WO1997036901A1 - Inhibitors of farnesyl-protein transferase - Google Patents

Inhibitors of farnesyl-protein transferase Download PDF

Info

Publication number
WO1997036901A1
WO1997036901A1 PCT/US1997/005304 US9705304W WO9736901A1 WO 1997036901 A1 WO1997036901 A1 WO 1997036901A1 US 9705304 W US9705304 W US 9705304W WO 9736901 A1 WO9736901 A1 WO 9736901A1
Authority
WO
WIPO (PCT)
Prior art keywords
substituted
alkyl
unsubstituted
aryl
hydrogen
Prior art date
Application number
PCT/US1997/005304
Other languages
French (fr)
Inventor
Neville J. Anthony
Robert P. Gomez
Samuel L. Graham
Original Assignee
Merck & Co., Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from GBGB9613462.2A external-priority patent/GB9613462D0/en
Priority claimed from GBGB9617277.0A external-priority patent/GB9617277D0/en
Application filed by Merck & Co., Inc. filed Critical Merck & Co., Inc.
Priority to EP97920000A priority Critical patent/EP0891361A1/en
Priority to JP9535534A priority patent/JP2000507590A/en
Priority to AU24301/97A priority patent/AU706150B2/en
Publication of WO1997036901A1 publication Critical patent/WO1997036901A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D233/00Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings
    • C07D233/54Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings having two double bonds between ring members or between ring members and non-ring members
    • C07D233/56Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings having two double bonds between ring members or between ring members and non-ring members with only hydrogen atoms or radicals containing only hydrogen and carbon atoms, attached to ring carbon atoms
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • 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
    • C07D233/54Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings having two double bonds between ring members or between ring members and non-ring members
    • C07D233/64Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings having two double bonds between ring members or between ring members and non-ring members with substituted hydrocarbon radicals attached to ring carbon atoms, e.g. histidine
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D233/00Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings
    • C07D233/54Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings having two double bonds between ring members or between ring members and non-ring members
    • C07D233/66Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings having two double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D233/70One oxygen atom
    • 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
    • C07D401/06Heterocyclic 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 carbon chain containing only aliphatic carbon atoms
    • 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
    • C07D403/10Heterocyclic 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 aromatic rings
    • 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
    • C07D403/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/06Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/06Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing three or more hetero rings

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, Ki4a-ras, Ki4b-ras and N-ras) are found in many human cancers, including colorectal carcinoma, exocrine pancreatic carcinoma, and myeloid leukemias. The protein products of these genes are defective in their GTPase activity and constitutively
  • 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 1 -Aaa 2 -Xaa” box (Cys is cysteine, Aaa is an aliphatic amino acid, the Xaa is any amino acid) (Willumsen et ai. Nature 310:583-586 (1984)).
  • this motif serves as a signal sequence for the enzymes farnesyl-protein transferase or geranylgeranyl-protein transferase, which catalyze the alkylation of the cysteine residue of the CAAX motif with a C 15 or C 20 isoprenoid, respectively.
  • the Ras protein is one of several proteins that are known to undergo post-translational farnesyl- ation.
  • 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 structure and function in addition to those listed above.
  • FPTase farnesyl-protein transferase
  • FPP farnesyl diphosphate
  • Ras protein substrates
  • Bisubstrate inhibitors and inhibitors of farnesyl-protein transferase that are non-competitive with the substrates have also been described.
  • 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 farnesyl-protein transferase enzyme, or may be purely competitive inhibitors (U.S. Patent 5,141 ,851 , University of Texas; N.E. Kohl et al., Science, 260: 1934-1937 (1993); Graham, et al., J. Med. Chem., 37, 725 (1994)).
  • 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.
  • farnesyl-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).
  • the present invention comprises arylheteroaryl- containing compounds which inhibit the farnesyl-protein transferase. Further contained in this invention are chemotherapeutic compositions containing these farnesyl transferase inhibitors and methods for their production.
  • the compounds of this invention are useful in the inhibition of farnesyl-protein transferase and the farnesylation of the oncogene protein Ras.
  • the inhibitors of farnesyl-protein transferase are illustrated by the formula A:
  • R 1 and R 2 are independently selected from:
  • substituent on the substituted C 1 -C 6 alkyl is selected from unsubstituted or substituted aryl, heterocyclic, C 3 -C 10 cycloalkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl,
  • R 3 , R 4 and R 5 are independently selected from:
  • substituted C 1 -C 6 alkyl is selected from unsubstituted or substituted aryl, unsubstituted or substituted heterocyclic, C 3 -C 10 cycloalkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl,
  • R 12 O-, R 11 S(O) m -, R 10 C(O)NR 10 -, (R 10 ) 2 NC(O)-, R 10 2 N-C(NR 10 )-, CN, R 10 C(O)-, N 3 , -N(R 10 ) 2 , and R 11 OC(O)-NR 10 -;
  • R 6a , R 6b , R 6c , R 6d and R 6e are independently selected from:
  • substituted C 1 -C 6 alkyl is selected from unsubstituted or substituted aryl, unsubstituted or substituted heterocyclic, C 3 -C 10 cycloalkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl,
  • R 7 is selected from: H; C 1 -4 alkyl, C 3-6 cycloalkyl, heterocycle, aryl aroyl, heteroaroyl, arylsulfonyl, heteroarylsulfonyl, unsubstituted or substituted with:
  • R 8 is independently selected from:
  • cyanophenyl heterocycle, C 3 -C 10 cycloalkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, perfluoroalkyl, F, Cl, Br, R 10 O-, R 1 1 S(O) m -, R 10 C(O)NH-, (R 10 ) 2 NC(O)-, R 10 2 N-C(NR 10 )-, CN, R 10 C(O)-, N 3 , -N(R 10 ) 2 , or R 10 OC(O)NH-; provided that when R 8 is heterocycle, attachment of R 8 to V is through a substitutable ring carbon;
  • R 9 is independently selected from:
  • R 10 is independently selected from hydrogen, C 1 -C 6 alkyl, benzyl,
  • R 11 is independently selected from C 1 -C 6 alkyl and aryl
  • R 1 2 is independently selected from hydrogen, C 1 -C 6 alkyl, C 1 -C 6
  • aralkyl C 1 -C 6 substituted aralkyl, C 1 -C 6 heteroaralkyl, C 1 -C 6 substituted heteroaralkyl, aryl, substituted aryl, heteroaryl, substituted heteraryl, C 1 -C 6 perfluoroalkyl, 2-aminoethyl and 2,2,2-trifluoroethyl;
  • V is selected from:
  • aryl d) C 1 -C 20 alkyl wherein from 0 to 4 carbon atoms are replaced with a heteroatom selected from O, S, and N, and e) C 2 -C 20 alkenyl,
  • 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 A 2 is S(O) m ;
  • V when V is heterocycle, attachment of V to R 8 and to A 1 is through a substitutable ring carbon;
  • W is a heterocycle
  • n is independently 0, 1 , 2, 3 or 4;
  • p is independently 0, 1 , 2, 3 or 4;
  • q 0, 1 , 2 or 3;
  • r is 0 to 5, provided that r is 0 when V is hydrogen; and t is 0 or 1 ; or the pharmaceutically acceptable salts thereof.
  • f(s) are independently N or N->O, and the remaining f's are independently CH;
  • R 1 is independently selected from: hydrogen, C 3 -C 10 cycloalkyl, R 10 O-, -N(R 10 ) 2 , F or C 1 -C 6 alkyl;
  • R 2 is independently selected from:
  • substituent on the substituted C 1 -C 6 alkyl is selected from unsubstituted or substituted aryl, heterocycle, C 3 -C 10 cycloalkyl, C 2 -C 6 alkenyl, R 10 O- and -N(R 10 ) 2 ;
  • R 3 , R 4 and R 5 are independently selected from:
  • R 6a , R 6b , R 6c , R 6d and R 6e are independently selected from:
  • substituted C 1 -C 6 alkyl wherein the substituent on the substituted C 1 -C 6 alkyl is selected from unsubstituted or substituted aryl, unsubstituted or substituted heterocyclic, C 3 -C 10 cycloalkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, R 12 O-, R 11 S(O) m -, R 10 C(O)NR 10 -, (R 10 ) 2 NC(O)-, R 10 2 N-C(NR 10 )-, CN,
  • R 7 is selected from: H; C 1 -4 alkyl, C 3-6 cycloalkyl, heterocycle, aryl, aroyl, heteroaroyl, arylsulfonyl, heteroarylsulfonyl, unsubstituted or substituted with:
  • R 8 is independently selected from:
  • aryl, substituted aryl, heterocycle C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 1 -C 6 perfluoroalkyl, F, Cl, R 10 O-, R 10 C(O)NR 10 -, CN, NO 2 , (R 10 ) 2 N-C(NR 10 )-, R 10 C(O)-, -N(R 10 ) 2 , or R 11 OC(O)NR 10 -, and c) C 1 -C 6 alkyl substituted by C 1 -C 6 perfluoroalkyl,
  • R 10 O-, R 10 C(O)NR 10 -, (R 10 ) 2 N-C(NR 10 )-, R 10 C(O)-, -N(R 10 ) 2 , or R 11 OC(O)NR 10 -; provided that when R 8 is heterocycle, attachment of R 8 to V is through a substitutable ring carbon;
  • R 9 is selected from:
  • R 10 is independently selected from hydrogen, C 1 -C 6 alkyl, benzyl,
  • R 11 is independently selected from C 1 -C 6 alkyl and aryl
  • R 12 is independently selected from hydrogen, C 1 -C 6 alkyl, C 1 -C 6 aralkyl, C 1 -C 6 substituted aralkyl, C 1 -C 6 heteroaralkyl, C 1 -C 6 substituted heteroaralkyl, aryl, substituted aryl, heteroaryl, substituted heteraryl, C 1 -C 6 perfluoroalkyl,
  • V is selected from:
  • heterocycle selected from pyrrolidinyl, imidazolyl,
  • 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 A 2 is S(O) m ;
  • W is a heterocycle selected from pyrrolidinyl, imidazolyl, imidazolinyl, pyridinyl, thiazolyl, oxazolyl, indolyl, quinolinyl, triazolyl or
  • n is independently 0, 1 , 2, 3 or 4;
  • r is 0 to 5, provided that r is 0 when V is hydrogen; and t is 0 or 1 ; or the pharmaceutically acceptable salts thereof.
  • f(s) are independently N or N->O, and the remaining f's are independently CH;
  • R 1 is selected from: hydrogen, C 3 -C 10 cycloalkyl, R 10 O-, -N(R 10 ) 2 , F or C 1 -C 6 alkyl;
  • R 2 is independently selected from:
  • substituent on the substituted C 1 -C 6 alkyl is selected from unsubstituted or substituted aryl, heterocycle, C 3 -C 10 cycloalkyl, C 2 -C 6 alkenyl, R 10 O- and -N(R 10 ) 2 ;
  • R 3 and R 4 are independently selected from:
  • substituted C 1 -C 6 alkyl wherein the substituent on the substituted C 1 -C 6 alkyl is selected from unsubstituted or substituted aryl, unsubstituted or substituted heterocyclic, C 3 -C 10 cycloalkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, R 12 O-, R 1 1 S(O) m -, R 10 C(O)NR 10 -, (R 10 ) 2 NC(O)-, R 10 2 N-C(NR 10 )-, CN, R 10 C(O)-, N 3 , -N(R 10 ) 2 , and R 11 OC(O)-NR 10 -;
  • R 6a , R 6b , R 6c , R 6d and R 6e are independently selected from:
  • R 12 O-, R 11 S(O) m -, R 10 C(O)NR 10 -, (R 10 ) 2 NC(O)-, R 10 2 N-C(NR 10 )., CN, NO 2 , R 10 C(O)-, N 3 , -N(R 10 ) 2 , or R 11 OC(O)NR 10 -,
  • R 8 is independently selected from:
  • R 8 when R 8 is heterocycle, attachment of R 8 to V is through a substitutable ring carbon;
  • R 9a and R 9b are independently hydrogen, C 1 -C 6 alkyl, trifluoromethyl and halogen;
  • R 10 is independently selected from hydrogen, C 1 -C 6 alkyl, benzyl,
  • R 1 1 is independently selected from C 1 -C 6 alkyl and aryl
  • R 12 is independently selected from hydrogen, C 1 -C 6 alkyl, C 1 -C 6
  • aralkyl C 1 -C 6 substituted aralkyl, C 1 -C 6 heteroaralkyl, C 1 -C 6 substituted heteroaralkyl, aryl, substituted aryl, heteroaryl, substituted heteraryl, C 1 -C 6 perfluoroalkyl, 2-aminoethyl and 2,2,2-trifluoroethyl;
  • V is selected from:
  • heterocycle selected from pyrrolidinyl, imidazolyl,
  • 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 A 2 is S(O) m ;
  • V is heterocycle, attachment of V to R 8 and to A 1 is through a substitutable ring carbon;
  • m is 0, 1 or 2;
  • n is independently 0, 1 , 2, 3 or 4;
  • p 0, 1 , 2, 3 or 4;
  • r is 0 to 5, provided that r is 0 when V is hydrogen; or the pharmaceutically acceptable salts thereof.
  • R 1 is selected from: hydrogen, C 3 -C 10 cycloalkyl, R 10 O-, -N(R 10 ) 2 , F or C 1 -C 6 alkyl;
  • R 2 is independently selected from:
  • substituent on the substituted C 1 -C 6 alkyl is selected from unsubstituted or substituted aryl, heterocycle, C 3 -C 10 cycloalkyl, C 2 -C 6 alkenyl, R 10 O- and -N(R 10 ) 2 ;
  • R 3 and R 4 are independently selected from:
  • R 6a , R 6b , R 6c , R 6d and R 6e are independently selected from:
  • R 8 is independently selected from:
  • R 8 when R 8 is heterocycle, attachment of R 8 to V is through a substitutable ring carbon;
  • R 9a and R 9b are independently hydrogen, C 1 -C 6 alkyl, trifluoromethyl and halogen;
  • R 10 is independently selected from hydrogen, C 1 -C 6 alkyl, benzyl,
  • R 11 is independently selected from C 1 -C 6 alkyl and aryl;
  • R 12 is independently selected from hydrogen, C 1 -C 6 alkyl, C 1 -C 6
  • aralkyl C 1 -C 6 substituted aralkyl, C 1 -C 6 heteroaralkyl, C 1 -C 6 substituted heteroaralkyl, aryl, substituted aryl, heteroaryl, substituted heteraryl, C 1 -C 6 perfluoroalkyl,
  • V is selected from:
  • 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 A 2 is S(O) m ;
  • V when V is heterocycle, attachment of V to R 8 and to A 1 is through a substitutable ring carbon;
  • n is independently 0, 1 , 2, 3 or 4;
  • p is 0, 1 , 2, 3 or 4, provided that p is not 0 if X is a bond or O;
  • r is 0 to 5, provided that r is 0 when V is hydrogen; or the pharmaceutically acceptable salts thereof.
  • R 1 is selected from: hydrogen, C 3 -C 10 cycloalkyl or C 1 -C 6 alkyl;
  • R 2 is independently selected from:
  • R 3 is selected from:
  • substituted C 1 -C 6 alkyl is selected from unsubstituted or substituted aryl, unsubstituted or substituted heterocyclic, C 3 -C 10 cycloalkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl,
  • R 12 O-, R 11 S(O) m -, R 10 C(O)NR 10 -, (R 10 ) 2 NC(O)-, R 10 2 N-C(NR 10 )-, CN, R 10 C(O)-, N 3 , -N(R 10 ) 2 , and R 11 OC(O)-NR 10 -;
  • R 4 is selected from H, halogen, C 1 -C 6 alkyl and CF 3 ;
  • R 6a , R 6b , R 6c , R6d and R6e are independently selected from: a) hydrogen,
  • R 8 is independently selected from:
  • R 9a and R 9b are independently hydrogen, halogen, CF 3 or methyl;
  • R 10 is independently selected from hydrogen, C 1 -C 6 alkyl, benzyl,
  • R 11 is independently selected from C 1 -C 6 alkyl and aryl;
  • R 12 is independently selected from hydrogen, C 1 -C 6 alkyl, C 1 -C 6 aralkyl, C 1 -C 6 substituted aralkyl, C 1 -C 6 heteroaralkyl, C 1 -C 6 substituted heteroaralkyl, aryl, substituted aryl, heteroaryl, substituted heteraryl, C 1 -C 6 perfluoroalkyl, 2-aminoethyl and 2,2,2-trifluoroethyl;
  • a 1 is selected from: a bond, -C(O)-, O, -N(R 10 )-, or S(O) m ;
  • n 0, 1 or 2;
  • p is 0, 1 , 2, 3 or 4; or the pharmaceutically acceptable salts thereof.
  • the inhibitors of farnesyl-protein transferase are illustrated by the formula E: wherein: from 1 -2 of f(s) are independently N or N->O, and the remaining f's are independently CH;
  • R 1 is selected from: hydrogen, C 3 -C 10 cycloalkyl, R 10 O-, -N(R 10 ) 2 , F or C 1 -C 6 alkyl;
  • R 2 is independently selected from:
  • R 3 is selected from:
  • R 10 2 N-C(NR 10 )-, CN, R 10 C(O)-, N 3 , -N(R 10 ) 2 , and R 1 1 OC(O)-NR 10 -;
  • R 4 is selected from H, halogen, C 1 -C 6 alkyl and CF 3 ;
  • R 6a , R 6b , R 6c ; R 6d and R 6e are independently selected from:
  • R 8 is independently selected from:
  • R 8 when R 8 is heterocycle, attachment of R 8 to V is through a substitutable ring carbon;
  • R 9a and R 9b are independently hydrogen, halogen, CF 3 or methyl;
  • R 10 is independently selected from hydrogen, C 1 -C 6 alkyl, benzyl,
  • R 11 is independently selected from C 1 -C 6 alkyl and aryl;
  • R 12 is independently selected from hydrogen, C 1 -C 6 alkyl, C 1 -C 6
  • aralkyl C 1 -C 6 substituted aralkyl, C 1 -C 6 heteroaralkyl, C 1 -C 6 substituted heteroaralkyl, aryl, substituted aryl, heteroaryl, substituted heteraryl, C 1 -C 6 perfluoroalkyl, 2-aminoethyl and 2,2,2-trifIuoroethyl;
  • n 0, 1 or 2;
  • p is 0, 1 , 2, 3 or 4, provided that p is not 0 if X is a bond or O; or the pharmaceutically acceptable salts thereof.
  • R 1 is selected from: hydrogen, C 3 -C 10 cycloalkyl or C 1 -C 6 alkyl;
  • R 2 is independently selected from:
  • R 3 is selected from:
  • substituted C 1 -C 6 alkyl wherein the substituent on the substituted C 1 -C 6 alkyl is selected from unsubstituted or substituted aryl, unsubstituted or substituted heterocyclic, C 3 -C 10 cycloalkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl,
  • R 4 is selected from H, halogen, CH 3 and CF 3 ;
  • R 6a , R 6b , R 6c , R 6d and R 6e are independently selected from:
  • R 3 , R 6a , R 6b , R 6c , R 6d or R 6e is through a substitutable heterocycle ring carbon
  • R 9a and R 9b are independently hydrogen, halogen, CF 3 or methyl
  • R 10 is independently selected from hydrogen, C 1 -C 6 alkyl, benzyl,
  • R 11 is independently selected from C 1 -C 6 alkyl and aryl;
  • R 1 2 is independently selected from hydrogen, C 1 -C 6 alkyl, C 1 -C 6
  • aralkyl C 1 -C 6 substituted aralkyl, C 1 -C 6 heteroaralkyl, C 1 -C 6 substituted heteroaralkyl, aryl, substituted aryl, heteroaryl, substituted heteraryl, C 1 -C 6 perfluoroalkyl,
  • p is 0, 1 , 2, 3 or 4; or the pharmaceutically acceptable salts thereof.
  • R 1 is selected from: hydrogen, C 3 -C 10 cycloalkyl, R 10 O-, -N(R 10 ) 2 , F or C 1 -C 6 alkyl;
  • R 2 is independently selected from:
  • R 3 is selected from:
  • substituted C 1 -C 6 alkyl is selected from unsubstituted or substituted aryl, unsubstituted or substituted heterocyclic, C 3 -C 10 cycloalkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl,
  • R 12 O-, R 11 S(O) m -, R 10 C(O)NR 10 -, (R 10 ) 2 NC(O)-, R 10 2 N-C(NR 10 )-, CN, R 10 C(O)-, N 3 , -N(R 10 ) 2 , and
  • R 1 1 OC(O)-NR 10 -;
  • R 4 is selected from H, halogen, CH 3 and CF 3 ;
  • R 6a , R 6b , R 6c , R 6d and R 6e are independently selected from:
  • substituted C 1 -C 6 alkyl wherein the substituent on the substituted C 1 -C 6 alkyl is selected from unsubstituted or substituted aryl, unsubstituted or substituted heterocyclic, C 3 -C 10 cycloalkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, R 12 O-, R 1 1 S(O) m -,
  • R 9a and R 9b are independently hydrogen, halogen, CF 3 or methyl;
  • R 10 is independently selected from hydrogen, C 1 -C 6 alkyl, benzyl,
  • R 11 is independently selected from C 1 -C 6 alkyl and aryl
  • R 12 is independently selected from hydrogen, C 1 -C 6 alkyl, C 1 -C 6 aralkyl, C 1 -C 6 substituted aralkyl, C 1 -C 6 heteroaralkyl, C 1 -C 6 substituted heteroaralkyl, aryl, substituted aryl, heteroaryl, substituted heteraryl, C 1 -C 6 perfluoroalkyl, 2-aminoethyl and 2,2,2-trifIuoroethyl;
  • a 1 is selected from: a bond, -C(O)-, O, -N(R 10 )-, or S(O) m ; m is 0, 1 or 2; and
  • n O or 1; or the pharmaceutically acceptable salts thereof.
  • Preferred compounds of the invention are:
  • 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 1 , R 2 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 and the alkyl portion of aralkyl and similar terms, 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.
  • cycloalkyl is intended to include non- aromatic cyclic hydrocarbon groups having the specified number of carbon atoms.
  • examples of cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl and the like.
  • Alkenyl groups include those groups having the specified number of carbon atoms and having one or several double bonds.
  • alkenyl groups include vinyl, allyl, isopropenyl, pentenyl, hexenyl, heptenyl, cyclopropenyl, cyclobutenyl, cyclopentenyl, cyclohexenyl, 1 -propenyl, 2-butenyl, 2-methyl-2-butenyl, isoprenyl, farnesyl, geranyl, geranylgeranyl and the like.
  • Alkynyl groups include those groups having the specified number of carbon atoms and having one triple bonds. Examples of alkynyl groups include acetylene, 2-butynyl, 2-pentynyl, 3-pentynyl and the like.
  • Halogen or "halo” as used herein means fluoro, chloro, bromo and iodo.
  • aryl and the aryl portion of aroyl and aralkyl, 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 structure.
  • heterocyclic elements include, but are not limited to, azepinyl, benzimidazolyl, benzisoxazolyl, benzofurazanyl, benzopyranyl, benzothiopyranyl, benzofuryl,
  • quinolinyl quinoxalinyl, tetrahydrofuryl, tetrahydroisoquinolinyl, tetrahydroquinolinyl, thiamorpholinyl, thiamorpholinyl sulfoxide, thiazolyl, thiazolinyl, thienofuryl, thienothienyl, and thienyl.
  • 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,
  • the substituted group is intended to mean a substituted Cl-8 alkyl, substituted C 2-8 alkenyl, substituted C 2-8 alkynyl, substituted aryl or substituted heterocycle from which the substituent(s) R 3 , R 4 , R 5 and R 6a-e are selected.
  • the substituted C 1 -8 alkyl, substituted C 3-6 cycloalkyl, substituted aroyl, substituted aryl, substituted heteroaroyl, substituted arylsulfonyl, substituted heteroarylsulfonyl and substituted heterocycle include moieties containing from 1 to 3 substituents in addition to the point of attachment to the rest of the compound.
  • substituted aryl substituted heterocycle
  • substituted cycloalkyl are intended to include the cyclic group which is substituted on a substitutable ring carbon atom with 1 or 2 substitutents selected from the group which includes but is not limited to F, Cl, Br, CF 3 , NH 2 , N(C 1 -C 6 alkyl) 2 , NO 2 , CN, (C 1 -C 6 alkyl)O-, -OH, (C 1 -C 6 alkyl)S(O) m -, (C 1 -C 6 alkyl)C(O)NH-, H 2 N-C(NH)-, (C 1 -C 6
  • Lines drawn into the ring systems from substituents means that the indicated bond may be attached to any of the substitutable ring carbon atoms.
  • the substituent illustrated by the structure is a simplified representation of a phenyl ring having five (5)
  • fused ring moieties may be further substituted by the remaining R 6a , R 6b , R 6c , R 6d and/or R 6e as defined
  • the moiety designated by the following structure represents an aromatic 6-membered heterocyclic ring and includes the following ring systems:
  • the aromatic 6-membered heterocyclic ring is a pyridyl ring.
  • R 1 and R 2 are independently selected from: hydrogen, R 11 C(O)O-, -N(R 10 ) 2 , R 10 C(O)NR 10 -, R 10 O- or unsubstituted or substituted C 1 -C 6 alkyl wherein the substituent on the substituted C 1 -C 6 alkyl is selected from unsubstituted or substituted phenyl, -N(R 10 ) 2 , R 10 O- and R 10 C(O)NR 10 -.
  • R 3 is selected from:
  • substituted C 1 -C 6 alkyl is selected from unsubstituted or substituted aryl, unsubstituted or substituted heterocyclic, C 3 -C 10 cycloalkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl,
  • R 12 O-, R 11 S(O) m -, R 10 C(O)NR 10 -, (R 10 ) 2 NC(O)-, R 10 2 N-C(NR 10 )-, CN, R 10 C(O)-, N 3 , -N(R 10 ) 2 , and R 11 OC(O)-NR 10 -.
  • R 4 is selected from: hydrogen, halogen, trifluoromethyl, trifluoromethoxy and C 1 -C 6 alkyl.
  • R 5 is hydrogen
  • R 6a , R 6b , R 6c , R 6d and R 6e are independently selected from:
  • -CH CH-CH 2 -, -(CH 2 ) 4 - and -(CH 2 ) 3 -.
  • R 8 is independently selected from: a) hydrogen, and
  • R 9 is hydrogen, halogen, CF 3 or methyl.
  • R 10 is selected from H, C 1 -C 6 alkyl and benzyl.
  • a 1 and A 2 are independently .selected from: a bond, -C(O)NR 10 -, -NR 10 C(O)-, O, -N(R 10 )-, -S(O) 2 N(R 10 )- and-
  • V is selected from hydrogen, heterocycle and aryl. More preferably, V is 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.
  • s is 0.
  • t is 1.
  • any substituent or variable e.g., R 1 , R 2 , R 9 , n, etc.
  • -N(R 10 ) 2 represents -NHH, -NHCH 3 , -NHC 2 H 5 , 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 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 -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.
  • Schemes 1 -12 illustrate synthesis of the instant aryl- heteroaryl compound which incorporate a preferred benzylimidazolyl sidechain.
  • a arylheteroaryl intermediate that is not commercially available may be synthesized by methods known in the art.
  • a suitably substituted phenyl boronic acid I may be reacted under Suzuki coupling conditions (Pure Appl.
  • a suitably substituted halogenated nicotinic acid such as 4-bromonicotinic acid
  • the acid may be reduced and the triflate of the intermediate alcohol III may be formed in situ and coupled to a suitably substituted benzylimidazolyl IV to provide, after deprotection, the instant compound V.
  • Schemes 2-4 illustrate other methods of synthesizing the key alcohol intermediates, which can then be processed as described in Scheme 1.
  • Scheme 2 illustrates the analogous series of arylheteroaryl alcohol forming reactions starting with the methyl nicotinate boronic acid and the "terminal" phenyl moiety employed in the Suzuki coupling as the halogenated reactant.
  • Such a coupling reaction is also compatible when one of the reactants incorporates a suitably protected hydroxyl functionality as illustrated in Scheme 3.
  • Negishi chemistry (Org. Synth., 66:67 (1988)) may also be employed to form the arylheteroaryl component of the instant compounds, as shown in Scheme 4.
  • a suitably substituted zinc bromide adduct may be coupled to a suitably substituted heteroaryl halide in the presence of nickel (II) to provide the arylheteroaryl VII.
  • the heteroaryl halide and the zinc bromide adduct may be selected based on the availability of the starting reagents.
  • Scheme 5 illustrates the preparation of a suitably substituted biphenylmethyl bromide which could also be utilized in the reaction with the protected imidazole as described in Scheme 1.
  • a suitably substituted imidazole may first be alkylated with a suitably substituted benzyl halide to provide intermediate VIII.
  • Scheme 7 illustrates synthesis of an instant compound wherein a non-hydrogen R 9b is incorporated in the instant compound.
  • a readily available 4-substituted imidazole IX may be selectively iodinated to provide the 5-iodoimidazole X. That imidazole may then be protected and coupled to a suitably substituted benzyl moiety to provide intermediate XI. Intermediate XI can then undergo the alkylation reactions that were described hereinabove.
  • Scheme 8 illustrates synthesis of instant compounds that incorporate a preferred imidazolyl moiety connected to the biaryl via an alkyl amino, sulfonamide or amide linker.
  • the 4-aminoalkyl- imidazole XII wherein the primary amine is protected as the phthali- mide, is selectively alkylated then deprotected to provide the amine XIII.
  • the amine XIII may then react under conditions well known in the art with various activated arylheteroaryl moieties to provide the instant compounds shown.
  • the suitably substituted phenol XIV may be reacted with methyl N-(cyano)methanimidate to provide the 4-phenoxyimidazole XV.
  • the intermediate XVI can undergo alkylation reactions as described for the benzylimidazoles hereinabove.
  • Scheme 10 illustrates an analogous series of reactions wherein the (CR 2 2 ) ⁇ X(CR 2 2 ) p linker of the instant compounds is oxygen.
  • a suitably substituted halopyridinol such as 3-chloro- 2-pyridinol
  • Intermediate XVI is then protected and, if desired to form a compound of a preferred embodiment, alkylated with a suitably protected benzyl.
  • the intermediate XVII can then be coupled to a aryl moiety by Suzuki chemistry to provide the instant compound.
  • a 1 (CR 1 2 ) n A 2 (CR 1 2 ) n linker is a substituted methylene may be synthesized by the methods shown in Scheme 1 1.
  • the N-protected imidazolyl iodide XVIII is reacted, under Grignard conditions with a suitably protected benzaldehyde to provide the alcohol XIX.
  • Acylation, followed by the alkylation procedure illustrated in the Schemes above (in particular, Scheme 1) provides the instant compound XX. If other R 1 substituents are desired, the acetyl moiety can be manipulated as illustrated in the Scheme.
  • the final product XXII may be isolated in the salt form, for example, as a trifluoroacetate, hydrochloride or acetate salt, among others.
  • the product diamine XXII can further be selectively protected to obtain XXIII, which can subsequently be reductively alkylated with a second aldehyde to obtain XXIV. Removal of the protecting group, and conversion to cyclized products such as the dihydroimidazole XXV can be accomplished by literature procedures.
  • the arylheteroaryl subunit reagent is reacted with an aldehyde which also has a protected hydroxyl group, such as XXVI in Scheme 14, the protecting groups can be subsequently removed to unmask the hydroxyl group (Schemes 14, 15).
  • 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 alkyl lithium reagents, to obtain secondary alcohols such as XXX.
  • the Boc protected amino alcohol XXVIII can also be utilized to synthesize 2-aziridinylmethylarylheteroaryl such as XXXIII (Scheme 16). Treating XXVIII with 1 ,1 '-sulfonyldiimidazole and sodium hydride in a solvent such as dimethylformamide led to the formation of aziridine XXXIII . The aziridine is reacted with a nucleophile, such as a thiol, in the presence of base to yield the ring- opened product XXXIV .
  • a nucleophile such as a thiol
  • arylheteroaryl subunit reagent can be reacted with aldehydes derived from amino acids such as O-alkylated tyrosines, according to standard procedures, to obtain compounds such as XL, as shown in Scheme 17.
  • R' is an aryl group
  • XL can first be hydrogenated to unmask the phenol, and the amine group deprotected with acid to produce XLI.
  • the amine protecting group in XL can be removed, and O-alkylated phenolic amines such as XLII 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.,
  • NF- 1 neurofibromin
  • the compounds of the instant invention inhibit farnesyl- protein transferase and the farnesylation of the oncogene protein Ras.
  • the instant compounds may also inhibit tumor angiogenesis, thereby affecting the growth of tumors (J. Rak et al. Cancer Research,
  • 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. Schafmer et al. American Journal of Pathology, 142: 1051-1060 (1993) and B. Cowley, Jr. et al.FASEB Journal, 2:A3160 (1988)).
  • the instant compounds may also be useful for the treatment of fungal infections.
  • the 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 corn 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.
  • 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 alternatively 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
  • 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.
  • pharmacolo- gically acceptable carriers e.g., saline
  • 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 farnesyl-protein transferase (FPTase) in a composition.
  • FPTase farnesyl-protein transferase
  • mixtures which comprise a known substrate of FPTase (for example a tetrapeptide having a cysteine at the amine terminus) and farnesyl pyrophosphate and, in one of the mixtures, a compound of the instant invention.
  • FPTase for example a tetrapeptide having a cysteine at the amine terminus
  • farnesyl pyrophosphate for example a tetrapeptide having a cysteine at the amine terminus
  • the chemical content of the assay mixtures may be determined by well known
  • 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 farnesyl- protein transferase, an excess amount of a known substrate of FPTase (for example a tetrapeptide having a cysteine at the amine terminus) and farnesyl 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 1 -Trityl-4-(4-cyanobenzyl)-imidazole
  • Step E 1 -(2-Phenylpyrid-5-ylmethyl)-5-(4-cyanobenzyI)
  • Step B 3-Phenyl-6-hydroxymethylpyridine
  • 3-phenyl-6-carboxypyridine 1.05g, 5.27 mmol
  • tetrahydrofuran 25 mL
  • 1.0 M lithium aluminum hydride in tetrahydrofuran
  • the reaction was allowed to stir at ambient temperature for 6 hours, cooled to 0°C, and quenched by dropwise addition of water (0.50 mL), 4 N aq. NaOH (0.50 mL), and water (1.5 mL).
  • Step C 1 -(3-Phenylpyrid-6-ylmethyl)-5-(4-cyanobenzyl)
  • Step B 2-(3-Trifluoromethoxyphenyl)-5-carboxy pyridine
  • Step D 1 -(2-(3-Trifluoromethoxyphenyl)-pyrid-5-ylmethyl)-
  • Step D 1 -(2-(2-Trifluoromethylphenyl)-pyrid-5-ylmethyI)-
  • Step B 3-Phenyl-2-chloro-6-methylpyridine and 3-phenyI-4- chloro-6-methylpyridine
  • Step D 1 -(3-Phenyl-2-chloropyrid-6-ylmethyl)-5-(4- cyanobenzyl)imidazole hydrochloride salt
  • Step B N-bis t-Butoxycarbonyl-2-Amino-3-Phenyl-6- methylpyridine
  • Step C 2-(bis t-butoxycarbonylamino)-3-phenyl-6- methylpyridine-N-oxide
  • Step D N-bis t-Butoxycarbonyl-2-amino-3-phenyl-6- acetoxymethylpyridine
  • Step F 1-(2-Amino-3-phenylpyrid-6-ylmethyl)-5-(4- cyanobenzyl)imidazole hydrochloride salt
  • the title compound was prepared using the procedure described for Example 3 step C using N-bis t-butoxycarbonyl-2-amino-3-phenyl- 6-hydroxymethylpyridine in place of 3-phenyl-6-hydroxymethyl- pyridine.
  • the free base was treated with TFA and triethylsilane to effect cleavage of the t-butoxycarbonyl groups which was followed by its conversion to the hydrochloride salt.
  • Bovine FPTase was assayed in a volume of 100 ⁇ l containing 100 mM N-(2-hydroxy ethyl) piperazine-N '-(2-ethane sulfonic acid) (HEPES), pH 7.4, 5 mM MgCl 2 , 5 mM dithiothreitol (DTT), 100 mM [ 3 H]-farnesyl diphosphate ([ 3 H]-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 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.
  • 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 mCi[ 35 Sjmethionine (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 MgCl 2 /1mM 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 MgCl 2 /1mM DTT/10 mg/ml aprotinen/2 mg/ml leupeptin/2 mg/ml antipain/0.5 mM PM
  • 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 nonfarnesylated ras proteins are compared to 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
  • 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.

