WO1999009985A1 - Inhibiteurs de prenyl-proteine-transferases - Google Patents

Inhibiteurs de prenyl-proteine-transferases Download PDF

Info

Publication number
WO1999009985A1
WO1999009985A1 PCT/US1998/017696 US9817696W WO9909985A1 WO 1999009985 A1 WO1999009985 A1 WO 1999009985A1 US 9817696 W US9817696 W US 9817696W WO 9909985 A1 WO9909985 A1 WO 9909985A1
Authority
WO
WIPO (PCT)
Prior art keywords
cyanobenzyl
substituted
imidazol
aryl
alkyl
Prior art date
Application number
PCT/US1998/017696
Other languages
English (en)
Inventor
Christopher J. Dinsmore
John H. Hutchinson
Theresa M. Williams
Original Assignee
Merck & Co., Inc.
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 GBGB9800975.6A external-priority patent/GB9800975D0/en
Application filed by Merck & Co., Inc. filed Critical Merck & Co., Inc.
Priority to EP98943406A priority Critical patent/EP1014984A1/fr
Priority to AU91213/98A priority patent/AU741725B2/en
Priority to CA002301770A priority patent/CA2301770A1/fr
Priority to JP2000507375A priority patent/JP2001513561A/ja
Publication of WO1999009985A1 publication Critical patent/WO1999009985A1/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/06Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system

