, 123.68, 125.74, 126.07, 130.22, 131.19, 131.31, 135.04, 138.21, 144.74, 170.75.

D. 4-nitro-2-(3,5-dimethylphenyl)benzoyl methionine methyl ester 4-nitro-2-(3,5-dimethylphenyl)benzoic acid (0.15 g, 0.55 mmol), methionine methyl ester hydrochloride salt (0.11 g, 0.55 mmol), EDCI
(0.11 g, 0.55 mmol), HOBT (0.07 g, 0.55 mmol) and Et3N (0.08 ml) in dry CH2Cl2 (2.2 ml) were reacted and worked up according to the procedure for N-BOC-4-aminobenzoyl methionine methyl ester in Example 1. After recrystallization from ethyl acetate and hexanes, 0.13 g (58.4~) of pure product was isolated. mp 122-124~C; lH NMR (CDCl3) 1.2-1.84 (lH, m), 1.85-1.97 (lH, m), 2.01 (3H, s), 2.05 (3H, t, J=7.7 Hz), 2.38 (6H, s), 3.70 (3H, s), 4.67-4.74 (lH, m), 6.03 (lH, d, J=7.9 Hz), 7.05 (2H, s), 7.09 (lH, s), 7.84-7.87 (lH, m), 7.84-7.87 (lH, m), 8.23-8.26 (2H, m); 13C NMR
(CDC13), 15.20, 21.26, 29.22, 31.15, 51.79, 52.57, 122.07, 25.11, 126.27, 130.03, 130.53, 137.77, 138.82, 140.29, 141.56, 148.41, 167.14, 171.53.

-CA 022072~2 1997-06-06 WO96/21456 PCT~S96101559 E. 4-amino-2-(3~5-dimethylphenyl)benzoyl methionine methyl ester 4-nitro-2-(3,5-dimethylphenyl)benzoyl methionine methyl ester (0.11 g, 0.26 mmol) was taken up in ethyl acetate (3.0 ml). To this mixture was added SnCl2 2H2O (0.30 g, 1.30 mmol) and the reaction was heated under nitrogen at reflux for 6h. The mixture was worked up as described for 4-amino-2-phenylbenzoyl methionine methyl ester in Example 2, to give 0.15 g of a yellow film that was wet with solvent. The material was otherwise pure by NMR and was used without further purification. lH NMR (CDCl3) 1.60-1.70 (lH, m), 1.80-1.90 (lH, m), 1.99 (3H, s), 2.05 (2H, t, J=7.6 Hz), 2.33 (6H, s), 3.64 (3H, s), 3.93 (2H, br s), 4.61-4.64 (lH, m), 5.82 (lH, d, J=7.7 Hz), 6.49 (lH, d, J=2.3 Hz), 6.62 (lH, dd, J=8.4 Hz, 2.4 Hz), 6.98 (2H, s), 7.00 (lH, s), 7.65 (lH, d, J=8.3 Hz); 13C NMR (CDCl3) 15.08, 21.17, 29.28, 31.49, 51.70, 52.18, 113.30, 115.94, 123.55, 126.36, 129.32, 131.23, 138.15, 140.72, 141.92, 148.40, 168.45, 172.01.

F. N-BOC-S-trityl-cysteine-4-amino-2-(3,5-dimethylphenyl)benzoyl methionine methyl ester 4-amino-2-(3,5-dimethylphenyl)benzoyl methionine methyl ester (0.lOg, 0.26 mmol) was dissolved into dry CH2C12 (1.4 ml) and it was allowed to stand. In another flask, N-BOC-S-trityl-Cys (0.12 g, 0.26 mmol) was dissolved into THF (4.4 ml) and was reacted with IBCF (0.03 ml) and Et3N (0.04 ml) as described above. The product was worked up as described for N-BOC-S-trityl-cysteine-4-aminobenzoyl methionine methyl ester in Example 1 and chromatographed on silica gel using a 1:1 hexanes and ethyl acetate elution mixture to give 0.12 g (56.0~) of pure material. lH NMR

CA 022072~2 1997-06-06 W O96/21456 PCTrUS96/01559 (CDCl3) 1.33 (9H, s), 1.61-1.68 (lH, m), 1.73-1.91 (4H, m), 1.96 (2H, t, J=7.6 Hz), 2.24 (6H, s), 2.57- 2.64 (2H, m), 3.57 (3H, s), 4.00 (lH, br s), 4.54-4.58 (lH, m), 5.84 (lH, d, J=7.8 Hz), 5.97 (lH, br d), 6.90 (lH, s), 6.92 (2H, s), 7.18-7.22 (9H, m), 7.27-7.40 (7H, m), 7.55 (lH, m), 7.61 (lH, m), 8.58 (lH, br s); 13C NMR (CDCl3) 15.11, 21.20, 27.79, 29.25, 31.28, 51.70, 52.28, 54.08, 60.32, 71.45, 80.75, 118.01, 120.80, 126.38, 126.82, 127.98, 129.41, 129.87, 130.22, 138.11, 139.18, 139.79, 141.06, 144.17, 168.38, 169.04, 171.82.

G. TFA Cysteine-4-amino-2-(3,5-dimethylphenyl)benzoyl methionine FTI275 N-BOC-S-trityl-cysteine-4-amino-2-(3,5-dimethylphenyl)benzoyl methionine methyl ester (0.12 g, 0.15 mmol) was placed into THF (0.9 ml) and was reacted with 0.5 M of LiOH (0.6 ml) at 0 ~C
as described above, followed by deprotection with TFA (1.5 ml) and Et3SiH (0.24 ml). Addition of excess scavenger does not appear to affect the result. The product was purified by preparative HPLC to give 23.8 mg (27.3~). mp 135-138~C; lH NMR
(CDCl3) 1.76-1.84 (lH, m), 2.00-2.17 (6H, m), 2.33 (6H, s), 3.05 (lH, dd, J=14.6 Hz, 7.3 Hz), 3.17 (lH, dd, J=14.6 Hz, J=4.9 Hz), 4.15 (lH, dd, J=7.3, 4.9 Hz), 4.45-4.48 (lH, m), 7.02 (3H, s), 7.53 (lH, d, J=8.0 Hz), 7.66 (2H, m); 13C (CD30D) 14.96, 21.51, 26.28, 30.91, 31.70, 53.03, 56.98, 119.27, 122.30, 127.52, 130.07, 130.57, 133.37, 139.28, 140.39, 141.29, 142.86, 166.89, 172.60, 174.81.

CA 022072~2 1997-06-06 WO96/21456 PCT~S96101559 ~Y~ SIS OF FTI-266 A. 4-amino-1-naphthoic acid 4-amino-l-naphthalenecarbonitrile (1.50 g, 8.91 mmol) was dissolved into a 50~ KOH solution (18 ml). The heterogeneous solution was heated at reflux for 2-3 days. Once the ~olutior~ becarl,e homogenous and TLC showed no more starting material, the deep red solution was cooled and poured over 200 ml of water. The solution was then filtered and the acid was precipitated with concentrated HCl. The red crystals were filtered and the filtrate was refiltered to give pink crystals. The first fraction was treated with activated carbon to remove some of the red color.
1.51 g (90.6~) of product was obtained. mp 169-171~C; lH NMR (CD30D) 6.69 (lH, d, J=8.2 Hz), 7.38-7.43 (lH, m), 7.48-7.54 (lH, m), 8.03 (lH, d, J=8.5 Hz), 8.13 (lH, d, J=8.2 Hz), 9.09 (lH, d, J=8.5 Hz); 13C NMR (CD30D) 107.39, 114.61, 122.99, 123.92, 125.21, 127.40, 128.48, 135.04, 151.35, 171.44; HRMS calc. for CllH7NO2, 187.0633; found, 187.0642.

B. N-BOC-4-amino-1-naphthoic acid 4-amino-1-naphthoic acid (0.86 g, 4.61 mmol) was dissolved into dioxane (9.2 ml) and 0.5 M NaOH
(9.2 ml). Di-t-butyl dicarbonate (1.11 g, 5.07 mmol) was added and the mixture was stirred overnight. The reaction mixture was worked up as described for N-BOC-4-aminobenzoic acid in Example 1 to give 0.76 g (56.7~) of reddish pink solid.
mp 194-195~C; lH NMR (CD30D) 1.56 (9H, s), 7.53-7.62 (2H, m), 7.79 (lH, d, J=8.1 Hz), 8.12 (lH, d, J=8.0 Hz), 8.22 ~lH, d, J=8.18 Hz), 9.02 (lH, d, J=8.9 Hz); 13C NMR (CD30D), 26.68, 81.62, 119.06, 123.40, 124.57, 127.03, 127.37, 128.49, 128.77, CA 022072~2 1997-06-06 W O96/21456 PCTrUS96/01559 131.89, 133.76, 139.86, 155.95, 170.73; anal.
calc. for Cl7Hl7NO4, C: 66.90, H: 5.96, N: 4.88;
found C: 66.49, H: 6.08, N: 4.79; m/ z (BI), 289;
HRMS calc. for Cl6Hl7NO4, 287.1158; found, 287.1151.

C. N-BOC-4-amino-1-naphthoyl methionine methyl ester N-BOC-4-amino-1-naphthoic acid (0.46 g, 1.60 mmol), methionine methyl ester hydrochloride (0.35 g, 1.76 mmol), EDCI (0.43 g, 1.76 mmol), HOBT (0.24 g, 1.76 mmol) and Et3N ( 0.27 ml) in CH2Cl2 (6.4 ml) were reacted as described for N-BOC-4-aminobenzoyl methioniné methyl ester in Example 1. After workup and recrystallization from ethyl acetate and hexanes, 0.44 g (63.6~) of pale pink crystals were obtained. mp 131-132~C; lH
NMR (CDC13) 1.57 (9H, s), 2.11-2.21 (4H, m), 2.29-2.41 (lH, m), 2.65 (2H, t, J=7.1 Hz), 3.83 (3H, s), 4.99-5.06 (lH, m), 6.68 (lH, d, J=8.0), 7.02 (lH, s), 7.56-7.59 (2H, m), 7.69 (lH, d, J=7.9 Hz), 7.87-7.90 (lH, m), 8.02 (lH, d, J=7.9 Hz), 8.44-8.48 (lH, m); l3C NMR (CDCl3) 15.56, 28.31, 30.19, 31.65, 52.06, 52.64, 81.17, 115.82, 120.18, 125.79, 126.37, 126.53, 127.18, 131.02, 135.65, 152.93, 169.04, 172.40; HRMS calc. for C22H28N2O5S, 432.1719; found 432.1702; m/ z (FA~3) 433 (M+l).

D. HCl 4-amino-1-naphthoyl methionine methyl ester N-BOC-4-amino-1-naphthoyl methionine methyl ester (0.57 g, 1.31 mmol) was deprotected with HCl/ ether to yield 0.31 g (64.1~) of white solid.
mp 178-181~C; lH NMR (CD30D) 2.08-2.16 (4H, m), 2.20-2.30 (lH, m), 2.57-2.75 (2H, m), 3.82 (3H, s), 4.87-4.91 (lH, m), 7.59 (lH, d, J=7.5 Hz), 7.67 (lH, d, J=7.5 Hz), 7.71-7.80 (2H, m), 8.03 (lH, dd, J=7.1 Hz, 2.0 Hz), 8.35 (lH, dd, J=6.8 Hz, 1.8 Hz); 13C NMR (CD30D) 15.23, 31.40, 53.01, CA 022072~2 1997-06-06 WO96/21456 PCT~S96101559 53.33, 119.90, 122.20, 126.15, 127.41, 127.77, 129.09, 129.31, 131.50, 132.33, 135.64, 171.77, 173.83; m/ z (FAB), 369 (M+l).

E. N-BOC-S-trityl-cysteine-4-amino-1-naphthoyl methionine methy~ ester N-BOC-S-trityl-Cys (0.31 g, 0.67 mmol) in dry THF (11.2 ml) was reacted with Et3N (0.10 ml) and IBCF (0.10 ml, 0~74 mmol) at -10 ~C as described above. HCl-4-amino-1-naphthoyl methionine methyl ester (0.25 g, 0.67 mmol) in dry CH2Cl2 (3.5 ml) was added and the mixture was stirred overnight under nitrogen. The mixture was worked up as described for N-BOC-S-trityl-cysteine-4-aminobenzoyl methionine methyl ester in Example 1, and the crude material was chromatographed on silica gel using a 2:1 mixture of hexanes and ethyl acetate to give 0.20g (37.5 ~) of pure product. lH NMR (CDCl3) 1.48 (9H, s), 2.10-2.20 (4H, m), 2.30-2.37 (lH, m), 2.63 (2H, t, J=7.4), 2.74 (lH, J=12.9 Hz, J=5.3 Hz), 2.90 (lH, J=12.9 Hz, 6.2 Hz), 3.81 (3H, s), 4.96-5.03 (2H, m), 6.77 (lH, d, J=8.0 Hz), 7.18-7.33 (llH, m), 7.44-7.56 (7H, m), 7.60 (lH, d, J=7.7 Hz), 7.88 (lH, d, J=8.0 Hz), 8.00 (lH, d, J=7.1 Hz), 8.37 (lH, d, J=8.4 Hz), 8.94 (lH, br s); 13C NMR (CDCl3) 15.23, 26.52, 31.41, 31.50, 52.98, 53.31, 56.79, 68.15, 122.52, 123.54, 126.16, 126.99, 128.03, 128.39, 129.52, 132.30, 134.04, 135.24, 168.08, 172.38, 173.94.

F. TFA cysteine-4-amino-1-naphthoyl methionine, N-BOC-S-trityl-cysteine-4-amino-1-naphthoyl methionine methyl ester (83.3 mg, 0.11 mmol) was taken up in THF (0.7 ml) and to this mixture was added 0.5 M LioH (0.43 ml) at 0~C. The mixture CA 022072~2 1997-06-06 W 096/21456 PCTrUS96/OlS59 was stirred at 0~C for 35 minutes. The solvent was removed in vacuo using a cold water bath. The residue was worked up as described for TFA-cysteine-4-amino-3-methylbenzoyl methionine in Example 2, and 74.1 mg of the free acid was obtained. This was then dissolved into CH2C12 (1 ml) and Et3SiH (0.015 ml) was added followed by TFA
(1 ml). The reaction mixture was stirred at room temperature for 1 h and worked up as further described for TFA-cysteine-4-amino-3-methylbenzoyl methionine in Example 2. After lyophilization, 42.4 mg of crude material was obtained which was then purified on the HPLC using 0.1~ TFA in water and acetonitrile. mp 121-125~C; [~]25D=+2.4O
(c=0.8, H2O); lH NMR (CD30D) 2.03-2.13 (4H, m), 2.22-2.36 (lH, m), 2.59-2.74 (2H, m), 3.16-3.33 (2H, m), 4.42 (lH, m), 4.84-4.89 ~lH, m), 7.57-7.61 (2H, m), 7.64 (lH, d, J=7.7 Hz), 7.70 (lH, d, J=7.7 Hz), 8.08-8.11 (lH, m), 8.29-8.32 (lH, m), 8.98 (lH, d, J=7.7 Hz); 13C NMR (CD30D) 15.19, 26.45, 31.50, 31.63, 53.20, 56.72, 122.~2, 123.43, 126.43, 126.12, 127.02, 127.96, 128.32, 129.49, 132.27, 134.15, 135.12, 168.11, 172.41, 175.17;
anal. calc. for C2lH23F3N3O6S2, C: 47.19, H: 4.34, N:
7.86; found, C: 46.53, H: 4.56, N: 7.59; Note:
difference for C is 0.65.

G. HCl cysteine-4-amino-1-naphthoyl methionine methyl ester FTI-270 HCl TFA cysteine-4-amino-1-naphthoyl methionine (0.12 g, 0.15 mmol) was dissolved in CH30H (4.3 ml). To this solution was added a solution of HgCl2 (0.23 g, 0.86 mmol) in CH30H (4.3 ml). The procedure was continued as described above and after HCl/ Et2O precipitation and several reprecipitations 31.0 mg (18.3 ~) of pure white material was obtained. mp sub 137~C, decomp 214-CA 022072~2 1997-06-06 WO96/21456 PCT~S96/01559 215~C; [~]25D=-32.oo (c=1 CH30H); lH NMR (CD30D) 2.12 (3H, s), 2.21-2.28 (lH, ~), 2.57-2.73 (3H, m), 3.20-3.34 (2H, m), 3.82 (3H, s), 4.39-4.43 (lH, m), 7.61-7.68 (3H, m), 7.78 (lH, d, J=7.7 Hz), 8.13-8.16 (lH, m), 8.28-8.32 (lH, m); 13C (CD30D) 15.23, 26.52, 31.41, 31.50, 52.98, 53.31, 56.79, 122.52, 123.54, 126.16, 126.99, 128.03, 128.39, 129.52, 132.30, 134.04, 135.24, 168.08, 172.38, 173.94.

~YNl~SIS OF FTI-254 A. N-Boc-S-trityl cy teinal Triethylamine (2.22 mL, 16 mmoL) and N,O-dimethylhydroxylamine hydrochloride (1.57 g, 16.1 mmol) were added to a solution of N-Boc-S-trityl cysteine (7.44 g, 16 mmol) in 85 mL of methylene chloride. This mixture was cooled in an ice bath and 1-(3-dimethy].aminopropyl)-3-ethylcarbodiimide hydrochloride (EDCI, 3.08 g, 16.0 mmol) and HOBT
(2.17 g, 16 mmol) was added. The mixture was stirred at 0~C for 1 hr and at room temperature for a further 10 hr. The mixture was extracted with methylene chloride and 0.5 N HCl. The organic layer was washed consecutively with 0.5 N
HCl, concentrated NaHCO3 and brine. The organic layer was dried and evaporated. The residue was purified by flash column chromatography (1.5 : 1 =
hexane : ethylacetate) to give a white foam (7.40 g, 91~). m.p. 59-60 ~C (decomp). lH NMR (CDCl3) ~
7.41 (m, 6H), 7.20-7.31 (m, 9H), 5.13 (d, 8.9 Hz, lH), 4.76 (br s, lH), 3.64 (s, 3H), 3.15 (s, 3H), 2.56 (dd, 4.7 and 12.1 Hz, lH), 2.39 (dd, 7.8 and 12.1 Hz, lH), 1.43 (s, 9H). 13C NMR (CDCl3) ~
170.7, 154.9, 144.2, 129.3, 127.6, 126.4, 79.3, 66.4, 61.2, 49.5, 33.8, 31.8, 28.1. This carboxyamide (2.02 g, 4.0 mmol) was dissolved in CA 022072~2 1997-06-06 30 mL of ether and cooled to -10 ~C. Lithium alllm;nllm hydride ~167 mg, 4.40 mmol) was added and the mixture was stirred for 15 min under the nitrogen. Then 40 mL of 0.5 N HCl was added and the solution was extracted with ether. The ether layer was washed with 0. 5 N HCl and dried. The evaporation of solvents gave a white foam (1. 80 g) which was used for further reaction without purification. The lH NMR spectrum of this compound was complex. The percentage of the aldehyde was about 65 - 70 ~, which was calculated according to the integration of the sharp singlet (~ 9.17) and the trityl peak (~ 7. 40, m, 6H; 7.28, m, 9H) .
Lowering the temperature to -45~C did not improve the aldehyde percentage.

B . 4 -N- ~2 (R) - tert-Butoxycarbonyl ~m; no_ 3 _ triphenylmethylthi~L~yl~m;n~henzoyl methionine methyl ester.
One equivalent of N-Boc-S-trityl cysteinal in 10 mL of methanol was added to a solution of 4-aminobenzoyl methionine methyl ester hydrochloride (1.7836 g, 5.6 mmol) in 60 mL of methanol and 4 mL
of glacial acetic acid. Sodium cyanoboronhydride (0.528 g, 8.40 mmol) was added to this deep colored solution at 0 ~C. The mixture was stirred at room temperature for 15 hr. After the evaporation of solvents, the residue was extracted with ethyl acetate and concentrated sodium bicarbonate. The organic phase was dried and the solvents were evaporated. The residue was purified through flash column chromatography (ethyl acetate / hexane = 1:1) to give a pure desired product (2.52 g, 65~ H NMR (CDCl3) ~ 7.63 (d, 8.6 Hz ~
2H), 7.43 (m, 6H), 7.21-7.32 (m, 9H), 6.73 (d, 7.6 Hz~ lH~ Met amide), 6.50 (d, 8.6 Hz~ 2H) ~ 4.91 (ddd, 5.1 Hz~ 5.3 Hz and 7.6 Hz~ lH~ Met ~ H) -- . ' CA 022072~2 1997-06-06 WO96/21456 PCT~S96101559 4.59 (d, 8.9 Hz, lH, Boc amide), 4.25-4.40 (br, lH, NHPh), 3.80 (m, lH, Cys ~ H), 3.78 (s, 3H, OCH3), 3.09 (d, 6.3 Hz, 2H, CH2NH), 2.55-2.60 (m, 2H, CH2SCPh3), 2.96 (d, 5.0 Hz, 2H, CH2SCH3), 2.23-2.28 (m, lH, Met CH2), 2.07-2.12 (m, lH, Met CH2), 2.09 (s, 3H, SCH3~, 1.43 (s, 9H, Boc).
;~
C. 4-N-t2(R)-Amino-3-mercaptopropyl]aminobenzoyl methionine methyl e~ter.
The fully protected 4-N-[2(R)-tert-Butoxycarbonylamino-3 triphenylmethylthiopropyl]
amino-benzoyl methionine methyl ester (1.31 g, 1.83 mmol) was dissolved into 20 mL of methanol.
To this solution wa~ added mercuric chloride (1.09 g, 4.04 mmol) in 5 mL of methanol. The mixture was refluxed for 20 min and then cooled down. The clear solution was removed and the solid precipitate was washed with 5 mL of methanol. The solid was dried and then suspended in 15 mL of methanol. The suspension was stirred and reacted with hydrogen sulfide gas for 1 hr. The black precipitate was removed by centrifugation. The clear solution was evaporated to dryness. The residue was dissolved in 6 mL of methylene chloride followed by the addition of 20 mL of 3N
HCl in ether. The white precipitate was filtered and dried to give a hydrochloride salt of the desired product (0.60 g, 73~). lH NMR (CD30D) ~
7.73 (d, 8.8 Hz, 2H), 6.75 (d, 8.8 Hz, 2H), 4.74 (dd, 4.9 Hz and 4.3 Hz, lH, Met ~ H), 3.72 (s, 3H, OCH3), 3.43-3.59 (m, 3H, CH2NH and Cys ~ H), 2.93 (dd, 3.9 Hz and 14.4 Hz, lH, CH2SH), 2.81 (dd, 5.2 Hz and 14.5 Hz, lH, CH2SH), 2.49-2.66 (m, 2H, CH2SCH3), 2.07-2.20 (m, 2H, Met CH2), 2.10 S, 3H, sCH3).

