EP0891360A1 - Inhibitors of farnesyl-protein transferase - Google Patents

Inhibitors of farnesyl-protein transferase

Info

Publication number
EP0891360A1
EP0891360A1 EP97917738A EP97917738A EP0891360A1 EP 0891360 A1 EP0891360 A1 EP 0891360A1 EP 97917738 A EP97917738 A EP 97917738A EP 97917738 A EP97917738 A EP 97917738A EP 0891360 A1 EP0891360 A1 EP 0891360A1
Authority
EP
European Patent Office
Prior art keywords
substituted
alkyl
aryl
cycloalkyl
unsubstituted
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
EP97917738A
Other languages
German (de)
French (fr)
Other versions
EP0891360A4 (en
Inventor
Christopher J. Dinsmore
Theresa M. Williams
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Merck and Co Inc
Original Assignee
Merck and 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 GBGB9609666.4A external-priority patent/GB9609666D0/en
Application filed by Merck and Co Inc filed Critical Merck and Co Inc
Publication of EP0891360A1 publication Critical patent/EP0891360A1/en
Publication of EP0891360A4 publication Critical patent/EP0891360A4/en
Withdrawn legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • 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
    • 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
    • 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
    • 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

Definitions

  • Ras proteins are part of a signalling pathway that links cell surface growth factor receptors to nuclear signals initiating cellular proliferation.
  • Biological and biochemical studies of Ras action indicate that Ras functions like a G-regulatory protein.
  • 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.
  • Mutated ras genes (Ha-ras, K 4a-ras, K ⁇ 4b-ras and N-ras) are found in many human cancers, including colorectal carcinoma, exocrine pancreatic carcinoma, and myeloid leukemias. The protein products of these genes are defective in their GTPase activity and constitutive ly transmit a growth stimulatory signal.
  • Ras must be localized to the plasma membrane for both normal and oncogenic functions. At least 3 post-translational modifications are involved with Ras membrane localization, and all 3 modifications occur at the C-terminus of Ras.
  • the Ras C-terminus contains a sequence motif termed a "CAAX” or "Cys-Aaa * -Aaa ⁇ -Xaa” box (Cys is cysteine, Aaa is an aliphatic amino acid, the Xaa is any amino acid) (Willumsen et al. Nature 3/0:583-586 (1984)).
  • this motif serves as a signal sequence for the enzymes famesyl-protein transferase or geranylgeranyl-protein transferase, which catalyze the alkylation of the cysteine residue of the CAAX motif with a C15 or C20 isoprenoid, respectively.
  • the Ras protein is one of several proteins that are known to undergo post-translational famesylation.
  • farnesylated proteins include the Ras-related GTP- binding proteins such as Rho, fungal mating factors, the nuclear lamins, and the gamma subunit of transducin. James, et al., J. Biol. Chem. 269, 14182 (1994) have identified a peroxisome associated protein Pxf which is also farnesylated. James, et al., have also suggested that there are farnesylated proteins of unknown stmcture and function in addition to those listed above.
  • Famesyl-protein transferase utilizes famesyl pyrophosphate to covalently modify the Cys thiol group of the Ras CAAX box with a famesyl 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 , 87:1541 -7545 ( 1990)).
  • Inhibition of famesyl pyrophosphate biosynthesis by inhibiting HMG-CoA reductase blocks Ras membrane localization in cultured cells.
  • direct inhibition of famesyl- protein transferase would be more specific and attended by fewer side effects than would occur with the required dose of a general inhibitor of isoprene biosynthesis.
  • FPTase famesyl-protein transferase
  • FPP famesyl diphosphate
  • Ras protein substrates
  • 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.
  • Such inhibitors may inhibit protein prenylation while serving as altemate substrates for the famesyl-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: 1934-1931 (1993); Graham, et al., J. Med. Chem., 37, 725 (1994)).
  • deletion of the thiol from a CAAX derivative has been shown to dramatically reduce the inhibitory potency of the compound.
  • the thiol group potentially places limitations on the therapeutic application of FPTase inhibitors with respect to pharmacokinetics, pharmacodynamics and toxicity. Therefore, a functional replacement for the thiol is desirable.
  • famesyl-protein transferase inhibitors are inhibitors of proliferation of vascular smooth muscle cells and are therefore useful in the prevention and therapy of arteriosclerosis and diabetic disturbance of blood vessels (JP H7- 1 12930).
  • an object of this invention to develop peptidomimetic compounds that do not have a thiol moiety, and that will inhibit famesyl-protein transferase and thus, the post-translational famesylation of proteins. It is a further object of this invention to develop chemotherapeutic compositions containing the compounds of this invention and methods for producing the compounds of this invention.
  • the present invention comprises peptidomimetic piperazine-containing compounds which inhibit the famesyl-protein transferase.
  • the instant compounds lack a thiol moiety and thus offer unique advantages in terms of improved pharmacokinetic behavior in animals, prevention of thiol-dependent chemical reactions, such as rapid autoxidation and disulfide formation with endogenous thiols, and reduced systemic toxicity.
  • chemotherapeutic compositions containing these famesyl transferase inhibitors and methods for their production are further contained in this invention.
  • the compounds of this invention are useful in the inhibition of famesyl-protein transferase and the famesylation of the oncogene protein Ras.
  • the inhibitors of famesyl-protein transferase are illustrated by the formula A:
  • Rl a and Rib are independently selected from: a) hydrogen, b) aryl, heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, C2- C6 alkynyl, R lOO-, R l l S(0) m -, R 10C(O)NR 10-, CN(RlO)2NC(0)-, R 10 2 N-C(NR 10)-, CN, N ⁇ 2, R lOc(O)-, N3, -N(RlO)2, or Rl l ⁇ C(O)NR l0-, c) unsubstituted or substituted C 1 -C6 alkyl wherein the substitutent on the substituted C1 -C6 alkyl is selected from unsubstituted or substituted aryl, heterocyclic, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, RlOO-, Rl lS(0)m
  • R2 and R ⁇ are independently selected from: H; unsubstituted or substituted Cl -8 alkyl, unsubstituted or substituted C2-8 alkenyl, unsubstituted or substituted C2-8 alkynyl, unsubstituted or substituted aryl,
  • halogen e) CN, f) aryl or heteroaryl, g) perfluoro-C 1 -4 alkyl, h) SR6 , S(0)R6a, S ⁇ 2R 6a ,
  • R3 and R5 are selected from H and CH3;
  • R2 and R3 or R4 and R ⁇ are attached to the same C atom and are combined to form - (CH2)u - wherein one of the carbon atoms is optionally replaced by a moiety selected from: O, S(0) m , -NC(O)-, and -10 -N ⁇ OR ⁇ O)- ;
  • R6, R7 and R ⁇ a are independently selected from: H; Cl -4 alkyl, C3-6 cycloalkyl, heterocycle, aryl, aroyl, heteroaroyl, arylsulfonyl, heteroarylsulfonyl, unsubstituted or substituted with: a) Cl -4 alkoxy, b) aryl or heterocycle, c) halogen, d) HO,
  • R6 and R7 may be joined in a ring; R7 and R ⁇ a m ay be joined in a ring;
  • R6a is selected from: C] -4 alkyl, C3-6 cycloalkyl, heterocycle, aryl, unsubstituted or substituted with: a) Cl -4 alkoxy, b) aryl or heterocycle, c) halogen, d) HO,
  • R8 is independently selected from: a) hydrogen, b) aryl, heterocycle, C3-C 10 cycloalkyl, C2-C6 alkenyl, C2-
  • R9 is selected from: a) hydrogen, b) alkenyl, alkynyl, perfluoroalkyl, F, Cl, Br, Rl*>0-, R 1 lS(0)m-, R 10 C(O)NRl0-, (Rl0) 2 NC(O)-, RIO2N- C(NRlO)-, CN, N02, R 10 C(O)-, N3, -N(RlO) 2 , or RllOC(O)NRl0-, and c) C1-C6 alkyl unsubstituted or substituted by perfluoroalkyl, F, Cl, Br, RlOO-, Rl lS(0) m -, R!0C(O)NR10-, (Rl ) 2 NC(O)-, Rl ⁇ 2N-C(NRlO)-, CN, RlOC(O)-, N3, -N(Rl )2, or Rl l ⁇ C(O)NRl0- ;
  • RlO is independently selected from hydrogen, C1-C6 alkyl, benzyl and aryl;
  • Rl 1 is independently selected from C1-C6 alkyl and aryl
  • V is selected from: a) hydrogen, b) heterocycle, c) aryl, d) C1 -C2O alkyl wherein from 0 to 4 carbon atoms are replaced with a a heteroatom selected from O, S, and N, and e) C2-C20 alkenyl, provided that V is not hydrogen if Al is S(0)m and V is not hydrogen if Al is a bond, n is 0 and A 2 is S(0)m,
  • W is a heterocycle
  • Z is selected from:
  • a unsubstituted or substituted group selected from aryl, heteroaryl, arylmethyl, heteroarylmethyl, arylsulfonyl, heteroarylsulfonyl, wherein the substituted group is substituted with one or more of the following: a) C l -4 alkyl, unsubstituted or substituted with:
  • R la is independently selected from: hydrogen or C1 -C6 alkyl
  • Rib is independently selected from: a) hydrogen, b) aryl, heterocycle, cycloalkyl, RlOO-, -N(R lO) or C2-C6 alkenyl, c) unsubstituted or substituted Ci -C ⁇ alkyl wherein the substitutent on the substituted C1 -C6 alkyl is selected from unsubstituted or substituted aryl, heterocycle, cycloalkyl, alkenyl, R lOO- and -N(RlO) 2;
  • R3 and R ⁇ are independently selected from H and CH3;
  • R2 and R ⁇ are independently selected from H; O or Cl -5 alkyl, unbranched or branched, unsubstituted or substituted with one or more of:
  • R°, R7 and R ⁇ are independently selected from: H; Cl-4 alkyl, C3-6 cycloalkyl, aryl, heterocycle, unsubstituted or substituted with: a) Cl -4 alkoxy, b) halogen, or c) aryl or heterocycle;
  • R6a i selected from: Cl-4 alkyl or C3-6 cycloalkyl, unsubstituted or substituted with: a) Cl-4 alkoxy, b) halogen, or c) aryl or heterocycle;
  • R8 is independently selected from: a) hydrogen, b) C 1 -C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C 1 -C6 perfluoroalkyl, F, Cl, RlOO-, R!0C(O)NR10-, CN, NO2,
  • R9 is selected from: a) hydrogen, b) C2-C6 alkenyl, C2-C6 alkynyl, Cl-C ⁇ perfluoroalkyl, F, Cl, RlOO-, RllS(0) m -, R!0C(O)NR10-, CN, NO2,
  • RlO is independently selected from hydrogen, C1-C6 alkyl, benzyl and aryl;
  • Rl 1 is independently selected from Cl-C ⁇ alkyl and aryl
  • V is selected from: a) hydrogen, b) heterocycle selected from pyrrolidinyl, imidazolyl, pyridinyl, thiazolyl, pyridonyl, 2-oxopiperidinyl, indolyl, quinolinyl, isoquinolinyl, and thienyl, c) aryl, d) C1 -C20 alkyl wherein from 0 to 4 carbon atoms are replaced with a a heteroatom selected from O, S, and N, and e) C2-C20 alkenyl, and provided that V is not hydrogen if Al is S(0)m and V is not hydrogen if A l is a bond, n is 0 and A 2 is S(0)
  • W is a heterocycle selected from pyrrolidinyl, imidazolyl, pyridinyl, thiazolyl, pyridonyl, 2-oxopiperidinyl, indolyl, quinolinyl, or isoquinolinyl;
  • Y is a bond or -CH2-
  • Z is selected from:
  • a unsubstituted or substituted group selected from aryl, heteroaryl, arylmethyl, heteroarylmethyl, arylsulfonyl, heteroarylsulfonyl, 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 6 R 7 , C3-6 cycloalkyl, aryl, heterocycle, HO, -S(0) m R 6a , or -C(0)NR6R7, b) aryl or heterocycle, c) halogen, d) OR6, e) NR6R7, f) CN, g) N ⁇ 2, h) CF3; i) -S(0) m R 6a , j) -C(0)NR6R7, or k) C3-C6 cycloalkyl; or 2) unsubstituted C1-C6 alkyl, substitute
  • Rla is selected from: hydrogen or C1-C6 alkyl
  • Rib is independently selected from: a) hydrogen, b) aryl, heterocycle, cycloalkyl, Rl°0-, -N(Rl°)2 or C2-C6 alkenyl, c) C1-C alkyl unsubstituted or substituted by aryl, heterocycle, cycloalkyl, alkenyl, RlOO-, or -N(RlO)2;
  • R3 and R ⁇ are independently selected from H and CH3;
  • R2 and R4 are independently selected from H; O or C 1-5 alkyl, unbranched or branched, unsubstituted or substituted with one or more of:
  • R6 and R 7 are independently selected from: a) hydrogen, b) Cl -C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, Cl -C6 perfluoroalkyl, F, Cl, RlOO-, R!0C(O)NR10-, CN, N02, (R10) 2 N-C(NR10)-, RlOC(O)-, RlO ⁇ C(O)-, -N(RlO)2, or R1IOC(O)NR10-, and c) C1-C6 alkyl substituted by C1-C6 perfluoroalkyl, Rl°0-,
  • R ⁇ i selected from: Cl-4 alkyl or C3-6 cycloalkyl, unsubstituted or substituted with: a) Cl-4 alkoxy, b) halogen, or c) aryl or heterocycle;
  • R8 is independently selected from: a) hydrogen, b) C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 perfluoroalkyl, F, Cl, RlOO-, R!0C(O)NR10-, CN, N02, (R!0)2N-C(NR10)-, RlOC(O)-, -N(RlO)2, or
  • R11OC(O)NR10- and c) C1-C6 alkyl substituted by Cl-C ⁇ perfluoroalkyl, Rl°0-, Rl0C(O)NRl0-, (R10) 2 N-C(NR10)-, RlOC(O)-, -N(RlO)2, O ⁇ RHOC(O)NR10-;
  • R ⁇ is hydrogen or methyl
  • RlO is independently selected from hydrogen, C1-C6 alkyl, benzyl and aryl;
  • Rl 1 is independently selected from C1-C6 alkyl and aryl
  • V is selected from: a) hydrogen, b) heterocycle selected from pyrrolidinyl, imidazolyl, pyridinyl, thiazolyl, pyridonyl, 2-oxopiperidinyl, indolyl, quinolinyl, isoquinolinyl, and thienyl, c) aryl, d) Cl -C20 alkyl wherein from 0 to 4 carbon atoms are replaced with a a heteroatom selected from O, S, and N, and e) C2-C20 alkenyl, and provided that V is not hydrogen if A ⁇ is S(0)m and V is not hydrogen if A 1 is a bond, n is 0 and A 2
  • Z is selected from: 1 ) a unsubstituted or substituted group selected from aryl, heteroaryl, arylmethyl, heteroarylmethyl, arylsulfonyl, heteroarylsulfonyl, wherein the substituted group is substituted with one or more of the following: a) Cl -4 alkyl, unsubstituted or substituted with: C l -4 alkoxy, NR6R7, C3-6 cycloalkyl, aryl, heterocycle, HO, -S(0) m R 6a , or -C(0)NR6R7, b) aryl or heterocycle, c) halogen, d) OR6, e) NR6R7, f) CN, g) N ⁇ 2, h) CF3; i) -S(0) m R6a, j) -C(0)NR6R7, or k) C3-C6 cycloalkyl; or 2) unsubstituted C1
  • the inhibitors of famesyl-protein transferase are illustrated by the formula C:
  • Rib is independently selected from: a) hydrogen, b) aryl, heterocycle, cycloalkyl, Rl°0-, -N(Rl°)2 or C2-C6 alkenyl, c) C1-C6 alkyl unsubstituted or substituted by aryl, heterocycle, cycloalkyl, alkenyl, RI ⁇ O-, or -N(R 0)2;
  • R3 and R ⁇ are independently selected from H and CH3;
  • R and R4 are independently selected from H; O or Ci-5 alkyl, unbranched or branched, unsubstituted or substituted with one or more of:
  • R6 and R 7 are independently selected from: a) hydrogen, b) C l -C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C 1 -C6 perfluoroalkyl, F, Cl, RlOO-, R!0C(O)NR10-, CN, N02,
  • R8 is independently selected from: a) hydrogen, b) Cl -C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, Cl -C6 perfluoroalkyl, F, Cl, RlOO-, R!0C(O)NR10-, CN, NO2, (RlO)2N-C(NR O)-, Rl°C(0)-, -N(RlO)2, or R11OC(O)NR10-, and c) C1-C6 alkyl substituted by Cj-C ⁇ perfluoroalkyl, Rl°0-, R!0C(O)NR1°-, (RlO)2N-C(NRlO)-, RlOC(O)-,
  • RlO is independently selected from hydrogen, C1-C6 alkyl, benzyl and aryl;
  • Rl 1 is independently selected from Cl -C alkyl and aryl
  • Z is selected from:
  • a unsubstituted or substituted group selected from aryl, heteroaryl, arylmethyl, heteroarylmethyl, arylsulfonyl, heteroarylsulfonyl, wherein the substituted group is substituted with one or more of the following: a) Cl-4 alkyl, unsubstituted or substituted with: Cl-4 alkoxy, R6R7, C3-6 cycloalkyl, aryl, heterocycle, HO, -S(0) m R 6a , or -C(0)NR6R7 5
  • Rib is independently selected from: a) hydrogen, b) aryl, heterocycle, cycloalkyl, R 10 ⁇ -, -N(R 1 °)2 or C2-C6 alkenyl, c) C1-C6 alkyl unsubstituted or substituted by aryl, heterocycle, cycloalkyl, alkenyl, RlOO-, or -N(Rl )2;
  • R2 and R ⁇ are independently selected from: hydrogen or Cl-C6 alkyl
  • R3 and R ⁇ are hydrogen
  • R6 and R 7 are independently selected from: a) hydrogen, b) C 1 -C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C 1 -C ⁇ perfluoroalkyl, F, Cl, Rl O-, R!0C(O)NR10-, CN, N ⁇ 2, (R10) 2 N-C(NR10)-, RlOc(O)-, Rl ⁇ C(O)-, -N(Rl )2, or Rll ⁇ C(O)NRl0-,and c) C1-C6 alkyl substituted by C1-C6 perfluoroalkyl, Rl°0-, Rl0C(O)NRl0-, (R10) 2 N-C(NR10 , RlOc(O)-,
  • R 10 ⁇ C(O)-, -N(R 10)2, or R 11 OC(0)NR 1 . ;
  • RlO is independently selected from hydrogen, C1-C6 alkyl, benzyl and aryl;
  • Rl 1 is independently selected from C1-C6 alkyl and aryl;
  • 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, R l , R2 etc.
  • its definition on each occurence is independent at every other occurence.
  • combinations of substituents/or variables are permissible only if such combinations result in stable compounds.
  • 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 1 1- 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 stmcture.
  • 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 -8 alkyl, substituted C2-8 alkenyl, substituted C2-8 alkynyl, substituted aryl or substituted heterocycle from which the substitutent(s) R and R3 are selected.
  • As used herein in the definition of R°, R oa , R7 an d R7a ? the substituted Cl -8 alkyl, substituted C3-6 cycloalkyl, substituted aroyl, substituted aryl, substituted heteroaroyl, substituted arylsulfonyl, substituted heteroarylsulfonyl and substituted heterocycle include moieties containing from 1 to 3 substitutents in addition to the point of attachment to the rest of the compound.
  • such substitutents are selected from the group which includes but is not limited to F, Cl, Br, CF3, NH2, N(Cl -C6 alkyl)2, N ⁇ 2, CN, (C1-C6 alkyl)0-, -OH, (C1 -C6 alkyl)S(0) m -, (C1 -C6 alkyl)C(0)NH-, H2N-C(NH)-, (C1-C6 alkyl)C(O)-, (C1 -C6 alkyl)OC(O)-, N3,(Cl -C6 alkyl)OC(0)NH-, phenyl, pyridyl, imidazolyl, oxazolyl, isoxazolyl, thiazolyl, thienyl, furyl, isothiazolyl and C1 -C2O alkyl.
  • R2 and R3 are combined to form - (CH2)u -» cyclic moieties are formed
  • cyclic moieties may optionally include a heteroatom(s).
  • heteroatom-containing cyclic moieties include, but are not limited to:
  • Rla and R ib are independently selected from: hydrogen, -N(R 10 )2, R l0C(O)NR l0- or unsubstituted or substituted C1 -C6 alkyl wherein the substituent on the substituted Cl -C6 alkyl is selected from unsubstituted or substituted phenyl, -N(RlO)2, R I ⁇ - and Rl°C(O)NRl0-.
  • R is selected from: H, ⁇ 1R 6 R 7 _ ⁇ -OR 6
  • R3 is selected from: hydrogen and Cl -C6 alkyl.
  • R4 and R ⁇ are hydrogen.
  • R6, R7 and R 7a is selected from: hydrogen, unsubstituted or substituted Cl-C6 alkyl, unsubstituted or substituted aryl and unsubstituted or substituted cycloalkyl.
  • R6a i unsubstituted or substituted C1-C6 alkyl, unsubstituted or substituted aryl and unsubstituted or substituted cycloalkyl.
  • R°- is hydrogen or methyl.
  • R a is hydrogen.
  • R IO is selected from H, C1 -C6 alkyl and benzyl.
  • Al and A2 are independently selected from: a bond, -C(0)NR lO-, -NR IOC(O)-, O, -N(R 10)-, -S(0)2N(R 10). and-
  • V is selected from hydrogen, heterocycle and aryl. More preferably, V is phenyl.
  • Y is a bond or -CH2-. More preferably, Y is -CH2-.
  • 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.
  • W is selected from imidazolinyl, imidazolyl, oxazolyl, pyrazolyl, pyyrohdinyl, thiazolyl and pyridyl. More preferably, W is selected from imidazolyl and pyridyl.
  • n and r are independently 0, 1 , or 2.
  • p is 1 , 2 or 3.
  • s is 0.
  • t is 1.
  • -N(R lO)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 - 15, 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 R2, R3, R4 ⁇ d R5; however the point of attachment to the ring is illustrative only and is not meant to be limiting.
  • Schemes 1 -15 The requisite intermediates are in some cases commercially available, or can be prepared according to literature procedures, for the most part.
  • Scheme 1 for example, the synthesis of 3-substituted-2- piperidone is outlined.
  • a suitably substituted 2-hydroxynicotinic acid I may be catalytically hydrogenated to provide the 2-piperidone II.
  • This intermediate may be coupled to a suitably substituted aryl moiety to provide the key intermediate III.
  • the ester of compound III can be saponified and the acid can be reacted with an amine such as IV to provide the instant compound V.
  • intermediate III can be deprotonated and reacted with an electrophile, such as VI, to provide the instant compound VII.
  • Compound VII can also be decarboxylated to provide the instant compound VIII.
  • Similar coupling to an aryl moiety, deprotonation and alkylation can be performed on a pyrrolidinone, as shown in Scheme 3, to provide the instant compound IX.
  • Scheme 3a illustrates an alternative synthesis of the pyrrolidinone compounds of the instant invention.