Landscapes

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

Abstract

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

Description

TITLE OF THE INVENTION
LNHIBITORS 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, Ki4a-ras, Ki4b-ras and N-ras) are found in many human cancers, including colorectal carcinoma, exocrine pancreatic carcinoma, and myeloid leukemias. The protein products of these genes are defective in their GTPase activity and constitutively
transmit a growth stimulatory signal.
Ras must be 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-Aaa1 -Aaa2-Xaa" box (Cys is cysteine, Aaa is an aliphatic amino acid, the Xaa is any amino acid) (Willumsen et ai. Nature 310:583-586 (1984)). Depending on the specific sequence, this motif serves as a signal sequence for the enzymes farnesyl-protein transferase or geranylgeranyl-protein transferase, which catalyze the alkylation of the cysteine residue of the CAAX motif with a C15 or C20 isoprenoid, respectively. (S. Clarke., Ann. Rev. Biochem. 61 :355-386 (1992); W.R. Schafer and J. Rine, Ann. Rev. Genetics 30:209-237 (1992)). The Ras protein is one of several proteins that are known to undergo post-translational farnesyl- ation. 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 structure and function in addition to those listed above.
Inhibition of farnesyl-protein transferase has been shown to block the growth of Ras-transformed cells in soft agar and to modify other aspects of their transformed phenotype. It has also been demonstrated that certain inhibitors of farnesyl-protein transferase selectively block the processing of the Ras oncoprotein intracellularly (N.E. Kohl et al., Science, 260: 1934- 1937 (1993) and G.L. James et al., Science, 260: 1937-1942 (1993). Recently, it has been shown that an inhibitor of farnesyl-protein transferase blocks the growth of ras-dependent tumors in nude mice (N.E. Kohl et al., Proc. Natl.
Acad. Sci U.S.A., 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 farnesyl-protein transferase in vivo has been demonstrated with lovastatin (Merck & Co., Rahway, NJ) and compactin (Hancock et al., ibid; Casey et al., ibid; Schafer et al., Science 245:379 (1989)). These drugs inhibit HMG-CoA reductase, the rate limiting enzyme for the production of polyisoprenoids including farnesyl pyrophosphate. Famesyl-protein transferase utilizes farnesyl pyrophosphate to covalently modify the Cys thiol group of the Ras CAAX box with a farnesyl group (Reiss et al., Cell, 62:81 -88 ( 1990); Schaber et al., J. Biol Chem., 265: 14701-14704 (1990); Schafer et al, Science, 249: 1 133-1 139 (1990); Marine et al, Proc. Natl. Acad. Sci USA, 87:7541 -7545 (1990)). Inhibition of farnesyl pyrophosphate biosynthesis by inhibiting HMG-CoA reductase blocks Ras membrane localization in cultured cells. However, direct inhibition of farnesyl- 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 farnesyl-protein transferase (FPTase) have been described in four general classes (S. Graham, Expert Opinion Ther. Patents, (1995) 5:1269-1285). The first are analogs of farnesyl diphosphate (FPP), while a second class of inhibitors is related to the protein substrates (e.g., Ras) for the enzyme. Bisubstrate inhibitors and inhibitors of farnesyl-protein transferase that are non-competitive with the substrates have also been described. The peptide derived inhibitors that have been described are generally cysteine containing molecules that are related to the CAAX motif that is the signal for protein prenylation. (Schaber et al., ibid; Reiss et. al., ibid; Reiss et al., PNAS, 88:732-736 (1991)). Such inhibitors may inhibit protein prenylation while serving as altemate substrates for the farnesyl-protein transferase enzyme, or may be purely competitive inhibitors (U.S. Patent 5,141 ,851 , University of Texas; N.E. Kohl et al., Science, 260: 1934-1937 (1993); Graham, et al., J. Med. Chem., 37, 725 (1994)). 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 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 farnesyl
protein transferase have also been disclosed (WO 95/09001
and EP 0 675 112 A1).
It has recently been reported that farnesyl-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 is, therefore, an object of this invention to develop low molecular weight compounds that will inhibit farnesyl-protein transferase and thus, the post-translational farnesylation 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 arylheteroaryl- containing compounds which inhibit the farnesyl-protein transferase. Further contained in this invention are chemotherapeutic compositions containing these farnesyl transferase inhibitors and methods for their production.
The compounds of this invention are illustrated by the formula A:
Figure imgf000006_0001
DETAILED DESCRIPTION OF THE INVENTION
The compounds of this invention are useful in the inhibition of farnesyl-protein transferase and the farnesylation of the oncogene protein Ras. In a first embodiment of this invention, the inhibitors of farnesyl-protein transferase are illustrated by the formula A:
Figure imgf000007_0001
wherein: from 1 -2 of f(s) are independently N or N->O, and the remaining f's are independently CH;
R1 and R2 are independently selected from:
a) hydrogen,
b) aryl, heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl,
C2-C6 alkynyl, R10O-, R11S(O)m-, R10C(O)NR10-, R11C(O)O-, (R10)2NC(O)-, R10 2N-C(NR10)-, CN, NO2, R10C(O)-, N3, -N(R10)2, or R11OC(O)NR10-,
c) unsubstituted or substituted C1 -C6 alkyl wherein the
substituent on the substituted C1 -C6 alkyl is selected from unsubstituted or substituted aryl, heterocyclic, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl,
R10O-, R11S(O)m-, R10C(O)NR10-, (R10)2NC(O)-, R10 2N-C(NR10)-, CN, R10C(O)-, N3, -N(R10)2, and R11OC(O)-NR10-;
R3, R4 and R5 are independently selected from:
a) hydrogen,
b) unsubstituted or substituted aryl, unsubstituted or
substituted heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, halogen, C1 -C6 perfluoroalkyl, R12O-, R11S(O)m-, R10C(O)NR10-, (R 10)2NC(O)-, R11C(O)O-, R10 2N-C(NR10)-, CN, NO2, R10C(O)-, N3, -N(R 10)2, or R11OC(O)NR10-,
c) unsubstituted C1 -C6 alkyl,
d) substituted C1 -C6 alkyl wherein the substituent on the
substituted C1 -C6 alkyl is selected from unsubstituted or substituted aryl, unsubstituted or substituted heterocyclic, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl,
R12O-, R11S(O)m-, R10C(O)NR10-, (R10)2NC(O)-, R10 2N-C(NR10)-, CN, R10C(O)-, N3, -N(R10)2, and R11OC(O)-NR10-;
R6a, R6b, R6c, R6d and R6e are independently selected from:
a) hydrogen,
b) unsubstituted or substituted aryl, unsubstituted or
substituted heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, halogen, C1 -C6 perfluoroalkyl, R12O-, R 1 1S(O)m-, R10C(O)NR10-, (R10)2NC(O)-, R11C(O)O-, R10 2N-C(NR10)-, CN, NO2, R10C(O)-, N3, -N(R10)2, or R11OC(O)NR10-,
c) unsubstituted C1 -C6 alkyl,
d) substituted C1 -C6 alkyl wherein the substituent on the
substituted C1 -C6 alkyl is selected from unsubstituted or substituted aryl, unsubstituted or substituted heterocyclic, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl,
R12O-, R11S(O)m-, R10C(O)NR10-, (R10)2NC(O)-, R10 2N-C(NR10)-, CN, R10C(O)-, N3, -N(R10)2, and R11OC(O)-NR10-; or any two of R6a, R6b, R6c, R6d and R6e on adjacent carbon atoms are combined to form a diradical selected from -CH=CH-CH=CH-, -CH=CH-CH2-, -(CH2)4- and -(CH2)3-; provided that when R3, R4, R5, R6a, R6b, R6c, R6d or R6e is unsubstituted or substituted heterocycle, attachment of R3, R4, R5, R6a, R6b, R6c, R6d or R6e to the 6-membered heteroaryl ring, or phenyl ring respectively, is through a substitutable heterocycle ring carbon;
R7 is 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,
Figure imgf000009_0001
f) -SO2R1 1
g) N(R 10)2 or
h) C1 -4 perfluoroalkyl;
R8 is independently selected from:
a) hydrogen,
b) aryl, substituted aryl, heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, perfluoroalkyl, F, Cl, Br, R10O-, R11S(O)m-, R10C(O)NR10-, (R10)2NC(O)-, R10 2N-C(NR10)-, CN, NO2, R10C(O)-, N3, -N(R 10)2, or R11OC(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, R10O-, R 1 1S(O)m-, R10C(O)NH-, (R10)2NC(O)-, R10 2N-C(NR10)-, CN, R10C(O)-, N3, -N(R 10)2, or R10OC(O)NH-; provided that when R8 is heterocycle, attachment of R8 to V is through a substitutable ring carbon;
R9 is independently selected from:
a) hydrogen,
b) alkenyl, alkynyl, perfluoroalkyl, F, Cl, Br,
R11O-, R11S(O)m-, R10C(O)NR10-, (R10)2NC(O)-, R10 2N-C(NR10)-, CN, NO2, R10C(O)-, N3, -N(R10)2, or R11OC(O)NR10-, and
c) C1 -C6 alkyl unsubstituted or substituted by perfluoroalkyl,
F, Cl, Br, R10O-, R11S(O)m-, R10C(O)NR10-, (R10)2NC(O)-, R10 2N-C(NR10)-, CN, R10C(O)-,
N3, -N(R10)2, or R11OC(O)NR10-; R10 is independently selected from hydrogen, C1 -C6 alkyl, benzyl,
2,2,2-trifluoroethyl and aryl; R11 is independently selected from C1 -C6 alkyl and aryl; R 1 2 is independently selected from hydrogen, C1 -C6 alkyl, C1 -C6
aralkyl, C1 -C6 substituted aralkyl, C1 -C6 heteroaralkyl, C1 -C6 substituted heteroaralkyl, aryl, substituted aryl, heteroaryl, substituted heteraryl, C1 -C6 perfluoroalkyl, 2-aminoethyl and 2,2,2-trifluoroethyl;
A 1 and A2 are independently selected from: a bond, -CH=CH-, -CΞC-,
-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 heteroatom selected from O, S, and N, and e) C2-C20 alkenyl,
provided that V is not hydrogen if A1 is S(O)m and V is not hydrogen if A1 is a bond, n is 0 and A2 is S(O)m;
provided that when V is heterocycle, attachment of V to R8 and to A1 is through a substitutable ring carbon;
W is a heterocycle;
X is a bond, -CH=CH-, O, -C(=O)-, -C(O)NR7-, -NR7C(O)-, -C(O)O-,
-OC(O)-, -C(O)NR7C(O)-, -NR7-, -S(O)2N(R10)-,
-N(R10)S(O)2- or -S(=O)m-; m is 0, 1 or 2;
n is independently 0, 1 , 2, 3 or 4;
p is independently 0, 1 , 2, 3 or 4;
q is 0, 1 , 2 or 3;
r is 0 to 5, provided that r is 0 when V is hydrogen; and t is 0 or 1 ; or the pharmaceutically acceptable salts thereof.
A preferred embodiment of the compounds of this invention is illustrated by the following formula A:
Figure imgf000011_0001
wherein: from 1-2 of f(s) are independently N or N->O, and the remaining f's are independently CH;
R1 is independently selected from: hydrogen, C3-C10 cycloalkyl, R10O-, -N(R 10)2, F or C1 -C6 alkyl;
R2 is independently selected from:
a) hydrogen,
b) aryl, heterocycle, C3-C10 cycloalkyl, R10O-, -N(R10)2, F or C2-C6 alkenyl,
c) unsubstituted or substituted C1 -C6 alkyl wherein the
substituent on the substituted C1 -C6 alkyl is selected from unsubstituted or substituted aryl, heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, R10O- and -N(R10)2;
R3, R4 and R5 are independently selected from:
a) hydrogen,
b) unsubstituted or substituted aryl, unsubstituted or
substituted heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, halogen, C1 -C6 perfluoroalkyl, R12O-, R11S(O)m-, R10C(O)NR10-, (R10)2NC(O)-, R10 2N-C(NR10)-, CN, NO2, R10C(O)-, N3, -N(R10)2, or R11OC(O)NR10-,
c) unsubstituted C1 -C6 alkyl;
d) substituted C1 -C6 alkyl wherein the substituent on the substituted C1 -C6 alkyl is selected from unsubstituted or substituted aryl, unsubstituted or substituted heterocyclic, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, R12O-, R1 1S(O)m-, R10C(O)NR10-, (R10)2NC(O)-, R10 2N-C(NR10)-, CN, R10C(O)-, N3, -N(R10)2, and R11OC(O)-NR10-; R6a, R6b, R6c, R6d and R6e are independently selected from:
a) hydrogen,
b) unsubstituted or substituted aryl, unsubstituted or
substituted heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, halogen, C1 -C6 perfluoroalkyl, R12O-, R11S(O)m-, R10C(O)NR10-, (R10)2NC(O)-, R10 2N-C(NR10)., CN, NO2, R10C(O)-, N3, -N(R10)2, or R11OC(O)NR10-,
c) unsubstituted C1 -C6 alkyl;
d) substituted C1 -C6 alkyl wherein the substituent on the substituted C1 -C6 alkyl is selected from unsubstituted or substituted aryl, unsubstituted or substituted heterocyclic, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, R12O-, R11S(O)m-, R10C(O)NR10-, (R 10)2NC(O)-, R10 2N-C(NR10)-, CN,
R10C(O)-, N3, -N(R10)2, and R11OC(O)-NR10-; or any two of R6a, R6b, R6c, R6d and R6e on adjacent carbon atoms are combined to form a diradical selected from -CH=CH-CH=CH-, -CH=CH-CH2-, -(CH2)4- and -(CH2)3-; provided that when R3, R4, R5, R6a, R6b, R6c, R6d or R6e is unsubstituted or substituted heterocycle, attachment of R3, R4, R5, R6a, R6b, R6c, R6d or R6e to the 6-membered heteroaryl ring, or phenyl ring respectively, is through a substitutable heterocycle ring carbon;
R7 is 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,
Figure imgf000014_0001
V f) - SO2R11
g) N(R10)2 or
h) C1 -4 perfluoroalkyl; R8 is independently selected from:
a) hydrogen,
b) aryl, substituted aryl, heterocycle, 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 R11OC(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 R11OC(O)NR10-; provided that when R8 is heterocycle, attachment of R8 to V is through a substitutable ring carbon;
R9 is selected from:
a) hydrogen,
b) C2-C6 alkenyl, C2-C6 alkynyl, C1 -C6 perfluoroalkyl, F, Cl, R11O-, R11S(O)m-, R10C(O)NR10-, (R10)2NC(O)-,
CN, NO2, (R10)2N-C(NR10)-, R10C(O)-, -N(R10)2, or R11OC(O)NR10-, and
c) C1 -C6 alkyl unsubstituted or substituted by C1 -C6
perfluoroalkyl, F, Cl, R10O-, R11S(O)m-, R10C(O)NR10- (R10)2NC(O)-, CN, (R10)2N-C(NR10)-, R10C(O)-,
-N(R 10)2, or R11OC(O)NR10-; R10 is independently selected from hydrogen, C1 -C6 alkyl, benzyl,
2,2,2-trifluoroethyl and aryl; R11 is independently selected from C1 -C6 alkyl and aryl; R12 is independently selected from hydrogen, C1 -C6 alkyl, C1 -C6 aralkyl, C1 -C6 substituted aralkyl, C1 -C6 heteroaralkyl, C1 -C6 substituted heteroaralkyl, aryl, substituted aryl, heteroaryl, substituted heteraryl, C1 -C6 perfluoroalkyl,
2-aminoethyl and 2,2,2-trifluoroethyI;
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,
imidazolinyl, pyridinyl, thiazolyl, oxazolyl, indolyl, quinolinyl, isoquinolinyl, triazolyl and thienyl, c) aryl,
d) C1 -C20 alkyl wherein from 0 to 4 carbon atoms are
replaced with 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;
provided that when V is heterocycle, attachment of V to R8 and to A1 is through a substitutable ring carbon; W is a heterocycle selected from pyrrolidinyl, imidazolyl, imidazolinyl, pyridinyl, thiazolyl, oxazolyl, indolyl, quinolinyl, triazolyl or
isoquinolinyl;
X is a bond, O, -C(=O)-, -CH=CH-, -C(O)NR7-, -NR7C(O)-, -NR7-,
-S(O)2N(R10)-, -N(R10)S(O)2- or -S(=O)m-; m is 0, 1 or 2;
n is independently 0, 1 , 2, 3 or 4;
p is independently 0, 1 , 2, 3 or 4; q is 0, 1 , 2 or 3;
r is 0 to 5, provided that r is 0 when V is hydrogen; and t is 0 or 1 ; or the pharmaceutically acceptable salts thereof.
A preferred embodiment of the compounds of this invention are illustrated by the formula B:
wherein: from 1-2 of f(s) are independently N or N->O, and the remaining f's are independently CH;
R 1 is selected from: hydrogen, C3-C10 cycloalkyl, R10O-, -N(R 10)2, F or C1 -C6 alkyl;
R2 is independently selected from:
a) hydrogen,
b) aryl, heterocycle, C3-C10 cycloalkyl, R10O-, -N(R10)2, F or C2-C6 alkenyl,
c) unsubstituted or substituted C1 -C6 alkyl wherein the
substituent on the substituted C1 -C6 alkyl is selected from unsubstituted or substituted aryl, heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, R10O- and -N(R10)2;
R3 and R4 are independently selected from:
a) hydrogen, b) unsubstituted or substituted aryl, unsubstituted or substituted heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, halogen, C1 -C6 perfluoroalkyl, R12O-, R1 1 S(O)m-, R10C(O)NR10-, (R10)2NC(O)-, R10 2N-C(NR10)-, CN, NO2, R10C(O)-, N3, -N(R 10)2, or R11OC(O)NR10-,
c) unsubstituted C1 -C6 alkyl,
d) substituted C1 -C6 alkyl wherein the substituent on the substituted C1 -C6 alkyl is selected from unsubstituted or substituted aryl, unsubstituted or substituted heterocyclic, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, R12O-, R 1 1 S(O)m-, R10C(O)NR10-, (R10)2NC(O)-, R10 2N-C(NR10)-, CN, R10C(O)-, N3, -N(R10)2, and R11OC(O)-NR10-;
R6a, R6b, R6c, R6d and R6e are independently selected from:
a) hydrogen,
b) unsubstituted or substituted aryl, unsubstituted or
substituted heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, halogen, C1 -C6 perfluoroalkyl,
R12O-, R11 S(O)m-, R10C(O)NR10-, (R10)2NC(O)-, R10 2N-C(NR10)., CN, NO2, R10C(O)-, N3, -N(R 10)2, or R11OC(O)NR10-,
c) unsubstituted C1 -C6 alkyl,
d) substituted C1 -C6 alkyl wherein the substituent on the substituted C1 -C6 alkyl is selected from unsubstituted or substituted aryl, unsubstituted or substituted heterocyclic, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, R12O-, R1 1S(O)m-, R10C(O)NR10-, (R10)2NC(O)-, R10 2N-C(NR10)-, CN, R10C(O)-, N3, -N(R10)2, and R11OC(O)-NR10-; or any two of R6a, R6b, R6c, R6d and R6e on adjacent carbon atoms are combined to form a diradical selected from -CH=CH-CH=CH-, -CH=CH-CH2-, -(CH2)4- and -(CH2)3-; provided that when R3, R4, R6a, R6b, R6c, R6d or R6e is
unsubstituted or substituted heterocycle, attachment of R3, R4, R6a, R6b, R6c, R6d or R6e to the 6-membered heteroaryl ring, or phenyl ring respectively, is through a substitutable heterocycle ring carbon;
R8 is independently selected from:
a) hydrogen,
b) aryl, substituted aryl, heterocycle, C1 -C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1 -C6 perfluoroalkyl, F, Cl, R10O-, R10C(O)NR10-, CN, NO2, (R 10)2N-C(NR10)-, R10C(O)-, -N(R10)2, or R11OC(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 R11OC(O)NR10-;
provided that when R8 is heterocycle, attachment of R8 to V is through a substitutable ring carbon;
R9a and R9b are independently hydrogen, C1 -C6 alkyl, trifluoromethyl and halogen; R10 is independently selected from hydrogen, C1 -C6 alkyl, benzyl,
2,2,2-trifluoroethyl and aryl;
R 1 1 is independently selected from C1 -C6 alkyl and aryl;
R12 is independently selected from hydrogen, C1 -C6 alkyl, C1 -C6
aralkyl, C1 -C6 substituted aralkyl, C1 -C6 heteroaralkyl, C1 -C6 substituted heteroaralkyl, aryl, substituted aryl, heteroaryl, substituted heteraryl, C1 -C6 perfluoroalkyl, 2-aminoethyl and 2,2,2-trifluoroethyl;
A 1 and A2 are independently selected from: a bond, -CH=CH-, -C≡C-,
-C(O)-, -C(O)NR10-, O, -N(R1 0)-, or S(O)m;
V is selected from:
a) hydrogen,
b) heterocycle selected from pyrrolidinyl, imidazolyl,
imidazolinyl, pyridinyl, thiazolyl, oxazolyl, indolyl, quinolinyl, isoquinolinyl, triazolyl and thienyl, c) aryl,
d) C1 -C20 alkyl wherein from 0 to 4 carbon atoms are
replaced with 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 A 1 is a bond, n is 0 and A2 is S(O)m;
provided that when V is heterocycle, attachment of V to R8 and to A 1 is through a substitutable ring carbon; X is a bond, -CH=CH-, -C(O)NR10-, -NR10C(O)-, -NR 1 0-, O or -C(=O)-; m is 0, 1 or 2;
n is independently 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.
Another preferred embodiment of the compounds of this invention are illustrated by the formula C:
Figure imgf000020_0001
wherein: from 1 -2 of f(s) are independently N or N->O, and the remaining f's are independently CH;
R 1 is selected from: hydrogen, C3-C10 cycloalkyl, R10O-, -N(R10)2, F or C1 -C6 alkyl; R2 is independently selected from:
a) hydrogen,
b) aryl, heterocycle, C3-C10 cycloalkyl, R 1 0O-, -N(R 10)2, F or C2-C6 alkenyl,
c) unsubstituted or substituted C1 -C6 alkyl wherein the
substituent on the substituted C1 -C6 alkyl is selected from unsubstituted or substituted aryl, heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, R10O- and -N(R 10)2;
R3 and R4 are independently selected from:
a) hydrogen,
b) unsubstituted or substituted aryl, unsubstituted or
substituted heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, halogen, C1 -C6 perfluoroalkyl, R12O-, R11S(O)m-, R10C(O)NR10-, CN(R10)2NC(O)-, R10 2N-C(NR10)-, CN, NO2, R10C(O)-, N3, -N(R10)2, or R11OC(O)NR10-,
c) unsubstituted C1 -C6 alkyl, d) substituted C1 -C6 alkyl wherein the substituent on the substituted C1 -C6 alkyl is selected from unsubstituted or substituted aryl, unsubstituted or substituted heterocyclic, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, R12O-, R11S(O)m-, R10C(O)NR10-, (R10)2NC(O)-, R10 2N-C(NR10)-, CN, R10C(O)-, N3, -N(R10)2, and R11OC(O)-NR10-;