CA 022072~2 1997-06-06 W O 96/21456 PCTrUS96/01559 D. 4-N-[2(R)-Amino-3-mercaptopropyl] ~m;nnhenzoyl methionine The fully protected peptide 4-N-[2 (R)-tert-Butoxycarbonylamino- 3- triphenylmethylthiopropyl]-aminobenzoyl methionine methyl ester (567 mg, 0.79 mmol) was dissolved into 3.0 mL of 0. 5 N lithium hydroxide and 3.0 mL of tetrahydrofuran. The mixture was stirred at 0~C for 1 hr. After the evaporation of solvents, the residue was dissolved in water and extracted with methylene chloride and lN hydrochloric acid. The organic phase was dried and the solvents were evaporated. The residue was dissolved in a mixture of lmL of methylene chloride and 2 mL of trifluoroacetic acid.
Triethylsilane was added dropwise until the deep brown color disappeared. The mixture was kept at rt for 1 hr. The solvents were evaporated and the residue was dried. This residue was dissolved in 1 mL of 1.7N HCl in acetic acid followed by the addition of 20 mL of 3N HCl in ether. The white precipitate was filtered and dried to give a hydrochloride salt of the desired product (159 mg, 46~). Analytical HPLC showed purity over 98~. lH
NMR (CD30D) ~ 7.74 (d, 8.7 Hz, 2H), 6.75 (d, 8.7 Hz, 2H), 4.73 (dd, 4.5 Hz and 4.7 Hz, lH, Met ~
H), 3.45-3.58 (m, 3H, CH2NH and Cys ~ H), 2.93 (dd, 4.5 Hz and 14.6 Hz, 1 H, CH2SH), 2.80 (dd, 5.3 Hz and 14.6 Hz, lH, CH2SH), 2.53-2.64 (m, 2H, CH2SCH3), 2.15-2.23 (m, lH, Met CH2), 2.07-2.13 (m, lH, Met CH2), 2.10 (s, 3H, SCH3).

SYnthesis of FTI-284 A. 4-Nitro-2-phenylbenzoyl-~l'(S)-methoxycarbonyl-3'-methylsulfonyl]propyl amide 4 -nitro- 2 -phenylbenzoyl methionine methyl ester (525 mg, 1.28 mmol), N-methylmorpholine CA 022072~2 1997-06-06 WO96/21456 PCT~S96101559 oxide (453 mg, 3.87 mmol) and 0.5 mL of osmium tetroxide (2.5 wt.~ solution in tert-butanol) were added to a mixture of 40 mL of acetone and 10 mL
of water. The mixture was stirred at rt overnight. After the addition of excess sodium sulfite, the reaction mixture was extracted with ethyl acetate and washed with concentrated sodium bicarbonate. The organic phase was dried and the solvents were evaporated to give a solid (570 mg, 100~ H NMR (CDC13) ~ 8.29 (d, 7.7 Hz, lH), 8.25 (s, lH), 7.83 (d, 7.7 Hz, lH), 7.43-7.55 (m, 5H), 6.15 (d, 7.3 Hz, lH, Met amide), 4.68 (ddd, 5.0 Hz, 5.1 Hz and 7.3 Hz, lH, Met ~ H), 3.70 (s, 3H, OCH3), 2.85 (s, 3H, SCH3), 2.69-2.81 (m, lH, CH2SO2), 2.58-2.66 (m, lH, CH2SO2), 2.21-2.33 (m, lH, Met CH2), 1.96-2.08 (m, lH, Met CH2).

B. 4-N-~2(R)-tert-Butoxycarbonylamino-3-triphenylmethylthiopropyl~ino-2-phenylbenzoyl-tl'(S)-methoxycal~o~,~1-3'-methylsulfonyl]propyl amide The 4-Nitro-2-phenylbenzoyl-[1'(S)-methoxycarbonyl-3'=-methylsulfonyl]propyl amide (430 mg, 1.02 mmol) was dissolved in 20 mL of methanol. A catalytic amount of 5~ palladium on carbon was added and the mixture was hydrogenated at 40 PSI for 1.5 hr. The mixture was filtered and the filtrate was evaporated to dryness to give 4-amino product t400 mg, 100~). lH NMR (CD30D) ~ 7.70 (d, 8.0 Hz, lH), 7.38-7.47 (m, 7H), 4.53 (dd, 4.6 Hz and 4.8 Hz, lH, Met ~ H), 3.72 (s, 3H, OCH3), 2.89 (s, 3H, SO2CH3), 2.79-2.85 (m, lH, CH2SO2), 2.58-2.68 (m, lH, CH2SO2), 2.19-2.29 (m, lH, Met CH2), 1.93-2.04 (m, lH, Met CH2). This amine was dissolved in 15 mL of methanol and 0.8 mL of acetic acid. One equivalent of N-Boc-S-trityl cysteinal was added followed by the addition of sodium cyanoboronhydride (97 mg, 1.5 eq). The CA 022072~2 1997-06-06 W O96/21456 PCTrUS96/015S9 mixture was stirred at rt overnight. After the evaporation of solvents, the residue was extracted with ethyl acetate and concentrated sodium bicarbonate. The organic phase was dried and solvents were evaporated. The residue was purified through flash column chromatography (ethyl acetate / hexane / methanol = 15:15:2) to give a pure product (500 mg, 60~). lH NMR (CDCl3) 7.64 (d, 8.5 Hz, lH), 7.37-7.46 (m, llH), 7.18-7.33(m, 9H), 6.53 (d, 8.5 Hz, lH), 6.34 (s, lH), 5.74 (d, 7.5 Hz, lH, Met amide), 4.64 (ddd, 4.9 Hz, 5.1 Hz and 7.5 Hz, lH, Met ~ H), 4.55 (d, 7.5 Hz, lH, Boc amide), 4.26 (br, lH, NHPh), 3.79 (m, lH, Cys ~ H), 3.68 (s, 3H, OCH3), 3.10 (t, 5.7 Hz, 2H, CH2NHPh), 2.84 (s, 3H, S02CH3), 2.62-2.82 (m, 2H, CH2SO2), 2.45 (d, 2H, CH2SCPh3), 2.19-2.27 (m, lH, Met CH2), 1.84-1.95 (m, lH, Met CH2), 1.41 (s, 9H).

C. 4-N-[2(R)-Amino-3-mercaptopropyl]amino-2-phenylbenzoyl-[l'(S)-methoxycarbonyl-3'-methylsul~onul]propyl amide, FTI-284 The fully protected peptide 4-N-[2(R)-tert-Butoxycarbonylamino-3-triphenylmethyl-thiopropyl]amino-2-phenylbenzoyl-[l'(S)-methoxycarbonyl-3~-methylsulfonyl]propyl amide(277 mg, 0.33 mmol) was dissolved into 5 mL of methanol. To this solution was added mercuric chloride (229 mg, 2.50 eq) in 2 mL of methanol.
The mixture was refluxed for 20 min. The precipitate was dried and then suspended in 10 mL
of methanol. This mixture was reacted with hydrogen sulfide gas. The reaction mixture was centrifuged and the clear solution was evaporated.
The residue was dissolved in 2 mL of methylene chloride followed by the addition of 20 mL of 3N
HCl in ether. The white precipitate was collected CA 022072~2 1997-06-06 W Og6/21456 PCTrUS96/01559 and dried to give a hydrochloride salt of the desired product (165 mg, 89~). lH NMR (CD30D) ~
7.44 (d, 8.4 Hz, lH), 7.32-7.40 (m, 5H), 6.77 (d, 8.4 Xz, lH), 6.68 (s, lH), 4.45 (dd, 4.5 Hz and 4.7 Hz, lH, Met ~ H), 3.69 (s, 3H, OCH3), 3.40-3.57 (m, 3H, CH2NHPh and Cys ~ H), 2.78-2.96 (m, 3H, CH2SH and CH2SO2), 2.89 (S, 3H, SO2CH3), 2.60-2.69 (m, lH, CH2SO2), 2.15-2.24 (m, lH, Met CH2), 1.91-2.02 (m, lH, Met CH2).

SYnthesis of FTI-277 A. 4-N-~2(R)-tert-Butoxycarbonyl-3-triphenylmethylthiopropyl]amino-2-phenylbenzoyl methionine methyl ester The coupling o~ 4-amino-2-phenylbenzoyl methionine methyl ester (3.88 g, 10 mmol) with one equivalent of N-Boc-S-trityl cysteinal in the presence of 1.5 equivalent of sodium cyanoboronhydride gave a crude mixture which was purified through flash column chromatography (ethyl acetate / hexane = 1:1) to give a pure desired product (5.83 g, 74~). lH NMR (CDCl3) ~
7.65 (d, 8.5 Hz, lH), 7.32-7.45 (m, llH), 7.18-7.30 (m, 9H), 6.50 (d, 8.5 Hz, lH), 6.33 (s, lH), 5.65 (d, 7.6 Hz, lH, Met amide), 4.62 (ddd, 5.0 Hz, 5.2 Hz and 7.6 Hz, lH, Met ~ H), 4.54 (d, 8.1 Hz, Boc amide), 4.18 (br, lH, NHPh), 3.78 (m, lH, Cys ~ H), 3.64 (s, 3H, OCH3), 3.10 (t, 6.1 Hz, 2H, CH2NHPh), 2.45 (d, 5.0 Hz, 2H, CH2SCPh3), 2.04-2.10 (m, 2H, CH2SCH3), 2.00 (s, 3H, SCH3), 1.81-1.92 (m, lH, Met CH2), 1.61-1.70 (m, lH, Met CH2), 1.40 (s, 9H). 13C NMR (CDCl3) ~ 172.0, 168.3, 155.7, 149.4, 144.3, 141.6, 14101, 131.3, 129.5, 128.7, 128.5, 127.9, 127.7, 126.8, 122.6, 113.6, 111.3, 79.8, 67.1, 52.2, 51.7, 49.5, 47.2, 34.3, 31.6, 29.4, 28.3, 15.2.

CA 022072~2 1997-06-06 W O96/21456 PCTrUS96/01559 B. 4-N-~2(R)-~m;no-3-mercaptG~ opyl]amino-2-phenylbenzoyl methionine methyl ester, FTI-277.
The fully protected peptide 4-N-[2(R)-tert-Butoxycarbonyl-3-triphenylmethyl-thiopropyl]amino-2-phenylbenzoyl methionine methyl ester(1.57 g, 2.0 mmol) was first reacted with mercuric chloride (1.36 g, 5.0 mmol) and then reacted with hydrogen sulfide gas in methanol to give a hydrochloride salt of the desired product (0.808 g, 84~).
Analytical HPLC showed purity over 98~. [~]2~D = -12.1~ (c=0.008, CH30H). lH NMR (CD30D) ~ 7.42 (d, 8.3 Hz, lH), 7.30-7.38 (m, 5H), 6.78 (d, 8.3 Hz, lH), 6.71 (s, lH), 4.47 (dd, 4.2 Hz and 5.1 Hz, lH, Met ~ H), 3.68 (s, 3H, OCH3), 3.44-3,54 (m, 3H, CH2NHPh and Cys ~ H), 2.94 (dd, 4.1 Hz and 14.6 Hz, lH, CH2SH), 2.81 (dd, 5.0 Hz and 14.6 Hz, lH, CH2SH), 2.12-2.22 (m, lH, CH2SCH3), 2.03-2.10 (m, lH, CH2SCH3), 2.00 (s, 3H, SCH3), 1.90-1.97 (m, lH, Met CH2), 1.73-1.82 (m, lH, Met CH2). 13C NMR
(CD30D) ~ 173.7, 173.4, 150.7, 143.5, 142.3, 131.2, 129.8, 129.5, 128.6, 125.6, 115.6, 112.2, 53.7, 53.2, 52.8, 45.0, 31.3, 30.8, 25.3, 15Ø

SYnthesis of FTI-276 4-N-~2(R)-Amino-3-mercaptopropyl] amino-2-phenylbenzoyl methionine The fully protected peptide 4-N-[2(R)-tert-Butoxycarbonyl-3-triphenylmethylthiopropyl]-amino-2-phenylbenzoyl methionine methyl ester (2.36 g, 3 mmol) was first reacted with lithium hydroxide and then with trifluoroacetic acid to give a crude product (1.30 g, 77~ yield, 85~ purity shown by HPLC) which was further purified through preparative HPLC to give a pure product (0.98 g, 75~) [~]2~D = -13.6~ (c=0.005, CH30H). 1H NMR
(CD30D) ~ 7.44 (d, 8.4 Hz, lH), 7.30-7.41 (m, 5H), CA 022072~2 1997-06-06 WO96/21456 PCT~S96/01559 6.75 (d, 8.4 Hz, lH), 6.68 (s, lH), 4.43 (dd, 4.2 Hz and 5.1 Hz, lH, Met ~ H), 3.44-3.58 (m, 3H, CH2NHPh and Cys ~ H), 2.95 (dd, 4.4 Hz and 14.5 Hz, lH, CH2SH), 2.83 (dd, 5.0 Hz and 14.5 Hz, lH, CH2SH), 2.14-2.23 (m, lH, CH2SCH3), 2.05-2.11 (m, lH, CH2SCH3), 2.00 (s, 3H, SCH3), 1.91-1.99 (m, lH, Met CH2), 1.72-1.81 (m, lH, Met CH2). 13C NMR
(CD30D) ~ 176.4, 173.5, 150.4, 143.0, 141.5, 131.0, 129.7, 129.4, 128.9, 124.6, 115.0, 112.2, 53.3, 44.4, 30.8, 30.1, 24.9, 14.8.
Other compounds of the invention (in particular those of claims [[14-18) are synthesizable by modifications of the procedure described for the 2-phenyl-4-aminobenzoic acid derivative of claim 3. In particular, modifications of the Suzuki couping method will allow the incorporation of an alkoxy-, chloro, bromo or methyl substituted phenyl group onto the 4-aminobenzoic acid spacer. As with the unsubstituted derivative, 4-nitro-2-bromotoluene will be coupled with the corresponding substituted phenyl boronic acid derivative (alkoxyphenyl or chloro-, bromo- or methylphenylboronic acid) under paladium catalyzed conditions. The appropriately substituted 2-(substituted) phenyl-4-nitro toluene derivative will be incorporated into the peptidomimetic synthesis as described for the 2-phenyl case.
In a similar way the precursor to the 2-naphthyl-, 2-thiophene-, 2-pyrrole-, and 2-pyridyl-4-aminobenzoic acid spacers can be prepared by reaction of 4-nitro-2-bromotoluene with naphthalene-2-boronic acid, thiophene-2-boronic acid, pyrrole-2-boronic acid, pyridine-2,3- or 4-boronic acid.

.

CA 022072~2 1997-06-06 WO96/21456 PCT~S96/01559 FTase and GGTase I Activity Assay FTase and GGTase I activities from 60,000 X g supernatants of human Burkitt lymphoma (Daudi) cells (ATCC, Rockville, MD, USA) were assayed as described previously for FTase (41). Briefly, 100 ~g of the supernatant was incubated in 50 mM Tris, pH 7.5, 50 ~M ZnCl2, 20 mM KCl and 1 mM
dithiothreitol (DTT). The reaction was incubated 0 at 30~C for 30 min with recombinant Ha-Ras-CVLS (11 ~M) and [3H]FPP (625 nM; 16.3 Ci/mmol) for FTase, and recombinant Ha-Ras-CVLL (5 ~M) and [~H]geranylgeranylpyrophosphate (525 nM; 19.0 Ci/mmol) for GGTase I. The peptidomimetics were mixed with FTase and GGTase before adding to the reaction mixture.

Ras and RaPlA Processinq Assay H-RasF cells (45) were seeded on day 0 in 100 mm Dishes (costar) in Dulbecco's modified Eagles medium (GIBCO) and allowed to grow to 40-60~
confluency. On days 1 and 2, cells were fed with 4 ml of medium per plate plus various concentra-tions of FTI-277 or vehicle. On day 3, cells were washed one time with ice cold PBS and were collected and lysed by incubation for 30-60 min on ice in lysis buffer (41). Lysates were cleared (14,000 rpm, 4~C, 15 min) and supernatants collected. Equal amounts of lysate were separated on a 12.5~ SDS-PAGE, transfered to nitrocellulose, and a western blot performed using a anti-Ras antibody (Y13-238, ATCC) or anti-RaplA antibody (Santa Cruz Biotechnology, Santa Cruz, CA).
Antibody reactions were visualized using peroxidase-conjugated goat anti-rat IgG for Y13-238 and peroxidase-conjugated goat anti-rabbit IgG

CA 022072~2 1997-06-06 W O 96/21456 PCTnUS96101559 for RaplA and an enhanced chemiluminescence detection system (EC~; Amersham Corp.) Co-immunoPrecipitation of Raf and Ras Cells were seeded on day 0 in 100 mm dishes in 10 ml Dulbecco's Modified Eagles Medium (GIBCO) supplemented with 10~ calf serum (Hyclone) and 1~ pen/strep (GIBCO). On days 1 and 2 cells were treated with FTI-277 (5 ~M) or vehicle (confluency of cells 40-60~). On day 3, cells were collected by centrifugation in ice cold PBS.
Cell pellets were then resuspended in ice cold hypotonic buffer (10 mM Tris, pH 7.5, 5 mM MgCl2, 1 mM DTT, 1 mM PMSF) and cells were sonicated to break up cell pellet to promote separation of cytosol and membrane. The cell suspension was then centrifuged at 2,000 rpm for 10 min to clear debris after which the supernatant was loaded in ultrocentrifuge tubes and spun for 30 min at 100,000 X g to SW Ti55 Rotor to separate membrane and cytosol fractions. The cytosol and membrane fractions were lysed on ice for 60 min in buffer containing 30 mM HEPES, pH 7.5, 1~ TX-100, 10~
glycerol, 10 mM NaCl, 5 mM MgCl2, 2 mM Na3VO4, 25 mM NaF, 1 mM EGTA, 10 ~M soybean trypsin inhibitor, 25 ~g/ml leupeptin, 10 ~g/ml aprotinin, 2 mM PMSF). The ]ysates were clarified by centrifugation. Equal amounts of cytosol and membrane fractions were immunoprecipitated using 50 ~l of a 25~ Protein-A Sepharose Cl-4B
suspension (Sigma) with 1 ~f/ml anti-c-Raf-1 (SC133, Santa Cruz Biotechnology, Santa Cruz, CA).
The samples were tumbled at 4~C for 60 min and then washed 5 times in 50 mM HEPES, pH 7.5, 100 mM
NaCl, 5 mM MgCl2, 0.1~ TX-100, 10~ glycerol, 20 mM
NaF. The final pellets were run on 12.5~ SDS--CA 022072~2 1997-06-06 WO96/21456 PCT~S96/01559 PAGE, transferred to nitrocellulose, and immunoblotted for the presence of Ras using anti-Ras antibody (Y13-238) and immunoblotted for the presence of Raf (c-Raf-1, SC133, Santa Cruz Biotechnology, Santa Cruz, CA). Detection was the same as above for Ras and RaplA processing.

Detection of GTP and GDP bound to Ras (FTI-277) H-RasF cells were seeded and treated as above for Ras/Raf interaction and Ras and RaplA
processing. On day 2, howevér, cells were labeled overnight with [32p] orthophosphate at 100 ~LCi/mo (Amersham PBS13) in 10 ml DMEM-phosphate supplemented with 10~ calf serum, 1 mg/ml BSA and 20 mM HEPES, pH 7.5. On day 3, the medium was removed and cells were washed one time in ice-cold PBS, scraped from the plate with a cell scraper, collected and centrifuged. The cell pellet was resuspended in ice-cold hypotonic buffer listed above and the cytosol and membrane fractions were separated according to the above description for Ra/Raf association. The cytosol and membrane fractions were lysed on ice for 60 min in 50 mM
Tris, pH 7.5, 5 mM MgCl2, 1~ Triton X-100 (TX-100), 0.5~ DOC, 0.05~ SDS, 500 mM NaCl, 1 mM EGTA, 10 ~g/ml aprotinin, 10 ~g/ml soybean trypsin inhibitor, 25 ~g/ml leupeptin, 1 mM DTT, 1 mg/ml BSA. Lysates were cleared and equal amounts of protein were immunoprecipitated using anti-Ras antibody (Y13-259) along with 30 ~l Protein A-Agarose goat anti-rat IgG complex (Oncogene Science) for 60 min at 4~C. Immunoprecipitates were washed 6 times in 50 mM HEPES, pH 7.5, 0.5 M
NaCl, 0.1~ TX-100, 0.0005 SDS, 5 mM MgCl2, drained using a syringe and bound nucleotide eluted in 12 ~l of 5 mM DTT, 5 mM EDTA, 0.2~ SDS, 0.5 mM GDP

WO96/21456 PCT~S96101559 and 0.5 mM GTP at 68~C for 20 min. Immune complexes were spun down quickly and 6 ~l of the supernatent was loaded onto PEI cellulose thin layer chromatography plates (20 cm X 20 cm).
Nucleotides were separated by chromatography in 78 g/linter ammonium formate, 9.6~ (v/v) concentrated HCl. Plates were analyzed by autoradiogram.

Analysis of Raf-I Kinase ActivitY
Raf-1 kinase was assayed by determining the ability of Raf to transfer phosphate from [~y_32p]
ATP to a 19-mer peptide containing an autophosphorylation site. Membrane and cytosol fraction isolation and Raf immunoprecipitates were washed three time.s with cold HEPES buffer and twice with kinase buffer (50 mM Tris, pH 7.5, 150 mM NaCl, 12 mM MnCl2, 1 mM DTT, 0.2~ Tween 20.
Immune complex kinase assays were performed by incubating immunoprecipitaes from membrane and cytosol fractions in 96 ~l of k;n~e buffer with 20 ~Ci of [~-32P]ATP (10 mCi/ml, Amersham) and 2 ~l of the Raf-1 substrate peptide (1 mg/ml, Promega) for 30 min at 25~C. The sequence of the Raf-1 substrate peptide is IVQQFGFQRRASDDGKLTD.
The phosphorylation reaction was terminated by spotting 50 ~l ali.quots of the assay mixture onto Whatman P81 for 40 min in 0.5~ orthophosphoric acid and air dried. The amount of 32p incorporated was determined by liquid scintillation counting.

Inhibition of FTa~e bY FTI-276 and other comPounds ? Fig. lB shows that FTI-276 inhibited the transfer of farnesyl from [3H]FPP to recombinant H-Ras-CVLS with an IC50 of 500 pM. FTI-249, the parent compound of FTI-276, inhibited FTase with CA 022072~2 1997-06-06 W O96121456 PCTrUS96/01559 an IC50 of 200,000 pM. Thus, a phenyl ring at the 2 position of the benzoic acid spacer increased inhibition potency of FTase by 400 fold confirming our prediction of a significant hydrophobic pocket within the CAAX binding site of FTase. This extremely potent inhibitor was also highly selective (100-fold) for FTase over the closely related GGTase I (Fig. lB). FTI-276 inhibited the transfer of geranylgeranyl from [3H]GG-PP to recombinant H-Ras-CVll with an IC50 of 50 nM (Fig.
lB). This 100-fold selectivity is superior to the 15-fold selectivity of the parent compound, FTI-249. Data for a number of other compounds of interest are shown in Table 1.

CA 022072~2 1997-06-06 WO 96121456 PCT~S96/015~9 Table 1 FTa~e GGTase I ICsc C __ ~ ICso [nm][nm] GG/F
O FTI

260 3-Me-CABAM 8259000 11 261 3-OMe-CABAM 255050000 20 .
272 2-Ph-CABAM 5 267 53 274 2-Ph- ~3AM-OMe 205030000 lS
0 275 2-Xy-CP~3AM 405 400 249 red.CABAM 2723967 15 254 red.CABAM-OMe 100019000 19 276 red.2-Ph-CA~3AM 0.5 57 114 277 red.2-Ph C1~3AM-OMe 50 1600 32 Inhibition of Ras Processina by FTI-277 To facilitate cellular uptake, FTI-277, the methylester of FTI-276, was used in experiments to measure inhibition of Ras processing. H-RasF
cells (NIH 3T3 cells transformed with oncogenic (61 leucine) H-Ras-CVLS (45) were treated with FTI-277 (0-50 ~M) and the lysates blotted with anti-Ras or anti-RaplA antibodies. As shown in Fig. 2A, concentrations as low as 10 nN inhibited Ras processing but concentrations as high as 10 ~M
did not inhbit processing of the geranylgeranylated RaplA. FTI-277 inhibited Ras processing with an IC50 of 100 nM. In contrast, the ICso of FTI-249 is 100 ~M, and the most potent CAAX peptidomimetics previously reported inhibited CA 022072~2 1997-06-06 W 096/21456 PCTrUS96/OlSS9 Ras processing at concentrations of 10 ~M or higher (44).
The selectivity of FTI-277 for farnesylation but not geranylgeranylation processing is further demonstrated in Fig 2B. H-RasGG cells (NIH 3T3 cells transformed with oncogenic (61 leucine) H-Ras-CVLL (45) were treated with FTI-277.
Processing of RasGG was not affected, whereas that of RasF was completely blocked. The processing of endogenous Ras is also blocked in pZIPneo cells (NIH 3T3 cells transfected with the same plasmid as H-RasF and H Ras FF except the vector contained no oncogenic Ras sequences) and Raf cells (NIH 3T3 cells transformed by an activated viral Raf (48)).