  • a suitably substituted 4-halobutanoyl chloride may be cyclized with an amine in a two step procedure and then selectively functionalized adjacent to the carbonyl to provide the instant compound having a preferred arylmethylimidazolyl moiety.
  • Scheme 3c illustrates preparation of the instant compounds wherein Y is S.
  • the suitably substituted thiomo ⁇ holinone may be selectively alkylated with reagents illustrated in Scheme 3 and hereinbelow and may also be oxidized subsequent to the alkylation as shown in the scheme.
  • Preparation of a thiomo ⁇ holinone intermediate having a substituent in the 5-position is illustrated in Scheme 3d.
  • Scheme 4 illustrates preparation of the instant copounds wherein the linker X is an amine.
  • the amine intermediate X may be alkylated with a suitably substituted electrophile.
  • the amine intermediate X can be reductively alkylated with a variety of aldehydes, such as XI.
  • 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 3).
  • 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 dichloroe thane, methanol or dimethylformamide.
  • the product XII can be deprotected to give the final compounds XIII with trifluoroacetic acid in methylene chloride.
  • the final product XIII is isolated in the salt form, for example, as a trifluoroacetate, hydrochloride or acetate salt, among others.
  • the product diamine XIII can further be selectively protected to obtain XIV, which can subsequently be reductively alkylated with a second aldehyde to obtain XV. Removal of the protecting group, and conversion to cyclized products such as the dihydroimidazole XVI can be accomplished by literature procedures.
  • the piperidone intermediate X can be reductively alkylated with other aldehydes such as l -trityl-4-imidazolyl- carboxaldehyde or l-trityl-4-imidazolylacetaldehyde, to give products such as XVII (Scheme 6).
  • the trityl protecting group can be removed from XVII to give XVIII, or alternatively, XVII can first be treated with an alkyl halide then subsequently deprotected to give the alkylated imidazole XIX.
  • the intermediate X can be acylated or sulfonylated by standard techniques.
  • the imidazole acetic acid XX can be converted to the acetate XXI by standard procedures, as shown in Scheme 7, and XXI can be first reacted with an alkyl halide, then treated with refluxing methanol to provide the regiospecifically alkylated imidazole acetic acid ester XXII.
  • Hydrolysis and reaction with intermediate X in the presence of condensing reagents such as l -(3- dimethylaminopropyl)-3-ethylcarbodiimide (EDC) leads to acylated products such as XXIV.
  • Scheme 7a illustrates the preparation of a suitably substituted benzylimidazolyl aldehyde intermediate whch may be reacted with the piperazinone and thiomo ⁇ holinone intermediates whose syntheses is described in Schemes 3b-3d (Scheme 7a and 7b).
  • the protecting groups can be subsequently removed to unmask the hydroxyl group (Schemes 8, 9).
  • 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 XXIX.
  • the fully deprotected amino alcohol XXX can be reductively alkylated (under conditions described previously) with a variety of aldehydes to obtain secondary amines, such as XXXI (Scheme 9), or tertiary amines.
  • the Boc protected amino alcohol XXVII can also be utilized to synthesize 2-aziridinylmethylpiperazines such as XXXII (Scheme 10). Treating XXVII with l ,l '-sulfonyldiimidazole and sodium hydride in a solvent such as dimethylformamide led to the formation of aziridine XXXII. The aziridine reacted in the presence of a nucleophile, such as a thiol, in the presence of base to yield the ring- opened product XXXIII.
  • a nucleophile such as a thiol
  • intermediate X can be reacted with aldehydes derived from amino acids such as O-alkylated tyrosines, according to standard procedures, to obtain compounds such as XXXIX, as shown in Scheme 1 1.
  • R' is an aryl group
  • XXXIX can first be hydrogenated to unmask the phenol, and the amine group deprotected with acid to produce XL. Altematively, the amine protecting group in XXXIX can be removed, and O-alkylated phenolic amines such as XLI produced.
  • the instant compounds are useful as pharmaceutical agents for mammals, especially for humans. These compounds may be administered to patients for use in the treatment of cancer.
  • Examples of the type of cancer which may be treated with the compounds of this invention include, but are not limited to, colorectal carcinoma, exocrine pancreatic carcinoma, myeloid leukemias and neurological tumors. Such tumors may arise by mutations in the ras genes themselves, mutations in the proteins that can regulate Ras activity (i.e., neurofibromin (NF- 1 ), neu, scr, abl , lck, fyn) or by other mechanisms.
  • the compounds of the instant invention inhibit famesyl- protein transferase and the famesylation 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 blindness related to retinal vascularization.
  • the compounds of this invention are also useful for inhibiting other proliferative diseases, both benign and malignant, wherein Ras proteins are aberrantly activated as a result of oncogenic mutation in other genes (i.e., the Ras gene itself is not activated by mutation to an oncogenic form) with said inhibition being accomplished by the 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 alFASEB Journal, 2:A3160 (1988)).
  • the instant compounds may also be useful for the treatment of fungal infections.
  • the compounds of this invention may be administered to mammals, preferably humans, either alone or, preferably, in combination with pharmaceutically acceptable carriers or diluents, optionally with known adjuvants, such as alum, in a pharmaceutical composition, according to standard pharmaceutical practice.
  • the compounds can be administered orally or parenterally, including the intravenous, intramuscular, intraperitoneal, subcutaneous, rectal and topical routes of administration.
  • the selected compound may be administered, for example, in the form of tablets or capsules, or as an aqueous solution or suspension.
  • carriers which are commonly used include lactose and co starch, and lubricating agents, such as magnesium stearate, are commonly added.
  • useful diluents include lactose and dried com starch.
  • aqueous suspensions are required for oral use, the active ingredient is combined with emulsifying and suspending agents. If desired, certain sweetening and/or flavoring agents may be added.
  • sterile solutions of the active ingredient are usually prepared, and the pH of the solutions should be suitably adjusted and buffered.
  • the total concentration of solutes should be controlled in order to render the preparation isotonic.
  • 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 instant compounds may be useful in combination with known anti-cancer and cytotoxic agents.
  • the instant compounds may be useful in combination with agents that are effective in the treatment and prevention of NF-1 , restinosis, polycystic kidney disease, infections of hepatitis delta and related viruses and fungal infections.
  • compositions of this invention include aqueous solutions comprising compounds of this invention and pharmacolo- gically acceptable carriers, e.g., saline, at a pH level, e.g., 7.4. The solutions may be introduced into a patient's blood-stream by local bolus injection.
  • 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, 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 are also useful as a component in an assay to rapidly determine the presence and quantity of famesyl-protein transferase (FPTase) in a composition.
  • FPTase famesyl-protein transferase
  • the composition to be tested may be divided and the two portions contacted with mixtures which comprise a known substrate of FPTase (for example a tetrapeptide having a cysteine at the amine terminus) and famesyl pyrophosphate and, in one of the mixtures, a compound of the instant invention.
  • the chemical content of the assay mixtures may be determined by well known immunological, radiochemical or chromatographic techniques. Because the compounds of the instant invention are selective inhibitors of FPTase, absence or quantitative reduction of the amount of substrate in the assay mixture without the compound of the instant invention relative to the presence of the unchanged substrate in the assay containing the instant compound is indicative of the presence of FPTase in the composition to be tested.
  • potent inhibitor compounds of the instant invention may be used in an active site titration assay to determine the quantity of enzyme in the sample.
  • a series of samples composed of aliquots of a tissue extract containing an unknown amount of famesyl-protein transferase, an excess amount of a known substrate of FPTase (for example a tetrapeptide having a cysteine at the amine terminus) and fa esyl pyrophosphate are incubated for an appropriate period of time in the presence of varying concentrations of a compound of the instant invention.
  • concentration of a sufficiently potent inhibitor i.e., one that has a Ki substantially smaller than the concentration of enzyme in the assay vessel
  • concentration of a sufficiently potent inhibitor i.e., one that has a Ki substantially smaller than the concentration of enzyme in the assay vessel
  • Step A 4-Cvanobenzyl-N ⁇ .-phthaloylhistamine
  • N ⁇ -Pivaloyloxymethyl-N ⁇ -phthaloylhistamine (4.55 g, 12.8 mmol) was prepared as previously described (J. C. Emmett, F. H. Holloway, and J. L. Turner, J. Chem. Soc, Perkin Trans. 1 , 1341 , (1979)).
  • ⁇ -Bromo-p-tolunitrile (3.77 g, 1 .2 mmol) was dissolved in acetonitrile (70 mL). The solution was heated at 55°C for 4 h, cooled to room temperature, and filtered to remove the white solid. The acetonitrile (30 mL) was concentrated to 1/2 its volume under reduced pressure and the solution was heated at 55°C overnight.
  • Step B 4-Cvanobenzyl histamine 4-Cyanobenzyl-N ⁇ -phthaloylhistamine (1.64 g, 4.61 mmol), and hydrazine (1.46 mL, 46.1 mmol) were dissolved in absolute ethanol (70 mL). The solution was concentrated after 1 hr and filtered to remove a white precipitate which was washed several times with ethanol. The filtrate was concentrated and the residue was chromatographed (silica gel, 10:90: 1 MeOH/CH2Cl2/NH4 ⁇ H) to give the title compound.
  • Step C 3-Carbethoxy- 1 -phenyl-2-piperidinone
  • Step D 3-Carboxy- 1 -pheny 1-2-piperidinone
  • Step E N-[2- ⁇ l -(4-Cyanobenzyl)-5-imidazolyl ⁇ ethyl]-3- carbamoyl- 1 -phenyl-2-piperidinone hydrochloride
  • Step D 4-(3-Hydroxy- 1 -propyl)-3-( 1 -triphenylmethyl-4- imidazolvDpropionate
  • Step E 1 -Pheny 1-2-piperdinone
  • Step F 3-[3- ⁇ l -triphenylmethyl-4-imidazolyl ⁇ - l -propyl]- l -phenyl- 2-piperidinone hydrochloride
  • a solution of 1 -phenyl-2-piperidinone from Step F in THF is added to a solution of one equivalent of LDA in THF at -78°C, and stirred for 30 min.
  • a solution of 4-(3-hydroxy-l-propyl)-3-( l - triphenylmethyl-4-imidazolyl)propionate in methylene chloride is cooled to -78°C under nitrogen.
  • One equivalent of ⁇ -butyl lithium followed by one equivalent of triflic anhydride is added, the reaction stirred for 10 min, then added to the LDA/l -phenyl-2-piperidinone solution.
  • the reaction is warmed to room temperature, and quenched with saturated ammonium chloride.
  • the reaction is then partitioned between ethyl acetate and saturated brine.
  • the organic phase is dried over magnesium sulfate, filtered and concentrated.
  • the title compound is isolated by chromatography on silica gel.
  • Step G 3-[3- ⁇ l-(4-Cyanobenzyl)-5-imidazolyl ⁇ -l-propyl]-l- phenyl-2-piperidinone hydrochloride
  • the product from Step F is dissolved in acetonitrile and one equivalent of 4-cyanobenzylbromide added.
  • the reaction is stirred at room temperature overnight, concentrated, and taken up in methanol.
  • the methanol solution is refluxed for 3 h, concentrated, and partitioned between ethyl acetate and saturated sodium bicarbonate solution.
  • the organic phase is washed with saturated brine and dried over magnesium sulfate.
  • the title compound is obtained after chromatography on silica gel, and conversion to the dihydrochloride salt.
  • Step A 1 -Phenyl-2-pyrrolidinone
  • the title compound is prepared according to the procedure described in Example 2, Step E, except using 2-pyrrolidinone in place of valerolactam.
  • the cmde product is purified by column chromatography.
  • Step B 3-[3- ⁇ l -triphenylmethyl-4-imidazolyl ⁇ -l -propyl]-l -phenyl- 2-pyrrolidinone
  • the title compound is prepared according to the procedure described in Example 2, Step F, except using 1 -phenyl-2-pyrrolidinone in place of 1 -phenyl-2-piperdinone.
  • the title compound is isolated by chromatography on silica gel.
  • Step C 3-[3- ⁇ l -(4-Cyanobenzyl)-5-imidazolyl ⁇ - l-propyl]- l - phenyl-2-pyrrolidinone hydrochloride
  • the title compound is prepared according to the procedure described in Example 2, Step G except using 3-[3- ⁇ l-triphenylmethyl-4- imidazolyl ⁇ -l -propyl]-l -phenyl-2-pyrrolidinone in place of 3-[3- ⁇ 1- triphenylmethyl-4-imidazolyl ⁇ -l -propyl]-l -phenyl-2-piperidinone.
  • the title compound is obtained after chromatography on silica gel, and conversion to the hydrochloride salt.
  • Step A 4-Carbomethoxy- 1 -pheny 1-2-pyrrolidinone
  • Step B 4-Hydroxymethy 1- 1 -phenyl-2-pyrrolidinone
  • Step C 4-Methoxymethyl- 1 -phenyl-2-pyrrolidinone
  • Step D ( ⁇ )cis- and ( ⁇ )/r ⁇ /2s-4-Methoxymethyl-l-phenyl-3-[3- ⁇ 1 -t riphenylmethyl-4-imidazolyl ⁇ - 1 -propyl]-2- pyrrolidinone
  • the title compound is prepared according to the procedure described in Example 2, Step F, except using 4-methoxymethyl-l - phenyl-2-pyrroIidinone in place of l-phenyl-2-piperdinone.
  • the title compounds are isolated by chromatography on silica gel.
  • Step E ( ⁇ )cis- and ( ⁇ )trans-3-[3- ⁇ 1 -(4-Cyanobenzyl)-5- imidazolyl ⁇ - 1 -propyl ]-4-methoxymethy 1- 1 -pheny 1-2- pyrrolidinone hydrochloride
  • the title compound is prepared according to the procedure described in Example 2, Step G except using cis -and trans-4- methoxymethyl- 1 -pheny l-3-[3- ⁇ 1 -triphenylmethyl-4-imidazolyl ⁇ - 1 - propyl]-2-pyrrolidinone_in place of 3-f3- ⁇ l -triphenylmethyl-4- imidazolyl ⁇ -l -propyl]- 1 -pheny 1-2-piperidinone.
  • the title compounds are separated by preparative HPLC and converted to their hydrochloride salts.
  • Step B 1 -Phenyl-2-pyrrolidinone
  • THF 157 mL
  • sodium hydride 50% dispersion in oil
  • the reaction was stirred at room temperature overnight.
  • An additional portion of sodium hydride was added (1 g) and the reaction continued to stir for 3 h.
  • the reaction was diluted with ethyl acetate and poured into 10% aqueous hydrogen chloride.
  • the organic phase was washed with saturated brine, and dried over sodium sulfate. Concentration in vacuo afforded the title compound.
  • Step C 3-(f-ButylcarboxymethyD- 1 -phenyl-2-pyrrolidinone
  • Step D 3-(2-CarboxymethyP- 1 -pheny 1-2-pyrrolidinone
  • Step E 3-(2-HvdroxyethvD- 1 -phenyl-2-pyrrolidinone
  • Step G 3-[2- ⁇ 5-(4-Cyanobenzyl)- 1 -imidazolyl ⁇ - 1 -ethyl]- 1 -phenyl-
  • Step B 1 -Benzy l-3-[2- ⁇ 5-(4-cyanobenzy 1)- 1 -imidazolyl ⁇ - 1 -ethy 1] - 2-pyrrolidinone hydrochloride
  • Step A Preparation of l -triphenylmethyl-4-(hydroxymethyl)- imidazole
  • Step B Preparation of l -triphenylmethyl-4-(acetoxymethyl)- imidazole
  • Step C Preparation of l-(4-cyanobenzyl)-5-(acetoxymethyl)- imidazole hydrobromide
  • 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 500 mL of methanol, and warmed to 60 °C. After two hours, the solution was reconcentrated in vacuo to provide a white solid which was triturated with hexane to remove soluble materials. Removal of residual solvents in vacuo provided the titled product hydrobromide as a white solid which was used in the next step without further purification.
  • Step D Preparation of l-(4-cyanobenzyl)-5-(hydroxymethyl)- imidazole
  • Step E Preparation of l-(4-cyanobenzyl)-5-imidazole- carboxaldehyde
  • Step F Preparation of N-(3-chlorophenyl)ethylenediamine hydrochloride To a solution of 3-chloroaniline (30.0 mL, 284 mmol) in 500 mL of dichloromethane at 0 °C was added dropwise a solution of 4 N HCI in 1 ,4-dioxane (80 mL, 320 mmol HCI). The solution was warmed to room temperature, then concentrated to dryness in vacuo to provide a white powder. A mixture of this powder with 2- oxazolidinone (24.6 g, 282 mmol) was heated under nitrogen atmosphere at 160 °C for 10 hours, during which the solids melted, and gas evolution was observed.
  • Step G Preparation of /V-( / ⁇ -butoxycarbonyl)-/V'-(3- chlorophenyDethylenediamine
  • the amine hydrochloride from Step F (ca. 282 mmol, cmde material prepared above) was taken up in 500 mL of THF and 500 mL of sat. aq. NaHC ⁇ 3 soln., cooled to 0 °C, and ⁇ i-tert- butylpyrocarbonate (61.6 g, 282 mmol) was added. After 30 h, the reaction was poured into EtOAc, washed with water and brine, dried (Na2S ⁇ 4), 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 ⁇ H2-(f -butoxycarbamoyl)ethyl]-/V-(3- chloropheny 1 )-2-chloroacetami de
  • Step I Preparation of 4-(/crt-butoxycarbonyl)- 1 -(3- chlorophenvl)-2-piperazinone
  • Step J Preparation of ( ⁇ )-4-(r -butoxy carbonyl)- 1 -(3- chloropheny)-3-[ 1 -(1 -(4-cyanobenzyl)-5-imidazolyl)- 1 -
  • the solution was dried (Na2S04), filtered, and concentrated in vacuo to provide the cmde product.
  • the product was purified by flash chromatography on silica gel (3-6% MeOH/CH2C12) to provide 60 mg of the titled product as a 9: 1 mixture of diastereomers.
  • Step K Preparation of ( ⁇ )- l -(3-chloropheny)-3-[ l -(l -(4- cyanobenzy l)-5-imidazolyl)- 1 -(hydroxy )methyl]-2- piperazinone hydrochloride
  • Bovine FPTase was assayed in a volume of 100 ⁇ l containing 100 mM N-(2- hydroxy ethyl) piperazine-/V'-(2-ethane sulfonic acid) (HEPES), pH 7.4, 5 mM MgCl2, 5 mM dithiothreitol (DTT), 100 mM [3H]-farnesyl diphosphate ([3H]-FPP; 740 CBq/mmol, New England Nuclear), 650 nM Ras-CVLS and 10 ⁇ g/ml FPTase at 31°C for 60 min. Reactions were initiated with FPTase and stopped with 1 ml of 1.0 M HCL in ethanol.
  • Precipitates were collected onto filter-mats using a TomTec Mach II cell harvestor, washed with 100% ethanol, dried and counted in an LKB ⁇ -plate counter.
  • the assay was linear with respect to both substrates, FPTase levels and time; less than 10% of the [3H]-FPP was utilized during the reaction period.
  • Purified compounds were dissolved in 100% dimethyl sulfoxide (DMSO) and were diluted 20-fold into the assay. Percentage inhibition is measured by the amount of incorporation of radioactivity in the presence of the test compound when compared to the amount of incorporation in the absence of the test compound.
  • DMSO dimethyl sulfoxide
  • Human FPTase was prepared as described by Omer et al., Biochemistry 32:5167-5176 (1993). Human FPTase activity was assayed as described above with the exception that 0.1 % (w/v) polyethylene glycol 20,000, 10 ⁇ M ZnCl 2 and 100 ⁇ M Ras-CVIM were added to the reaction mixture. Reactions were performed for 30 min., stopped with 100 ⁇ l of 30% (v/v) trichloroacetic acid (TCA) in ethanol and processed as described above for the bovine enzyme.
  • TCA trichloroacetic acid
  • the cell line used in this assay is a v-ras line derived from either Ratl or NIH3T3 cells, which expressed viral Ha-ras p21.
  • the assay is performed essentially as described in DeClue, J.E. et aL, Cancer Research 51 :712-717, (1991 ). Cells in 10 cm dishes at 50-75% confluency are treated with the test compound (final concentration of solvent, methanol or dimethyl sulfoxide, is 0.1 %).
  • the cells After 4 hours at 37°C, the cells are labelled in 3 ml methionine-free DMEM supple- meted with 10% regular DMEM, 2% fetal bovine serum and 400 mCif35s]methionine (1000 Ci/mmol). After an additional 20 hours, the cells are lysed in 1 ml lysis buffer (1 % NP40/20 mM HEPES, pH 7.5/5 mM MgCl2/lmM DTT/10 mg/ml aprotinen/2 mg/ml leupeptin/2 mg/ml antipain/0.5 mM PMSF) and the lysates cleared by centrifugation at 100,000 x g for 45 min.
  • 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
  • the immunoprecipitates are washed four times with IP buffer (20 nM HEPES, pH 7.5/1 mM EDTA/1 % Triton X- 100.0.5% deoxycholate/0.1%/SDS/0.1 M 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 nonfamesylated ras proteins are compared to determine the percent inhibition of famesyl 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 se m) 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.