R6a, R6b, R6c, R6d and R6e are independently selected from:
a) hydrogen,
b) unsubstituted or substituted aryl, unsubstituted or
substituted heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, halogen, C1 -C6 perfluoroalkyl, R12O-, R11S(O)m-, R10C(O)NR10-, CN(R10)2NC(O)-, R10 2N-C(NR10)-, CN, NO2, R10C(O)-, N3, -N(R10)2, or R11OC(O)NR10-,
c) unsubstituted C1 -C6 alkyl,
d) substituted C1 -C6 alkyl wherein the substituent on the substituted C1 -C6 alkyl is selected from unsubstituted or substituted aryl, unsubstituted or substituted heterocyclic, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, R12O-, R11S(O)m-, R10C(O)NR10-, (R10)2NC(O)-, R10 2N-C(NR10)-, CN, R10C(O)-, N3, -N(R10)2, and R11OC(O)-NR10-; or any two of R6a, R6b, R6c, R6d and R6e on adjacent carbon atoms are combined to form a diradical selected from -CH=CH-CH=CH-, -CH=CH-CH2-, -(CH2)4- and -(CH2)3-; provided that when R3, R4, R6a, R6b, R6c, R6d or R6e is
unsubstituted or substituted heterocycle, attachment of R3, R4, R6a, R6b, R6c, R6d or R6e to the 6-membered heteroaryl ring, or phenyl ring respectively, is through a substitutable heterocycle ring carbon; R8 is independently selected from:
a) hydrogen,
b) aryl, substituted aryl, heterocycle, C1 -C6 alkyl, C2-C6
alkenyl, C2-C6 alkynyl, C1 -C6 perfluoroalkyl, F, Cl, R10O-, R10C(O)NR10-, CN, NO2, (R 10)2N-C(NR10)-, R10C(O)-, -N(R10)2, or R11OC(O)NR10-, and c) C1 -C6 alkyl substituted by C1 -C6 perfluoroalkyl, R10O-, R10C(O)NR10-, (R 10)2N-C(NR10)-, R10C(O)-,
-N(R10)2, or R1 1OC(O)NR10-;
provided that when R8 is heterocycle, attachment of R8 to V is through a substitutable ring carbon;
R9a and R9b are independently hydrogen, C1 -C6 alkyl, trifluoromethyl and halogen; R10 is independently selected from hydrogen, C1 -C6 alkyl, benzyl,
2,2,2-trifluoroethyl and aryl; R11 is independently selected from C1 -C6 alkyl and aryl;
R12 is independently selected from hydrogen, C1 -C6 alkyl, C1 -C6
aralkyl, C1 -C6 substituted aralkyl, C1 -C6 heteroaralkyl, C1 -C6 substituted heteroaralkyl, aryl, substituted aryl, heteroaryl, substituted heteraryl, C1 -C6 perfluoroalkyl,
2-aminoethyl and 2,2,2-trifluoroethyl;
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, imidazolinyl, pyridinyl, thiazolyl, oxazolyl, indolyl, quinolinyl, isoquinolinyl, triazolyl and thienyl, c) aryl,
d) C1-C20 alkyl wherein from 0 to 4 carbon atoms are
replaced with 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 A 1 is a bond, n is 0 and A2 is S(O)m;
provided that when V is heterocycle, attachment of V to R8 and to A1 is through a substitutable ring carbon;
X is a bond, -CH=CH-, -C(O)NR10-, -NR10C(O)-, -NR10-, O or
-C(=O)-; m is 0, 1 or 2;
n is independently 0, 1 , 2, 3 or 4;
p is 0, 1 , 2, 3 or 4, provided that p is not 0 if X is a bond or O; 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 farnesyl-protein transferase are illustrated by the formula D:
Figure imgf000023_0001
wherein: from 1-2 of f(s) are independently N or N->0, and the remaining f's are independently CH; R 1 is selected from: hydrogen, C3-C10 cycloalkyl or C1 -C6 alkyl;
R2 is independently selected from:
a) hydrogen,
b) aryl, heterocycle, C3-C10 cycloalkyl, R10O-, -N(R10)2, F or C2-C6 alkenyl,
c) C1 -C6 alkyl unsubstituted or substituted by aryl,
heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, R10O-, or -N(R10)2; R3 is selected from:
a) hydrogen,
b) unsubstituted or substituted aryl, unsubstituted or
substituted heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, halogen, C1 -C6 perfluoroalkyl, R12O-, R11S(O)m-, R10C(O)NR10-, (R10)2NC(O)-, R10 2N-C(NR10)-, CN, NO2, R10C(O)-, N3, -N(R10)2, or R 1 1 OC(O)NR10-,
c) unsubstituted C1 -C6 alkyl,
d) substituted C1 -C6 alkyl wherein the substituent on the
substituted C1 -C6 alkyl is selected from unsubstituted or substituted aryl, unsubstituted or substituted heterocyclic, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl,
R12O-, R11S(O)m-, R10C(O)NR10-, (R10)2NC(O)-, R10 2N-C(NR10)-, CN, R10C(O)-, N3, -N(R10)2, and R11OC(O)-NR10-;
R4 is selected from H, halogen, C1 -C6 alkyl and CF3;
R6a, R6b, R6c, R6d and R6e are independently selected from: a) hydrogen,
b) unsubstituted or substituted aryl, unsubstituted or
substituted heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, halogen, C1 -C6 perfluoroalkyl, R12O-, R11S(O)m-, R10C(O)NR10-, (R10)2NC(O)-, R10 2N-C(NR10)-, CN, NO2, R10C(O)-, N3, -N(R10)2, or R11OC(O)NR10-,
c) unsubstituted C1 -C6 alkyl,
d) substituted C1 -C6 alkyl wherein the substituent on the substituted C1 -C6 alkyl is selected from unsubstituted or substituted aryl, unsubstituted or substituted heterocyclic, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, R12O-, R11S(O)m-, R10C(O)NR10-, (R10)2NC(O)-, R10 2N-C(NR10)-, CN, R10C(O)-, N3, -N(R 10)2, and R11OC(O)-NR10-; or any two of R6a, R6b, R6c, R6d and R6e on adjacent carbon atoms are combined to form a diradical selected from -CH=CH-CH=CH-, -CH=CH-CH2-, -(CH2)4- and -(CH2)3-; provided that when R3, R6a, R6b, R6c, R6d or R6e is
unsubstituted or substituted heterocycle, attachment of R3, R6a, R6b, R6c, R6d or R6e to the 6-membered heteroaryl ring, or phenyl ring respectively, is through a substitutable heterocycle ring carbon;
R8 is independently selected from:
a) hydrogen,
b) aryl, substituted aryl, heterocycle, 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 R11OC(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 R1 1OC(O)NR10-;
provided that when R8 is heterocycle, attachment of R8 to V is through a substitutable ring carbon;
R9a and R9b are independently hydrogen, halogen, CF3 or methyl; R10 is independently selected from hydrogen, C1 -C6 alkyl, benzyl,
2,2,2-trifluoroethyl and aryl; R11 is independently selected from C1 -C6 alkyl and aryl;
R 12 is independently selected from hydrogen, C1 -C6 alkyl, C1 -C6 aralkyl, C1 -C6 substituted aralkyl, C1 -C6 heteroaralkyl, C1 -C6 substituted heteroaralkyl, aryl, substituted aryl, heteroaryl, substituted heteraryl, C1 -C6 perfluoroalkyl, 2-aminoethyl and 2,2,2-trifluoroethyl; A 1 is selected from: a bond, -C(O)-, O, -N(R10)-, or S(O)m;
X is a bond, -CH=CH-, -C(O)NR10-, -NR10C(O)-, -NR10-, O or -C(=O)-, n is 0 or 1 ; provided that n is not 0 if A1 is a bond, O,
-N(R10)- or S(O)m;
m is 0, 1 or 2; and
p is 0, 1 , 2, 3 or 4; or the pharmaceutically acceptable salts thereof.
In another more preferred embodiment of this invention, the inhibitors of farnesyl-protein transferase are illustrated by the formula E:
Figure imgf000027_0001
wherein: from 1 -2 of f(s) are independently N or N->O, and the remaining f's are independently CH;
R 1 is selected from: hydrogen, C3-C10 cycloalkyl, R10O-, -N(R10)2, F or C1 -C6 alkyl; R2 is independently selected from:
a) hydrogen,
b) aryl, heterocycle, C3-C10 cycloalkyl, R10O-, -N(R 10)2, F or C2-C6 alkenyl,
c) C1 -C6 alkyl unsubstituted or substituted by aryl,
heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, R10O-, or
-N(R10)2;
R3 is selected from:
a) hydrogen,
b) unsubstituted or substituted aryl, unsubstituted or
substituted heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, halogen, C1 -C6 perfluoroalkyl, R12O-, R11S(O)m-, R10C(O)NR10-, (R 10)2NC(O)-, R10 2N-C(NR10)-, CN, NO2, R10C(O)-, N3, -N(R10)2, or R1 1OC(O)NR10-,
c) unsubstituted C1 -C6 alkyl, d) substituted C1 -C6 alkyl wherein the substituent on the substituted C1 -C6 alkyl is selected from unsubstituted or substituted aryl, unsubstituted or substituted heterocyclic, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, R12O-, R11S(O)m-, R10C(O)NR10-, (R10)2NC(O)-,
R 10 2N-C(NR10)-, CN, R10C(O)-, N3, -N(R10)2, and R 1 1OC(O)-NR10-;
R4 is selected from H, halogen, C1 -C6 alkyl and CF3;
R6a, R6b, R6c; R6d and R6e are independently selected from:
a) hydrogen,
b) unsubstituted or substituted aryl, unsubstituted or
substituted heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, halogen, C1 -C6 perfluoroalkyl, R12O-, R 1 1 S(O)m-, R10C(O)NR10-, (R10)2NC(O)-, R10 2N-C(NR10)-, CN, NO2, R10C(O)-, N3, -N(R10)2, or R11OC(O)NR10-,
c) unsubstituted C1 -C6 alkyl,
d) substituted C1 -C6 alkyl wherein the substituent on the
substituted C1 -C6 alkyl is selected from unsubstituted or substituted aryl, unsubstituted or substituted heterocyclic, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, R12O-, R 1 1 S(O)m-, R10C(O)NR10-, (R10)2NC(O)-, R10 2N-C(NR10)-; CN, R10C(O)-, N3, -N(R10)2, and R11OC(O)-NR10-; or any two of R6a, R6b, R6c, R6d and R6e on adjacent carbon atoms are combined to form a diradical selected from -CH=CH-CH=CH-, -CH=CH-CH2-, -(CH2)4- and -(CH2)3-; provided that when R3, R6a, R6b, R6c, R6d or R6e is
unsubstituted or substituted heterocycle, attachment of R3, R6a, R6b, R6c5 R6d or R6e to the 6-membered heteroaryl ring, or phenyl ring respectively, is through a substitutable heterocycle ring carbon;
R8 is independently selected from:
a) hydrogen,
b) aryl, substituted aryl, heterocycle, 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 R11OC(O)NR10-, and c) C1 -C6 alkyl substituted by C1 -C6 perfluoroalkyl, R10O-, R10C(O)NR10-5 (R10)2N-C(NR10)-, R10C(O)-,
-N(R10)2, or R11 OC(O)NR10-;
provided that when R8 is heterocycle, attachment of R8 to V is through a substitutable ring carbon;
R9a and R9b are independently hydrogen, halogen, CF3 or methyl; R10 is independently selected from hydrogen, C1 -C6 alkyl, benzyl,
2,2,2-trifluoroethyl and aryl; R11 is independently selected from C1 -C6 alkyl and aryl;
R12 is independently selected from hydrogen, C1 -C6 alkyl, C1 -C6
aralkyl, C1 -C6 substituted aralkyl, C1 -C6 heteroaralkyl, C1 -C6 substituted heteroaralkyl, aryl, substituted aryl, heteroaryl, substituted heteraryl, C1 -C6 perfluoroalkyl, 2-aminoethyl and 2,2,2-trifIuoroethyl;
X is a bond, -CH=CH-, -C(O)NR10-, -NR10C(O)-, -NR10-, O or -C(=O)-; n is 0 or 1 ;
m is 0, 1 or 2; and
p is 0, 1 , 2, 3 or 4, provided that p is not 0 if X is a bond or O; or the pharmaceutically acceptable salts thereof.
In a further embodiment of this invention, the inhibitors of farnesyl-protein transferase are illustrated by the formula F:
Figure imgf000030_0001
wherein: from 1 -2 of f(s) are independently N or N->O, and the remaining f's are independently CH;
R 1 is selected from: hydrogen, C3-C10 cycloalkyl or C1 -C6 alkyl;
R2 is independently selected from:
a) hydrogen,
b) aryl, heterocycle, C3-C10 cycloalkyl, R10O-, -N(R1 0)2 or
F,
c) C1 -C6 alkyl unsubstituted or substituted by aryl,
heterocycle, C3-C10 cycloalkyl, R10O-, or -N(R 10)2; R3 is selected from:
a) hydrogen,
b) unsubstituted or substituted aryl, unsubstituted or
substituted heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, halogen, C1 -C6 perfluoroalkyl, R 12O-, R11 S(O)m-, R10C(O)NR10-, (R10)2NC(O)-, R10 2N-C(NR10)-, CN, NO2, R10C(O)-, N3, -N(R10)2, or R11OC(O)NR10-, c) unsubstituted C1 -C6 alkyl,
d) substituted C1 -C6 alkyl wherein the substituent on the substituted C1 -C6 alkyl is selected from unsubstituted or substituted aryl, unsubstituted or substituted heterocyclic, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl,
R 1 2O-, R11S(O)m-, R10C(O)NR10-, (R10)2NC(O)-, R10 2N-C(NR10)-, CN, R10C(O)-, N3, -N(R10)2, and R11OC(O)-NR10-; R4 is selected from H, halogen, CH3 and CF3;
R6a, R6b, R6c, R6d and R6e are independently selected from:
a) hydrogen,
b) unsubstituted or substituted aryl, unsubstituted or
substituted heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, halogen, C1 -C6 perfluoroalkyl, R1 2O-, R11S(O)m-, R10C(O)NR10-, (R10)2NC(O)-, R10 2N-C(NR10)-, CN, NO2, R10C(O)-, N3, -N(R10)2, or R11OC(O)NR10-,
c) unsubstituted C1 -C6 alkyl,
d) substituted C1 -C6 alkyl wherein the substituent on the substituted C1 -C6 alkyl is selected from unsubstituted or substituted aryl, unsubstituted or substituted heterocyclic, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, R12O-, R11S(O)m-, R10C(O)NR10-, (R10)2NC(O)-, R10 2N-C(NR10)-, CN, R10C(O)-, N3, -N(R10)2, and R11OC(O)-NR10-; or any two of R6a, R6b, R6c, R6d and R6e on adjacent carbon atoms are combined to form a diradical selected from -CH=CH-CH=CH-,
-CH=CH-CH2-, -(CH2)4- and -(CH2)3-; provided that when R3, R6a, R6b, R6c, R6d or R6e is
unsubstituted or substituted heterocycle, attachment of R3, R6a, R6b , R6c, R6d or R6e to the 6-membered heteroaryl ring, or phenyl ring respectively, is through a substitutable heterocycle ring carbon; R9a and R9b are independently hydrogen, halogen, CF3 or methyl; R10 is independently selected from hydrogen, C1 -C6 alkyl, benzyl,
2,2,2-trifluoroethyl and aryl; R11 is independently selected from C1 -C6 alkyl and aryl;
R 1 2 is independently selected from hydrogen, C1 -C6 alkyl, C1 -C6
aralkyl, C1 -C6 substituted aralkyl, C1 -C6 heteroaralkyl, C1 -C6 substituted heteroaralkyl, aryl, substituted aryl, heteroaryl, substituted heteraryl, C1 -C6 perfluoroalkyl,
2-aminoethyl and 2,2,2-trifluoroethyl;
X is a bond, -CH=CH-, -C(O)NR10-, -NR10C(O)-, -NR 1 0-, O or -C(=O)-; m is 0, 1 or 2; and
p is 0, 1 , 2, 3 or 4; or the pharmaceutically acceptable salts thereof.
In a further embodiment of this invention, the inhibitors of farnesyl-protein transferase are illustrated by the formula G:
Figure imgf000032_0001
wherein: from 1 -2 of f(s) are independently N or N->O, and the remaining f's are independently CH;
R 1 is selected from: hydrogen, C3-C10 cycloalkyl, R10O-, -N(R10)2, F or C1 -C6 alkyl;
R2 is independently selected from:
a) hydrogen,
b) aryl, heterocycle or C3-C10 cycloalkyl,
c) C1 -C6 alkyl unsubstituted or substituted by aryl,
heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, R10O-, or -N(R10)2;
R3 is selected from:
a) hydrogen,
b) unsubstituted or substituted aryl, unsubstituted or
substituted heterocycle, C3-C10 cycloalkyl, C2-C6 a lkenyl, C2-C6 alkynyl, halogen, C1 -C6 perfluoroalkyl, R12O-, R11S(O)m-, R10C(O)NR10-, (R10)2NC(O)-, R10 2N-C(NR10)-, CN, NO2, R10C(O)-, N3, -N(R 10)2, or R11OC(O)NR10-,
c) unsubstituted C1 -C6 alkyl,
d) substituted C1 -C6 alkyl wherein the substituent on the
substituted C1 -C6 alkyl is selected from unsubstituted or substituted aryl, unsubstituted or substituted heterocyclic, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl,
R12O-, R11S(O)m-, R10C(O)NR10-, (R10)2NC(O)-, R10 2N-C(NR10)-, CN, R10C(O)-, N3, -N(R10)2, and
R1 1OC(O)-NR10-;
R4 is selected from H, halogen, CH3 and CF3; R6a, R6b, R6c, R6d and R6e are independently selected from:
a) hydrogen,
b) unsubstituted or substituted aryl, unsubstituted or
substituted heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, halogen, C1 -C6 perfluoroalkyl, R12O-, R11S(O)m-, R10C(O)NR10-, (R10)2NC(O)-, R10 2N-C(NR10)-, CN, NO2, R10C(O)-, N3, -N(R10)2, or R11OC(O)NR10-,
c) unsubstituted C1 -C6 alkyl,
d) substituted C1 -C6 alkyl wherein the substituent on the substituted C1 -C6 alkyl is selected from unsubstituted or substituted aryl, unsubstituted or substituted heterocyclic, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, R12O-, R 1 1 S(O)m-,
R10C(O)NR10-, (R10)2NC(O)-, R10 2N-C(NR10)-, CN,
R10C(O)-, N3, -N(R10)2, and R 1 1 OC(O)-NR 1 0-; or any two of R6a, R6b, R6c, R6d and R6e on adjacent carbon atoms are combined to form a diradical selected from -CH=CH-CH=CH-, -CH=CH-CH2-, -(CH2)4- and -(CH2)3-; provided that when R3, R6a, R6b, R6c, R6d or R6e is
unsubstituted or substituted heterocycle, attachment of R3, R6a, R6b, R6c, R6d or R6e to the 6-membered heteroaryl ring, or phenyl ring respectively, is through a substitutable heterocycle ring carbon;
R9a and R9b are independently hydrogen, halogen, CF3 or methyl; R10 is independently selected from hydrogen, C1 -C6 alkyl, benzyl,
2,2,2-trifluoroethyl and aryl; R11 is independently selected from C1 -C6 alkyl and aryl; R12 is independently selected from hydrogen, C1 -C6 alkyl, C1 -C6 aralkyl, C1 -C6 substituted aralkyl, C1 -C6 heteroaralkyl, C1 -C6 substituted heteroaralkyl, aryl, substituted aryl, heteroaryl, substituted heteraryl, C1 -C6 perfluoroalkyl, 2-aminoethyl and 2,2,2-trifIuoroethyl;
A 1 is selected from: a bond, -C(O)-, O, -N(R10)-, or S(O)m; m is 0, 1 or 2; and
n is O or 1; or the pharmaceutically acceptable salts thereof.
Preferred compounds of the invention are:
1 -(2-Phenylpyrid-5-ylmethyl)-5-(4-cyanobenzyI)imidazole
1 -(2-Phenyl-N-Oxopyrid-5-ylmethyl)-5-(4-cyanobenzyl)imidazole 1 -(3-Phenylpyrid-6-ylmethyl)-5-(4-cyanobenzyl)imidazole
1 -(3-Phenyl-N-Oxopyrid-6-ylmethyl)-5-(4-cyanobenzyl)imidazole
1-(2-(3-Trifluoromethoxyphenyl)-pyrid-5-ylmethyl)-5-(4- cyanobenzyl)imidazole
1 -(2-(2-Trifluoromethylphenyl)-pyrid-5-ylmethyl)-5-(4- cyanobenzyl)imidazole
1 -(3-Phenyl-2-Chloropyrid-6-ylmethyl)-5-(4-cyanobenzyl)imidazoIe 1 -(3-Phenyl-4-chloropyrid-6-ylmethyl)-5-(4-cyanobenzyl)imidazole and
1 -(2-Amino-3-phenylpyrid-6-ylmethyl)-5-(4-cyanobenzyl)imidazole or a pharmaceutically acceptable salt thereof.
Specific examples of the compounds of the instant invention are:
1 -(2-Phenylpyrid-5-ylmethyl)-5-(4-cyanobenzyl)imidazole
Figure imgf000036_0001
1 -(2-(2-Trifluoromethylphenyl)-pyrid-5-ylmethyl)-5-(4- cyanobenzyI)imidazole
Figure imgf000036_0002
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, R1 , 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" and the alkyl portion of aralkyl and similar terms, 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.
As used herein, "cycloalkyl" is intended to include non- aromatic cyclic hydrocarbon groups having the specified number of carbon atoms. Examples of cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl and the like.
"Alkenyl" groups include those groups having the specified number of carbon atoms and having one or several double bonds.
Examples of alkenyl groups include vinyl, allyl, isopropenyl, pentenyl, hexenyl, heptenyl, cyclopropenyl, cyclobutenyl, cyclopentenyl, cyclohexenyl, 1 -propenyl, 2-butenyl, 2-methyl-2-butenyl, isoprenyl, farnesyl, geranyl, geranylgeranyl and the like.
"Alkynyl" groups include those groups having the specified number of carbon atoms and having one triple bonds. Examples of alkynyl groups include acetylene, 2-butynyl, 2-pentynyl, 3-pentynyl and the like.
"Halogen" or "halo" as used herein means fluoro, chloro, bromo and iodo.
As used herein, "aryl," and the aryl portion of aroyl and aralkyl, 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 structure. 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, isoindohnyl, isoquinolinyl, isothiazolidinyl, isothiazolyl, isothiazolidinyl, morpholinyl,
naphthyridinyl, oxadiazolyl, 2-oxoazepinyI, oxazolyl, 2- oxopyrrolidinyl, pyridyl, pyrazinyl, pyrazolidinyl, pyrazolyl,
pyridazinyl, pyrimidinyl, pyrrolidinyl, pyrrolyl, quinazolinyl,
quinolinyl, quinoxalinyl, tetrahydrofuryl, tetrahydroisoquinolinyl, tetrahydroquinolinyl, thiamorpholinyl, 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, isoindohnyl, 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 R3, R4, R5 and R6a-e, the term "the substituted group" is intended to mean a substituted Cl-8 alkyl, substituted C2-8 alkenyl, substituted C2-8 alkynyl, substituted aryl or substituted heterocycle from which the substituent(s) R3, R4, R5 and R6a-e are selected.
As used herein in the definition of R7, the substituted C1 -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 substituents in addition to the point of attachment to the rest of the compound.
As used herein, when no specific substituents are set forth, the terms "substituted aryl", "substituted heterocycle" and "substituted cycloalkyl" are intended to include the cyclic group which is substituted on a substitutable ring carbon atom with 1 or 2 substitutents selected from the group which includes but is not limited to F, Cl, Br, CF3, NH2, N(C1 -C6 alkyl)2, NO2, CN, (C1 -C6 alkyl)O-, -OH, (C1 -C6 alkyl)S(O)m-, (C1 -C6 alkyl)C(O)NH-, H2N-C(NH)-, (C1 -C6
alkyl)C(O)-, (C1 -C6 alkyl)OC(O)-, N3,(C1 -C6 alkyl)OC(O)NH-, phenyl, pyridyl, imidazolyl, oxazolyl, isoxazolyl, thiazolyl, thienyl, furyl, isothiazolyl and C1 -C20 alkyl.
Lines drawn into the ring systems from substituents (such as from R3, R4, Q etc.) means that the indicated bond may be attached to any of the substitutable ring carbon atoms.
The substituent illustrated by the structure
Figure imgf000039_0001
is a simplified representation of a phenyl ring having five (5)