Me~h~n;~m of disruPtion of Ras oncoqenic siqnallinq bY FTI-277 Ras relays biological information from tyrosine kinase receptors to the nucleus by activation of a cacade of MAPKs (reviewed in 29-31). Upon growth factor stimulation, Ras becomesGTP bound and is then able to recruit the ser/thr kinase c-Raf-l to the plasma membrane where it is activated. c-Raf-l then phosphorylates and activates MEK, a dual thr/tyr kinase, which activates MAPK. Recently, epidermal growth factor has been shown to induce association of Raf with Ras (46).
In order to determine the mechanism by which FTI-277 disrupts Ras oncogenic signaling, NIH 3T3 cells were transfected with activated (GTP-locked) Ras and the effects of FTI-277 on the interaction of Ras with its immediate effector, Raf, were investigated. Various NIH 3T3 cell transfectants (pZIPneo, H-RasF, and H-RasGG) were treated with vehicle or FTI-277, membrane and cytosolic fractions were isolated and immunoprecipitated CA 022072~2 1997-06-06 WO96121456 PCT~S96/01559 with anti-Raf antibody as described above. Raf did not associate with Ras in pZIPneo cells which did not contain GTP-locked Ras, as shown in Fig.
3. In contrast, H-RasF and H-RasGG cells contain Ras/Raf complexes in the membrane, but not in the cytosolic fractions, as shown in Fig. 3.
Treatment of these cells with FTI-277 resulted in the accumulation of Ras/Raf complexes in the cytosolic but not membrane fractions of H-RasF
cells, but not in the H-RasGG cells (Fig 3).
Thus, the disrupt.ion of Ras/Raf interaction at the cell membrane and accumulation of these complexes in the cytoplasm occurred only in Ras-F but not Ras-GG cells, in agreement with the Ras processing selectivity results of Fig. 2. Thus, these results demonstrate that inhibition with FTI-277 results in the accumulation of non-farnesylated cytosolic Ras that is capable of binding to Raf.
The fact that non-processed Ras can associate with Raf in a non-membranous cytoplasmic environment was confirmed by ~ransfecting NIH 3T3 cells with a GTP-locked Ras that lacks a farnesylated site and, therefore, r~m~; n~ in the cytoplasm (Ras mutant with a 61 leucine oncogenic mutation and a 186 serine mutation) and showing that these cells contained only cytoplasmic Ras/Raf complexes when immunoprecipitated with Raf and blotted with antiRas antibodies (Fig. 3). In short, farnesylation is not required for Ras to bind to Raf.

Det~m;n~tion of nucleotide state of Ras The fact that Raf binds Ras-GTP with much higher affinity than Ras-GDP was used to determine the nucleotide state of Ras in the cytoplasmic Ras/Raf complexes, as described above. In Ras-F

CA 022072~2 1997-06-06 WO96/21456 PCT~S96/01559 cells, only membrane fractions contained GTP-locked Ras, as shown in Fig 4A. Upon treatment with FTI-277, however, the non-farnesylated cytosolic Ras was found to be GTP bound. Thus, binding of GTP to 61 leucine Ras does not require Ras processing and subsequent plasma membrane association. The ser/thr kinase activity of Raf in Ras/Raf complexes was then determined by immunoprecipitating Raf and assaying for its ability to phosphorylate a l9-mer autophosphorylated peptide. Fig. 4B shows that oncogenic Ras-F induced activation of Raf in the plasma membrane and that treatment with FTI-277 suppressed this activation. More importantly, the cytoplasmic Ras/Raf complexes that were induced by FTI-277 (Fig. 3) had basal levels of Raf kinase activity that were comparable to those of the parental NIH 3T3 cell line pZIPneo (Fig. 4B).
Taken together, Figures 3 and 4 demonstrate that oncogenic transformation with GTP-locked Ras results in the constitutive recruitment to the plasma membrane and subsequent activation of Raf.
Furthermore, FTase inhibition by FTI-277 suppresses this activation by inducing the accumulation of Ras/Raf complexes in the cytoplasm where Ras is GTP-bound but Raf kinase is not activated. The fact that Raf kinase is not activated when bound to Ras in a non-membranous environment is consistent with recent reports that indicate that Raf activation requires an as yet to be determined activating factor at the plasma membrane (47).
Experiments were then performed to investigate the effects of FTI-277 on oncogenic Ras activation of MAPK, a Raf downstream signalling event (29-31). Oncogenic activation of MAPK can be easily detected since activated MAPK

-CA 022072~2 1997-06-06 WO96/21456 PCT~S96101559 migrates slower in SDS-PAGE. Fig. 5A shows that NIH 3T3 cells transfected with pZIPneo contain only inactive MAPK but that upon transformation with oncogenic H-Ras, MAPK is activated (Fig. 5A).
Pretreatment with FTI-277 results in a concentration dependent inhibition of oncogenic Ras activation of MAPK. Concentrations as low as 300 nM were effective and the block was complete at l ~M. Taken together, Figs. 3 and 5 demonstrate that at least 50~ inhibition of Ras processing is required for complete suppression of MAPK activation but that less than a lO0~
inhibition of Ras processing is required for complete suppression of MAPK activation by Ras. A
series of NIH 3T3 cell lines transformed with various oncogenes was used to determine whether the inhibition of MAPK activation is due to selectively antagonizing Ras function. Fig. 5B
shows that FTI-277 was able to block H-RasF but not H-RasGG activation of MAPK. This is consistent with its ability to inhibit H-RasF but not H-RasGG processing (Fig. 2). Selectivity of FTI-277 towards antagonizing Ras-dependent activation of MAPK was substantiated by using NIH
3T3 cells where MAPK is constitutively activated by transformation with the Raf oncogene. Fig.
5B shows that oncogenic Raf activation of MAPK is not blocked by FTI-277 even though processing of endogenous Ras was inhibited in these cells.
Similar results were also obtained with FTI-276 (Fig. 6). Taken together these results clearly demonstrate that FTI-276 and FTI-277 are highly effective and selective in disrupting contitutive Ras-specific activation of MAPK.
Thus, FTI-277 is an extremely potent and highly selective FTase inhibitor. This compound inhibited Ras processing with concentrations as CA 022072~2 1997-06-06 W O96/21456 PCTrUS96/015S9 low as 10 nM and processing was blocked at 1 ~M.
The most potent inhibitor previously reported BZA-5B, blocked Ras processing only at 150 ~M (44).

Antitumor Efficacy and SelectivitY of FTI-276 and FTI-277 In order to demonstrate the efficacy of these inhibitors as anticancer agents and show that they can inhibit tumor growth of human tumors which have multiple and complex genetic alterations, antitumor efficacy experiments were performed using a human tumor cell line. A
critical issue connected with the potential use of the compounds of the invention is whether the growth of hnm~n tumors which harbor K-Ras mutations can be blocked. This is important for further development of FTase inhibitors as anticancer drugs since K-Ras mutations are most common in human cancers and since K-Ras processing is more difficult to inhibit than the processing of the less prevalent H-Ras (1-3, 15).
Furthermore, the majority of human tumors have multiple genetic alternations; notably a delation in the tumor suppressor gene p53 is most prevalent. It is therefore extremely important to determine whether or not inhibition of Ras function is sufficient to halt the growth of human tumors which harbor K-Ras mutation as well as deletions in p53.
To evaluate the antitumor efficacy of FTI-276, a nude mouse xenograft model was used. In this model, tumors from two human lung carcinoma cell lines are implanted subcutaneously. One of these (Calu-1) harbors a K-Ras oncogenic mutation and has a deletion of the tumor suppressor gene p53. The other human lung carcinoma (NCI-H180) has no Ras mutations. Thirty two days after s.c.

CA 022072~2 1997-06-06 W O96J21456 PCTrUS96/01559 implantation when the tumors reached sizes of 60 to 80 mm3, the mice were randomly separated into control and treated groups (4 animals per group;
each animal had a tumor on both the right and the left flank). Figure 7A shows that tumors from control animals treated with saline once daily starting on day 36 grew to an average size of 566 mm3 over a period of 64 days from tumor implantation. In contrast, tumors treated once daily with FTI-276 (50 mg/kg) grew very little and the average tumor size was 113 mm3 (Fig. 7A). In another experiment, FTI-277, the methylester of FTI-276, inhibited the growth of Calu-I cells to the same extent (Figure 8). Although the animals were treated once daily with 50 mg/kg for 36 days (total cumulative does of 1.8 g/kg), no weight loss was observed and the animals appeared normal with no evidence of gross toxicity. This lack of toxicity was also observed in separate experiments where the dose was escalated to 180 mg/kg once daily. Thus, FTI-276 and FTI-277 essentially blocked tumor growth of Calu-I carcinoma with no evidence of gross toxicity.
The effect of FTI-276 on the tumor growth of another human lung carcinoma, NCI-H810, that does not harbor an oncogenic Ras mutation was also determined. Figure 7B shows that tumors from animals treated with saline or FTI-276 grew at a similar rate. Over a period of 14 days of treatment the average tumor size of the control and FTI-276 treated groups were 919 mm3 and 815 mm3, respectively. These results clearly demonstrate that in contrast to Calu-1, NCI-H810 carcinomas were not sensitive to FTI-276 treatment suggesting that FTI-276 inhibition of tumor growth of hl7m;7n lung carcinomas is Ras-dependent.

CA 022072~2 1997-06-06 W O 96/21456 PCTrUS96/015S9 Furthermore, FTI-276 inhibited tumor growth even though Calu-1 does not express p53.
To further establish the selectivity of FTI-276 to inhibit selectively Ras-dependent tumors, the anti-tumor efficacy of FTI-276 and FTI-277 against H-RasF and Raf transformed NIH 3T3 in the same nude mouse xenograft model was ~x~m; ned~
Figure 9 shows that a once daily injection of FTI-276 or FTI-277 (50 mg/kg) inhibited tumor growth of H-RasF transformed NIH 3T3 cells. In contrast, an identical treatment regimen with FTI-276 and FTI-277 had no effect on the growth of Raf-transformed NIH 3T3 cells (Fig. 10), further confirming the conclusion from the results of Figures 7 and 8 that FTI-276 and FTI-277 are selective for Ras-dependent tumors.
In addition, the question of whether FTI-276 inhibition of tumor growth correlated with inhibition of Ras processing in vivo was addressed. To so this, mice having subcutaneous H-RasF cells were treated with various doses of FTI-276 (0, 10, 50 and 100 mg/kg) and tumor size and Ras processing in the HRasF tumors in vivo were ~m; ned. Figure llA shows that throughout the 11 day treatment period, FTI-276 inhibited tumor growth in a dose dependent fashion. The tumor sizes at the end of 17 days were 2490 mm3 for saline, 1793 mm3 for 10 mg/kg, 1226 mm3 for 50 mg/kg and 624 mm3 for 100 mg/kg treated animals.
To determine the levels of inhibition of Ras processing, the animals were sacrificed 5 hrs after the last injection, the tumors were excised and processed for immunoblotting with anti-Ras antibody as described in legend to Fig. 11.
Tumors from control animals contained only fully processed Ras which migrates faster in SDS-PAGE
gels (Fig. llB). As the dose of FTI-276 increases CA 022072~2 l997-06-06 W O96/21456 PCTnUS96/015S9 from 10 to 100 mg/kg there was a progressive accumulation of unprocessed Ras which was paralleled by a decrease in the relative ratio of fully processed Ras. Thus, the extent of tumor growth inhibition correlated with the extent of inhibition of Ras processing. Furthermore, the inhibition of Ras processing in vivo was selective in that FTI-276 did not inhibit RaplA processing even at 100 mg/kg.
II. FarnesyltranRferase Inhibitors of the type C~
Compounds of another major embodiment of the invention are represented by formula II. Several examples are shown in Figure 12. These and other compounds of this embodiment may be prepared using - procedures which are conventional in the art. For example, compounds 4 and 5 of Figure 12 may be prepared by reductive amination of 4-amino-3'-tert.butoxy-carbonyl biphenyl or 4-amino-4'-tert.butoxy carbonyl biphenyl, respectively, with N-Boc-S-trityl cysteinal followed by deprotection with, for example, trifluoroacetic acid and purification.
This embodiment of the invention is illustrated but not limited by the following examples:

The compound C-4ABA-Met of formula (2) (see Figure 12) was prepared as described in reference (27). The protected form of the peptidomimetic (2a) was prepared through the reductive amination of 4-aminobenzoyl methionine methyl ester and N-Boc-S-trityl cysteinal in methanol solution cont~;n;ng NaBH3CN and 5~ acetic acid. This reaction gave the N-Boc-S-trityl, methyl ester of (2a) with a yield of 65~. The protected -CA 022072~2 1997-06-06 W O96121456 PCTrUS96/01559 peptidomimetic was deesterified by LiOH in THF and then deprotected by trifluoroacetic acid in methylene chloride with two equivalents of triethylsilane to give crude (2a) which was purified by reverse phase HPLC. The biphenyl-based peptidomimetic (8) was prepared by the reductive amination of 4-amino-3'-methyl biphenyl with N-Boc-S-trityl cysteinal, to give the N-Boc-S-trityl derivatives of (8~), which was then deprotected by trifluoroace~ic acid and purified by reverse phase HPLC. The peptidomimetics (4) and (5) were prepared from the reductive amination of 4-amino-3'-tert.butoxycarbonyl biphenyl and 4-amino-4'-tert.butoxycarbonylbiphenyl, respectively, with N-Boc-S-trityl cysteinal, to give the N-Boc-S-trityl, tert-butyl ester of (4) and (5). Deprotection by trifluoroacetic acid and purification by reverse phase HPLC gave pure (4) and (5).

Synthesi~
The basic approach used for the preparation of the compounds of the invention is illustrated in Scheme 1 with the synthesis of compound 4.
[Compound numbers in the following discussion refer to Schemes 1 and 2.]

WO 96/21456 PCTIUS96/OlSS9 Scheme 1. Representative Synthesis of FTase Tnhihi~r~
02N~Br + ,B,~ a ~ 02N~ . ozN ~3 ~ ~ ~ d O2N ~
BocNH 18 COOBut, COOBut HS~

y cY~teinai 17, (3) NaB(cN)H3 (~) TFAPYEei3dsinH~H2O; (c) (1) (COCI)2, (2) ~ert-butyl alcoh I B L
S~h.~n~ 2. Synthesis of Compounds 11 and 12 ~ a ~ b Ph3CS~'o_~
H3C COOBu' N3 COOBut BocNH N COOBut ~d ¦c Ph3CS~ ~ HS~_~o_~
BocNH N COOBut H2N N COOH

~c HS~

CA 022072~2 1997-06-06 W O96/214S6 PCTrUS96/01559 l-Bromo-4-nitrobenzene was coupled to 3-methylphenylboronic acid (34) through an modified Suzuki coupling (30) to afford compound 14 (35).
Compound 14 was oxidized to carboxylic acid 15 which was converted to the acid chloride and reacted with lithium tert-butoxide (36) to give the tert-butyl ester 16. Reduction of 16 by hydrogenation and subseqent reductive amination (37) of the resulting amine with N-Boc-S-trityl-cysteinal 17 (38) gave the fully protected derivative 18, which was deprotected by trifluoroacetic acid in the presence of triethylsilane (39). Compound 4 was purified by reverse phase HPLC and isolated as its trifluoroacetate salt by lyophilization.
The synthesis of 11 and 12 is described in Scheme 2. Compound 19 was made from 3-methyl-3'-carboxybiphenyl (itself formed from aryl-aryl coupling of methyl 3-bromobenzoate with 3-methylphenylboronic acid followed by a saponification) via the same method as compound 16. Bromination of 19 followed by reaction with sodium azide gave 20 which as catalytically hydrogenated to give the corresponding amine.
Reaction of Boc-trityl protected cysteine with this amine through the mixed anhydride method gave 21, while compound 22 was made by reductive amination with Boc-trityl protected cysteinal.

Experimental lH and 13C NMR spectrum were recorded on a Bruker AM-300 spectrometer. Chemical shifts were reported in ~ (ppm) relative to tetramethylsilane.
All coupling constants were described in Hz.
Elemental analyses were performed by Atlantic Microlab Inc., Georgia. Optical rotations were measured on a Perkin-Elmer 241 polarimeter.

CA 022072~2 1997-06-06 WO96/21456 PCT~S96101559 Concentrations are expressed in g/mL. Flash column chromatography was performed on silica gel (40-63 ~m) under a pressure about 4 psi. Solvents were obtained from commercial suppliers and purified as following: tetrahydrofuran and ether were distilled from sodium benzophenone ketyl, methylene chloride was distilled over lithium aluminum hydride. Preparative HPLC was performed using a Waters 600 E controller and a Waters 490 E
Multi-Wavelength W detector with a 25x10 cm Delta-Pak C-18 300 A cartridge column inside a Waters 25x10 cm Radial Compréssion Module.
Analytical HPLC was performed using a ~ln;n HP
Controller and a R~-n;n W-C detector with a R~;n;n 250x4.6 mm 5 ~m Microsorb C-18 column.
High resolution mass spectra (HRMS) and low resolution ma~s speçtra (TRMS) were perfo~r..ed on a Varian MAT CH-5 and VG 7070 mass spectrometer.
The purity of all the synthesized inhibitors was more than 98~ as indicated by analytical HPLC.

A. 4-Nitro-3'-methylbiphenyl (14).
To a mixture of 4-nitrobenzene (3.0 g, 14.8 mmol) and 3-methylphenylboronic acid (2.06 g, 15.1 mmol) in 35 mL of acetone and 40 mL of water was added K2CO3-1.5H2O (5.93 g, 37.5 mmol) and Pd(OAc)2 (101 mg, 0.50 mmol). The deep black mixture was refluxed for 6 hr and then cooled. The mixture was extracted with ether and the organic layer was passed through a layer of celite. The pale yellow solution was dried over Na2SO4 and evaporated to dryness. The residue was recrystallized from hot methanol to give pale yellow crystals (2.68 g, 85~). m.p.59-60 ~C.lH NMR (CDCl3) ~ 8.26 (d, 8.7 Hz, 2H), 7.70 (d, 8.7 Hz, 2H), 7.41 (m, 3H), 7.26 (d, 7.1Hz, lH), 2.43 (s, lH). 13C NMR (CDCl3) ~
147.6, 146.8, 138.8, 138.6, 129.6, 128.9, 128.0, _ 99 _ CA 022072~2 l997-06-06 W 096/21456 PCTrUS96/OlS59 127.6, 124.4, 123.9, 21.4. LRMS (EI) for Cl3H1lNO2213 (M+, intensity 100); HRMS (EI) calcd 213.0789, obsd 213.0778.

B. 4-Nitro-3'-carboxybiphenyl (15).
Compound 14 (2.31 g, 10 mmol) was suspended in a mixture of lOmL of pyridine and 20 mL of water. The mixture was heated to refluxing and then KMnO4 (7.9 g, 50 mmol) was added in portions.
This mixture was refluxed for 1 hr and then stirred at room temperature for 4 hr. The hot mixture was filtered and the black solid was washed with hot water. The filtrate was acidified with 6 N HCl. The precipitate was collected and dried (2.16 g, 8990-). m.p. 265~C (decomp). lH NMR
(DMSO-d6) ~i 11.1-11.4 (br s, COOH), 8.32 (d, 8.7 Hz, 2H), 8.27 (S, lH), 8.02 (m, 4H), 7.66 (t, 7.8 Hz, lH) . 13C NMR (DMSO-d6) ~ 167.1, 148.9, 145.6, 138.2, 131.8, 131.5, 129.6 (br), 127.9, 124.2 (br). LRMS (EI) for Cl3Hg04N 243 (M+, 100), 152 (60); HRMS (EI) calcd 243.0531, obsd 243.0544.
Anal. (C13HgNO4) C, H, N.

C. 4-Nitro-3'-tert-butoxycarbonylbiphenyl (16).
To a solution of 15 (1. 215 g, 5 mmol) in 30 mL of methylene chloride was added oxalyl chloride (0.65 mL, 7.45 mmol) and one drop of DMF. The mixture was stirred until no further bubbling was observed. The clear solution was exaporated to dryness to give the crude acid chloride. To another flask containing 7.0 mL of tert-butanol was added n-BuLi (1. 8 M in hexane, 2.8 mL, 5.04 mmol) under a water bath. The turbid solution was stirred for 5 min at room temperature and then the above acid chloride in 20 mL of THF was added through a dropping funnel. The mixture was stirred overnight before the solvents were ~ - .
CA 022072~2 1997-06-06 WO96/21456 PCT~S96101559 evaporated. The residue was dissolved into methylene chloride and washed with 0.5 N NaOH.
The organic layer was dried over MgSO4 and evaporated. The residue was recrystallized from methanol to give pale yellow crystals (851 mg, 57~). m.p. 110.5-111.0~C. lH NMR (CDCl3) ~ 8.32 (d, 7.8 Hz, 2H), 8.24 (s, lH), 8.06 (d, 7.7 Hz, lH), 7.77 (m, 3H), 7.56 (t, 7.7 Hz, lH), 1.63 (s, 9H). 13C NMR (CDCl3) ~ 165.1, 147.1, 146.5, 138.7, 132.8, 131.0, 12g.6, 129.0, 128.2, 127.8, 124.0, 81.4, 28.0, LRMS (EI) for C17H17O4N 299 (M+, 20), 243 (70), 266 (30), 152 (25; HRMS (EI) calcd 299.1157, obsd 299.1192. Anal. (C1,H17NO4) C, H~ N-D. N-Boc-S-trityl cy~teinal (17).
To a solution o~ N-Boc-S-trityl cysteine (7.44 g, 16 mmol) in 85 mL of methylene chloride was added triethylamine (2.22 mL, 16 mmoL) and N,O-dimethylhydroxylamine hydrochloride (1.57 g, 16.1 mmol). This mixture was cooled in an ice bath and 1-(3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride (EDCI, 3.08 g, 16.0 mmol) and HOBT (2.17 g, 16 mmol) was added.
The mixture was stirred at 0~C ~or 1 hr and at room temperature for a further 10 hr. The mixture was extracted with methylene chloride and 0.5 N
HCl. The organic layer was washed consecutively with 0.5 N HCl, concentrated NaHCO3 and brine. The organic layer was dried and evaporated. The residue was purified by flash column chromatography (1.5:1=hexane:ethylacetate) to give a white foam (7.40 g, 91~).m.p.59-60~(decomp). 1H
NMR (CDCl3) ~ 7.41 (m, 6H), 7.20-7.31 (m, 9H), 5.13 (d, 8.9 Hz, lH), 4.76 (br s, lH), 3.64 (s, 3H), 3.15 (s, 3H), 2.56 (dd, 4.7 and 12.1 Hz, lH), 2.39 (dd, 7.8 and 12.1 Hz, lH), 1.43 (s, 9H). 13C NMR
(CDCl3 ~ 170.7, 154.9, 144.2, 129.3, 127.6, 126.4, CA 022072~2 1997-06-06 W O96/21456 PCTrUS96/015~9 79.3, 66.4, 61.2, 49.5, 33.8, 31.8, 28.1. This carboxyamide (2.02 g, 4.0 mmol) was dissolved in 30 mL of ether and cooled to -10~c. Lithium aluminum hydride (167 mg, 4.40 mmol) was added and the mixture was stirred for 15 min under the nitrogen. Then 40 mL of 0.5 N HCl was added and the solution was extracted with ether. The ether layer was washed with 0.5 N HCl and dried. The evaporation of solvents gave a white foam (1.80 g) which was used for further reaction without purification. The lH NMR spectrum of this compound was complex. The percentage of the aldehyde was about 65-70~, which was calculated according to the integration of the sharp singlet (~ 9.17) and the trityl peak (~ 7.40, m, 6H;7.28,m,9H).
Lowering the temperature to -45~C did not improve the aldehyde percentage.

