EP0862435A1 - Inhibitoren der farnesyl-protein-transferase - Google Patents

Inhibitoren der farnesyl-protein-transferase

Info

Publication number
EP0862435A1
EP0862435A1 EP96942798A EP96942798A EP0862435A1 EP 0862435 A1 EP0862435 A1 EP 0862435A1 EP 96942798 A EP96942798 A EP 96942798A EP 96942798 A EP96942798 A EP 96942798A EP 0862435 A1 EP0862435 A1 EP 0862435A1
Authority
EP
European Patent Office
Prior art keywords
substituted
piperidine
cyanobenzyl
imidazol
ylethyl
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Withdrawn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
EP96942798A
Other languages
English (en)
French (fr)
Other versions
EP0862435A4 (de
Inventor
Byeong M. Kim
Anthony W. Shaw
Samuel L. Graham
S. Jane Desolms
Terrence M. Ciccarone
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 GBGB9604311.2A external-priority patent/GB9604311D0/en
Application filed by Merck and Co Inc filed Critical Merck and Co Inc
Publication of EP0862435A1 publication Critical patent/EP0862435A1/de
Publication of EP0862435A4 publication Critical patent/EP0862435A4/de
Withdrawn legal-status Critical Current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
    • C07D233/00Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings
    • C07D233/04Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings having one double bond between ring members or between a ring member and a non-ring member
    • C07D233/20Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings having one double bond between ring members or between a ring member and a non-ring member with substituted hydrocarbon radicals, directly attached to ring carbon atoms
    • C07D233/24Radicals substituted by nitrogen atoms not forming part of a nitro radical