substituents (hydrogens and/or non-hydrogens) and may also be represented by the structure
Figure imgf000040_0001
The moiety described as
Figure imgf000040_0002
where any two of R6a, R6b, R6c R6d and R6e on adjacent carbon atoms are combined to form a diradical selected from -CH=CH-CH=CH, -CH=CH-CH-, -(CH2)4- and -(CH2)4- includes the following
structures:
Figure imgf000040_0003
It is understood that such fused ring moieties may be further substituted by the remaining R6a, R6b, R6c, R6d and/or R6e as defined
hereinabove.
The moiety designated by the following structure
Figure imgf000040_0004
represents an aromatic 6-membered heterocyclic ring and includes the following ring systems:
Figure imgf000041_0001
The moiety designated by the following structure
Figure imgf000041_0002
represents an aromatic 6-membered heterocyclic ring and includes the following ring systems:
Figure imgf000041_0003
wherein it is understood that one of the ring carbon atoms is substituted with
Figure imgf000041_0004
Preferably, the aromatic 6-membered heterocyclic ring is a pyridyl ring.
Preferably, R 1 and R2 are independently selected from: hydrogen, R11C(O)O-, -N(R10)2, R10C(O)NR10-, R10O- or unsubstituted or substituted C1 -C6 alkyl wherein the substituent on the substituted C1 -C6 alkyl is selected from unsubstituted or substituted phenyl, -N(R 10)2, R10O- and R10C(O)NR10-.
Preferably, R3 is selected from:
a) hydrogen,
b) C3-C10 cycloalkyl, halogen, C1 -C6 perfluoroalkyl, R 1 2O-,
CN, NO2, R10C(O)- or -N(R10)2,
c) unsubstituted C1 -C6 alkyl,
d) substituted C1 -C6 alkyl wherein the substituent on the
substituted C1 -C6 alkyl is selected from unsubstituted or substituted aryl, unsubstituted or substituted heterocyclic, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl,
R12O-, R11 S(O)m-, R10C(O)NR10-, (R10)2NC(O)-, R10 2N-C(NR 10)-, CN, R10C(O)-, N3, -N(R 10)2, and R11OC(O)-NR10-.
Preferably, R4 is selected from: hydrogen, halogen, trifluoromethyl, trifluoromethoxy and C1 -C6 alkyl.
Preferably, R5 is hydrogen.
Preferably, R6a, R6b, R6c, R6d and R6e are independently selected from:
a) hydrogen,
b) C3-C10 cycloalkyl, halogen, C1 -C6 perfluoroalkyl, R 12O-, R1 1S(O)m-, CN, NO2, R10C(O)- or -N(R10)2, c) unsubstituted C1 -C6 alkyl;
d) substituted C1 -C6 alkyl wherein the substituent on the
substituted C1 -C6 alkyl is selected from unsubstituted or substituted aryl, C3-C10 cycloalkyl, R12O-, R11S(O)m-, R10C(O)- or -N(R 10)2; or any two of R6a, R6b, R6c, R6d and R6e on adjacent carbon atoms are combined to form a diradical selected from -CH=CH-CH=CH-,
-CH=CH-CH2-, -(CH2)4- and -(CH2)3-.
Preferably, R8 is independently selected from: a) hydrogen, and
b) aryl, substituted aryl, heterocycle, substituted heterocycle, C1 -C6 perfluoroalkyl or CN.
Preferably, R9 is hydrogen, halogen, CF3 or methyl. Preferably, R10 is selected from H, C1 -C6 alkyl and benzyl.
Preferably, A1 and A2 are independently .selected from: a bond, -C(O)NR10-, -NR10C(O)-, O, -N(R10)-, -S(O)2N(R10)- and-
N(R10)S(O)2-.
Preferably, V is selected from hydrogen, heterocycle and aryl. More preferably, V is 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 s is 0.
Preferably t is 1.
Preferably, the moiety
Figure imgf000043_0001
is selected from:
Figure imgf000044_0001
It is intended that the definition of any substituent or variable (e.g., R1 , R2, R9, n, etc.) at a particular location in a molecule be independent of its definitions elsewhere in that molecule. Thus, -N(R 10)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 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 -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. Substituents R3, R6 and R8, as shown in the Schemes, represent the substituents R3, R4, R5 R6a, R6b, R6c, R6d, R6e and R8; although only one such R3, R6 or R8 is present in the intermediates and products of the schemes, it is understood that the reactions shown are also applicable when such aryl or heteroaryl moieties contain multiple substituents.
These reactions may be employed in a linear sequence to provide the compounds of the invention or they may be used to synthesize fragments which are subsequently joined by the alkylation reactions described in the Schemes. The reactions described in the Schemes are illustrative only and are not meant to be limiting. Other reactions useful in the preparation of heteroaryl moieties are described in "Comprehensive Organic Chemistry, Volume 4: Heterocyclic Compounds" ed. P.G. Sammes, Oxford (1979) and references therein. Aryl-aryl coupling is generally described in "Comprehensive Organic Functional Group Transformations," Katritsky et al. eds., pp 472-473, Pergamon Press (1995).
Synopsis of Schemes 1 -21:
The requisite intermediates are in some cases commercially available, or can be prepared according to literature procedures, for the most part. Schemes 1 -12 illustrate synthesis of the instant aryl- heteroaryl compound which incorporate a preferred benzylimidazolyl sidechain. Thus, in Scheme 1 , for example, a arylheteroaryl intermediate that is not commercially available may be synthesized by methods known in the art. Thus, a suitably substituted phenyl boronic acid I may be reacted under Suzuki coupling conditions (Pure Appl. Chem., 63:419 (1991 )) with a suitably substituted halogenated nicotinic acid, such as 4-bromonicotinic acid, to provide the arylheteroaryl carboxylic acid II. The acid may be reduced and the triflate of the intermediate alcohol III may be formed in situ and coupled to a suitably substituted benzylimidazolyl IV to provide, after deprotection, the instant compound V.
Schemes 2-4 illustrate other methods of synthesizing the key alcohol intermediates, which can then be processed as described in Scheme 1. Thus, Scheme 2 illustrates the analogous series of arylheteroaryl alcohol forming reactions starting with the methyl nicotinate boronic acid and the "terminal" phenyl moiety employed in the Suzuki coupling as the halogenated reactant. Such a coupling reaction is also compatible when one of the reactants incorporates a suitably protected hydroxyl functionality as illustrated in Scheme 3.
Negishi chemistry (Org. Synth., 66:67 (1988)) may also be employed to form the arylheteroaryl component of the instant compounds, as shown in Scheme 4. Thus, a suitably substituted zinc bromide adduct may be coupled to a suitably substituted heteroaryl halide in the presence of nickel (II) to provide the arylheteroaryl VII. The heteroaryl halide and the zinc bromide adduct may be selected based on the availability of the starting reagents.
Scheme 5 illustrates the preparation of a suitably substituted biphenylmethyl bromide which could also be utilized in the reaction with the protected imidazole as described in Scheme 1.
As illustrated in Scheme 6, the sequence of coupling reactions may be modified such that the aryl-heteroaryl bond is formed last. Thus, a suitably substituted imidazole may first be alkylated with a suitably substituted benzyl halide to provide intermediate VIII.
Intermediate VIII can then undergo Suzuki type coupling to a suitably substituted phenyl boronic acid.
Scheme 7 illustrates synthesis of an instant compound wherein a non-hydrogen R9b is incorporated in the instant compound. Thus, a readily available 4-substituted imidazole IX may be selectively iodinated to provide the 5-iodoimidazole X. That imidazole may then be protected and coupled to a suitably substituted benzyl moiety to provide intermediate XI. Intermediate XI can then undergo the alkylation reactions that were described hereinabove. Scheme 8 illustrates synthesis of instant compounds that incorporate a preferred imidazolyl moiety connected to the biaryl via an alkyl amino, sulfonamide or amide linker. Thus, the 4-aminoalkyl- imidazole XII, wherein the primary amine is protected as the phthali- mide, is selectively alkylated then deprotected to provide the amine XIII. The amine XIII may then react under conditions well known in the art with various activated arylheteroaryl moieties to provide the instant compounds shown.
Compounds of the instant invention wherein the A1 (CR1 2)nA2(CR 1 2)n linker is oxygen may be synthesized by methods known in the art, for example as shown in Scheme 9.
The suitably substituted phenol XIV may be reacted with methyl N-(cyano)methanimidate to provide the 4-phenoxyimidazole XV.
After selective protection of one of the imidazolyl nitrogens, the intermediate XVI can undergo alkylation reactions as described for the benzylimidazoles hereinabove.
Scheme 10 illustrates an analogous series of reactions wherein the (CR2 2)ρX(CR2 2)p linker of the instant compounds is oxygen. Thus, a suitably substituted halopyridinol, such as 3-chloro- 2-pyridinol, is reacted with methyl N-(cyano)methanimidate to provide intermediate XVI. Intermediate XVI is then protected and, if desired to form a compound of a preferred embodiment, alkylated with a suitably protected benzyl. The intermediate XVII can then be coupled to a aryl moiety by Suzuki chemistry to provide the instant compound.
Compounds of the instant invention wherein the
A1 (CR 1 2)nA2(CR1 2)n linker is a substituted methylene may be synthesized by the methods shown in Scheme 1 1. Thus, the N-protected imidazolyl iodide XVIII is reacted, under Grignard conditions with a suitably protected benzaldehyde to provide the alcohol XIX. Acylation, followed by the alkylation procedure illustrated in the Schemes above (in particular, Scheme 1) provides the instant compound XX. If other R1 substituents are desired, the acetyl moiety can be manipulated as illustrated in the Scheme. Addition of various nucleophiles to an imidazolyl aldehyde may also be employed to form a substituted alkyl linker between the biheteroaryl and the preferred W (imidazolyl) as shown in Scheme 12. Thus an aryllithium can be reacted with pyridine to form the 2- substituted N-lithio-1 ,2-dihydropyridine XXa. Intermediate XXa can then react with a aldehyde to provide a suitably substituted instant compound. Similar substituent manipulation as shown in Scheme 1 1 may be performed on the fully functionalized compound which incorporates an R2 hydroxyl moiety.
Figure imgf000048_0001
Figure imgf000049_0001
Figure imgf000050_0001
Figure imgf000051_0001
Figure imgf000052_0001
Figure imgf000053_0001
Figure imgf000054_0001
Figure imgf000055_0001
Figure imgf000056_0001
Figure imgf000057_0001
Figure imgf000058_0001
Figure imgf000059_0001
Figure imgf000060_0001
Figure imgf000061_0001
Schemes 13-21 illustrate reactions wherein the moiety
Figure imgf000062_0001
incorporated in the compounds of the instant invention is represented by other than a substituted imidazole-containing group.
Thus, the intermediates whose synthesis are illustrated in
Schemes hereinabove and other arylheteroaryl intermediates obtained commercially or readily synthesized, can be coupled with a variety of aldehydes. 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. Lithioheteroaryl chemistry may be utilized, as shown in Scheme 13, to incorporate the arylheteroaryl moiety. Thus, a suitably substituted arylheteroaryl N-lithio reagent is reacted with an aldehyde to provide the C-alkylated instant compound XXI. Compound XXI can be deoxygenated by methods known in the art, such as a catalytic
hydrogention, then deprotected with trifluoroacetic acid in methylene chloride to give the final compound XXII. The final product XXII may be isolated in the salt form, for example, as a trifluoroacetate, hydrochloride or acetate salt, among others. The product diamine XXII can further be selectively protected to obtain XXIII, which can subsequently be reductively alkylated with a second aldehyde to obtain XXIV. Removal of the protecting group, and conversion to cyclized products such as the dihydroimidazole XXV can be accomplished by literature procedures.
If the arylheteroaryl subunit reagent is reacted with an aldehyde which also has a protected hydroxyl group, such as XXVI in Scheme 14, the protecting groups can be subsequently removed to unmask the hydroxyl group (Schemes 14, 15). 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 alkyl lithium reagents, to obtain secondary alcohols such as XXX.
In addition, the fully deprotected amino alcohol XXXI can be
reductively alkylated (under conditions described previously) with a variety of aldehydes to obtain secondary amines, such as XXXII (Scheme 15), or tertiary amines.
The Boc protected amino alcohol XXVIII can also be utilized to synthesize 2-aziridinylmethylarylheteroaryl such as XXXIII (Scheme 16). Treating XXVIII with 1 ,1 '-sulfonyldiimidazole and sodium hydride in a solvent such as dimethylformamide led to the formation of aziridine XXXIII . The aziridine is reacted with a nucleophile, such as a thiol, in the presence of base to yield the ring- opened product XXXIV .
In addition, the arylheteroaryl subunit reagent can be reacted with aldehydes derived from amino acids such as O-alkylated tyrosines, according to standard procedures, to obtain compounds such as XL, as shown in Scheme 17. When R' is an aryl group, XL can first be hydrogenated to unmask the phenol, and the amine group deprotected with acid to produce XLI. Alternatively, the amine protecting group in XL can be removed, and O-alkylated phenolic amines such as XLII produced.
Schemes 18-21 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 incorporate other heterocyclic moieties for variable W are also well known in the art.
Figure imgf000064_0001
Figure imgf000065_0001
Figure imgf000066_0001
Figure imgf000067_0001
Figure imgf000068_0001
Figure imgf000069_0001
Figure imgf000070_0001
Figure imgf000071_0001
Figure imgf000072_0001
Figure imgf000073_0001
Figure imgf000074_0001
Figure imgf000075_0001
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 farnesyl- protein transferase and the farnesylation 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. Schafmer et al. American Journal of Pathology, 142: 1051-1060 (1993) and B. Cowley, Jr. et al.FASEB Journal, 2:A3160 (1988)).
The instant compounds may also be useful for the treatment of fungal infections.
The 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 corn 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 alternatively 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 farnesyl-protein transferase (FPTase) in a composition. Thus the composition to be tested may be divided and the two
portions contacted with mixtures which comprise a known substrate of FPTase (for example a tetrapeptide having a cysteine at the amine terminus) and farnesyl pyrophosphate and, in one of the mixtures, a compound of the instant invention. After the assay mixtures are incubated for an sufficient period of time, well known in the art, to allow the FPTase to farnesylate the substrate, the chemical content of the assay mixtures may be determined by well known
immunological, radiochemical or chromatographic techniques.
Because the compounds of the instant invention are selective
inhibitors of FPTase, absence or quantitative reduction of the amount of substrate in the assay mixture without the compound of the instant invention relative to the presence of the unchanged substrate in the assay 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 farnesyl-protein transferase and quanti- tating 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 farnesyl- protein transferase, an excess amount of a known substrate of FPTase (for example a tetrapeptide having a cysteine at the amine terminus) and farnesyl 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 1 -(2-Phenylpyrid-5-ylmethyl)-5-(4-cyanobenzyl)imidazole
hydrochloride salt
Step A: 1 -Trityl-4-(4-cyanobenzyl)-imidazole
To a suspension of activated zinc dust (3.57g, 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 a-bromo-p-tolunitrile (9.33g, 47.6 mmol) in THF (100 mL) was added dropwise over a period of 10 minutes. The reaction was then allowed to stir at 20°C for 6 hours and bis(triphenylphosphine)Nickel II chloride (2.4g, 3.64 mmol) and 4-iodo-1-tritylimidazole ( 15.95g, 36.6 mmol, S. V. Ley, et al.,
J. Org. Chem. 56, 5739 ( 1991 )) were added in one portion.The resulting mixture was stirred 16 hours at 20°C and then quenched by addition of sat. aq. NH4CI solution (100 mL) and the mixture stirred for 2 hours. Saturated aq. NaHCO3 solution was added to give a pH of 8 and the solution was extracted with EtOAc (2 x 250 mL), dried, (MgSO4) and the solvent evaporated in vacuo. The residue was chromatographed (Silica gel, 0-20% EtOAc in CH2CI2 to afford the title compound as a white solid.
1H NMR (CDCl3, 400MHz) δ 7.54 (2H, d, J=7.9Hz), 7.38(1H, s), 7.36-7.29 ( 1 1 H, m), 7.15-7.09(6H, m), 6.58(1H, s), and 3.93(2H, s)ppm. Step B: 2-Phenyl-5-methylpyridine
A mixture of 2-bromo-5-methylpyridine (2.00 g, 1 1.63 mmol), phenylboronic acid (1.56 g, 12.79 mmol), barium hydroxide (5.50g, 17.4 mmol), DME (80 mL) and water (15 mL) was purged with dry argon. Tetrakis(triphenylphosphine)palladium(0) (672 mg, 0.58 mmol) was added, and the resultant solution was stirred at 80°C for 4 hours. The solvents were evaporated in vacuo, and the residue partitioned between EtOAc and water and acidified with 1M aq. HCl. The aqueous extract was separated, and extracted with EtOAc. The organic extracts were combined, washed with NaHCO3 and 5% aq. Na2S2O3, dried (Na2SO4), filtered and the solvent evaporated in vacuo. The residue was purified by chromatography (Silica gel, CH2CI2) to afford the title compound.
1H NMR (CDCI3, 400MHz) δ 8.52 (1H, s), 7.96(2H, d, J=7.0Hz), 7.63(1H, d, J=8.0Hz), 7.55(1H, brd, J=8.0Hz), 7.50-7.35(3H, m), and 2.37(3H, s) ppm.
Step C: 2-Phenyl-5-carboxypyridine
A suspension of 2-phenyl-5-methyl pyridine (1.03g, 6.09 mmol) and potassium permanganate (2.89g, 18.3 mmol), in water (25 mL) was heated at reflux for 2 hours. The reaction was allowed to cool to ambient temperature and filtered through celite to remove the solids. Acetic acid (1 mL) was added to the colourless filtrate and the product was collected as a white solid by filtration. 1 H NMR (CD3OD, 400MHz) δ 9.18(1H, s), 8.41 (1H, dd, 2.2 and 8.2Hz), 8.08-8.02(2H, m), 7.97(1 H, dd, J=8.2 and 0.7Hz) and 7.56- 7.46(3H, m) ppm.
Step D: 2-Phenyl-5-hydroxymethylpyridine
To a solution of 2-phenyl-5-carboxypyridine (520 mg,
2.61 mmol) in tetrahydrofuran (10 mL) at 0°C was added 1.0 M lithium aluminum hydride in tetrahydrofuran (2.61 mL, 2.61 mmol) over 10 minutes. The reaction was allowed to stir at ambient temperature for 16 hours, cooled to 0°C, and quenched by dropwise addition of water (0.20 mL), 4 N aq. NaOH (0.20 mL), and water (0.60 mL). The reaction was filtered through a pad of Celite and the filtrate evaporated in vacuo. The residue was chromatographed (silica gel, 0-5% MeOH in CH2CI2) to afford the title compound. 1H NMR (CDCI3, 400MHz) δ 8.66(1H, s), 7.97(2H, d, J=7.9Hz), 7.82-7.70(2H, m), 7.52-7.38(3H, m), 4.77(2H, s) and 1.89(1H, brs) ppm.
Step E: 1 -(2-Phenylpyrid-5-ylmethyl)-5-(4-cyanobenzyI)
imidazole hydrochloride salt
To a solution of 2-phenyl-5-hydroxymethylpyridine (264 mg, 1.43 mmol) and diisopropylethylamine (0.522 mL, 3.00 mmol) in dichloromethane (10 mL) at -78°C was added trifluoro- methanesulfonic anhydride (0.252 mL, 1.50 mmol) and the mixture stirred at -78°C for 15 minutes. To this mixture was added a solution of 1 -trityl-4-(4-cyanobenzyl)imidazole (608 mg , 1.43 mmol) in dichloromethane (9 mL). The mixture was allowed to warm to ambient temperature and stirred for 16 hours. The solvent was evaporated in vacuo. The residue was dissolved in methanol (15 mL), heated at reflux for 1 hour, and the solvent evaporated in vacuo. The residue was partitioned between dichloromethane and sat. aq. NaHCO3 solution. The organic layer was dried,
(Na2SO4) and the solvent evaporated in vacuo. The residue was chromatographed (Silica gel, 0-5% NH4OH in CH2CI2). The amine was converted to the HCl salt by treatment with 1.0M HCl in aqueous acetonitrile. Evaporation of the solvent in vacuo afforded the title compound as a white solid.
FAB MS 351 (MH+)
1H NMR (CD3OD, 400MHz) δ 8.38(1H, d, J=2.4Hz),7.97(2H, m), 7.64(1H, d, J=8.2Hz), 7.60(1H, s), 7.56-7.40(5H, m), 7.28-7.20(1H, m), 7.17(2H, d, J=8.0Hz), 6.97(1H, s), 4.96(2H, s) and 3.89(2H, s) ppm.