Abstract

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

Description

FTITLE OF THE INVENTION
INHIBITORS OF FARNESYL-PROTEIN TRANSFERASE
BACKGROUND OF THE INVENTION The Ras proteins (Ha-Ras, Ki4a-Ras, Ki4b-Ras and N-Ras) are part of a signalling pathway that links cell surface growth factor receptors to nuclear signals initiating cellular proliferation. Biological and biochemical studies of Ras action indicate that Ras functions like a G-regulatory protein. In the inactive state, Ras is bound to GDP. Upon growth factor receptor activation Ras is induced to exchange GDP for GTP and undergoes a conformational change. The GTP-bound form of Ras propagates the growth stimulatory signal until the signal is terminated by the intrinsic GTPase activity of Ras, which returns the protein to its inactive GDP bound form (D.R. Lowy and D.M. Willumsen, Ann. Rev. Biochem. 62:851 -891 (1993)). Mutated ras genes (Ha-ras, K 4a-ras, K\4b-ras and N-ras) are found in many human cancers, including colorectal carcinoma, exocrine pancreatic carcinoma, and myeloid leukemias. The protein products of these genes are defective in their GTPase activity and constitutive ly transmit a growth stimulatory signal.
Ras must be localized to the plasma membrane for both normal and oncogenic functions. At least 3 post-translational modifications are involved with Ras membrane localization, and all 3 modifications occur at the C-terminus of Ras. The Ras C-terminus contains a sequence motif termed a "CAAX" or "Cys-Aaa * -Aaa^-Xaa" box (Cys is cysteine, Aaa is an aliphatic amino acid, the Xaa is any amino acid) (Willumsen et al. Nature 3/0:583-586 (1984)). Depending on the specific sequence, this motif serves as a signal sequence for the enzymes famesyl-protein transferase or geranylgeranyl-protein transferase, which catalyze the alkylation of the cysteine residue of the CAAX motif with a C15 or C20 isoprenoid, respectively. (S. Clarke., Ann. Rev. Biochem. 67 :355-386 (1992); W.R. Schafer and J. Rine, Ann. Rev. Genetics 30:209-231 (1992)). The Ras protein is one of several proteins that are known to undergo post-translational famesylation. Other farnesylated proteins include the Ras-related GTP- binding proteins such as Rho, fungal mating factors, the nuclear lamins, and the gamma subunit of transducin. James, et al., J. Biol. Chem. 269, 14182 (1994) have identified a peroxisome associated protein Pxf which is also farnesylated. James, et al., have also suggested that there are farnesylated proteins of unknown stmcture and function in addition to those listed above.
Inhibition of famesyl-protein transferase has been shown to block the growth of Ras-transformed cells in soft agar and to modify other aspects of their transformed phenotype. It has also been demonstrated that certain inhibitors of famesyl-protein transferase selectively block the processing of the Ras oncoprotein intracellularly (N.E. Kohl et a , Science, 260:1934-1937 ( 1993) and G.L. James et al, Science, 260: 1931- 1942 (1993). Recently, it has been shown that an inhibitor of famesyl-protein transferase blocks the growth of ras- dependent tumors in nude mice (N.E. Kohl et al, Proc. Natl. Acad. Sci U.S.A., 97 :9141 -9145 ( 1994) and induces regression of mammary and salivary carcinomas in ras transgenic mice (N.E. Kohl et al, Nature Medicine, 1 :792-797 (1995). Indirect inhibition of famesyl-protein transferase in vivo has been demonstrated with lovastatin (Merck & Co., Rahway, NJ) and compactin (Hancock et al., ibid; Casey et al, ibid; Schafer et al, Science 245:319 (1989)). These drugs inhibit HMG-CoA reductase, the rate limiting enzyme for the production of polyisoprenoids including famesyl pyrophosphate. Famesyl-protein transferase utilizes famesyl pyrophosphate to covalently modify the Cys thiol group of the Ras CAAX box with a famesyl 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 , 87:1541 -7545 ( 1990)). Inhibition of famesyl pyrophosphate biosynthesis by inhibiting HMG-CoA reductase blocks Ras membrane localization in cultured cells. However, direct inhibition of famesyl- protein transferase would be more specific and attended by fewer side effects than would occur with the required dose of a general inhibitor of isoprene biosynthesis.
Inhibitors of famesyl-protein transferase (FPTase) have been described in two general classes. The first are analogs of famesyl diphosphate (FPP), while the second class of inhibitors is related to the protein substrates (e.g., Ras) for the enzyme. The peptide derived inhibitors that have been described are generally cysteine containing molecules that are related to the CAAX motif that is the signal for protein prenylation. (Schaber et al, ibid; Reiss et. al, ibid; Reiss et al., PNAS, 88:132-136 (1991 )). Such inhibitors may inhibit protein prenylation while serving as altemate substrates for the famesyl-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: 1934-1931 (1993); Graham, et al., J. Med. Chem., 37, 725 (1994)). In general, deletion of the thiol from a CAAX derivative has been shown to dramatically reduce the inhibitory potency of the compound. However, the thiol group potentially places limitations on the therapeutic application of FPTase inhibitors with respect to pharmacokinetics, pharmacodynamics and toxicity. Therefore, a functional replacement for the thiol is desirable.
It has recently been reported that famesyl-protein transferase inhibitors are inhibitors of proliferation of vascular smooth muscle cells and are therefore useful in the prevention and therapy of arteriosclerosis and diabetic disturbance of blood vessels (JP H7- 1 12930).
It has recently been disclosed that certain tricyclic compounds which optionally incorporate a piperidine moiety are inhibitors of FPTase (WO 95/10514, WO 95/10515 and WO 95/10516). Imidazole-containing inhibitors of famesyl protein transferase have also been disclosed (WO 95/09001 and EP 0 675 1 12 A 1 ).
It is, therefore, an object of this invention to develop peptidomimetic compounds that do not have a thiol moiety, and that will inhibit famesyl-protein transferase and thus, the post-translational famesylation of proteins. It is a further object of this invention to develop chemotherapeutic compositions containing the compounds of this invention and methods for producing the compounds of this invention.
SUMMARY OF THE INVENTION
The present invention comprises peptidomimetic piperazine-containing compounds which inhibit the famesyl-protein transferase. The instant compounds lack a thiol moiety and thus offer unique advantages in terms of improved pharmacokinetic behavior in animals, prevention of thiol-dependent chemical reactions, such as rapid autoxidation and disulfide formation with endogenous thiols, and reduced systemic toxicity. Further contained in this invention are chemotherapeutic compositions containing these famesyl transferase inhibitors and methods for their production.
The compounds of this invention are illustrated by the formula A:
- A , 1 /PCDRll 1 a ι a 2)
DETAILED DESCRIPTION OF THE INVENTION
The compounds of this invention are useful in the inhibition of famesyl-protein transferase and the famesylation of the oncogene protein Ras. In a first embodiment of this invention, the inhibitors of famesyl-protein transferase are illustrated by the formula A:
wherein:
Rl a and Rib are independently selected from: a) hydrogen, b) aryl, heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, C2- C6 alkynyl, R lOO-, R l l S(0)m-, R 10C(O)NR 10-, CN(RlO)2NC(0)-, R 102N-C(NR 10)-, CN, Nθ2, R lOc(O)-, N3, -N(RlO)2, or Rl lθC(O)NR l0-, c) unsubstituted or substituted C 1 -C6 alkyl wherein the substitutent on the substituted C1 -C6 alkyl is selected from unsubstituted or substituted aryl, heterocyclic, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, RlOO-, Rl lS(0)m-, R 1 °C(0)NR 10-, (R lO)2NC(0)-, R !02N-
C(NRlO)-, CN, Rl C(O)-, N3, -N(RlO)2, and R HθC(O)-
NR 10-;
R2 and R^ are independently selected from: H; unsubstituted or substituted Cl -8 alkyl, unsubstituted or substituted C2-8 alkenyl, unsubstituted or substituted C2-8 alkynyl, unsubstituted or substituted aryl,
unsubstituted or substituted heterocycle, wherein the substituted group is substituted with one or more of:
1) aryl or heterocycle, unsubstituted or substituted with: a) Cl -4 alkyl,
d) halogen, e) CN, f) aryl or heteroaryl, g) perfluoro-C 1 -4 alkyl, h) SR6 , S(0)R6a, Sθ2R6a,
2) C3-6 cycloalkyl,
3) OR6, 4) SR6a, S(0)R6a, or S02R6a,
5) — NR6R7
Re
7)
N NR7R7a
T O O^ . R6R7
8) T O
9) — O^OR6
T O
11 ) — S02- NR6R7
1 4> XT0* o
15) N3,
16) F, or
17) perfluoro-C1.4-alkyl; or
5 R3 and R5 are selected from H and CH3;
R2 and R3 or R4 and R^ are attached to the same C atom and are combined to form - (CH2)u - wherein one of the carbon atoms is optionally replaced by a moiety selected from: O, S(0)m, -NC(O)-, and -10 -N^OR ΪO)- ; R6, R7 and R^a are independently selected from: H; Cl -4 alkyl, C3-6 cycloalkyl, heterocycle, aryl, aroyl, heteroaroyl, arylsulfonyl, heteroarylsulfonyl, unsubstituted or substituted with: a) Cl -4 alkoxy, b) aryl or heterocycle, c) halogen, d) HO,
f) — S02R11 , or g) N(Rl O)2; or
R6 and R7 may be joined in a ring; R7 and R^a may be joined in a ring;
R6a is selected from: C] -4 alkyl, C3-6 cycloalkyl, heterocycle, aryl, unsubstituted or substituted with: a) Cl -4 alkoxy, b) aryl or heterocycle, c) halogen, d) HO,
f) -S02R11 , or g) N(R l )2;
R8 is independently selected from: a) hydrogen, b) aryl, heterocycle, C3-C 10 cycloalkyl, C2-C6 alkenyl, C2-
C6 alkynyl, perfluoroalkyl, F, Cl, Br, R lOO-, R l l S(0)m- RlOC(0)NRl -, (Rl0)2NC(O)-, Rl 2N-C(NRlO)-, CN, Nθ2, Rl°C(0)-, N3, -N(RlO)2, or Rl lθC(O)NRl0-, and c) Cl-Cό alkyl unsubstituted or substituted by aryl, cyanophenyl, heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, perfluoroalkyl, F, Cl, Br, RlOO-,
Rl lS(0)m-, R °C(0)NH-, (Rl ) NC(0)-, Rl02N- C(NRl )-, CN, Rl C(O)-, N3, -N(RlO)2, or Rl0θC(O)NH-;
R9 is selected from: a) hydrogen, b) alkenyl, alkynyl, perfluoroalkyl, F, Cl, Br, Rl*>0-, R1 lS(0)m-, R10C(O)NRl0-, (Rl0)2NC(O)-, RIO2N- C(NRlO)-, CN, N02, R10C(O)-, N3, -N(RlO)2, or RllOC(O)NRl0-, and c) C1-C6 alkyl unsubstituted or substituted by perfluoroalkyl, F, Cl, Br, RlOO-, Rl lS(0)m-, R!0C(O)NR10-, (Rl )2NC(O)-, Rlθ2N-C(NRlO)-, CN, RlOC(O)-, N3, -N(Rl )2, or Rl lθC(O)NRl0-;
RlO is independently selected from hydrogen, C1-C6 alkyl, benzyl and aryl;
Rl 1 is independently selected from C1-C6 alkyl and aryl;
Al and A^ are independently selected from: a bond, -CH=CH-, -C≡C-, -C(O)-, -C(O)NRl0-, -NRlOc(O)-, O, -N(RlO)-, -S(0)2N(RlO)-, -N(Rl0)S(O)2-, or S(0)m;
V is selected from: a) hydrogen, b) heterocycle, c) aryl, d) C1 -C2O alkyl wherein from 0 to 4 carbon atoms are replaced with a a heteroatom selected from O, S, and N, and e) C2-C20 alkenyl, provided that V is not hydrogen if Al is S(0)m and V is not hydrogen if Al is a bond, n is 0 and A 2 is S(0)m,
W is a heterocycle;
X is a bond, -CH2-, -C(=0)-, -C(0)NR6-, -NR6C(0)-, -NR6- or -S(=0)m-;
Y is a bond, -CH2-, -NH-, -S(=0)m- or O;
Z is selected from:
1 ) a unsubstituted or substituted group selected from aryl, heteroaryl, arylmethyl, heteroarylmethyl, arylsulfonyl, heteroarylsulfonyl, wherein the substituted group is substituted with one or more of the following: a) C l -4 alkyl, unsubstituted or substituted with:
C l -4 alkoxy, NR6R7, C3-6 cycloalkyl, aryl, heterocycle, HO, -S(0)mR6a, or -C(0)NR6R7, b) aryl or heterocycle, c) halogen, d) OR6, e) NR6R7, O CN, g) Nθ2, h) CF3; i) -S(0)mR6a, j) -C(0)NR6R7, or k) C3-C6 cycloalkyl; or
2) unsubstituted C1 -C6 alkyl, substituted C1-C6 alkyl, unsubstituted C3-C6 cycloalkyl or substituted C3-C6 cycloalkyl,wherein the substituted C1 -C6 alkyl and substituted C3-C6 cycloalkyl is substituted with one or two of the following: a) Cl -4 alkoxy, b) NR6R7, c) C3-6 cycloalkyl, d) -NR6C(0)R7, e) HO, f) -S(0)mR6a, g) halogen, or h) perfluoroalkyl;
or the pharmaceutically acceptable salts thereof.
A preferred embodiment of the compounds of this invention is illustrated by the following formula:
wherein: R la is independently selected from: hydrogen or C1 -C6 alkyl;
Rib is independently selected from: a) hydrogen, b) aryl, heterocycle, cycloalkyl, RlOO-, -N(R lO) or C2-C6 alkenyl, c) unsubstituted or substituted Ci -Cβ alkyl wherein the substitutent on the substituted C1 -C6 alkyl is selected from unsubstituted or substituted aryl, heterocycle, cycloalkyl, alkenyl, R lOO- and -N(RlO)2;
R3 and R^ are independently selected from H and CH3;
NR6R7
R2 and R^ are independently selected from H; O or Cl -5 alkyl, unbranched or branched, unsubstituted or substituted with one or more of:
1 ) aryl,
2) heterocycle,
3) OR6,
4) SR6a, S02R6a, or
5) \ ,NR6R7
T O ; and any two of R2, R , R45 and R5 are optionally attached to the same carbon atom;
R°, R7 and R^ are independently selected from: H; Cl-4 alkyl, C3-6 cycloalkyl, aryl, heterocycle, unsubstituted or substituted with: a) Cl -4 alkoxy, b) halogen, or c) aryl or heterocycle;
R6a i selected from: Cl-4 alkyl or C3-6 cycloalkyl, unsubstituted or substituted with: a) Cl-4 alkoxy, b) halogen, or c) aryl or heterocycle;
R8 is independently selected from: a) hydrogen, b) C 1 -C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C 1 -C6 perfluoroalkyl, F, Cl, RlOO-, R!0C(O)NR10-, CN, NO2,
(RlO)2N-C(NRlO)-, RlOC(O)-, -N(RlO)2, or RllθC(O)NRl0-,and c) C1-C6 alkyl substituted by -C6 perfluoroalkyl, Rl°0-, R!0C(O)NR10-, (R10)2N-C(NR10)-, RlOC(O)-, -N(RlO)2, or Rl 10C(0)NR10-;
R9 is selected from: a) hydrogen, b) C2-C6 alkenyl, C2-C6 alkynyl, Cl-Cό perfluoroalkyl, F, Cl, RlOO-, RllS(0)m-, R!0C(O)NR10-, CN, NO2,
(RlO)2N-C(NR O)-, RlOC(O)-, -N(Rl°)2, or RllθC(O)NRl0-, and c) Cl -C alkyl unsubstituted or substituted by C l -C6 perfluoroalkyl, F, Cl, Rl<0-, R1 ^(O , R10C(0)NR10-, CN, (R10) N-C(NR10)-, RlOC(O)-, -N(RlO)2, or
RllθC(O)NRl0-;
RlO is independently selected from hydrogen, C1-C6 alkyl, benzyl and aryl;
Rl 1 is independently selected from Cl-Cό alkyl and aryl;
Al and A^ are independently selected from: a bond, -CH=CH-, -C≡C-, -C(O)-, -C(O)NRl0-, O, -N(RlO)-, or S(0)m; V is selected from: a) hydrogen, b) heterocycle selected from pyrrolidinyl, imidazolyl, pyridinyl, thiazolyl, pyridonyl, 2-oxopiperidinyl, indolyl, quinolinyl, isoquinolinyl, and thienyl, c) aryl, d) C1 -C20 alkyl wherein from 0 to 4 carbon atoms are replaced with a a heteroatom selected from O, S, and N, and e) C2-C20 alkenyl, and provided that V is not hydrogen if Al is S(0)m and V is not hydrogen if A l is a bond, n is 0 and A 2 is S(0)m;
W is a heterocycle selected from pyrrolidinyl, imidazolyl, pyridinyl, thiazolyl, pyridonyl, 2-oxopiperidinyl, indolyl, quinolinyl, or isoquinolinyl;
X is -CH2- or -C(=0)-;
Y is a bond or -CH2-;
Z is selected from:
1) a unsubstituted or substituted group selected from aryl, heteroaryl, arylmethyl, heteroarylmethyl, arylsulfonyl, heteroarylsulfonyl, wherein the substituted group is substituted with one or more of the following: a) Cl -4 alkyl, unsubstituted or substituted with: Cl -4 alkoxy, NR6R7, C3-6 cycloalkyl, aryl, heterocycle, HO, -S(0)mR6a, or -C(0)NR6R7, b) aryl or heterocycle, c) halogen, d) OR6, e) NR6R7, f) CN, g) Nθ2, h) CF3; i) -S(0)mR6a, j) -C(0)NR6R7, or k) C3-C6 cycloalkyl; or 2) unsubstituted C1-C6 alkyl, substituted C1 -C6 alkyl, unsubstituted C3-C6 cycloalkyl or substituted C3-C6 cycloalkyl,wherein the substituted C1 -C6 alkyl and substituted C3-C6 cycloalkyl is substituted with one or two of the following: a) Cl-4 alkoxy, b) NR6R7, c) C3-6 cycloalkyl, d) -NR6C(0)R7, e) HO, f) -S(0)mR6a, g) halogen, or h) perfluoroalkyl;
or the pharmaceutically acceptable salts thereof.
A preferred embodiment of the compounds of this invention are illustrated by the formula B: wherein:
Rla is selected from: hydrogen or C1-C6 alkyl;
Rib is independently selected from: a) hydrogen, b) aryl, heterocycle, cycloalkyl, Rl°0-, -N(Rl°)2 or C2-C6 alkenyl, c) C1-C alkyl unsubstituted or substituted by aryl, heterocycle, cycloalkyl, alkenyl, RlOO-, or -N(RlO)2;
R3 and R^ are independently selected from H and CH3;
\ ^NR6R7
R2 and R4 are independently selected from H; O or C 1-5 alkyl, unbranched or branched, unsubstituted or substituted with one or more of:
1) aryl,
2) heterocycle,
3) OR6, 4) SR6a, Sθ2R6a, or
5) \ .NR6R7
T O : and any two of R2, R3? 4; ancj R5 are optionally attached to the same carbon atom;
R6 and R7 are independently selected from: a) hydrogen, b) Cl -C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, Cl -C6 perfluoroalkyl, F, Cl, RlOO-, R!0C(O)NR10-, CN, N02, (R10)2N-C(NR10)-, RlOC(O)-, RlOθC(O)-, -N(RlO)2, or R1IOC(O)NR10-, and c) C1-C6 alkyl substituted by C1-C6 perfluoroalkyl, Rl°0-,
Rl0C(O)NRl0-, (R10)2N-C(NR10)-, RlOC(O)-, R 1 OθC(O)-, -N(R 10)2, or R 11 OC(0)NR 10-;
R^ i selected from: Cl-4 alkyl or C3-6 cycloalkyl, unsubstituted or substituted with: a) Cl-4 alkoxy, b) halogen, or c) aryl or heterocycle;
R8 is independently selected from: a) hydrogen, b) C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 perfluoroalkyl, F, Cl, RlOO-, R!0C(O)NR10-, CN, N02, (R!0)2N-C(NR10)-, RlOC(O)-, -N(RlO)2, or
R11OC(O)NR10-, and c) C1-C6 alkyl substituted by Cl-Cό perfluoroalkyl, Rl°0-, Rl0C(O)NRl0-, (R10)2N-C(NR10)-, RlOC(O)-, -N(RlO)2, OΓRHOC(O)NR10-;
R^ is hydrogen or methyl;
RlO is independently selected from hydrogen, C1-C6 alkyl, benzyl and aryl;
Rl 1 is independently selected from C1-C6 alkyl and aryl;
Al and A^ are independently selected from: a bond, -CH=CH-, -C≡C- -C(O)-, -C(O)NRl0_, O, -N(Rl0)-, 0r S(0)m; V is selected from: a) hydrogen, b) heterocycle selected from pyrrolidinyl, imidazolyl, pyridinyl, thiazolyl, pyridonyl, 2-oxopiperidinyl, indolyl, quinolinyl, isoquinolinyl, and thienyl, c) aryl, d) Cl -C20 alkyl wherein from 0 to 4 carbon atoms are replaced with a a heteroatom selected from O, S, and N, and e) C2-C20 alkenyl, and provided that V is not hydrogen if A ^ is S(0)m and V is not hydrogen if A 1 is a bond, n is 0 and A 2 is S(0)m,
X is -CH2- or -C(=0)-;
Y is a bond, -CH2-, -NH-, -S(=0)m- or O;
Z is selected from: 1 ) a unsubstituted or substituted group selected from aryl, heteroaryl, arylmethyl, heteroarylmethyl, arylsulfonyl, heteroarylsulfonyl, wherein the substituted group is substituted with one or more of the following: a) Cl -4 alkyl, unsubstituted or substituted with: C l -4 alkoxy, NR6R7, C3-6 cycloalkyl, aryl, heterocycle, HO, -S(0)mR6a, or -C(0)NR6R7, b) aryl or heterocycle, c) halogen, d) OR6, e) NR6R7, f) CN, g) Nθ2, h) CF3; i) -S(0)mR6a, j) -C(0)NR6R7, or k) C3-C6 cycloalkyl; or 2) unsubstituted C1 -C6 alkyl, substituted C1 -C6 alkyl, unsubstituted C3-C6 cycloalkyl or substituted C3-C6 cycloalkyl,wherein the substituted C1 -C6 alkyl and substituted C3-C6 cycloalkyl is substituted with one or two of the following: a) Cl -4 alkoxy, b) NR6R7, c) C3-6 cycloalkyl, d) -NR6C(0)R7, e) HO, f) -S(0)mR6a, g) halogen, or h) perfluoroalkyl;