Definitions

  • Ras protein is 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. Willumsen, Ann. Rev. Biochem. (52:851 - 891 (1993)).
  • ras genes are found in many human cancers, including colorectal carcinoma, exocrine pancreatic carcinoma, and myeloid leukemias.
  • the protein products of these genes are defective in their GTPase activity and constitutively transmit a growth stimulatory signal.
  • 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 570:583-586 (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 C ⁇ s or C20 isoprenoid, respectively.
  • the Ras protein is one of several proteins that are known to undergo post- translational famesylation.
  • Other famesylated 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 famesylated.
  • James, et al. have also suggested that there are famesylated proteins of unknown structure 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, 87:7541-7545 (1990)).
  • Inhibition of famesyl pyrophosphate biosynthesis by inhibiting HMG-CoA reductase blocks Ras membrane localization in cultured cells.
  • 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 alternate 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, 260:1934-1937 (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- 112930). It has also 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). It is, therefore, an object of this invention to develop novel 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 piperidine, 1 ,4-dihydropyridine and 1,2,3,4-tetrahydropyridine compounds which inhibit the famesyl-protein transferase. Furthermore, these compounds differ from such heterocyclic compounds previously described as inhibitors of famesyl-protein transferase with respect to the position of substituents about the nitrogen containing ring. Further contained in this invention are chemotherapeutic compositions containing these famesyl transferase inhibitors and methods for their production.
  • 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:
  • Rla, Rib anc j Rlc are independently selected from: a) hydrogen, b) unsubstituted or substituted aryl, unsubstituted or substituted heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, R 0-, R9s(0) m -, R8c(0)NR8-, CN, NO2, (R8)2N-C(NR8)-, R8C(0)-, R80C(0)-, N3, -N(R8)2, or R9 ⁇ C(0)NR8-, c) C1-C alkyl unsubstituted or substituted by unsubstituted or substituted aryl, heterocyclic, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, R80-, R9S(0) m -, R8C(0)NR -, CN, (R8)2N-C(NR8)
  • R2 is selected from: H; unsubstituted or substituted Cl-8 alkyl, unsubstituted or substituted C2-8 alkenyl, unsubstituted or substituted aryl,
  • R3 is selected from: H; ° O
  • R4 is independently selected from: a) hydrogen, b) unsubstituted or substituted aryl, unsubstituted or substituted heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, perfluoroalkyl, F, CI, Br, R 0-, R9S(0) m -
  • R5 is independently selected from: a) hydrogen, b) C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, perfluoroalkyl, F, CI, Br, R80-, R9s(0) m -, R8C(0)NR8-, CN, N ⁇ 2, (R8)2N-C-(NR8)-, R8C(0)-, R8 ⁇ C(0)-, N3, -N(R8)2, or R9 ⁇ C(0)NR8-, and c) C1-C alkyl, unsubstituted or substituted by perfluoroalkyl,
  • R6, R7 and R 7a are independently selected from: H; Cl -4 alkyl, C3-6 cycloalkyl, heterocycle, aryl, Cl-4 perfluoroalkyl, unsubstituted or substituted with one or two substituents selected from: a) Cl-4 alkoxy, b) substituted or unsubstituted aryl or substituted or unsubstituted heterocycle,
  • R6 and R 7 may be joined in a ring; R7 and R 7a may be joined in a ring;
  • R is independently selected from hydrogen, Cl-C alkyl, benzyl, 2,2,2-trifluoroethyl and aryl;
  • R9 is independently selected from C1-C6 alkyl and aryl
  • RlO is selected from: H; R c(0)-; R9s(0) m -; unsubstituted or substituted Cl-4 alkyl, unsubstituted or substituted C3-6 cycloalkyl, unsubstituted or substituted heterocycle, unsubstituted or substituted aryl, substituted aroyl, unsubstituted or substituted heteroaroyl, substituted arylsulfonyl, unsubstituted or substituted heteroarylsulfonyl, wherein the substituted group is substituted with one or two substituents selected from: a) Cl_4 alkoxy, b) aryl or heterocycle, c) halogen, d) HO, ,R £ e)
  • V is selected from: a) hydrogen, b) heterocycle, c) aryl, d) C l -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 Al is S(0)m and V is not hydrogen if A is a bond, n is 0 and A ⁇ is S(0)m;
  • W is a heterocycle
  • n 0, 1 , 2, 3 or 4
  • p 0, 1, 2, 3 or 4
  • q IS 0, 1 , 2, 3 or 4
  • r is 0 to 5, provided that r is 0 when V is hydrogen; s is 1 or 2; t is 0 or 1 ;
  • R a and Rlc are independently selected from: hydrogen, C3-C10 cycloalkyl, R 0-, -N(R8)2, F or C1-C6 alkyl;
  • R b is independently selected from: a) hydrogen, b) unsubstituted or substituted aryl, unsubstituted or substituted heterocycle, C3-C6 cycloalkyl, R 8 0-, -N(R 8 )2 or C2-C6 alkenyl, c) C1-C6 alkyl unsubstituted or substituted by unsubstituted or substituted aryl, heterocycle, C3-C6 cycloalkyl, C2-C6 alkenyl, R Q-, or -N(R8)2;
  • R2 is selected from: a) Cl-8 alkyl, unsubstituted or substituted with one or more of:
  • R5 is selected from: a) hydrogen, b) C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, Cl-C ⁇ perfluoroalkyl, F, CI, R 8 0-, R9s(0) m -, R8C(0)NR8-, CN, N ⁇ 2, (R8)2N-C(NR8)-, R8C(0)-, R8 ⁇ C(0)-, -N(R8)2, or
  • R9 ⁇ C(0)NR8- and c) C 1 -C6 alkyl unsubstituted or substituted by C 1 -C ⁇ perfluoroalkyl, F, CI, R 8 0-, R9S(0) m -, R8C(0)NR8-, CN, (R8)2N-C(NR8)-, R8c(0)-, R8 ⁇ C(0)-, -N(R8)2, or R90C(0)NR8-;
  • R6, R7 and R ⁇ a are independently selected from:
  • R is independently selected from hydrogen, Cl-C ⁇ alkyl, benzyl, 2,2,2-trifluoroethyl and aryl;
  • R9 is independently selected from Cl-C ⁇ alkyl and aryl;
  • RlO is selected from: H; R8C(0)-; R9s(0) m -; unsubstituted or substituted Cl-4 alkyl, unsubstituted or substituted C3-6 cycloalkyl, unsubstituted or substituted heterocycle, unsubstituted or substituted aryl, substituted aroyl, unsubstituted or substituted heteroaroyl, substituted arylsulfonyl, unsubstituted or substituted heteroarylsulfonyl, wherein the substituted group is substituted with one or two substituents selected from: a) Cl-4 alkoxy, b) aryl or heterocycle, c) halogen, d) HO,
  • V is selected from: a) heterocycle selected from pyrrolidinyl, imidazolyl, pyridinyl, thiazolyl, pyridonyl, 2-oxopiperidinyl, indolyl, quinolinyl, isoquinolinyl, and thienyl, and b) aryl;
  • W is a heterocycle selected from pyrrolidinyl, imidazolyl, pyridinyl, thiazolyl, pyridonyl, 2-oxopiperidinyl, indolyl, quinolinyl, or isoquinolinyl;
  • Rla and Rlc are independently selected from: hydrogen, C3-C10 cycloalkyl, R80-, -N(R8)2, F or Cl-C ⁇ alkyl;
  • R b is independently selected from: a) hydrogen, b) aryl, heterocycle, C3-C10 cycloalkyl, R80-, -N(R8)2, F or C2-C6 alkenyl, c) unsubstituted or substituted Cl-C ⁇ alkyl wherein the substituent on the substituted Cl-C ⁇ alkyl is selected from unsubstituted or substituted aryl, heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, R80- and -N(R8)2;
  • R is selected from: H; unsubstituted or substituted Cl-8 alkyl, unsubstituted or substituted C2-8 alkenyl, unsubstituted or substituted aryl,
  • substituted group is substituted with one or more of: 1 ) aryl or heterocycle, unsubstituted or substituted with one or two groups selected from: a) Cl-4 alkyl,
  • R4 is independently selected from: a) hydrogen, b) aryl, substituted aryl, heterocycle, substituted heterocycle, Cl-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, Cl -C ⁇ perfluoroalkyl, F, CI, R8O-, R8C(0)NR8-, CN, N ⁇ 2, (R8)2N-C(NR8)-, R8C(0)-, -N(R8)2, or R9oC(0)NR8-, and c) Cl-C ⁇ alkyl substituted by Cl-C ⁇ perfluoroalkyl, R80-, R8C(0)NR 8 -, (R8) 2 N-C(NR8)-, R8C(0)-, -N(R8)2, or R90C(0)NR8-;
  • R ⁇ a and R ⁇ b are independently hydrogen, Cl-C ⁇ alkyl, cyclopropyl, trifluoromethyl and halogen;
  • R6, R7 and R 7a are independently selected from:
  • R is independently selected from hydrogen, Cl -C ⁇ alkyl, 2,2,2- trifluoroethyl, benzyl and aryl;
  • R is independently selected from Cl -C ⁇ alkyl and aryl
  • RlO is selected from: H; R 8 C(0)-; R 9 S(0) m -; unsubstituted or substituted Cl-4 alkyl, unsubstituted or substituted C3-6 cycloalkyl, unsubstituted or substituted heterocycle, unsubstituted or substituted aryl, substituted aroyl, unsubstituted or substituted heteroaroyl, substituted arylsulfonyl, unsubstituted or substituted heteroarylsulfonyl, wherein the substituted group is substituted with one or two substituents selected from: a) C]-4 alkoxy, b) aryl or heterocycle, c) halogen, d) HO,
  • V is selected from: a) hydrogen, b) heterocycle selected from pyrrolidinyl, imidazolyl, imidazolinyl, pyridinyl, thiazolyl, pyridonyl, 2- oxopiperidinyl, oxazolyl, indolyl, quinolinyl, isoquinolinyl, triazolyl and thienyl, c) aryl, d) C 1-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 is a bond, n is 0 and A2 is S(0) m ;
  • r is 0 to 5, provided that r is 0 when V is hydrogen;
  • R a and Rlc are independently selected from: hydrogen, C3-C10 cycloalkyl, R 0-, -N(R8) , F or Cl -C ⁇ alkyl;
  • R b is independently selected from: a) hydrogen, b) aryl, heterocycle, C3-C10 cycloalkyl, R O-, -N(R 8 )2, F or
  • C2-C6 alkenyl c) unsubstituted or substituted Cl-C ⁇ alkyl wherein the substituent on the substituted Cl-C ⁇ alkyl is selected from unsubstituted or substituted aryl, heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, R80- and -N(R8)2;