EXAMPLE 2 1 -(2-Phenyl-N-Oxopyrid-5-ylmethyl)-5-(4-cyanobenzyl)imidazole hydrochloride salt
1 -(2-Phenylpyrid-5-ylmethyl)-5-(4-cyanobenzyl) imidazole hydochloride (66.7mg, 0.159 mmol) was partitioned between CH2Cl2 (1mL) and sat. aq. Na2CO3 (1 mL). The organic layer was separated, dried, (MgSO4) and the solvent evaporated in vacuo. The residue was dissolved in CH2CI2 (2 mL), 3-chloro- perbenzoic acid (109 mg, 0.506 mmol) was added and the solution stirred at ambiant temperature for 16 hours. The reaction was partitioned between CH2Cl2(5mL) and sat. aq. Na2CO3 (2mL) and the organic layer separated, dried, (MgSO4) and the solvent evaporated in vacuo. The residue was chromatographed (Silica gel 4-10% MeOH in CH2Cl2). The amine was converted to the HCl salt by treatment with 1.0M HCl in aqueous acetonitrile. Evaporation of the solvent in vacuo afforded the title compound as a white solid. 1H NMR (CD3OD, 400MHz) δ 9.18(1H, s), 8.13(1H,s), 7.80- 7.20(12H,m), 5.53(2H,s) and 4.28(2H,s) ppm.
EXAMPLE 3
1 -(3-Phenylpyrid-6-ylmethyl)-5-(4-cyanobenzyl)imidazoIe
hydrochloride salt Step A: 3-Phenyl-6-carboxypyridine
A suspension of 3-phenyI-6-methyl pyridine (1.99g, 1 1.78 mmol) and potassium permanganate (7.65, 48.6 mmol), in water (50 mL) was heated at reflux for 16 hours. The reaction was allowed to cool to ambient temperature and filtered through celite to remove the solids. Acetic acid (2 mL) was added to the colourless filtrate and the product was collected as a white solid by filtration. 1H NMR (CD3OD, 400MHz) δ 8.86(1H, s), 8.15(2H,m),
7.70(2H,d, J=6.7Hz) and 7.60-7.30(3H,m) ppm. Step B: 3-Phenyl-6-hydroxymethylpyridine To a solution of 3-phenyl-6-carboxypyridine (1.05g, 5.27 mmol) in tetrahydrofuran (25 mL) at 0°C was added 1.0 M lithium aluminum hydride in tetrahydrofuran (10.0 mL, 10.0 mmol) over 10 minutes. The reaction was allowed to stir at ambient temperature for 6 hours, cooled to 0°C, and quenched by dropwise addition of water (0.50 mL), 4 N aq. NaOH (0.50 mL), and water (1.5 mL). The reaction was filtered through a pad of Celite and the filtrate evaporated in vacuo. The residue was chromatographed (silica gel, 0-5% MeOH in CH2CI2) to afford the title compound. 1H NMR (CDCI3, 400MHz) δ 8.79(1 H, d, J=1.0Hz), 7.88(1H, dd, J=8.6 and 1.5Hz), 7.58(2H,d, J=6.7Hz), 7.49(2H,t, J=7.0Hz), 7.41 (1H,t, J=7.0Hz), 7.33(1H,d, J=7.6Hz), 4.83(2H,s) and
3.75(1H,brs) ppm. Step C: 1 -(3-Phenylpyrid-6-ylmethyl)-5-(4-cyanobenzyl)
imidazole hydrochloride salt
To a solution of 3-phenyl-6-hydroxymethylpyridine (192 mg, 1.04 mmol) and diisopropylethylamine (0.360 mL, 2.07 mmol) in dichloromethane (8 mL) at -78°C was added trifluoro- methanesulfonic anhydride (0.180 mL, 1.07 mmol) and the mixture stirred at -78°C for 1 hour. To this mixture was added a solution of 1 -trityl-4-(4-cyanobenzyl)imidazole (441 mg , 1.04 mmol) in dichloromethane (9 mL). The mixture was allowed to warm to ambient temperature and stirred for 4 hours. The solvent was evaporated in vacuo. The residue was dissolved in methanol (10 mL), heated at reflux for 1 hour, and the solvent evaporated in vacuo. The residue was partitioned between dichloromethane and sat. aq. NaHCO3 solution. The organic layer was dried, (Na2SO4) and the solvent evaporated in vacuo. The residue was chroma- tographed (Silica gel, EtOAc and then 5% MeOH in CH2Cl2 ). The amine was converted to the HCl salt by treatment with 1.0M HCl in aqueous acetonitrile. Evaporation of the solvent in vacuo afforded the title compound as a white solid. FAB HRMS exact mass calcd for C23H19N4 351.160972 (MH+); found 351.161206.
1H NMR (CD3OD, 400MHz) δ 9.20(1H, d, J=1.4Hz), 8.75(1H, d, J=2.2Hz), 8.16(1H, d, J=8.20), 7.66 (2H, d, J=8.4Hz), 7.60-7.40(7H, m), 7.26(2H, d, J=8.0Hz), 5.73(2H, s) and 4.27(2H, s) ppm.
Anal. Calcd. for C23H1 8N4·2.00 H Cl-0.80 H2O:
C, 63.1 1 ; H, 4.97; N, 12.80.
Found: C, 63.10; H, 4.97; N, 12.95. EXAMPLE 4
1-(3-Phenyl-N-Oxopyrid-6-ylmethyl)-5-(4-cyanobenzyl)imidazole hydrochloride salt
1 -(3-Phenylpyrid-6-ylmethyl)-5-(4-cyanobenzyl) imidazole hydochloride (100.0mg, 0.236mmol) was partitioned between CH2Cl2(2mL) and sat. aq. Na2CO3 (lmL). The organic layer was separated, dried, (MgSO4) and the solvent evaporated in vacuo. The residue was dissolved in CH2Cl2 (2 mL), 3-chloroperbenzoic acid (143mg, 0.472 mmol) was added and the solution stirred at ambient temperature for 16 hours. The reaction was partitioned between CH2Cl2 (5mL) and sat. aq. Na2CO3 (2mL) and the organic layer separated, dried, (MgSO4) and the solvent evaporated in vacuo. The residue was chromatographed (Silica gel 4-10% MeOH in
CH2Cl2The amine was converted to the HCl salt by treatment with 1.0M HCl in aqueous acetonitrile. Evaporation of the solvent in vacuo afforded the title compound as a white solid.
1H NMR free base (CDCI3, 400MHz) δ 8.44(1H, d, J=1.5Hz), 7.63(1H,s), 7.60-7.20(10H,m), 7.03(1H,s), 6.35(1H,d, J=8.2Hz), 5.29(2H,s) and 3.96(2H,s) ppm.
EXAMPLE 5
1 -(2-(3-Trifluoromethoxyphenyl)-pyrid-5-yImethyl)-5-(4- cyanobenzyl)imidazole hydrochloride salt Step A: 2-(3-Trifluoromethoxyphenyl)-5-methylpyridine
To a solution of 3-bromotrifluoromethoxybenzene (0.590mL, 4.00 mmol) in THF ( 12 mL) at -78°C was added t-butyl lithium (4.71mL, of a 1.7M solution in pentane, 8.00 mmol. After 10 minutes zinc chloride(4.0mL, of a 1 M solution in diethylether, 4.00 mmol) was added. The reaction was stirred for 10 minutes at -78°C and then allowed to warm to 0°C and stirred for 30minutes. This solution was added via cannula to a solution of 2-bromo-5- methyl pyridine and bis(triphenylphosphine) Nickel II chloride. The reaction stirred for 1 hour at 0°C and then at ambient temperature for a furthur 1 hour. Saturated ammonium hydroxide solution (3 mL) was added and the mixture stirred until homogenous, extracted with Et2O and the organic extracts washed with saturated brine, dried (MgSO4) and evaporated in vacuo. The residue was
chromatographed (Silica gel, 25-50% CH2Cl2 in hexanes).
1H NMR (CD3OD, 400MHz) δ 8.48(1H, s),7.93(1H, brd, J=8.0Hz), 7.87(1 H, s), 7.79(2H, d, J=8.0Hz), 7.74(2H, d, J=8.0Hz), 7.56(1 H, t, J=8.0Hz), 7.32(1H, brd, J=8.0Hz) and 2.40(3H, s) ppm.
Step B: 2-(3-Trifluoromethoxyphenyl)-5-carboxy pyridine
A solution of 2-(3-TrifluoromethoxyphenyI)-5- methylpyridine (2.35g, 2.22 mmol) and tetrabutylammonium permanganate (1.904, 0.012mol), in pyridine (8 mL) was heated at 75°C for 16 hours. The cooled reaction was filtered through celite to remove the solids. The solid was washed with EtOAc and MeOH and the filtrate evaporated in vacuo to afford the title compound of sufficient purity to be used in the next step. Step C: 2-(3-Trifluoromethoxyphenyl)-5- hydroxymethylpyridine
To a solution of 2-(3-trifluoromethoxyphenyl)-5- carboxy pyridine (2.0 g, 7.06 mmol) in tetrahydrofuran ( 15 mL) at 0°C was added 1.0 M lithium aluminum hydride in tetrahydrofuran (7.07 mL, 7.07 mmol) over 10 minutes. The reaction was allowed to stir at ambient temperature for 4 hours, cooled to 0°C, and quenched by dropwise addition of saturated Na2SO4 (1.0 mL). The reaction was diluted with diethylether, filtered through a pad of Celite and the filtrate evaporated in vacuo. The residue was chromatographed (silica gel, 50% EtOAc in hexanes) to afford the title compound.
1H NMR (CD3OD, 400MHz) δ 8.62(1 H, d, J=1.0Hz), 8.00- 7.84(H,m), 7.57(1H, t, J=8.0Hz), 7.33(1H,brd, J=8.0Hz) and
4.84(2H,s) ppm.
Step D: 1 -(2-(3-Trifluoromethoxyphenyl)-pyrid-5-ylmethyl)-
5-(4-cyanobenzyl)imidazole hydrochloride salt
To a solution of 2-(3-trifluoromethoxyphenyl)-5- hydroxymethylpyridine (66 mg, 0.25 mmol), diisopropylethylamine (0.085 mL, 0.49 mmol), and 1 -trityl-4-(4-cyanobenzyl)imidazole (105 mg , 0.25 mmol) in dichloromethane (1.4 mL) at -78°C was added trifluoromethanesulfonic anhydride (0.041 mL, 0.25 mmol) and the mixture stirred at -78°C for 1 hour. The reaction was allowed to warm to ambient temperature and stirred for 4 hours. The solvent was evaporated in vacuo. The residue was dissolved in methanol (15 mL), heated at reflux for 1 hour, and the solvent evaporated in vacuo. The residue was partitioned between dichloromethane and sat. aq. Na2CO3 solution. The organic layer was dried, (Na2SO4) and the solvent evaporated in vacuo. The residue was chromatographed (Silica gel, 3% MeOH in CH2Cl2). The amine was converted to the HCl salt by treatment with 1.0M HCl in aqueous acetonitrile. Evaporation of the solvent in vacuo afforded the title compound as a white solid.
1 H NMR (CD3OD, 400MHz) δ 9.23(1H, s), 8.67(1H,s), 8.18- 8.04(2H, m), 8.00-7.90(2H,m), 7.74(1H, t, J=7.9Hz), 7.62-7.50(4H, m), 7.31 (2H, d, J=7.9Hz), 5.71 (2H, s), 4.29(2H, s) ppm. FAB HRMS exact mass calcd for C24H18N4 OF3 435.143271 (MH+); found 435.144474.
Anal. Calcd. for C24H17N4 OF3 ·2.00 HCl:
C, 56.82; H, 3.77; N, 1 1.04.
Found: C, 56.50; H, 3.88; N, 10.86.
EXAMPLE 6
1 -(2-(2-Trifluoromethylphenyl)-pyrid-5-ylmethyl)-5-(4- cyanobenzyl)imidazole hydrochloride salt
Step A: 2-(2-Trifluoromethylphenyl)-5-methylpyridine
To a solution of 2 bromo-5-methyl pyridine
(1.81g, 10.53 mmol) and barium hydroxide (4.97 g, 15.78 mmol) in water (15 mL) was added DME (80 mL). This mixture was treated sequentially with 2-(trifluoromethyI)phenylboronic acid
(2.00g, 10.53 mmol) and palladium tetrakis(triphenylphosphine)
(553 mg, 0.48 mmol) and the mixture warmed to 80°C for 48 hours.
Water (100mL) was added and the pH of the solution was adjusted to 10 and extracted with EtOAc (3X200mL).
The organic extracts were combined, washed with brine, dried (MgSO4), and the solvent evaporated in vacuo. The residue was chromatographed (Silica gel, 50% -100% CH2Cl2 in hexanes) to afford the title compound.
1H NMR (CDCI3, 400MHz) δ 8.52(1 H, s), 7.75(1H, d, J=7.9Hz),
7.64-7.44(4H, m), 7.32(1 H, d, J=7.9Hz) and 2.40(3H,s) ppm.
Step B: 2-(2-Trifluoromethylphenyl)-5-carboxypyridine
A suspension of 2-(2-Trifluoromethylphenyl)-5- methylpyridine (0.40g, 1.68 mmol) and potassium permanganate (1.60g, 10.1 mmol), in water (10 mL) was heated at reflux for 16 hours. The reaction was filtered hot through celite to remove the solids. Acetic acid was added to the colourless filtrate to yield a pH of 5 and the resulting suspension was extracted with CH2Cl2.washed with water (10 mL), dried, (MgSθ4), and the solvent evaporated in vacuo to afford the title compound.
1H NMR (CD3OD, 400MHz) δ 9.34(1H, s), 8.41 (1H,d, J=8.2Hz),
7.80(1 H,d, J=7.9Hz) and 7.70-7.50(4H,m) ppm.
Step C: 2-(2-Trifluoromethylphenyl)-5-hydroxymethylpyridine
To a solution of 2-(2-Trifluoromethylphenyl)-
5-carboxypyridine (220 mg, 1.23 mmol) in tetrahydrofuran
(10 mL) at 0°C was added 1.0 M lithium aluminum hydride in tetrahydrofuran (1.23 mL, 1.23 mmol) over 10 minutes. The reaction was allowed to stir at ambient temperature for 16 hours, cooled to 0°C, and quenched by dropwise addition of water (0.05 mL), 2.5 N aq. NaOH (0.05 mL), and water (0.15 mL). Sodium sulfate was added, the reaction filtered through a pad of Celite and the filtrate evaporated in vacuo. The residue was chromatographed
(silica gel, CH2Cl2 then EtOAc) to afford the title compound.
1H NMR (CDCI3, 400MHz) δ 8.63(1 H, s), 7.80-7.40(6H,m) and
4.77(2H, s) ppm. Step D: 1 -(2-(2-Trifluoromethylphenyl)-pyrid-5-ylmethyI)-
5-(4-cyanobenzyl)imidazole hydrochloride salt
The title compound was prepared using the procedure described for Example 5, step D using 2-(2-trifluoromethylphenyl)
-5-hydroxymethylpyridine from Step C in place of 2-(3-trifluoro- methoxyphenyI)-5-hydroxymethylpyridine.
1 H NMR (CD3OD, 400MHz) δ 9.17( 1H, s), 8.42(1 H,s), 8.00-
7.40(11H, m), 5.60(2H, s), 4.26(2H, s) ppm.
FAB MS 419 (MH+)
Anal. Calcd. for C24H17N4 F3 ·2.95 HCl. 0.6 EtOAc:
C, 54.78; H, 4.31 ; N, 9.68.
Found: C, 54.79; H, 4.18; N, 9.68.
EXAMPLE 7 1 -(3-Phenyl-2-Chloropyrid-6-ylmethyl)-5-(4-cyanobenzyl)imidazole hydrochloride salt
Step A: 3-Phenyl-6-methylpyridine N-oxide
A solution of 3-phenyl -6-me thyl pyridine (2.36g, 13.95 mmol), in CH2Cl2 (40 mL) at 0°C was treated with MCPBA (3.58g, 13.95 mmol) for 1 hour. Saturated aq. Na2CO3 (50 mL) was added and the reaction was extracted with CH2Cl2 (20 mL). The organic extracts were dried (MgSO4), and the solvent evaporated in vacuo to afford the title compound.
1 H NMR (CDCl3, 400MHz) δ 8.53(1H, s), 7.60-7.20(7H, m) and 2.57(3H, s) ppm.
Step B: 3-Phenyl-2-chloro-6-methylpyridine and 3-phenyI-4- chloro-6-methylpyridine
A solution of 3-phenyl-6-methyl pyridine-N-Oxide (1.42g, 7.66 mmol), in P2O5 (50 mL) at 0°C was at 80°C for 3 hours. The reaction was allowed to cool to room temperature and then poured over ice (400g). Saturated aq. Na2CO3 was added until the pH of the solution was 8 and the reaction was extracted with CH2Cl2 (3X250 mL). The organic extracts were dried (MgSO4), and the solvent evaporated in vacuo. The residue was chromatographed (silica gel, 10-20% EtOAc in CH2Cl2 to afford 3-Phenyl-2-chloro- 6-methylpyridine (First eluted)
1H NMR (CDCI3, 400MHz) δ 7.56(1H, d, J=7.6Hz), 7.60- 7.30(5H,m), 7.15(1H,d, J=7.6Hz) and 2.59(3H, s) ppm.
3-Phenyl-4-chloro-6-methylpyridine (Second eluted).
1H NMR (CDCl3, 400MHZ) δ 8.43(1 H, s), 7.60-7.40(5H,m), 7.29(1H,s) and 2.59(3H, s) ppm.
Step C: 3-Phenyl-2-chloro-6-bromomethylpyridine
A solution of 3-Phenyl-2-chloro-6-methylpyridine (0.094g, 0.462 mmol), NBS (0.086g, 0.485 mmol) and AIBN
(0.008g, 0.046mmol) in CCl4 (3 mL) were heated at reflux for 2 hours. The solvent was evaporated and the residue chroma- tographed(Silica gel, 100% CH2Cl2 to afford the title compound. 1H NMR (CDCl3, 400MHz) δ 7.68(1H, d, J=7.6Hz), 7.60- 7.40(6H,m), and 4.56(2H, s) ppm.
Step D: 1 -(3-Phenyl-2-chloropyrid-6-ylmethyl)-5-(4- cyanobenzyl)imidazole hydrochloride salt
To 1 -trityl-4-(4-Cyanobenzyl)-imidazole (88.4mg, 0.208 mmol) in acetonitrile (1 mL) was added 3-phenyl-2-chloro- 6-bromomethylpyridine (53.5mg, 0.189 mmol) and the mixture heated at 65°C for 16 hours. The residue was dissolved in methanol (3 ml) and heated at reflux for 2 hours, cooled and evaporated to dryness. The residue was partitioned between sat. aq. Na2CO3 solution and CH2Cl2. The organic layer was dried, (MgSO4) and the solvent evaporated in vacuo. The residue was chromatographed (Silica gel, 2.5-3% MeOH in CH2Cl2) to afford the free base which was converted to the HCl salt by treatment with one equivalent of HCl in aqueous acetonitrile. Evaporation of solvent in vacuo afforded the title compound as a white powder.
1H NMR (CD3OD, 400MHz) δ 9.11 (1H, s), 7.64(1H.d, J=7.7Hz), 7.55(2H,d, J=8.2Hz), 7.51 (1H,s), 7.50-7.34(5H,m), 7.32-7.20(3H, m), 5.56(2H, s), 4.27(2H, s) ppm.
Anal. Calcd. for C23H17CIN4 ·1.00 HCl. 0.6 EtOAc:
C, 54.78; H, 4.31 ; N, 9.68.
Found: C, 54.79; H, 4.18; N, 9.68.
EXAMPLE 8
1 -(3-Phenyl-4-chIoropyrid-6-ylmethyl)-5-(4-cyanobenzyl)imidazoIe hydrochloride salt
The title compound was prepared using the procedure described for Example 7, steps C and D using 3-phenyl-4-chloro-6-methylpyridine in place of 3-phenyl-6-methyl pyridine. Anal. Calcd. for C24H17N4 Cl·1.00 HCl. 0.30 H2O:
C, 64.74; H, 4.39; N, 13.13.
Found: C, 64.82; H, 4.52; N, 12.93. EXAMPLE 9
1 -(2-Amino-3-phenylpyrid-6-ylmethyl)-5-(4-cyanobenzyl)imidazole hydrochloride salt
Step A: 2-Amino-3-Phenyl-6-methylpyridine
A solution of 3-phenyl-6-methyl pyridine
(0.815 g, 4.82 mmol), and sodium amide (752mg, 19.3mmol) in diethylaniline (10mL) was heated at 180°C for 72 hours. The reaction was cooled and quenched with ice (lOOg), and the mixture extracted with EtOAc. The organic extract was washed with brine (50 mL) , dried (MgSO4), silica gel (100g) was added and the solvent evaporated in vacuo.
The material was loaded onto a column and chromatographed (Silica gel, eluting with 0-100% EtOAc in CH2Cl2) to afford the title compound.
1H NMR (CDCI3, 400MHz) δ 7.50-7.20(6H, m) 6.61 ( l H,d, J=7.0Hz), and 2.42(3H, s) ppm.
Step B: N-bis t-Butoxycarbonyl-2-Amino-3-Phenyl-6- methylpyridine
A solution of 2-amino-3-phenyl-6-methyl pyridine (1.21 g, 6.57 mmol), di t-butylcarbonate(3.58g, 16.4 mmol), triethylamine (2.29 mL, 16.4 mmol) and DMAP (0.803g, 6.57 mmol) in CH2Cl2 (20mL) were heated at 65°C for 16 hours. The reactionwas diluted with sat. aq. Na2C03 and extracted with CH2Cl2 The solvent was evaporated in vacuo. and the residue chromatographed (Silica gel, eluting with 20% EtOAc in CH2Cl2) to afford the title compound. 1H NMR (CDCI3, 400MHz) δ 7.62( 1 H, d, J=7.7Hz), 7.41 -7.30(5H, m), 7.19(1H, d, J=7.7Hz), 2.59(3H, s) and 1.28(18H, s) ppm.
Step C: 2-(bis t-butoxycarbonylamino)-3-phenyl-6- methylpyridine-N-oxide
A solution of N-bis t-butoxycarbonyI-2-amino-3-phenyl-
6-methylpyridine (0.215g, 0.56 mmol), in CH2Cl2 (4 mL) at 0°C was treated with MCPBA (0.220g , 0.727 mmol) for 1 hour. Saturated aq. Na2CO3 (50 mL) was added and the reaction was extracted with CH2Cl2 (2X50 mL). The organic extracts were dried (MgSO4), and the solvent evaporated in vacuo. The residue was chromatographed (Silica gel, eluting with 100% EtOAc to afford the title compound. 1 H NMR (CDCI3, 400MHz) δ 7.44-7.36(6H,m), 7.13(1H, d,
J=7.7Hz), 2.56(3H, s) and 1.31(18H, s) ppm. Step D: N-bis t-Butoxycarbonyl-2-amino-3-phenyl-6- acetoxymethylpyridine
A solution of 2-(bis t-butoxycarbonylamino)-3-phenyl- 6-methylρyridine-N-oxide (0.223g, 0.557 mmol), in acetic anhydride (5 mL) was heated at 65°C for 24 hours. The solvent was evaporated in vacuo and the residue chromatographed (30-50%EtOAc in hexanes) to afford the title compound.
1 H NMR (CDCl3, 400MHz) δ 7.74(1H, d, J=7.7Hz), 7.50-7.30(6H, m), 5.25(2H, s), 2.17(3H, s) and 1.28(18H, s) ppm. Step E: N-bis t-Butoxycarbonyl-2-amino-3-phenyl-6- hydroxymethylpyridine
A solution of 2-(bis t-butoxycarbonylamino)-3-phenyl- 6-acetoxymethylpyridine (0.040g, 0.09 mmol), THF (1.3 mL) was treated with Lithium hydroxide (1M solution in water 0.271 ml, 0.271 mmol) at room temperature for 16 hours. The reaction was diluted with water and extracted with CH2Cl2. The organic extracts were dried (MgSO4),and the solvent evaporated in vacuo to afford the title compound.
1H NMR (CDCI3, 400MHz) δ 7.74(1H, d, J=7.8 Hz), 7.44-7.33(5H, m), 7.31( 1H,brd, J=7.8Hz), 4.81 (2H, s), and 1.29(18H, s) ppm.
Step F: 1-(2-Amino-3-phenylpyrid-6-ylmethyl)-5-(4- cyanobenzyl)imidazole hydrochloride salt
The title compound was prepared using the procedure described for Example 3 step C using N-bis t-butoxycarbonyl-2-amino-3-phenyl- 6-hydroxymethylpyridine in place of 3-phenyl-6-hydroxymethyl- pyridine. In this case the free base was treated with TFA and triethylsilane to effect cleavage of the t-butoxycarbonyl groups which was followed by its conversion to the hydrochloride salt.
1H NMR (CD3OD, 400MHz) δ 9.23(1 H, s), 7.80-7.20(H, m), 6.96(l H,s), 6.65(1H,d, J=7.6Hz), 5.66(2H, s), 4.33(2H, s) ppm.
Anal. Calcd. for C23H19N5·1.00 HCl. 0.95 H2O 0.35 EtOAc:
C, 60.26; H, 5.33; N, 14.40.
Found: C, 60.04; H, 5.10; N, 14.45.
EXAMPLE 10
In vitro inhibition of ras farnesyl 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 al., J. Biol. Chem. 265: 14701 -14704 ( 1990), Pompliano. et al.. Biochemistry 31 :3800 (1992) and Gibbs et al., PNAS U.S.A. 86:6630-6634 ( 1989), respectively. Bovine FPTase was assayed in a volume of 100 μl containing 100 mM N-(2-hydroxy ethyl) piperazine-N '-(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 nM 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 compound of the instant invention described in the above Examples 1-9 were tested for inhibitory activity against human FPTase by the assay described above and were found to have IC50 of ≤50 μM.
EXAMPLE 1 1 In vivo ras farnesylation assay
The cell line used in this assay is a v-ras line derived from either Ratl or NIH3T3 cells, which expressed viral Ha-ras p21. The assay is performed essentially as described in DeClue, J.E. et 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 mCi[35Sjmethionine (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 MgCl2/1mM 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 nonfarnesylated ras proteins are compared to
determine the percent inhibition of farnesyl transfer to protein.
EXAMPLE 12
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 Rat1 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 104 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.