n is 0, 1 , 2, 3 or 4; p is 0, 1 , 2, 3 or 4; and r is 0 to 5, provided that r is 0 when V is hydrogen;
or the pharmaceutically acceptable salts thereof.
In a more preferred embodiment of this invention, the inhibitors of famesyl-protein transferase are illustrated by the formula C:
wherein:
Rib is independently selected from: a) hydrogen, b) aryl, heterocycle, cycloalkyl, Rl°0-, -N(Rl°)2 or C2-C6 alkenyl, c) C1-C6 alkyl unsubstituted or substituted by aryl, heterocycle, cycloalkyl, alkenyl, RI^O-, or -N(R 0)2;
R3 and R^ are independently selected from H and CH3;
R and R4 are independently selected from H; O or Ci-5 alkyl, unbranched or branched, unsubstituted or substituted with one or more of:
R6 and R7 are independently selected from: a) hydrogen, b) C l -C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C 1 -C6 perfluoroalkyl, F, Cl, RlOO-, R!0C(O)NR10-, CN, N02,
(Rl0)2N-C(NRl0)-, Rl°C(0)-, Rl°OC(0)-, -N(RlO)2, or RllθC(O)NRl0-, and c) Cl-Cό alkyl substituted by Cl -C6 perfluoroalkyl, R 1°0-, Rl0C(O)NRl0-, (R10)2N-C(NR10)-, RlOC(O)-, RlOθC(O)-, -N(Rl0)2,orRllOC(O)NRl0-; R6 is selected from:
Cl-4 alkyl or C3-6 cycloalkyl, unsubstituted or substituted with: a) Cl-4 alkoxy, b) halogen, or c) aryl or heterocycle;
R8 is independently selected from: a) hydrogen, b) Cl -C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, Cl -C6 perfluoroalkyl, F, Cl, RlOO-, R!0C(O)NR10-, CN, NO2, (RlO)2N-C(NR O)-, Rl°C(0)-, -N(RlO)2, or R11OC(O)NR10-, and c) C1-C6 alkyl substituted by Cj-Cό perfluoroalkyl, Rl°0-, R!0C(O)NR1°-, (RlO)2N-C(NRlO)-, RlOC(O)-,
-N(R 10)2, or R 11 OC(0)NR 10-;
RlO is independently selected from hydrogen, C1-C6 alkyl, benzyl and aryl;
Rl 1 is independently selected from Cl -C alkyl and aryl;
Xis -CH2- or -C(=0)-;
Y is a bond, -CH2-, -NH-, -S(=0)m- or O;
Z is selected from:
1) a unsubstituted or substituted group selected from aryl, heteroaryl, arylmethyl, heteroarylmethyl, arylsulfonyl, heteroarylsulfonyl, wherein the substituted group is substituted with one or more of the following: a) Cl-4 alkyl, unsubstituted or substituted with: Cl-4 alkoxy, R6R7, C3-6 cycloalkyl, aryl, heterocycle, HO, -S(0)mR6a, or -C(0)NR6R75
or the pharmaceutically acceptable salts thereof.
In a second more preferred embodiment of this invention, the inhibitors of famesyl-protein transferase are illustrated by the formula D: Z
wherein:
Rib is independently selected from: a) hydrogen, b) aryl, heterocycle, cycloalkyl, R 10θ-, -N(R 1 °)2 or C2-C6 alkenyl, c) C1-C6 alkyl unsubstituted or substituted by aryl, heterocycle, cycloalkyl, alkenyl, RlOO-, or -N(Rl )2;
R2 and R^ are independently selected from: hydrogen or Cl-C6 alkyl;
R3 and R^ are hydrogen;
R6 and R7 are independently selected from: a) hydrogen, b) C 1 -C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C 1 -Cβ perfluoroalkyl, F, Cl, Rl O-, R!0C(O)NR10-, CN, Nθ2, (R10)2N-C(NR10)-, RlOc(O)-, Rl θC(O)-, -N(Rl )2, or RllθC(O)NRl0-,and c) C1-C6 alkyl substituted by C1-C6 perfluoroalkyl, Rl°0-, Rl0C(O)NRl0-, (R10)2N-C(NR10 , RlOc(O)-,
R 10θC(O)-, -N(R 10)2, or R 11 OC(0)NR 1 .;
RlO is independently selected from hydrogen, C1-C6 alkyl, benzyl and aryl;
Rl 1 is independently selected from C1-C6 alkyl and aryl; X is -CH2- or -C(=0)-;
Y is a bond, -CH2-, -NH-, -S(=0)m- or O;
or the pharmaceutically acceptable salts thereof. The preferred compounds of this invention are as follows:
N-[2-{ l -(4-Cyanobenzyl)-5-imidazolyl }ethyl]-3-carbamoyl-l -phenyl-2- piperidinone
3-f 3- { 1 -(4-Cyanobenzyl)-5-imidazolyl } - 1 -propyl]- 1 -phenyl-2- piperidinone
3-[3- { 1 -(4-Cyanobenzyl)-5-imidazolyl } - 1 -propyll- 1 -phenyl-2- pyrrolidinone
(±)cis--3-[3- { 1 -(4-Cyanobenzyl)-5-imidazolyl } - 1 -propy l]-4- methoxymethyl-l -phenyl-2-pyrrolidinone
(±)trans-3-[3- { 1 -(4-Cyanobenzyl)-5-imidazolyl } - 1 -propyl]-4- methoxymethyl-l -phenyl-2-pyrrolidinone
3-[2- { 5-(4-Cyanobenzyl)- 1 -imidazolyl } - 1 -ethyl]- 1 -phenyl-2- pyrrolidinone
1 -Benzyl-3-12- { 5-(4-cyanobenzyl)- 1 -imidazolyl } - 1 -ethyl] -2- pyrrolidinone and
(±)- 1 -(3-chloropheny)-3-[ 1 -( 1 -(4-cyanobenzyl)- 5-imidazolyl)-l-(hydroxy)methyl]-2-piperazinone
or the pharmaceutically acceptable salts or optical isomers thereof.
Specific examples of the compounds of the invention are:
N-[2-{ l -(4-Cyanobenzyl)-5-imidazolyl } ethyl] -3-carbamoyl- l -phenyl-2- piperidinone
3-[2-{ 5-(4-Cyanobenzyl)-l -imidazolyl } -1 -ethyl]- 1 -phenyl-2- pyrrolidinone
1 -Benzyl-3-[2- { 5-(4-cyanobenzyl)- 1 -imidazolyl } - 1 -ethyl] -2- pyrrolidinone
or the pharmaceutically acceptable salts thereof.
The compounds of the present invention may have asymmetric centers and occur as racemates, racemic mixtures, and as individual diastereomers, with all possible isomers, including optical isomers, being included in the present invention. When any variable (e.g. aryl, heterocycle, R l , R2 etc.) occurs more than one time in any constituent, its definition on each occurence is independent at every other occurence. Also, combinations of substituents/or variables are permissible only if such combinations result in stable compounds.
As used herein, "alkyl" is intended to include both branched and straight-chain saturated aliphatic hydrocarbon groups having the 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.
As used herein, "aryl" is intended to mean any stable monocyclic or bicyclic carbon ring of up to 7 members in each ring, wherein at least one ring is aromatic. Examples of such aryl elements include phenyl, naphthyl, tetrahydronaphthyl, indanyl, biphenyl, phenanthryl, anthryl or acenaphthyl.
The term heterocycle or heterocyclic, as used herein, represents a stable 5- to 7-membered monocyclic or stable 8- to 1 1- 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 stmcture. Examples of such heterocyclic elements include, but are not limited to, azepinyl, benzimidazolyl, benzisoxazolyl, benzofurazanyl, benzopyranyl, benzothiopyranyl, benzofuryl, benzothiazolyl, benzothienyl, benzoxazolyl, chromanyl, cinnolinyl, dihydrobenzofuryl, dihydrobenzothienyl, dihydrobenzothiopyranyl, dihydrobenzothiopyranyl sulfone, furyl, imidazolidinyl, imidazolinyl, imidazolyl, indolinyl, indolyl, isochromanyl, isoindolinyl, isoquinolinyl, isothiazolidinyl, isothiazolyl, isothiazolidinyl, morpholinyl, naphthyridinyl, oxadiazolyl, 2-oxoazepinyl, oxazolyl, 2-oxopiperazinyl, 2-oxopiperdinyl, 2-oxopyrrolidinyl, piperidyl, piperazinyl, pyridyl, pyrazinyl, pyrazolidinyl, pyrazolyl, pyridazinyl, pyrimidinyl, pyrrolidinyl, pyrrolyl, quinazolinyl, quinolinyl, quinoxalinyl, tetrahydrofuryl, tetrahydroisoquinolinyl, tetrahydroquinolinyl, thiamo holinyl, thiamorpholinyl sulfoxide, thiazolyl, thiazolinyl, thienofuryl, thienothienyl, and thienyl.
As used herein, "heteroaryl" is intended to mean any stable monocyclic or bicyclic carbon ring of up to 7 members in each ring, wherein at least one ring is aromatic and wherein from one to four carbon atoms are replaced by heteroatoms selected from the group consisting of N, O, and S. Examples of such heterocyclic elements include, but are not limited to, benzimidazolyl, benzisoxazolyl, benzofurazanyl, benzopyranyl, benzothiopyranyl, benzofuryl, benzothiazolyl, benzothienyl, benzoxazolyl, chromanyl, cinnolinyl, dihydrobenzofuryl, dihydrobenzothienyl, dihydrobenzothiopyranyl, dihydrobenzothiopyranyl sulfone, furyl, imidazolyl, indolinyl, indolyl, isochromanyl, isoindolinyl, isoquinolinyl, isothiazolyl, naphthyridinyl, oxadiazolyl, pyridyl, pyrazinyl, pyrazolyl, pyridazinyl, pyrimidinyl, pyrrolyl, quinazolinyl, quinolinyl, quinoxalinyl, tetrahydroisoquinolinyl, tetrahydroquinolinyl, thiazolyl, thienofuryl, thienothienyl, and thienyl.
As used herein in the definition of R2 and R^, the term "the substituted group" intended to mean a substituted -8 alkyl, substituted C2-8 alkenyl, substituted C2-8 alkynyl, substituted aryl or substituted heterocycle from which the substitutent(s) R and R3 are selected.
As used herein in the definition of R°, Roa, R7 and R7a? the substituted Cl -8 alkyl, substituted C3-6 cycloalkyl, substituted aroyl, substituted aryl, substituted heteroaroyl, substituted arylsulfonyl, substituted heteroarylsulfonyl and substituted heterocycle include moieties containing from 1 to 3 substitutents in addition to the point of attachment to the rest of the compound. Preferably, such substitutents are selected from the group which includes but is not limited to F, Cl, Br, CF3, NH2, N(Cl -C6 alkyl)2, Nθ2, CN, (C1-C6 alkyl)0-, -OH, (C1 -C6 alkyl)S(0)m-, (C1 -C6 alkyl)C(0)NH-, H2N-C(NH)-, (C1-C6 alkyl)C(O)-, (C1 -C6 alkyl)OC(O)-, N3,(Cl -C6 alkyl)OC(0)NH-, phenyl, pyridyl, imidazolyl, oxazolyl, isoxazolyl, thiazolyl, thienyl, furyl, isothiazolyl and C1 -C2O alkyl. When R2 and R3 are combined to form - (CH2)u -» cyclic moieties are formed. Examples of such cyclic moieties include, but are not limited to:
In addition, such cyclic moieties may optionally include a heteroatom(s). Examples of such heteroatom-containing cyclic moieties include, but are not limited to:
Lines drawn into the ring systems from substituents (such as from R , R , R4 etc ) indicate that the indicated bond may be attached to any of the substitutable ring carbon atoms.
Preferably, Rla and R ib are independently selected from: hydrogen, -N(R 10)2, R l0C(O)NR l0- or unsubstituted or substituted C1 -C6 alkyl wherein the substituent on the substituted Cl -C6 alkyl is selected from unsubstituted or substituted phenyl, -N(RlO)2, R I Ø- and Rl°C(O)NRl0-. Preferably, R is selected from: H, \ 1R6R7 _ ^^-OR6
O O and an unsubstituted or substituted group, the group selected from Cl -8 alkyl, C2-8 alkenyl and C2-8 alkynyl; wherein the substituted group is substituted with one or more of: 1 ) aryl or heterocycle, unsubstituted or substituted with: a) Cl -4 alkyl,
d) halogen, 2) C3-6 cycloalkyl,
3) OR6,
4) SR6a, S(0)R6a, Sθ2R6a,
5) — NR6R7
0\ . NR6R7
8) T O
9) O^ .OR
T O
11) — S02- NR6R7
o
15) N3l or
16) F.
Preferably, R3 is selected from: hydrogen and Cl -C6 alkyl.
Preferably, R4 and R^ are hydrogen.
Preferably, R6, R7 and R7a is selected from: hydrogen, unsubstituted or substituted Cl-C6 alkyl, unsubstituted or substituted aryl and unsubstituted or substituted cycloalkyl. Preferably, R6a i unsubstituted or substituted C1-C6 alkyl, unsubstituted or substituted aryl and unsubstituted or substituted cycloalkyl.
Preferably, R°- is hydrogen or methyl. Most preferably, Ra is hydrogen. Preferably, R IO is selected from H, C1 -C6 alkyl and benzyl.
Preferably, Al and A2 are independently selected from: a bond, -C(0)NR lO-, -NR IOC(O)-, O, -N(R 10)-, -S(0)2N(R 10). and-
Preferably, V is selected from hydrogen, heterocycle and aryl. More preferably, V is phenyl.
Preferably, Y is a bond or -CH2-. More preferably, Y is -CH2-. Preferably, 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. Preferably, W is selected from imidazolinyl, imidazolyl, oxazolyl, pyrazolyl, pyyrohdinyl, thiazolyl and pyridyl. More preferably, W is selected from imidazolyl and pyridyl.
Preferably, n and r are independently 0, 1 , or 2. Preferably p is 1 , 2 or 3. Preferably s is 0.
Preferably t is 1. Preferably, the moiety
is selected from:
It is intended that the definition of any substituent or variable (e.g., Rl a, R° n, etc.) at a particular location in a molecule be independent of its definitions elsewhere in that molecule. Thus,
-N(R lO)2 represents -NHH, -NHCH3, -NHC2H5, etc. It is understood that substituents and substitution patterns on the compounds of the instant invention can be selected by one of ordinary skill in the art to provide compounds that are chemically stable and that can be readily synthesized by techniques known in the art, as well as those methods set forth below, from readily available starting materials.
The pharmaceutically acceptable salts of the compounds of this invention include the conventional non-toxic salts of the compounds of this invention as formed, e.g., from non-toxic inorganic or organic acids. For example, such conventional non-toxic salts include those derived from inorganic acids such as hydrochloric, hydrobromic, sulfuric, sulfamic, phosphoric, nitric and the like: and the salts prepared from organic acids such as acetic, propionic, succinic, glycolic, stearic, lactic, malic, tartaric, citric, ascorbic, pamoic, maleic, hydroxymaleic, phenylacetic, glutamic, benzoic, salicylic, sulfanilic, 2-acetoxy-benzoic, fumaric, toluenesulfonic, methanesulfonic, ethane disulfonic, oxalic, isethionic, trifluoroacetic and the like.
The pharmaceutically acceptable salts of the compounds of this invention can be synthesized from the compounds of this invention which contain a basic moiety by conventional chemical methods. Generally, the salts are prepared either by ion exchange chromatography or by reacting the free base with stoichiometric amounts or with an excess of the desired salt-forming inorganic or organic acid in a suitable solvent or various combinations of solvents. Reactions used to generate the compounds of this invention are prepared by employing reactions as shown in the Schemes 1 - 15, 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 R2, R3, R4 ^d R5; however the point of attachment to the ring is illustrative only and is not meant to be limiting.
These reactions may be employed in a linear sequence to provide the compounds of the invention or they may be used to synthesize fragments which are subsequently joined by the reductive alkylation reactions described in the Schemes.
Synopsis of Schemes 1 -15: The requisite intermediates are in some cases commercially available, or can be prepared according to literature procedures, for the most part. In Scheme 1 , for example, the synthesis of 3-substituted-2- piperidone is outlined. Thus a suitably substituted 2-hydroxynicotinic acid I may be catalytically hydrogenated to provide the 2-piperidone II. This intermediate may be coupled to a suitably substituted aryl moiety to provide the key intermediate III. The ester of compound III can be saponified and the acid can be reacted with an amine such as IV to provide the instant compound V.
Alternatively, as shown in Scheme 2, intermediate III can be deprotonated and reacted with an electrophile, such as VI, to provide the instant compound VII. Compound VII can also be decarboxylated to provide the instant compound VIII. Similar coupling to an aryl moiety, deprotonation and alkylation can be performed on a pyrrolidinone, as shown in Scheme 3, to provide the instant compound IX. Scheme 3a illustrates an alternative synthesis of the pyrrolidinone compounds of the instant invention. Thus a suitably substituted 4-halobutanoyl chloride may be cyclized with an amine in a two step procedure and then selectively functionalized adjacent to the carbonyl to provide the instant compound having a preferred arylmethylimidazolyl moiety.
Preparation of the instant compounds wherein Y is NH is illustrated in Schem 3b. Thus the suitably substituted N- aminoethylaniline is prepared and undergoes a two step cyclization with chloroacetyl chloride to provide the protected piperazinone. This intermediate can undergo selective alkylation reactions such as illustrated in Scheme 3 and hereinbelow. After incorporation of a moiety at the 3-position, the N-protecting group may be removed by methods known in the art, such as with HCI in ethyl acetate.
Scheme 3c illustrates preparation of the instant compounds wherein Y is S. The suitably substituted thiomoφholinone may be selectively alkylated with reagents illustrated in Scheme 3 and hereinbelow and may also be oxidized subsequent to the alkylation as shown in the scheme. Preparation of a thiomoφholinone intermediate having a substituent in the 5-position is illustrated in Scheme 3d.
Scheme 4 illustrates preparation of the instant copounds wherein the linker X is an amine. As shown in the Scheme the amine intermediate X may be alkylated with a suitably substituted electrophile. The amine intermediate X can be reductively alkylated with a variety of aldehydes, such as XI. 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 3). 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 dichloroe thane, methanol or dimethylformamide. The product XII can be deprotected to give the final compounds XIII with trifluoroacetic acid in methylene chloride. The final product XIII is isolated in the salt form, for example, as a trifluoroacetate, hydrochloride or acetate salt, among others. The product diamine XIII can further be selectively protected to obtain XIV, which can subsequently be reductively alkylated with a second aldehyde to obtain XV. Removal of the protecting group, and conversion to cyclized products such as the dihydroimidazole XVI can be accomplished by literature procedures.
Alternatively, the piperidone intermediate X can be reductively alkylated with other aldehydes such as l -trityl-4-imidazolyl- carboxaldehyde or l-trityl-4-imidazolylacetaldehyde, to give products such as XVII (Scheme 6). The trityl protecting group can be removed from XVII to give XVIII, or alternatively, XVII can first be treated with an alkyl halide then subsequently deprotected to give the alkylated imidazole XIX. Alternatively, the intermediate X can be acylated or sulfonylated by standard techniques. The imidazole acetic acid XX can be converted to the acetate XXI by standard procedures, as shown in Scheme 7, and XXI can be first reacted with an alkyl halide, then treated with refluxing methanol to provide the regiospecifically alkylated imidazole acetic acid ester XXII. Hydrolysis and reaction with intermediate X in the presence of condensing reagents such as l -(3- dimethylaminopropyl)-3-ethylcarbodiimide (EDC) leads to acylated products such as XXIV.
Scheme 7a illustrates the preparation of a suitably substituted benzylimidazolyl aldehyde intermediate whch may be reacted with the piperazinone and thiomoφholinone intermediates whose syntheses is described in Schemes 3b-3d (Scheme 7a and 7b).
If the intermediate X is reductively alkylated with an aldehyde which also has a protected hydroxyl group, such as XXV in Scheme 8, the protecting groups can be subsequently removed to unmask the hydroxyl group (Schemes 8, 9). 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 XXIX. In addition, the fully deprotected amino alcohol XXX can be reductively alkylated (under conditions described previously) with a variety of aldehydes to obtain secondary amines, such as XXXI (Scheme 9), or tertiary amines.
The Boc protected amino alcohol XXVII can also be utilized to synthesize 2-aziridinylmethylpiperazines such as XXXII (Scheme 10). Treating XXVII with l ,l '-sulfonyldiimidazole and sodium hydride in a solvent such as dimethylformamide led to the formation of aziridine XXXII. The aziridine reacted in the presence of a nucleophile, such as a thiol, in the presence of base to yield the ring- opened product XXXIII. In addition, the intermediate X can be reacted with aldehydes derived from amino acids such as O-alkylated tyrosines, according to standard procedures, to obtain compounds such as XXXIX, as shown in Scheme 1 1. When R' is an aryl group, XXXIX can first be hydrogenated to unmask the phenol, and the amine group deprotected with acid to produce XL. Altematively, the amine protecting group in XXXIX can be removed, and O-alkylated phenolic amines such as XLI produced.