  • R2 is selected from: H; unsubstituted or substituted Cl-8 alkyl, unsubstituted or substituted C2-8 alkenyl, unsubstituted or substituted
  • aryl unsubstituted or substituted heterocycle, O ' and -S(0)2R 6 , wherein the substituted group is substituted with one or more of: 1 ) aryl or heterocycle, unsubstituted or substituted with one or two groups selected from: a) Cl-4 alkyl,
  • R3 is selected from: H; O O
  • R4 is independently selected from: a) hydrogen, b) aryl, substituted aryl, heterocycle, substituted heterocycle,
  • Cl-C ⁇ alkyl C2-C6 alkenyl, C2-C6 alkynyl, Cl-C ⁇ perfluoroalkyl, F, CI, R 0-, R8C(0)NR 8 -, CN, NO2, (R8)2N-C(NR8)-, R8C(0)-, -N(R8)2, or R9 ⁇ C(0)NR8-, and c) Cl-C ⁇ alkyl substituted by Cl-C ⁇ perfluoroalkyl, R s O-,
  • R ⁇ a and R ⁇ b are independently hydrogen, Cl -C ⁇ alkyl, cyclopropyl, trifluoromethyl and halogen;
  • R6, R7 and R a are independently selected from:
  • R8 is independently selected from hydrogen, Cl-C ⁇ alkyl, 2,2,2- trifluoroethyl, benzyl and aryl;
  • R9 is independently selected from Cl-C ⁇ alkyl and aryl
  • RlO is selected from: H; R8C(0)-; R S(0) m -; unsubstituted or substituted Cl-4 alkyl, unsubstituted or substituted C3-6 cycloalkyl, unsubstituted or substituted heterocycle, unsubstituted or substituted aryl, substituted aroyl, unsubstituted or substituted heteroaroyl, substituted arylsulfonyl, unsubstituted or substituted heteroarylsulfonyl, wherein the substituted group is substituted with one or two substituents selected from: a) Cl-4 alkoxy, b) aryl or heterocycle, c) halogen, d) HO,
  • V is selected from: a) hydrogen, b) heterocycle selected from pyrrolidinyl, imidazolyl, imidazolinyl, pyridinyl, thiazolyl, pyridonyl, 2- oxopiperidinyl, oxazolyl, indolyl, quinolinyl, isoquinolinyl, triazolyl and thienyl, c) aryl, d) C l -C20 alkyl wherein from 0 to 4 carbon atoms are replaced with a a heteroatom selected from O, S, and N, and e) C2-C2O alkenyl, and 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 A2 is S(0) m ;
  • Rla and Rlc are independently selected from: hydrogen, C3-C10 cycloalkyl or Cl-C ⁇ alkyl;
  • R b is independently selected from: a) hydrogen, b) aryl, heterocycle, C3-C10 cycloalkyl, R 0-, -N(R8)2, F or C2-C6 alkenyl, c) Cl-C ⁇ alkyl unsubstituted or substituted by aryl, heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, R 8 0-, or
  • R is selected from: H; unsubstituted or substituted Cl-8 alkyl,
  • aryl or heterocycle unsubstituted or substituted with one or two groups selected from: a) Cl-4 alkyl,
  • R3 is selected from: H; O O
  • R4 is independently selected from: a) hydrogen, b) aryl, substituted aryl, heterocycle, substituted heterocycle,
  • Cl -C ⁇ alkyl C2-C6 alkenyl, C2-C6 alkynyl, Cl-C ⁇ perfluoroalkyl, F, CI, R8O-, R8C(0)NR8-, CN, NO2, (R8)2N-C(NR8)-, R ⁇ C(O)-, -N(R8)2, or R OC(0)NR8-, and c) Cl-C ⁇ alkyl substituted by Cl -C ⁇ perfluoroalkyl, R80-,
  • R ⁇ and R ⁇ b are independently hydrogen, ethyl, cyclopropyl or methyl
  • R6, R7 and R7a are independently selected from:
  • R is independently selected from hydrogen, Cl-C ⁇ alkyl, 2,2,2- trifluoroethyl, benzyl and aryl;
  • R9 is independently selected from Cl-C ⁇ alkyl and aryl
  • RlO is selected from: H; R 8 C(0)-; R9S(0) m -; unsubstituted or substituted Cl-4 alkyl, wherein the substituted alkyl group is substituted with one or two substituents selected from: a) Cl-4 alkoxy, b) aryl or heterocycle, c) halogen, d) HO,
  • a l is selected from: a bond, -C(O)-, O, -N(R8)-, or S(0) m ;
  • n is 0 or 1 ; provided that n is not 0 if A* is a bond, O, -N(R8)-, or S(0) m ; m is 0, 1 or 2; p is 0, 1 , 2, 3 or 4; and q is 0 or 1 ;
  • the inhibitors of famesyl-protein transferase are illustrated by the formula E: wherein:
  • Rl and Rl are independently selected from: hydrogen, R 0-, -N(R8)2, F, C3-C10 cycloalkyl or Cl-C ⁇ alkyl;
  • R b is independently selected from: a) hydrogen, b) aryl, heterocycle, C3-C10 cycloalkyl, R8 ⁇ -, -N(R 8 )2, F or C2-C6 alkenyl, c) Cl-C ⁇ alkyl unsubstituted or substituted by aryl, heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, R 0-, or
  • R2 is selected from: H; unsubstituted or substituted Cl -8 alkyl,
  • R3 is selected from: H;
  • R4 is independently selected from: a) hydrogen, b) aryl, substituted aryl, heterocycle, substituted heterocycle,
  • Cl -C ⁇ alkyl C2-C6 alkenyl, C2-C6 alkynyl, Cl-C ⁇ perfluoroalkyl, F, CI, R s O-, R8c(0)NR8-, CN, NO2, (R8) 2 N-C(NR8)-, R8C(0)-, -N(R8)2, or R9 ⁇ C(0)NR8-, and c) Cl -C ⁇ alkyl substituted by Cl -C ⁇ perfluoroalkyl, R s O-,
  • R5 and R b are independently hydrogen, ethyl, cyclopropyl or methyl
  • R6, R7 and R7a are independently selected from:
  • R is independently selected from hydrogen, Cl-C ⁇ alkyl, 2,2,2- trifluoroethyl, benzyl and aryl;
  • R9 is independently selected from Cl-C ⁇ alkyl and aryl
  • RlO is selected from: H; R8c(0)-; R9S(0) m -; unsubstituted or substituted Cl-4 alkyl, wherein the substituted alkyl group is substituted with one or two substituents selected from: a) Cl-4 alkoxy, b) aryl or heterocycle,
  • Rla and Rlc are independently selected from: hydrogen, C3-C10 cycloalkyl or Cl-C ⁇ alkyl;
  • Rib is independently selected from: a) hydrogen, b) aryl, heterocycle, C3-C10 cycloalkyl, R 8 0-, -N(R 8 )2 or F, c) Cl-C ⁇ alkyl unsubstituted or substituted by aryl, heterocycle, C3-C10 cycloalkyl, R 0-, or -N(R8)2;
  • R2 is selected from: H; unsubstituted or substituted Cl-8 alkyl,
  • unsubstituted or substituted aryl wherein the substituted group is substituted with one or more of: 1 ) aryl or heterocycle, unsubstituted or substituted with one or two groups selected from: a) Cl-4 alkyl,
  • R3 is selected from: H; O
  • R5 and R b are independently hydrogen, ethyl, cyclopropyl or methyl
  • R6, R7 and R7a are independently selected from:
  • R8 is independently selected from hydrogen, Cl-C ⁇ alkyl, 2,2,2- trifluoroethyl, benzyl and aryl;
  • R9 is independently selected from Cl-C ⁇ alkyl and aryl
  • RlO is selected from: H; R8c(0)-; R9S(0) m -; unsubstituted or substituted Cl-4 alkyl, wherein the substituted alkyl group is substituted with one or two substituents selected from: a) Cl-4 alkoxy, b) aryl or heterocycle, c) halogen, d) HO,
  • Rla and Rlc are independently selected from: hydrogen, R80-, -N(R8)2, F, C3-C10 cycloalkyl or Cl-C ⁇ alkyl;
  • R b is independently selected from: a) hydrogen, b) aryl, heterocycle or C3-C10 cycloalkyl, c) Cl-C ⁇ alkyl unsubstituted or substituted by aryl, heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, R 8 ⁇ -, or -N(R8)2; R2 is selected from: H; unsubstituted or substituted Cl-8 alkyl,
  • substituted group is substituted with one or more of: 1) aryl or heterocycle, unsubstituted or substituted with one or two groups selected from: a) Cl -4 alkyl,
  • R5a and R5b are independently hydrogen, ethyl, cyclopropyl or methyl
  • R6, R7 and R7a are independently selected from:
  • R is independently selected from hydrogen, Cl -C ⁇ alkyl, 2,2,2- trifluoroethyl, benzyl and aryl;
  • R9 is independently selected from C] -C ⁇ alkyl and aryl
  • RlO is selected from: H; R 8 C(0)-; R9S(0) m -; unsubstituted or substituted Cl-4 alkyl, wherein the substituted alkyl group is substituted with one or two substituents selected from: a) Cl-4 alkoxy, b) aryl or heterocycle, c) halogen, d) HO,
  • A is selected from: a bond, -C(O)-, O, -N(R8)-, or S(0) m ;
  • n 0, 1 or 2
  • n 0 or 1
  • p is 1, 2 or 3
  • q is 0 or 1 ;
  • the compounds of the present invention may have asymmetric centers and occur as racemates, racemic mixtures, and as individual diastereomers, with all possible isomers, including optical isomers, being included in the present invention.
  • any variable e.g. aryl, heterocycle, Rl a , R4 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, bicyclic or tricyclic carbon ring of up to 7 members in each ring, wherein at least one ring is aromatic.
  • monocyclic and bicyclic aryl elements include phenyl, naphthyl, tetrahydronaphthyl, indanyl, biphenyl, phenanthryl, anthryl or acenaphthyl.
  • tricyclic aryl elements include 10,11- dihydro-5H-dibenzo[a,d]cyclohepten-5-yl (which is also known as dibenzylsuberyl), 9-fluorenyl and 9,10-dihydroanthracen-9-yl.
  • "aryl” is a monocyclic or bicyclic carbon ring.
  • heterocycle or heterocyclic represents a stable 5- to 7-membered monocyclic or stable 8- to 11- membered bicyclic heterocyclic ring or stable 13- to 15-membered tricyclic heterocyclic ring, which is either saturated or unsaturated, and which consists of carbon atoms and from one to four heteroatoms selected from the group consisting of N, O, and S, and including any bicyclic group in which any of the above-defined heterocyclic rings is fused to a benzene ring.
  • the heterocyclic ring may be attached at any heteroatom or carbon atom which results in the creation of a stable structure.
  • Examples of monocyclic and bicyclic 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, mo ⁇ holinyl, naphthyridinyl,
  • tricyclic heterocyclic elements include, but are not limited to, 6,l l-dihydro-5H- benzo[5,6]cyclohepta[l,2-b]pyridine, 9,10-dihydro-4H-3-thia- benzo[f]azulen-4-yl and 9-xanthenyl.
  • the 6,1 l-dihydro-5H- benzo[5,6]cyclohepta[l,2-b]pyridine moiety has the following structure:
  • heterocyclic is a monocyclic or bicyclic moiety.
  • heteroaryl is intended to mean any stable monocyclic, bicyclic or tricyclic carbon ring of up to 7 members in each ring, wherein at least one ring is aromatic and wherein from one to four carbon atoms are replaced by heteroatoms selected from the group consisting of N, O, and S.
  • Examples of monocyclic and bicyclic heteroaryl elements include, but are not limited to, benzimidazolyl, benzisoxazolyl, benzofurazanyl, benzopyranyl, benzothiopyranyl, benzofuryl, benzothiazolyl, benzothienyl, benzoxazolyl, chromanyl, cinnolinyl, dihydrobenzofuryl, dihydrobenzothienyl, dihydrobenzothiopyranyl, dihydrobenzothiopyranyl sulfone, furyl, imidazolyl, indolinyl, indolyl, isochromanyl, isoindolinyl, isoquinolinyl, isothiazolyl, naphthyridinyl, oxadiazolyl, pyridyl, pyrazinyl, pyrazolyl, pyridazinyl, pyrimidinyl, pyrroly
  • tricyclic heteroaryl elements include, but are not limited to, 6,1 l-dihydro-5H- benzo[5,6]cyclohepta[l,2-b]pyridine.
  • heteroaryl is a monocyclic or bicyclic moiety.
  • substituted aryl As used herein, the terms “substituted aryl”, “substituted heterocycle” and “substituted cycloalkyl” are intended to include the cyclic group containing from 1 to 3 substitutents in addition to the point of attachment to the rest of the compound.
  • Such substitutents are preferably selected from the group which includes but is not limited to F, CI, Br, CF3, NH2, N(Cl-C6 alkyl)2, N ⁇ 2, CN, (Cl-C ⁇ alkyDO-, -OH, (Cl-C ⁇ alkyl)S(0) m -, (Cl-C ⁇ alkyl)C(0)NH-, H2N-C(NH)-, (Cl - C ⁇ alkyl)C(O)-, (Cl-C ⁇ alkyl)OC(O)-, N3,(Cl-C ⁇ alkyl)OC(0)NH- and C1-C20 alkyl.
  • R and R7 or R7 and R7a are combined to form a ring, cyclic amine moieties are formed. Examples of such cyclic moieties include, but are not limited to:
  • Such cyclic moieties may optionally include another heteroatom(s).
  • heteroatom-containing cyclic amine moieties include, but are not limited to:
  • Lines drawn into the ring systems from substituents indicate that the indicated bond may be attached to any of the substitutable ring carbon atoms.
  • Rla and Rib are independently selected from: hydrogen, -N(R8)2, R8C(0)NR8- or Cl-C ⁇ alkyl which is unsubstituted or substituted by -N(R 8 )2, R 8 0- or R S C(0)NR8-.
  • R2 is selected from: a) Cl-8 alkyl, unsubstituted or substituted with one or more of:
  • R2 comprises at least one unsubstituted or substituted phenyl.
  • R4 is selected from: hydrogen, perfluoroalkyl, F, CI, Br, R8 ⁇ -, R9s(0) m -, CN, N02, R82N-C(NR )-, R8C(0)-, N3, -N(R8)2, R9 ⁇ C(0)NR8- and Cl-C ⁇ alkyl.
  • R is hydrogen.
  • R7b is Cl -C ⁇ alkyl substituted with hydrogen or an unsubstituted or substituted aryl group.
  • R is selected from H, Cl-C ⁇ alkyl and benzyl.
  • a ⁇ and A2 are independently selected from: a bond, -C(0)NR8-, -NR8C(0)-, O, -N(R8)-, -S(0)2N(R8)- and-
  • V is selected from hydrogen, heterocycle and aryl.
  • W is imidazolyl.
  • n, p and r are independently 0, 1 , or 2.
  • t is 1.
  • 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.
  • any substituent or variable e.g., Rla, Z, n, etc.
  • -N(R8)2 represents -NH2, -NHCH3, -NHC2H5, etc.
  • 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 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.
  • DMAP 4-Dimethylaminopyridine
  • DME 1,2-Dimethoxyethane
  • the compounds of this invention are prepared by employing reactions as shown in the Schemes 1-21, 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. While stereochemistry is shown in the Schemes, a person of ordinary skill in the art would understand that the illustrated compounds represent racemic mixtures which may be separated at a subsequent purification step or may be utilized as the racemic mixture.
  • the monocarboxylic acid 2 can be treated with an appropriately substituted amine in the presence of a suitable coupling reagent, such as EDC/HOBT, and the like, to provide the 5- carboxynipecotamide 3.
  • a suitable coupling reagent such as EDC/HOBT, and the like
  • the suitably substituted 5- carboxynipecotamide is then deprotected and the piperidine nitrogen can then be reductively alkylated to provide intermediate 4.
  • the remaining ester moiety is saponified and then similarly functionalized with another suitably substituted amine to provide the bisamidopiperidine 5.
  • the monocarboxylic acid 2 can undergo a Curtius rearrangment to provide the piperidine 6 after catalytic reduction. Subsequent amide formation provides intermediate 7, which is then subjected to the reactions illustrated in Scheme 1 to provide compound 8 of the instant invention.
  • the instant invention also includes 1 ,4-dihydropyridine and 1,2,3,4-tetrahydropyridine analogs of the piperidine compounds whose syntheses are described above.
  • Scheme 4 illustrates the synthetic route to the intermediates 11 and 13 which correspond to the saturated ring intermedate 2 illustrated in Scheme 1.
  • the appropriately substituted pyridine may be N-alkylated to provide the quaternary intermediate 9.
  • Subsequent reduction of this intermediate provides the 1 ,4-dihydropyridine 10, which can be selectively hydrolized to the key intermediate 11.
  • the 1 ,4-dihydropyridine 10 can be further reduced to provide the enantiomeric mixture of tetrahydropyridines 12, which can be hydrolized and resolved by chromatography to provide the key intermediate 13 (and the enantiomer which is not illustrated).
  • Intermediates 11 and 13 can then undergo synthetic modifications as described hereinabove in Schemes 1- 3.
  • Schemes 5-7 illustrate the syntheses of 1,3-disubstituted piperidines of the instant invention wherein the "X" moiety is other than an amido moiety.
  • the reactions illustrated therein may be modified by using appropriate protecting groups and reagents well known to one skilled in the art to provide 1,3,5-trisubstituted piperidines of the instant invention.
  • Scheme 5 illustrates the syntheses of compounds of the instant invention wherein "X" is -S- or -S ⁇ 2--
  • a racemic nipecotate 14 can be resolved by the selective crystallization of chiral tartrate salts and is then reductively alkylated to provide the ester 15.
  • Intermediate 15 is reduced to the alcohol 16, activated and treated with a suitable thioacetate to provide the thioester 17.
  • the thiol is then generated and may be alkylated and optionally oxidized to provide compounds 18 and 19 of the instant invention.
  • the intermediate 16 may be selectively oxidized back to an aldehyde, which can then be utilized to reductively alkylate a suitably substituted amine to provide the instant compound 20.
  • the secondary amine of 20 can be further functionalized as illustrated.
  • the activated alcohol can also be reacted with a suitably substituted imidazolyl to provide compounds of the instant invention wherein "X" is a bond, as shown in Scheme 7.
  • Scheme 8 illustrates the syntheses of compounds of the instant invention wherein R2 is an aryl moiety.
  • RS C CH2- is R2 or a protected precursor thereof; and RSb_ i s R6 O ⁇ a protected precusor thereof; and
  • R- is a "substituent" or a protected precusor thereof.
  • the selectively protected intermediate 20 utilized in the synthesis illustrated in Scheme 9 can be reductively alkylated with a variety of aldehydes, such as 21.
  • 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.
  • the reductive alkylation can be accomplished at pH 5-7 with a variety of reducing agents, such as sodium triacetoxyborohydride or sodium cyanoborohydride in a solvent such as dichloroethane, methanol or dimethylformamide.
  • the ester product 22 can be deprotected with trifluoroacetic acid in methylene chloride to give the substituted diamine 23. That diamine may be isolated in the salt form, for example, as a trifluoroacetate, hydrochloride or acetate salt, among others.
  • the product diamine 23 can be further selectively protected and reductively alkylated with a second aldehyde to obtain an analogous tertiary amine.
  • the diamine 23 can be cyclized to obtain intermediates such as the dihydroimidazole 24 by procedures known in the literature.
  • the ester 24 can then be utilized in a reaction such as illustrated in Scheme 3 hereinabove or can be converted to the amine 26, via the azido intermediate 25.
  • Scheme 10 illustrates preparation of aralkyl imidazolyl intermediates 31 that can be utilized in reactions such as illustrated in Scheme 3.
  • imidazole acetic acid 27 can be converted to the protected acetate 29 by standard procedures, and 29 can be first reacted with an alkyl halide, then treated with refluxing methanol to provide the regiospecifically alkylated imidazole acetic acid ester 30. Hydrolysis provides the acetic acid 31.
  • intermediate 31 can be converted into the homologous amine 34 via the azido intermediate 33, as shown in Scheme 1 1. This amine can then be utilized in reactions such as illustrated in Scheme 1.
  • the Boc protected phthalimidyl alcohol 39 can also be utilized to synthesize 2-aziridinylmethylamines such as 42 (Scheme 13). Treating 39 with l,l'-sulfonyldiimidazole and sodium hydride in a solvent such as dimethylformamide led to the formation of aziridine 42. The aziridine may then be reacted in the presence of a nucleophile, such as a thiol, in the presence of base to yield, after deprotection, the ring- opened intermediate amine 43.