Definitions

  • the present invention relates to certain compounds that are useful for the inhibition of prenyl-protein transferases and the treatment of cancer.
  • the invention relates to prenyl- protein transferase inhibitors which are efficacious in vivo as inhibitors of geranylgeranyl-protein transferase type I (GGTase-I) and that inhibit the cellular processing of both the H-Ras protein and the K4B-Ras protein.
  • Prenylation of proteins by prenyl-protein transferases represents a class of post-translational modification (Glomset, J. A., Gelb, M. H., and Farnsworth, C. C. (1990). Trends Biochem. Sci. 15, 139-142; Maltese, W. A. (1990). FASEB J. 4, 3319-3328). This modification typically is required for the membrane localization and function of these proteins.
  • Prenylated proteins share characteristic C-terminal sequences including CAAX (C, Cys; A, an aliphatic amino acid; X, another amino acid), XXCC, or XCXC.
  • GGPTase farnesyl-protein transferase
  • GGPTase-I geranylgeranyl- protein transferase type I
  • GGPTase-II geranylgeranyl-protein transferase type-II
  • FPTase farnesylates CaaX-containing proteins that end with Ser, Met, Cys, Gin or Ala.
  • CaaX tetrapeptides comprise the minimum region required for interaction of the protein substrate with the enzyme.
  • the enzymological characterization of these three enzymes has demonstrated that it is possible to selectively inhibit one with little inhibitory effect on the others (Moores, S. L., Schaber, M. D., Mosser, S. D., Rands, E., O'Hara, M. B., Garsky, V. M., Marshall, M. S., Pompliano, D. L., and Gibbs, J. B., J. Biol. Chem., 266:17438 (1991), U.S. Pat. No. 5,470,832).
  • the Ras protein is part of a signaling pathway that links cell surface growth factor receptors to nuclear signals initiating cellular proliferation.
  • Biological and biochemical studies of Ras action indicate that Ras functions like a G-regulatory protein.
  • Ras In the inactive state, Ras is bound to GDP.
  • Ras Upon growth factor receptor activation, Ras is induced to exchange GDP for GTP and undergoes a conformational change.
  • the GTP-bound form of Ras propagates the growth stimulatory signal until the signal is terminated by the intrinsic GTPase activity of Ras, which returns the protein to its inactive GDP bound form (D.R. Lowy and D.M. Willumsen, Ann. Rev. Biochem. 62:851-891 (1993)).
  • Ras Activation of Ras leads to activation of multiple intracellular signal transduction pathways, including the MAP Kinase pathway and the Rho/Rac pathway (Joneson et al., Science 271:810-812).
  • Mutated ras genes are found in many human cancers, including colorectal carcinoma, exocrine pancreatic carcinoma, and myeloid leukemias. The protein products of these genes are defective in their GTPase activity and constitutively transmit a growth stimulatory signal.
  • the Ras protein is one of several proteins that are known to undergo post-translational modification.
  • Farnesyl-protein transferase utilizes farnesyl pyrophosphate to covalently modify the Cys thiol group of the Ras CAAX box with a farnesyl group (Reiss et al., Cell, 62:81-88 (1990); Schaber et al., J. Biol. Chem., 265:14701-14704 (1990); Schafer et al., Science, 249:1133-1139 (1990); Manne et al., Proc. Natl. Acad. Sci USA, 87:7541-7545 (1990)).
  • Ras must be localized to the plasma membrane for both normal and oncogenic functions. At least 3 post-translational modifications are involved with Ras membrane localization, and all 3 modifications occur at the C-terminus of Ras.
  • the Ras C-terminus contains a sequence motif termed a "CAAX” or "Cys-Aaa -Aaa -Xaa” box (Cys is cysteine, Aaa is an aliphatic amino acid, the Xaa is any amino acid) (Willumsen et al., Nature 310:583-586 (1984)).
  • this motif serves as a signal sequence for the enzymes farnesyl-protein transferase or geranylgeranyl-protein transferase, which catalyze the alkylation of the cysteine residue of the CAAX motif with a Cl5 or C20 isoprenoid, respectively.
  • farnesylated proteins include the Ras-related GTP- binding proteins such as RhoB, fungal mating factors, the nuclear lamins, and the gamma subunit of transducin.
  • RhoB Ras-related GTP- binding proteins
  • James, et al. have also suggested that there are farnesylated proteins of unknown structure and function in addition to those listed above.
  • Inhibitors of farnesyl-protein transferase (FPTase) have been described in two general classes.
  • the first class includes analogs of farnesyl diphosphate (FPP), while the second is related to protein substrates (e.g., Ras) for the enzyme.
  • FPP farnesyl diphosphate
  • protein substrates e.g., Ras
  • the peptide derived inhibitors that have been described are generally cysteine containing molecules that are related to the CAAX motif that is the signal for protein prenylation. (Schaber et al., ibid; Reiss et. al., ibid; Reiss et al., PNAS, 88:732-736 (1991)). Such inhibitors may inhibit protein prenylation while serving as alternate substrates for the farnesyl-protein transferase enzyme, or may be purely competitive inhibitors (U.S. Patent 5,141,851, University of Texas; N.E. Kohl et al., Science, 260:1934-1937 (1993); Graham, et al., J. Med. Chem., 37, 725 ( 1994)
  • H-ras is an abbreviation for Harvey-ras.
  • K4A-ras and K4B-ras are abbreviations for the Kirsten splice variants of ras that contain the 4A and 4B exons, respectively.
  • HMG-CoA reductase the rate limiting enzyme for the production of polyisoprenoids
  • Inhibition of farnesyl pyrophosphate biosynthesis by inhibiting HMG-CoA reductase blocks Ras membrane localization in cultured cells.
  • a composition which comprises such an inhibitor compound is used in the present invention to treat cancer.
  • the present invention comprises peptidomimetic piperazinone-containing compounds which inhibit the prenyl-protein transferases: farnesyl-protein transferase and geranylgeranyl-protein transferase type I. Further contained in this invention are chemotherapeutic compositions containing these prenyl transferase inhibitors and methods for their production.
  • the compounds of this invention are useful in the inhibition of prenyl-protein transferases and the prenylation of the oncogene protein Ras.
  • the inhibitors of prenyl-protein transferases are illustrated by the formula I:
  • R ⁇ - a is selected from: hydrogen or Ci -C ⁇ alkyl
  • Rl° is independently selected from: a) hydrogen, b) aryl, heterocycle, cycloalkyl, Rl°O-, -N(R 10 )2 or C2-C6 alkenyl, c) C1-C6 alkyl unsubstituted or substituted by aryl, heterocycle, cycloalkyl, alkenyl, Rl ⁇ O-, or -N(RlO)2;
  • R 3 and R 4 selected from H and CH3;
  • R ⁇ is selected fromH; unsubstituted or substituted aryl, unsubstituted or substituted heteroaryl,
  • Ci-5 alkyl unbranched or branched, unsubstituted or substituted with one or more of:
  • Y O : and R2 and R 3 are optionally attached to the same carbon atom;
  • R6 and R' 7 are independently selected from:
  • R ⁇ a is selected from: Ci-4 alkyl or C3-6 cycloalkyl, unsubstituted or substituted with: a) Ci-4 alkoxy, b) halogen, or c) aryl or heterocycle;
  • R8 is independently selected from: a) hydrogen, b) Ci-C ⁇ alkyl, C 2 -C 6 alkenyl, C -C 6 alkynyl, Ci -C ⁇ perfluoroalkyl, F, Cl, RlOO-, R 10 C(O)NR 10 -, CN, NO2, (R!0)2N-C(NR 1 0)-, R 10 C(O)-, -N(RlO) 2 , or R OC(O)NRl0-, and c) C1-C6 alkyl substituted by Ci-C ⁇ perfluoroalkyl, R 10 O-,
  • R 10 C(O)NR 10 -, (Rl°)2N-C(NRlO)-, RlOC(O)-, -N(RlO)2, or R11OC(O)NR10-;
  • R ⁇ a is hydrogen or methyl
  • RIO is independently selected from hydrogen, Ci-C ⁇ alkyl, C1-C6 perfluoroalkyl, 2,2,2-trifluoroethyl, benzyl and aryl;
  • RU is independently selected from Ci-C ⁇ alkyl and aryl
  • Z is selected from:
  • a unsubstituted or substituted group selected from aryl, heteroaryl, arylmethyl, heteroaryl ethyl, arylsulfonyl, heteroarylsulfonyl, wherein the substituted group is substituted with one or more of the following: a) Cl-4 alkyl, unsubstituted or substituted with:
  • Rl° is independently selected from: a) hydrogen, b) aryl, heterocycle, cycloalkyl, RlOO-, -N(R 10 )2 or C2-C6 alkenyl, c) C ⁇ -C6 alkyl unsubstituted or substituted by aryl, heterocycle, cycloalkyl, alkenyl, RI ⁇ O-, or -N(RlO)2;
  • R2 is selected from H; unsubstituted or substituted aryl or Cl-5 alkyl, unbranched or branched, unsubstituted or substituted with one or more of:
  • SR 6a ; R 6 and R 7 are independently selected from: Cl-4 alkyl, aryl, and heteroaryl, unsubstituted or substituted with: a) Cl-4 alkoxy, b) halogen, c) perfluoro-Ci-4 alkyl, or d) aryl or heteroaryl;
  • R a is selected from:
  • Cl-4 alkyl unsubstituted or substituted with: a) C ⁇ -4 alkoxy, or b) aryl or heteroaryl;
  • is independently selected from: a) hydrogen, b) Ci-C ⁇ lkyl, C2-C6 alkenyl, C2-C6 alkynyl, Ci-C ⁇ perfluoroalkyl, F, Cl, R 10 O-, R10C(O)NR10-, CN, NO2, (R 1 0)2N-C(NRlO)-, RlOc(O)-, -N(R 10 )2, or RH ⁇ C(O)NR 10 -, and c) Ci-C ⁇ alkyl substituted by C ⁇ -C6 perfluoroalkyl, Rl ⁇ O-, R 1 0C(O)NR 10 -, (R 10 )2N-C(NRlO)-, R 10 C(O)-,
  • RlO is independently selected from hydrogen, Ci-C ⁇ alkyl, benzyl and aryl;
  • RU is independently selected from Ci-C ⁇ alkyl and aryl
  • Z is an unsubstituted or substituted group selected from aryl, arylmethyl and arylsulfonyl, wherein the substituted group is substituted with one or more of the following: a) Cl-4 alkyl, unsubstituted or substituted with: Cl-4 alkoxy, NR R 7 , C3.6 cycloalkyl, unsubstituted or substituted aryl, heterocycle, HO, -S(O) m R 6a , or
  • the compounds of the instant invention differ from previously disclosed piperazinone-containing compounds, (PCT Publ. No. WO 97/30343 - October 3, 1996; PCT Publ. No. WO 97/36593 - October 9, 1997; PCT Publ. No. WO 97/36592 - October 9, 1997) that were described as selective inhibitors of farnesyl-protein transferase, in that the instant compounds are dual inhibitors of farnesyl-protein transferase and geranylgeranyl-protein transferase type I (GGTase-I).
  • the compounds of the instant invention inhibit FPTase in vitro (Example 15) at an IC 50 of less than 1 mM, inhibit GGTase-I in vitro (Example 16) at an IC 50 of less than 1 mM and inhibited the cellular processing (farnesylation) of H-Ras (Example 17) at an IC 50 of less than 1 mM.
  • the compounds of the present invention may have asymmetric centers and occur as racemates, racemic mixtures, and as individual diastereomers, with all possible isomers, including optical isomers, being included in the present invention.
  • any variable e.g. aryl, heterocycle, Rl, R2 etc.
  • any variable e.g. aryl, heterocycle, Rl, R2 etc.
  • alkyl is intended to include both branched and straight-chain saturated aliphatic hydrocarbon groups having the specified number of carbon atoms; “alkoxy” represents an alkyl group of indicated number of carbon atoms attached through an oxygen bridge.
  • Halogen or “halo” as used herein means fluoro, chloro, bromo and iodo.
  • aryl is intended to mean any stable monocyclic or bicyclic carbon ring of up to 7 members in each ring, wherein at least one ring is aromatic.
  • aryl elements include phenyl, naphthyl, tetrahydronaphthyl, indanyl, biphenyl, phenanthryl, anthryl or acenaphthyl.
  • heterocycle or heterocyclic represents a stable 5- to 7-membered monocyclic or stable 8- to 11-membered bicyclic heterocyclic ring which is either saturated or unsaturated, and which consists of carbon atoms and from one to four heteroatoms selected from the group consisting of N, O, and S, and including any bicyclic group in which any of the above-defined heterocyclic rings is fused to a benzene ring.
  • the heterocyclic ring may be attached at any heteroatom or carbon atom which results in the creation of a stable structure.
  • heterocyclic elements include, but are not limited to, azepinyl, benzimidazolyl, benzisoxazolyl, benzofurazanyl, benzopyranyl, benzothiopyranyl, benzofuryl, benzothiazolyl, benzothienyl, benzoxazolyl, chromanyl, cinnolinyl, dihydrobenzofuryl, dihydrobenzothienyl, dihydrobenzothiopyranyl, dihydrobenzothiopyranyl sulfone, furyl, imidazolidinyl, imidazolinyl, imidazolyl, indolinyl, indolyl, isochromanyl, isoindolinyl, isoquinolinyl, isothiazolidinyl, isothiazolyl, isothiazolidinyl, morpholinyl, naphthyridinyl, oxadiazolyl,
  • heteroaryl is intended to mean any stable monocyclic or bicyclic carbon ring of up to 7 members in each ring, wherein at least one ring is aromatic and wherein from one to four carbon atoms are replaced by heteroatoms selected from the group consisting of N, O, and S.
  • heterocyclic elements include, but are not limited to, benzimidazolyl, benzisoxazolyl, benzofurazanyl, benzopyranyl, benzothiopyranyl, benzofuryl, benzothiazolyl, benzothienyl, benzoxazolyl, chromanyl, cinnolinyl, dihydrobenzofuryl, dihydrobenzothienyl, dihydrobenzothiopyranyl, dihydrobenzothiopyranyl sulfone, furyl, imidazolyl, indolinyl, indolyl, isochromanyl, isoindolinyl, isoquinolinyl, isothiazolyl, naphthyridinyl, oxadiazolyl, pyridyl, pyrazinyl, pyrazolyl, pyridazinyl, pyrimidinyl, pyrrolyl, quinazolin
  • the substituted group intended to mean a substituted Cl-8 alkyl, substituted C2-8 alkenyl, substituted C2-8 a lkynyl, substituted aryl or substituted heterocycle from which the substituent(s) R 2 and R 3 are selected.
  • the substituted Cl-8 a lkyl, substituted C3-6 cycloalkyl, substituted aroyl, substituted aryl, substituted heteroaroyl, substituted arylsulfonyl, substituted heteroarylsulfonyl and substituted heterocycle include moieties containing from 1 to 3 substituents in addition to the point of attachment to the rest of the compound.
  • substituents are selected from the group which includes but is not limited to F, Cl, Br, CF3, NH 2 , N(Ci-C6 a lkyl)2, NO 2 , CN, (Ci-C 6 alkyDO-, -OH, (Ci-C ⁇ alkyl)S(O , (Ci-C ⁇ alkyl)C(O)NH-, H2N-C(NH)-, (Ci-C 6 alkyl)C(O)-, (C1-C6 alkyl)OC(O)-, N3,(C ⁇ -C ⁇ alkyl)OC(O)NH-, phenyl, pyridyl, imidazolyl, oxazolyl, isoxazolyl, thiazolyl, thienyl, furyl, isothiazolyl and C1-C20 alkyl.
  • Lines drawn into the ring systems from substituents indicate that the indicated bond may be attached to any of the substitutable ring carbon atoms.
  • Rl a and Rl° are independently selected from: hydrogen, -N(R 10 )2, R 10 C(O)NR 1 0- or unsubstituted or substituted C1-C6 alkyl wherein the substituent on the substituted C1-C6 alkyl is selected from unsubstituted or substituted phenyl, -N(RlO)2, Rl ⁇ O- and R 10 C(O)NRl0-.
  • R 2 is selected from: H,
  • R4 is hydrogen
  • R 6 and R 7 are selected from: hydrogen, unsubstituted or substituted C1-C6 alkyl, unsubstituted or substituted aryl and unsubstituted or substituted C3-C6 cycloalkyl.
  • R 6a is unsubstituted or substituted Cl-C6-
  • R ⁇ is hydrogen
  • RlO is selected from H, C1-C6 alkyl and benzyl.
  • a ⁇ and A 2 are independently selected from: a bond, -C(O)NR 10 -, -NRlOC(O)-, O, -N(RIO)., -S(O)2N(R10). and -N(R 10 )S(O)2-.
  • a 1 and A 2 are a bond.
  • V is selected from hydrogen, heterocycle and aryl. More preferably, V is phenyl.
  • Z is selected from unsubstituted or substituted phenyl, unsubstituted or substituted naphthyl, unsubstituted or substituted pyridyl, unsubstituted or substituted furanyl and unsubstituted or substituted thienyl. More preferably, Z is unsubstituted or substituted phenyl.
  • n and r are independently 0, 1, or 2.
  • p is 1, 2 or 3.
  • the moiety is 1, 2 or 3.
  • any substituent or variable e.g., RJ- a , R", n, etc.
  • -N(RlO)2 represents -NHH, -NHCH3, -NHC2H5, etc. It is understood that substituents and substitution patterns on the compounds of the instant invention can be selected by one of ordinary skill in the art to provide compounds that are chemically stable and that can be readily synthesized by techniques known in the art, as well as those methods set forth below, from readily available starting materials.
  • the pharmaceutically acceptable salts of the compounds of this invention include the conventional non-toxic salts of the compounds of this invention as formed, e.g., from non-toxic inorganic or organic acids.