E. 4-N-[2(R)-tert-butoxycarbonylamino-3-triphenylmethylthipropyl]amino-3'-~ert-butoxycarbonylibiphenyl (18).
Compound 16 (768 mg, 2.56 mmol) was dissolvedin THF. A catalytic amount of 10~ Pd on activated carbon (78 mg) was added. The mixture was hydrogenated (40 psi) for 30 min. The black mixture was passed through a thin layer of celite and the pale yellow solution was evaporated. The residue was dissolved in 10 mL of methanol. To this solution was added 0.5 mL of acetic acid and a solution of the same equivalents of aldehyde 17 (according to the lH NMR determination) in 6 mL of methanol. Sodium cyanoborohydride (241 mg, 3.84 mmol, 1.5 eq) was added and the mixture was stirred overnight. After the evaporation of solvents, the residue was extracted with ethyl acetate and concentrated sodium bicarbonate. The organic layer was dried and evaporated. The CA 022072~2 1997-06-06 WO96/21456 PCT~S96/~1559 residue was puri~ied by flash column chromatography (3.5:1-hexane:THF) to give a white foam (1.09 g, 61~). m.p. 75.0-76.0 ~C(decomp).
[~]25D=-2.13 (c=0.01, CH3COOC2H5). lH NMR (CDC13) 8.14 (s, lH), 7.86 (d, 7.7 Hz, lH), 7.66 (d, 7.8 Hz, lH), 7.40 (m, 9H), 7.22-7.30 (m, 9H), 6.61 (d, 8.5 Hz, 2H), 4.58 (d, 7.1 Hz, lH), 3.83 (br m, 2H, Cys ~ proton and the amine), 3.12 (br m, 2H, CH2N), 2.48 (br, m, 2H, CH2S), 1.60 (s, 9H), 1.44 (s, 9H).
13C NMR (CDc13) ~ 165.9, 155.6, 147.5, 144.4, 141.2, 132.3, 130.1, 129.5, 129.2, 128.5, 128.0, 127.9, 127.1, 126.8, 112.9, 80.9, 79.7, 67.0, 49.4, 47.1, 34.3, 28.3, 28.2 (expect 14 aromatic C, observed 13). Anal. (C44H48N2O4S 1.2H2O) C, H, N, S.

F. 4-N-~2~R)-amino-3-mercaptopropyl]amino-3'-carboxybiphenyl (4).
Compound 18 (600 mg, 0.85 mmol) was dissolved in 2mL of TFA and 2 mL of methylene chloride.
Triethylsilane was added dropwise to the deep brown mixture until the brown color had disappeared. The mixture was then kept at room temperature for 1 hr. Then solvents were evaporatee and the residue was dried under vacuum.
The solid was triturated with 30 mL of ether and 3mL of 3 N Hcl in ether. The whité precipitate was filtered and washed with ether to obtain a crude product (270 mg, 84~). This crude product was dissolved into 30 mL of dilute Hcl solution (O.OlN) and was lyophilized. Analytical HPLC
showed the purity to be 95~.m.p. 105-106 ~C
(decomp). [~]25D=+13.16 (c=0.01 in methanol. lJ M<R
(CD30D ~ 8.18 (s, lH), 7.89 (d, 7.7 Hz, lH), 7.78 (d, 7.3 Hz, lH), 7.49 (m, 3H), 6.82 (d, 8.5 Hz, 2H), 3.56 (m, 2H CHN and CH2N), 3.42 (dd, 8.9 and 15.2 Hz, lH, CH2S). 13C NMR (D2O and CD30D) CA 022072~2 1997-06-06 W O96/21456 PCTrUS96101S59 171.1, 147.1, 141.4, 131.9, 130.9, 130.1, 128.8, 128.5, 127.0, 115.2, 53.2, 45.4, 25.0 LRMS (FAB, glycerol) for Cl6Hl8N2O2S (M+l) 303. Anal.
(Cl6Hl8N2O2S 2HCl) C, H, N, S. Further purification by preparative HPLC (Waters C-18, 40~
acetonitrile, 60~ water, 0.1~ TFA, 40 min gradient) gave product 4 (120 mg) with a purity over 99.9~.

G. 3-Methyl-3'-tert-butoxycarbonylbiphenyl (19).
The coupling of 3-methylphenylboronic acid with 3-bromobenzoic acid methyl ester gave a 3-methyl-3'-methoxycarbonylbiphenyl (79~ yield), which was then hydrolyzed to yield a 3-methyl-3'-carboxybiphenyl (97~ yield). Compound 19 (an oil) was prepared from this acid using the same method as for the preparation of compound 16 (65~ yield).
H NMR (CDl3) ~ 8.21 (s, lH), 7.95 (d, 7.8 Hz, lH), 7.73 (d, 6.6 Hz), lH), 7.46 (m, 3H), 7.35 (t, 7.5 Hz, lH), 7.20 (d, 7.4 Hz, lH), 2.43 (s, 3H), 1.62 (s, 9H). 13C NMR (CDCl3) ~ 165.6, 141.3, 140.2, 138.3, 132.4, 140.0, 128.7, 128.5, 128.3, 128.0, 127.9, 124.2, 81.0, 28.1, 21.4. LRMS (EI) for Cl8H20O2 268 (M+, 35), 212(100), 195 (20); HRMS (EI) calcd 268.1463, obsd 268.1458.

H. 3-Azido-3'-tert-butoxycarbonylbiphenyl (20).
Compound 19 (2.18 g, 8.13 mmol) and N-bromosuccinimide (1.70 g, 9.50 mmol) was suspended in 60 mL of CCl4. Dibenzoyl peroxide (20 mg) was added and the mixture was refluxed for 1.5 hr.
After removing the solid, the filtrate was washed with concentrated sodium bicarbonate and dried over sodium sulfate. lH NMR showed the crude material contained 70~ of monobrominated and 30 of dibrominated product. This material was dissolved in 20 mL of DMSO and sodium azide (3.70 CA 022072~2 1997-06-06 W O96/21456 PCTrUS96/01559 g, 57 mmol) was added. The mixture was heated to 80 ~C for 4 hr before being poured into a mixture of methylene chloride and water. The organic layer was dried and evaporated. The residue was purified by flash column chromatography (5% of ethyl acetate in hexane) to give 20 (2.14 g, 78~, two steps) as colorless oil. lH NMR (CDCl3) ~ 8.22 (s, lH), 8.00 (d, 7.7 Hz, lH), 7.76 (d, 8.2 Hz, lH), 7.58 (m, 2H), 7.50 (m, 2H), 7.33 (d, 7.6 Hz, lH), 4.43 (s, 2H), 1.62 (s, 9H). 13C NMR (CDC13) 165.2, 140.5, 140.3, 135.8, 132.3, 130.7, 129.1, 128.5, 128.2, 127.8, 127.1, i26.7, 126.6, 80.9, 54.3, 27.8.

I. N-Boc-S-trityl- cy8 teinyl-3- Im;nQmP thyl-3'-tert-butoxycarbonylbiphenyl (21).
Compound 20 (0.75 g, 2.43 mmol) was dissolved in 30 mL of methanol. A catalytic amount of 5 palladium on barium sulfate (0.30 g) was added.
The mixture was hydrogenated at 1 atm for 5 hr.
The catalyst was removed by filtration and the methanol was evaporated. This residue was dissolved in 40 mL of methylene chloride. N-Boc-S-trityl cysteine (1.12 g, 2.43 mmol) was added at 0~C followed by EDCI (1 eq) and HOBT (1 eq). The mixture was stirred for 24 hr. After workup and evaporation of solvents, the residue was purified by flash column chromatography (hexane:ethyl acetate=3.2:1) to give 21 (570 mg, 44~). m.p. 84-86~C. lH NMR (CDC13) ~ 8.17 (s, lH), 7.95 (d, 7.7 Hz, lH), 7.70 (d, 7.7 Hz, lH), 7.50-7.30 (m, 9H), 7.30-7.10 (m, llH), 6.44 (br, lH), 4.86 (br, lH), 4.45 (d, 4.0 Hz, 2H, CH2Ph), 3.87 (br, lH, Cys ~
H), 2.75 (dd, 7.2 and 12.8 Hz, lH, CH2S), 2.55 (dd, S.3 and 12.8 Hz, lH, CH2S), 1.62 (s, 9H), 1.36 (s, 9H). Anal. (C45H~8N2O5S) C, H, N, S.

CA 022072~2 1997-06-06 W O 96/21456 PCTrUS96/01559 J. Cysteinyl-3 _~m;n~m~thyl -3'-carboxybiphenyl (11) .
Compound 21 (150 mg) was deprotected using the same method as for the preparation of compound , 4. Final purification by preparative HPLC gave 11 as a white solid (42 mg, 46%). m.p. 88-89 ~C
(decomp). lH MNR (CD30D) ~ 8.26 (s, lH), 8.01 (d, 7.7 Hz, lH), 7.86 (d, 7.7 Hz, lH), 7.64 (s, lH), 7.56 (m, 2H), 7.46 (t, 7.6 Hz, lH), 7.35 (d, 7.6 Hz, lH), 4.53 (s, 2H), 4.00 (t, 5.2 Hz, lH, Cys ~
H), 3.06 (dd, 14.6 and 5.2 Hz, lH, CH2S), 2.97 (dd, 14.6 and 6.8 Hz, lH, CH2S). LRMS (EI) for Cl7Hl8N2O3S 331 (M+l, 8), 281 (100), 226 (75), Anal.
(Cl7Hl8N2o3s-Hcl-o.6H2o) C, H, N.

R. 3-N-t2(R)-tert-Butoxycarbonylamino-3-triphenylmethylthiopropyl~ ~m;n~me thyl-3'-tert-buto xy-carbonylbiphenyl (22).
The azide 20 (900 mg, 2.91 mmol) was dissolved in 20 mL of methanol. A catalytic amount of 5~ Pd on barium sulfate (90 mg) was added. This mixture was hydrogenated at 1 atm overnight. The catalyst was removed and the methanol was evaporated. The rern~in;ng residue was dissolved in a mixture of 0.5 N HCl (20 mL) and ether (20 mL). The aqueous phase was neutralized with 1 N NaOH and extracted into methylene chloride. After the evaporation of solvents, a viscous oil was obtained (600 mg, 73~). lH NMR (CDCl3) ~ 8.22 (s, IH), 7.97 (d, 7.8 Hz, lH), 7.75 (d, 7.7 Hz, lH), 7.57 (s, lH), 7.50 (m, 2H), 7.43 (t, 7.7 Hz, lH), 7.33 (d, 7.4 Hz, lH), 3.96 (s, 2H), 1.62 (s, 9H), 1.46 (br s, 2H, NH2). This amine (581 mg, 2.05 mmol) was dissolved in 10 mL of methanol and 0.5 mL of acetic acid before N-Boc-S-tritylcysteinal (leq, according to H NMR determination of the aldehyde percentage) CA 022072~2 1997-06-06 W O96/21456 ~CTrUS96/01559 was added. Sodium cyanoborohydride (193 mg, 1.50 eq) was added to the above solution and the mixture was stirred at room temperature overnight.
After workup, the crude residue was purified by flash column chromatography (1: 1 = ethyl acetate:
hexane) to give a white foam (602 mg, 41~). m.p.
66-68 ~C (decomp). lH NMR (CDCl3) ~ 8.21 (s, lH), 7.96 (d, 7.7 Hz, lH), 7.73 (d, 8.0 Hz, lH), 7.37-7.51 (m, lOH), 7.15-7.31 (m, lOH), 4.69 (br d, lH), 3.75 (br s, 3H, PhCH2N and Cys ~ H), 2.68 (dd, 6.0 and 12;3 Hz, lH, CH2S), 2.56 (dd, 5.5 and 12.3 Hz, lH, CH2S) r 2.47 (m, lH, CH2N), 2.35 (m, lH, CH2N), 1.62 (s, 9H), 1.42 (s, 9H), 1.12 (br s, lH, NH).

L. 3-N-t2(R)-amino-3-mercaptopropyl]
~;n~m~thyl-3'-carboxybiphenyl (12).
Compound 22 (480 mg, 0.672 mmol) was dissolved in a mixture of 2 mL of methylene chloride and 2 mL of trifluoroacetic acid.
Several drops of triethylsilane were added until the deep brown color had disappeared. This mixture was kept at room temperature for 1.5 hr, and then the solvents were evaporated, and the residue was dried under vacuum. The solid residue was dissolved in 1 mL of acetic acid and 2 mL of HCl (1.7 M) in acetic acid. Finally 5 mL of HCl (3 M) in ether and 10 mL of ether were added. The white precipitate was washed with dry ether and dried to give a hydrochloride salt (215 mg, 81~).
lH NMR (D2O) ~ 8.16 (s, lH), 7.94 (d, 7.7 Hz, lH), 7.85 (d, 7.7 Hz, lH), 7.70 (s, 2H), 7.55 (t, 7.8 Hz, 2H), 7.46 (d, 7.5 Hz, lH), 4.36 (s, 2H, PhCH2), 3.81 (m, lH, Cys ~ H), 3.57 (dd, 5.7 and 13.7 Hz, lH, CH2N), 3.44 (dd, 6.5 and 13.7 Hz, lH, CH2N), 2.97 (dd, 5.3 and 15.1 Hz, lH, CH2S), 2.86 (dd, 5.9 and 15.1 Hz, lH, CH2S).

CA 022072~2 1997-06-06 W O 96/21456 PCTrUS96/01~59 M. 2-Methoxy-4-nitro-3'-tert-butoxycarbonyl-biphenyl (23).
The coupling of l-bromo-2-methoxy-4-nitrobenzene with 3-methylphenylboronic acid followed by the oxidation gave the 2-methoxy-4-nitro-3'-carboxybiphenyl. The reaction of acid chloride with lithium tert-butoxide gave 23 (3 steps, 35~). m.p. 88.0-88.5 ~C. lH NMR (CDCl3) ~
8.13 (s, lH), 8.00 (d, 7.7 Hz, lH), 7.89 (d, 8.3 Hz, lH), 7.81 (s, lH), 7.69 (d, 7.7 Hz, lX), 7.48 (m, 2H), 3.90 (s, 3H), 1.60 (s, 9H). 13C NMR
(CDCl3) ~ 165.2, 156.7, 148.0, 136.3, 135.2, 133.2, 132.0, 130.8, 130.1, 129.0, 127.9, 115.8, 106.0, 81.1, 55.9, 27.9. LRMS (EI) for Cl8HlgNO5 329 (M+, 30), 273 (100).

N. 2-Methoxy-4-N-[2(R)-N-tert-butoxycarbonyl-amino-3-triphenylmethylthiopropyl]amino-3'-tert-butoxycarbonylbiphenyl (24).
Compound 24 was prepared using the same method as for the preparation of compound 18 (yield 63~). m.p. 76.0-77.0 ~C (decomp). [~]25D=-11.25 (c=O.Ol, CH3COOC2H5). lH NMR (CDCl3) ~ 8.09 (s , lH), 7.86 (d, 7.0 Hz, lH), 7.65 (d, 7.0 Hz, lH), 7.37 (t, 7.7 Hz, lH), 7.43 (m, 6H), 7.21-7.32 (m. 9H), 7.11 (d, 8.1 HZ, lH), 6.21 (s, lH), 6.18 (d, 8.1 Hz, lH), 4.58 (d, 6.1 Hz, lH), 3.86 (br s, lH), 3.76 (s and m, 4H), 3.14 (br d, 4.9 Hz, 2H), 2.49 (br d, 5.1 Hz, 2H), 1.59 (s, 9H), 1.43 (s, 9H). 13C NMR (CDC13) ~ 165.9, 157.3, 155.5, 148.8, 144.3, 138.9, 133.3, 131.5, 131.2, 130.0, 129.4, 127.8, 127.5, 126.7, 118.7, 104.7, 96.2, 80.5, 79.4, 66.8, 55.2, 49.3, 47.0, 34.1, 28.2, 28.1.
Anal. (C45H50N2O5S) C, H, N, S.

O. 2-Methoxy-4-N-~2(R)-amino-3-mercaptopropyl]
amino-3~-carboxybiphenyl (10).

-CA 022072~2 1997-06-06 WO96/21456 PCT~S96/01559 Compound 10 was obtained from the deprotection of compound 24. m.p. 120-121 ~C
(decomp). [tx]25D=+12 .62 (c=O.Ol, in methanol). lH
NMR (CD30D) ~ 8.09 (s, lH), 7.89 (d,7.8 Hz, lH), 7.67 (d, 7.8 Hz, lH), 7.43 (t, 7.7 Hz, lH), 7.20 (d, 8.1 Hz, lH), 6.56 (s, lH), 6.53 (d, 8.1 Hz, lH), 3.81 (s, 3H), 3.60 (m, 2H, Cys ~ H and CH2N), 3.48 (m, lH, CH2N), 2.96 (dd, 4.9 and 13.7 Hz, lH, CH2S), 2.86 (dd, 5.4 and 13.7 Hz, lH, CH2S). 13C
NMR (D20 and CD30D) ~ 171.1, 158.2, 149.3, 139.7, 135.1, 132.2, 131.1, 130.4, 129.4, 128.4, 120.5, 106.2 (broad, due to deuterium exchange), 98.8, 56.3, 53.4, 45.1, 24.9. LRMS (EI) for Cl7H20N2O3S 332 (M+). Anal. (Cl7H2oNzO3S 1.2HCl H2O) C, H, N, S.

P. Cysteinyl-4-amino-3'-carboxybiphenyl (6).
Compound 6 was purified through preparative HPLC. Purity was shown to be over 99~. m.p.
120.0-121.0 ~C. lH NMR (CD30D) ~ 8.25 (s, lH), 7.98 (d, 7.6 Hz, lH), 7.84 (d, 7.7 Hz, lH), 7.74 (d, 7.0 Hz, 2H), 7.54 (t, 7.7 Hz, lH), 7.66 (d, 8.6 Hz, 2H), 4.16 (~, 5.0 Hz, lH), 3.19 (dd, 5.20 and 14.8 Hz, lH), 3.07 (dd, 7.7 and 14.7 Hz, lH); 13C
NMR (CD30D) ~ 169.8, 166.7, 141.9, 138.7, 137.8, 132.6, 132.2, 130.2, 129.5, 128.8, 128.5, 121.6, 56.9, 26.4. LRMS (El) for Cl6Hl6O3N2S 316 (M+, 25), 213 (100); HRMS (EI) calcd 316.0882, obsd 316.0867. Anal. (Cl6Hl6N2O3S-CF3COOH-H2O) C, H, N.

Q. 4-N-~2(R)-Amino-3-mercaptopropyl]amino-2'-carboxybiphenyl (3~.
m.p. 129-130 ~C (decomp). [~x]25D=+12 .58 (c=0.01, CH30H). lH NMR (CD30D) ~ 7.73 (d, 7.6 Hz, lH), 7.50 (d, 7.6 Hz, lH), 7.35 (m, 2H), 7.21 (d, 8.5 Hz, 2H), 6.86 (d, 8.5 Hz, 2H), 3.58 (m, 2H), 3.46 (m, lH), 2.97 (dd, 4.8 and 14.6 Hz, lH), 2.86 (dd, 5.4 and 14.6 Hz, lH). 13C NMR (D2O and CD30D) CA 022072~2 1997-06-06 W O 96/21456 PCTnUS96/01559 174.3, 146.3, 141.9, 132.8, 132.7, 131.4, 130.6, 130.2, 127.9, 115.3, 52.9, 45.8, 25Ø LRMS (FAB, glycerol) for C16H18N202S (M+l) 303. Anal.
(Cl6Hl8N2o2s-l.6Hcl) C, H, N, S.

R. 4-N-t2(R)-Amino-3-mercaptopropyl]a_ino-4'-carboxybiphenyl (5).
m.p. 260 ~C (decomp). [~x]25D=+12 .20 (c=0.01, CH30H). lH NMR (CD30D) ~ 8.03 (d, 8.5 Hz, 2H), 7.66 (d, 8.4 Hz, 2H), 7.56 (d, 8.4 Hz, 2H), 6.85 (d, 8.5 Hz, 2H), 3.57 (m, 2H), 3.45 (m, lH), 2.98 (dd, 4.8 and 14.5 Hz, lH), 2.85 (dd, 5.7 and 14.5 Hz, lH). 13C NMR (D20 and CD300) ~ 169.8, 146.4, 146.1, 133.6, 131.4, 129.8, 129.4, 127.1, 117.0, 53.3, 47.2, 25.5. LRMS (EI~ for C16H1802N2S 302 (M ~ 15)~
285 (15), 226 (100), 213 (50). HRMS (EI) calcd 302.1088, obsd 302.1089. Anal. (Cl6Hl8N2O2S 2HCl) C, H, N.

S. 4-N-[2(R)Amino-3 -Mercaptu~Lo~yl] ~m; nohiphenyl (7).
m.p. 216 ~C (decomp). [~]25D=+13.27 (c=O.Ol, CH30H). lH NMR (CD30D) a! 7.54 (m, 4H), 7.39 (m, 2H), 7.26 (m, lH), 6.82 (br s, 2H), 3.56 (br m, 2H), 3.45 (m, lH), 2.98 (m, lH), 2.87 (m, lH); 13C
NMR (CD30D) ~ 144.8, 141.8, 135.7, 129.9, 129.1, 127.8, 127.4, 117.4, 53.2, 47.6, 25.5. LRMS (EI) for Cl5Hl8N2S 258 (M+, 15), 182 (100); HRMS (EI) calcd 258.1190, obsd 258.1183. Anal.
(Cl5Hl8N2S-1.6HCl) C, H, N, S.

T. 4-N-[2(R)-Amino-3-mercaptopropyl]amino-3'-methylbiphenyl (8).
lH NMR (CD30D) ~ 7.50 (d, 8.2 Hz, 2H), 7.35 (m, 2H), 7.27 (t, 7.6 Hz, lH), 7.08 (d, 7.3 Hz, lH), 6.95 (d, 8.2 Hz, 2H), 3.60 (m, 2H), 3.46 (m, lH), 2.99 (dd, 4.9 and 14.6 Hz, lH), 2.88 (dd, 5.5 CA 022072~2 1997-06-06 WO961214S6 PCT~S96/015~9 and 14.6 Hz, lH), 2.37 (s, 3H). LRMS (EI) for Cl6H20N2S 272 (M+, 15), 196 (100). HRMS (EI) calcd 272.1341, obsd 272.1347.