  • a nucleophile such as a thiol
  • amines such as 48 derived from amino acids such as O-alkylated tyrosines can be prepared according to standard procedures as shown in Scheme 14. Illustrated is a procedure where the amine moiety is derived from the azide of an intermediate such as 47.
  • Schemes 15-18 illustrate syntheses of suitably substituted alkanols useful in the syntheses of the instant compounds wherein the variable W is present as a pyridyl moiety.
  • the hydroxyl moiety of such intermediates may be converted into the corresponding amine, as illustrated in Scheme 15 or may be converted to a suitable leaving group, as illustrated in Scheme 17.
  • Similar synthetic strategies for preparing alkanols that incorporate other heterocyclic moieties for variable W are also well known in the art.
  • Al(CRl a 2)nA2(CRl a 2)n linker is a substituted methylene may be synthesized by the methods shown in Scheme 19.
  • the N-protected imidazolyl iodide 50 is reacted, under Grignard conditions with a suitably protected benzaldehyde to provide the alcohol 51.
  • Acylation, followed by the alkylation procedure illustrated in the Schemes above (in particular, Scheme 7) provides the instant compound 52. If other R 1 substituents are desired, the acetyl moiety can be manipulated as illustrated in the Scheme.
  • Scheme 20 illustrates synthesis of an instant compound wherein a non-hydrogen R ⁇ is incorporated in the instant compound.
  • a readily available 4-substituted imidazole 53 may be selectively iodinated to provide the 5-iodoimidazole 54. That imidazole may then be protected and coupled to a suitably substituted benzyl moiety to provide intermediate 55. Intermediate 55 can then undergo the alkylation reactions that were described hereinabove.
  • R' is R >1 a a or a protected precursor thereof
  • R' is (R 4 ) r -V- or a protected precursor thereol SCHEME 14
  • R'CH 2 - is R 8 or a protected precursor thereof
  • 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, ser, abl, lck, fyn) or by other mechanisms.
  • the compounds of the instant invention inhibit farnesyl- 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 poly cystic kidney disease (D.L. Schaffner et al. American Journal of Pathology, 142:1051-1060 (1993) and B. Cowley, Jr. et 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 com 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 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 intramuscular 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 farnesyl-protein transferase (FPTase) in a composition.
  • FPTase farnesyl-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 famesyl 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 B Preparation of Piperidine-cw, rra «5-3,5-dicarboxylic acid methyl ester hydrochloride
  • Step C Preparation of l-(t-Butoxycarbonyl)piperidine-c/s and
  • Piperidine-3,5-dicarboxylic acid methyl ester hydrochloride (7.11 g, 29.9 mmol) was dissolved in THF (60 mL) and H2 ⁇ (60 mL).
  • Sodium bicarbonate (13.81 g, 0.164 mol) was added followed by di-tert-butyl dicarbonate (9.79 g, 44.9 mmol).
  • the mixture was stirred at ambient temperature for 5 hrs.
  • the THF was removed under reduced pressure, and the solution was extracted with CH2CI2 (3x100 mL). The combined CH2CI2 layers were washed with brine and dried (MgS ⁇ 4).
  • Step D Preparation of l-(t-Butoxycarbonyl)- s-3- methoxycarbonyl-piperidine-5-carboxylic acid l-(t-Butoxycarbonyl)piperidine-c/_?-3,5-dicarboxylic acid methyl ester(0.761 g, 2.52 mmol) was dissolved in DME (6 mL) and H2 ⁇ (6 mL) followed by addition of LiOH H2 ⁇ (0.106 g, 2.52 mmol). The mixture was stirred at ambient temperature overnight. The DME was removed under reduced pressure, taken up in EtOAc and water, acidified to pH 3, and extracted with EtOAc (10X). The EtOAc layers were combined, dried (MgS04), filtered, and concentrated to dryness to give the title compound after chromatography (silica gel, 1-3% MeOH CH2Cl2).
  • NY-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. I, 1341, (1979)).
  • ⁇ -Bromo-p-tolunitrile (3.77 g, 19.2 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 F Preparation of l-(t-Butoxycarbonyl)-c/s-3-methoxy- carbonyl-5-[N-( 1 -(4-cyanobenzyl)- 1 H-imidazol-5- ylethyDcarbonyll-piperidine 1 -t-Butoxycarbonyl-c/s-3-methoxycarbonyl-piperidine-5- carboxylic acid (1.45 g, 5.06 mmol), 3-(4-cyanobenzyl) histamine ( 1.14 g, 5.06 mmol), HOBT (0.72 g, 5.31 mmol), EDC (1.02 g, 5.31 mmol), Et3N (0.63 mL, 4.55 mmol) were dissolved in DMF (20 mL).
  • Step D Following the procedure of Steps D-F but substituting the l-(t-butoxycarbonyl)piperidine-tr ⁇ ns-3,5-dicarboxylic acid methyl ester prepared as described in Step C for the l-(t-Butoxycarbonyl)piperidine- ct5-3,5-dicarboxylic acid methyl ester utilized in Step D provided l-(t- butoxycarbonyl)-fr ⁇ ns-3-methoxy-carbonyl-5-[N-(l -(4-cyanobenzyl)- lH-imidazol-5-ylethyl)carbamoyl]-piperidine.
  • Step A Preparation of c/s-3-Methoxycarbonyl-5-[N-(l-(4- cyanobenzyl)-lH-imidazol-5-ylethyl)carbamoyll piperidine
  • Step B Preparation of l-Phenethyl-ris-3-methoxycarbonyl-5-[N-
  • the solution was purified on a RP HPLC VYDAC column (0.1% TFA in CH3CN: 0.1 % TFA in H2O, 5:95 to 95:5 gradient) and lyophilized to give the title compound as cis diastereomers.
  • the solution was heated at 90°C for 4 h.
  • the solution was diluted with EtOAc and was washed with Sat. NaHC ⁇ 3 solution, water, and brine.
  • the organics were dried (MgS ⁇ 4), filtered, and concentrated to give the title compound without further purification.
  • Step B Preparation of l-(t-Butoxycarbonyl)- ' s-3-methoxy- carbonyl-5-amino piperidine l-(t-Butoxycarbonyl)- s-3-methoxycarbonyl-5- (benzyloxycarbonyl)amino piperidine (2.55 g, 6.51 mmol) was dissolved in EtOAc (75 mL), treated with Pd/C (510 mg) and shaken on a Parr apparatus at 45 psi overnight. The reaction mixture was filtered through celite, concentrated, and chromatographed (silica gel, 2%
  • Step C Preparation of lH-Imidazole-4- acetic acid methyl ester hydrochloride
  • Step D Preparation of l-(Triphenylmethyl)-lH-imidazol-4-ylacetic acid methyl ester
  • Step E Preparation of [l-(4-Cyanobenzyl)-lH-imidazol-5-yl]acetic acid methyl ester
  • the precipitated imidazolium salts were combined, suspended in methanol (100 ml) and heated to reflux for 30min. After this time, the solvent was removed in vacuo, the resulting residue was suspended in EtOAc (75ml) and the solid isolated by filtration and washed (EtOAc). The solid was treated with sat aq NaHC ⁇ 3 (300ml) and CH2CI2 (300ml) and stirred at room temperature for 2 hr.
  • Step F Preparation of [1 -(4-cyanobenzyl)- lH-imidazol-5-yl]acetic acid
  • Step G Preparation of l-(t-Butoxycarbonyl)-cw-3-methoxy- carbonyl-5-[N-( 1 -(4-cyanobenzyl)- 1 H-imidazol-5- yPacetylaminol-piperidine l-(t-Butoxycarbonyl)- s-3-methoxycarbonyl-5-amino piperidine (134 mg, 0.520 mmol), [1 -(4-cyanobenzyl)- lH-imidazol-5- yl]acetic acid (147 mg, 0.520 mmol), HOBT (73,8 mg, 0.546 mmol), EDC (104 mg, 0.546 mmol), and Et3N (65.2 uL, 0.468 mmol) were dissolved in DMF (4mL) and stirred at ambient temperature overnight.
  • Step B Preparation of l-Phenethyl- s-3-methoxycarbonyl-5-[N-
  • Step A Preparation of 1 -(2.2-Diphenylethyl)-3-carboxy piperidine Nipecotic acid (300 mg, 2.38 mmol), diphenylacetaldehyde (1.26 mL, 7.13 mmol), sodium cyanoborohydride (448 mg, 7.13 mmol), and HOAc (204 uL, 3.57 mmol) were dissolved in MeOH (20 mL) and stirred at ambient temperature overnight. The solution was concentrated under reduced pressure, take up in ether and IN NaOH, extract with ether (3X), acidify the aqueous layer with IN HCI, and extract with EtOAc (3X). The EtOAc layers were dried (MgS ⁇ 4) and concentrated to give the title compound without further purification.
  • Step B Preparation of l -(2,2-Diphenylethyl)-3-[N-(l-(4- cyanobenzyP-lH-imidazol-5-ylethyl)carbamoyll piperidine l-(2,2-Diphenylethyl)-3-carboxy piperidine(472 mg, 1,52 mmol),3-(4-cyanobenzyl) histamine (456 mg, 1.52 mmol) (EXAMPLE 1, Step E) HOBT (216 mg, 1.60 mmol), EDC (307 mg, 1.60 mmol), and Et3N (637 uL, 4.57 mmol) were dissolved in DMF (10 mL) and was stirred overnight at ambient temperature.
  • Step A Preparation of S-(-)-Ethyl nipecotate
  • Racemic ethyl nipecotate (122.5 g, 0.78 mol) was resolved with D-tartaric acid (117 g, 0.78 mol) in 95% EtOH (611 mL) following the procedure described by P. Magnus et al. (J. Org. Chem. 1991, 56, 1166-1170) to give S-(-)-ethyl nipecotate.
  • Step B Preparation of Ethyl l-(t-butoxycarbonyl)piperidine-3(S)- carboxylate
  • Step C Preparation of l-(tert-Butoxycarbonyl)piperidine-3(S)- carboxylic acid
  • Step D Preparation of l-(tert-Butyloxycarbonyl)-3(S)-[N- l- (4- cyanobenzvP-lH-imidazol-5-ylethyl)carbamoyll piperidine 1 -(tert-Butoxycarbonyl)piperidine-3(S)-carboxylic acid (5.06 g, 0.022 mol) and 3-(4-cyanobenzyl)histamine (Example 1 , Step E) (6.6 g, 0.022 mol) were dissolved in DMF (30 mL) at ambient temperature and treated with EDC ( 5.07 g, 0.026 mol), HOBT ( 3.58 g, 0.26 mol), and N-methylmorpholine (12.12 mL, 0.11 mol).
  • reaction mixture was partitioned between EtOAc(500 mL)- aq satd NaHC0 3 soln, the organic layer separated, washed with satd NaHC0 3 soln, H 2 0, brine, dried (MgS0 4 ), filtered and concentrated to give the title compound which was used without further purification.