  • such conventional non-toxic salts include those derived from inorganic acids such as hydrochloric, hydrobromic, sulfuric, sulfamic, phosphoric, nitric and the like: and the salts prepared from organic acids such as acetic, propionic, succinic, glycolic, stearic, lactic, malic, tartaric, citric, ascorbic, pamoic, maleic, hydroxymaleic, phenylacetic, glutamic, benzoic, salicylic, sulfanilic, 2-acetoxy-benzoic, fumaric, toluenesulfonic, methanesulfonic, ethane disulfonic, oxalic, isethionic, trifluoroacetic and the like.
  • the pharmaceutically acceptable salts of the compounds of this invention can be synthesized from the compounds of this invention which contain a basic moiety by conventional chemical methods.
  • the salts are prepared either by ion exchange chromatography or by reacting the free base with stoichiometric amounts or with an excess of the desired salt-forming inorganic or organic acid in a suitable solvent or various combinations of solvents.
  • Reactions used to generate the compounds of this invention are prepared by employing reactions as shown in the Schemes 1-11, in addition to other standard manipulations such as ester hydrolysis, cleavage of protecting groups, etc., as may be known in the literature or exemplified in the experimental procedures.
  • Substituent R as shown in the Schemes, represents the substituents R 2 , R 3 , R 4 , and R ⁇ » however the point of attachment to the ring is illustrative only and is not meant to be limiting.
  • Piperazin-5-ones can be prepared as shown in Scheme 1.
  • the protected suitably substituted amino acid IV can be converted to the corresponding aldehyde V by first forming the amide and then reducing it with LAH.
  • Reductive amination of Boc-protected amino aldehydes V gives rise to compound VI.
  • the intermediate VI can be converted to a piperazinone by acylation with chloroacetyl chloride to give VII, followed by base-induced cyclization to VIII.
  • Deprotection, followed by reductive alkylation with a protected imidazole carboxalde- hyde leads to IX, which can be alkylated with an arylmethylhalide to give the imidazolium salt X.
  • Final removal of protecting groups by either solvolysis with a lower alkyl alcohol, such as methanol, or treatment with triethylsilane in methylene chloride in the presence of trifluoroacetic acid gives the final product XI.
  • the intermediate VIII can be reductively alkylated with a variety of aldehydes, such as XII.
  • the aldehydes can be prepared by standard procedures, such as that described by O. P. Goel, U. Krolls, M. Stier and S. Kesten in Organic Syntheses. 1988, 67, 69-75, from the appropriate amino acid (Scheme 2).
  • the reductive alkylation can be accomplished at pH 5-7 with a variety of reducing agents, such as sodium triacetoxyborohydride or sodium cyanoborohydride in a solvent such as dichloroethane, methanol or dimethylformamide.
  • the product XIII can be deprotected to give the final compounds XIV with trifluoro- acetic acid in methylene chloride.
  • the final product XIV is isolated in the salt form, for example, as a trifluoroacetate, hydrochloride or acetate salt, among others.
  • the product diamine XIV can further be selectively protected to obtain XV, which can subsequently be reductively alkylated with a second aldehyde to obtain XVI. Removal of the protecting group, and conversion to cyclized products such as the dihydroimidazole XVII can be accomplished by literature procedures.
  • the imidazole acetic acid XVIII can be converted to the acetate XIX by standard procedures, and XIX can be first reacted with an alkyl halide, then treated with refluxing methanol to provide the regiospecifically alkylated imidazole acetic acid ester XX (Scheme 3).
  • Hydrolysis and reaction with piperazinone VIII in the presence of condensing reagents such as l-(3-dimethylaminopropyl)- 3-ethylcarbodiimide (EDC) leads to acylated products such as XXI.
  • the piperazinone VIII is reductively alkylated with an aldehyde which also has a protected hydroxyl group, such as XXII in Scheme 4, the protecting groups can be subsequently removed to unmask the hydroxyl group (Schemes 4, 5).
  • the alcohol can be oxidized under standard conditions to e.g. an aldehyde, which can then be reacted with a variety of organometallic reagents such as Grignard reagents, to obtain secondary alcohols such as XXIV.
  • the fully deprotected amino alcohol XXV can be reductively alkylated (under conditions described previously) with a variety of aldehydes to obtain secondary amines, such as XXVI (Scheme 5), or tertiary amines.
  • the Boc protected amino alcohol XXIII can also be utilized to synthesize 2-aziridinylmethylpiperazinones such as XXVII (Scheme 6). Treating XXIII with l,l'-sulfonyldiimidazole and sodium hydride in a solvent such as dimethylformamide led to the formation of aziridine XXVII. The aziridine reacted in the presence of a nucleophile, such as a thiol, in the presence of base to yield the ring-opened product XXVIII.
  • a nucleophile such as a thiol
  • piperazinone VIII can be reacted with aldehydes derived from amino acids such as O-alkylated tyrosines, according to standard procedures, to obtain compounds such as XXX (Scheme 7).
  • R' is an aryl group
  • XXX can first be hydrogenated to unmask the phenol, and the amine group deprotected with acid to produce XXXI.
  • the amine protecting group in XXX can be removed, and O-alkylated phenolic amines such as XXXII produced.
  • Scheme 8 illustrates the use of an optionally substituted homoserine lactone XXXIII to prepare a Boc-protected piperazinone XXXVII.
  • Intermediate XXXVII may be deprotected and reductively alkylated or acylated as illustrated in the previous Schemes.
  • the hydroxyl moiety of intermediate XXXVII may be mesylated and displaced by a suitable nucleophile, such as the sodium salt of ethane thiol, to provide an intermediate XXXVIII.
  • Intermediate XXXVII may also be oxidized to provide the carboxylic acid on intermediate IXL, which can be utilized form an ester or amide moiety.
  • N-Aralkyl-piperazin-5-ones can be prepared as shown in Scheme 9. Reductive amination of Boc-protected amino aldehydes V (prepared from III as described previously) gives rise to compound XL. This is then reacted with bromoacetyl bromide under Schotten- Baumann conditions; ring closure is effected with a base such as sodium hydride in a polar aprotic solvent such as dimethylformamide to give XLI. The carbamate protecting group is removed under acidic conditions such as trifluoroacetic acid in methylene chloride, or hydrogen chloride gas in methanol or ethyl acetate, and the resulting piperazine can then be carried on to final products as described in Schemes 1-7.
  • the isomeric piperazin-3-ones can be prepared as described in Scheme 10.
  • the imine formed from arylcarboxamides XLII and 2-aminoglycinal diethyl acetal (XLIII) can be reduced under a variety of conditions, including sodium triacetoxyborohydride in dichloroethane, to give the amine XLIV.
  • Amino acids I can be coupled to amines XLIV under standard conditions, and the resulting amide XLV when treated with aqueous acid in tetrahydrofuran can cyclize to the unsaturated XL VI.
  • Catalytic hydrogenation under standard conditions gives the requisite intermediate XL VII, which is elaborated to final products as described in Schemes 1-7.
  • amino acids of the general formula IL which have a sidechain not found in natural amino acids may be prepared by the reactions illustrated in Scheme 11 starting with the readily prepared imine XLVIII.
  • the instant compounds are useful as pharmaceutical agents for mammals, especially for humans. These compounds may be administered to patients for use in the treatment of cancer.
  • Examples of the type of cancer which may be treated with the compounds of this invention include, but are not limited to, colorectal carcinoma, exocrine pancreatic carcinoma, myeloid leukemias and neurological tumors. Such tumors may arise by mutations in the ras genes themselves, mutations in the proteins that can regulate Ras activity (i.e., neurof ⁇ bromin (NF-1), neu, src, abl, lck, fyn) or by other mechanisms.
  • the compounds of the instant invention inhibit prenyl- protein transferase and the prenylation of the oncogene protein Ras.
  • the instant compounds may also inhibit tumor angiogenesis, thereby affecting the growth of tumors (J. Rak et al. Cancer Research, 55: 4575-4580 (1995)). Such anti-angiogenesis properties of the instant compounds may also be useful in the treatment of certain forms of vision deficit related to retinal vascularization.
  • the compounds of this invention are also useful for inhibiting other proliferative diseases, both benign and malignant, wherein Ras proteins are aberrantly activated as a result of oncogenic mutation in other genes (i.e., the Ras gene itself is not activated by mutation to an oncogenic form) with said inhibition being accomplished by the administration of an effective amount of the compounds of the invention to a mammal in need of such treatment.
  • a component of NF-1 is a benign proliferative disorder.
  • the instant compounds may also be useful in the treatment of certain viral infections, in particular in the treatment of hepatitis delta and related viruses (J.S. Glenn et al. Science, 256:1331-1333 (1992).
  • the compounds of the instant invention are also useful in the prevention of restenosis after percutaneous transluminal coronary angioplasty by inhibiting neointimal formation (C. Indolfi et al. Nature medicine, 1:541-545(1995).
  • the instant compounds may also be useful in the treatment and prevention of polycystic kidney disease (D.L. Schaffner et al. American Journal of Pathology, 142:1051-1060 (1993) and B. Cowley, Jr. et al.FASEB Journal, 2:A3160 (1988)).
  • the instant compounds may also be useful for the treatment of fungal infections.
  • the instant compounds may also be useful as inhibitors of proliferation of vascular smooth muscle cells and therefore useful in the prevention and therapy of arteriosclerosis and diabetic vascular pathologies.
  • the compounds of this invention may be administered to mammals, preferably humans, either alone or, preferably, in combination with pharmaceutically acceptable carriers, excipients or diluents, in a pharmaceutical composition, according to standard pharmaceutical practice.
  • the compounds can be administered orally or parenterally, including the intravenous, intramuscular, intraperitoneal, subcutaneous, rectal and topical routes of administration.
  • compositions containing the active ingredient may be in a form suitable for oral use, for example, as tablets, troches, lozenges, aqueous or oily suspensions, dispersible powders or granules, emulsions, hard or soft capsules, or syrups or elixirs.
  • Compositions intended for oral use may be prepared according to any method known to the art for the manufacture of pharmaceutical compositions and such compositions may contain one or more agents selected from the group consisting of sweetening agents, flavoring agents, coloring agents and preserving agents in order to provide pharmaceutically elegant and palatable preparations. Tablets contain the active ingredient in admixture with non-toxic pharmaceutically acceptable excipients which are suitable for the manufacture of tablets.
  • excipients may be for example, inert diluents, such as calcium carbonate, sodium carbonate, lactose, calcium phosphate or sodium phosphate; granulating and disintegrating agents, for example, microcrystalline cellulose, sodium crosscarmellose, corn starch, or alginic acid; binding agents, for example starch, gelatin, polyvinyl- pyrrolidone or acacia, and lubricating agents, for example, magnesium stearate, stearic acid or talc.
  • the tablets may be uncoated or they may be coated by known techniques to mask the unpleasant taste of the drug or delay disintegration and absorption in the gastrointestinal tract and thereby provide a sustained action over a longer period.
  • a water soluble taste masking material such as hydroxypropylmethyl- cellulose or hydroxypropylcellulose, or a time delay material such as ethyl cellulose, cellulose acetate buryrate may be employed.
  • Formulations for oral use may also be presented as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent, for example, calcium carbonate, calcium phosphate or kaolin, or as soft gelatin capsules wherein the active ingredient is mixed with water soluble carrier such as polyethyleneglycol or an oil medium, for example peanut oil, liquid paraffin, or olive oil.
  • an inert solid diluent for example, calcium carbonate, calcium phosphate or kaolin
  • water soluble carrier such as polyethyleneglycol or an oil medium, for example peanut oil, liquid paraffin, or olive oil.
  • Aqueous suspensions contain the active material in admixture with excipients suitable for the manufacture of aqueous suspensions.
  • excipients are suspending agents, for example sodium carboxymethylcellulose, methylcellulose, hydroxypropylmethyl- cellulose, sodium alginate, polyvinyl-pyrrolidone, gum tragacanth and gum acacia; dispersing or wetting agents may be a naturally-occurring phosphatide, for example lecithin, or condensation products of an alkylene oxide with fatty acids, for example polyoxyethylene stearate, or condensation products of ethylene oxide with long chain aliphatic alcohols, for example heptadecaethylene-oxycetanol, or condensation products of ethylene oxide with partial esters derived from fatty acids and a hexitol such as polyoxyethylene sorbitol monooleate, or condensation products of ethylene oxide with partial esters derived from fatty acids and hexitol anhydrides, for example polyethylene sorbitan
  • the aqueous suspensions may also contain one or more preservatives, for example ethyl, or n-propyl p-hydroxybenzoate, one or more coloring agents, one or more flavoring agents, and one or more sweetening agents, such as sucrose, saccharin or aspartame.
  • preservatives for example ethyl, or n-propyl p-hydroxybenzoate
  • coloring agents for example ethyl, or n-propyl p-hydroxybenzoate
  • flavoring agents such as sucrose, saccharin or aspartame.
  • sweetening agents such as sucrose, saccharin or aspartame.
  • Oily suspensions may be formulated by suspending the active ingredient in a vegetable oil, for example arachis oil, olive oil, sesame oil or coconut oil, or in mineral oil such as liquid paraffin.
  • the oily suspensions may contain a thickening agent, for example beeswax, hard paraffin or cetyl alcohol.
  • Sweetening agents such as those set forth above, and flavoring agents may be added to provide a palatable oral preparation.
  • These compositions may be preserved by the addition of an anti-oxidant such as butylated hydroxyanisol or alpha-tocopherol.
  • Dispersible powders and granules suitable for preparation of an aqueous suspension by the addition of water provide the active ingredient in admixture with a dispersing or wetting agent, suspending agent and one or more preservatives.
  • Suitable dispersing or wetting agents and suspending agents are exemplified by those already mentioned above. Additional excipients, for example sweetening, flavoring and coloring agents, may also be present. These compositions may be preserved by the addition of an anti-oxidant such as ascorbic acid.
  • the pharmaceutical compositions of the invention may also be in the form of an oil-in-water emulsions.
  • the oily phase may be a vegetable oil, for example olive oil or arachis oil, or a mineral oil, for example liquid paraffin or mixtures of these.
  • Suitable emulsifying agents may be naturally-occurring phosphatides, for example soy bean lecithin, and esters or partial esters derived from fatty acids and hexitol anhydrides, for example sorbitan monooleate, and condensation products of the said partial esters with ethylene oxide, for example polyoxyethylene sorbitan monooleate.