Schemes 12-15 illustrate syntheses of suitably substituted aldehydes useful in the syntheses of the instant compounds wherein the variable W is present as a pyridyl moiety. Similar synthetic strategies for preparing alkanols that incoφorate other heterocyclic moieties for variable W are also well known in the art.
SCHEME1
OH
SCHEME 1 (continued^)
SCHEME2
SCHEME 3
sub
SCHEME 3a
TFA, CH2CI2
SCHEME 3a (continued)
1)Tf20, iPr2NEt
SCHEME 3b
SCHEME 3c
Cl CICH COCI \ Na2S
R CICH2COCI \ Ms2O
Et3N HO N-Z Et3N
CH2CI2 / ^
Cl O
SCHEME4
SCHEME5
Boc
XIII
SCHEME 5 (continued)
SCHEME 6
SCHEME 7
XXI
XXII
XXIII
SCHEME 7 (continued)
XXIV sub
SCHEME 7a
SCHEME 7a (continued)
SCHEME 7b
i. LiHMDS, THF
SCHEME 8
XXV
SCHEME 8 (continued)
XXVIII
SCHEME9
XXVII
XXXII
XXXIII
SCHEME 1 1
XXXIV
XXXV
XXXVI
XXXVII XXXVIII
SCHEME 1 1 (continued)
XXXVIII
XL SCHEME 12
NaBH4 (excess)
SCHEME 13
SCHEME 14
NaBH4 (excess)
excess NaBH4
The instant compounds are useful as pharmaceutical agents for mammals, especially for humans. These compounds may be administered to patients for use in the treatment of cancer. Examples of the type of cancer which may be treated with the compounds of this invention include, but are not limited to, colorectal carcinoma, exocrine pancreatic carcinoma, myeloid leukemias and neurological tumors. Such tumors may arise by mutations in the ras genes themselves, mutations in the proteins that can regulate Ras activity (i.e., neurofibromin (NF- 1 ), neu, scr, abl , lck, fyn) or by other mechanisms. The compounds of the instant invention inhibit famesyl- protein transferase and the famesylation 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 blindness related to retinal vascularization.
The compounds of this invention are also useful for inhibiting other proliferative diseases, both benign and malignant, wherein Ras proteins are aberrantly activated as a result of oncogenic mutation in other genes (i.e., the Ras gene itself is not activated by mutation to an oncogenic form) with said inhibition being accomplished by the administration of an effective amount of the compounds of the invention to a mammal in need of such treatment. For example, a component of NF- 1 is a benign proliferative disorder. The instant compounds may also be useful in the treatment of certain viral infections, in particular in the treatment of hepatitis delta and related viruses (J.S. Glenn et al. Science, 256: 1331 -1333 ( 1992).
The compounds of the instant invention are also useful in the prevention of restenosis after percutaneous transluminal coronary angioplasty by inhibiting neointimal formation (C. Indolfi et al. Nature medicine, 1 :541-545(1995).
The instant compounds may also be useful in the treatment and prevention of polycystic kidney disease (D.L. Schaffner et al. American Journal of Pathology, 142: 1051-1060 (1993) and B. Cowley, Jr. et alFASEB Journal, 2:A3160 (1988)).
The instant compounds may also be useful for the treatment of fungal infections. The compounds of this invention may be administered to mammals, preferably humans, either alone or, preferably, in combination with pharmaceutically acceptable carriers or diluents, optionally with known adjuvants, such as alum, in a pharmaceutical composition, according to standard pharmaceutical practice. The compounds can be administered orally or parenterally, including the intravenous, intramuscular, intraperitoneal, subcutaneous, rectal and topical routes of administration.
For oral use of a chemotherapeutic compound according to this invention, the selected compound may be administered, for example, in the form of tablets or capsules, or as an aqueous solution or suspension. In the case of tablets for oral use, carriers which are commonly used include lactose and co starch, and lubricating agents, such as magnesium stearate, are commonly added. For oral administration in capsule form, useful diluents include lactose and dried com starch. When aqueous suspensions are required for oral use, the active ingredient is combined with emulsifying and suspending agents. If desired, certain sweetening and/or flavoring agents may be added. For intramuscular, intraperitoneal, subcutaneous and intravenous use, sterile solutions of the active ingredient are usually prepared, and the pH of the solutions should be suitably adjusted and buffered. For intravenous use, the total concentration of solutes should be controlled in order to render the preparation isotonic.
The compounds of the instant invention may also be co- administered with other well known therapeutic agents that are selected for their particular usefulness against the condition that is being treated. For example, the instant compounds may be useful in combination with known anti-cancer and cytotoxic agents. Similarly, the instant compounds may be useful in combination with agents that are effective in the treatment and prevention of NF-1 , restinosis, polycystic kidney disease, infections of hepatitis delta and related viruses and fungal infections.
If formulated as a fixed dose, such combination products employ the compounds of this invention within the dosage range described below and the other pharmaceutically active agent(s) within its approved dosage range. Compounds of the instant invention may altematively be used sequentially with known pharmaceutically acceptable agent(s) when a combination formulation is inappropriate. The present invention also encompasses a pharmaceutical composition useful in the treatment of cancer, comprising the administration of a therapeutically effective amount of the compounds of this invention, with or without pharmaceutically acceptable carriers or diluents. Suitable compositions of this invention include aqueous solutions comprising compounds of this invention and pharmacolo- gically acceptable carriers, e.g., saline, at a pH level, e.g., 7.4. The solutions may be introduced into a patient's blood-stream by local bolus injection.
As used herein, the term "composition" is intended to encompass a product comprising the specified ingredients in the specific amounts, as well as any product which results, directly or indirectly, from combination of the specific ingredients in the specified amounts.
When a compound according to this invention is administered into a human subject, the daily dosage will normally be determined by the prescribing physician with the dosage generally varying according to the age, weight, and response of the individual patient, as well as the severity of the patient's symptoms.
In one exemplary application, a suitable amount of compound is administered to a mammal undergoing treatment for cancer. Administration occurs in an amount between about 0.1 mg/kg of body weight to about 60 mg/kg of body weight per day, preferably of between 0.5 mg/kg of body weight to about 40 mg/kg of body weight per day.
The compounds of the instant invention are also useful as a component in an assay to rapidly determine the presence and quantity of famesyl-protein transferase (FPTase) in a composition. Thus the composition to be tested may be divided and the two portions contacted with mixtures which comprise a known substrate of FPTase (for example a tetrapeptide having a cysteine at the amine terminus) and famesyl pyrophosphate and, in one of the mixtures, a compound of the instant invention. After the assay mixtures are incubated for an sufficient period of time, well known in the art, to allow the FPTase to famesylate the substrate, the chemical content of the assay mixtures may be determined by well known immunological, radiochemical or chromatographic techniques. Because the compounds of the instant invention are selective inhibitors of FPTase, absence or quantitative reduction of the amount of substrate in the assay mixture without the compound of the instant invention relative to the presence of the unchanged substrate in the assay containing the instant compound is indicative of the presence of FPTase in the composition to be tested.
It would be readily apparent to one of ordinary skill in the art that such an assay as described above would be useful in identifying tissue samples which contain famesyl-protein transferase and quantitating the enzyme. Thus, potent inhibitor compounds of the instant invention may be used in an active site titration assay to determine the quantity of enzyme in the sample. A series of samples composed of aliquots of a tissue extract containing an unknown amount of famesyl-protein transferase, an excess amount of a known substrate of FPTase (for example a tetrapeptide having a cysteine at the amine terminus) and fa esyl pyrophosphate are incubated for an appropriate period of time in the presence of varying concentrations of a compound of the instant invention. The concentration of a sufficiently potent inhibitor (i.e., one that has a Ki substantially smaller than the concentration of enzyme in the assay vessel) required to inhibit the enzymatic activity of the sample by 50% is approximately equal to half of the concentration of the enzyme in that particular sample. EXAMPLES
Examples provided are intended to assist in a further understanding of the invention. Particular materials employed, species and conditions are intended to be further illustrative of the invention and not limitative of the reasonable scope thereof.
EXAMPLE 1
N-[2- { l -(4-Cyanobenzyl)-5-imidazolyl }ethyl]-3-carbamoyl- l -phenyl-2- piperidinone hydrochloride
Step A: 4-Cvanobenzyl-N≤.-phthaloylhistamine
N^-Pivaloyloxymethyl-Nα-phthaloylhistamine (4.55 g, 12.8 mmol) was prepared as previously described (J. C. Emmett, F. H. Holloway, and J. L. Turner, J. Chem. Soc, Perkin Trans. 1 , 1341 , (1979)). α-Bromo-p-tolunitrile (3.77 g, 1 .2 mmol) was dissolved in acetonitrile (70 mL). The solution was heated at 55°C for 4 h, cooled to room temperature, and filtered to remove the white solid. The acetonitrile (30 mL) was concentrated to 1/2 its volume under reduced pressure and the solution was heated at 55°C overnight. The solution was cooled and filtered to give a white solid. The volume of the filtrate was reduced to 10 mL, the solution was heated at 55°C for 1 hr, then cooled to room temperature, diluted with EtOAc (25 mL) and filtered to obtain additional white solid. The solids were combined, dried, and dissolved in methanol (100 mL) which was saturated with ammonia gas while the temperature was maintained below 30°C. The solution was stirred for 1 hr, concentrated to dryness, and extracted with CH2CI2 (3x200 mL), dried (MgS04), concentrated, and chromatographed (silica gel, 10:90: 1 MeOH/CH2Cl2/NH4θH) to give the title compound
Step B: 4-Cvanobenzyl histamine 4-Cyanobenzyl-Nα-phthaloylhistamine (1.64 g, 4.61 mmol), and hydrazine (1.46 mL, 46.1 mmol) were dissolved in absolute ethanol (70 mL). The solution was concentrated after 1 hr and filtered to remove a white precipitate which was washed several times with ethanol. The filtrate was concentrated and the residue was chromatographed (silica gel, 10:90: 1 MeOH/CH2Cl2/NH4θH) to give the title compound.
Step C: 3-Carbethoxy- 1 -phenyl-2-piperidinone
A mixture of 3-carbethoxy-2-piperidinone (1.00 g, 5.84 mmol), triphenylbismuth (3.85 g, 8.77 mmol), copper(II)acetate (1.58 g, 8.77 mmol) and triethylamine (1.22 mL, 8.77 mmol) was stirred for
17 h at 20°C in dichloromethane (25 mL). The reaction mixture was adsorbed onto silica gel and eluted with 30-50% ethyl acetate/hexane.
The title compund was obtained as an oil.
Step D: 3-Carboxy- 1 -pheny 1-2-piperidinone
The product from Step C (0.551 g, 2.56 mmol) was dissolved in methanol (3 mL) and 5% aqueous sodium hydroxide added
(3 mL). After 1 h, methanol was removed in vacuo, and the residue partitioned between ethyl acetate and 10% aqueous HCI. The organic layer was washed with saturated brine, and dried over MgS04. The title compound was obtained as a white solid.
Step E: N-[2-{ l -(4-Cyanobenzyl)-5-imidazolyl }ethyl]-3- carbamoyl- 1 -phenyl-2-piperidinone hydrochloride
The product from Step D (0.223 g, 1.01 mmol) was dissolved in dimethylformamide (5 mL). To this solution was added 4- cyanobenzyl histamine dihydrochloride (0.220 g, 0.84 mmol), EDC • HCI (0.191 g, 1.00 mmol) and 1-hydroxybenzotriazole (0.135 g, 1.00 mmol). The pH was adjusted to 7.5 with triethylamine. After 16 h, the reaction was poured into water and extracted with ethyl acetate. The organic phase was washed with saturated brine and dried over magnesium sulfate. The crude product was chromatographed on silica gel with 10% methanol in methylene chloride. The purified product was converted to the hydrochloride salt with 4N HCI in dioxane. The title compound was obtained as a white solid. FAB ms (m+l) 428. Anal. Calc. for C25H25N5O2 0.55 H2O: C, 63.37; H, 5.76; N, 14.78. Found: C, 63.37; H, 6.05; N, 13.28.
EXAMPLE 2
3-[3- { 1 -(4-Cyanobenzy l)-5-imidazolyl } - 1 -propyl]- 1 -phenyl-2- piperidinone hydrochloride
Step A: Methyl 3-(4-imidazolyl)-2-propenoate
A suspension of urocanic acid (5.0 g) in methanol at 0 °C was saturated with HCI gas. The reaction was stirred for one hour, then concentrated in vacuo to dryness to provide the title compound .
Step B: Methyl 3-(4-imidazolyl)propionate
To a solution of methyl 3-(4-imidazolyl)-2-propenoate (5.3 g) in methanol at room temperature was added 10% palladium on carbon (20 mg) under a blanket of argon.. A balloon of hydrogen was applied, and the reaction was stirred for one hour. The solution was filtered, and then concentrated in vacuo to dryness to provide the title compound. Step C: Methyl 3-( 1 -triphenylmethyl-4-imidazol vDpropionate
To a solution of methyl 3-(4-imidazoIyl)propionate (1.77 g) in 10 mL of dry DMF at room temperature was added triethylamine (3.25 mL). A white solid precipitated from the solution. Chlorotriphenylmethane (2.65 g) in 12 mL of DMF was added dropwise. The reaction mixture was stirred for 20 hours, then poured into 40% ethyl acetate/hexane solution and washed with two portions of water. The organic layer was extracted with sat. aq. NaHCθ3 solution and brine, then dried (Na2Sθ4), filtered, and concentrated in vacuo to provide the title compound.
Step D: 4-(3-Hydroxy- 1 -propyl)-3-( 1 -triphenylmethyl-4- imidazolvDpropionate
The product from Step C (0.063 g, 0.159 mmol) was dissolved in THF and cooled to 0°C under nitrogen. A solution of lithium aluminum hydride in THF (1 M, 0.180 mL) was added and the reaction stirred at 0°C for 20 min. The reaction was quenched with saturated sodium potassium tartrate solution and extracted with ethyl acetate. The title compound was obtained as a clear oil.
Step E: 1 -Pheny 1-2-piperdinone
A mixture of valerolactam (1.00 g, 10.18 mmol), triphenylbismuth (6.72 g, 15.27 mmol), copper(II)acetate (2.77 g, 15.27 mmol) and triethylamine (2.12 mL, 15.27 mmol) was stirred for 17 h at 20°C in dichloromethane (25 mL). The reaction mixture was adsorbed onto silica gel and chromatographed with 40% ethyl acetate/methylene chloride. The title compund was obtained as an oil.
Step F: 3-[3-{ l -triphenylmethyl-4-imidazolyl }- l -propyl]- l -phenyl- 2-piperidinone hydrochloride
A solution of 1 -phenyl-2-piperidinone from Step F in THF is added to a solution of one equivalent of LDA in THF at -78°C, and stirred for 30 min. A solution of 4-(3-hydroxy-l-propyl)-3-( l - triphenylmethyl-4-imidazolyl)propionate in methylene chloride is cooled to -78°C under nitrogen. One equivalent of ι-butyl lithium followed by one equivalent of triflic anhydride is added, the reaction stirred for 10 min, then added to the LDA/l -phenyl-2-piperidinone solution. The reaction is warmed to room temperature, and quenched with saturated ammonium chloride. The reaction is then partitioned between ethyl acetate and saturated brine. The organic phase is dried over magnesium sulfate, filtered and concentrated. The title compound is isolated by chromatography on silica gel.
Step G: 3-[3-{ l-(4-Cyanobenzyl)-5-imidazolyl }-l-propyl]-l- phenyl-2-piperidinone hydrochloride
The product from Step F is dissolved in acetonitrile and one equivalent of 4-cyanobenzylbromide added. The reaction is stirred at room temperature overnight, concentrated, and taken up in methanol. The methanol solution is refluxed for 3 h, concentrated, and partitioned between ethyl acetate and saturated sodium bicarbonate solution. The organic phase is washed with saturated brine and dried over magnesium sulfate. The title compound is obtained after chromatography on silica gel, and conversion to the dihydrochloride salt.
EXAMPLE 3
3-[3- { 1 -(4-Cyanobenzyl)-5-imidazolyl } - 1 -propylj- 1 -phenyl-2- pyrrolidinone hydrochloride
Step A: 1 -Phenyl-2-pyrrolidinone
The title compound is prepared according to the procedure described in Example 2, Step E, except using 2-pyrrolidinone in place of valerolactam. The cmde product is purified by column chromatography.
Step B: 3-[3-{ l -triphenylmethyl-4-imidazolyl }-l -propyl]-l -phenyl- 2-pyrrolidinone
The title compound is prepared according to the procedure described in Example 2, Step F, except using 1 -phenyl-2-pyrrolidinone in place of 1 -phenyl-2-piperdinone. The title compound is isolated by chromatography on silica gel.
Step C: 3-[3-{ l -(4-Cyanobenzyl)-5-imidazolyl } - l-propyl]- l - phenyl-2-pyrrolidinone hydrochloride
The title compound is prepared according to the procedure described in Example 2, Step G except using 3-[3-{ l-triphenylmethyl-4- imidazolyl }-l -propyl]-l -phenyl-2-pyrrolidinone in place of 3-[3-{ 1- triphenylmethyl-4-imidazolyl } -l -propyl]-l -phenyl-2-piperidinone. The title compound is obtained after chromatography on silica gel, and conversion to the hydrochloride salt.
EXAMPLE 4
(±)cis- and (±)trans-3-[3-{ l-(4-CyanobenzyI)-5-imidazolyl } -l -propyl]- 4-methoxymethyl- 1 -phenyl-2-pyrrolidinone hydrochloride
Step A: 4-Carbomethoxy- 1 -pheny 1-2-pyrrolidinone
Commercially available 4-carboxy- 1 -phenyl-2- pyrrolidinone in 10% methanol in toluene is treated with an excess of trimethylsilyldiazomethane for 30 min. Excess diazomethane reagent is destroyed with glacial acetic acid. The solvents are removed in vacuo and the title compound is isolated.
Step B: 4-Hydroxymethy 1- 1 -phenyl-2-pyrrolidinone
The product from Step A in THF is added to a 0°C solution of 2.5 eq lithium aluminum hydride in THF, under a nitrogen atmosphere. After 30 min the reaction is quenched with sodium hydrogen sulfate, and filtered through a bed of celite. The filtrate is concentrated and the residue partitioned between 2% aqueous sodium hydrogen sulfate and ethyl acetate. The organic layer is washed with saturated brine and dried over magnesium sulfate. Filtration and concentration provides the title compound.