  • Step E Preparation of 3(S)-[N-(1 -(4-cyanobenzyl)- lH-imidazole-5- ethyPcarbamoyll piperidine dihydrochloride l-(tert-Butyloxycarbonyl)-3(S)-[N-l- (4-cyanobenzyl )- lH-imidazol-5-ylethyl)carbamoyl] piperidine (9.25 g, 0.021 mol) was dissolved in EtOAc (500 mL) with stirring at 0°C. in an ice- water bath.
  • Step F Preparation of 2-(3-Chlorophenyl)-2-phenyl oxirane
  • NaH 50% dispersion in mineral oil
  • DMSO 40 mL
  • To this reaction mixture was added trimethylsulfoxonium iodide (10.56 g, 0.048 mol) through a solid addition funnel over 15 min.
  • a solution of 3-chlorobenzophenone (8.66 g, 0.04 mol) in DMSO (15 mL) was added dropwise, and the mixture was heated at 55 °C.
  • Step H Preparation of l-(2-(3-Chlorophenyl)-2-phenylethyl)-3- (S)-[N-(1 -(4-cyanobenzyl)- lH-imidazol-5- ylethyPcarbamoyll piperidine 3(S)-[N-( 1 -(4-cyanobenzyl)- 1 H-imidazol-5- ylethyl)carbamoyl] piperidine dihydrochloride (0.256 g, 0.623 mmol) was dissolved in MeOH (10 mL), the pH adjusted to 5 with Et N, and 2- (3-chlorophenyl)-2-phenylcarboxaldehyde (0.427 g, 1.87 mmol) and NaCNBH 3 (0.078 g, 1.25 mmol) were added.
  • Step A Preparation of Ethyl l-[2-(2-pyridyl)-2-phenyl-2- hydroxyethvn piperidine-3(S)- carboxylate
  • Step B Preparation of l-[2-(2-Pyridyl)-2-phenyl-2-hydroxyethylj piperidine-3(S)- carboxylic acid
  • Step C Preparation of l-[2-(2-Pyridyl)-2-phenyl-2-hydroxyethyl]-
  • Step A Preparation of Ethyl l-(2-pyridylethyl) piperidine-3(S )- carboxylate
  • Step B Preparation of l-(2-Pyridylethyl) piperidine-3(S)- carboxylic acid
  • Step C Preparation of l-(2-Pyridylethyl)-3(S)-[N-(l-(4- cyanobenzyP- 1 H-imidazol-5-ylethyl)carbamoyl1 piperidine Following the procedure of Example 15, Step C, the title compound was prepared. FAB MS (M+l) 413.
  • Step A Preparation of Ethyl l-phenyl-(S)-piperidine carboxylate
  • Step B Preparation of l-Phenyl-(S)- piperidine carboxylic acid
  • Step C Preparation of l-Phenyl-3(S)-[N-(l-(4-cyanobenzyl)-lH- imidazol-5-ylethyl)carbamoyll piperidine
  • Step A Preparation of Ethyl l-(2,2-Dipheny lethyl)-3(S)- piperidine carboxylate
  • Step B Preparation of l-(2,2-Diphenylethyl)-3(S)-hydroxymethyl- piperidine
  • Ethyl l-(2,2-Diphenylethyl)-3(S)-piperidine carboxylate (4.90 g, 0.014 mol) dissolved in dry ether (40mL) was added to a suspension of lithium aluminum hydride (1.93 g, 0.051 mol) in dry ether (40mL). The solution was refluxed until the starting material was consumed and then was quenched with saturated potassium sodium tartrate solution (lOOmL) and stirred for 2 hr. The layers were separated and the aqueous layer extracted with ether (2X). The combined organic extracts were dried (MgS0 4 ) and concentrated to yield the title compound without further purification.
  • Step C Preparation of l-(2.2-Di ⁇ henylethyl)-3(S)-tosyloxymethyl- piperidine l-(2,2-Diphenylethyl)-3(S)-hydroxymethyl-piperidine (1.01 g, 3.41 mmol) was dissolved in dry pyridine (25 mL) and tosyl chloride (0.684 g, 3.58 mmol) was added to the solution After stirring at ambient temperature for 18 hr, the solution was concentrated, the residue taken up in EtOAc, washed with sat. NaHC0 3 solution, H 2 0, brine, and dried (MgS0 4 ). Filtration and concentration gave the title compound without further purification.
  • Step D Preparation of l -(2,2-Diphenylethyl)-3(S)- acetylthiomethyl-piperidine l-(2,2-Diphenylethyl)-3(S)-tosyloxymethyl-piperidine (1.35 g, 3.23 mmol), potassium thiol acetate (1.47g, 12.9 mmol) and DMF (30mL) were heated at 100°C for 4 hr. The solution was poured into ice and the resulting green precipitate was dissolved in ETOAc. The organics were washed with H 2 0 (2X), sat. NaHC0 3 solution, brine, dried (MgS04) and concentrated to give the title compound without further purification.
  • Step D Preparation of the disulfide of l-(2,2-Diphenylethyl)-3(S)- mercaptomethyl-piperidine l-(2,2-Diphenylethyl)-3(S)-acetylthiomethyl-piperidine (0.523 g, 1.47 mmol), EtOH (20 mL), and NaOH (0.5N, 20mL) was refluxed 3 hr. The solution was concentrated to remove ethanol and the remaining aqueous solution was decanted from the green oil. The oil was washed with water (2X) and decanted. The oil was dried under reduced pressure to give the title compound without further purification.
  • Step E Preparation of l-(2,2-Diphenylethyl)-3(S)- mercaptomethyl-piperidine
  • Step F Preparation of l-Triphenylmethyl-4-(hydroxymethyl)- imidazole
  • Step G Preparation of l-Triphenylmethyl-4-(acetoxymethyl)- imidazole
  • Step H Preparation of l-(4-Cyanobenzyl)-5-(acetoxymethyl)- imidazole hydrobromide l-Triphenylmethyl-4-(acetoxymethyl)-imidazole (85.8 g, 225 mmol) and 4-cyanobenzyl bromide (50.1 g, 232 mmol) in EtOAc (500mL) were stirred at 60 °C for 20 hr, 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 2 hrs, cooled to room temperature, and filtered again.
  • the filtrate was concentrated in vacuo to a volume (100 mL), reheated at 60 °C for another 2hrs, cooled to room temperature, and concentrated in vacuo to provide a pale yellow solid. All of the solid material was combined, dissolved in methanol (500mL), and warmed to 60 °C. After 2hrs, the solution was concentrated 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 I Preparation of l-(4-Cyanobenzyl)-5-(hydroxymethyl)- imidazole l-(4-Cyanobenzyl)-5-(acetoxymethyl)-imidazole hydrobromide (50.4 g, 150 mmol) in 3: 1 THF/water (1.5 L) at 0 °C was added lithium hydroxide monohydrate (18.9 g, 450 mmol). After lhr, 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 (Na2S ⁇ 4), filtered, and concentrated in vacuo to provide the crude product as a pale yellow fluffy solid which was sufficiently pure for use in the next step without further purification.
  • Step J Preparation of l-(4-Cyanobenzyl)-5-(chloromethyl)- imidazole
  • Step K Preparation of l-(2,2-Diphenylethyl)-3(S)-[N-(l-(4- cyanobenzyl)- 1 H-imidazol-5-ylethylthiomethvn piperidine l-(2,2-Diphenylethyl)-3(S)-mercaptomethyl-piperidine (0.322 mmol), l-(4-cyanobenzyl)-5-(chloromethyl)-imidazole (0.1 17 g, 0.386 mmol), and diisopropylethylamine (0.168 mL, 0.966 mmol) were dissolved in CH 2 C1 2 (lOmL) and refluxed overnight.
  • Step A Preparation of l-(tert-Butyloxycarbonyl)-3(S)-[N-(l-(4- cyanobenzyl)- 1 H-imidazol-5-ylethyl)-N-methylcarbamoyl] piperidine l-(tert-Butyloxycarbonyl)-3(S)-[N-( 1 -(4-cyanobenzyl)- 1 H- imidazol-5-ylethyl)carbamoyl] piperidine (Example 11, Step D)(0.100 g, 0.228 mmol) was dissolved in dry DMF (5mL) and cooled in a ice bath.
  • Step B Preparation of 1-3(S)-[N-(1 -(4-cyanobenzyl)- lH-imidazole-
  • Step C Preparation of l-(2,2-Diphenylethyl)-3(S)-[N-l-(4- cyanobenzyl)- 1 H-imidazol-5-ylethyl)-N-methylcarbamoyl] piperidine
  • Step A Preparation of l-(2,2-Diphenylethyl)-3(S)-piperidine carboxaldehyde 1 -(2,2-Diphenylethyl)-3(S)-hydroxymethyl-piperidine
  • Step B Preparation of l-(2,2-Diphenylethyl)-3(S)-[N-( 1-
  • Step A Preparation of l-Trityl-4-(4-cyanobenzyl)-imidazole
  • Step B Preparation of l-(2,2-Diphenylethyl)-3(S)-[5-(4- cyanobenzyl)-l H-imidazol- 1-ylmethyl] piperidine bis trifluoroacetate bistrifluoroacetate
  • Step A Preparation of 5-(4-cyanobenzyl)-l H-imidazol- 1- ylethylphthalimide
  • Step B Preparation of 5-(4-cyanobenzyl)-l-(2-aminoethyl)- lH- imidazole
  • Step C Preparation of l-(2,2-Diphenylethyl)-3(S)-[5-(4- cvanobenzyl)- 1 H-imidazol- 1 - ylethylcarbamoyll piperdine
  • Bovine FPTase was assayed in a volume of 100 ⁇ l containing 100 mM 7V-(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 ([ ⁇ Hj-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 [ 3 H]-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 inco ⁇ oration of radioactivity in the presence of the test compound when compared to the amount of inco ⁇ oration 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 nM 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. The compounds of the instant invention that are described in Example 1-24 were tested for inhibitory activity against human FPTase by the assay described above and were found to have IC50 of ⁇ 10 ⁇ M.
  • 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 mCi[35S]methionine (1000 Ci/mmol). After an additional 20 hours, the cells are lysed in 1 ml lysis buffer (1% NP40/20 mM HEPES, pH 7.5/5 mM MgCl2/l M 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/l M 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 famesylated 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 4 cells per plate (35 mm in diameter) in a 0.3% top agarose layer in medium A (Dulbecco's modified Eagle's medium supplemented with 10% fetal bovine serum) over a bottom agarose layer (0.6%). Both layers contain 0.1 % methanol or an appropriate concentration of the instant compound (dissolved in methanol at 1000 times the final concentration used in the assay).
  • the cells are fed twice weekly with 0.5 ml of medium A containing 0.1 % methanol or the concentration of the instant compound. Photomicrographs are taken 16 days after the cultures are seeded and comparisons are made.
EP96942798A 1995-11-22 1996-11-18 Inhibitoren der farnesyl-protein-transferase Withdrawn EP0862435A4 (de)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US749895P 1995-11-22 1995-11-22
US7498P 1995-11-22
GBGB9604311.2A GB9604311D0 (en) 1996-02-29 1996-02-29 Inhibitors of farnesyl-protein transferase
GB9604311 1996-02-29
PCT/US1996/018811 WO1997018813A1 (en) 1995-11-22 1996-11-18 Inhibitors of farnesyl-protein transferase