U. 4-N-[2(R)-~;nQ-3-mercaptopropyl~amino-3'-methoxyca ~o..ylbiphenyl (9).
m.p. 86-89 ~C (decomp). lH NMR (CD30D) ~ 8.18 (s, lH), 7.90 (d, 7.7 Hz, lH), 7.79 (d, 6.6 Hz, lH), 7.55 (d, 8.6 Hz, 2H), 7.49 (t, 7.7 Hz, lH), 7.01 (d, 8.6 Hz, 2H), 3.92 (s, 3H), 3.543.65 (m, 2H), 3.44-3.52 (m, lH), 2.97 (dd, 4.7 and 14.7 Hz, lH), 2.88 (dd, 5.5 and 14.7 Hz, lH). LRMS (EI) for Cl7H20O2N2S 316 (M+, 15), 299 (20), 240 (100). HRMS
(EI) calcd 316.1240, obsd 316.1239. Anal.
(Cl7H20N2O2S-2HCl) C, H, N, S.

CA 022072~2 1997-06-06 PCTrUS96/01559 Table 2 Table of Microanalysis Data C~mpd Fonnulae C%(o~,obs) H%(c~,obs) N%(c~,obs) S%(c~,obs) 3 C16HIgN2O2S-1.6HCl 53.27(53AI) 5.44(5.78) 7.77(7.35) 8.87(8.47) 4 C16HIgN2O2S-2HCl 51.20(51.60) 5.37(5.30) 7.47(7.07) 8.53(8.22) C16HlgN2O2S-2HC1 51.20(51.62) 5.37(5.56) 7.47(7.00) 6 C16H16N2O3S CF3COOH-H2O 48.21(48.24) 4.24(4.20) 6.25(6.32) 7 ClsHIgN2S-1.6HCl 56.89(57.04) 6.19(6.46) 8.85(8.74) 10.11(10.03) 9 C17H20N2o2s-2Hcl 52.44(52.84) 5.65(5.92) 7.19(7.37) 8.22(8.53) C17H20N2o3s-l-2Hcl-H2o 51.80(51.91) 5.89(5.96) 7.11(6.81) 8.12(7.77) 11 C17HlgN2O3s-Hcl-0.6H2O 54.07(54.11) 5.35(5.39) 7.42(7.35) C13HgNO4 64.19(64.05) 3.70(3.75) 5.76(5.80) 16 C17Hl7NO4 68.23(68.07) 5.68(5.73) 4.68(4.64) 18 ~1~8~2O4sl.2H2o 73.17(72.82) 6.98(6.83) 3.88(3.87) 4.43(4.50 21 C4sH48N2Oss 74.14(73.74) 6.64(6.74) 3.84(3-79) 4.39(4-32) 24 C4sHsoN2OsS 73.97(73.72) 6.85(7.04) 3.83(3.66) 4.38(4.32) CA 022072~2 1997-06-06 Table 3 Examples of Pepti~ tics of the In~ention Compound Structure 2 FTI-232 Cys-4-aminobenzoyl-Met 2a FTI-249 red.Cys-4-aminobenzoyl-Met 3 FTI-273 red.Cys-4-amino-2'-carboxybiphenyl 4 FTI-265 red.Cys-4-amino-3~-carboxybiphenyl FTI-271 red.Cys-4-amino-4'-carboxybiphenyl 6 FTI-278 Cys-4-amino-3~-carboxybiphenyl 7 FTI-268 red.Cys-4-aminobiphenyl 8 FTI-263 red.Cys-4-amino-3'-methylbiphenyl 9 FTI-259 red.Cys-4-amino-3'-carboxymethylbiphenyl 10 FTI-281 red.Cys-4-amino-2-OMe-3'-carboxybiphenyl 11 FTI-285 red.Cys-4-amino-2-phenyl-3'-methylbiphenyl 12 FTI-238 Cys-3-~r;n~ -thyl-3'-carboxybiphenyl FTI-283 red.Cys-3-~r;nsm~thyl-3'-carboxybiphenyl FTI-282 (DL)4-(2,3-~i~r; n~propyl)-amino-3' carboxybiphenyl FTI-288 red.Cys-4-amino-2-OPr-3'-carboxybiphenyl FTI-289 red.Cys-4-amino-2-phenyl-3'-carboxybiphenyl FTI-2s1 4-(3-~m;no~l~nyl)-amino-3'-carboxybiphenyl FTI-292 (L)4(2,3-~;~m;nopropyl)-amino-3'-carboxybiphenyl FTI-295 4-(Ethylsulfonyl-3-~m;no~l~nyl)-amino-3~-carb biphenyl FTI-296 4-(Vinylsulfonyl-3-~m;no~l~nyl)-amino-3'-carboxybiphenyl FTI-2102 red.Cys-4-amino-3'-tetrazolylbiphenyl CA 022072~2 l997-06-06 W O96/21456 PCTrUS96/01559 Number designations used for compounds of the invention discussed below is shown in Table 3.

FTase and GGTase I Activity AssaY
Human Burkitt lymphoma (Daudi) cells (ATCC, Rockville, MD) were grown in suspension in RPMI
1640 medium containing 10~ fetal bovine serum (FBS) and 1~ Pen-Strep in a humidified 10~ CO2 incubator at 37~C. The cells were harvested and sonicated in 50 mM Tris, pH 7.5, 1 mM EDTA, 1 mM
EGTA, 25 ~g/ml leupeptin, 1 mM
phenylmethylsulfonyl fluoride. Homogenates were then spun at 12,000 x g and the resulting supernatant further spun at 60,000 x g. The supernatant was assayed for both FTase and GGTase I. Briefly, 100 ~g of the supernatants was incubated in 50 mM Tris, pH 7.5, 50 ~M ZnCl2, 20 mM
KCl, 3 mM MgCl2 and 1 mM DTT. For FTase assays, the reaction was incubated at 37~C for 30 minutes with recombinant H-Ras-CVLS (11 ~M) and [3H] FPP
(625 nM; 16.3 Ci/mmol). For GGTase assays, the reaction was also incubated for 30 minutes at 37~C
but with recombinant H-Ras-CVLL (5 ~M) and [3H]
GGPP (525 nM; 19.0 Ci/mmol). The reaction was stopped and passed through glass fiber filters to separate free and incorporated label. For inhibition studies, the peptidomimetics were premixed with FTase or GGTase I prior to adding the remainder of the reaction mixture.
Recombinant H-Ras-CVLS was prepared as described previously (26) from bacteria (31). Recombinant H-Ras-CVLL was prepared from bacteria (32).

CA 022072~2 1997-06-06 W O 96121456 PCTnUS96/01559 Peptidomimetics FarnesYlation Assay The ability of human Burkitt lymphoma~ (Daudi) FTase to farnesylate peptides and peptidomimetics was determined as described previously (34, 35).
Briefly, 25 ~l of reaction mixture containing 50 ~g of 60,000 xg supernatants and 20 ~M
peptidomimetic in 50 mM Tris, pH 7.5, 50 ~M ZnCl2, 20 mM KCl, 3 mM MgCl2, 1 mM DTT and 0.2~ octyl~-D-glucoside was incubated for 30 minutes at 37~C, then spotted onto silica gel G TLC sheets (20 x 20 cm, Brinkmann Instruments), and developed with n-propanol/5 N ammonium hydroxide/water (6:1:1).
The dried sheets were sprayed with En3Hance (DuPont NEN) and exposed to x-ray film for detection of [3H] farnesylated products.

Ras and Ra~lA Processin~ Assay EJ3 cells were treated with peptidomimetics or vehicle for 20-24 h. Cells were lysed in lysis buffer (10 mM Na2HPO4, pH 7.25, 150 mM NaCl, 0.1 sodium dodecyl sulfate, 1~ Triton X-100, 12 mM
sodium deoxycholate, 1 mM NaF, 0.2~ NaN3, 2 mM
PMSF, 25 ~g/ml leupeptin) and the lysates were cleared by spinning at 13,000 rpm for 15 minutes.
Ras protein was immunoprecipitated overnight at 4~C with 50 ~g of anti-Ras antibody (Y13-259;
hybridoma from ATCC, Rockville, MD) along with 30 ~1 Protein A-agarose goat anti-rat IgG complex (Oncogene Science, Uniondale, NY).
Immunoprecipitates were washed 4 times with lysis buffer and the bound proteins were released by heating for 5 minutes in 40 ~l SDS-PAGE sample buffer and subsequently electrophoresed on a 12.5 SDS-PAGE. Proteins were transferred onto nitrocellulose and subsequently blocked with 5 CA 022072~2 1997-06-06 W 096/21456 PCT~US96/01559 non-fat dry milk in PBS (containing 1~ Tween 20 (PBS-T) and probed with Y13-259 (50 ~g/ml in 3 non-fat dry milk in PBS-T). Positive antibody reactions were visualized using peroxidase-conjugated goat anti-rat IgG (Oncogene Science, Uniondale, NY) and an enhanced chemiluminescence detection system (ECL; Amersham).
For RaplA processing assays, 50 ~g of cell lysates were electrophoresed as described above for Ras processing and transferred to nitrocellulose. These membranes were then blocked with 5~ milk in Tris-buffered saline, pH 8.0, containing 0.5~ Tween-20 and probed with anti-RaplA (1 ~g/ml in 5~ milk/TBS-T; Santa Cruz Biotechnology, Santa Cruz, CA). Antibody reactions were visualized using peroxidase-conjugated goat anti-rabbit IgG (Oncogene) and ECL
chemiluminescence as described above.

Structural Modelinq (Fiqure 13) The calculation of the energy minimized conformations was carried out using the AMBER
force field within the MacroModel program, version 3.5a.

The potency of the peptidomimetics of Figure 12 and Table 3 for inhibiting partially purified FTase was evaluated by determining their ability to inhibit the transfer of farnesyl to recombinant H-Ras as described above. The results are summarized in Table 4, which indicates the ICsos obtained for FTase activity and GGTase-I activity, and the selectivity for a number of peptidomimetics of the invention. The IC5Q values given in Table 4 represent inhibition of FTase and GGTase I in vitro by the listed compounds.

-CA 022072~2 1997-06-06 W O 96/21456 PCTnUS96101559 Table 4 In vitro Activity of CAAX Mimetic Inhibitors of FTase lr FTase ICso GGTase-I ICso Inhibitor (nM) (nM)Selectivity Substrate FTI-232 150 lS00 10 FTI-249 300 ~ 4400 15 FTI-273 543(3)~ 140,000 (2)~258 ndb FTI-265 114 (10) lOo,000 (6)877 no FTI-271 4575(4) ~100,000 (2)~22 no FTI-278 13,500(2) lOo,OOo (2) 7 nd 1 0 FTI-268 1,070(3) ~100,000 (3)93 no FTI-263 710 (3) ,100,000 t3)141 no FTI-259 917 (3) ,100,000 (3)109 no FTI-281 40 (6) 43,600 (5) 1090 ~ nd FTI-238 100,000 (2) ,100,000 (3) ~1 no FTI-283 11,000 (1) 35,000 (2) 3 nd FTI-285 2075 (4) 8500 (2) 4 FTI-282 50,000 (1) ~lOO,oO0 (1),2 FTI-288 41 (6) 2375 (4) 59 FTI-289 16 (5) 643 (4) 40 2 0 FTI-291 210,000 (1) 0 FTI-292 60,000 (1~ 0 FTI-295 200,000 (1) ~l,OoO,000 (1) ~5 FTI-296 430,000 (1) ~l,oO0,000 (1) ~2 FTI-2102 30 (1) 5,000 (1) 187 ~Numbers in parentheses indicate number of determinations.
Where no number is given, at least two determinations were made.

., ~nd indicates not determined.

CA 022072~2 l997-06-06 W O96/21456 PCTrUS96/01559 The results obtained showed that compound 2, i.e. Cys-4ABA-Met (1-10 ~M) inhibited FTase in a concentration-dependent manner with an IC50 of 150 nM (FTI-232, Table 4). This value is similar to the previously reported IC50 values for CVIM and Cys-4ABA-Met (35). Reduction of the amide bond between cysteine and aminobenzoic acid gave the red-Cys-4ABA-Met (2a, FTI-249) which had an IC50 of 300 nM. However, replacing the methionine and the C-terminal amide bond in (2a) by another aromatic ring to obtain the biphenyl-based peptidomimetic (4) improved potency by twofold (FTI-265, Table 4). Peptidomimetic 4 had an IC50 of 114 nM towards partially purified FTase from human Burkitt lymphoma cells and 50 nM towards rat brain FTase purified to homogeneity. Thus, despite major structural differences between the compound CVIM
(1) and 4, the latter (4) retained the potent FTase inhibitory activity of the tetrapeptide CVIM
(1) and the peptide mimetics 2 and 2a.
As noted, Figures 14A and 14B graphically illustrate the results of FTase and GGTase I
inhibition studies. In these studies, partially purified FTase and GGTase I were incubated with the peptidomimetics to be tested and their ability to transfer [3H] farnesyl to H-Ras-CVLS (FTase) and [3H] geranylgeranyl to H-Ras CVLL (CCTase I) was determined as described. Figure 14A shows FTase inhibition by: ~, (4) and ~, (5) while Figure 14B
plots FTase (~) and GGTase I (-) inhibition by (4). Each curve is representative of at least four independent experiments.
Geranylgeranylation is a more common protein prenylation than farnesylation (49). It is, therefore, advantageous for CAAX peptidomimetics targeting farnesylation to have high selectivity towards inhibiting FTase compared to GGTase. In CA 022072~2 1997-06-06 WO96/21456 PCT~S96101~59 the CAAX tetrapeptides, the X position determines whether the cysteine thiol will be farnesylated by FTase or geranylgeranylated by GGTase I. Those proteins or peptides with Leu or Ile at the X
position are geranylgeranylated. As shown in Table 4, the present compounds do not significantly inhibit GGTase I and demonstrate much greater selectivity for FTase.
Figure 14B shows that compound 4, which is a potent FTase inhibitor, is a very poor GGTase I
inhibitor. The ability of compound 4 to inhibit the transfer of geranylgeranyl to Ras-CVLL (ICso =
lO0,000 nM) was found to be 877-fold less than that of 4 to inhibit the transfer of farnesyl to Ras-CVLS (IC50 = 114 nM) (Table 4). This selectivity was much more pronounced than in the peptidomimetics 2 and 2a which were more selective for FTase relative to GGTase I by only lO and 15-fold, respectively. It is also noted that the free carboxylate of compound 4 is not responsible for this selectivity since replacement of this group by a methyl in compound 8 did not increase affinity towards GGTase I (Table 4). These results indicate that the FTase and GGTase I
binding sites are quite different and that differences between Leu, Ile and Met side chains cannot be the only predictors of selectivity.
Regardless of the explanation, it is clear that the compounds of the invention are much more selective to inhibition of FTase.
Besides having poor cellular uptake and being rapidly degraded, another disadvantage of natural CAAX peptides is that they are farnesylated by FTase. This results in metabolic inactivation since farnesylated CAAX derivatives are no longer inhibitors of FTase (34). Figure 15 shows that the natural peptide CVLS (carboxyl terminal CAAX

CA 022072~2 1997-06-06 of H-Ras) is farnesylated by FTase from Burkitt lymphoma cells. Replacing the tripeptide VLS with 4-amino-3'-hydroxycarbonylbiphenyl, as in 4 did not affect potency towards FTase inhibition (Table 4) but prevented farnesylation of the cysteine thiol (Figure 15). None of the peptidomimetics of L
the invention is metabolically-inactivated by FTase (Figure 15). Thus, although A~X tripeptides are not necessary for potent FTase inhibition, they appear to be required for farnesylation.
With reference to Figure 15, it is to be noted that the transfer of [3H] farnesyl to peptides and peptidomimetic by FTase was determined by silica G TLC as described below.
FPP, F-peptide, and ORIGIN designate farnesyl pyrophosphate, farnesylated peptide and origin, respectively. Figure 15 shows: Lane 1, FPP only;
lane 2, FPP and CVLS but no FTase; lane 3, FPP and FTase but not peptide. Lanes 4-9 all contained FTase and FPP with lane 4, CVIM; lane 5, CVLS;
lane 6, compound 2a; lane 7, compound 4; lane 8, compound 5; lane 9, compound 8. The results shown indicate that the compounds of the invention are not farnesylated in contrast to the CAAX
compounds. Data given are representative of two independent experiments.
The foregoing results show that the novel peptidomimetics described herein have two very important features, namely, they are potent FTase inhibitors, and they are resistant to metabolic inactivation by FTase. Another important feature is that the present compounds inhibit Ras processing in whole cells. This is shown by the following with reference to Figure 16 which illustrates Ras and RaplA processing. To this end, Ras transformed 3T3 cells were treated with inhibitors, lysed and the lysate A) CA 022072~2 1997-06-06 W O 96/21456 PCTrUSg6/01559 ;mmllnoprecipitated with anti-Ras antibody or B) separated by SDS-PAGE. Immunoprecipitates from A) were separated by SDS-PAGE and blotted with anti-Ras antibody whereas samples from B) were blotted with anti-RaplA antibody as described hereafter.
9 Figure 16 shows: Lane 1, control; lane 2, lovastatin; lane 3, reduced 2a (200 ~M); lane 4, 4 (100 ~M); lane 5, 4 (50 ~M); lane 6, 4 (25 ~M);
lane 7, 5; lane 8, 8. Data are representative of 3 independent experiments. Farnesylated Ras runs faster than unprocessed Ras on SDS-PAGE (23-25, 28, 29). Figure 16A (lane 1) shows that cells treated with vehicle contain only processed Ras whereas cells treated with lovastatin (lane 2) contained both processed and unprocessed Ras indicating that lovastatin inhibited Ras processing. Lovastatin~ an HMG-CoA reductase inhibitor which inhibits the biosynthesis of farnesylpyrophosphate and geranylgeranylpyrophosphate, is used routinely as a positive control for inhibition of processing of both geranylgeranylated and farnesylated proteins (36, 37, 39, 40, 51). Cells treated with reduced Cys-4ABA-Met 3 in its free carboxylate forms did not inhibit Ras processing. However, in contrast, the corresponding methyl ester of 2a (200 ~M) inhibited FTase (Figure 16A, lane 3). This is consistent with previous work that showed that neutralization of the carboxylate of CAAX peptides enhances their ability to inhibit Ras processing (37, 40, 51). Although compound 4 has a free carboxylate negative charge, it was able to enter cells and potently inhibit Ras processing (lane 4, 100 ~M compound 4). It was found that compound 4 inhibited Ras processing with concentrations as low as 50 ~M (lane 5), whereas its corresponding parent compound 2a did not inhibit Ras processing CA 022072~2 1997-06-06 W096/21456 PCT~S96/015~9 at concentrations as high as 200 ~M. Compound 4 was as potent as the methylester of its parent compound (2a) (Figure 16A, lane 3). Furthermore, 4 appears to be the first CAAX peptidomimetic that effectively inhibits Ras processing in whole cells directly without relying on cellular enzymes for activation. The hydrophobic character of the biphenyl group apparently compensates for the free carboxylate negative charge thus allowing the peptidomimetic to penetrate membranes and promoting its cellular uptake.
The selectivity of the present Ras farnesylation inhibitors has also been investigated by determining their ability to inhibit processing of RaplA, a small G-protein that is geranylgeranylated (49, 50). Cells were - treated with lovastatin or peptidomimetics exactly as described for Ras processing experiments.
Lysates were then separated by SDS-PAGE and immunoblotted with anti-RaplA antibody as described below. Control cells contained only the geranylgeranylated RaplA (Figure 16B, lane 1) whereas lovastatin-treated cells contained both processed and unprocessed forms of RaplA
indicating, as expected, that lovastatin inhibited the processing of RaplA (Figure 16B, lane 2).
Compound 4, which inhibited Ras processing, was not able to inhibit RaplA geranylgeranylation (Figure 16B, lanes 4-6). Compounds 5 and 8 also did not inhibit RaplA processing (Figure 16B, lanes 7 and 8).
Structures for a number of compounds of the invention are summarized in Table 3, and their relative effectiveness in inhibiting FTTase and GGTase shown in Table 4. The activity of the inhibitors is reported in Table 4 as IC50 values, the concentration at which FTase or GGTase I

CA 022072~2 l997-06-06 W O 96/21456 PCTrUS96/01559 activity was inhibited by 50~. Some of the inhibitors were further characterized for their ability to serve as substrates for farneylsation by thin layer chromatography, as shown in Table 4 (41).
The published sequence dependence studies on FTase have shown a strong preference for methionine in the terminal position of CAAX. In the compounds of the present invention, no methionine residue is present and the tripeptide AAX is completely replaced by a simple hydrophobic moiety. The most potent inhibitor in the CAAX
series is Cys-Ile-Phe-Met (18, 22) with an ICso value of 30 nM. Peptidomimetic inhibitor 10 is as potent as CIFM despite the large difference between their structures. These results confirm the hydrophobic strate~r for AAX replacer~,ent according to the invention.

As previously reported, the incorporation of an aromatic amino acid into the A2 position of CA1A2X (such as CIFM) prevents the tetrapeptide from serving as a substrate for farneyslation (22). Table 4 shows that the designed non-peptide CAAX mimetics (such as compound 4) are not substrates for farnesylation. This lack of farnesylation by FTase may be due to the inhibitor binding to the enzyme in a conformation that does not permit farnesyl transfer to the thiol group.

III. Geranylgeranyl tranfera~e Inhibitors The carboxyl ~erminal CAA~ tetrapeptide of Ras is a substrate for FTase and serves as a target for designing inhibitors of this enzyme with potentiai anticancer activity (33). Our earlier application describes a highly potent (ICso=500 pM) inhibitor of FTase, FTI-276 (Fig. 17) CA 022072~2 1997-06-06 WO96/21456 PCT~S96/01559 (66). Its cell-permeable methyl ester FTI-277 inhibits H-Ras processing in whole cells with an IC50 of 100 nM (66). Furthermore, FTI-276 is highly selective (100-fold) for FTase over GGTase I (Table 5).

In Vitro ~ICso, nM) In Vivo ProcP~sin~ (ICso, uM) FTase GGTase I H-Ras K-Ras RaplA
0 FTI-276 0.5 50 FTI-277 0.1 10 50 GGTI-287 25 5 _GGTI-286 ~30 2 2 GGTI-297 270 40 GGTI-298 ~20 3 3 Sy~thesis of FTase and GGTase I Inhibi tors Peptidomimetics FTI-276 and FTI-277 were prepared as described above. GGTase I inhibitors GGTI-287 and GGTI-286 were prepared from 2-phenyl-4-nitrobenzoic acid (66) by reaction with L-leucine methyl ester followed by reduction with stannous chloride. The resulting 4-amino-2-phenylbenzoyl leucine methyl ester was reacted with N-Boc-S-trityl-cysteinal and deprotected by procedures similar to those described for the FTase inhibitors (66) to give GGTI-286 and GGTI-287 as their hydrochloride salts.