Step C: 4-Methoxymethyl- 1 -phenyl-2-pyrrolidinone
The product from Step C in THF is added to 1.5 eq sodium hydride and 1 eq methyl iodide in THF at 0°C. After 3h the reaction is quenched with water and extracted with ethyl acetate. The ethyl acetate is washed with saturated brine, dried over magnesium sulfate, filtered and concentrated. The title compound is isolated after column chromatography.
Step D: (±)cis- and (±)/rø/2s-4-Methoxymethyl-l-phenyl-3-[3-{ 1 -t riphenylmethyl-4-imidazolyl } - 1 -propyl]-2- pyrrolidinone
The title compound is prepared according to the procedure described in Example 2, Step F, except using 4-methoxymethyl-l - phenyl-2-pyrroIidinone in place of l-phenyl-2-piperdinone. The title compounds are isolated by chromatography on silica gel.
Step E: (±)cis- and (±)trans-3-[3-{ 1 -(4-Cyanobenzyl)-5- imidazolyl } - 1 -propyl ]-4-methoxymethy 1- 1 -pheny 1-2- pyrrolidinone hydrochloride
The title compound is prepared according to the procedure described in Example 2, Step G except using cis -and trans-4- methoxymethyl- 1 -pheny l-3-[3- { 1 -triphenylmethyl-4-imidazolyl } - 1 - propyl]-2-pyrrolidinone_in place of 3-f3-{ l -triphenylmethyl-4- imidazolyl }-l -propyl]- 1 -pheny 1-2-piperidinone. The title compounds are separated by preparative HPLC and converted to their hydrochloride salts.
EXAMPLE 5
3-[2- { 5-(4-Cyanobenzyl)- 1 -imidazolyl } - 1 -ethyl]- 1 -phenyl-2- pyrrolidinone hydrochloride
Step A: N-Phenyl 4-chlorobutyramide
A solution of aniline ( 10.0 mL, 1 10 mmol) and triethylamine (18.4 mL, 132 mmol) in methylene chloride (100 mL) was cooled to 0°C under argon, and 4-chlorobutyrylchloride (14.8 mL, 132 mmol) added dropwise. The reaction was stirred at room temperature for 1.5 h, then extracted with 1M potassium hydrogen sulfate. The organic phase was separated, washed with saturated brine, and dried over sodium sulfate. After concentration in vacuo, the white solid was triturated with hexane and filtered, giving the title compound.
Step B: 1 -Phenyl-2-pyrrolidinone The product from Step A (21.7 g, 1 10 mmol) was dissolved in THF (157 mL) under argon, and sodium hydride (60% dispersion in oil) (5.01 g, 125 mmol) added slowly portionwise. The reaction was stirred at room temperature overnight. An additional portion of sodium hydride was added (1 g) and the reaction continued to stir for 3 h. The reaction was diluted with ethyl acetate and poured into 10% aqueous hydrogen chloride. The organic phase was washed with saturated brine, and dried over sodium sulfate. Concentration in vacuo afforded the title compound.
Step C: 3-(f-ButylcarboxymethyD- 1 -phenyl-2-pyrrolidinone
Diisopropylamine (0.722 mL, 41.0 mmol) in THF 950 mL) was cooled under argon to -78°C and a solution of n- butyllithium in hexane (15.1 mL of 2.5 M solution, 37.8 mmol) was added. After stirring at 0°C for 15 min, this solution was transferred via cannula to a -78°C solution of the product from Step B (5.08 g, 31.5 mmol) in THF (108 mL). When the addition was complete, the reaction was stirred at 0°C for lh, then cooled to -78°C and /-butyl bromoacetate (5.12 mL, 34.7 mmol) added via syringe. The reaction was stirred at -78°C for 2 h, then allowed to warm to room temperature ovemight. The reaction was quenched with 10% aqueous HCI and extracted with ethyl acetate. The organic layer was washed with saturated brine and dried over sodium sulfate. The title compound was obtained after chromatography on silica gel with 20% ethyl acetate in hexane.
Step D: 3-(2-CarboxymethyP- 1 -pheny 1-2-pyrrolidinone
The product from Step C (2.37 g, 8.60 mmol) was stirred in methylene chloride containing 2% water and 10% trifluoroacetic acid (60 mL). Subsequently, additional water ( 1 mL) and trifluoroacetic acid (5 mL) was added and the reaction stirred at room temperature ovemight. The solvent was removed in vacuo, and the residue partitioned between saturated ammonium chloride and ethyl acetate. The combined organic layers were washed with saturated brine, and dried over sodium sulfate. Filtration and evaporation of solvent gave the title compound.
Step E: 3-(2-HvdroxyethvD- 1 -phenyl-2-pyrrolidinone
Ethyl chloroformate (0.201 mL, 2.10 mmol) was added to a 0°C solution of triethylamine ( 0.294 mL, 2.10 mmol) and the product from Step D (0.420 g, 1.91 mmol) in THF (10 mL). The reaction was stirred for 1.5 h, and sodium borohydride added (0.217 g, 5.74 mmol). The reaction was stirred at room temperature ovemight, then quenched with saturated sodium bicarbonate solution. The layers were separated and the aqueous phase extracted with additional ethyl acetate. The combined organic phases were washed with saturated brine and dried over sodium sulfate. The title compound was obtained after purification on silica gel with 0-70% ethyl acetate in hexane. Step F: l -Trityl-4-(4-cvanobenzyl)-imidazole.
To a suspension of activated zinc dust (3.57 g, 54.98 mmol) in THF (50 mL) was added dibromoethane (0.315 mL, 3.60 mmol) and the reaction stirred under argon at 20°C. The suspension was cooled to 0°C and α-bromo-p-toluinitrile (9.33 g, 47.6 mmol) in THF (100 mL) was added dropwise over a period of 10 min. The reaction was then allowed to stir at 20°C for 6 h and bis(triphenylphosphine)Nickel II chloride (2.4 g, 3.64 mmol) and 4 -iodotrityl imidazole (15.95 g, 36.6 mmol) was added in one portion. The resulting mixture was stirred 16 h at 20°C and then quenched by addition of saturated ammonium chloride solution (100 mL) and the mixture stirred for 2 h. Saturated sodium bicarbonate solution was added to give a pH of 8 and the solution was extracted with ethyl acetate ( 2 x 250 mL), dried with magnesium sulfate, and the solvent evaporated in vacuo. The residue was chromatographed on silica gel with 0-20% ethyl acetate in methylene chloride to afford the title compound as a white solid. iH NMR δ CDC13 (7.54 (2H,d, J=7.9Hz), 7.38(lH,s), 7.36-7.29 (H H,m), 7.15-7.09(6H,m), 6.58( l H,s), and 3.93(2H,s)ppm.
Step G: 3-[2- { 5-(4-Cyanobenzyl)- 1 -imidazolyl } - 1 -ethyl]- 1 -phenyl-
2-pyrrolidinone hydrochloride
A solution of the products from Step E (0.100 g, 0.487 mmol) and Step F (0.207 g, 0.487 mmol) and diisopropylethyl amine (0.094 mL, 0.535 mmol) in methylene chloride (2 mL) was cooled to -78°C under argon. Trifluoromethanesulfonic acid anhydride (0.084 mL, 0.501 mmol) was added and the reaction warmed to room temperature over a period of two hours. The reaction was stirred at room temperature ovemight, and then the solvent removed in vacuo. The residue was dissolved in methanol (3 mL) and refluxed for 2 h. The methanol was evaporated, and the residue partitioned between ethyl acetate and saturated sodium bicarbonate solution. The organic phase was washed with saturated brine, and dried over sodium sulfate. The crude product was chromatographed on silica gel with 50% acetone in methylene chloride followed by 10% methanol in chloroform. The title compound was obtained after conversion to the HCI salt. FAB ms: 371 (M+l ). Anal. Calc for C23H22N4O • 1.55 HCI - 0.30 H20, C, 63.89; H, 5.63; N.I2.96. Found: C, 63.88; H, 5.64; N, 12.82.
EXAMPLE 6
1 -Benzyl-3-[2- { 5-(4-cyanobenzyl)- 1 -imidazolyl } - 1 -ethyl] -2- pyrrolidinone hydrochloride
Step A: N-Benzyl 4-chlorobutyramide
The title compound was prepared according to the procedure described in Example 5, Step A, using benzylamine in place of aniline.
Step B: 1 -Benzy l-3-[2- { 5-(4-cyanobenzy 1)- 1 -imidazolyl } - 1 -ethy 1] - 2-pyrrolidinone hydrochloride
Using the product of Step A, the title compound was prepared according to the procedure described in Example 5, Steps B-G. FAB ms: 385 (M+l ). Anal. Calc for C24H24N4O • 2.3 HCI • 0.60 Et2O, C, 61.83; H, 6.35; N.10.93. Found: C, 61.85; H, 5.97; N, 10.61.
EXAMPLE 7 Preparation of (±)-l -(3-chloropheny)-3-l l -(l -(4-cyanobenzyl)- 5-imidazolvD- 1 -(hvdroxy)methyl1-2-piperazinone hydrochloride
Step A: Preparation of l -triphenylmethyl-4-(hydroxymethyl)- imidazole
To a solution of 4-(hydroxymethyl)imidazole hydrochloride (35.0 g, 260 mmol) in 250 mL of dry DMF at room temperature was added triethylamine (90.6 mL, 650 mmol). A white solid precipitated from the solution. Chlorotriphenylmethane (76.1 g, 273 mmol) in 500 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
Alcohol from Step A (260 mmol, prepared above) was suspended in 500 mL of pyridine. Acetic anhydride (74 mL, 780 mmol) was added dropwise, and the reaction was stirred for 48 hours during which it became homogeneous. The solution was poured into 2 L of EtOAc, washed with water (3 x 1 L), 5% aq. HCI soln. (2 x 1 L), sat. aq. NaHC03, and brine, then dried (Na2Sθ4), filtered, and concentrated in vacuo to provide the cmde product. The acetate was isolated as a white powder which was sufficiently pure for use in the next reaction.
Step C: Preparation of l-(4-cyanobenzyl)-5-(acetoxymethyl)- imidazole hydrobromide
A solution of the product from Step B (85.8 g, 225 mmol) and α-bromo- -tolunitrile (50.1 g, 232 mmol) in 500 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 500 mL of methanol, and warmed to 60 °C. After two hours, the solution was reconcentrated in vacuo to provide a white solid which was triturated with hexane to remove soluble materials. Removal of residual solvents in vacuo provided the titled product hydrobromide as a white solid which was used in the next step without further purification.
Step D: Preparation of l-(4-cyanobenzyl)-5-(hydroxymethyl)- imidazole
To a solution of the acetate from Step C (50.4 g, 150 mmol) in 1.5 L of 3: 1 THF/water at 0 °C was added lithium hydroxide monohydrate (18.9 g, 450 mmol). After one hour, the reaction was concentrated in vacuo, diluted with EtOAc (3 L), and washed with water, sat. aq. NaHC03 and brine. The solution was then dried (Na2Sθ4), filtered, and concentrated in vacuo to provide the cmde product as a pale yellow fluffy solid which was sufficiently pure for use in the next step without further purification.
Step E: Preparation of l-(4-cyanobenzyl)-5-imidazole- carboxaldehyde
To a solution of the alcohol from Step D (21.5 g, 101 mmol) in 500 mL of DMSO at room temperature was added triethylamine (56 mL, 402 mmol), then Sθ3-pyridine complex (40.5 g, 254 mmol). After 45 minutes, the reaction was poured into 2.5 L of EtOAc, washed with water (4 x 1 L) and brine, dried (Na2S04), filtered, and concentrated in vacuo to provide the aldehyde as a white powder which was sufficiently pure for use in the next step without further purification.
Step F: Preparation of N-(3-chlorophenyl)ethylenediamine hydrochloride To a solution of 3-chloroaniline (30.0 mL, 284 mmol) in 500 mL of dichloromethane at 0 °C was added dropwise a solution of 4 N HCI in 1 ,4-dioxane (80 mL, 320 mmol HCI). The solution was warmed to room temperature, then concentrated to dryness in vacuo to provide a white powder. A mixture of this powder with 2- oxazolidinone (24.6 g, 282 mmol) was heated under nitrogen atmosphere at 160 °C for 10 hours, during which the solids melted, and gas evolution was observed. The reaction was allowed to cool, forming the crude diamine hydrochloride salt as a pale brown solid. Step G: Preparation of /V-( /τ-butoxycarbonyl)-/V'-(3- chlorophenyDethylenediamine
The amine hydrochloride from Step F (ca. 282 mmol, cmde material prepared above) was taken up in 500 mL of THF and 500 mL of sat. aq. NaHCθ3 soln., cooled to 0 °C, and άi-tert- butylpyrocarbonate (61.6 g, 282 mmol) was added. After 30 h, the reaction was poured into EtOAc, washed with water and brine, dried (Na2Sθ4), 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 ΛH2-(f -butoxycarbamoyl)ethyl]-/V-(3- chloropheny 1 )-2-chloroacetami de
A solution of the product from Step G (77 g, ca. 282 mmol) and triethylamine (67 mL, 480 mmol) in 500 mL of CH2CI2 was cooled to 0 °C. Chloroacetyl chloride (25.5 mL, 320 mmol) was added dropwise, and the reaction was maintained at 0 °C with stirring. After 3 h, another portion of chloroacetyl chloride (3.0 mL) was added dropwise. After 30 min, the reaction was poured into EtOAc (2 L) and washed with water, sat. aq. NH4CI soln, sat. aq. NaHCθ3 soln., and brine. The solution was dried (Na2Sθ4), filtered, and concentrated in vacuo to provide the chloroacetamide as a brown oil which was used in the next step without further purification.
Step I: Preparation of 4-(/crt-butoxycarbonyl)- 1 -(3- chlorophenvl)-2-piperazinone
To a solution of the chloroacetamide from Step H (ca. 282 mmol) in 700 mL of dry DMF was added K2CO3 (88 g, 0.64 mol). The solution was heated in an oil bath at 70-75 °C for 20 hours, cooled to room temperature, and concentrated in vacuo to remove ca. 500 mL of DMF. The remaining material was poured into 33% EtOAc/hexane, washed with water and brine, dried (Na2Sθ4), filtered, and concentrated in vacuo to provide the product as a brown oil. This material was purified by silica gel chromatography (25-50% EtOAc/hexane) to yield pure titled product.
Step J: Preparation of (±)-4-(r -butoxy carbonyl)- 1 -(3- chloropheny)-3-[ 1 -(1 -(4-cyanobenzyl)-5-imidazolyl)- 1 -
(hydroxy )methy 11 -2-piperazinone
To a solution of the piperazinone from Step I (98.0 mg, 0.316 mmol) in 1.5 mL of THF at -78 °C was added dropwise 1 M lithium hexamethyldisilazide solution (0.332 mL, 0.332 mmol). After 20 minutes, a solution of the aldehyde from Step E (76 mg, 0.360 mmol) in 0.5 mL of THF was added. After 30 min, the reaction was quenched with sat aq. NH4C1 soln., poured into EtOAc, and washed with sat. aq. NaHCθ3 soln., and brine. The solution was dried (Na2S04), filtered, and concentrated in vacuo to provide the cmde product. The product was purified by flash chromatography on silica gel (3-6% MeOH/CH2C12) to provide 60 mg of the titled product as a 9: 1 mixture of diastereomers.
Step K: Preparation of (±)- l -(3-chloropheny)-3-[ l -(l -(4- cyanobenzy l)-5-imidazolyl)- 1 -(hydroxy )methyl]-2- piperazinone hydrochloride
To a solution of the piperazinone from Step J (60 mg, 0.1 15 mmol) in 2 mL of dichloromethane at 0 °C was added trifluoroacetic acid (1.0 mL), and the reaction was allowed to warm to room temperature. After 6 hours, the solution was concentrated in vacuo, taken up in CH2C12, washed with dilute aq. NaHCθ3 soln., dried (Na2Sθ4), filtered, and concentrated in vacuo to provide the cmde product. After conversion to the HCI salt, the product was isolated (62 mg) as a 9: 1 mixture of diastereomers. FAB ms: 422 (M+l ). Anal. Calc for C22H20C1N5O2 • 2.10 HCI • 1.00 H20: C, 51.16; H, 4.70; O, 13.56. Found: C, 51.21 ; H, 4.70; O, 12.66.
EXAMPLE 8 ln vitro inhibition of ras famesyl transferase
Assays of famesyl-protein transferase. Partially purified bovine FPTase and Ras peptides (Ras-CVLS, Ras-CVIM and Ras-CAIL) were prepared as described by Schaber et aJ , J. Biol. Chem. 265 : 14701 - 14704 (1990), Pompliano, et al., Biochemistry 31 :3800 (1992) and Gibbs et ah, PNAS U.S.A. 86:6630-6634 (1989), respectively. Bovine FPTase was assayed in a volume of 100 μl containing 100 mM N-(2- hydroxy ethyl) piperazine-/V'-(2-ethane sulfonic acid) (HEPES), pH 7.4, 5 mM MgCl2, 5 mM dithiothreitol (DTT), 100 mM [3H]-farnesyl diphosphate ([3H]-FPP; 740 CBq/mmol, New England Nuclear), 650 nM Ras-CVLS and 10 μg/ml FPTase at 31°C for 60 min. Reactions were initiated with FPTase and stopped with 1 ml of 1.0 M HCL in ethanol. Precipitates were collected onto filter-mats using a TomTec Mach II cell harvestor, washed with 100% ethanol, dried and counted in an LKB β-plate counter. The assay was linear with respect to both substrates, FPTase levels and time; less than 10% of the [3H]-FPP was utilized during the reaction period. Purified compounds were dissolved in 100% dimethyl sulfoxide (DMSO) and were diluted 20-fold into the assay. Percentage inhibition is measured by the amount of incorporation of radioactivity in the presence of the test compound when compared to the amount of incorporation in the absence of the test compound.
Human FPTase was prepared as described by Omer et al., Biochemistry 32:5167-5176 (1993). Human FPTase activity was assayed as described above with the exception that 0.1 % (w/v) polyethylene glycol 20,000, 10 μM ZnCl2 and 100 ΠM Ras-CVIM were added to the reaction mixture. Reactions were performed for 30 min., stopped with 100 μl of 30% (v/v) trichloroacetic acid (TCA) in ethanol and processed as described above for the bovine enzyme.
The compounds of the instant invention described in the above Examples 1 and 5-7 were tested for inhibitory activity against human FPTase by the assay described above and were found to have IC50 of <50 μM. EXAMPLE 9
In vivo ras famesylation assay The cell line used in this assay is a v-ras line derived from either Ratl or NIH3T3 cells, which expressed viral Ha-ras p21. The assay is performed essentially as described in DeClue, J.E. et aL, Cancer Research 51 :712-717, (1991 ). Cells in 10 cm dishes at 50-75% confluency are treated with the test compound (final concentration of solvent, methanol or dimethyl sulfoxide, is 0.1 %). After 4 hours at 37°C, the cells are labelled in 3 ml methionine-free DMEM supple- meted with 10% regular DMEM, 2% fetal bovine serum and 400 mCif35s]methionine (1000 Ci/mmol). After an additional 20 hours, the cells are lysed in 1 ml lysis buffer (1 % NP40/20 mM HEPES, pH 7.5/5 mM MgCl2/lmM DTT/10 mg/ml aprotinen/2 mg/ml leupeptin/2 mg/ml antipain/0.5 mM PMSF) and the lysates cleared by centrifugation at 100,000 x g for 45 min. Aliquots of lysates containing equal numbers of acid-precipitable counts are bought to 1 ml with IP buffer (lysis buffer lacking DTT) and immunoprecipitated with the ras-specific monoclonal antibody Y 13-259 (Furth, M.E. et al, J. Virol. 43:294-304, (1982)). Following a 2 hour antibody incubation at 4°C, 200 ml of a 25% suspension of protein A-Sepharose coated with rabbit anti rat IgG is added for 45 min. The immunoprecipitates are washed four times with IP buffer (20 nM HEPES, pH 7.5/1 mM EDTA/1 % Triton X- 100.0.5% deoxycholate/0.1%/SDS/0.1 M 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 nonfamesylated ras proteins are compared to determine the percent inhibition of famesyl transfer to protein.
EXAMPLE 10
In vivo growth inhibition assay To determine the biological consequences of FPTase inhibition, the effect of the compounds of the instant invention on the anchorage-independent growth of Ratl cells transformed with either a v-ras, v-raf, or v-mos oncogene is tested. Cells transformed by v-Raf and v-Mos maybe included in the analysis to evaluate the specificity of instant compounds for Ras-induced cell transformation.
Rat 1 cells transformed with either v-ras, v-raf, or v-mos are seeded at a density of 1 x 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 se m) 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.