Publications (2)

Publication Number Publication Date
EP0862435A1 true EP0862435A1 (de) 1998-09-09
EP0862435A4 EP0862435A4 (de) 1999-02-03

Family

ID=26308829

Family Applications (1)

Application Number Title Priority Date Filing Date
EP96942798A Withdrawn EP0862435A4 (de) 1995-11-22 1996-11-18 Inhibitoren der farnesyl-protein-transferase

Country Status (5)

Country Link
EP (1) EP0862435A4 (de)
JP (1) JP2000500502A (de)
AU (1) AU704139B2 (de)
CA (1) CA2238081A1 (de)
WO (1) WO1997018813A1 (de)

Families Citing this family (86)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2243507A1 (en) * 1996-01-30 1997-08-07 Christopher J. Dinsmore Inhibitors of farnesyl-protein transferase
US5891889A (en) * 1996-04-03 1999-04-06 Merck & Co., Inc. Inhibitors of farnesyl-protein transferase
WO1997038665A2 (en) * 1996-04-03 1997-10-23 Merck & Co., Inc. Inhibitors of farnesyl-protein transferase
US6117641A (en) 1996-04-11 2000-09-12 Mitotix, Inc. Assays and reagents for identifying anti-fungal agents and uses related thereto
JP2001513622A (ja) 1996-04-11 2001-09-04 マイトティックス インコーポレーテッド 抗菌剤の同定のためのアッセイおよび試薬、並びにそれらに関連する利用
US6727082B1 (en) 1996-04-11 2004-04-27 Gpc Biotech Inc. Assays and reagents for identifying anti-fungal agents, and uses related thereto
US5965570A (en) * 1996-09-13 1999-10-12 Schering Corporation Tricyclic piperidinyl compounds useful as inhibitors of farnesyl-protein transferase
US5972966A (en) * 1996-12-05 1999-10-26 Merck & Co., Inc. Inhibitors of farnesyl-protein transferase
US5932590A (en) * 1996-12-05 1999-08-03 Merck & Co., Inc. Inhibitors of farnesyl-protein transferase
US5977134A (en) * 1996-12-05 1999-11-02 Merck & Co., Inc. Inhibitors of farnesyl-protein transferase
US6015817A (en) * 1996-12-05 2000-01-18 Merck & Co., Inc. Inhibitors of farnesyl-protein transferase
US5958940A (en) * 1997-09-11 1999-09-28 Schering Corporation Tricyclic compounds useful as inhibitors of farnesyl-protein transferase
WO1999027928A1 (en) * 1997-12-04 1999-06-10 Merck & Co., Inc. Inhibitors of farnesyl-protein transferase
FR2780892B1 (fr) * 1998-07-08 2001-08-17 Sod Conseils Rech Applic Utilisation d'inhibiteurs de prenyltransferases pour preparer un medicament destine a traiter les pathologies qui resultent de la fixation membranaire de la proteine g heterotrimerique
US6432959B1 (en) 1998-12-23 2002-08-13 Schering Corporation Inhibitors of farnesyl-protein transferase
FR2788274B1 (fr) * 1999-01-11 2001-02-09 Aventis Pharma Sa Nouveaux derives de polyhydroxypyrazines, leur preparation et les compositions pharmaceutiques qui les contiennent
US6677332B1 (en) 1999-05-25 2004-01-13 Sepracor, Inc. Heterocyclic analgesic compounds and methods of use thereof
US6635661B2 (en) 2000-05-25 2003-10-21 Sepracor Inc. Heterocyclic analgesic compounds and methods of use thereof
US7361666B2 (en) 1999-05-25 2008-04-22 Sepracor, Inc. Heterocyclic analgesic compounds and methods of use thereof
WO2001068604A2 (en) 2000-03-14 2001-09-20 Sepracor, Inc. 3-substituted piperidines comprising urea functionality, and methods of use thereof
AU2001216218A1 (en) * 2000-05-25 2001-12-11 Sepracor, Inc. Heterocyclic analgesic compounds and method of use thereof
EP1296978A2 (de) 2000-06-30 2003-04-02 Bristol-Myers Squibb Company N-ureidoheterocycloaklylpiperidine als modulatoren der chemokinrezeptoraktivität
KR100432577B1 (ko) * 2002-02-08 2004-05-24 주식회사유한양행 이미다졸 유도체의 제조방법
US6924285B2 (en) 2002-03-30 2005-08-02 Boehringer Ingelheim Pharma Gmbh & Co. Bicyclic heterocyclic compounds, pharmaceutical compositions containing these compounds, their use and process for preparing them
JP4137159B2 (ja) 2003-06-17 2008-08-20 ファイザー・インク セロトニン及びノルアドレナリン再取り込み阻害薬としてのn−ピロリジン−3−イル−アミド誘導体
GB0317665D0 (en) 2003-07-29 2003-09-03 Astrazeneca Ab Qinazoline derivatives
WO2005026150A1 (en) 2003-09-16 2005-03-24 Astrazeneca Ab Quinazoline derivatives as tyrosine kinase inhibitors
ATE550019T1 (de) 2005-05-17 2012-04-15 Merck Sharp & Dohme Cis-4-ä(4-chlorophenyl)sulfonylü-4-(2,5- difluorophenyl)cyclohexanepropansäure zur behandlug von krebs
ES2430139T3 (es) 2005-12-30 2013-11-19 Novartis Ag Compuestos de piperidina 3,5-sustituido como inhibidores de renina
GB0603041D0 (en) 2006-02-15 2006-03-29 Angeletti P Ist Richerche Bio Therapeutic compounds
EP2698157B1 (de) 2006-09-22 2015-05-20 Merck Sharp & Dohme Corp. Verfahren zur Behandlung von Fettsäure-Synthese-Hemmern
US20110218176A1 (en) 2006-11-01 2011-09-08 Barbara Brooke Jennings-Spring Compounds, methods, and treatments for abnormal signaling pathways for prenatal and postnatal development
EP1921070A1 (de) 2006-11-10 2008-05-14 Boehringer Ingelheim Pharma GmbH & Co. KG Bicyclische Heterocyclen, diese Verbindungen enthaltende Arzneimittel, deren Verwendung und Verfahren zu ihrer Herstelllung
EP2109608B1 (de) 2007-01-10 2011-03-23 Istituto di Richerche di Biologia Molecolare P. Angeletti S.p.A. Amidsubstituierte indazole als poly-(adp-ribose)-polymerase (parp)-hemmer
WO2008093737A1 (ja) 2007-01-31 2008-08-07 Dainippon Sumitomo Pharma Co., Ltd. アミド誘導体
WO2008095847A1 (de) 2007-02-06 2008-08-14 Boehringer Ingelheim International Gmbh Bicyclische heterocyclen, diese verbindungen enthaltende arzneimittel, deren verwendung und verfahren zu ihrer herstellung
MX2009009304A (es) 2007-03-01 2009-11-18 Novartis Ag Inhibidores de cinasa pim y metodos para su uso.
CN101754965B (zh) 2007-05-21 2014-03-19 诺华股份有限公司 Csf-1r抑制剂、组合物及使用方法
ES2541107T3 (es) 2007-06-25 2015-07-16 Novartis Ag Derivados de N5-(2-etoxietil)-N3-(2-piridinil)-3,5-piperidindicarboxamida para su uso como inhibidores de renina
US8389553B2 (en) 2007-06-27 2013-03-05 Merck Sharp & Dohme Corp. 4-carboxybenzylamino derivatives as histone deacetylase inhibitors
WO2009098061A1 (de) 2008-02-07 2009-08-13 Boehringer Ingelheim International Gmbh Spirocyclische heterocyclen, diese verbindungen enthaltende arzneimittel, deren verwendung und verfahren zu ihrer herstellung
AU2009247782C1 (en) 2008-05-13 2013-09-19 Astrazeneca Ab Fumarate salt of 4- (3-chloro-2-fluoroanilino) -7-methoxy-6- { [1- (N-methylcarbamoylmethyl) piperidin- 4-yl] oxy } quinazoline
JP5539351B2 (ja) 2008-08-08 2014-07-02 ベーリンガー インゲルハイム インターナショナル ゲゼルシャフト ミット ベシュレンクテル ハフツング シクロヘキシルオキシ置換ヘテロ環、これらの化合物を含有する医薬、およびそれらを生成するための方法
EP2413932A4 (de) 2009-04-01 2012-09-19 Merck Sharp & Dohme Hemmer der akt-aktivität
US8765747B2 (en) 2009-06-12 2014-07-01 Dana-Farber Cancer Institute, Inc. Fused 2-aminothiazole compounds
US8658639B2 (en) 2009-06-24 2014-02-25 Dainippon Sumitomo Pharma Co., Ltd N-substituted-cyclic amino derivative
EP2488028B1 (de) 2009-10-14 2020-08-19 Merck Sharp & Dohme Corp. Substituierte piperidine zur erhöhung der p53-aktivität und ihre verwendung
EP2519517B1 (de) 2009-12-29 2015-03-25 Dana-Farber Cancer Institute, Inc. Typ-ii-raf-kinase-hemmer
BR112012023021A2 (pt) 2010-03-16 2016-05-31 Dana Farber Cancer Inst Inc compostos de indazol e seus usos
WO2011163330A1 (en) 2010-06-24 2011-12-29 Merck Sharp & Dohme Corp. Novel heterocyclic compounds as erk inhibitors
CN103068980B (zh) 2010-08-02 2017-04-05 瑟纳治疗公司 使用短干扰核酸(siNA)的RNA干扰介导的联蛋白(钙粘蛋白关联蛋白质),β1(CTNNB1)基因表达的抑制
KR102072631B1 (ko) 2010-08-17 2020-02-03 시르나 쎄러퓨틱스 인코퍼레이티드 짧은 간섭 핵산 (siNA)을 사용한 B형 간염 바이러스 (HBV) 유전자 발현의 RNA 간섭 매개 억제
US8883801B2 (en) 2010-08-23 2014-11-11 Merck Sharp & Dohme Corp. Substituted pyrazolo[1,5-a]pyrimidines as mTOR inhibitors
WO2012030685A2 (en) 2010-09-01 2012-03-08 Schering Corporation Indazole derivatives useful as erk inhibitors
US9242981B2 (en) 2010-09-16 2016-01-26 Merck Sharp & Dohme Corp. Fused pyrazole derivatives as novel ERK inhibitors
EP3766975A1 (de) 2010-10-29 2021-01-20 Sirna Therapeutics, Inc. Rna-interferenz-vermittelte inhibition von genexpression unter verwendung kurzer interferierender nukleinsäure (sina)
WO2012087772A1 (en) 2010-12-21 2012-06-28 Schering Corporation Indazole derivatives useful as erk inhibitors
US20140046059A1 (en) 2011-04-21 2014-02-13 Piramal Enterprises Limited Process for the preparation of morpholino sulfonyl indole derivatives
US9023865B2 (en) 2011-10-27 2015-05-05 Merck Sharp & Dohme Corp. Compounds that are ERK inhibitors
EP2822935B1 (de) 2011-11-17 2019-05-15 Dana-Farber Cancer Institute, Inc. Inhibitoren der c-jun-n-terminal-kinase (jnk)
US20150299696A1 (en) 2012-05-02 2015-10-22 Sirna Therapeutics, Inc. SHORT INTERFERING NUCLEIC ACID (siNA) COMPOSITIONS
WO2014052563A2 (en) 2012-09-28 2014-04-03 Merck Sharp & Dohme Corp. Novel compounds that are erk inhibitors
EP2909194A1 (de) 2012-10-18 2015-08-26 Dana-Farber Cancer Institute, Inc. Hemmer der cyclinabhängigen kinase 7 (cdk7)
US10000483B2 (en) 2012-10-19 2018-06-19 Dana-Farber Cancer Institute, Inc. Bone marrow on X chromosome kinase (BMX) inhibitors and uses thereof
WO2014063061A1 (en) 2012-10-19 2014-04-24 Dana-Farber Cancer Institute, Inc. Hydrophobically tagged small molecules as inducers of protein degradation
ES2651347T3 (es) 2012-11-28 2018-01-25 Merck Sharp & Dohme Corp. Composiciones y métodos para el tratamiento del cáncer
CA2895504A1 (en) 2012-12-20 2014-06-26 Merck Sharp & Dohme Corp. Substituted imidazopyridines as hdm2 inhibitors
US9540377B2 (en) 2013-01-30 2017-01-10 Merck Sharp & Dohme Corp. 2,6,7,8 substituted purines as HDM2 inhibitors
CN105073728A (zh) 2013-03-15 2015-11-18 全球血液疗法股份有限公司 化合物及其用于调节血红蛋白的用途
WO2015034925A1 (en) 2013-09-03 2015-03-12 Moderna Therapeutics, Inc. Circular polynucleotides
EP3057956B1 (de) 2013-10-18 2021-05-05 Dana-Farber Cancer Institute, Inc. Polycyclische inhibitoren der cyclin-dependent-kinase 7 (cdk7)
US20160264551A1 (en) 2013-10-18 2016-09-15 Syros Pharmaceuticals, Inc. Heteroaromatic compounds useful for the treatment of prolferative diseases
EA201992707A1 (ru) 2013-11-18 2020-06-30 Глобал Блад Терапьютикс, Инк. Соединения и их применения для модуляции гемоглобина
WO2015164614A1 (en) 2014-04-23 2015-10-29 Dana-Farber Cancer Institute, Inc. Janus kinase inhibitors and uses thereof
WO2015164604A1 (en) 2014-04-23 2015-10-29 Dana-Farber Cancer Institute, Inc. Hydrophobically tagged janus kinase inhibitors and uses thereof
JO3589B1 (ar) 2014-08-06 2020-07-05 Novartis Ag مثبطات كيناز البروتين c وطرق استخداماتها
WO2016105528A2 (en) 2014-12-23 2016-06-30 Dana-Farber Cancer Institute, Inc. Inhibitors of cyclin-dependent kinase 7 (cdk7)
US10550121B2 (en) 2015-03-27 2020-02-04 Dana-Farber Cancer Institute, Inc. Inhibitors of cyclin-dependent kinases
WO2016201370A1 (en) 2015-06-12 2016-12-15 Dana-Farber Cancer Institute, Inc. Combination therapy of transcription inhibitors and kinase inhibitors
EP4019515A1 (de) 2015-09-09 2022-06-29 Dana-Farber Cancer Institute, Inc. Hemmer von cyclinabhängigen kinasen
JOP20190055A1 (ar) 2016-09-26 2019-03-24 Merck Sharp & Dohme أجسام مضادة ضد cd27
US10851164B2 (en) 2017-04-13 2020-12-01 Aduro Biotech Holdings, Europe B.V. Anti-SIRPα antibodies
US10947234B2 (en) 2017-11-08 2021-03-16 Merck Sharp & Dohme Corp. PRMT5 inhibitors
WO2019148412A1 (en) 2018-02-01 2019-08-08 Merck Sharp & Dohme Corp. Anti-pd-1/lag3 bispecific antibodies
WO2020033282A1 (en) 2018-08-07 2020-02-13 Merck Sharp & Dohme Corp. Prmt5 inhibitors
WO2020033284A1 (en) 2018-08-07 2020-02-13 Merck Sharp & Dohme Corp. Prmt5 inhibitors