Claims

WHAT IS CLAIMED IS:
1. A compound which inhibits farnesyl-protein transferase of the formula A:
Figure imgf000098_0001
wherein: from 1 -2 of f(s) are independently N or N->O, and the remaining f's are independently CH;
R1 and R2 are independently selected from:
a) hydrogen,
b) aryl, heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, R10O-, R11S(O)m-, R10C(O)NR10-, R 1 1C(O)O-, (R10)2NC(O)-, R 10 2N-C(NR10)-, CN, NO2, R10C(O)-, N3, -N(R10)2, or R11OC(O)NR10-, c) unsubstituted or substituted C1 -C6 alkyl wherein the substituent on the substituted C1 -C6 alkyl is selected from unsubstituted or substituted aryl, heterocyclic, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl,
R 10O-, R11 S(O)m-, R10C(O)NR10-, (R 10)2NC(O)-, R10 2N-C(NR10)-, CN, R10C(O)-, N3, -N(R10)2, and R11OC(O)-NR10-; R3, R4 and R5 are independently selected from: a) hydrogen,
b) unsubstituted or substituted aryl, unsubstituted or
substituted heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, halogen, C1 -C6 perfluoroalkyl, R12O-, R1 1 S(O)m-, R10C(O)NR10-, (R10)2NC(O)-, R1 1 C(O)O-, R10 2N-C(NR10)-, CN, NO2, R10C(O)-, N3, -N(R10)2, or R11OC(O)NR10-,
c) unsubstituted C1 -C6 alkyl,
d) substituted C1 -C6 alkyl wherein the substituent on the
substituted C1 -C6 alkyl is selected from unsubstituted or substituted aryl, unsubstituted or substituted heterocyclic, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl,
R12O-, R11 S(O)m-, R10C(O)NR10-, (R10)2NC(O)-, R10 2N-C(NR10)-, CN, R10C(O)-, N3, -N(R 10)2, and R11OC(O)-NR10-;
R6a, R6b, R6c, R6d and R6e are independently selected from:
a) hydrogen,
b) unsubstituted or substituted aryl, unsubstituted or
substituted heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, halogen, C1 -C6 perfluoroalkyl, R1 2O-, R 11S(O)m-, R10C(O)NR10-, (R10)2NC(O)-, R11C(O)O-, R10 2N-C(NR10)-, CN, NO2, R10C(O)-, N3, -N(R10)2, or R11OC(O)NR10-,
c) unsubstituted C1 -C6 alkyl,
d) substituted C1 -C6 alkyl wherein the substituent on the
substituted C1 -C6 alkyl is selected from unsubstituted or substituted aryl, unsubstituted or substituted heterocyclic, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl,
R12O-, R11 S(O)m-, R10C(O)NR10., (R10)2NC(O)-, R10 2N-C(NR10)-, CN, R10C(O)-, N3, -N(R10)2, and R11OC(O)-NR10-; or any two of R6a, R6b, R6c, R6d and R6e on adjacent carbon atoms are combined to form a diradical selected from -CH=CH-CH=CH-, -CH=CH-CH2-, -(CH2)4- and -(CH2)3-; provided that when R3, R4, R5, R6a, R6b, R6c, R6d or R6e is unsubstituted or substituted heterocycle, attachment of R3, R4, R5, R6a, R6b, R6c, R6d or R6e to the 6-membered heteroaryl ring, or phenyl ring respectively, is through a substitutable heterocycle ring carbon;
R7 is 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,
Figure imgf000100_0001
f) — SO2R11
g) N(R 10)2 or
h) C1 -4 perfluoroalkyl;
R8 is independently selected from:
a) hydrogen,
b) aryl, substituted aryl, heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, perfluoroalkyl, F, Cl, Br, R10O-, R 1 1 S(O)m-, R10C(O)NR10-, (R 10)2NC(O)-, R10 2N-C(NR10)-, CN, NO2, R10C(O)-, N3, -N(R 10)2, or R11OC(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, R10O-, R11S(O)m-, R10C(O)NH-, (R10)2NC(O)-,
R10 2N-C(NR10)-, CN, R10C(O)-, N3, -N(R10)2, or R10OC(O)NH-;
provided that when R8 is heterocycle, attachment of R8 to V is through a substitutable ring carbon;
R9 is independently selected from:
a) hydrogen,
b) alkenyl, alkynyl, perfluoroalkyl, F, Cl, Br, R11O-,
R11S(O)m-, R10C(O)NR10-, (R10)2NC(O)-, R10 2N-C(NR10)-, CN, NO2, R10C(O)-, N3, -N(R10)2, or R11OC(O)NR10-, and
c) C1 -C6 alkyl unsubstituted or substituted by perfluoroalkyl, F, Cl, Br, R10O-, R1 1S(O)m-, R10C(O)NR10-, (R10)2NC(O)-, R10 2N-C(NR10)-, CN, R10C(O)-,
N3, -N(R10)2, or R11OC(O)NR10-; R10 is independently selected from hydrogen, C1 -C6 alkyl, benzyl,
2,2,2-trifluoroethyI and aryl; R11 is independently selected from C1 -C6 alkyl and aryl;
R12 is independently selected from hydrogen, C1 -C6 alkyl, C1 -C6
aralkyl, C1 -C6 substituted aralkyl, C1 -C6 heteroaralkyl, C1 -C6 substituted heteroaralkyl, aryl, substituted aryl, heteroaryl, substituted heteraryl, C1 -C6 perfluoroalkyl, 2-aminoethyl and 2,2,2-trifluoroethyl;
A1 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 heteroatom selected from O, S, and N, and e) C2-C20 alkenyl,
provided that V is not hydrogen if A1 is S(O)m and V is not hydrogen if A1 is a bond, n is 0 and A2 is S(O)m;
provided that when V is heterocycle, attachment of V to R8 and to A1 is through a substitutable ring carbon;
W is a heterocycle;
X is a bond, -CH=CH-, O, -C(=O)-, -C(O)NR7-, -NR7C(O)-, -C(O)O-,
-OC(O)-, -C(O)NR7C(O)-, -NR7-, -S(O)2N(R10)-,
-N(R10)S(O)2- or -S(=O)m-; m is 0, 1 or 2;
n is independently 0, 1 , 2, 3 or 4;
p is independently 0, 1 , 2, 3 or 4;
q is 0, 1 , 2 or 3;
r is 0 to 5, provided that r is 0 when V is hydrogen; and t is 0 or 1 ; or a pharmaceutically acceptable salt thereof.
2. The compound according to Claim 1 of the formula A:
Figure imgf000103_0001
wherein: from 1 -2 of f(s) are independently N or N->0, and the remaining f's are independently CH;
R 1 is independently selected from: hydrogen, C3-C10 cycloalkyl, R10O-, -N(R10)2, F or C1 -C6 alkyl; R2 is independently selected from:
a) hydrogen,
b) aryl, heterocycle, C3-C10 cycloalkyl, R10O-, -N(R10)2, F or C2-C6 alkenyl,
c) unsubstituted or substituted C1 -C6 alkyl wherein the
substituent on the substituted C1 -C6 alkyl is selected from unsubstituted or substituted aryl, heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, R10O- and -N(R10)2;
R3, R4 and R5 are independently selected from:
a) hydrogen,
b) unsubstituted or substituted aryl, unsubstituted or
substituted heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, halogen, C1 -C6 perfluoroalkyl, R12O-, R11S(O)m-, R10C(O)NR10-, (R 10)2NC(O)-, R10 2N-C(NR10)-, CN, NO2, R10C(O)-, N3, -N(R10)2, or R11OC(O)NR10-,
c) unsubstituted C1 -C6 alkyl;
d) substituted C1 -C6 alkyl wherein the substituent on the
substituted C1 -C6 alkyl is selected from unsubstituted or substituted aryl, unsubstituted or substituted heterocyclic, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, R12O-, R1 1S(O)m, R10C(O)NR10-, (R10)2NC(O)-, R10 2N- C(NR10)-, CN, R10C(O)-, N3, -N(R10)2, and R11OC(O)- NR10-;
R6a, R6b, R6c, R6d and R6e are independently selected from:
a) hydrogen,
b) unsubstituted or substituted aryl, unsubstituted or
substituted heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, halogen, C1 -C6 perfluoroalkyl, R 1 2O-, R 1 1S(O)m-, R10C(O)NR10-, (R10)2NC(O)-, R10 2N-C(NR10)-, CN, NO2, R10C(O)-, N3, -N(R10)2, or R11OC(O)NR10-,
c) unsubstituted C1 -C6 alkyl;
d) substituted C1 -C6 alkyl wherein the substituent on the substituted C1 -C6 alkyl is selected from unsubstituted or substituted aryl, unsubstituted or substituted heterocyclic, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, R12O-, R11S(O)m-, R10C(O)NR10-, (R10)2NC(O)-, R10 2N-C(NR10)-, CN,
R10C(O)-, N3, -N(R10)2, and R11OC(O)-NR10-, or any two of R6a, R6b, R6c, R6d and R6e on adjacent carbon atoms are combined to form a diradical selected from -CH=CH-CH=CH-, -CH=CH-CH2-, -(CH2)4- and -(CH2)3-; provided that when R3, R4, R5, R6a, R6b, R6c, R6d or R6e is unsubstituted or substituted heterocycle, attachment of R3, R4, R5, R6a, R6b, R6c, R6d or R6e to the 6-membered heteroaryl ring, or phenyl ring respectively, is through a substitutable heterocycle ring carbon;
R7 is 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,
Figure imgf000105_0001
f) -SO2R11
g) N(R10)2 or
h) C1 -4 perfluoroalkyl; R8 is independently selected from:
a) hydrogen,
b) aryl, substituted aryl, heterocycle, 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 R11OC(O)NR10-, and c) C1 -C6 alkyl substituted by C1 -C6 perfluoroalkyl, R10O-, R10C(O)NR10-, (R 10)2N-C(NR10)-, R10C(O)-,
-N(R10)2, or R11OC(O)NR10-;
provided that when R8 is heterocycle, attachment of R8 to V is through a substitutable ring carbon;
R9 is selected from:
a) hydrogen,
b) C2-C6 alkenyl, C2-C6 alkynyl, C1 -C6 perfluoroalkyl, F, Cl, R11O-, R 1 1S(O)m-, R10C(O)NR10-, (R10)2NC(O)-, CN, NO2, (R10)2N-C(NR10)-, R10C(O)-, -N(R10)2, or R11OC(O)NR10-, and
c) C1 -C6 alkyl unsubstituted or substituted by C1 -C6
perfluoroalkyl, F, Cl, R10O-, R11S(O)m-, R10C(O)NR10-, (R10)2NC(O)-, CN, (R10)2N-C(NR10)-, R10C(O)-, -N(R10)2, or R11OC(O)NR10-; R10 is independently selected from hydrogen, C1-C6 alkyl, benzyl,
2,2,2-trifIuoroethyl and aryl; R11 is independently selected from C1 -C6 alkyl and aryl;
R 1 2 is independently selected from hydrogen, C1 -C6 alkyl, C1 -C6
aralkyl, C1 -C6 substituted aralkyl, C1 -C6 heteroaralkyl, C1 -C6 substituted heteroaralkyl, aryl, substituted aryl, heteroaryl, substituted heteraryl, C1 -C6 perfluoroalkyl, 2-aminoethyl and 2,2,2-trifluoroethyl;
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,
imidazolinyl, pyridinyl, thiazolyl, oxazolyl, indolyl, quinolinyl, isoquinolinyl, triazolyl and thienyl, c) aryl,
d) C1 -C20 alkyl wherein from 0 to 4 carbon atoms are
replaced with 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;
provided that when V is heterocycle, attachment of V to R8 and to A1 is through a substitutable ring carbon; W is a heterocycle selected from pyrrolidinyl, imidazolyl, imidazolinyl, pyridinyl, thiazolyl, oxazolyl, indolyl, quinolinyl, triazolyl or
isoquinolinyl;
X is a bond, O, -C(=O)-, -CH=CH-, -C(O)NR7-, -NR7C(O)-, -NR7-,
-S(O)2N(R10)-, -N(R10)S(O)2- or -S(=O)m-; m is 0, 1 or 2;
n is independently 0, 1 , 2, 3 or 4;
p is independently 0, 1 , 2, 3 or 4;
q is 0, 1 , 2 or 3;
r is 0 to 5, provided that r is 0 when V is hydrogen; and t is 0 or 1; or a pharmaceutically acceptable salt thereof.
3. The compound according to Claim 1 of the formula B
Figure imgf000107_0001
wherein: from 1 -2 of f(s) are independently N or N->O, and the remaining f's are independently CH; R1 is selected from: hydrogen, C3-C10 cycloalkyl, R10O-, -N(R10)2, F or C1 -C6 alkyl; R2 is independently selected from:
a) hydrogen,
b) aryl, heterocycle, C3-C10 cycloalkyl, R10O-, -N(R10)2, F or C2-C6 alkenyl,
c) unsubstituted or substituted C1 -C6 alkyl wherein the
substituent on the substituted C1 -C6 alkyl is selected from unsubstituted or substituted aryl, heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, R10O- and -N(R10)2; R3 and R4 are independently selected from:
a) hydrogen,
b) unsubstituted or substituted aryl, unsubstituted or
substituted heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, halogen, C1 -C6 perfluoroalkyl, R12O-, R1 1 S(O)m-, R10C(O)NR10-, (R10)2NC(O)-, R10 2N-C(NR10)-, CN, NO2, R10C(O)-, N3, -N(R10)2, or R11OC(O)NR10-,
c) unsubstituted C1 -C6 alkyl,
d) substituted C1 -C6 alkyl wherein the substituent on the substituted C1 -C6 alkyl is selected from unsubstituted or substituted aryl, unsubstituted or substituted heterocyclic, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, R12O-, R11S(O)m-, R10C(O)NR10-, (R10)2NC(O)-, R10 2N-C(NR10)-, CN, R10C(O)-, N3, -N(R10)2, and R11OC(O)-NR10-;
R6a, R6b, R6c, R6d and R6e are independently selected from:
a) hydrogen,
b) unsubstituted or substituted aryl, unsubstituted or
substituted heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, halogen, C1 -C6 perfluoroalkyl, R12O-, R11S(O)m-, R10C(O)NR10-, (R10)2NC(O)-, R10 2N-C(NR10)-, CN, NO2, R10C(O)-, N3, -N(R10)2, or R11OC(O)NR10-, c) unsubstituted C1 -C6 alkyl,
d) substituted C1 -C6 alkyl wherein the substituent on the
substituted C1 -C6 alkyl is selected from unsubstituted or substituted aryl, unsubstituted or substituted heterocyclic, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl,
R1 2O-, R11S(O)m-, R10C(O)NR10-, (R10)2NC(O)-, R10 2N-C(NR10)-, CN, R10C(O)-, N3, -N(R10)2, and R11OC(O)-NR10-; or any two of R6a, R6b, R6c, R6d and R6e on adjacent carbon atoms are combined to form a diradical selected from -CH=CH-CH=CH-, -CH=CH-CH2-, -(CH2)4- and -(CH2)3-; provided that when R3, R4, R6a, R6b, R6c, R6d or R6e is
unsubstituted or substituted heterocycle, attachment of
R3, R4, R6a, R6b, R6c, R6d or R6e to the 6-membered heteroaryl ring, or phenyl ring respectively, is through a substitutable heterocycle ring carbon; R8 is independently selected from:
a) hydrogen,
b) aryl, substituted aryl, heterocycle, C1 -C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1 -C6 perfluoroalkyl, F, Cl, R10O-, R10C(O)NR10-, CN, NO2, (R 10)2N-C(NR10)-, R10C(O)-, -N(R10)2, or R11OC(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 R11OC(O)NR10-;
provided that when R8 is heterocycle, attachment of R8 to V is through a substitutable ring carbon;
R9a and R9b are independently hydrogen, C1 -C6 alkyl, trifluoromethyl and halogen; R10 is independently selected from hydrogen, C1 -C6 alkyl, benzyl, 2,2,2-trifIuoroethyl and aryl; R11 is independently selected from C1 -C6 alkyl and aryl;
R12 is independently selected from hydrogen, C1 -C6 alkyl, C1 -C6
aralkyl, C1 -C6 substituted aralkyl, C1 -C6 heteroaralkyl, C1 -C6 substituted heteroaralkyl, aryl, substituted aryl, heteroaryl, substituted heteraryl, C1 -C6 perfluoroalkyl, 2-aminoethyl and 2,2,2-trifluoroethyl;
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,
imidazolinyl, pyridinyl, thiazolyl, 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 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 A1 is a bond, n is 0 and A2 is S(O)m;
provided that when V is heterocycle, attachment of V to R8 and to A1 is through a substitutable ring carbon;
X is a bond, -CH=CH-, -C(O)NR10-, -NR10C(O)-, -NR10-, O or -C(=O)-; m is 0, 1 or 2;
n is independently 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.
4. The compound according to Claim 1 of the formula C:
Figure imgf000111_0001
wherein: from 1 -2 of f(s) are independently N or N->0, and the remaining f's are independently CH;
R 1 is selected from: hydrogen, C3-C10 cycloalkyl, R10O-, -N(R10)2, F or C1 -C6 alkyl; R2 is independently selected from:
a) hydrogen,
b) aryl, heterocycle, C3-C10 cycloalkyl, R10O-, -N(R 10)2, F or C2-C6 alkenyl,
c) unsubstituted or substituted C1 -C6 alkyl wherein the
substituent on the substituted C1 -C6 alkyl is selected from unsubstituted or substituted aryl, heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, R10O- and -N(R 10)2;
R3 and R4 are independently selected from:
a) hydrogen,
b) unsubstituted or substituted aryl, unsubstituted or
substituted heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, halogen, C1 -C6 perfluoroalkyl, R12O-, R1 1S(O)m-, R10C(O)NR10-, CN(R 10)2NC(O)-, R10 2N-C(NR10)-, CN, NO2, R10C(O)-, N3, -N(R10)2, or R 1 1 OC(O)NR10-,
c) unsubstituted C1 -C6 alkyl,
d) substituted C1 -C6 alkyl wherein the substituent on the substituted C1 -C6 alkyl is selected from unsubstituted or substituted aryl, unsubstituted or substituted heterocyclic. C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, R12O-, R 1 1 S(O)m-, R10C(O)NR10-, (R10)2NC(O)-, R10 2N-C(NR10)-, CN, R10C(O)-, N3, -N(R10)2, and R 1 1 OC(O)-NR10-;
R6a, R6b, R6c, R6d and R6e are independently selected from:
a) hydrogen,
b) unsubstituted or substituted aryl, unsubstituted or
substituted heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, halogen, C1 -C6 perfluoroalkyl, R12O-, R11S(O)m-, R10C(O)NR10-, CN(R10)2NC(O)-, R10 2N-C(NR10)-, CN, NO2, R10C(O)-, N3, -N(R10)2, or R11OC(O)NR10-,
c) unsubstituted C1 -C6 alkyl ,
d) substituted C1 -C6 alkyl wherein the substituent on the substituted C1 -C6 alkyl is selected from unsubstituted or substituted aryl, unsubstituted or substituted heterocyclic, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, R12O-, R 1 1S(O)m-, R10C(O)NR10-, (R10)2NC(O)-, R10 2N-C(NR10)-, CN, R10C(O)-, N3, -N(R10)2, and R11OC(O)-NR10-; or any two of R6a, R6b, R6c, R6d and R6e on adjacent carbon atoms are combined to form a diradical selected from -CH=CH-CH=CH-, -CH=CH-CH2-, -(CH2)4- and -(CH2)3-; provided that when R3, R4, R6a, R6b, R6c, R6d or R6e is unsubstituted or substituted heterocycle, attachment of R3, R4, R6a, R6b, R6c, R6d or R6e to the 6-membered heteroaryl ring, or phenyl ring respectively, is through a substitutable heterocycle ring carbon;
R8 is independently selected from:
a) hydrogen,
b) aryl, substituted aryl, heterocycle, 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 R11OC(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 R11OC(O)NR10-;
provided that when R8 is heterocycle, attachment of R8 to V is through a substitutable ring carbon;
R9a and R9b are independently hydrogen, C1 -C6 alkyl, trifluoromethyl and halogen; R10 is independently selected from hydrogen, C1 -C6 alkyl, benzyl,
2,2,2-trifluoroethyl and aryl; R11 is independently selected from C1 -C6 alkyl and aryl;
R12 is independently selected from hydrogen, C1 -C6 alkyl, C1 -C6