Claims

WHAT IS CLAIMED IS:
1. A compound which inhibits farnesyl-protein transferase of the formula A:
wherein:
R 1 a and R1 b are independently selected from:
a) hydrogen,
b) aryl, heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, C2- C6 alkynyl, R10O-, R 1 l S(O)m-, R10C(O)NR10-, CN(R10)2NC(O)-, R10 2N-C(NR 10)-, CN, NO2, R10C(O)-, N3, -N(R10)2, or R 1 1OC(O)NR 10-, c) unsubstituted or substituted C1 -C6 alkyl wherein the
substitutent on the substituted C1 -C6 alkyl is selected from unsubstituted or substituted aryl, heterocyclic, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, R10O-, R1 1S(O)m-, R10C(O)NR10-, (R10)2NC(OK R10 2N- C(NR 10)-, CN, R10C(O)-, N3, -N(R 10)2, and R 11OC(O)-
NR10-;
R2 and R4 are independently selected from: H; unsubstituted or
substituted C1 -8 alkyl, unsubstituted or substituted C2-8 alkenyl,
unsubstituted or substituted C2-8 alkynyl, unsubstituted or substituted aryl, unsubstituted or substituted heterocycle, wherein the substituted group is substituted with one or more of:
1 ) aryl or heterocycle, unsubstituted or substituted with a) C1 -4 alkyl,
b) (CH2)pOR6,
c) (CH2)pNR6R7,
d) halogen,
e) CN,
f) aryl or heteroaryl,
g) perfluoro-C1 -4 alkyl,
h) SR6a S(O)R6a, SO2R6a,
2) C3-6 cycloalkyl,
3) OR6,
4) SR6a, S(O)R6a, or SO2R6a,
R3 and R5 are selected from H and CH3;
R2 and R3 or R4 and R5 are attached to the same C atom and are combined to form - (CH2)u - wherein one of the carbon atoms is optionally replaced by a moiety selected from: O, S(O)m, -NC(O)-, and -N(COR10). ; R6, R7 and R7a are independently selected from: H; C1 -4 alkyl, C3-6 cycloalkyl, heterocycle, aryl, aroyl, heteroaroyl, arylsulfonyl, heteroarylsulfonyl, unsubstituted or substituted with:
a) C1 -4 alkoxy,
b) aryl or heterocycle,
c) halogen,
d) HO,
f) — S02R11 , or
g) N(R10)2; or
R6 and R7 may be joined in a ring;
R7 and R7a may be joined in a ring;
R6a is selected from: C1 -4 alkyl, C3-6 cycloalkyl, heterocycle, aryl, unsubstituted or substituted with:
a) C1 -4 alkoxy,
b) aryl or heterocycle,
c) halogen,
d) HO,
f) — SO2R11 , or
g) N(R10)2;
R8 is independently selected from:
a) hydrogen,
b) aryl, heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-
C6 alkynyl, perfluoroalkyl, F, Cl, Br, R 1 0O-, R 1 1 S(O)m- R10C(O)NR10-, (R10)2NC(O)-, R10 2N-C(NR10)-, CN, NO2 , R 10C(O)-, N3, -N(R10)2, or R 1 1OC(O)NR10-, and c) C1 -C6 alkyl unsubstituted or substituted by aryl,
cyanophenyl, heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, perfluoroalkyl, F, Cl, Br, R 10O-,
R1 1S(O)m-, R10C(O)NH-, (R10)2NC(O)-, R10 2N- C(NR10)-, CN, R10C(O)-, N3, -N(R10)2, or R10OC(O)NH-; R9 is selected from:
a) hydrogen,
b) alkenyl, alkynyl, perfluoroalkyl, F, Cl, Br, R10O-,
R1 1S(O)m-, R10C(O)NR10-, (R10)2NC(O)-, R10 2N- C(NR10)-, CN, NO2, R 1 0C(O)-, N3, -N(R10)2, or
R 11OC(O)NR10-, and
c) C1 -C6 alkyl unsubstituted or substituted by perfluoroalkyl, F, Cl, Br, R10O-, R1 1 S(O)m-, R 10C(O)NR10-, (R10)2NC(O)-, R10 2N-C(NR10)-, CN, R10C(O)-, N3, -N(R10)2, or R 1 1 OC(O)NR10-; R10 is independently selected from hydrogen, C 1-C6 alkyl, benzyl and aryl;
R1 1 is independently selected from C1 -C6 alkyl and aryl;
A 1 and A2 are independently selected from: a bond, -CH=CH-, -C≡C-,
-C(O)-, -C(O)NR10-, -NR10C(O)-, O, -N(R10)-,
-S(O)2N(R10)-, -N(R10)S(O)2-, or S(O)m; V is selected from:
a) hydrogen,
b) heterocycle,
c) aryl, d) C1 -C20 alkyl wherein from 0 to 4 carbon atoms are replaced with a a heteroatom selected from O, S, and N, and
e) C2-C20 alkenyl,
provided that V is not hydrogen if A1 is S(O)m and V is not hydrogen if A 1 is a bond, n is 0 and A2 is S(O)m;
W is a heterocycle; X is a bond, -CH2-, -C(=O)-, -C(O)NR6-, -NR6C(O)-, -NR6- or
-S(=O)m-;
Y is a bond, -CH2-, -NH-, -S(=O)m- or O; Z is selected from:
1) a unsubstituted or substituted group selected from aryl,
heteroaryl, arylmethyl, heteroarylmethyl, arylsulfonyl, heteroarylsulfonyl, wherein the substituted group is substituted with one or more of the following:
a) C1 -4 alkyl, unsubstituted or substituted with:
C1 -4 alkoxy, NR6R7, C3-6 cycloalkyl, aryl, heterocycle, HO, -S(O)mR6a, or -C(O)NR6R7, b) aryl or heterocycle,
c) halogen,
d) OR6,
e) NR6R7,
f) CN,
g) NO2,
h) CF3;
i) -S(O)mR6a,
j) -C(O)NR6R7, or
k) C3-C6 cycloalkyl; or
2) unsubstituted C1 -C6 alkyl, substituted C1 -C6 alkyl,
unsubstituted C3-C6 cycloalkyl or substituted C3-C6 cycloalkyl,wherein the substituted C1 -C6 alkyl and substituted C3-C6 cycloalkyl is substituted with one or two of the following:
a) C1 -4 alkoxy,
b) NR6R7,
c) C3-6 cycloalkyl,
d) -NR6C(O)R7,
e) HO,
f) -S(O)mR6a,
g) halogen, or
h) perfluoroalkyl; m is 0, 1 or 2;
n is 0, 1 , 2, 3 or 4;
p is 0, 1 , 2, 3 or 4;
q is 1 or 2;
r is 0 to 5, provided that r is 0 when V is hydrogen;
t is 0 or 1 ; and
u is 4 or 5; or a pharmaceutically acceptable salt thereof.
2. The compound according to Claim 1 of the formula A wherein Y is a bond, -CH2- or O.
3. The compound according to Claim 1 of the formula
wherein:
R 1 a is independently selected from: hydrogen or C1 -C6 alkyl;
R 1 b is independently selected from:
a) hydrogen,
b) aryl, heterocycle, cycloalkyl, R10O-, -N(R10)2 or C2-C6 alkenyl,
c) unsubstituted or substituted C1 -C6 alkyl wherein the
substitutent on the substituted C1 -C6 alkyl is selected from unsubstituted or substituted aryl, heterocycle, cycloalkyl, alkenyl, R10O- and -N(R10)2; R3 and R5 are independently selected from H and CH3;
R2 and R4 are independently selected from H; or C1 -5 alkyl, unbranched or branched, unsubstituted or substituted with one or more of:
1 ) aryl,
2) heterocycle,
3) OR6,
4) SR6a, SO2R6a, or
5)
and any two of R2, R3, R4 and R5 are optionally attached to the same carbon atom; R6, R7 and R7a are independently selected from:
H; C1 -4 alkyl, C3-6 cycloalkyl, aryl, heterocycle,
unsubstituted or substituted with:
a) C1 -4 alkoxy,
b) halogen, or
c) aryl or heterocycle;
R6a is selected from:
C1 -4 alkyl or C3-6 cycloalkyl,
unsubstituted or substituted with:
a) C1 -4 alkoxy,
b) halogen, or
c) aryl or heterocycle;
R8 is independently selected from:
a) hydrogen,
b) C1 -C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1 -C6
perfluoroalkyl, F, Cl, R10O-, R10C(O)NR10-, CN, NO2, (R10)2N-C(NR10)-, R10C(O)-, -N(R 10)2, or R 11OC(O)NR10-, and
c) C1 -C6 alkyl substituted by C1 -C6 perfluoroalkyl, R10O-, R10C(O)NR10-, (R10)2N-C(NR10)-, R10C(O)-, -N(R10)2, or R 11OC(O)NR10-;
R9 is selected from:
a) hydrogen,
b) C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 perfluoroalkyl, F, Cl, R10O-, R1 1S(O)m-, R10C(O)NR10-, CN, NO2,
(R10)2N-C(NR10)-, R10C(O)-, -N(R10)2, or R 11OC(O)NR10-, and
c) C1 -C6 alkyl unsubstituted or substituted by C1 -C6
perfluoroalkyl, F, Cl, R10O-, R 1 1 S(O)m-, R10C(O)NR10-, CN, (R10)2N-C(NR10)-, R10C(O)-, -N(R 10)2, or
R 11OC(O)NR10-; R10 is independently selected from hydrogen, C1 -C6 alkyl, benzyl and aryl; R11 is independently selected from C1 -C6 alkyl and aryl;
A 1 and A2 are independently selected from: a bond, -CH=CH-, -C≡C-,
-C(O)-, -C(O)NR10-, O, -N(R10)-, or S(O)m;
V is selected from:
a) hydrogen,
b) heterocycle selected from pyrrolidinyl, imidazolyl,
pyridinyl, thiazolyl, pyridonyl, 2-oxopiperidinyl, indolyl, quinolinyl, isoquinolinyl, and thienyl,
c) aryl,
d) C1 -C20 alkyl wherein from 0 to 4 carbon atoms are
replaced with a a heteroatom selected from O, S, and N, and
e) C2-C20 alkenyl, and
provided that V is not hydrogen if A 1 is S(O)m and V is not hydrogen if A 1 is a bond, n is 0 and A2 is S(O)m; W is a heterocycle selected from pyrrolidinyl, imidazolyl, pyridinyl, thiazolyl, pyridonyl, 2-oxopiperidinyl, indolyl, quinolinyl, or
isoquinolinyl;
X is -CH2- or -C(=O)-; Y is a bond or -CH2-; Z is selected from: 1) a unsubstituted or substituted group selected from aryl, heteroaryl, arylmethyl, heteroarylmethyl, arylsulfonyl, heteroarylsulfonyl, wherein the substituted group is substituted with one or more of the following:
a) C1 -4 alkyl, unsubstituted or substituted with:
C1 -4 alkoxy, NR6R7, C3-6 cycloalkyl, aryl, heterocycle, HO, -S(O)mR6a, or -C(O)NR6R7, b) aryl or heterocycle,
c) halogen,
d) OR6 '
e) NR6R7,
f) CN,
g) NO2,
h) CF3;
i) -S(O)mR6a,
j) -C(O)NR6R7, or
k) C3-C6 cycloalkyl; or
2) unsubstituted C1 -C6 alkyl, substituted C1 -C6 alkyl,
unsubstituted C3-C6 cycloalkyl or substituted C3-C6 cycloalkyl,wherein the substituted C1 -C6 alkyl and substituted C3-C6 cycloalkyl is substituted with one or two of the following:
a) C1 -4 alkoxy,
b) NR6R7,
c) C3-6 cycloalkyl,
d) -NR6C(O)R7,
e) HO,
f) -S(O)mR6a,
g) halogen, or
h) perfluoroalkyl; m is 0, 1 or 2;
n is 0, 1, 2, 3 or 4;
p is 0, 1 , 2, 3 or 4; r is 0 to 5, provided that r is 0 when V is hydrogen;
t is 0 or 1 ; and
u is 4 or 5; or a pharmaceutically acceptable salt thereof.
4. The compound according to Claim 1 of the formula B:
wherein:
R 1 a is selected from: hydrogen or C1 -C6 alkyl; R1 b is independently selected from:
a) hydrogen,
b) aryl, heterocycle, cycloalkyl, R10O-, -N(R10)2 or C2-C6 alkenyl,
c) C1 -C6 alkyl unsubstituted or substituted by aryl,
heterocycle, cycloalkyl, alkenyl, R10O-, or -N(R 10)2;
R3 and R5 are independently selected from H and CH3;
6 7
R2 and R4 are independently selected from H; or C1 -5 alkyl, unbranched or branched, unsubstituted or substituted with one or more of:
1 ) aryl,
2) heterocycle, 3) OR6,
4) SR6a, SO2R6a, or
5)
and any two of R2, R3, R4, and R5 are optionally attached to the same carbon atom;
R6 and R7 are independently selected from:
a) hydrogen,
b) C1 -C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1 -C6
perfluoroalkyl, F, Cl, R10O-, R10C(O)NR10-, CN, NO2,
(R10)2N-C(NR10)-, R10C(O)-, R10OC(O)-, -N(R10)2, or R 1 1OC(O)NR10-, and
c) C1 -C6 alkyl substituted by C1 -C6 perfluoroalkyl, R10O-, R10C(O)NR10-, (R10)2N-C(NR10)-, R10C(O)-,
R10OC(O)-, -N(R10)2, or R 11OC(O)NR10-;
R6a is selected from:
C1 -4 alkyl or C3-6 cycloalkyl,
unsubstituted or substituted with:
a) C1 -4 alkoxy,
b) halogen, or
c) aryl or heterocycle;
R8 is independently selected from:
a) hydrogen,
b) C1 -C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1 -C6
perfluoroalkyl, F, Cl, R10O-, R10C(O)NR10., CN, NO2, (R10)2N-C(NR10)-, R10C(O)-, -N(R10)2, or R 11OC(O)NR10-, and
c) C1 -C6 alkyl substituted by C1 -C6 perfluoroalkyl, R10O-, R10C(O)NR10-, (R10)2N-C(NR10)-, R10C(O)-,
-N(R 10)2, or R 11OC(O)NR10-; R9a is hydrogen or methyl; R10 is independently selected from hydrogen, C1 -C6 alkyl, benzyl and aryl; R11 is independently selected from C1 -C6 alkyl and aryl;
A1 and A2 are independently selected from: a bond, -CH=CH-, -C≡C-,
-C(O)-, -C(O)NR10-, O, -N(R10)-, or S(O)m;
V is selected from:
a) hydrogen,
b) heterocycle selected from pyrrolidinyl, imidazolyl,
pyridinyl, thiazolyl, pyridonyl, 2-oxopiperidinyl, indolyl, quinolinyl, isoquinolinyl, and thienyl,
c) aryl,
d) C1 -C20 alkyl wherein from 0 to 4 carbon atoms are
replaced with a a heteroatom selected from O, S, and N, and
e) C2-C20 alkenyl, and
provided that V is not hydrogen if A1 is S(O)m and V is not hydrogen if A1 is a bond, n is 0 and A2 is S(O)m;
X is -CH2- or -C(=O)-;
Y is a bond, -CH2-, -NH-, -S(=O)m- or O;
Z is selected from:
1) a unsubstituted or substituted group selected from aryl,
heteroaryl, arylmethyl, heteroarylmethyl, arylsulfonyl, heteroarylsulfonyl, wherein the substituted group is substituted with one or more of the following:
a) C1 -4 alkyl, unsubstituted or substituted with: C1 -4 alkoxy, NR6R7, C3-6 cycloalkyl, aryl, heterocycle, HO, -S(O)mR6a, or -C(O)NR6R7, b) aryl or heterocycle,
c) halogen,
d) OR6,
e) NR6R7,
f) CN,
g) NO2,
h) CF3;
i) -S(O)mR6a,
j) -C(O)NR6R7, or
k) C3-C6 cycloalkyl; or
2) unsubstituted C1 -C6 alkyl, substituted C1 -C6 alkyl,
unsubstituted C3-C6 cycloalkyl or substituted C3-C6 cycloalkyl,wherein the substituted C1 -C6 alkyl and substituted C3-C6 cycloalkyl is substituted with one or two of the following:
a) C1 -4 alkoxy,
b) NR6R7,
c) C3-6 cycloalkyl,
d) -NR6C(O)R7,
e) HO,
f) -S(O)mR6a,
g) halogen, or
h) perfluoroalkyl; m is 0, 1 or 2;
n is 0, 1 , 2, 3 or 4;
p is 0, 1 , 2, 3 or 4; and
r is 0 to 5, provided that r is 0 when V is hydrogen; or a pharmaceutically acceptable salt thereof.
5. The compound according to Claim 1 of the formula
C:
wherein:
R 1 b is independently selected from:
a) hydrogen,
b) aryl, heterocycle, cycloalkyl, R10O-, -N(R 10)2 or C2-C6 alkenyl,
c) C1 -C6 alkyl unsubstituted or substituted by aryl,
heterocycle, cycloalkyl, alkenyl, R10O-, or -N(R10)2;
R3 and R5 are independently selected from H and CH3;
R2 and R4 are independently selected from H; or C1 -5 alkyl, unbranched or branched, unsubstituted or substituted with one or more of:
1 ) aryl,
2) heterocycle,
3) OR6,
4) SR6a, SO2R6a, or
5)
and any two of R2, R3, R4, and R5 are optionally attached to the same carbon atom; R6 and R7 are independently selected from:
a) hydrogen,
b) C1 -C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1 -C6
perfluoroalkyl, F, Cl, R10O-, R10C(O)NR10-, CN, NO2, (R10)2N-C(NR10)-, R10C(O)-, R10OC(O)-, -N(R10)2, or R 11OC(O)NR10-, and
c) C1 -C6 alkyl substituted by C1 -C6 perfluoroalkyl, R10O-, R10C(O)NR10-, (R10)2N-C(NR10)-, R10C(O)-,
R10OC(O)-, -N(R10)2, or R 11OC(O)NR10-;
R6a is selected from:
C1 -4 alkyl or C3-6 cycloalkyl,
unsubstituted or substituted with:
a) C1 -4 alkoxy,
b) halogen, or
c) aryl or heterocycle;
R8 is independently selected from:
a) hydrogen,
b) C1 -C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1 -C6
perfluoroalkyl, F, Cl, R10O-, R10C(O)NR10-, CN, NO2, (R10)2N-C(NR10)-, R10C(O)-, -N(R10)2, or R 11OC(O)NR10-, and
c) C1 -C6 alkyl substituted by C1 -C6 perfluoroalkyl, R10O-, R10C(O)NR10-, (R10)2N-C(NR10)-, R10C(O)-,
-N(R10)2, or R 11OC(O)NR10-; R10 is independently selected from hydrogen, C1 -C6 alkyl, benzyl and aryl; R11 is independently selected from C1 -C6 alkyl and aryl; X is -CH2- or -C(=O)-; Y is a bond, -CH2-, -NH-, -S(=O)m- or O;
Z is selected from:
1) a unsubstituted or substituted group selected from aryl,
heteroaryl, arylmethyl, heteroarylmethyl, arylsulfonyl, heteroarylsulfonyl, wherein the substituted group is substituted with one or more of the following:
a) C1 -4 alkyl, unsubstituted or substituted with:
C1 -4 alkoxy, NR6R7, C3-6 cycloalkyl, aryl, heterocycle, HO, -S(O)mR6a, or -C(O)NR6R7, b) aryl or heterocycle,
c) halogen,
d) OR6 '
e) NR6R7>
f) CN,
g) NO2,
h) CF3;
i) -S(O)mR6a,
j) -C(O)NR6R7, or
k) C3-C6 cycloalkyl; or
2) unsubstituted C1 -C6 alkyl, substituted C1 -C6 alkyl,
unsubstituted C3-C6 cycloalkyl or substituted C3-C6 cycloalkyl, wherein the substituted C1 -C6 alkyl and substituted C3-C6 cycloalkyl is substituted with one or two of the following:
a) C1 -4 alkoxy,
b) NR6R7,
c) C3-6 cycloalkyl,
d) -NR6C(O)R7,
e) HO,
f) -S(O)mR6a,
g) halogen, or
h) perfluoroalkyl; m is 0, 1 or 2; and
p is 0, 1 , 2, 3 or 4; or a pharmaceutically acceptable salt thereof.
6. The compound according to Claim 1 of the formula
D:
wherein:
R 1 b is independently selected from:
a) hydrogen,
b) aryl, heterocycle, cycloalkyl, R 10O-, -N(R 10)2 or C2-C6 alkenyl,
c) C1-C6 alkyl unsubstituted or substituted by aryl,
heterocycle, cycloalkyl, alkenyl, R 10O-, or -N(R 10)2;
R2 and R4 are independently selected from: hydrogen or C1 -C6 alkyl; R3 and R5 are hydrogen;
R6 and R7 are independently selected from:
a) hydrogen,
b) C1 -C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1 -C6
perfluoroalkyl, F, Cl, R10O-, R10C(O)NR10-, CN, NO2,
(R 10)2N-C(NR 10)-, R10C(O)-, R10OC(O)-, -N(R10)2, or R 11OC(O)NR10-, and c) C1-C6 alkyl substituted by C1 -C6 perfluoroalkyl, R10O-, R1 0C(O)NR 10-, (R10)2N-C(NR10)-, R 10C(O)-,
R10OC(O)-, -N(R10)2, or R1 1OC(O)NR10-; R10 is independently selected from hydrogen, C1-C6 alkyl, benzyl and aryl;
R 1 1 is independently selected from C1 -C6 alkyl and aryl; X is -CH2- or -C(=O)-;
Y is a bond, -CH2-, -NH-, -S(=O)m- or O;
Z is mono- or bicyclic aryl, mono- or bicyclic heteroaryl, mono- or bicyclic arylmethyl, mono- or bicyclic heteroarylmethyl, mono- or bicyclic arylsulfonyl, mono- or bicyclic heteroarylsulfonyl, unsubstituted or substituted with one or two of the following:
1 ) C1 -4 alkyl, unsubstituted or substituted with:
a) C1 -4 alkoxy,
b) NR6R7,
c) C3-6 cycloalkyl,
d) aryl or heterocycle,
e) HO,
f) -S(O)mR6, or
g) -C(O)NR6R7,
2) aryl or heterocycle,
3) halogen,
4) OR6,
5) NR6R7,
6) CN,
7) NO2,
8) CF3;
9) -S(O)mR6 10) -C(O)NR6R7, or
11) C3-C6 cycloalkyl; m is 0, 1 or 2; and
pis 0, 1, 2, 3 or 4; or the pharmaceutically acceptable salts thereof.
7. A compound which inhibits farnesyl-protein transferase which is:
N-[2-{1-(4-Cyanobenzyl)-5-imidazolyl}ethyl]-3-carbamoyl-1-phenyl-2- piperidinone 3-[3-{1-(4-Cyanobenzyl)-5-imidazolyI}-1-propyl]-1-phenyl-2- piperidinone
3-[3-{1-(4-Cyanobenzyl)-5-imidazolyl}-1-propyl]-1-phenyl-2- pyrrolidinone
(±)cis—3-[3-{1-(4-Cyanobenzyl)-5-imidazolyl}-1-propyl]-4- methoxymethyl-1-phenyl-2-pyrrolidinone
(±)trans-3-[3-{1-(4-Cyanobenzyl)-5-imidazolyl}-1-propyl]-4- methoxymethyl-1-phenyl-2-pyrrolidinone
3-[ 2-{5-(4-Cyanobenzyl)-1-imidazolyl}-1-ethyl]-1-phenyl-2- pyrrolidinone 1-Benzyl-3-[2-{5-(4-cyanobenzyl)-1-imidazolyl}-1-ethyl]-2- pyrrolidinone or
(±)-1-(3-chloropheny)-3-[1-(1-(4-cyanobenzyl)- 5-imidazolyl)-1-(hydroxy)methyl]-2-piperazinone or a pharmaceutically acceptable salt or optical isomer thereof.
8. The compound according to Claim 7 which is: N-[2- { 1 -(4-Cyanobenzyl)-5-imidazolyl }ethyl]-3-carbamoyl-1-pheny1-2- piperidinone
or a pharmaceutically acceptable salt or optical isomer thereof.
9. The compound according to Claim 7 which is:
3-[2-{ 5-(4-Cyanobenzyl)-1 -imidazolyl } -1 -ethyl]-1-phenyl-2- pyrrolidinone
or a pharmaceutically acceptable salt or optical isomer thereof.
10. The compound according to Claim 7 which is:
1-Benzyl-3-[2-{5-(4-cyanobenzyl)-1-imidazolyl}-1-ethyl] -2- pyrrolidinone
or a pharmaceutically acceptable salt or optical isomer thereof.
1 1. A pharmaceutical composition comprising a pharmaceutical carrier, and dispersed therein, a therapeutically effective amount of a compound of Claim 1.
12. A pharmaceutical composition comprising a pharmaceutical carrier, and dispersed therein, a therapeutically effective amount of a compound of Claim 3.
13. A pharmaceutical composition comprising a pharmaceutical carrier, and dispersed therein, a therapeutically effective amount of a compound of Claim 4.
14. A pharmaceutical composition comprising a pharmaceutical carrier, and dispersed therein, a therapeutically effective amount of a compound of Claim 7.
15. A method for inhibiting farnesyl-protein transferase which comprises administering to a mammal in need thereof a
therapeutically effective amount of a composition of Claim 11.
16. A method for inhibiting farnesyl-protein transferase which comprises administering to a mammal in need thereof a
therapeutically effective amount of a composition of Claim 12.
17. A method for inhibiting farnesyl-protein transferase which comprises administering to a mammal in need thereof a
therapeutically effective amount of a composition of Claim 13.
18. A method for inhibiting farnesyl-protein transferase which comprises administering to a mammal in need thereof a
therapeutically effective amount of a composition of Claim 14.
19. A method for treating cancer which comprises administering to a mammal in need thereof a therapeutically effective amount of a composition of Claim 1 1.
20. A method for treating cancer which comprises administering to a mammal in need thereof a therapeutically effective amount of a composition of Claim 12.
21. A method for treating cancer which comprises administering to a mammal in need thereof a therapeutically effective amount of a composition of Claim 13.
22. A method for treating cancer which comprises administering to a mammal in need thereof a therapeutically effective amount of a composition of Claim 14.
23. A method for treating neurofibromin benign proliferative disorder which comprises administering to a mammal in need thereof a therapeutically effective amount of a composition of Claim 1 1.
24. A method for treating blindness related to retinal vascularization which comprises administering to a mammal in need thereof a therapeutically effective amount of a composition of Claim 1 1.
25. A method for treating infections from hepatitis delta and related viruses which comprises administering to a mammal in need thereof a therapeutically effective amount of a composition of Claim 10.
26. A method for preventing restenosis which comprises administering to a mammal in need thereof a therapeutically effective amount of a composition of Claim 1 1.
27. A method for treating polycystic kidney disease which comprises administering to a mammal in need thereof a
therapeutically effective amount of a composition of Claim 1 1.
28. A pharmaceutical composition made by combining the compound of Claim 1 and a pharmaceutically acceptable carrier.
29. A process for making a pharmaceutical composition comprising combining a compound of Claim 1 and a pharmaceutically acceptable carrier.
EP97917738A 1996-04-03 1997-03-27 Inhibitors of farnesyl-protein transferase Withdrawn EP0891360A4 (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US1479896P 1996-04-03 1996-04-03
US14798P 1996-04-03
GB9609666 1996-05-09
GBGB9609666.4A GB9609666D0 (en) 1996-05-09 1996-05-09 Inhibitors of farnesyl-protein transferase
PCT/US1997/005235 WO1997036900A1 (en) 1996-04-03 1997-03-27 Inhibitors of farnesyl-protein transferase