A. 4-Amino-2-phenylbenzoyl-(S)-methionine methyl ester hydrochloride To a mixture of 70 mL of acetone and 85 mL of water was added 2-bromo-4-nitrotoluene 6.84 g (30 mmol), phenylboronic acid 3.84 g (31.5 mmol), potassium carbonate 10.35 g (75 mmol) and palladium acetate 336 mg (1.5 mmol). The mixture was refluxed for 10 hr and then extracted with ether and dilute hydrochloric acid. After evaporating solvents, the solid residue was recrystallized from methanol to give 5.64 g of 4-CA 022072~2 1997-06-06 WO96/21456 PCT~S96/01559 nitro-2-phenyltoluene (88~ yield). lH NMR (CDC13) 8.09-8.11 (m, 2H), 7.40-7.49 (m, 4H), 7.30-7.33 (m, 2H), 2.37 (s, 3H).
The above 4-~itro-2-phenyltoluene (4.4~ g, 21 mmol) was suspended in 21 mL of pyridine and 42 mL
J of water. The mixture was heated to boiling followed by addition of potassium permanganate (19.8, 126 mmol). The mixture was refluxed for 2 hr and then filtered to remove the solids. The filtrate was acidi.fied with 6N HC1 to give 4.48 g of 4-nitro-2-phenylbenzoic acid (89~ yield). lH
NMR (CDC13) ~ 8.25-8.33 (m, 2H0, 8.08 (d, 8.9 Hz, lH), 7.41-7.51 (m, 3H), 7.31-7.39 (m, 2H).
The above 4-nitro-2-phenylbenzoic acid (2.43 g, 10 mmol) was suspended in 50 mL of methylene chloride. To this solution was added (L)-methionine methyl ester hydrochloride (2.0 g, 10 mmol), triethylamine (1.38 mL, 10 mmol), 1-(3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride (EDCI, 2.01 g, 10.5 mmol), 1-hydroxybenzotriazole (HOBT, 1.35 g, 10 mmol). The mixture was stirred for 12 hr and then extracted with methylene chloride and lN hydrochloric acid.
After the evaporation of solvents, the residue was recrystallized from ethyl acetate and hexane to give 3.22 g of 4-nitro-2-phenylbenzolyl-(L)-methionine methyl ester (yield 83~). lH NMR
(CDCl3) ~ 8.24-8.28 (m, 2H), 7.85 (d, 8.9 Hz, lH), 7.43-7.52 (m, 5H), 6.01 (d, 7.5 Hz, lH), 4.69 (ddd, lH), 3.68 (s, 3H), 2.05 (m, 2H), 1.98 (s, 3H), 1.88-1.96 (m, lH), 1.72-1.81 (m, lH).
The 4-nitro-2-phenylbenzoyl-(L)-methionine methyl ester (3.04 g, 7.83 mmol) was dissolved into 100 mL of ethyl acetate followed by the addition of stannous chloride hydrate (8.84 g, 39 mmol). The mixture was refluxed for 2 hr and then extracted with a mixture of ethyl acetate and CA 022072~2 1997-06-06 W O 96/21456 PCTrUS96/01559 concentrated sodium bicarbonate. After the evaporation of solvents, the residue was dissolved in methylene chloride followed by addition of 3N
hydrogen chloride in ether. The solid was <
filtered and dried to give 2.96 g of 4-amino-2-phenylbenzoyl-(L)-methionine methyl ester hydrochloride (yield 96~). lH NMR (CD30D) ~ 7.65 (d, 8.1 Hz, lH), 739-7.46 (m, 7H), 4.53 (dd, 4.3 and 9.5 Hz, lH), 3.69 (s, 3H), 2.15-2.23 (m, lH), 2.00 (s, 3H), 1.93-2.11 (m, 2H), 1.74-1.83 (m, lH); 13C NMR (CD30D) ~ 173.4, 171.7, 143.4, 140.1, 137.4, 134.0, 130.9, 129.7, 129.4, 125.5, 122.7, 53.0, 52.9, 31.3, 30.9, 15.1.

B. 4-~2(R)-tert-butoxycarbonylamino-3-triphenylmethylthiopropyl~amino-2-phenylbenzoyl-(S)-methionine methyl ester To a mixture of 4-amino-2-phenylbenzoyl-(S)-methionine methyl ester hydrochloride (1.27 g, 3.22 mmol) in 20 mL of methanol was added N-Boc-S-trityl-(L)-cysteinal (1.0 eq, according to lH NMR
determination of aldehyde percentage) and sodium cyanoborohydride (400 mg, 2.0 eq). The mixture was stirred for 12 hr. After the evaporation of solvents, the residue was extracted with ethyl acetate and concentrated sodium bicarbonate.
After removing solvents, the residue was purified through flash column chromatography (1:1 = hexane:
ethyl acetate, silica) to give the product 1.67 g (yield 67~). lH NMR (CDC13) ~ 7.65 (d, 8.6 Hz, lH), 7.34-7.42 (m, llH), 7.18-7.29 (m, 9H), 6.52 (dd, 2.3 and 8.1 Hz, lH), 6.34 (d, 2.3 Hz, lH), 5.65 (d, 7.7 Hz, lH), 4.64 (ddd, lH), 4.55 (d, 8.1 Hz, lH), 4.19 (br t, lH), 3.78 (br m, lH), 3.64 (s, 3H), 3.09 (t, 6.1 Hz, 2H), 2.44 (m, 2H), 2.04-2.10 (m, 2H), 2.00 (s, 3H), 1.81-1.90 (m, lH), 1.60-1.70 (m, lH), 1.41 (s, 9H)i 13C NMR (CDCl3) CA 022072~2 1997-06-06 WO96/21456 PCT~S96/01559 172.0, 168.3, 155.7, 149.4, 144.3, 141.6, 141.1, 131.3, 129.5, 128.7, 128.5, 127.9, 127.7, 126.8, 122.6, 113.6, 111.3, 79.8, 67.1, 52.2, 51.7, 49.5, 47.2, 34.3, 31.6, 29.4, 28.2, 15.2.

C. 4-t2(R)-amino-3-mercaptopropyl~amino-2-phenylbenzoyl-(S)-methionine methyl eRter hydrochloride The above N-Boc-S-trityl protected peptide methyl ester (900 mg) was dissolved in 5 mL of methanol. To this mixture was added a solution of mercuric chloride (774 mg, 2.50 eq) in 5 mL of methanol. The mixture was refluxed for 20 min.
The precipitate was collected and dried. This solid was suspended in 10 mL of methanol and reacted with gaseous hydrogen sulfide. After the removal of black solid, the clear solution was evaporated to dryness. The residue was then dissolved in methylene chloride followed by addition of 3N hydrogen chloride in ether. The white solid was collected and dried to give the pure product 476 mg ~yield 81~). lH NMR (CD30D) 7.42 (d, 8.4 Hz, lH), 7.30-7.38 (m, 5H), 7.77 (d, 8.4 Hz, lH), 6.71 (s, lH), 4.48 (dd, 4.2 and 5.1 Hz, lH), 3.68 (s, 3H), 3.44-3.58 (m, 3H), 2.90-2.95 (dd, 4.1 and 14.5 Hz, lH), 2.79-2.85 (dd, 4.7 and 14.5 Hz, lH), 2.18-2.22 (m, lH), 2.03-2.16 (m, lH), 2.00 (s, 3H), 1.91-1.97 (m, lH), 1.73-1.82~(m, lH); 13C NMR (CD3OD) ~ 173.7, 173.4, 150.7, 143.5, 142.3, 131.2, 129.8, 129.5, 128.6, 125.6, 115.~, 112.2, 53.7, 53.2, 52.8, 45.0, 31.4, 30.9, 25.3, 15Ø
D. 4-t2(R)-amino-3-mercaptopropyl]amino-2-phenylbenzoyl-(S)-methionine The N-Boc-S-trityl protected peptide methyl ester (500 mg) was hydrolyzed with 2.0 eq of lithium hydroxide at 0~C for 1 hr. The product CA 022072~2 1997-06-06 W O 96/21456 PCTrUS96101559 was deprotected with trifluoroacetic acid (2 mL) in methylene chloride (1 mL). Triethylsilane was added dropwise until the deep yellow color disappeared. The mixture was kept at r.t. for 1.5 hr. After the evaporation of solvents, the residue was dried and washed with dry ether. The solid was purified through preparative HPLC to give a pure product 270 mg (yield 78~ H NMR
(CD30D) ~ 7.44 (d, 8.4 Hz, lH), 7.30-7.39 (m, 5H), 6.75 (d, 8.4 Hz, lH), 6.67 (s, lH), 4.45 (dd, 4.2 and 5.1 Hz, lH), 3.42-3.58 (m, 3H), 2.90 (dd, 4.3 and 14.5 Hz, lH), 2.81 (dd, 5.5 and 14.5 Hz, lH), 2.17-2.23 (m, lH), 2.09-2.15 (m, lH), 2.00 (s, 3H), 1.90-1.99 (m, lH), 1.71-1.81 (m, lH); 13C NMR
(CD30D) ~ 176.4, 173.5, 150.4, 143.0, 141.5, 131.0, 129.7, 129.4, 128.9, 124.6, 115.0, 112.3, 53.3, 49.6, 44.4, 30.8, 30.1, 24.9, 14.8.

E. 4-Nitro-2-phenylbenzoyl-(S)-leucine methyl ester This compound was prepared through the coupling of 4-nitro-2-phenylbenzoic acid with (L)-leucine methyl ester hydrochloride as for the preparation of the methionine derivative (see Example 26, section A). lH NMR (CDCl3) ~ 8.24-8.26 (m, 2H), 7.86 (d, 8.7 Hz, lH), 7.41-7.46 (m, 5H), 5.71 (d, 7.4 Hz, lH), 4.57 (ddd, lH), 3.67 (s, 3H), 1.37-1.46 (m, lH), 1.08-1.25 (m, 2H), 0.78 (dd, 6H).

F. 4-[2(R)-tert-butoxycarbonyl-3-triphenylmethylthiopropyl]amino-2-phenylbenzoyl-(S)-leucine methyl ester This compound was prepared using the same method as for the preparation of methionine derivative (See Example 26, section B), using 4-amino-2-phenylbenzoyl-(S)-leucine methyl ester and N-Boc-S-trityl-(L)-cysteinal as starting CA 022072~2 1997-06-06 WO96/21456 PCT~S96/01559 materials. lH NMR (CDCl3) ~ 7.68 (d, 8.6 Hz, lH), 7.33-7.41 (m, llH), 7.17-7.29 (m, 9H), 6.50 (d, 8.6 Hz, lH), 6.31 (s, lH), 5.43 (d, 7.8 Hz, lH), 4.60 (d, 6.1 Hz, lH), 4.47 (ddd, lH), 4.19 (br t, lH), 3.77 (br m, lH), 3.62 (s, 3H), 3.09 (t, 5.9 Hz, 2H), 2.45 (br m, 2H), 1.40 (s, 9H), 1.27-1.33 (m, lH), 1.03-1.18 (m, 2H), 0.75 (dd, 6H); 13C
(CDCl3) ~ 173.2, 168.2, 155.6, 149.4, 144.4, 141.7, 141.2, 131.4, 129.5, 128.8, 128.5, 127.9, 127.6, 126.8, 122.7, 113.6, 111.3, 79.6, 67.1, 51.9, 50.9, 49.5, 47.1, 41.2, 34.3, 28.3, 24.4, 22.7, 21.8.

G. 4-[2(R)-amino-3-mercaptopropyl]amino-2-phenylbenzoyl-(S)-leucine methyl ester hydrochloride This compound was prepared with the same method as for the preparation of methionine derivative (see Example 26, section C), using 4-[2(R)-tert-butoxycarbonyl-3-triphenylmethylthiopropyl]amino-2-phenylbenzoyl-(S)-leucine methyl ester and mercuric chloride. lH
NMR (CD30D) ~ 7.42 (d, 8.5 Hz, lH), 7.31-7.38 (m, 5H), 6.76 (d, 8.5 Hz, lH), 6.68 (s, lH), 4.33 (t, 7.8 Hz, lH), 3.67 (s, 3H), 3.46-3.55 (m, 3H), 2.95 (dd, 4.4 and 14.5 Hz, lH), 2.81 (dd, 5.1 and 14.5 Hz, lH), 1.44 (t, 7.6 Hz, 2H), 1.18-1.25 (m, lH), 0.76-0.83 (dd, 4.1 and 6.6 Hz, 6H).

H. 4-t2(R)-amino-3-mercaptopropyl]amino-2-phenylbenzoyl-(S)-leucine This compound was prepared with the same method as for the preparation of the methionine derivative (see Example 26, section D), using 4-[2(R)-amino-3-mercaptopropyl]amino-2-phenylbenzoyl-(S)-leucine methyl ester hydrochloride and lithium hydroxide. lH NMR
(CD30D) ~ 7.42 (d, 8.5 Hz, lH), 7.29-7.38 (m, 5H), CA 022072~2 l997-06-06 PCTrUS96/01559 6.73 (d, 8.5 Hz, lH), 6.66 (s, lH), 4.32 (dd, 3.3 and 5.9 Hz, lH), 3.41-3.57 (m, 3H), 2.94 (dd, 4.3 and 14.5 Hz, lH), 2.78 (5.2 and 14.5 Hz, lH), 1.45 (t, 6.7 Hz, 2H), 1.17-1.26 (m, lH), 0.78-0.83 (t, 8.5 Hz, 6H).

I. 4-Nitro-2-naphthylbenzoic acid The coupling of 4-nitro-2-bromobenzoic acid methyl ester (1.92 g, 7.4 mmol) with 1-naphthylboronic acid (2.53 g, 14.7 mmol) in the presence of anhydrous sodium phosphate (3.64 g, 22.2 mmol) and palladium tetrakistriphenylphosphine (426 mg, 0.368 mmol) in 50 mL of DMF at 100~C gave the 4-nitro-2-naphthylbenzoic acid methyl ester (1.66 g, 73~ yield). lH NMR (CDCl3) ~ 8.34 (d, 8.5 Hz, lH), 8.28 (s, lH), 8.14 (d, 8.5 Hz, lH), 7.92 (d, 8.2 Hz, 2H), 7.47-7.56 (m, 2H), 7.41 (d, 3.8 Hz, 2H), 7.34 (d, 6.8 Hz, lH), 3.40 (s, 3H). After the hydrolysis of methyl ester, 1.44 g of product was collected (yield 91~). lH NMR ~CDCl3) ~ 8.33 (d, 8.6 Hz, lH), 8.23 (s, lH), 8.17 (d, 8.6 Hz, lH), 7.88 (d, 8.2 Hz, 2H), 7.46-7.52 (m, 2H), 7.38-7.42 (m, 2H), 7.33 (d, 7.0 Hz, lH); 13C NMR
(CD3COCD3) ~ 167.2, 150.1, 143.1, 139.0, 138.2, 134.4, 132.5, 132.1, 129.2, 127.3, 126.7, 125.8, 125.9, 123.3.

J. 4-Nitro-2-naphthylbenzoyl-(S)-methionine methyl ester The coupling of 4-nitro-2-naphthylbenzoic acid with (L)-methionine methyl ester in the presence of EDCI and HOBT provided the desired product (yield 95~). TLC of the product showed single spot, but lH NMR showed the presence of diastereomers caused by the restricted rotation between naphthyl and phenyl rings. lH NMR ( CDCl3 ) 8.33-8.38 (m, lH), 8.26 (ss, lH), 8.14 (d, 8.5 -CA 022072~2 1997-06-06 W O96/21456 PCTrUS96/01559 Hz, 0.5H), 8.00 (d, 8.5 Hz, 0.5H), 7.94-7.98 (m, 2H), 7.42-7.65 (m, 5H), 5.98 (t, lH), 4.42 (m, lH), 3.56 (s, 1.5~), 3.51 (s, 1.5H), 1.83 (s, 1.5H), 1.74 (s, 1.5H), 1.56-1.64 (m, lH), 1.33-1.45 (m, 2H), 1.09-1.14 (m, lH).

R. 4-~2(R)-tert--butoxycarbonyl-3-triphenylmethylthiopropyl]~ino-2-naphthyl-(S)-methionine methyl e~ter The reduction of 4-nitro-2-naphthylbenzoyl-(L)-methionine methyl ester gave a quantitative yield of the amino derivative which was reacted with N-Boc-S-trityl cysteinal in the presence of sodium cyanoborohydride. After flash column chromatography (1:1 = ethyl acetate : hexane) purification, a desired product was obtained (yield 40~). TLC showed single spot, but lH NMR
showed diastereomers caused by the restricted rotation between the naphthyl and phenyl rings. lH
NMR (CDC13) ~ 7.84--7.96 (m, 3H), 7.49-7.66 (m, 4H), 7.37-7.43 (m, 7H), 7.14-7.27 (m, 9H), 6.60-6.63 (d, 8.6 Hz, lH), 6.33 (m, lH), 5.67 (d, 7.8 Hz, 0.6H), 5.60 (d, 7.8 Hz, 0.4H), 4.56 (br d, 6.2 Hz, lH), 4.35-4.44 (m, lH), 4.30 (br, lH), 3.78 (br m, lH), 3.55 (s, l.9H), 3.38 (s, l.lH), 3.06 (t, 5.8 Hz, 2H), 2.44 (m, 2H), 1.90 (s, lH), 1.79 (s, 2H), 1.57-1.68 (m, 0.5H), 1.36-1.45 (m, lOH), 1.23-1.32 (m, 2H), 0.94-0.98 (m, 0.7H).

L. 4-t2(R)-amino--3-mercaptopropyl]amino-2-naphthylbenzoyl-(S)-methionine This compound was prepared from the N-Boc-S-trityl protected form (section K) by saponi~ication followed with acidic cleavage by trifluoroacetic acid. The pure compound was obtained through preparative HPLC. lH NMR showed complicated diastereomers caused by the restricted CA 022072~2 1997-06-06 W O96/21456 PCT~US96/01559 rotation of aryl-aryl bond. lH NMR (CD30D) 7.86-7.94 (m, 2H), 7.73 (d, 8.6 Hz, 0.6H), 7.35-7.67 (m, 5.4H), 6.83-6.88 (m, lH), 6.63-6.67 (m, lH), 4.17-4.23 (m, lH), 3.41-3.58 (m, 3H), 2.91 (dd, 4.2 and 14.5 Hz, lH), 2.80 (dd, 5.3 and 14.5 Hz, lH), 1.82 (s, 1.4H), 1.80 (s, 1.6H), 1.65-1.77 (m, lH), 1.41-1.52 (m, 2H), 1.09-1.32 (m, lH).

M. 4-Nitro-2-naphthylbenzoyl-(S)-leucine methyl ester This compound was prepared with the same method as for the preparation of the methionine derivative (section J) using 4-nitro-2-naphthylbenzoic acid, (S)-leucine methyl ester, EDCI and HOBT. lH NMR (CDC13) ~ 8.34-8.39 (m, lH), 8.25 (s, lH), 8.18 (d, 8.6 Hz, 0.6H), 8.02 (d, 8.6 Hz, 0.4H), 7.91-8.00 (m, 2H), 7.62 (t, 7.0 Hz, 0.6H), 7.48-7.58 (m, 3H), 7.41 (t, 7.0 Hz, 1.4H), 5.71 (d, 7.9 Hz, 0.6H), 5.60 (d, 7.9 Hz, 0.4H), 4.29 (m, lH), 3.57 (s, 1.7H), 3.52 (s, 1.3H), 1.05-1.11 (m, 0.5H), 0.88-0.97 (m, 0.7H), 0.69-0.78 (m, 0.5H), 0.41-0.59 (m, 7.0H), 0.19-0.26 (m, 0.6H).

N. 4-[2(R)-tert-butoxycarbonylamino-3-triphenylmethylthiopropyl]amino-2-naphthylbenzoyl-(S)-leucine methyl ester This compound was prepared with the same method as for the preparation of the methionine derivative (section K). lH NMR (CDCl3) ~ 7.85-8.00 (m, 3H), 7.47-7.67 (m, 4H), 7.39-7.43 (m, 7H), 7.14-7.37 (m, 9H), 6.61 (d, 8.6 Hz, lH), 6.32 (s, lH), 5.46 (d, 7.6 Hz, 0.6H), 5.36 (d, 7.6 Hz, 0.4H), 4.55 (d, 7.2 Hz, lH), 4.20-4.27 (m, 2H), 3.76 (br, lH), 3.56 (s, 2H), 3.38 (s, lH), 3.06 (t, 5.9 Hz, 2H), 2.43 (m, 2H), 1.36-1.43 (m, 9H), CA 022072~2 1997-06-06 W O 96/21456 PCTrUS96/01559 0.81-1.03 (m, lH), 0.55-0.67 (m, 2.8H), 0.36-0.45 (m, 4.7H), 0.00-0.09 (m, 0.6H).

O. 4-[2(R)amino--3-mercaptopropyl]amino-2-naphthylbenzoyl-(S) -leucine methyl ~ 5 ester This compound was prepared from the N-Boc-S-trityl methyl ester of the corresponding compound, using the method of section L. lH NMR
(CDCl3) ~ 7.98 (d, 8.5 Hz, 0.6H), 7.84-7.90 (m, 2.4H), 7.65 (d, 8.5 Hz, 0.4H), 7.43-7.58 (m, 3.6H), 7.34-7.39 (m, lH), 6.72 (m, lH), 6.45 (ss, lH), 5.46 (d, ~.8 Hz, 0.6H), 5.40 (d, 7.7 Hz, 0.4H), 4.64 (m, lH), 4.23 (m, lH), 3.54 (s, 2H), 3.30 (s, lH), 3.25 (m, lH), 2.97-3.06 (m, 2H), 2.67 (dd, 3.7 and 13.1 Hz, lH), 2.47 (dd, 6.5 and 13.2 Hz, lH), 1.45-1.65 (br s, 2H), 0.81-1.03 (m, 1.2H), 0.54-0.67 (m, 3H), 0.36-0.39 (m, 4.3H), 0.00-0.10 (m, 0.7H).

EXAMP~E 27 FTase and GGTase I Activity Assay FTase and GGTase I activities from 60,000 x g supernatants of human Burkitt lymphoma (Daudi) cells (ATCC, Rockville, MD) were assayed exactly as described previously for FTase (41).
Inhibition studies were performed by determ; n; ng the ability of Ras CAAX peptidomimetics to inhibit the transfer of 13H]-farnesyl and 13H]-geranylgeranyl from [3H]FPP and [3H]GGPP to H-ras-CVLS and H-Ras-CVLL, respectively (41).

Ras and RapIA Processing Assay H-Ras cells (45) and K-Ras4B Cells (32) were kind gifts from Dr. ~h~nn; ng Der and Dr. Adrienne Cox (University of North Carolina, Chapel Hill).

CA 022072~2 1997-06-06 W O96t21456 PCTrUS96/01559 Means of obtaining these cell lines will be easily recognized by the skilled practitioner.
Cells were seeded on day 0 in 100 mm dishes in Dulbecco's modified Eagles medium supplemented with 10~ calf serum and 1~ penicillin-streptomycin. On days 1 and 2, cells were refed with medium containing various concentrations of FTI-277, GGTI-286 or vehicle (10 mM DTT in DMSO).
On day 3, cells were washed and lysed in lysis buffer containing 50 mM HEPES, pH 7.5, 10 mM NaCl, 1~ TX-100, 10~ glycerol, 5 mM MgCl2, 1 mM EGTA, 25 ~g/ml leupeptin, 2 mM PMSF, 2 mM Na3VO4, 1 mg/ml soybean trypsin inhibitor! 10 ~g/ml aprotinin, 6.4 mg/ml Sigma-104~ phosphatase substrate. Lysates were cleared (14,000 rpm, 4~C, 15 min) and equal amounts of protein were separated on a 12.5% SDS-PAGE, transferred to nitrocellulose, and immunoblotted using an anti-Ras antibody (Y13-259, ATCC) or an anti-RapIA antibody (SC-65, Santa Cruz Biotechnology, Santa Cruz, CA). Antibody reactions were visualized using either peroxidase-conjugated goat anti-rat lgG (for Y13-259), or peroxidase-conjugated goat anti-rabbit lgG (for RaplA) and an enhanced chemiluminescence detection (ECL, Amersham Corp.), as described previously (41).