aralkyl, C1 -C6 substituted aralkyl, C1 -C6 heteroaralkyl, C1 -C6 substituted heteroaralkyl, aryl, substituted aryl, heteroaryl, substituted heteraryl, C1 -C6 perfluoroalkyl,
2-aminoethyl and 2,2,2-trifluoroethyl;
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,
imidazolinyl, pyridinyl, thiazolyl, oxazolyl, indolyl, quinolinyl, isoquinolinyl, triazolyl and thienyl, c) aryl,
d) C1 -C20 alkyl wherein from 0 to 4 carbon atoms are
replaced with 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;
provided that when V is heterocycle, attachment of V to R8 and to A1 is through a substitutable ring carbon;
X is a bond, -CH=CH-, -C(O)NR10-, -NR10C(O)-, -NR10-, O or -C(=O)-; m is 0, 1 or 2;
n is independently 0, 1 , 2, 3 or 4;
p is 0, 1 , 2, 3 or 4, provided that p is not 0 if X is a bond or O; 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 3 of the formula D:
Figure imgf000114_0001
wherein: from 1-2 of f(s) are independently N or N->O, and the remaining f's are independently CH;
R1 is selected from: hydrogen, C3-C10 cycloalkyl or C1 -C6 alkyl;
R2 is independently selected from:
a) hydrogen,
b) aryl, heterocycle, C3-C10 cycloalkyl, R10O-, -N(R 10)2, F or C2-C6 alkenyl,
c) C1 -C6 alkyl unsubstituted or substituted by aryl,
heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, R10O-, or -N(R10)2;
R3 is selected from:
a) hydrogen,
b) unsubstituted or substituted aryl, unsubstituted or
substituted heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, halogen, C1 -C6 perfluoroalkyl, R12O-, R11S(O)m-, R10C(O)NR10-, (R10)2NC(O)-, R10 2N-C(NR10)-, CN, NO2, R10C(O)-, N3, -N(R10)2, or R11OC(O)NR10-,
c) unsubstituted C1 -C6 alkyl,
d) substituted C1 -C6 alkyl wherein the substituent on the
substituted C1 -C6 alkyl is selected from unsubstituted or substituted aryl, unsubstituted or substituted heterocyclic, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl,
R12O-, R 1 1 O(O)m-, R10C(O)NR10-, (R10)2NC(O)-, R10 2N-C(NR10)-, CN, R10C(O)-, N3, -N(R10)2, and
R 1 1OC(O)-NR10-;
R4 is selected from H, halogen, C1 -C6 alkyl and CF3; R6a, R6b, R6c, R6d and R6e are independently selected from:
a) hydrogen,
b) unsubstituted or substituted aryl, unsubstituted or
substituted heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, halogen, C1 -C6 perfluoroalkyl, R12O-, R1 1S(O)m-, R10C(O)NR10-, (R10)2NC(O)-, R10 2N-C(NR10)-, CN, NO2, R10C(O)-, N3, -N(R10)2, or R 1 1 OC(O)NR10-,
c) unsubstituted C1 -C6 alkyl,
d) substituted C1 -C6 alkyl wherein the substituent on the
substituted C1 -C6 alkyl is selected from unsubstituted or substituted aryl, unsubstituted or substituted heterocyclic, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, R12O-, R 1 1 S(O)m-, R10C(O)NR10-, (R10)2NC(O)-, R10 2N-C(NR10)-, CN, R10C(O)-, N3, -N(R10)2, and
R1 1OC(O)-NR10-; or any two of R6 a, R6b, R6c, R6d and R6e on adjacent carbon atoms are combined to form a diradical selected from -CH=CH-CH=CH-, -CH=CH-CH2-, -(CH2)4- and -(CH2)3-; provided that when R3, R6a, R6b, R6c, R6d or R6e is
unsubstituted or substituted heterocycle, attachment of R3, R6a, R6b, R6c, R6d or R6e to the 6-membered heteroaryl ring, or phenyl ring respectively, is through a substitutable heterocycle ring carbon;
R8 is independently selected from:
a) hydrogen,
b) aryl, substituted aryl, heterocycle, 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 R11OC(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 R11OC(O)NR10-;
provided that when R8 is heterocycle, attachment of R8 to V is through a substitutable ring carbon;
R9a and R9b are independently hydrogen, halogen, CF3 or methyl; R10 is independently selected from hydrogen, C1 -C6 alkyl, benzyl,
2,2,2-trifιuoroethyl and aryl; R11 is independently selected from C1 -C6 alkyl and aryl;
R12 is independently selected from hydrogen, C1 -C6 alkyl, C1 -C6 aralkyl, C1 -C6 substituted aralkyl, C1 -C6 heteroaralkyl, C1 -C6 substituted heteroaralkyl, aryl, substituted aryl, heteroaryl, substituted heteraryl, C1 -C6 perfluoroalkyl, 2-aminoethyl and 2,2,2-trifiuoroethyl; A 1 is selected from: a bond, -C(O)-, O, -N(R10)-, or S(O)m;
X is a bond, -CH=CH-, -C(O)NR10-, -NR10C(O)-, -NR10-, O or -C(=O)-, n is 0 or 1; provided that n is not 0 if A1 is a bond, O,
-N(R10)- or S(O)m;
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 4 of the formula E:
Figure imgf000118_0001
wherein: from 1 -2 of f(s) are independently N or N->O, and the remaining f's are independently CH;
R1 is selected from: hydrogen, C3-C10 cycloalkyl, R10O-, -N(R10)2, F or C1 -C6 alkyl; R2 is independently selected from:
a) hydrogen,
b) aryl, heterocycle, C3-C10 cycloalkyl, R10O-, -N(R10)2, F or C2-C6 alkenyl,
c) C1 -C6 alkyl unsubstituted or substituted by aryl,
heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, R10O-, or
-N(R 10)2;
R3 is selected from:
a) hydrogen,
b) unsubstituted or substituted aryl, unsubstituted or
substituted heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, halogen, C1 -C6 perfluoroalkyl, R12O-, R11S(O)m-, R10C(O)NR10-, (R 10)2NC(O)-, R10 2N-C(NR10)-, CN, NO2, R10C(O)-, N3, -N(R10)2, or R11OC(O)NR10-,
c) unsubstituted C1 -C6 alkyl, d) substituted C1 -C6 alkyl wherein the substituent on the substituted C1 -C6 alkyl is selected from unsubstituted or substituted aryl, unsubstituted or substituted heterocyclic, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, R12O-, R1 1S(O)m-, R10C(O)NR10-, (R10)2NC(O)-, R10 2N-C(NR10)-, CN, R10C(O)-, N3, -N(R10)2, and R11OC(O)-NR10-;
R4 is selected from H, halogen, C1 -C6 alkyl and CF3;
R6a, R6b, R6c, R6d and R6e are independently selected from:
a) hydrogen,
b) unsubstituted or substituted aryl, unsubstituted or
substituted heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, halogen, C1 -C6 perfluoroalkyl, R12O-, R11S(O)m-, R10C(O)NR10-, (R10)2NC(O)-, R10 2N-C(NR10)-, CN, NO2, R10C(O)-, N3, -N(R10)2, or R11OC(O)NR10-,
c) unsubstituted C1 -C6 alkyl,
d) substituted C1 -C6 alkyl wherein the substituent on the substituted C1 -C6 alkyl is selected from unsubstituted or substituted aryl, unsubstituted or substituted heterocyclic, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, R12O-, R11S(O)m-, R10C(O)NR10-, (R10)2NC(O)-, R10 2N-C(NR10)-, CN, R10C(O)-, N3, -N(R10)2, and R11OC(O)-NR10-; or any two of R6a, R6b, R6C, R6d and R6e on adjacent carbon atoms are combined to form a diradical selected from -CH=CH-CH=CH-, -CH=CH-CH2-, -(CH2)4- and -(CH2)3-; provided that when R3, R6a, R6b, R6c, R6d or R6e is
unsubstituted or substituted heterocycle, attachment of R3, R6a, R6b, R6c, R6d or R6e to the 6-membered heteroaryl ring, or phenyl ring respectively, is through a substitutable heterocycle ring carbon;
R8 is independently selected from:
a) hydrogen,
b) aryl, substituted aryl, heterocycle, 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 R11OC(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 R11OC(O)NR10-;
provided that when R8 is heterocycle, attachment of R8 to V is through a substitutable ring carbon;
R9a and R9b are independently hydrogen, halogen, CF3 or methyl; R10 is independently selected from hydrogen, C1 -C6 alkyl, benzyl,
2,2,2-trifluoroethyl and aryl;
R 1 1 is independently selected from C1 -C6 alkyl and aryl;
R12 is independently selected from hydrogen, C1 -C6 alkyl, C1 -C6
aralkyl, C1 -C6 substituted aralkyl, C1 -C6 heteroaralkyl, C1 -C6 substituted heteroaralkyl, aryl, substituted aryl, heteroaryl, substituted heteraryl, C1 -C6 perfluoroalkyl, 2-aminoethyl and 2,2,2-trifluoroethyl;
X is a bond, -CH=CH-, -C(O)NR10-, -NR10C(O)-, -NR10-, O or -C(=O)-; n is 0 or 1;
m is 0, 1 or 2; and
p is 0, 1 , 2, 3 or 4, provided that p is not 0 if X is a bond or O; or a pharmaceutically acceptable salt thereof.
7. The compound according to Claim 5 of the formula F:
Figure imgf000121_0001
wherein: from 1 -2 of f(s) are independently N or N->O, and the remaining f's are independently CH;
R 1 is selected from: hydrogen, C3-C10 cycloalkyl or C1 -C6 alkyl;
R2 is independently selected from:
a) hydrogen,
b) aryl, heterocycle, C3-C10 cycloalkyl, R10O-, -N(R10)2 or
F,
c) C1 -C6 alkyl unsubstituted or substituted by aryl,
heterocycle, C3-C10 cycloalkyl, R10O-, or -N(R10)2; R3 is selected from:
a) hydrogen,
b) unsubstituted or substituted aryl, unsubstituted or
substituted heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, halogen, C1 -C6 perfluoroalkyl, R12O-, R1 1S(O)m-, R10C(O)NR10-, (R10)2NC(O)-, R10 2N-C(NR10)-, CN, NO2, R10C(O)-, N3, -N(R10)2, or R11OC(O)NR10-, c) unsubstituted C1 -C6 alkyl,
d) substituted C1 -C6 alkyl wherein the substituent on the substituted C1 -C6 alkyl is selected from unsubstituted or substituted aryl, unsubstituted or substituted heterocyclic, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, R12O-, R11S(O)m-, R10C(O)NR10-, (R10)2NC(O)-, R10 2N-C(NR10)-, CN, R10C(O)-, N3, -N(R10)2, and R11OC(O)-NR10-; R4 is selected from H, halogen, CH3 and CF3;
R6a, R6b, R6c, R6d and R6e are independently selected from:
a) hydrogen,
b) unsubstituted or substituted aryl, unsubstituted or
substituted heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, halogen, C1 -C6 perfluoroalkyl, R 12O-, R11S(O)m-, R10C(O)NR10-, (R10)2NC(O)-, R10 2N-C(NR10)-, CN, NO2, R10C(O)-, N3, -N(R10)2, or R11OC(O)NR10-,
c) unsubstituted C1 -C6 alkyl,
d) substituted C1 -C6 alkyl wherein the substituent on the
substituted C1 -C6 alkyl is selected from unsubstituted or substituted aryl, unsubstituted or substituted heterocyclic, C3-C1 0 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, R 1 2O-, R11S(O)m-, R10C(O)NR10-, (R10)2NC(O)-, R10 2N-C(NR10)-, CN, R10C(O)-, N3, -N(R10)2, and R11OC(O)-NR10-; or any two of R6a, R6b, R6c, R6d and R6e on adjacent carbon atoms are combined to form a diradical selected from -CH=CH-CH=CH-,
-CH=CH-CH2-, -(CH2)4- and -(CH2)3-; provided that when R3, R6a, R6b, R6c, R6d or R6e is
unsubstituted or substituted heterocycle, attachment of R3, R6a, R6b, R6c, R6d or R6e to the 6-membered heteroaryl ring, or phenyl ring respectively, is through a substitutable heterocycle ring carbon; R9a and R9b are independently hydrogen, halogen, CF3 or methyl; R10 is independently selected from hydrogen, C1 -C6 alkyl, benzyl,
2,2,2-trifluoroethyl and aryl; R11 is independently selected from C1 -C6 alkyl and aryl;
R12 is independently selected from hydrogen, C1 -C6 alkyl, C1 -C6
aralkyl, C1 -C6 substituted aralkyl, C1 -C6 heteroaralkyl, C1 -C6 substituted heteroaralkyl, aryl, substituted aryl, heteroaryl, substituted heteraryl, C1 -C6 perfluoroalkyl,
2-aminoethyl and 2,2,2-trifluoroethyl;
X is a bond, -CH=CH-, -C(O)NR10-, -NR10C(O)-, -NR10-, O or -C(=O)-; m is 0, 1 or 2; and
p is 0, 1 , 2, 3 or 4; or a pharmaceutically acceptable salt thereof.
8. The compound according to Claim 6 of the formula G:
Figure imgf000123_0001
wherein: from 1-2 of f(s) are independently N or N->0, and the remaining f's are independently CH;
R 1 is selected from: hydrogen, C3-C10 cycloalkyl, R10O-, -N(R10)2, F or C1 -C6 alkyl;
R2 is independently selected from:
a) hydrogen,
b) aryl, heterocycle or C3-C10 cycloalkyl,
c) C1 -C6 alkyl unsubstituted or substituted by aryl,
heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, R10O-, or -N(R10)2;
R3 is selected from:
a) hydrogen,
b) unsubstituted or substituted aryl, unsubstituted or
substituted heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, halogen, C1 -C6 perfluoroalkyl,
R 1 2O-, R 1 1 S(O)m-, R10C(O)NR10-, (R 10)2NC(O)-,
R 10 2N-C(NR10)-, CN, NO2, R10C(O)-, N3, -N(R 10)2, or R11OC(O)NR10-,
c) unsubstituted C1 -C6 alkyl,
d) substituted C1 -C6 alkyl wherein the substituent on the
substituted C1 -C6 alkyl is selected from unsubstituted or substituted aryl, unsubstituted or substituted heterocyclic, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl,
R12O-, R11S(O)m-, R10C(O)NR10-, (R10)2NC(O)-, R10 2N-C(NR10)-, CN, R10C(O)-, N3, -N(R10)2, and
R1 1OC(O)-NR10-;
R4 is selected from H, halogen, CH3 and CF3; R6a, R6b, R6c, R6d and R6e are independently selected from:
a) hydrogen,
b) unsubstituted or substituted aryl, unsubstituted or
substituted heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, halogen, C1 -C6 perfluoroalkyl, R12O-, R11S(O)m-, R10C(O)NR10-, (R10)2NC(O)-, R10 2N-C(NR10)-, CN, NO2, R10C(O)-, N3, -N(R10)2, or R11OC(O)NR10-,
c) unsubstituted C1 -C6 alkyl,
d) substituted C1 -C6 alkyl wherein me substituent on the substituted C1 -C6 alkyl is selected from unsubstituted or substituted aryl, unsubstituted or substituted heterocyclic, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, R12O-, R11S(O)m-, R10C(O)NR10-, (R10)2NC(O)-, R10 2N-C(NR10)-, CN, R10C(O)-, N3, -N(R10)2, and R11OC(O)-NR10-; or any two of R6a, R6b, R6c, R6d and R6e on adjacent carbon atoms are combined to form a diradical selected from -CH=CH-CH=CH-, -CH=CH-CH2-, -(CH2)4- and -(CH2)3-; provided that when R3, R6a, R6b, R6c, R6d or R6e is
unsubstituted or substituted heterocycle, attachment of R3, R6a, R6b, R6c, R6d or R6e to the 6-membered heteroaryl ring, or phenyl ring respectively, is through a substitutable heterocycle ring carbon;
R9a and R9b are independently hydrogen, halogen, CF3 or methyl; R10 is independently selected from hydrogen, C1 -C6 alkyl, benzyl,
2,2,2-trifluoroethyl and aryl;
R1 1 is independently selected from C1 -C6 alkyl and aryl; R12 is independently selected from hydrogen, C1 -C6 alkyl, C1 -C6 aralkyl, C1 -C6 substituted aralkyl, C1 -C6 heteroaralkyl, C1 -C6 substituted heteroaralkyl, aryl, substituted aryl, heteroaryl, substituted heteraryl, C1 -C6 perfluoroalkyl, 2-aminoethyl and 2,2,2-trifluoroethyl;
A1 is selected from: a bond, -C(O)-, O, -N(R10)-, or S(O)m; m is 0, 1 or 2; and
n is 0 or 1 ; or a pharmaceutically acceptable salt thereof.
9. A compound which inhibits farnesyl-protein transferase which is:
1 -(2-Phenylpyrid-5-ylmethyl)-5-(4-cyanobenzyl)imidazole 1 -(2-Phenyl-N-Oxopyrid-5-yImethyl)-5-(4-cyanobenzyl)imidazole 1 -(3-Phenylpyrid-6-ylmethyl)-5-(4-cyanobenzyl)imidazole 1-(3-Phenyl-N-Oxopyrid-6-ylmethyl)-5-(4-cyanobenzyl)imidazole
1 -(2-(3-Trifluoromethoxyphenyl)-pyrid-5-ylmethyl)-5-(4- cyanobenzyl)imidazole
1-(2-(2-Trifluoromethylphepyl)-pyrid-5-ylmethyl)-5-(4- cy anobenzyl)imidazole
1 -(3-Phenyl-2-Chloropyrid-6-ylmethyl)-5-(4-cyanobenzyl)imidazole 1 -(3-Phenyl-4-chloropyrid-6-ylmethyl)-5-(4-cyanobenzyl)imidazoIe or 1-(2-Amino-3-phenylpyrid-6-ylmethyl)-5-(4-cyanobenzyl)imidazole or a pharmaceutically acceptable salt thereof.
10. The compound according to Claim 9 which is:
1 -(2-Phenylpyrid-5-ylmethyl)-5-(4-cyanobenzyl)imidazole
Figure imgf000127_0001
or a pharmaceutically acceptable salt thereof.
11. The compound according to Claim 9 which is:
1 -(2-(2-Trifluoromethylphenyl)-pyrid-5-ylmethyI)-5-(4- cyanobenzyl)imidazole
Figure imgf000128_0001
or a pharmaceutically acceptable salt thereof.
12. A pharmaceutical composition comprising a pharmaceutical carrier, and dispersed therein, a therapeutically effective amount of a compound of Claim 1.
13. A pharmaceutical composition comprising a pharmaceutical carrier, and dispersed therein, a therapeutically effective amount of a compound of Claim 3.
14. A pharmaceutical composition comprising a pharmaceutical carrier, and dispersed therein, a therapeutically effective amount of a compound of Claim 4.
15. A pharmaceutical composition comprising a pharmaceutical carrier, and dispersed therein, a therapeutically effective amount of a compound of Claim 9.
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 inhibiting farnesyl-protein transferase which comprises administering to a mammal in need thereof a
therapeutically effective amount of a composition of Claim 15.
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 cancer which comprises administering to a mammal in need thereof a therapeutically effective amount of a composition of Claim 15.
24. 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 12.
25. 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 12.
26. 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 12.
27. A method for preventing restenosis which comprises administering to a mammal in need thereof a therapeutically effective amount of a composition of Claim 12.
28. 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 12.
29. A pharmaceutical composition made by combining the compound of Claim 1 and a pharmaceutically acceptable carrier.
30. A process for making a pharmaceutical composition comprising combining a compound of Claim 1 and a pharmaceutically acceptable carrier.
PCT/US1997/005304 1996-04-03 1997-04-01 Inhibitors of farnesyl-protein transferase WO1997036901A1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
EP97920000A EP0891361A1 (en) 1996-04-03 1997-04-01 Inhibitors of farnesyl-protein transferase
JP9535534A JP2000507590A (en) 1996-04-03 1997-04-01 Farnesyl-protein transferase inhibitor
AU24301/97A AU706150B2 (en) 1996-04-03 1997-04-01 Inhibitors of farnesyl-protein transferase