  • the emulsions may also contain sweetening, flavouring agents, preservatives and antioxidants.
  • Syrups and elixirs may be formulated with sweetening agents, for example glycerol, propylene glycol, sorbitol or sucrose. Such formulations may also contain a demulcent, a preservative, flavoring and coloring agents and antioxidant.
  • sweetening agents for example glycerol, propylene glycol, sorbitol or sucrose.
  • Such formulations may also contain a demulcent, a preservative, flavoring and coloring agents and antioxidant.
  • compositions may be in the form of a sterile injectable aqueous solutions.
  • acceptable vehicles and solvents that may be employed are water, Ringer's solution and isotonic sodium chloride solution.
  • the sterile injectable preparation may also be a sterile injectable oil-in-water microemulsion where the active ingredient is dissolved in the oily phase.
  • the active ingredient may be first dissolved in a mixture of soybean oil and lecithin. The oil solution then introduced into a water and glycerol mixture and processed to form a microemulation.
  • the injectable solutions or microemulsions may be introduced into a patient's blood-stream by local bolus injection. Alternatively, it may be advantageous to administer the solution or microemulsion in such a way as to maintain a constant circulating concentration of the instant compound.
  • a continuous intravenous delivery device may be utilized.
  • An example of such a device is the Deltec CADD-PLUSTM model 5400 intravenous pump.
  • the pharmaceutical compositions may be in the form of a sterile injectable aqueous or oleagenous suspension for intramuscular and subcutaneous administration. This suspension may be formulated according to the known art using those suitable dispersing or wetting agents and suspending agents which have been mentioned above.
  • the sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally-acceptable diluent or solvent, for example as a solution in 1,3-butane diol.
  • sterile, fixed oils are conventionally employed as a solvent or suspending medium.
  • any bland fixed oil may be employed including synthetic mono- or diglycerides.
  • fatty acids such as oleic acid find use in the preparation of injectables.
  • Compounds of Formula A may also be administered in the form of a suppositories for rectal administration of the drug.
  • These compositions can be prepared by mixing the drug with a suitable non- irritating excipient which is solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum to release the drug.
  • suitable non- irritating excipient include cocoa butter, glycerinated gelatin, hydrogenated vegetable oils, mixtures of polyethylene glycols of various molecular weights and fatty acid esters of polyethylene glycol.
  • compositions, ointments, jellies, solutions or suspensions, etc., containing the compound of Formula A are employed.
  • topical application shall include mouth washes and gargles.
  • the compounds for the present invention can be administered in intranasal form via topical use of suitable intranasal vehicles and delivery devices, or via transdermal routes, using those forms of transdermal skin patches well known to those of ordinary skill in the art.
  • the dosage administration will, of course, be continuous rather than intermittent throughout the dosage regimen.
  • composition is intended to encompass a product comprising the specified ingredients in the specific amounts, as well as any product which results, directly or indirectly, from combination of the specific ingredients in the specified amounts.
  • the daily dosage will normally be determined by the prescribing physician with the dosage generally varying according to the age, weight, sex and response of the individual patient, as well as the severity of the patient's symptoms.
  • a suitable amount of compound is administered to a mammal undergoing treatment for cancer.
  • Administration occurs in an amount between about 0.1 mg/kg of body weight to about 60 mg/kg of body weight per day, preferably of between 0.5 mg/kg of body weight to about 40 mg/kg of body weight per day.
  • the compounds of the instant invention may also be co-administered with other well known therapeutic agents that are selected for their particular usefulness against the condition that is being treated.
  • the compounds of the instant invention may also be co-administered with other well known cancer therapeutic agents that are selected for their particular usefulness against the condition that is being treated. Included in such combinations of therapeutic agents are combinations of the instant farnesyl-protein transferase inhibitors and an antineoplastic agent. It is also understood that such a combination of antineoplastic agent and inhibitor of farnesyl-protein transferase may be used in conjunction with other methods of treating cancer and/or tumors, including radiation therapy and surgery.
  • antineoplastic agent examples include, in general, microtubule-stabilizing agents (such as paclitaxel (also known as Taxol®), docetaxel (also known as Taxotere®), epothilone A, epothilone B, desoxy epothilone A, desoxyepothilone B or their derivatives); microtubule-disruptor agents; alkylating agents, anti-metabolites; epidophyllotoxin; an antineoplastic enzyme; a topoisomerase inhibitor; procarbazine; mitoxantrone; platinum coordination complexes; biological response modifiers and growth inhibitors; hormonal/anti- hormonal therapeutic agents and haematopoietic growth factors.
  • microtubule-stabilizing agents such as paclitaxel (also known as Taxol®), docetaxel (also known as Taxotere®), epothilone A, epothilone B, desoxy epothilone A, desoxyepoth
  • Example classes of antineoplastic agents include, for example, the anthracycline family of drugs, the vinca drugs, the mitomycins, the bleomycins, the cytotoxic nucleosides, the taxanes, the epothilones, discodermolide, the pteridine family of drugs, diynenes and the podophyllotoxins.
  • Particularly useful members of those classes include, for example, doxorubicin, carminomycin, daunorubicin, aminopterin, methotrexate, methopterin, dichloro-methotrexate, mitomycin C, porfiromycin, 5-fluorouracil, 6-mercaptopurine, gemcitabine, cytosine arabinoside, podophyllotoxin or podo-phyllotoxin derivatives such as etoposide, etoposide phosphate or teniposide, melphalan, vinblastine, vincristine, leurosidine, vindesine, leurosine, paclitaxel and the like.
  • antineoplastic agents include estramustine, cisplatin, carboplatin, cyclophosphamide, bleomycin, tamoxifen, ifosamide, melphalan, hexamethyl melamine, thiotepa, cytarabin, idatrexate, trimetrexate, dacarbazine, L-asparaginase, camptothecin, CPT-11, topotecan, ara-C, bicalutamide, flutamide, leuprolide, pyridobenzoindole derivatives, interferons and interleukins.
  • the preferred class of antineoplastic agents is the taxanes and the preferred antineoplastic agent is paclitaxel.
  • Radiation therapy including x-rays or gamma rays which are delivered from either an externally applied beam or by implantation of tiny radioactive sources, may also be used in combination with the instant inhibitor of farnesyl-protein transferase alone to treat cancer.
  • compounds of the instant invention may also be useful as radiation sensitizers, as described in WO 97/38697, published on October 23, 1997, and herein incorporated by reference.
  • the instant compounds may also be useful in combination with other inhibitors of parts of the signaling pathway that links cell surface growth factor receptors to nuclear signals initiating cellular proliferation.
  • the instant compounds may be utilized in combination with farnesyl pyrophosphate competitive inhibitors of the activity of farnesyl-protein transferase or in combination with a compound which has Raf antagonist activity.
  • the instant compounds may also be co-administered with compounds that are selective inhibitors of geranylgeranyl protein transferase or farnesyl-protein transferase.
  • such administration can be orally or parenterally, including intravenous, intramuscular, intraperitoneal, subcutaneous, rectal and topical routes of administration. It is preferred that such administration be orally. It is more preferred that such administration be orally and simultaneously.
  • the protein substrate-competitive inhibitor and farnesyl pyrophosphate-competitive inhibitor are administered sequentially, the administration of each can be by the same method or by different methods.
  • the instant compounds may also be useful in combination with an integrin antagonist for the treatment of cancer, as described in U.S. Ser. No. 09/055,487, filed April 6, 1998, which is incorporated herein by reference.
  • an integrin antagonist refers to compounds which selectively antagonize, inhibit or counteract binding of a physiological ligand to an integrin(s) that is involved in the regulation of angiogenisis, or in the growth and invasiveness of tumor cells.
  • the term refers to compounds which selectively antagonize, inhibit or counteract binding of a physiological ligand to the avb3 integrin, which selectively antagonize, inhibit or counteract binding of a physiological ligand to the avb ⁇ integrin, which antagonize, inhibit or counteract binding of a physiological ligand to both the avb3 integrin and the avb ⁇ integrin, or which antagonize, inhibit or counteract the activity of the particular integrin(s) expressed on capillary endothelial cells.
  • the term also refers to antagonists of the avb ⁇ , avb8, albl, a2bl, a ⁇ bl, a ⁇ bl and a6b4 integrins.
  • the term also refers to antagonists of any combination of avb3, avb ⁇ , avb6, avb8, albl, a2bl, a ⁇ bl, a ⁇ bl and a6b4 integrins.
  • the instant compounds may also be useful with other agents that inhibit angiogenisis and thereby inhibit the growth and invasiveness of tumor cells, including, but not limited to angiostatin and endostatin.
  • the instant compounds may be useful in combination with agents that are effective in the treatment and prevention of NF-1, restenosis, polycystic kidney disease, infections of hepatitis delta and related viruses and fungal infections.
  • combination products employ the combinations of this invention within the dosage range described below and the other pharmaceutically active agent(s) within its approved dosage range.
  • Combinations of the instant invention may alternatively be used sequentially with known pharmaceutically acceptable agent(s) when a multiple combination formulation is inappropriate.
  • Step A Preparation of l-triphenylmethyl-4-(hydroxymethyl)- imidazole To a solution of 4-(hydroxymethyl)imidazole hydrochloride (3 ⁇ .O g, 260 mmol) in 2 ⁇ 0 mL of dry DMF at room temperature was added triethylamine (90.6 mL, 6 ⁇ 0 mmol). A white solid precipitated from the solution. Chlorotriphenylmethane (76.1 g, 273 mmol) in ⁇ OO mL of DMF was added dropwise. The reaction mixture was stirred for 20 hours, poured over ice, filtered, and washed with ice water. The resulting product was slurried with cold dioxane, filtered, and dried in vacuo to provide the titled product as a white solid which was sufficiently pure for use in the next step.
  • Step B Preparation of l-triphenylmethyl-4-(acetoxymethyl)- imidazole
  • Step C Preparation of l-(4-cyanobenzyl)- ⁇ -(acetoxymethyl)- imidazole hydrobromide
  • a solution of the product from Step B (8 ⁇ .8 g, 22 ⁇ mmol) and a-bromo-p-tolunitrile ( ⁇ .1 g, 232 mmol) in ⁇ OO mL of EtOAc was stirred at 60°C for 20 hours, during which a pale yellow precipitate formed.
  • the reaction was cooled to room temperature and filtered to provide the solid imidazolium bromide salt.
  • the filtrate was concentrated in vacuo to a volume 200 mL, reheated at 60°C for two hours, cooled to room temperature, and filtered again.
  • the filtrate was concentrated in vacuo to a volume 100 mL, reheated at 60°C for another two hours, cooled to room temperature, and concentrated in vacuo to provide a pale yellow solid. All of the solid material was combined, dissolved in ⁇ OO mL of methanol, and warmed to 60°C.
  • Step D Preparation of l-(4-cyanobenzyl)-5-(hydroxymethyl)- imidazole
  • Step E Preparation of l-(4-cyanobenzyl)- ⁇ - imidazolecarboxaldehyde
  • Step F Preparation of N-(3-chlorophenyl)ethylenediamine hydrochloride To a solution of 3-chloroaniline (30.0 mL, 284 mmol) in
  • the amine hydrochloride from Step F (ca. 282 mmol, crude material prepared above) was taken up in 500 mL of THF and 500 mL of sat. aq. NaHCO3 soln., cooled to 0°C, and di-tert- butylpyrocarbonate (61.6 g, 282 mmol) was added. After 30 h, the reaction was poured into EtOAc, washed with water and brine, dried (Na2SO4), filtered, and concentrated in vacuo to provide the titled carbamate as a brown oil which was used in the next step without further purification.
  • Step H Preparation of N-[2-(tert-butoxycarbamoyl)ethyl]-N-(3- chlorophenyl)-2-chloroacetamide
  • Step J Preparation of l-(3-chlorophenyl)-2-piperazinone Through a solution of Boc-protected piperazinone from Step I (17.19 g, 5 ⁇ .4 mmol) in ⁇ OO L of EtOAc at -78°C was bubbled anhydrous HCl gas. The saturated solution was warmed to 0°C, and stirred for 12 hours. Nitrogen gas was bubbled through the reaction to remove excess HCl, and the mixture was warmed to room temperature. The solution was concentrated in vacuo to provide the hydrochloride as a white powder. This material was taken up in 300 mL of CH2CI2 and treated with dilute aqueous NaHCO3 solution.
  • the aqueous phase was extracted with CH2CI2 (8 x 300 mL) until tic analysis indicated complete extraction.
  • the combined organic mixture was dried (Na2SO4), filtered, and concentrated in vacuo to provide the titled free amine as a pale brown oil.
  • Step K Preparation of l-(3-chlorophenyl)-4-[l-(4-cyanobenzyl)-5- imidazolylmethyl] -2-piperazinone dihydrochloride
  • 4A powdered molecular sieves 10 g
  • sodium triacetoxy- borohydride 17.7 g, 83.3 mmol
  • the imidazole carboxaldehyde from Step E of Example 1 (11.9 g, 56.4 mmol) was added, and the reaction was stirred at 0°C. After 26 hours, the reaction was poured into
  • Examples 2 and 3 were prepared using the above protocol, which describes the synthesis of the structurally related compound Table 1 lists other compounds of the instant invention that were prepared using the procedure described in Example 1. In Step F, the appropriately substituted aniline was used in place of 3- chloroaniline.
  • Step D Preparation of Methyl 4-Cyano-3-methoxybenzoate Sodium hydride (9 g, 0.24 mol as 60% wt. disp. mineral oil) was aded to a solution of the phenol from Step C (36.1 g, 204 mmol) in 400 mL of dry DMF at room temperature. Iodomethane was added
  • Step G Preparation of l-(4-cyano-3-methoxybenzyl)- ⁇ - (acetoxymethyl)-imidazole hydrobromide
  • the titled product was prepared by reacting the bromide from Step F (21.7 g, 96 mmol) with the imidazole product from Step B of Example 1 (34.9 g, 91 mmol) using the procedure outlined in Step C of Example 1.
  • the crude product was triturated with hexane to provide the titled product hydrobromide.
  • Step H Preparation of l-(4-cyano-3-methoxybenzyl)- ⁇ -
  • the titled product was prepared by hydrolysis of the acetate from Step G (19.43 g, 68.1 mmol) using the procedure outlined in Step D of Example 1.
  • the crude titled product was isolated in modest yield (11 g, 66% yield). Concentration of the aqueous extracts provided solid material (ca. 100 g) which contained a significant quantity of the titled product , as judged by H NMR spectroscopy.
  • Step I Preparation of l-(4-cyano-3-methoxybenzyl)- ⁇ - imidazolecarboxaldehvde
  • the titled product was prepared by oxidizing the alcohol from Step H (11 g, 4 ⁇ mmol) using the procedure outlined in Step E of Example 1.
  • the titled aldehyde was isolated as a white powder which was sufficiently pure for use in the next step without further purification.
  • Step J Preparation of l-(3-chlorophenyl)-4-[l-(4-cyano-3- methoxybenzyl)- ⁇ -imidazolylmethyl] -2-piperazinone dihydrochloride