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0197840A1 (de) * 1985-03-26 1986-10-15 Institut National De La Sante Et De La Recherche Medicale (Inserm) (Imidazolyl-4)-piperidine, ihre Herstellung und Anwendung als Heilmittel
WO1991009030A1 (en) * 1989-12-12 1991-06-27 Rhone-Poulenc Sante 2-substituted 4,5-diphenyl-imidazoles
WO1993012107A1 (en) * 1991-12-18 1993-06-24 Schering Corporation Imidazoylalkyl substituted with a six membered nitrogen containing heterocyclic ring
WO1995000507A1 (en) * 1993-06-22 1995-01-05 Knoll Ag Azolyl-cyclic amine derivatives with immunomodulatory activity
EP0675112A1 (de) * 1994-03-31 1995-10-04 Bristol-Myers Squibb Company Imidazol-enthaltende Farnesyl-Protein-Transferase-Inhibitoren

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB889225A (de) * 1959-07-01
FR2710066B1 (fr) * 1993-09-14 1995-10-20 Synthelabo Dérivés de 1-[2-amino-5-[1-(triphénylméthyl-1H-imidazol-4-yl]-1-oxopentyl]piperidine, leur préparation et leur utilisation comme intermédiaires de synthèse.

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0197840A1 (de) * 1985-03-26 1986-10-15 Institut National De La Sante Et De La Recherche Medicale (Inserm) (Imidazolyl-4)-piperidine, ihre Herstellung und Anwendung als Heilmittel
WO1991009030A1 (en) * 1989-12-12 1991-06-27 Rhone-Poulenc Sante 2-substituted 4,5-diphenyl-imidazoles
WO1993012107A1 (en) * 1991-12-18 1993-06-24 Schering Corporation Imidazoylalkyl substituted with a six membered nitrogen containing heterocyclic ring
WO1995000507A1 (en) * 1993-06-22 1995-01-05 Knoll Ag Azolyl-cyclic amine derivatives with immunomodulatory activity
EP0675112A1 (de) * 1994-03-31 1995-10-04 Bristol-Myers Squibb Company Imidazol-enthaltende Farnesyl-Protein-Transferase-Inhibitoren

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See also references of WO9718813A1 *

Also Published As

Publication number Publication date
JP2000500502A (ja) 2000-01-18
EP0862435A4 (de) 1999-02-03
WO1997018813A1 (en) 1997-05-29
CA2238081A1 (en) 1997-05-29
AU1162697A (en) 1997-06-11
AU704139B2 (en) 1999-04-15

Similar Documents

Publication Publication Date Title
EP0862435A1 (de) Inhibitoren der farnesyl-protein-transferase
US5817678A (en) Inhibitors of farnesyl-protein transferase
US6127366A (en) Inhibitors of farnesyl-protein transferase
US5756528A (en) Inhibitors of farnesyl-protein transferase
US6066738A (en) Inhibitors of farnesyl-protein transferase
US5914341A (en) Inhibitors of farnesyl-protein transferase
US5872135A (en) Inhibitors of farnesyl-protein transferase
US5968965A (en) Inhibitors of farnesyl-protein transferase
WO1996030343A1 (en) Inhibitors of farnesyl-protein transferase
WO1997036889A1 (en) Inhibitors of farnesyl-protein transferase
EP0841919A1 (de) Inhibitoren der farnesyl-protein transferase
EP0891360A1 (de) Inhibitoren der farnesyl-protein transferase
US5780492A (en) Inhibitors of farnesyl-protein transferase
EP0897303A1 (de) Inhibitoren von farnesylprotein-transferase
AU715667B2 (en) Inhibitors of farnesyl-protein transferase
EP0880320A1 (de) Inhibitoren der farnesyl-protein transferase
EP0891352A1 (de) Inhibitoren der farnesyl-protein transferase
WO1996034010A2 (en) Inhibitors of farnesyl-protein transferase
WO1997036593A1 (en) Inhibitors of farnesyl-protein transferase
EP0891353A1 (de) Farnesyl-protein transferase inhibitoren
US5627202A (en) Inhibitors of farnesyl-protein transferase
US5972984A (en) Inhibitors of farnesyl-protein transferase
AU2555797A (en) Inhibitors of farnesyl-protein transferase
WO1997027853A1 (en) Inhibitors of farnesyl-protein transferase
EP0900081A1 (de) Inhibitoren der 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: 19980622

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

A4 Supplementary search report drawn up and despatched
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

RIN1 Information on inventor provided before grant (corrected)

Inventor name: CICCARONE, TERRENCE, M.

Inventor name: DESOLMS, S., JANE

Inventor name: GRAHAM, SAMUEL, L.

Inventor name: SHAW, ANTHONY, W.

Inventor name: KIM, BYEONG, M.

17Q First examination report despatched

Effective date: 20001006

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

18D Application deemed to be withdrawn

Effective date: 20010418