M~P ~inase Tmm~7nohlotting Cells were treated with FTI-277, GGTI-286, or vehicle and lysed as previously described for Ras and RaplA processing. Equal amounts of protein were separated on a 15~ SDS-PAGE, transferred to nitrocellulose, and immunoblotted using an anti-MAP kinase antibody (erk2, monoclonal, UB1, Lake Placid, NY). Antibody reactions were visualized using peroxidase-conjugated donkey anti-mouse IgG

CA 022072~2 1997-06-06 W O96/21456 PCTrUS96/01559 (Jackson ImmunoResearch Laboratories Inc., West Grove, PA) and an enhanced chemiluminescence detection system (ECL, Amersham Corp.) ..
EXAMP~E 30 Tnh i hi tion of GGTase I by GGTI-286 GGTI-287 potently inhibited GGTase I in vitro (IC50=5 nM) and was selective towards inhibiting GGTase I over FTase (ICso=25 nM) (Table 5). Thus, the substitution of methionine in FTI-276 by a leucine in GGTI-287 (Fig. 17) increased the potency towards GGTase I by approximately 10-fold (Table 5). More importantly, it reversed the selectivity from a FTase to a GGTase I-specific inhibitor by a factor of 500 (Table 5). To determine whether this selectivity is respected in whole cells, the cell-permeable methyl ester derivative of GGTI-287, GGTI-286 (Fig. 17), was synthesized and used to treat NIH 3T3 cells which overexpress oncogenic H-Ras-CVLS (31). Cell lysates were electrophoresed on SDS-PAGE and immunoblotted with an anti-Ras antibody as described in Example 28. Figure 18 shows that accumulation of unprocessed H-Ras did not occur at concentrations lower than 30 ~M GGTI-286.
Therefore, GGTI-286 is not a good inhibitor of H-Ras processing in whole cells. However, GGTI-286 was a very potent inhibitor of the processing of the geranylgeranylated RaplA protein (IC50=2 ~M) (Fig. 18). Thus, GGTI-286 is more than 15-fold selective for inhibition of geranylgeranylation over farnesylation processing (Table 5). This data is in direct contrast to the FTase specific inhibitor FTI-277 which inhibited H-Ras and RapIA
processing with IC50s of 100 nM and 50 ~M, respectively (Fig. 18). Thus, GGTI-286 is 25-fold CA 022072~2 1997-06-06 W O 96/21456 PCTrUS96/01559 more potent than FTI-277 at inhibiting geranylgeranylation in whole cells (Table 5).

Tnh i h; tion of GGTase I by ~GTI-297 and GGTI-298 To determine the effect of replacing the phenyl substituent with a naphthyl on GGTase I
inhibition, 4-[2(R)-amino-3-mercaptopropyl] amino-2-naphthyl benzoyl-(L)-leucine (GGTI-297) and its methylester (GGTI-298) were tested. GGTI-297 inhibited GGTase I in vitro with an ICso of 40 nM
and was selective towards inhibiting GGTase I over FTase ( ICso = 270 nM) (Fig. 21, Table 5). Thus, the substitution of phenyl in GGTI-287 by naphthyl in GGTI-297 decreased the potency towards GGTase I by 8 fold and towards FTase by over 10-fold.
However, more importantly, the selectivity for GGTase I over FTase increased from 5-fold (GGTI-287) to 7-fold (GGTI-297), as shown in Table 5.
This selectivity was also respected in vivo since concentrations as high as ~0 ~M did not inhibit H-Ras processing whereas RaplA and Ras4B were completely blocked at 10 ~M GGTI-298 (Table 5).

Inhibi tion of R-Ras4B Function by GGTI-286 The ability of GGTI-286 to inhibit the processing and signaling of oncogenic K-Ras4B was then evaluated. NIH 3T3 cells which overexpress oncogenic K-Ras4B (32) were treated either GGTI-286 (0-30 ~M) or FTI-277 (0-30 ~M) and the lysates were immunoblotted with an anti-Ras antibody as described under Example 28. Figure 19 shows that GGTI-286 inhibited potently the processing of K-Ras4B with an ICso of 2 ~M. The ability of GGTI-286 to inhibit the processing of K-Ras4B was much closer to its ability to inhibit the processing of - - -CA 022072~2 1997-06-06 WO96121456 PCT~S96/01559 geranylgeranylated RaplA (IC50=2 ~M) than that of farnesylated H-Ras (IC50~30 ~M)(Fig. 18) (Table 5).
This suggested that K-Ras4B might be geranylgeranylated. Consistent with this is the fact that K-Ras4B processing was very resistant to the FTase-specific inhibitor FT-277 (IC50=lO ~M) (Fig. l9). Furthermore, GGTI-286 inhibited K-Ras4B processing at concentrations (1-3 ~M) (Fig.
l9) that had no e~fect on the processing of farnesylated H-Ras (Fig. 18).

Effects of GGTI-2~6 on Oncogenic ~-Ras 4B
Constituti~re Activation of ~P Rinase To determine whether inhibition of K-Ras4B
processing by GGTI-286 results in disruption of oncogenic signaling, the ability of GGTI-286 to antagonize oncogenic K-Ras 4B constitutive activation of MAP kinase was e~m;ned. Activated MAP kinase is hyperphosphorylated and migrates slower than hypophosphorylated (inactive) MAP
kinase on SDS-PAGE (43, 66~. Figure 20 shows that K-Ras4B transformed cells contained mainly activated MAP kinase. Treatment of these cells with the FTase-specific inhibitor FTI-277 (0-30 ~M) did not inhibi~ MAP kinase activation until 30 ~M (Fig. 20). In contrast, GGTI-286 inhibited MAP
kinase activation with an IC50 of l ~M and the block was complete at lO ~M. Thus, GGTI-286 blocked oncogenic K-Ras4B MAP kinase activation at a concentration (lO ~M) where FTI-277 had no effect. In contrast, oncogenic H-Ras activation of MAP kinase was inhibited only slightly by GGTI-286 whereas FTI-277 completely blocked this activation at 3 ~M (Fig. 20). Furthermore, GGTI-286 blocked K-Ras4~ activation of MAP kinase at a CA 022072~2 l997-06-06 W O 96/21456 PCTrUS96/01559 concentration (10 ~M) that had little effect on H-Ras activation of MAP kinase (Fig. 20).

Anti tumor Efficacy The above examples demonstrate that GGTI-286 is a potent and highly selective inhibitor of K-Ras4B processing and activation of oncogenic signalling. In order to demonstrate the efficacy of these inhibitors as anticancer agents, K-Ras4B
transformed NIH-3T3 cells were implanted subcutaneously in nude mice. When the tumors reached sizes of 50-100 mm3, the mice were randomly separated into control and treated groups (5 animals per group, each animal had a tumor on both the right and the left flank). Figure 22 shows that tumors from control ~n;m~l S treated with saline once daily grew to an average size of 2900 mm3 over a period of two weeks. In contrast, tumors from animals treated once daily with GGTI-286 (25 mg/kg or 50 mg/kg) grew to a size of 1600 mm3 or 900 mm3, respectively (Fig. 22). Thus, GGTI-286 inhibited tumor growth by 50~ and 70~, respectively.
In summary, the data clearly identifies GGTI-286 not only as a potent antagonist of K-Ras4B
oncogenic signaling in cultured cells, but also as an inhibitor of tumor growth in whole animals.
References cited herein are listed below for convenience and are hereby incorporated by reference. ~=

-CA 022072~2 1997-06-06 WO96/21456 PCT~S96101559 R~ ~S

1. Barbacid) M., Annu. Rev. Biochem., 56:779-828, (1987) 2. Grand, R.J.A. and Owen, D., Biochem. J., 279:609-631, (1991) 3. Barbacid, M., Important Advances in Oncoloqy, eds. Devita, Hellman &
Rosenberg (Lippincott, Philadelphia, PA), pp. 3-22, (1986) 4. Mulcahy, L.S., Smith, M.R., and Stacey, D.W., Nature, 313:241-243, (1985) 5. Noda, M., Ko, M., Ogura, A., Liu, D.G., Amano, T., Takano, T. and Ikawa, Y., Nature, 318:73-75, (1985) 6. Bar-Sagi, D. and Feramisco, J.R., Cell, 42:841-8~8, (1985) 7. Kat~oka; T~; ~owers, S~, McG~ll, C., Fasano, O., Strathern, J., Broach, J.
and Wigler, M., Cell, 37:437-445, (1984) 8. Willumsen, B.M., Christensen, A., Hubbert, N.C., Papageorge, A.G. and Lowy, D.R., Nature, 310:583-586, (1984) 9. Willumsen, B.M., Norris, K., Papageorge, A.G., Hubbert, N.C. and Lowy, D.R., EMBO
J., 3:2581-2585, (1984) 10. Hancock, J.F., Magee, A.I., Childs, J.E.
and Marshall, C.J., Cell, 57:1167-1177, (1989) 11. Gutierrez, L., Magee, A.I., Marshall, C.J. and Hancock, J.F., EMBO J, 8:1093-1098, (1989) 12. Casey, P.J., Solski, P.A., Der, C.J. and Buss, J.E., Proc. Natl. Acad. Sci. USA, 86:8323-8327, (1989) 13. Jackson, J.H., Cochrane, C.G., Bourne, J.R., Solski, P.A., Buss, J.E. and Der, C.J., Proc. Natl. Acad. Sci. USA, 87:3042-3046, (1990) 14. Hancock, J.F., Paterson, H. and Marshall, J.C., Cell, 63:133-139, (1990) CA 022072~2 1997-06-06 W O96121456 PCTrUS96/01559 15. Reiss, Y., Goldstein, J.L., Seabra, M.C., Casey, P.J. and Brown, M.S., Cell, 62:81-88, (1990) 16. Reiss, Y., Seabra, M.C., Goldstein, J.L.
and Brown, M.S., METHODS: A Companion to Methods in EnzYmolo~Y, 1:241-245, (1990) 17. Reiss, Y., Seabra, M.C., Armstrong, S.A., Slaughter, C.A., Goldstein, J.L.
and Brown, M.S., J. Biol. Chem., 266:10672-10877, (1991) 18. Reiss, Y., Stradley, S.J., Gierasch,-L.M., Brown, M.S. and Goldstein, J.L., Proc. Natl. Acad. Sci. USA, 88:732-736, (1991) 19. Manne, V., Roberts, D., Tobin, A., O'Rourke, E., DeVirgillio, M., Meyres, C., Ahmed, N.,-Kurz, E., Resh, M., Kung, H.F. and Barbacid, M., Proc. Natl. Acad.
Sci. USA, 87:7541-7545, (1990) 20. Gibbs, J.B., Cell, 65:1-4, (1991) 21. Moores, S.L., Schaber,M.D., Mosser, S.D., Rands, E., O'Hara, M.B., Garsky, V.M., Marshall,M.S., Pompliano, D.L. and Gibbs, J.B., J. Biol. Chem., 266:14603-14610, (1991) 22. Goldstein, J.L., Brown, M.S., Stradley, S.J., Reiss, Y. and Gierasch, L.M., J.
Biol. Chem., 266:15575-15578, (1991) 23. Brown, M.S., Goldstein, J.L., Paris, K.J., Burnier, J.P. and Marsters, J.C., Proc. Natol. Acad. Sci. USA, 89:8313-8316, (1992) 24. Pompliano, D.L., Rands, E., Schaber, M.D., Mosser, S.D., Neville, J.A. and Gibbs, J.B., Biochemistry, 31:3800-3807, (1992) 25. Lacal, J.D., Santos, E., Notario, V., Barbacid, M., Yamazaki, S., Kung, H.F., Se~m~n.q, O., McAndrew, S. and Crowl, R., Proc. Natl. Acad. Sci. USA, 81:5305-5309, (1984) 26. Stewart, F.H.C., Aus. J. Chem., 36:2511, (1983) CA 022072~2 1997-06-06 WO96/21456 PCT~S96101559 27. Brown, M.J., Milano, P.D., Lever, D.C., Epstein, W.W. and Poulter, C.D., J. Am.
Chem. Soc., 113:3176, (1991) 28. Yang, C.C., Marlowe, C.K. and Kania, R., J. Am. Chem. Soc., 113:3177, (1991) 29. McCormick, F. (1993) Nature 363:15-16.
30. McCormick, F. (1994) Current Opinion in Genetics & Development 4:71-76 31. Marsha].l, C.J. (1994) Current Opinion in Genetics & Development 4: 82-89 32. Kato, K., Cox, A.D., Hisaka, M.M., Graham, S.M., Buss, J.E., and Der, C.J.
(1992) Proc. Natl. Acad. Sci. USA
89:6403-6407.
33. Gibbs, J.B., Oliff, A., and Kohl, N.E.
(1994) Cell 77:175-178.
34. Nigam, M., Seong, C., Qian, Y., Hamilton, A.D., and Sebti, S. M. (1993) J. Biol. Chem. 268:20695-20698.
35. Qian, Y., Blaskovich, M.A., Saleem, M., Seong, C., Wathen, S.P., Hamilton, A.D., and Sebti, S.M. (1994) J. Biol. Chem.
269:12410-12413.
36. Qian, Y., Blaskovich, M.A., Seong, C.M., Vogt, A., Hamilton, A.D., and Sebti, S.M. (1994) Bioorq. Med. Chem. Lett 4:2579-2584.
37. Kohl, N.E., Mosser, S.D., deSolms, S.J., Giuliani, E.A., Pompliano, D.L., Graham, S.L., Smith, R.L., Scolnick, E.M., Oliff, A., and Gibbs, J.B. (1993) Science 260:1934-1937.
38. James, G.L., Goldstein, J.L., Brown, M.S., Rawson, T.E., Somers, T.C., McDowell, R.S., Crowley, C.W., Lucas, B.K., Levinson, A.D., and Marsters, J.C., Jr. (1993) Science 260:193-194.
39. Graham, S.L., deSolms, S.J., Giuliani, E.A., Kohl, N.E., Mosser, S.D., Oliff, A.I., Pompliano, D.L., Rands, E.
Breslin, M.J., Deana, A.A., Garsky, r V.M., Scholz, T.H., Gibbs, J.B., and CA 022072~2 1997-06-06 W O 96121456 PCTrUS96/01559 Smith, R.L. (1994) J. Med. Chem. 37:725-732.
40. Garcia, A.M., Rowell, C. Ackerman, K., Kowalczyk, J.J., and Lewis, M.D. (1993) J. Biol. Chem. 268:18415-18418.
41. Vogt, A., Qian, Y., Blaskovich, M.A., Fossum, R.D., Hamilton, A.D., and Sebti, S.M. (1995) J. Biol. Chem. 270:660-664.
42. Kohl, N.E., Wilson, F.R., Mosser, S.D., Giuliani, E., deSolms, S.J., Conner, M.W., Anthony, N.J., Holtz, W.J., Gomez, R.P., Lee, T.J., and et al. (1994) Proc.
Natl. Acad. Sci. USA 91:9141-9145.
43. Cox, A.D., Garcia, A.M., Westwick, J.K., Kowalczyk, J.J., Lewis, M.D., Brenner, D.A., and Der, C.J. (1994) J. Biol.
Chem. 269:19203-19206.
44. James, G.L., Brown, M.S., Cobb, M.H., and Goldstein, J.L. (1994) J. Biol.
Chem. 269:277705-277714.
45. Cox, A.D., Hisaka, M.M., Buss, J.E., and Der, C.J. (1992) Mol. Cell. Biol.
12:2606-2615.
46. Hallberg, B. Rayter, S.I., and Downward, J. (1994) J. Biol. Chem. 269:3913-3916.
47. Leevers, S.J., Paterson, H.F., and Marshall, C.J. (1994) Nature 369:411-414.
48. Stanton, V. P., Jr., Nichols, D. W., Laudano, A. P., Cooper, G. M. (1989) Mol. Cell. Blol.
9: 639-647.
49. Casey, P., J. Lipid. Res., 88:1731-1740 (1992) 50. Cox et al, Curr. OP. Cell Biol., 4:1008-1016 (1992) 51. Goldstein et al, Science, 260:1937-1942 (1993) 52. Watanabe et al, Syn. Lett., 3:207-210 (1992) CA 022072~2 1997-06-06 WO96/21456 PCT~S961015S9 53. St~adley et al, Biochemistry, 32:12586-12590 (1993~
54. Gardino, J. et al, J. Orq. Chem., 49:
5237-5243 (1984) 55. Wallow, T. I. et al. J. Orq. Chem.: 59, 5034-5037 (1994) 56. Crowther, G. P. et al., Orq. Syn., 51:
96-100 (1971) 57. Fincham, C. I. et al., J. Med. Chem., 35: 1472-1484 (1992) 58. Goel, O. P. et al., Org. Syn., 67: 69-75 (1989) 59. Pearson, D. A. et al., Tetrahedron Lett., 30: 2739-2742 (1989) 60. Casey, P.J., Solski, P.A., Der, C.J., and Buss, J.E. (1989) Cell 57, 1167-1177 61. Seabra M ~.j RgiQ-~, Y., ~aQey, P.J., Brown, M.S., and Goldstein, J.L. (1991) Cell 65, 429-434 62. Zhang, F.L., Diehl, R.E., Kohl, N.e., Gibbs, J.B., Giros, B., Casey, P.J., and Omer, C.A. 91994) J. Biol. Chem. 269, 63. 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 64. Yokoyama, K., Goodwin, G.W., Ghomashchi, F., Glomser, J.A., and Gelb, M.H. (1991) Proc. Nati. Acad. Sci. USA 88, 5302-5306 65. Trueblood, C.E., Ohya, Y., and Rine, J.
(1993) Mol. Cell. Biol. 13, 4260-4275 66. Lerner, E.G., Qian, Y., Blaskovich, M.A., Fossum, R.D., Vogt A., Sun, J., Cox, A.D., Der, C.J., Hamilton, A.D., and Sebti, S.M. (1995) ~. Biol. Chem.
270, 26802-26806 67. James, G.L., Goldstein, L.L., and Brown, M.S. (1995) ~. Biol. Chem. 270, 6221-W O 96/21456 PCTrUS96/01559 68. Sun, J., Qian, Y., Hamilton, A.D., and Sebti, S.M. (1995) Cancer Research 55, 4243-4247.
69. Moomaw, J.P. and Casey, P.J. (1992) J.
Biol. Chem. 267, 17438-17443 It will be appreciated that various modifications may be made in the invention as described above without departing from the scope and intent of the invention as defined in the following claims wherein:

Claims (76)

1. A peptidomimetic of the formula:

C.beta.X
wherein C is a cysteinyl moiety, in a reduced or nonreduced state;
X is an amino acid; and .beta. is a residue of an aminobenzoic acid or an aminonaphthoic acid.
2. A peptidomimetic according to claim 1 wherein the cysteinyl moiety is in the reduced state.
3. A peptidomimetic according to claim 2 wherein .beta. is 2-phenyl-4-aminobenzoic acid.
4. A peptidomimetic according to claim 1 of the formula:

5. A peptidomimetic according to claim 1 wherein .beta. is a substituted 4-aminobenzoic acid.
6. A peptidomimetic according to claim 1 wherein X is methionine or phenylalanine.
7. A pharmaceutical composition comprising a peptidomimetic according to claim 1 and a pharmaceutically acceptable carrier.
8. A method of inhibiting farnesyltransferase in a host where the farnesyltransferase is present which comprises administering to the host an effective amount of a peptidomimetic according to claim 1.
9. A peptidomimetic according to claim 1 in the form of a pro-drug.
10. A pro-drug according to claim 9 wherein the pro-drug comprises a compound as defined with one or more terminal amino, sulfhydryl and acid groups functionalized with a lipophilic, esterase-sensitive moiety.
11. A pro-drug according to claim 9 wherein the terminal amino and sulfhydryl groups of the cysteine C are functionalized by benzyloxy carbonyl groups or other cleavable lipophilic groups and a terminal carboxylic acid group is esterified.
12. A pro-drug according to claim 11 wherein the carboxylic acid group is esterified as the methyl ester.
13. A compound of the formula:

wherein R represents H, CH3, or any lipophilic esterase-sensitive moiety, and R1 represents H or a substituted or unsubstituted phenyl group.
14. A compound according to claim 13 wherein R1 is an unsubstituted phenyl group, or an alkoxy-, chloro-, bromo- or methyl- substituted phenyl group.
15. A compound according to claim 13 wherein R1 is chosen from the group consisting of a 3,5 dimethylphenyl radical, a thiophene radical, a naphthyl radical, a pyrrole radical, a pyridyl radical, an alkyl radical, and an alkoxy radical.
16. A compound of the formula wherein R represents H, CH3 or any lipophilic esterase-sensitive moiety, and R1 represents H or a substituted or unsubstituted phenyl group.
17. A compound according to claim 15 wherein R1 is an unsubstituted phenyl group, or an alkoxy-, chloro-, bromo- or methyl- substituted phenyl group.
18. A compound according to claim 16 wherein R1 is a 3,5 dimethylphenyl radical.
19. A compound of the formula wherein R represents H, CH3 or any lipophilic esterase-sensitive moiety.
20. A compound of the formula wherein R represents H, CH3 or any lipophilic esterase-sensitive moiety, and R1 represents H, CH3 or OCH3.
21. A method of inhibiting farnesyltransferase in a host wherein the farnesyltransferase is present which comprises administering to the host an effective amount of a peptidomimetic according to claim 13.
22. A method of treating cancer comprising administering to a patient in need of such treatment an effective amount of a peptidomimetic according to claim 1 or 13.
23. A peptidomimetic according to claim 1 wherein .beta. is a residue of an aminobenzoic acid.
24. A peptidomimetic according to claim 23 wherein X is leucine or isoleucine.
25. A peptidomimetic according to claim 24 wherein .beta. is a substituted 4-aminobenzoic acid.
26. A peptidomimetic according to claim 25 wherein the cysteinyl moiety is in the reduced state.
27. A peptidomimetic according to claim 26 wherein the 4-aminobenzoic acid is substituted at the 2- and/or 3- position of the phenyl ring by an alkyl, alkoxy, aryl, or naphthyl group, or a heterocyclic or heteroaromatic ring.
28. A peptidomimetic according to claim 26 wherein .beta. is 2-phenyl-4-aminobenzoic acid or 2-naphthyl-4-aminobenzoic acid.
29. A peptidomimetic according to claim 28 wherein L is leucine.
30. A pharmaceutical composition comprising a peptidomimetic according to claim 24 and a pharmaceutically acceptable carrier.
31. A method of inhibiting geranylgeranyltransferase in a host where the geranylgeranyltransferase is present which comprises administering to the host an effective amount of a peptidomimetic according to claim 24.
32. A peptidomimetic according to claim 24 in the form of a pro-drug.
33. A pro-drug according to claim 32 wherein the pro-drug comprises a compound as defined with one or more terminal amino, sulfhydryl and acid groups functionalized with a lipophilic, esterase-sensitive moiety.
34. A pro-drug according to claim 33 wherein a terminal carboxylic acid group is esterified.
35. A pro-drug according to claim 34 wherein the carboxylic acid group is esterified as the methyl ester.
36. A method of inhibiting geranylgeranyltransferase in a host where geranylgeranyltransferase is present which comprises administering to the host an effective amount of the compound of claim 35.
37. A method of treating cancer comprising administering to a patient in need of such treatment an effective amount of a peptidomimetic according to claim 24.
38. A method of treating cancer comprising administering to a patient in need of such treatment an effective amount of a peptidomimetic according to claim 35.
39. A compound of the formula C°B
wherein C° stands for A representing O or 2H, and Ro representing SH, NH2, or CxHy-SO2-NH-, wherein CxHy is a straight chain saturated or unsaturated hydrocarbon, with x being between 1 and 20 and y between 3 and 41, inclusive; and B stands for -NHR, wherein R is an aryl group.
40. A compound according to claim 39 wherein R is a biphenyl group.
41. A compound according to claim 39 wherein C° is 3-mercapto-2-amino-propylamino.
42. A compound according to claim 41 wherein R is a biphenyl group.
43. A compound according to claim 42 wherein R is a biphenyl group substituted with -COOH.
44. A compound according to claim 42 wherein R is a biphenyl group substituted with lower alkyl or oxyalkyl.
45. A compound according to claim 44 wherein the lower alkyl is methyl.
46. A compound according to claim 39 of the formula:

wherein A is O or 2H; R1 is hydrogen or COOH; R2 is hydrogen, COOH, COOCH3, CH3 or tetrazolyl; R3 is hydrogen or COOH; and R4 is hydrogen, OCH3, OC3H7 or a phenyl group.
47. A compound according to claim 46 wherein R1 is hydrogen; R2 is hydrogen, COOH or CH3; R3 is hydrogen or COOH, R2 being COOH or CH3 when R3 is hydrogen; and R4 is hydrogen.
48. A compound according to claim 47 wherein R2 i s COOH and R3 is hydrogen.
49. A compound according to claim 46 wherein R1 and R3 are H, R2 is COOH or CH3, and R4 is a phenyl or n-oxypropyl group.
50. A compound according to claim 46 wherein R1, R3, and R4 are H; and R2 is tetrazolyl.
51. A compound according to claim 40, wherein Ro is CxHy,-SO2-NH-, x is 2-16 and y is 3 to 33.
52. A compound according to claim 51, wherein Ro is selected from the group consisting of C2H5-SO2-NH-, CH2=CH-SO2-NH-, and CH3(CH2)15-SO2-NH-.
53. A compound according to claim 40, wherein Ro is NH2-.
54. A compound according to claim 53, wherein R is a 2'carboxy-biphenyl group or 1 methoxy-2'-carboxy-biphenyl group.
55. A compound according to claim 39 wherein Ro is SH and B is 3 aminomethyl-3'-carboxy-biphenyl.
56. A method of inhibiting p21ras farnesyltransferase in a host in need of such inhibition which comprises administering to said host an effective amount of a compound according to claim 39.
57. A pharmaceutical composition comprising a compound according to claim 39 and a pharmaceutically acceptable carrier therefor.
58. A method of inhibiting p21ras farnesyltransferase in a host in need of such inhibition which comprises administering to said host an effective amount of a compound according to claim 41.
59. A pharmaceutical composition comprising a compound according to claim 41 and a pharmaceutically acceptable carrier therefor.
60. A peptidomimetic of formula:

C,.beta.X
wherein C represents cysteine;
X is an amino acid; and .beta. is a non-peptide aminoalkyl- or amino-substituted aliphatic or aromatic carboxylic acid or a heterocyclic monocarboxylic acid.
61. A peptidomimetic according to claim 60 wherein .beta. is an aminobenzoic or aminoalkanoic acid.
62. A peptidomimetic of the formula:
C.DELTA. .
wherein C is cysteine and .DELTA. is an aryl or heterocyclic substituent which does not include a peptide amino acid but corresponds essentially in size with a tetrapeptide of the formula CA1A2X
wherein C is cysteine and A1, A2 and X are peptide amino acids.
63. A peptidomimetic according to claim 65 comprising cysteine joined directly to an aminomethyl-bicyclic arylcarboxylic acid.
64. A compound according to claim 63 wherein comprises a biphenyl group.
65. A compound according to claim 64 wherein .DELTA. is the radical of 3-aminomethyl-biphenyl-3-carboxylic acid.
66. A compound of the formula:
.DELTA.1 C .DELTA., .DELTA.1 CA1A2 X
or .DELTA.1 C-.beta.-X
wherein C is cysteine;
A1, A2 and X are peptide amino acids;
.DELTA. is an aryl or heterocyclic substituent which does not include a peptide amino acid;
.beta. is a non-peptide aminoalkyl- or amino-substituted aliphatic or aromatic carboxylic acid or hetercarboxylic acid; and .DELTA.1 is a phosphonate group joined to C
through the cysteine S atom.
67. A compound according to claim 66 of the formula:
.DELTA.1 C-AMBA-X

wherein .DELTA.1, C and X have the meanings given in claim 56 and AMBA is an aminomethyl-benzoic acid.
68. A compound according to claim 67 wherein AMBA is 3-aminomethyl benzoic acid.
69. A compound of the formula:
.beta.1C.DELTA. , .beta.1CA1A2X
or .beta.1C-.beta.-X

wherein C is cysteine; A1, A2 and X are peptide amino acids;
.DELTA. is an aryl or heterocyclic substituent which does not include a peptide amino acid;
.beta. is a non-peptide aminoalkyl- or amino-substituted aliphatic or aromatic carboxylic acid or heterocarboxylic acid; and .beta.1 is an aryl or aralkyl group joined to C through the cysteine S atom.
70. A compound according to claim 69 of the formula:
.beta.1 C-AMBA-X

wherein C, X and .beta.1 have the meanings given in claim 70 and AMBA is an aminomethyl benzoic acid.
71. A compound according to claim 70 wherein AMBA is 3-aminomethylbenzoic acid.
72. A compound of the formula:

or
73. A method of improving peptide or peptidomimetic uptake into cells which comprises utilizing the peptide in a functionalized form such that terminal amino, sulfhydryl and acid groups are functionalized by lipophilic or hydrophobic, esterase-sensitive moieties.
74. A method according to claim 73 for inhibiting tumor cell growth in a host which comprises administering to the host an effective amount of a peptide or peptidomimetic wherein one or more terminal amino, sulfhydryl and carboxylic acid groups have been functionalized with a lipophilic or hydrophobic esterase-sensitive moiety.
75. A method according to claim 74 wherein the terminal amino and sulfhydryl groups are functionalized by benzyloxy carbonyl groups and the carboxylic acid group is esterified.
76. A compound represented by one of formulas (A-L):

(a) (B) (C) (D) (E) (F) (G) (H) (I) or (J) (K) (L) wherein R1' is i) hydrogen;
ii) lower alkyl iii) alkenyl;
iv) alkoxy;
v) thioalkoxyi vi) halo;
vii) haloalkyl;
viii) aryl-L2-, wherein L2 is absent, -CH2-, -CH2CH2-, -CH(CH3)-, -O-, -S(O)q wherein q is 0, 1, or 2, -N(R')- wherein R' is hydrogen or lower alkyl, or -C(O)- and aryl is selected from the group consisting of phenyl, naphthyl, tetrahydronaphthyl, indanyl and indenyl and the aryl group is unsubstituted or substituted; or ix) heterocyclic-L3- wherein L3 is absent, -CH2-, -CH2CH2-, -CH(CH3)-, -O-, -S(O)q wherein q is 0, 1 or 2, -N(R')- wherein R' is hydrogen or loweralkyl, or -C(O)- and heterocyclic is a monocyclic heterocyclic wherein the heterocyclic is unsubstituted or substituted with one, two, or three substituents independently selected from the group consisting of loweralkyl, hydroxy, hydroxyalkyl, halo, nitro, oxo (=O), amino, N-protected amino, alkoxy, thioalkoxy and haloalkyl;

R1a is hydrogen or lower alkyl;

R2' is i) wherein R12a is hydrogen, loweralkyl or -C(O)O-R13, wherein R13 is hydrogen or a carboxy-protecting group and R12b is hydrogen or loweralkyl, with the proviso that R12a and R12b are not both hydrogen, ii) -C(O)NH-CH(R14)-C(O)OR15 wherein R14 is a) loweralkyl, b) cycloalkyl, c) cycloalkylalkyl, d) alkoxyalkyl, e) thioalkoxyalkyl, f) hydroxyalkyl, g) aminoalkyl, h) carboxyalkyl, i) alkoxycarbonylalkyl, j) arylalkyl or k) alkylsulfonylalkyl and R15 is hydrogen or a carboxy-protecting group or .
iii) R3' is where A represents O or 2H, and Ro represents SH, NH2, or CxHy-SO2-NH-, wherein CxHy is a straight chain saturated or unsaturated hydrocarbon, with x being between 1 and 20 and y between 3 and 41, inclusive; and B stands for -NHR, where R is an aryl group.
and L1 is -NH-;
or pharmaceutically acceptable salts or prodrugs thereof.
CA2207252A 1995-01-12 1996-01-11 Inhibitors of prenyl transferases Expired - Lifetime CA2207252C (en)

Applications Claiming Priority (9)

Application Number Priority Date Filing Date Title
US08/371,682 US5705686A (en) 1993-05-18 1995-01-12 Inhibition of farnesyl transferase
US08/371,682 1995-01-12
US08/451,839 US5834434A (en) 1993-05-18 1995-05-30 Inhibitors of farnesyltransferase
US08/451,839 1995-05-30
US55255495A 1995-11-03 1995-11-03
US08/552,554 1995-11-03
US08/582,076 1996-01-02
US08/582,076 US6011175A (en) 1993-05-18 1996-01-02 Inhibition of farnesyltransferase
PCT/US1996/001559 WO1996021456A1 (en) 1995-01-12 1996-01-11 Inhibitors of prenyl transferases

Publications (2)

Publication Number Publication Date
CA2207252A1 true CA2207252A1 (en) 1996-07-18
CA2207252C CA2207252C (en) 2014-02-25

Family

ID=27503084

Family Applications (1)

Application Number Title Priority Date Filing Date
CA2207252A Expired - Lifetime CA2207252C (en) 1995-01-12 1996-01-11 Inhibitors of prenyl transferases

Country Status (6)

Country Link
EP (1) EP0794789A4 (en)
JP (2) JP3929069B2 (en)
AU (1) AU4915796A (en)
CA (1) CA2207252C (en)
MX (1) MX9705273A (en)
WO (1) WO1996021456A1 (en)

Families Citing this family (67)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5962243A (en) * 1990-04-18 1999-10-05 Board Of Regents, The University Of Texas System Methods for the identification of farnesyltransferase inhibitors
US8003342B1 (en) 1990-04-18 2011-08-23 Board Of Regents, The University Of Texas System Method for identifying farnesyl transferase inhibitors
US6117641A (en) 1996-04-11 2000-09-12 Mitotix, Inc. Assays and reagents for identifying anti-fungal agents and uses related thereto
US6727082B1 (en) 1996-04-11 2004-04-27 Gpc Biotech Inc. Assays and reagents for identifying anti-fungal agents, and uses related thereto
ATE220110T1 (en) 1996-04-11 2002-07-15 Mitotix Inc ASSAY AND REAGANTS FOR IDENTIFYING FUNGICIDAL ACTIVE INGREDIENTS AND THEIR USES
US5773455A (en) * 1996-06-28 1998-06-30 Biomeasure, Incorporated Inhibitors of prenyl transferases
US5990277A (en) * 1996-09-03 1999-11-23 Yissum Research Development Company Of The Herbrew University Of Jerusalem Semipeptoid farnesyl protein transferase inhibitors and analogs thereof
US20030060434A1 (en) 1997-02-18 2003-03-27 Loretta Nielsen Combined tumor suppressor gene therapy and chemotherapy in the treatment of neoplasms
GB2323842A (en) * 1997-04-04 1998-10-07 Ferring Bv Pyridine derivatives
GB2323841A (en) * 1997-04-04 1998-10-07 Ferring Bv Group Holdings Pyridine derivatives with anti-tumor and anti-inflammatory activity
WO1998050031A1 (en) * 1997-05-07 1998-11-12 University Of Pittsburgh Inhibitors of protein isoprenyl transferases
FR2780974B1 (en) * 1998-07-08 2001-09-28 Sod Conseils Rech Applic USE OF IMIDAZOPYRAZINE DERIVATIVES FOR THE PREPARATION OF A MEDICAMENT FOR TREATING CONDITIONS RESULTING FROM THE FORMATION OF HETEROTRIMETER G PROTEIN
FR2780892B1 (en) * 1998-07-08 2001-08-17 Sod Conseils Rech Applic USE OF PRENYLTRANSFERASE INHIBITORS FOR THE PREPARATION OF A MEDICINAL PRODUCT FOR TREATING CONDITIONS RESULTING FROM MEMBRANE FIXATION OF HETEROTRIMERIC PROTEIN
US6423519B1 (en) * 1998-07-15 2002-07-23 Gpc Biotech Inc. Compositions and methods for inhibiting fungal growth
DE19851714A1 (en) * 1998-11-05 2000-05-11 Knoell Hans Forschung Ev Amides of cysteine as inhibitors of farnesyl transferase
EP1745800A4 (en) * 2004-04-26 2009-11-18 Ono Pharmaceutical Co Novel blt2-mediated disease, and blt2 binding agent and compound
ES2382814T3 (en) 2005-05-17 2012-06-13 Merck Sharp & Dohme Ltd. Cis-4 - [(4-chlorophenyl) sulfonyl] -4- (2,5-difluorophenyl) cyclohexanopropanoic acid for cancer treatment
GB0603041D0 (en) 2006-02-15 2006-03-29 Angeletti P Ist Richerche Bio Therapeutic compounds
LT2010528T (en) 2006-04-19 2017-12-27 Novartis Ag 6-o-substituted benzoxazole and benzothiazole compounds and methods of inhibiting csf-1r signaling
CA2664113C (en) 2006-09-22 2013-05-28 Merck & Co., Inc. Use of platencin and platensimycin as fatty acid synthesis inhibitors to treat obesity, diabetes and cancer
US20110218176A1 (en) 2006-11-01 2011-09-08 Barbara Brooke Jennings-Spring Compounds, methods, and treatments for abnormal signaling pathways for prenatal and postnatal development
BRPI0806245B1 (en) 2007-01-10 2022-01-25 Msd Italia S.R.L. Compounds of formula i and their uses
BRPI0808523A2 (en) 2007-03-01 2014-08-19 Novartis Vaccines & Diagnostic PIM KINASE INHIBITORS AND METHODS OF USE
CA2685967A1 (en) 2007-05-21 2008-11-21 Novartis Ag Csf-1r inhibitors, compositions, and methods of use
EP2170076B1 (en) 2007-06-27 2016-05-18 Merck Sharp & Dohme Corp. 4-carboxybenzylamino derivatives as histone deacetylase inhibitors
US8691825B2 (en) 2009-04-01 2014-04-08 Merck Sharp & Dohme Corp. Inhibitors of AKT activity
WO2010144909A1 (en) 2009-06-12 2010-12-16 Novartis Ag Fused heterocyclic compounds and their uses
WO2011046771A1 (en) 2009-10-14 2011-04-21 Schering Corporation SUBSTITUTED PIPERIDINES THAT INCREASE p53 ACTIVITY AND THE USES THEREOF
CA2784807C (en) 2009-12-29 2021-12-14 Dana-Farber Cancer Institute, Inc. Type ii raf kinase inhibitors
CN103080093A (en) 2010-03-16 2013-05-01 达纳-法伯癌症研究所公司 Indazole compounds and their uses
WO2011163330A1 (en) 2010-06-24 2011-12-29 Merck Sharp & Dohme Corp. Novel heterocyclic compounds as erk inhibitors
CA2805265A1 (en) 2010-08-02 2012-02-09 Merck Sharp & Dohme Corp. Rna interference mediated inhibition of catenin (cadherin-associated protein), beta 1 (ctnnb1) gene expression using short interfering nucleic acid (sina)
WO2012024170A2 (en) 2010-08-17 2012-02-23 Merck Sharp & Dohme Corp. RNA INTERFERENCE MEDIATED INHIBITION OF HEPATITIS B VIRUS (HBV) GENE EXPRESSION USING SHORT INTERFERING NUCLEIC ACID (siNA)
US8883801B2 (en) 2010-08-23 2014-11-11 Merck Sharp & Dohme Corp. Substituted pyrazolo[1,5-a]pyrimidines as mTOR inhibitors
US8946216B2 (en) 2010-09-01 2015-02-03 Merck Sharp & Dohme Corp. Indazole derivatives useful as ERK inhibitors
EP2615916B1 (en) 2010-09-16 2017-01-04 Merck Sharp & Dohme Corp. Fused pyrazole derivatives as novel erk inhibitors
WO2012058210A1 (en) 2010-10-29 2012-05-03 Merck Sharp & Dohme Corp. RNA INTERFERENCE MEDIATED INHIBITION OF GENE EXPRESSION USING SHORT INTERFERING NUCLEIC ACIDS (siNA)
WO2012087772A1 (en) 2010-12-21 2012-06-28 Schering Corporation Indazole derivatives useful as erk inhibitors
JP2014514321A (en) 2011-04-21 2014-06-19 メルク・シャープ・アンド・ドーム・コーポレーション Insulin-like growth factor 1 receptor inhibitor
US9023865B2 (en) 2011-10-27 2015-05-05 Merck Sharp & Dohme Corp. Compounds that are ERK inhibitors
WO2013074986A1 (en) 2011-11-17 2013-05-23 Dana-Farber Cancer Institute, Inc. Inhibitors of c-jun-n-terminal kinase (jnk)
EP2836482B1 (en) * 2012-04-10 2019-12-25 The Regents of The University of California Compositions and methods for treating cancer
US20150299696A1 (en) 2012-05-02 2015-10-22 Sirna Therapeutics, Inc. SHORT INTERFERING NUCLEIC ACID (siNA) COMPOSITIONS
WO2014052563A2 (en) 2012-09-28 2014-04-03 Merck Sharp & Dohme Corp. Novel compounds that are erk inhibitors
US10112927B2 (en) 2012-10-18 2018-10-30 Dana-Farber Cancer Institute, Inc. Inhibitors of cyclin-dependent kinase 7 (CDK7)
WO2014063054A1 (en) 2012-10-19 2014-04-24 Dana-Farber Cancer Institute, Inc. Bone marrow on x chromosome kinase (bmx) inhibitors and uses thereof
WO2014063061A1 (en) 2012-10-19 2014-04-24 Dana-Farber Cancer Institute, Inc. Hydrophobically tagged small molecules as inducers of protein degradation
MX363243B (en) 2012-11-28 2019-03-14 Merck Sharp & Dohme Compositions and methods for treating cancer.
RU2690663C2 (en) 2012-12-20 2019-06-05 Мерк Шарп И Доум Корп. Substituted imidazopyridines as hdm2 inhibitors
US9540377B2 (en) 2013-01-30 2017-01-10 Merck Sharp & Dohme Corp. 2,6,7,8 substituted purines as HDM2 inhibitors
US20160194368A1 (en) 2013-09-03 2016-07-07 Moderna Therapeutics, Inc. Circular polynucleotides
JP6491202B2 (en) 2013-10-18 2019-03-27 デイナ ファーバー キャンサー インスティチュート,インコーポレイテッド Polycyclic inhibitors of cyclin dependent kinase 7 (CDK 7)
ES2676734T3 (en) 2013-10-18 2018-07-24 Syros Pharmaceuticals, Inc. Heteroatomic compounds useful for the treatment of proliferative diseases
US9862688B2 (en) 2014-04-23 2018-01-09 Dana-Farber Cancer Institute, Inc. Hydrophobically tagged janus kinase inhibitors and uses thereof
US10017477B2 (en) 2014-04-23 2018-07-10 Dana-Farber Cancer Institute, Inc. Janus kinase inhibitors and uses thereof
JO3589B1 (en) 2014-08-06 2020-07-05 Novartis Ag Protein kinase c inhibitors and methods of their use
WO2016105528A2 (en) 2014-12-23 2016-06-30 Dana-Farber Cancer Institute, Inc. Inhibitors of cyclin-dependent kinase 7 (cdk7)
CA2978518C (en) 2015-03-27 2023-11-21 Nathanael S. Gray Inhibitors of cyclin-dependent kinases
EP3307728A4 (en) 2015-06-12 2019-07-17 Dana Farber Cancer Institute, Inc. Combination therapy of transcription inhibitors and kinase inhibitors
EP4019515A1 (en) 2015-09-09 2022-06-29 Dana-Farber Cancer Institute, Inc. Inhibitors of cyclin-dependent kinases
JOP20190055A1 (en) 2016-09-26 2019-03-24 Merck Sharp & Dohme Anti-cd27 antibodies
AU2018252546A1 (en) 2017-04-13 2019-10-10 Sairopa B.V. Anti-SIRPα antibodies
US10947234B2 (en) 2017-11-08 2021-03-16 Merck Sharp & Dohme Corp. PRMT5 inhibitors
WO2019148412A1 (en) 2018-02-01 2019-08-08 Merck Sharp & Dohme Corp. Anti-pd-1/lag3 bispecific antibodies
WO2020033284A1 (en) 2018-08-07 2020-02-13 Merck Sharp & Dohme Corp. Prmt5 inhibitors
EP3833668A4 (en) 2018-08-07 2022-05-11 Merck Sharp & Dohme Corp. Prmt5 inhibitors
WO2023250063A1 (en) * 2022-06-22 2023-12-28 Bioventures, Llc Method of mitigating radiation injury with geranylgeranyl transferase inhibitors

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5602098A (en) * 1993-05-18 1997-02-11 University Of Pittsburgh Inhibition of farnesyltransferase

Also Published As

Publication number Publication date
CA2207252C (en) 2014-02-25
JP4138826B2 (en) 2008-08-27
JP3929069B2 (en) 2007-06-13
JPH10512266A (en) 1998-11-24
AU4915796A (en) 1996-07-31
JP2007016035A (en) 2007-01-25
WO1996021456A1 (en) 1996-07-18
EP0794789A1 (en) 1997-09-17
MX9705273A (en) 1998-06-30
EP0794789A4 (en) 1999-05-26

Similar Documents

Publication Publication Date Title
CA2207252C (en) Inhibitors of prenyl transferases
US5965539A (en) Inhibitors of prenyl transferases
US5602098A (en) Inhibition of farnesyltransferase
AU678625B2 (en) Inhibitors of farnesyl-protein transferase
Sebti et al. Inhibitors of prenyl transferases
AU759492B2 (en) Functionalized alkyl and alkenyl side chain derivatives of glycinamides as farnesyl transferase inhibitors
US11530182B2 (en) YAP1 inhibitors that target the interaction of YAP1 with Oct4
CA2137455A1 (en) Inhibitors of farnesyl protein transferase
RU2092492C1 (en) Derivatives of hydrazine or their salts, pharmaceutical agent, aminoalkylhydrazines or their salts
GB1589933A (en) Amino acid derivatives
NZ257766A (en) Farnesyl-protein transferase inhibitors and their use
NO854516L (en) NEW 5-AMINO-4-HYDROXYVALERYL DERIVATIVES.
EP0048763A1 (en) Sulfur-containing acylamino acids
NO167744B (en) ANALOGY PROCEDURE FOR THE PREPARATION OF PURE INHIBITOR Peptides.
US5834434A (en) Inhibitors of farnesyltransferase
AU714072B2 (en) Combinations of inhibitors of farnesyl-protein transferase
Qian et al. Selective inhibition of type-I geranylgeranyltransferase in vitro and in whole cells by CAAL peptidomimetics
US4500467A (en) Benzoylthio compounds, their preparation and their use as drugs
Qian et al. Probing the hydrophobic pocket of farnesyltransferase: aromatic substitution of CAAX peptidomimetics leads to highly potent inhibitors
US5705686A (en) Inhibition of farnesyl transferase
deSolms et al. Pseudodipeptide inhibitors of protein farnesyltransferase
US6011175A (en) Inhibition of farnesyltransferase
EP0322633A1 (en) Mercapto-acylamino acid antihypertensives
Kunze Phosphono Analogues of Glutathione as New Inhibitors of Glutathione S‐Transferases
El Oualid et al. Synthesis and biological evaluation of protein: geranylgeranyltransferase I inhibitors based on the CaaX Box: incorporation of sugar amino acids as dipeptide isosters

Legal Events

Date Code Title Description
EEER Examination request
EEER Examination request

Effective date: 20021120

MKEX Expiry

Effective date: 20160111