  • the titled product was prepared by reductive alkylation of the aldehyde from Step I (8 ⁇ 9 mg, 3. ⁇ 6 mmol) and the amine (hydrochloride) from Step K of Example 1 (800 mg, 3.24 mmol) using the procedure outlined in Step H of Example 1. Purification by flash column chromatography through silica gel ( ⁇ 0%-7 ⁇ % acetone CH2CI2) and conversion of the resulting white foam to its dihydrochloride salt provided the titled product as a white powder. FAB ms (m+1) 437.
  • Steps F-J of Example 1 This amine (1.7 ⁇ g, ⁇ .93 mmol) was coupled to the aldehyde from Step I of Example 6 (l. ⁇ 7 g, 6. ⁇ 2 mmol) using the procedure outlined in Step H of Example 1. Purification by flash column chromatography through silica gel (60%- 100% acetone CH2CI2) and conversion of the resulting white foam to its dihydrochloride salt provided the titled product as a white powder.
  • Steps A-E Preparation of (R)- ⁇ -[(benzyloxy)methyl]-l-(3- ehlorophenyl)-2-piperazinone hydrochloride: The titled compound was prepared using an adaptation of the following protocol, which describes the synthesis of the structurally related compound ⁇ (S)-n-butyl-l-(2,3-dimethylphenyl)- 2-piperazinone hydrochloride.
  • N-Boc-Ser(OBn)-OH was used instead of 2(S)-(butoxycarbonylamino)hexanoic acid.
  • Step A N-Methoxy-N-methyl 2(S)-(tert-butoxycarbonylamino)- hexanamide
  • Step B 2(S)-(tert-Butoxycarbonylamino)hexanal
  • Step C N-(2,3-Dimethylphenyl)-2(S)-(tert-butoxycarbonylamino)- hexanamine
  • 2,3-Dimethylaniline (8.32 mL, 68.3 mmol) was dissolved in dichloroethane under nitrogen. Acetic acid was added to obtain pH ⁇ , and sodium triacetoxyborohydride (17.2 g, 80.8 mmol) and crushed molecular sieves (4 g) were added.
  • a solution of the product from Step B (13.3 g, 62.1 mmol) in dichloroethane (80 mL) was added slowly dropwise at 20°C. The reaction was stirred overnight, then quenched with saturated sodium bicarbonate solution. The aqueous layer was removed, the organic phase washed with saturated brine and dried over magnesium sulfate. Crystallization from hexane gave the title compound.
  • Step D 4-tert-Butoxycarbonyl- ⁇ (S)-n-butyl-l-(2,3- dimethylphenyl)-2-piperazinone
  • Step E ⁇ (S)-n-Butyl-l-(2.3-dimethylphenyl)-2-piperazinone
  • ethyl acetate ⁇ O mL
  • Step F Preparation of (R)-5-[(Benzyloxy)methyl]-l-(3- chlorophenyl)-4-[l-(4-cyanobenzyl)- ⁇ -imidazolylmethyl]-
  • Step B Preparation of methyl 4-cyano-3-(trifluoromethoxy) benzoate
  • Step C Preparation of 4-cyano-3-(trifluoromethoxy) benzyl alcohol
  • Step D Preparation of l-[4-cyano-3-(trifluoromethoxy)benzyl]-
  • Step E Preparation of l-[4-cyano-3-(trifluoromethoxy)benzyl]- ⁇
  • Step F Preparation of l-[4-cyano-3-(trifluoromethoxy)benzyl] imidazole- ⁇ -carboxaldehvde
  • the titled compound was prepared from the product of Step E (1.31 g, 4.41 mmol) using the procedure described in Step E of Example 1. This provided the titled product.
  • Step G Preparation of l-(3-Chlorophenyl)-4-[l-(4-cyano-3-
  • Step A Preparation of l-[3-(trifluoromethoxy)phenyl]-2- piperazinone hydrochloride
  • the titled compound was prepared from 3-
  • Step B Preparation of 4-[l-(4-cyano-3-(trifluoromethoxy)benzyl)- ⁇ -imidazolylmethyl] - 1- [3-( trifluoromethoxy)phenyl] -2- piperazinone dihydrochloride
  • the titled compound was prepared from the product of Step A and the product of Step F of Example 9 using the procedure described in Step K of Example 1. Purification by silica gel column chromatography ( ⁇ 0-6 ⁇ % acetone/dichloro methane) and conversion to the dihydrochloride salt using excess ethereal HCl solution gave the titled product as a white powder. FAB ms (m+1) ⁇ 40.2.
  • Step C Preparation of l-(4-cyano-3-fluorobenzyl)-5-(acetoxymethyl)- imidazole hydrobromide
  • Step D Preparation of l-(4-cyano-3-fluorobenzyl)-5- (hydroxymethyl)imidazole
  • Step E Preparation of l-(4-cyano-3-fluorobenzyl)-5- imidazolecarboxaldehyde
  • DMSO DMSO
  • triethylamine 5.6 mL, 40 mmol
  • SO3-pyridine complex 3.89 g, 25 mmol
  • the reaction was poured into EtOAc, washed with water and brine, dried (Na2SO4), filtered, and concentrated in vacuo to provide the aldehyde as a pale yellow powder which was sufficiently pure for use in the next step without further purification.
  • Step F Preparation of l-(3-Chlorophenyl)-4-[l-(4-cyano-3- fluorobenzyl)- ⁇ -imidazolylmethyl] - 2-piperazinone dihydrochloride
  • the titled compound was prepared from the product of
  • Example 11 product To a solution of the Example 11 product ( ⁇ 2 mg, 0.12 mmol) in 1 mL of DMF was added sodium thiomethoxide (17 mg, 0.24 mmol). After ca. 16 hours, the reaction was diluted with EtOAc and extracted with saturated NaHCO 3 solution and brine, dried over sodium sulfate, filtered, and concentrated in vacuo. Purification by silica gel preparative thin-layer chromatography (2 x O. ⁇ mm, 10% CHCl 3 /methanol) and conversion to the dihydrochloride salt using excess ethereal HCl solution gave the titled product as a white powder.
  • Example 11 To a solution of the Example 11 product ( ⁇ O mg, 0.12 mmol) in 1 mL of DMSO was added phenol (33 mg, 0.35 mmol), followed by cesium carbonate (114 mg, 0.35 mmol). After ca. 16 hours, the reaction was diluted with EtOAc and extracted with water and brine, dried over sodium sulfate, filtered, and concentrated in vacuo. Purification by silica gel preparative thin- layer chromatography (2 x 0.5 mm, 90:10:1 CHCl 3 /methanol/NH 4 OH) and conversion to the dihydrochloride salt using excess ethereal HCl solution gave the titled product as a white powder. FAB ms (m+1) 498.2.
  • Isoprenyl-protein transferase activity assays are carried out at 30 °C unless noted otherwise.
  • a typical reaction contains (in a final volume of ⁇ O mL): [ 3 H]farnesyl diphosphate, Ras protein , ⁇ O mM HEPES, pH 7.5, 5 mM MgCl2, ⁇ mM dithiothreitol, 10 mM ZnCl2, 0.1% polyethyleneglycol (PEG) (l ⁇ ,000-20,000 mw) and isoprenyl-protein transferase.
  • PEG polyethyleneglycol
  • the FPTase employed in the assay is prepared by recombinant expression as described in Omer, C.A., Krai, A.M., Diehl, R.E., Prendergast, G.C., Powers, S., Allen,
  • inhibitors are prepared as concentrated solutions in 100% dimethyl sulfoxide and then diluted 20-fold into the enzyme assay mixture.
  • Substrate concentrations for inhibitor IC ⁇ O determinations are as follows: FTase, 650 nM Ras-CVLS (SEQ.ID.NO.: 1), 100 nM farnesyl diphosphate.
  • the modified geranylgeranyl-protein transferase inhibition assay is carried out at room temperature.
  • a typical reaction contains (in a final volume of ⁇ O mL): [ H]geranylgeranyl diphosphate, biotinylated
  • Ras peptide ⁇ O mM HEPES, pH 7.5, a modulating anion (for example 10 mM glycerophosphate or 5mM ATP), 5 mM MgCl2, 10 mM ZnCl2, 0.1%
  • the GGTase-type I enzyme employed in the assay is prepared as described in U.S. Pat. No. 5,470,832, incorporated by reference.
  • the Ras peptide is derived from the K4B-Ras protein and has the following sequence: biotinyl-GKKKKKKSKTKCVIM (single amino acid code) (SEQ.ID.NO.: 2).
  • Reactions are initiated by the addition of GGTase and stopped at timed intervals (typically 15 min) by the addition of 200 mL of a 3 mg/mL suspension of streptavidin SPA beads (Scintillation Proximity Assay beads, Amersham) in 0.2 M sodium phosphate, pH 4, containing 50 mM EDTA, and 0.5% BSA. The quenched reactions are allowed to stand for 2 hours before analysis on a Packard TopCount scintillation counter.
  • streptavidin SPA beads Scintillation Proximity Assay beads
  • the cell line used in this assay is a v-ras line derived from either Ratl or NIH3T3 cells, which expressed viral Ha-ras p21.
  • the assay is performed essentially as described in DeClue, J.E. et al. , Cancer Research 51:712-717. (1991). Cells in 10 cm dishes at 50-7 ⁇ % confluency are treated with the test compound (final concentration of solvent, methanol or dimethyl sulfoxide, is 0.1%).
  • the cells are labeled in 3 ml methionine-free DMEM supple-mented with 10% regular DMEM, 2% fetal bovine serum and 400 mCi[3 ⁇ S]methionine (1000 Ci/mmol).
  • the cells are lysed in 1 ml lysis buffer (1% NP40/20 mM HEPES, pH 7.5/5 mM MgCl2/lmM DTT/10 mg/ml aprotinen/2 mg/ml leupeptin/2 mg/ml antipain/0.5 mM PMSF) and the lysates cleared by centrifugation at 100,000 x g for 4 ⁇ min.
  • the immuno- precipitates are washed four times with IP buffer (20 nM HEPES, pH 7.5/1 mM EDTA/1% Triton X-100.0.5% deoxycholate/0.1%/SDS/0.1 M NaCl) boiled in SDS-PAGE sample buffer and loaded on 13% acrylamide gels. When the dye front reached the bottom, the gel is fixed, soaked in Enlightening, dried and autoradiographed. The intensities of the bands corresponding to farnesylated and nonfarnesylated ras proteins are compared to determine the percent inhibition of farnesyl transfer to protein.
  • IP buffer 20 nM HEPES, pH 7.5/1 mM EDTA/1% Triton X-100.0.5% deoxycholate/0.1%/SDS/0.1 M NaCl
  • Rat 1 cells transformed with either v-ras, v-raf, or v-mos are seeded at a density of 1 x 10 ⁇ cells per plate (35 mm in diameter) in a 0.3% top agarose layer in medium A (Dulbecco's modified Eagle's medium supplemented with 10% fetal bovine serum) over a bottom agarose layer (0.6%). Both layers contain 0.1% methanol or an appropriate concentration of the instant compound (dissolved in methanol at 1000 times the final concentration used in the assay).
  • the cells are fed twice weekly with 0.5 ml of medium A containing 0.1% methanol or the concentration of the instant compound.
  • Photomicrographs are taken 16 days after the cultures are seeded and comparisons are made.
  • the SEAP reporter plasmid, pDSElOO was constructed by ligating a restriction fragment containing the SEAP coding sequence into the plasmid pCMV-RE-AKI.
  • the SEAP gene is derived from the plasmid pSEAP2-Basic (Clontech, Palo Alto, CA).
  • the plasmid pCMV- RE-AKI was constructed by Deborah Jones (Merck) and contains 5 sequential copies of the 'dyad symmetry response element' cloned upstream of a 'CAT-TATA' sequence derived from the cytomegalovirus immediate early promoter.
  • the plasmid also contains a bovine growth hormone poly-A sequence.
  • the plasmid, pDSElOO was constructed as follows. A restriction fragment encoding the SEAP coding sequence was cut out of the plasmid pSEAP2-Basic using the restriction enzymes EcoRl and Hpal. The ends of the linear DNA fragments were filled in with the Klenow fragment of E. coli DNA Polymerase I. The 'blunt ended' DNA containing the SEAP gene was isolated by electrophoresing the digest in an agarose gel and cutting out the 1694 base pair fragment. The vector plasmid pCMV-RE-AKI was linearized with the restriction enzyme Bgl- II and the ends filled in with Klenow DNA Polymerase I.
  • the SEAP DNA fragment was blunt end ligated into the pCMV-RE-AKI vector and the ligation products were transformed into DH ⁇ -alpha E. coli cells (Gibco-BRL). Transformants were screened for the proper insert and then mapped for restriction fragment orientation. Properly oriented recombinant constructs were sequenced across the cloning junctions to verify the correct sequence. The resulting plasmid contains the SEAP coding sequence downstream of the DSE and CAT-TATA promoter elements and upstream of the BGH poly-A sequence.
  • the SEAP repotrer plasmid, pDSElOl is also constructed by ligating a restriction fragment containing the SEAP coding sequence into the plasmid pCMV-RE-AKI.
  • the SEAP gene is derived from plasmid pGEM7zf(-)/SEAP.
  • the plasmid pDSElOl was constructed as follows: A restriction fragment containing part of the SEAP gene coding sequence was cut out of the plasmid pGEM7zf(-)/SEAP using the restriction enzymes Apa I and KpnI. The ends of the linear DNA fragments were chewed back with the Klenow fragment of E. coli DNA Polymerase I. The "blunt ended" DNA containing the truncated SEAP gene was isolated by electrophoresing the digest in an agarose gel and cutting out the 1910 base pair fragment. This 1910 base pair fragment was ligated into the plasmid pCMV-RE-AKI which had been cut with Bgl-II and filled in with E. coli Klenow fragment DNA polymerase.
  • the plasmid pCMV-RE-AKI is derived from plasmid pCMVIE-AKI-DHFR (Whang , Y., Silberklang, M., Morgan, A., Munshi, S., Lenny, A.B., Ellis, R.W., and Kieff, E. (1987) J. Virol., 61, 1796-1807) by removing an EcoRI fragment containing the DHFR and Neomycin markers.
  • the plasmid pGEM7zf(-)/SEAP was constructed as follows.
  • the SEAP gene was PCRed, in two segments from a human placenta cDNA library (Clontech) using the following oligos.
  • Antisense strand N-terminal SEAP 5' GAGAGAGCTCGAGGTTAACCCGGGT
  • Sense strand C-terminal SEAP 5' GAGAGAGTCTAGAGTTAACCCGTGGTCC CCGCGTTGCTTCCT 3' (SEQ.ID.NO.: ⁇ )
  • Antisense strand C-terminal SEAP ⁇ ' GAAGAGGAAGCTTGGTACCGCCACTG GGCTGTAGGTGGTGGCT 3' (SEQ.ID.NO. :6)
  • the N-terminal oligos (SEQ.ID.NO.: 4 and SEQ.ID.NO.: ⁇ ) were used to generate a l ⁇ 60 bp N-terminal PCR product that contained EcoRI and Hpal restriction sites at the ends.
  • the Antisense N-terminal oligo (SEQ.ID.NO.: 4) introduces an internal translation STOP codon within the SEAP gene along with the Hpal site.
  • SEQ.ID.NO.: ⁇ and SEQ.ID.NO.: 6 were used to amplify a 412 bp C- terminal PCR product containing Hpal and Hindlll restriction sites.
  • the sense strand C-terminal oligo (SEQ.ID.NO.: ⁇ ) introduces the internal STOP codon as well as the Hpal site.
  • the N-terminal amplicon was digested with EcoRI and Hpal while the C-terminal amplicon was digested with Hpal and Hindlll.
  • the two fragments comprising each end of the SEAP gene were isolated by electrophoresing the digest in an agarose gel and isolating the l ⁇ 60 and 412 base pair fragments.
  • An expression plasmid constitutively expressing the SEAP protein was created by placing the sequence encoding a truncated SEAP gene downstream of the cytomegalovirus (CMV) IE-1 promoter.
  • the expression plasmid also includes the CMV intron A region ⁇ ' to the SEAP gene as well as the 3' untranslated region of the bovine growth hormone gene 3' to the SEAP gene.
  • the plasmid pCMVIE-AKI-DHFR (Whang et al, 1987) containing the CMV immediate early promoter was cut with EcoRI generating two fragments. The vector fragment was isolated by agarose electrophoresis and religated. The resulting plasmid is named pCMV- AKI.
  • the cytomegalovirus intron A nucleotide sequence was inserted downstream of the CMV IE1 promter in pCMV-AKI.
  • the intron A sequence was isolated from a genomic clone bank and subcloned into pBR322 to generate plasmid pl6T-286.
  • the intron A sequence was mutated at nucleotide 18 ⁇ 6 (nucleotide numbering as in Chapman, B.S., Thayer, R.M., Vincent, K.A. and Haigwood, N.L., Nuc.Acids Res. 19, 3979-3986) to remove a Sad restriction site using site directed mutagenesis.
  • the mutated intron A sequence was PCRed from the plasmid pl6T-287 using the following oligos.
  • Sense strand ⁇ ' GGCAGAGCTCGTTTAGTGAACCGTCAG 3' (SEQ.ID.NO.: 7)
  • Antisense strand ⁇ ' GAGAGATCTCAAGGACGGTGACTGCAG 3' (SEQ.ID.NO.: 8)
  • oligos generate a 991 base pair fragment with a Sad site incorporated by the sense oligo and a Bgl-II fragment incorporated by the antisense oligo.
  • the PCR fragment is trimmed with Sad and Bgl-II and isolated on an agarose gel.
  • the vector pCMV-AKI is cut with Sad and Bgl-II and the larger vector fragment isolated by agarose gel electrophoresis.
  • the two gel isolated fragments are ligated at their respective Sad and Bgl-II sites to create plasmid pCMV-AKI- InA.
  • the DNA sequence encoding the truncated SEAP gene is inserted into the pCMV-AKI-InA plasmid at the Bgl-II site of the vector.
  • the SEAP gene is cut out of plasmid pGEM7zf(-)/SEAP (described above) using EcoRI and Hindlll. The fragment is filled in with Klenow DNA polymerase and the 1970 base pair fragment isolated from the vector fragment by agarose gel electrophoresis.