Publications (2)

Publication Number Publication Date
EP0891360A1 true EP0891360A1 (en) 1999-01-20
EP0891360A4 EP0891360A4 (en) 2002-05-15

Family

ID=26309289

Family Applications (1)

Application Number Title Priority Date Filing Date
EP97917738A Withdrawn EP0891360A4 (en) 1996-04-03 1997-03-27 Inhibitors of farnesyl-protein transferase

Country Status (5)

Country Link
EP (1) EP0891360A4 (en)
JP (1) JP2000507585A (en)
AU (1) AU715603B2 (en)
CA (1) CA2250353A1 (en)
WO (1) WO1997036900A1 (en)

Families Citing this family (26)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5972966A (en) * 1996-12-05 1999-10-26 Merck & Co., Inc. Inhibitors of farnesyl-protein transferase
EP1035849A4 (en) * 1997-12-04 2001-09-12 Merck & Co Inc Inhibitors of farnesyl-protein transferase
EP1058683B1 (en) * 1998-02-02 2002-12-04 Lg Chemical Limited Farnesyl transferase inhibitors having a piperidine structure and process for preparation thereof
KR100388790B1 (en) * 1998-03-06 2003-10-04 주식회사 엘지생명과학 Pyridin-2-one derivative useful as farnesyl transferase inhibitor
HRP990246A2 (en) 1998-08-07 2000-06-30 Du Pont Pharm Co Succinoylamino benzodiazepines as inhibitors of a beta protein production
NZ525513A (en) 1998-08-07 2004-09-24 Pont Pharmaceuticals Du Succinoylamino lactams as inhibitors of Abeta protein production
BR9917082A (en) 1998-12-24 2001-11-06 Du Pont Pharm Co Compounds that inhibit the production of "beta" proteins, a method of treating neurological disorders associated with the production of "beta" -amyloid, pharmaceutical composition and, method of inhibiting y-secretase activity
AU4181000A (en) * 1999-04-02 2000-10-23 Du Pont Pharmaceuticals Company Novel lactam inhibitors of matrix metalloproteinases, tnf-alpha, and aggrecanase
US6503902B2 (en) 1999-09-13 2003-01-07 Bristol-Myers Squibb Pharma Company Hydroxyalkanoylaminolactams and related structures as inhibitors of a β protein production
US6960576B2 (en) 1999-09-13 2005-11-01 Bristol-Myers Squibb Pharma Company Hydroxyalkanoylaminolactams and related structures as inhibitors of Aβ protein production
CA2387493A1 (en) 1999-10-08 2001-04-19 Lorin Andrew Thompson Amino lactam sulfonamides as inhibitors of a.beta. protein production
AU2001239791A1 (en) 2000-02-17 2001-08-27 Du Pont Pharmaceuticals Company Succinoylamino carbocycles and heterocycles as inhibitors of abeta protein production
US6495540B2 (en) 2000-03-28 2002-12-17 Bristol - Myers Squibb Pharma Company Lactams as inhibitors of A-β protein production
EP1268433A1 (en) 2000-04-03 2003-01-02 Bristol-Myers Squibb Pharma Company Cyclic lactams as inhibitors of a-beta-protein production
US6713476B2 (en) 2000-04-03 2004-03-30 Dupont Pharmaceuticals Company Substituted cycloalkyls as inhibitors of a beta protein production
EP1289966A1 (en) 2000-04-11 2003-03-12 Bristol-Myers Squibb Pharma Company SUBSTITUTED LACTAMS AS INHIBITORS OF A$g(b) PROTEIN PRODUCTION
BR0106717A (en) 2000-06-01 2002-04-16 Bristol Myers Squibb Pharma Co Compounds, pharmaceutical composition and uses of innovative lactam compounds
US6710058B2 (en) 2000-11-06 2004-03-23 Bristol-Myers Squibb Pharma Company Monocyclic or bicyclic carbocycles and heterocycles as factor Xa inhibitors
US6974869B2 (en) 2001-09-18 2005-12-13 Bristol-Myers Squibb Pharma Company Piperizinones as modulators of chemokine receptor activity
TW200307667A (en) 2002-05-06 2003-12-16 Bristol Myers Squibb Co Sulfonylaminovalerolactams and derivatives thereof as factor Xa inhibitors
US7846963B2 (en) 2003-05-17 2010-12-07 Korea Research Institute Of Bioscience And Biotechnology 2-oxo-heterocyclic compounds and the pharmaceutical compositions comprising the same
JP4914719B2 (en) * 2003-05-17 2012-04-11 コリア リサーチ インスティテュート オブ バイオサイエンス アンド バイオテクノロジー Novel 2-oxoheterocyclic compound and pharmaceutical composition containing the same
JP4799408B2 (en) * 2003-07-15 2011-10-26 コリア リサーチ インスティテュート オブ バイオサイエンス アンド バイオテクノロジー Use of novel 2-oxoheterocyclic compounds and pharmaceutical compositions containing them
US20070123508A1 (en) * 2005-05-27 2007-05-31 Roger Olsson PAR2-modulating compounds and their use
CN102548986A (en) 2009-06-05 2012-07-04 链接医药公司 Aminopyrrolidinone derivatives and uses thereof
WO2015130958A1 (en) * 2014-02-26 2015-09-03 Howard University 5-oxopyrrolidine derivatives as hiv integrase inhibitors

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0236940A2 (en) * 1986-03-07 1987-09-16 Ciba-Geigy Ag Alpha-heterocycle substituted tolunitriles
WO1993012108A1 (en) * 1991-12-18 1993-06-24 Schering Corporation Imidazolyl or imidazoylalkyl substituted with a four or five membered nitrogen containing heterocyclic ring
WO1993012107A1 (en) * 1991-12-18 1993-06-24 Schering Corporation Imidazoylalkyl substituted with a six membered nitrogen containing heterocyclic ring

Family Cites Families (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
IL69407A (en) * 1982-08-23 1987-10-20 Warner Lambert Co 2(1h)-pyridinones,their preparation and pharmaceutical compositions containing them
DE3336024A1 (en) * 1983-10-04 1985-04-18 Boehringer Ingelheim KG, 6507 Ingelheim 4-AMINO-L-BENZYL-PYRROLIDINONE AND ITS ACID ADDITION SALTS, METHOD FOR THEIR PRODUCTION AND MEDICINAL PRODUCTS
DE3433953A1 (en) * 1984-09-15 1986-03-27 A. Nattermann & Cie GmbH, 5000 Köln NEW, 3,4-DIHYDRO-2 (1H) -PYRIDONE AND 2 (1H) -PYRIDONE, INCLUDING AN IMIDAZOLE GROUP, METHOD FOR THE PRODUCTION THEREOF AND MEDICINAL PRODUCTS CONTAINING THE SAME
US5015653A (en) * 1989-07-10 1991-05-14 Hoffmann-La Roche Inc. [(5-oxo)-2-pyrrolidinyl)methyl]cyclohexaneacetamides, compositions and use
US5055467A (en) * 1989-11-13 1991-10-08 Allergan, Inc. Pharmaceutical epinephrine-pilocarpine compounds
US5264449A (en) * 1989-11-13 1993-11-23 Allergan, Inc. N-substituted derivatives of 3R,4R-ethyl-[(1-methyl-1H-imidazol-5-yl)methyl]-2-pyrrolidinone
TW218017B (en) * 1992-04-28 1993-12-21 Takeda Pharm Industry Co Ltd

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0236940A2 (en) * 1986-03-07 1987-09-16 Ciba-Geigy Ag Alpha-heterocycle substituted tolunitriles
WO1993012108A1 (en) * 1991-12-18 1993-06-24 Schering Corporation Imidazolyl or imidazoylalkyl substituted with a four or five membered nitrogen containing heterocyclic ring
WO1993012107A1 (en) * 1991-12-18 1993-06-24 Schering Corporation Imidazoylalkyl substituted with a six membered nitrogen containing heterocyclic ring

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See also references of WO9736900A1 *

Also Published As

Publication number Publication date
WO1997036900A1 (en) 1997-10-09
AU715603B2 (en) 2000-02-03
EP0891360A4 (en) 2002-05-15
CA2250353A1 (en) 1997-10-09
AU2598897A (en) 1997-10-22
JP2000507585A (en) 2000-06-20

Similar Documents

Publication Publication Date Title
US5919785A (en) Inhibitors of farnesyl-protein transferase
AU716338B2 (en) Inhibitors of farnesyl-protein transferase
US5854265A (en) Biheteroaryl inhibitors of farnesyl-protein transferase
US6066738A (en) Inhibitors of farnesyl-protein transferase
AU715603B2 (en) Inhibitors of farnesyl-protein transferase
US5968965A (en) Inhibitors of farnesyl-protein transferase
US5869682A (en) Inhibitors of farnesyl-protein transferase
US5859035A (en) Arylheteroaryl inhibitors of farnesyl-protein transferase
US5874452A (en) Biheteroaryl inhibitors of farnesyl-protein transferase
US5885995A (en) Inhibitors of farnesyl-protein transferase
EP0891356A1 (en) Inhibitors of farnesyl-protein transferase
US5925651A (en) Inhibitors of farnesyl-protein transferase
EP0897303A1 (en) Inhibitors of farnesyl-protein transferase
AU715667B2 (en) Inhibitors of farnesyl-protein transferase
US5780492A (en) Inhibitors of farnesyl-protein transferase
EP0891357A1 (en) Inhibitors of farnesyl-protein transferase
WO1997036585A1 (en) Inhibitors of farnesyl-protein transferase
EP0891343A1 (en) Inhibitors of farnesyl-protein transferase
AU707416B2 (en) Inhibitors of farnesyl-protein transferase
AU2660797A (en) Inhibitors of farnesyl-protein transferase
EP0921801A1 (en) Inhibitors of farnesyl-protein transferase
WO1997036892A1 (en) Inhibitors of farnesyl-protein transferase
US6001835A (en) Inhibitors of farnesyl-protein transferase
EP0935464A1 (en) Inhibitors of farnesyl-protein transferase
EP0900081A1 (en) Inhibitors of farnesyl-protein transferase

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 19981103

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AT BE CH DE DK ES FI FR GB GR IE IT LI LU NL PT SE

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

A4 Supplementary search report drawn up and despatched

Effective date: 20020328

AK Designated contracting states

Kind code of ref document: A4

Designated state(s): AT BE CH DE DK ES FI FR GB GR IE IT LI LU NL PT SE

RIC1 Information provided on ipc code assigned before grant

Free format text: 7C 07D 417/00 A, 7C 07D 403/02 B, 7A 61K 31/44 B, 7A 61K 31/415 B, 7C 07D 401/12 B, 7C 07D 403/06 B, 7A 61K 31/50 B, 7C 07D 401/12 J, 7C 07D 233:00 J, 7C 07D 211:00 J, 7C 07D 403/06 K, 7C 07D 233:00 K, 7C 07D 207:00 K, 7C 07D 403/06 L, 7C 07D 241:00 L, 7C 07D 233:00 L

18D Application deemed to be withdrawn

Effective date: 20011002