  • the pCMV-AKI-InA vector is prepared by digesting with Bgl-II and filling in the ends with Klenow DNA polymerase. The final construct is generated by blunt end ligating the SEAP fragment into the pCMV-AKI-InA vector.
  • Transformants were screened for the proper insert and then mapped for restriction fragment orientation. Properly oriented recombinant constructs were sequenced across the cloning junctions to verify the correct sequence.
  • the resulting plasmid named pCMV-SEAP, contains a modified SEAP sequence downstream of the cytomegalovirus immediately early promoter IE-1 and intron A sequence and upstream of the bovine growth hormone poly-A sequence.
  • the plasmid expresses SEAP in a constitutive manner when transfected into mammalian cells.
  • a DNA fragment containing viral-H-ras can be PCRed from plasmid "H-l” (Ellis R. et al. J. Virol. 36, 408, 1980) or "HB-11 (deposited in the ATCC under Budapest Treaty on August 27, 1997, and designated ATCC 209,218) using the following oligos.
  • Antisense ⁇ 'CACATCTAGATCAGGACAGCACAGACTTGCAGC 3'. (SEQ.ID.NO.: 10)
  • the sense strand oligo also optimizes the 'Kozak' translation initiation sequence immediately ⁇ ' to the ATG start site.
  • cysteine 186 would be mutated to a serine by substituting a G residue for a C residue in the C-terminal antisense oligo.
  • the PCR primer oligos introduce an Xhol site at the ⁇ ' end and a Xbal site at the 3'end.
  • the Xhol-Xbal fragment can be ligated into the mammalian expression plasmid pCI (Promega) cut with Xhol and Xbal. This results in a plasmid in which the recombinant myr-viral-H-ras gene is constitutively transcribed from the CMV promoter of the pCI vector.
  • a viral-H-ras clone with a C-terminal sequence encoding the amino acids CVLL can be cloned from the plasmid "H-l” (Ellis R. et al. J. Virol. 36, 408, 1980) or "HB-11 (deposited in the ATCC under Budapest Treaty on August 27, 1997, and designated ATCC 209,218) by PCR using the following oligos.
  • Antisense strand ⁇ 'CACTCTAGACTGGTGTCAGAGCAGCACACACTTGCAGC-3' (SEQ.ID.NO.: 12)
  • the sense strand oligo optimizes the 'Kozak' sequence and adds an Xhol site.
  • the antisense strand mutates serine 189 to leucine and adds an Xbal site.
  • the PCR fragment can be trimmed with Xhol and Xbal and ligated into the Xhol-Xbal cut vector pCI (Promega). This results in a plasmid in which the mutated viral-H-ras-CVLL gene is constitutively transcribed from the CMV promoter of the pCI vector.
  • the human c-H-ras gene can be PCRed from a human cerebral cortex cDNA library (Clontech) using the following oligonucleotide primers. ⁇ '-GAGAGAATTCGCCACCATGACGGAATATAAGCTGGTGG-3' (SEQ.ID.NO.: 13)
  • Antisense strand ⁇ '-GAGAGTCGACGCGTCAGGAGAGCACACACTTGC-3' (SEQ.ID.NO.: 14)
  • the primers will amplify a c-H-ras encoding DNA fragment with the primers contributing an optimized 'Kozak' translation start sequence, an EcoRI site at the N-terminus and a Sal I stite at the C-terminal end.
  • the c-H-ras fragment can be ligated ligated into an EcoRI -Sal I cut mutagenesis vector pAlter-1 (Promega). Mutation of glutamine-61 to a leucine can be accomplished using the manufacturer's protocols and the following oligonucleotide: ⁇ '-CCGCCGGCCTGGAGGAGTACAG-3' (SEQ.ID.NO.: l ⁇ )
  • the mutated c-H-ras-Leu61 can be excised from the pAlter-1 vector, using EcoRI and Sal I, and be directly ligated into the vector pCI (Promega) which has been digested with EcoRI and Sal I.
  • the new recombinant plasmid will constitutively transcribe c-H-ras-Leu61 from the CMV promoter of the pCI vector.
  • the human c-N-ras gene can be PCRed from a human cerebral cortex cDNA library (Clontech) using the following oligonucleotide primers.
  • Sense strand ⁇ '-GAGAGAATTCGCCACCATGACTGAGTACAAACTGGTGG-3' (SEQ.ID.NO.: 16)
  • Antisense strand ⁇ '-GAGAGTCGACTTGTTACATCACCACACATGGC-3' (SEQ.ID.NO.:
  • the primers will amplify a c-N-ras encoding DNA fragment with the primers contributing an optimized 'Kozak' translation start sequence, an EcoRI site at the N-terminus and a Sal I stite at the C-terminal end.
  • the c-N-ras fragment can be ligated into an EcoRI -Sal I cut mutagenesis vector pAlter-1 (Promega). Mutation of glycine-12 to a valine can be accomplished using the manufacturer's protocols and the following oligonucleotide:
  • the mutated c-N-ras-Val-12 can be excised from the pAlter- 1 vector, using EcoRI and Sal I, and be directly ligated into the vector pCI (Promega) which has been digested with EcoRI and Sal I.
  • the new recombinant plasmid will constitutively transcribe c-N-ras-Val-12 from the CMV promoter of the pCI vector.
  • the human c-K-ras gene can be PCRed from a human cerebral cortex cDNA library (Clontech) using the following oligonucleotide primers.
  • Sense strand ⁇ '-GAGAGGTACCGCCACCATGACTGAATATAAACTTGTGG-3' (SEQ.ID.NO.: 19)
  • Antisense strand ⁇ '-CTCTGTCGACGTATTTAC ATAATTACACACTTTGTC-3' (SEQ.ID.NO.: 20)
  • the primers will amplify a c-K-ras encoding DNA fragment with the primers contributing an optimized 'Kozak' translation start sequence, a Kpnl site at the N-terminus and a Sal I stite at the C-terminal end.
  • the c-K-ras fragment can be ligated into a Kpnl - Sal I cut mutagenesis vector pAlter-1 (Promega).
  • Mutation of cysteine- 12 to a valine can be accomplished using the manufacturer's protocols and the following oligonucleotide: ⁇ '-GTAGTTGGAGCTGTTGGCGTAGGC-3' (SEQ.ID.NO.: 21)
  • the mutated c-K-ras-Val-12 can be excised from the pAlter-
  • Human C33A cells (human epitheial carcenoma - ATTC collection) are seeded in 10cm tissue culture plates in DMEM + 10% fetal calf serum + IX Pen/Strep + IX glutamine + IX NEAA. Cells are grown at 37°C in a ⁇ % CO2 atmosphere until they reach ⁇ O -80% of confluency. The transient transfection is performed by the CaPO4 method (Sambrook et al., 1989). Thus, expression plasmids for H-ras, N-ras, K-ras, Myr-ras or H-ras-CVLL are co-precipitated with the DSE- SEAP reporter construct.
  • the cells are washed with PBS and trypsinized with 1ml of 0.0 ⁇ % trypsin.
  • the 1 ml of trypsinized cells is diluted into 10ml of phenol red free DMEM + 0.2% charcoal stripped calf serum + IX (Pen/Strep, Glutamine and NEAA ).
  • Transfected cells are plated in a 96 well microtiter plate (lOOml/well) to which drug, diluted in media, has already been added in a volume of 100ml. The final volume per well is 200ml with each drug concentration repeated in triplicate over a range of half-log steps.
  • the heat treated media is assayed for alkaline phosphatase by a luminescence assay using the luminescence reagent CSPD® (Tropix, Bedford, Mass.).
  • a volume of ⁇ O ml media is combined with 200 ml of CSPD cocktail and incubated for 60 minutes at room temperature.
  • Luminesence is monitored using an ML2200 microplate luminometer (Dynatech). Luminescence reflects the level of activation of the fos reporter construct stimulated by the transiently expressed protein.
  • PSN-1 human pancreatic carcinoma
  • viral -K4B-ras- transformed Ratl cells are used for analysis of protein processing.
  • Subconfluent cells in 100 mm dishes are fed with 3.5 ml of media (methionine-free RPMI supplemented with 2% fetal bovine serum or cysteine-free/methionine-free DMEM supplemented with 0.035 ml of 200 mM glutamine (Gibco), 2% fetal bovine serum, respectively) containing the desired concentration of test compound, lovastatin or solvent alone.
  • media methionine-free RPMI supplemented with 2% fetal bovine serum or cysteine-free/methionine-free DMEM supplemented with 0.035 ml of 200 mM glutamine (Gibco), 2% fetal bovine serum, respectively
  • Test compounds are prepared as lOOOx concentrated solutions in DMSO to yield a final solvent concentration of 0.1%. Following incubation at 37°C for two hours 204 ⁇ Ci/ml [ 3 ⁇ S]Pro-Mix (Amersham, cell labeling grade) is added.
  • the cells are incubated at 37°C for an additional period of time (typically 6 to 24 hours). The media is then removed and the cells are washed once with cold PBS. The cells are scraped into 1 ml of cold PBS, collected by centrifugation (10,000 x g for 10 sec at room temperature), and lysed by vortexing in 1 ml of lysis buffer (1% Nonidet P-40, 20 mM HEPES, pH 7.5, 150 mM NaCl, 1 mM EDTA, 0.5% deoxycholate, 0.1% SDS, 1 mM DTT, 10 ⁇ g/ml AEBSF, 10 ⁇ g/ml aprotinin, 2 ⁇ g/ml leupeptin and 2 ⁇ g/ml antipain). The lysate is then centrifuged at l ⁇ ,000 x g for 10 min at 4°C and the supernatant saved.
  • lysis buffer 1% Nonidet P-40, 20 mM HEPES,
  • Ki4B-Ras For immunoprecipitation of Ki4B-Ras, samples of lysate supernatant containing equal amounts of protein are utilized. Protein concentration is determined by the bradford method utilizing bovine serum albumin as a standard. The appropriate volume of lysate is brought to 1 ml with lysis buffer lacking DTT and 8 ⁇ g of the pan Ras monoclonal antibody, Y13-2 ⁇ 9, added. The protein/antibody mixture is incubated on ice at 4°C for 24 hours. The immune complex is collected on pansorbin (Calbiochem) coated with rabbit antiserum to rat IgG (Cappel) by tumbling at 4°C for 4 ⁇ minutes.
  • pansorbin Calbiochem
  • the pellet is washed 3 times with 1 ml of lysis buffer lacking DTT and protease inhibitors and resuspended in 100 ml elution buffer (10 mM Tris pH 7.4, 1% SDS).
  • the Ras is eluted from the beads by heating at 9 ⁇ °C for ⁇ minutes, after which the beads are pelleted by brief centrifugation (l ⁇ ,000 x g for 30 sec. at room temperature).
  • the supernatant is added to 1 ml of Dilution Buffer 0.1% Triton X-100, ⁇ mM EDTA, ⁇ O mM NaCl, 10 mM Tris pH 7.4) with 2 mg Kirsten-ras specific monoclonal antibody, c-K-ras Ab-1 (Calbiochem).
  • the second protein antibody mixture is incubated on ice at 4°C for 1-2 hours.
  • the immune complex is collected on pansorbin (Calbiochem) coated with rabbit antiserum to rat IgG (Cappel) by tumbling at 4°C for 4 ⁇ minutes.
  • the pellet is washed 3 times with 1 ml of lysis buffer lacking DTT and protease inhibitors and resuspended in Laemmli sample buffer.
  • the Ras is eluted from the beads by heating at 9 ⁇ °C for ⁇ minutes, after which the beads are pelleted by brief centrifugation.
  • the supernatant is subjected to SDS-PAGE on a 12% acrylamide gel (bis- acrylamide:acrylamide, 1:100), and the Ras visualized by fluorography.
  • Rapl processing inhibition assay Protocol A Cells are labeled, incubated and lysed as described in
  • Rapl For immunoprecipitation of Rapl, samples of lysate supernatant containing equal amounts of protein are utilized. Protein concentration is determined by the bradford method utilizing bovine serum albumin as a standard. The appropriate volume of lysate is brought to 1 ml with lysis buffer lacking DTT and 2 ⁇ g of the Rapl antibody, Rapl/Krevl (121) (Santa Cruz Biotech), is added. The protein/antibody mixture is incubated on ice at 4°C for 1 hour. The immune complex is collected on pansorbin (Calbiochem) by tumbling at 4°C for 4 ⁇ minutes.
  • the pellet is washed 3 times with 1 ml of lysis buffer lacking DTT and protease inhibitors and resuspended in 100 ml elution buffer (10 mM Tris pH 7.4, 1% SDS).
  • the Rapl is eluted from the beads by heating at 9 ⁇ °C for ⁇ minutes, after which the beads are pelleted by brief centrifugation (l ⁇ ,000 x g for 30 sec. at room temperature). The supernatant is added to 1 ml of Dilution Buffer (0.1%
  • Triton X-100 5 mM EDTA, 50 mM NaCl, 10 mM Tris pH 7.4 with 2 mg Rapl antibody, Rapl/Krevl (121) (Santa Cruz Biotech).
  • the second protein/antibody mixture is incubated on ice at 4°C for 1-2 hours.
  • the immune complex is collected on pansorbin (Calbiochem) by tumbling at 4°C for 4 ⁇ minutes.
  • the pellet is washed 3 times with 1 ml of lysis buffer lacking DTT and protease inhibitors and resuspended in Laemmli sample buffer.
  • the Rapl is eluted from the beads by heating at 9 ⁇ °C for ⁇ minutes, after which the beads are pelleted by brief centrifugation.
  • the supernatant is subjected to SDS-PAGE on a 12% acrylamide gel (bis- acrylamide:acrylamide, 1:100), and the Rapl visualized by fluorography.
  • PSN-1 cells are passaged every 3-4 days in 10cm plates, splitting near-confluent plates 1:20 and 1:40. The day before the assay is set up, ⁇ x 10 6 cells are plated on l ⁇ cm plates to ensure the same stage of confluency in each assay.
  • the media for these cells is RPMI 1640 (Gibco), with l ⁇ % fetal bovine serum and lx Pen/Strep antibiotic mix.
  • cells are collected from the l ⁇ cm plates by trypsinization and diluted to 400,000 cells/ml in media. O. ⁇ ml of these diluted cells are added to each well of 24-well plates, for a final cell number of 200,000 per well. The cells are then grown at 37°C overnight.
  • the compounds to be assayed are diluted in DMSO in 1/2-log dilutions.
  • the range of final concentrations to be assayed is generally 0.1-100 ⁇ M. Four concentrations per compound is typical.
  • the compounds are diluted so that each concentration is lOOOx of the final concentration (i.e., for a lO ⁇ M data point, a lOmM stock of the compound is needed).
  • RNAse/ DNase mix is added per well. This mix is lmg/ml DNasel (Worthington Enzymes), 0.25mg/ml Rnase A (Worthington Enzymes), 0.5M Tris-HCl pH8.0 and 50mM MgCl 2 .
  • the plate is left on ice for 10 minutes. Samples are then either loaded on the gel, or stored at -70°C until use. Each assay plate (usually 3 compounds, each in 4- point titrations, plus controls) requires one 15-well 14% Novex gel. 25 ⁇ l of each sample is loaded onto the gel. The gel is run at 15mA for about 3. ⁇ hours. It is important to run the gel far enough so that there will be adequate separation between 21kd (Rapl) and 29kd (Rab6). The gels are then transferred to Novex pre-cut PVDF membranes for 1.5 hours at 30V (constant voltage).
  • the membranes are blocked overnight in 20ml Western blocking buffer (2% nonfat dry milk in Western wash buffer (PBS + 0.1% Tween-20). If blocked over the weekend, 0.02% sodium azide is added. The membranes are blocked at 4°C with slow rocking.
  • Western blocking buffer 2% nonfat dry milk in Western wash buffer (PBS + 0.1% Tween-20). If blocked over the weekend, 0.02% sodium azide is added. The membranes are blocked at 4°C with slow rocking.
  • the blocking solution is discarded and 20ml fresh blocking solution containing the anti Rapla antibody (Santa Cruz Biochemical SC1482) at 1:1000 (diluted in Western blocking buffer) and the anti Rab6 antibody (Santa Cruz Biochemical SC310) at 1:5000 (diluted in Western blocking buffer) are added.
  • the membranes are incubated at room temperature for 1 hour with mild rocking.
  • the blocking solution is then discarded and the membrane is washed 3 times with Western wash buffer for l ⁇ minutes per wash.
  • ECF detection reagent About 2ml per gel of the Amersham ECF detection reagent is placed on an overhead transparency (ECF) and the PVDF membranes are placed face-down onto the detection reagent. This is incubated for one minute, then the membrane is placed onto a fresh transparency sheet.
  • ECF overhead transparency
  • the developed transparency sheet is scanned on a phosphorimager and the Rapla Minimum Inhibitory Concentration is determined from the lowest concentration of compound that produces a detectable Rapla Western signal.
  • the Rapla antibody used recognizes only unprenylated/unprocessed Rapla, so that the precence of a detectable Rapla Western signal is indicative of inhibition of Rapla prenylation.
  • mice in each oncogene group are randomly assigned to a vehicle, compound or combination treatment group. Animals are dosed subcutaneously starting on day 1 and daily for the duration of the experiment. Alternatively, the farnesyl-protein transferase inhibitor may be administered by a continuous infusion pump. Compound, compound combination or vehicle is delivered in a total volume of 0.1 ml. Tumors are excised and weighed when all of the vehicle-treated animals exhibited lesions of 0.5 - 1.0 cm in diameter, typically 11-15 days after the cells were injected. The average weight of the tumors in each treatment group for each cell line is calculated.

Landscapes

  • Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • General Chemical & Material Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Cardiology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Virology (AREA)
  • Communicable Diseases (AREA)
  • Oncology (AREA)
  • Ophthalmology & Optometry (AREA)
  • Urology & Nephrology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Plural Heterocyclic Compounds (AREA)

Abstract

L'invention concerne des composés qui inhibent les prényl-protéine-transférases, la farnésyl-protéine-transférase et la géranylgéranyl-protéine-transférase de type I, de même que la prénylation de la protéine oncogène Ras. L'invention concerne également des compositions chimiothérapeutiques renfermant les composés considérés, ainsi que des procédés permettant d'inhiber la farnésyl-protéine-transférase, la géranylgéranyl-protéine-transférase de type I et la prénylation de la protéine oncogène Ras.
PCT/US1998/017696 1997-08-27 1998-08-26 Inhibiteurs de prenyl-proteine-transferases WO1999009985A1 (fr)

Priority Applications (4)

Application Number Priority Date Filing Date Title
EP98943406A EP1014984A1 (fr) 1997-08-27 1998-08-26 Inhibiteurs de prenyl-proteine-transferases
AU91213/98A AU741725B2 (en) 1997-08-27 1998-08-26 Inhibitors of prenyl-protein transferase
CA002301770A CA2301770A1 (fr) 1997-08-27 1998-08-26 Inhibiteurs de prenyl-proteine-transferases
JP2000507375A JP2001513561A (ja) 1997-08-27 1998-08-26 プレニル蛋白トランスフェラーゼ阻害薬

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US5708097P 1997-08-27 1997-08-27
US60/057,080 1997-08-27
GBGB9800975.6A GB9800975D0 (en) 1998-01-16 1998-01-16 Inhibitors of prenyl-protein transferase
GB9800975.6 1998-01-16

Publications (1)

Publication Number Publication Date
WO1999009985A1 true WO1999009985A1 (fr) 1999-03-04

Family

ID=26312960

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1998/017696 WO1999009985A1 (fr) 1997-08-27 1998-08-26 Inhibiteurs de prenyl-proteine-transferases

Country Status (5)

Country Link
EP (1) EP1014984A1 (fr)
JP (1) JP2001513561A (fr)
AU (1) AU741725B2 (fr)
CA (1) CA2301770A1 (fr)
WO (1) WO1999009985A1 (fr)

Cited By (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001060815A1 (fr) * 2000-02-18 2001-08-23 Merck & Co., Inc. Inhibiteurs de prenyle proteine transferases
EP1158984A1 (fr) * 1998-10-29 2001-12-05 Merck & Co., Inc. Inhibiteurs de prenyl-proteine transferase
US6335343B1 (en) 1999-03-03 2002-01-01 Merck & Co., Inc. Inhibitors of prenyl-protein transferase
US6355643B1 (en) 1999-03-03 2002-03-12 Merck & Co., Inc. Inhibitors of prenyl-protein transferase
US6358956B1 (en) 1999-03-03 2002-03-19 Merck & Co., Inc. Inhibitors of prenyl-protein transferase
US6376496B1 (en) 1999-03-03 2002-04-23 Merck & Co., Inc. Inhibitors of prenyl-protein transferase
US6413964B1 (en) 2000-01-12 2002-07-02 Desolms S. Jane Inhibitors of prenyl-protein transferase
WO2003017939A2 (fr) * 2001-08-24 2003-03-06 Yale University Composes de piperazinone utilises comme agents antitumoraux et anticancereux et procedes de traitement
US6534506B2 (en) 2000-04-10 2003-03-18 Merck & Co., Inc. Inhibitors of prenyl-protein transferase
US6562823B1 (en) 1998-07-02 2003-05-13 Merck & Co., Inc. Inhibitors of prenyl-protein transferase
US6593455B2 (en) 2001-08-24 2003-07-15 Tripep Ab Tripeptide amides that block viral infectivity and methods of use thereof
US6951861B1 (en) 1999-10-07 2005-10-04 Smithkline Beechan Corporation Chemical compounds
US7012129B2 (en) 2001-09-19 2006-03-14 Tripep Ab Antiviral composition comprising glycine amide
US7060702B2 (en) 2000-10-17 2006-06-13 Smithkline Beecham Corporation Chemical compounds
US7189713B2 (en) 2002-02-08 2007-03-13 Glaxo Group Limited Piperidine derivatives
US7276509B2 (en) 2002-02-08 2007-10-02 Glaxo Group Limited Piperidine derivatives and their use as antagonists of tachykinins
USRE39921E1 (en) 1999-10-07 2007-11-13 Smithkline Beecham Corporation Chemical compounds
US7482365B2 (en) 2002-02-08 2009-01-27 Glaxo Group Limited Piperidylcarboxamide derivatives and their use in the treatment of tachykinin-mediated diseases
AU2007229363B2 (en) * 2001-08-24 2009-06-04 University Of South Florida Piperazinone Compounds as Anti-Tumor and Anti-Cancer Agents and the Methods of Treatment
EP3068390A1 (fr) * 2013-11-12 2016-09-21 Merck Sharp & Dohme Corp. Dérivés de triazole et d'imidazole à liaison pipéridine ou pipérazine et leurs procédés d'utilisation pour améliorer la pharmacocinétique d'un médicament

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP4965130B2 (ja) * 2006-01-26 2012-07-04 日本臓器製薬株式会社 乾式直打速崩壊性錠剤

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1997036593A1 (fr) * 1996-04-03 1997-10-09 Merck & Co., Inc. Inhibiteurs de la farnesyle-proteine transferase

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1997036593A1 (fr) * 1996-04-03 1997-10-09 Merck & Co., Inc. Inhibiteurs de la farnesyle-proteine transferase

Cited By (33)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6562823B1 (en) 1998-07-02 2003-05-13 Merck & Co., Inc. Inhibitors of prenyl-protein transferase
EP1158984A1 (fr) * 1998-10-29 2001-12-05 Merck & Co., Inc. Inhibiteurs de prenyl-proteine transferase
EP1158984A4 (fr) * 1998-10-29 2003-05-21 Merck & Co Inc Inhibiteurs de prenyl-proteine transferase
US6355643B1 (en) 1999-03-03 2002-03-12 Merck & Co., Inc. Inhibitors of prenyl-protein transferase
US6358956B1 (en) 1999-03-03 2002-03-19 Merck & Co., Inc. Inhibitors of prenyl-protein transferase
US6376496B1 (en) 1999-03-03 2002-04-23 Merck & Co., Inc. Inhibitors of prenyl-protein transferase
US6335343B1 (en) 1999-03-03 2002-01-01 Merck & Co., Inc. Inhibitors of prenyl-protein transferase
US7625904B2 (en) 1999-10-07 2009-12-01 Smithkline Beecham Corporation Methods for the treatment of sleep disorders
US7071196B2 (en) 1999-10-07 2006-07-04 Smithkline Beecham Croporation Chemical compounds
US7345041B2 (en) 1999-10-07 2008-03-18 Smithkline Beecham Corporation Chemical compounds
USRE39921E1 (en) 1999-10-07 2007-11-13 Smithkline Beecham Corporation Chemical compounds
US6951861B1 (en) 1999-10-07 2005-10-04 Smithkline Beechan Corporation Chemical compounds
US6413964B1 (en) 2000-01-12 2002-07-02 Desolms S. Jane Inhibitors of prenyl-protein transferase
WO2001060815A1 (fr) * 2000-02-18 2001-08-23 Merck & Co., Inc. Inhibiteurs de prenyle proteine transferases
US6534506B2 (en) 2000-04-10 2003-03-18 Merck & Co., Inc. Inhibitors of prenyl-protein transferase
US7294630B2 (en) 2000-10-17 2007-11-13 Smithkline Beecham Corporation Piperazinyl piperidine tachykinin antagonists
US7648990B2 (en) 2000-10-17 2010-01-19 Glaxosmithkline Llc Chemical compounds
US7119092B2 (en) 2000-10-17 2006-10-10 Smithkline Beecham Corporation Chemical compounds
US7060702B2 (en) 2000-10-17 2006-06-13 Smithkline Beecham Corporation Chemical compounds
US6593455B2 (en) 2001-08-24 2003-07-15 Tripep Ab Tripeptide amides that block viral infectivity and methods of use thereof
US7763620B2 (en) 2001-08-24 2010-07-27 Yale University Piperazinone compounds as anti-tumor and anti-cancer agents and methods of treatment
AU2002332640B2 (en) * 2001-08-24 2007-11-08 University Of South Florida Piperazinone compounds as anti-tumor and anti-cancer agents and methods of treatment
WO2003017939A3 (fr) * 2001-08-24 2003-11-13 Univ Yale Composes de piperazinone utilises comme agents antitumoraux et anticancereux et procedes de traitement
WO2003017939A2 (fr) * 2001-08-24 2003-03-06 Yale University Composes de piperazinone utilises comme agents antitumoraux et anticancereux et procedes de traitement
AU2007229363B2 (en) * 2001-08-24 2009-06-04 University Of South Florida Piperazinone Compounds as Anti-Tumor and Anti-Cancer Agents and the Methods of Treatment
US7012129B2 (en) 2001-09-19 2006-03-14 Tripep Ab Antiviral composition comprising glycine amide
US7482365B2 (en) 2002-02-08 2009-01-27 Glaxo Group Limited Piperidylcarboxamide derivatives and their use in the treatment of tachykinin-mediated diseases
US7189713B2 (en) 2002-02-08 2007-03-13 Glaxo Group Limited Piperidine derivatives
US7652012B2 (en) 2002-02-08 2010-01-26 Glaxo Group Limited 2-(R)-(4-fluoro-2-methyl-phenyl)-4-(S)-((8aS)-6-oxo-hexahydro-pyrrolo[1,2-a]-pyrazin-2-yl)-piperidine-1-carboxylic acid [1-(R)-3,5-bis-trifluoromethyl-phenyl)-ethyl]-methylamide maleate and pharmaceutical compositions thereof
US7276509B2 (en) 2002-02-08 2007-10-02 Glaxo Group Limited Piperidine derivatives and their use as antagonists of tachykinins
EP3068390A1 (fr) * 2013-11-12 2016-09-21 Merck Sharp & Dohme Corp. Dérivés de triazole et d'imidazole à liaison pipéridine ou pipérazine et leurs procédés d'utilisation pour améliorer la pharmacocinétique d'un médicament
EP3068390A4 (fr) * 2013-11-12 2017-05-10 Merck Sharp & Dohme Corp. Dérivés de triazole et d'imidazole à liaison pipéridine ou pipérazine et leurs procédés d'utilisation pour améliorer la pharmacocinétique d'un médicament
US10745377B2 (en) 2013-11-12 2020-08-18 Merck Sharp & Dohme Corp. Piperidine or piperazine linked imidazole and triazole derivatives and methods of use

Also Published As

Publication number Publication date
EP1014984A1 (fr) 2000-07-05
AU9121398A (en) 1999-03-16
JP2001513561A (ja) 2001-09-04
AU741725B2 (en) 2001-12-06
CA2301770A1 (fr) 1999-03-04

Similar Documents

Publication Publication Date Title
US6387903B1 (en) Inhibitors of prenyl-protein transferase
US6562823B1 (en) Inhibitors of prenyl-protein transferase
AU741725B2 (en) Inhibitors of prenyl-protein transferase
US6358985B1 (en) Inhibitors of prenyl-protein transferase
US20020052363A1 (en) Inhibitors of prenyl-protein transferase
US6358956B1 (en) Inhibitors of prenyl-protein transferase
EP1165084A1 (fr) Inhibiteurs de prenyl-proteine transferases
US20020052380A1 (en) Inhibitors of prenyl-protein transferase
US20020010184A1 (en) Inhibitors of prenyl-protein transferase
US6410534B1 (en) Inhibitors of prenyl-protein transferase
US6376496B1 (en) Inhibitors of prenyl-protein transferase
US6329376B1 (en) Inhibitors of prenyl-protein transferase
US20020193283A1 (en) Inhibitors of prenyl-protein transferase
US6335343B1 (en) Inhibitors of prenyl-protein transferase
US6355643B1 (en) Inhibitors of prenyl-protein transferase
AU762440B2 (en) Inhibitors of prenyl-protein transferase
US6413964B1 (en) Inhibitors of prenyl-protein transferase
WO1999010329A1 (fr) Inhibiteurs de prenyl-proteine-transferase
WO1999010525A1 (fr) Procede pour le traitement du cancer

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AL AM AU AZ BA BB BG BR BY CA CN CU CZ EE GE HR HU ID IL IS JP KG KR KZ LC LK LR LT LV MD MG MK MN MX NO NZ PL RO RU SG SI SK SL TJ TM TR TT UA US UZ VN YU

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): GH GM KE LS MW SD SZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
WWE Wipo information: entry into national phase

Ref document number: 09463917

Country of ref document: US

WWE Wipo information: entry into national phase

Ref document number: 1998943406

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2301770

Country of ref document: CA

Ref country code: CA

Ref document number: 2301770

Kind code of ref document: A

Format of ref document f/p: F

Ref country code: JP

Ref document number: 2000 507375

Kind code of ref document: A

Format of ref document f/p: F

NENP Non-entry into the national phase

Ref country code: KR

WWE Wipo information: entry into national phase

Ref document number: 91213/98

Country of ref document: AU

WWP Wipo information: published in national office

Ref document number: 1998943406

Country of ref document: EP

WWG Wipo information: grant in national office

Ref document number: 91213/98

Country of ref document: AU

WWW Wipo information: withdrawn in national office

Ref document number: 1998943406

Country of ref document: EP