Applications Claiming Priority (8)

Application Number Priority Date Filing Date Title
US1459296P 1996-04-03 1996-04-03
US60/014,592 1996-04-03
GB9613462.2 1996-06-27
GBGB9613462.2A GB9613462D0 (en) 1996-06-27 1996-06-27 Inhibitors of farnesyl-protein transferase
US2264796P 1996-07-24 1996-07-24
US60/022,647 1996-07-24
GB9617277.0 1996-08-16
GBGB9617277.0A GB9617277D0 (en) 1996-08-16 1996-08-16 Inhibitors of farnesyl-protein transferase

Publications (1)

Publication Number Publication Date
WO1997036901A1 true WO1997036901A1 (en) 1997-10-09

Family

ID=27451473

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1997/005304 WO1997036901A1 (en) 1996-04-03 1997-04-01 Inhibitors of farnesyl-protein transferase

Country Status (5)

Country Link
EP (1) EP0891361A1 (en)
JP (1) JP2000507590A (en)
AU (1) AU706150B2 (en)
CA (1) CA2249607A1 (en)
WO (1) WO1997036901A1 (en)

Cited By (36)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999028315A1 (en) * 1997-11-28 1999-06-10 Lg Chemical Ltd. Imidazole derivatives having an inhibitory activity for farnesyl transferase and process for preparation thereof
US5939439A (en) * 1996-12-30 1999-08-17 Merck & Co., Inc. Inhibitors of farnesyl-protein transferase
WO1999065898A1 (en) * 1998-06-16 1999-12-23 Societe De Conseils De Recherches Et D'applications Scientifiques Sas Imidazolyl derivatives
US6093737A (en) * 1996-12-30 2000-07-25 Merck & Co., Inc. Inhibitors of farnesyl-protein transferase
EP1035850A1 (en) * 1997-12-04 2000-09-20 Merck & Co., Inc. Inhibitors of farnesyl-protein transferase
US6127390A (en) * 1997-10-02 2000-10-03 Merck & Co., Inc. Inhibitors of prenyl-protein transferase
EP1045843A1 (en) * 1997-12-04 2000-10-25 Merck & Co., Inc. Inhibitors of farnesyl-protein transferase
EP1169320A1 (en) * 1999-04-13 2002-01-09 LG Chem Investment, Ltd. Farnesyl transferase inhibitors having a pyrrole structure and process for preparation thereof
US6420555B1 (en) 1998-06-16 2002-07-16 Societe De Conseils De Recherches Et D'applications Scientifiques, S.A.S. Imidazolyl derivatives
WO2002074747A1 (en) * 2000-11-30 2002-09-26 Abbott Laboratories Farnesyltransferase inhibitors
US6627629B2 (en) 2000-06-30 2003-09-30 Bristol-Myers Squibb Pharma N-ureidoheterocycloalkyl-piperidines as modulators of chemokine receptor activity
EP1420015A1 (en) * 1999-06-11 2004-05-19 Societe De Conseils De Recherches Et D'applications Scientifiques S.A.S. Imidazolyl derivatives
US6984643B2 (en) 2002-07-02 2006-01-10 Roche Palo Alto Llc 2,5-substituted pyrimidine derivatives-CCR-3 receptor antagonists
US7211595B2 (en) 2000-11-30 2007-05-01 Abbott Laboratories Farnesyltransferase inhibitors
US7649004B2 (en) 2004-07-23 2010-01-19 Pfizer, Inc. Pyridine derivatives
US7902373B2 (en) 2006-12-19 2011-03-08 Pfizer Inc Nicotinamide derivatives
US7968536B2 (en) 2007-06-29 2011-06-28 Millennium Pharmaceuticals, Inc. Heterocyclic compounds useful as RAF kinase inhibitors
US8067589B2 (en) 2007-02-26 2011-11-29 Pfizer Inc Heterocyclic compounds useful in treating diseases and conditions
US8293752B2 (en) 2007-06-29 2012-10-23 Millennium Pharmaceuticals, Inc. Compounds useful as Raf kinase inhibitors
CN102844302A (en) * 2010-04-06 2012-12-26 日本曹达株式会社 Nitrogen-containing heterocyclic compound and method for producing same
US8410144B2 (en) 2009-03-31 2013-04-02 Arqule, Inc. Substituted indolo-pyridinone compounds
US8536185B2 (en) 2008-09-22 2013-09-17 Cayman Chemical Company, Incorporated Multiheteroaryl compounds as inhibitors of H-PGDS and their use for treating prostaglandin D2 mediated diseases
US8946278B2 (en) 2007-02-07 2015-02-03 Glaxosmithkline Llc Inhibitors of AkT activity
WO2016018702A1 (en) * 2014-07-28 2016-02-04 Merck Sharp & Dohme Corp. FACTOR XIa INHIBITORS
US9255108B2 (en) 2012-04-10 2016-02-09 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US9359365B2 (en) 2013-10-04 2016-06-07 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US9708348B2 (en) 2014-10-03 2017-07-18 Infinity Pharmaceuticals, Inc. Trisubstituted bicyclic heterocyclic compounds with kinase activities and uses thereof
US9751888B2 (en) 2013-10-04 2017-09-05 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US9775844B2 (en) 2014-03-19 2017-10-03 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US9951069B1 (en) 2017-01-11 2018-04-24 Rodin Therapeutics, Inc. Bicyclic inhibitors of histone deacetylase
US10160761B2 (en) 2015-09-14 2018-12-25 Infinity Pharmaceuticals, Inc. Solid forms of isoquinolinones, and process of making, composition comprising, and methods of using the same
US10421756B2 (en) 2015-07-06 2019-09-24 Rodin Therapeutics, Inc. Heterobicyclic N-aminophenyl-amides as inhibitors of histone deacetylase
US10759806B2 (en) 2016-03-17 2020-09-01 Infinity Pharmaceuticals, Inc. Isotopologues of isoquinolinone and quinazolinone compounds and uses thereof as PI3K kinase inhibitors
US10919914B2 (en) 2016-06-08 2021-02-16 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US10919902B2 (en) 2015-07-06 2021-02-16 Alkermes, Inc. Hetero-halo inhibitors of histone deacetylase
US11225475B2 (en) 2017-08-07 2022-01-18 Alkermes, Inc. Substituted pyridines as inhibitors of histone deacetylase

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2014201038B2 (en) * 2010-04-06 2015-08-06 Nippon Soda Co., Ltd. Nitrogen-containing heterocyclic compound and method for producing same

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4713387A (en) * 1984-06-25 1987-12-15 Toyama Chemical Co., Ltd. Vasodilating and platelet aggregation inhibiting 1,4 dihydropyridines with an imidazolyl or pyridyl containing ester
US5428164A (en) * 1990-12-28 1995-06-27 Neurogen Corporation Certain 4-aryl substituted piperazinyl and piperidinylmethyl phenylimidazole derivatives; a new class of dopamine receptor subtype specific ligands
US5587390A (en) * 1992-08-07 1996-12-24 Istituto Luso Farmaco D'italia S.P.A. Imidazole derivatives having a II antagonist activity
US5633376A (en) * 1990-12-28 1997-05-27 Neurogen Corporation Certain aminomethyl phenylimidazole derivatives; and 4-aryl substituted piperazinyl and piperidinylmethyl phenylimidazole derivatives; a new class of dopamine receptor subtype ligands

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4713387A (en) * 1984-06-25 1987-12-15 Toyama Chemical Co., Ltd. Vasodilating and platelet aggregation inhibiting 1,4 dihydropyridines with an imidazolyl or pyridyl containing ester
US5428164A (en) * 1990-12-28 1995-06-27 Neurogen Corporation Certain 4-aryl substituted piperazinyl and piperidinylmethyl phenylimidazole derivatives; a new class of dopamine receptor subtype specific ligands
US5633376A (en) * 1990-12-28 1997-05-27 Neurogen Corporation Certain aminomethyl phenylimidazole derivatives; and 4-aryl substituted piperazinyl and piperidinylmethyl phenylimidazole derivatives; a new class of dopamine receptor subtype ligands
US5587390A (en) * 1992-08-07 1996-12-24 Istituto Luso Farmaco D'italia S.P.A. Imidazole derivatives having a II antagonist activity

Cited By (69)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5939439A (en) * 1996-12-30 1999-08-17 Merck & Co., Inc. Inhibitors of farnesyl-protein transferase
US6077853A (en) * 1996-12-30 2000-06-20 Merck & Co., Inc. Inhibitors of farnesyl-protein transferase
US6093737A (en) * 1996-12-30 2000-07-25 Merck & Co., Inc. Inhibitors of farnesyl-protein transferase
US6127390A (en) * 1997-10-02 2000-10-03 Merck & Co., Inc. Inhibitors of prenyl-protein transferase
US6268363B1 (en) 1997-11-28 2001-07-31 Lg Chemical Ltd. Imidazole derivatives having an inhibitory activity for farnesyl transferase and process for preparation thereof
US6518429B2 (en) 1997-11-28 2003-02-11 Lg Chemical, Ltd. Imidazole derivatives having an inhibitory activity for farnesyl transferase and process for preparation thereof
US6472526B1 (en) 1997-11-28 2002-10-29 Lg Chemical Ltd. Imidazole derivatives having an inhibitory activity for farnesyl transferase and process for preparation thereof
WO1999028315A1 (en) * 1997-11-28 1999-06-10 Lg Chemical Ltd. Imidazole derivatives having an inhibitory activity for farnesyl transferase and process for preparation thereof
EP1045843A1 (en) * 1997-12-04 2000-10-25 Merck & Co., Inc. Inhibitors of farnesyl-protein transferase
EP1045843A4 (en) * 1997-12-04 2001-10-24 Merck & Co Inc Inhibitors of farnesyl-protein transferase
EP1035850A4 (en) * 1997-12-04 2001-09-12 Merck & Co Inc Inhibitors of farnesyl-protein transferase
EP1035850A1 (en) * 1997-12-04 2000-09-20 Merck & Co., Inc. Inhibitors of farnesyl-protein transferase
US6420555B1 (en) 1998-06-16 2002-07-16 Societe De Conseils De Recherches Et D'applications Scientifiques, S.A.S. Imidazolyl derivatives
US6509336B1 (en) 1998-06-16 2003-01-21 Societe De Conseils De Recherches Et D'applications Scientifiques, S.A.S. Imidazolyl derivatives
WO1999065898A1 (en) * 1998-06-16 1999-12-23 Societe De Conseils De Recherches Et D'applications Scientifiques Sas Imidazolyl derivatives
EP1169320A1 (en) * 1999-04-13 2002-01-09 LG Chem Investment, Ltd. Farnesyl transferase inhibitors having a pyrrole structure and process for preparation thereof
EP1169320A4 (en) * 1999-04-13 2002-10-30 Lg Chem Investment Ltd Farnesyl transferase inhibitors having a pyrrole structure and process for preparation thereof
EP1420015A1 (en) * 1999-06-11 2004-05-19 Societe De Conseils De Recherches Et D'applications Scientifiques S.A.S. Imidazolyl derivatives
US6949546B2 (en) 2000-06-30 2005-09-27 Bristol-Myers Squibb Pharma Company N-ureidoheterocycloalkyl-piperidines as modulators of chemokine receptor activity
US6627629B2 (en) 2000-06-30 2003-09-30 Bristol-Myers Squibb Pharma N-ureidoheterocycloalkyl-piperidines as modulators of chemokine receptor activity
WO2002074747A1 (en) * 2000-11-30 2002-09-26 Abbott Laboratories Farnesyltransferase inhibitors
US7211595B2 (en) 2000-11-30 2007-05-01 Abbott Laboratories Farnesyltransferase inhibitors
US7323570B2 (en) 2000-11-30 2008-01-29 Abbott Laboratories Farnesyltransferase inhibitors
US6984643B2 (en) 2002-07-02 2006-01-10 Roche Palo Alto Llc 2,5-substituted pyrimidine derivatives-CCR-3 receptor antagonists
US7649004B2 (en) 2004-07-23 2010-01-19 Pfizer, Inc. Pyridine derivatives
US7902373B2 (en) 2006-12-19 2011-03-08 Pfizer Inc Nicotinamide derivatives
US8946278B2 (en) 2007-02-07 2015-02-03 Glaxosmithkline Llc Inhibitors of AkT activity
US8067589B2 (en) 2007-02-26 2011-11-29 Pfizer Inc Heterocyclic compounds useful in treating diseases and conditions
US8802657B2 (en) 2007-06-29 2014-08-12 Millennium Pharmaceuticals, Inc. Compounds useful as Raf kinase inhibitors
US9556177B2 (en) 2007-06-29 2017-01-31 Millennium Pharmaceuticals, Inc. Substituted 1,3-thiazoles as synthetic intermediates for preparation of Raf kinase inhibitors
US8293752B2 (en) 2007-06-29 2012-10-23 Millennium Pharmaceuticals, Inc. Compounds useful as Raf kinase inhibitors
US7968536B2 (en) 2007-06-29 2011-06-28 Millennium Pharmaceuticals, Inc. Heterocyclic compounds useful as RAF kinase inhibitors
US9920048B2 (en) 2007-06-29 2018-03-20 Millennium Pharmaceuticals, Inc. Substituted pyrimidines for inhibiting Raf kinase activity
US8536185B2 (en) 2008-09-22 2013-09-17 Cayman Chemical Company, Incorporated Multiheteroaryl compounds as inhibitors of H-PGDS and their use for treating prostaglandin D2 mediated diseases
US9126973B2 (en) 2008-09-22 2015-09-08 Cayman Chemical Company, Incorporated Multiheteroaryl compounds as inhibitors of H-PGDS and their use for treating prostaglandin D2 mediated diseases
US8410144B2 (en) 2009-03-31 2013-04-02 Arqule, Inc. Substituted indolo-pyridinone compounds
US20140163235A1 (en) * 2010-04-06 2014-06-12 Nippon Soda Co., Ltd. Nitrogen-containing heterocyclic compound and method for producing same
US8981107B2 (en) * 2010-04-06 2015-03-17 Nippon Soda Co., Ltd. Nitrogen-containing heterocyclic compound and method for producing same
CN102844302A (en) * 2010-04-06 2012-12-26 日本曹达株式会社 Nitrogen-containing heterocyclic compound and method for producing same
US9255108B2 (en) 2012-04-10 2016-02-09 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US9359365B2 (en) 2013-10-04 2016-06-07 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US9751888B2 (en) 2013-10-04 2017-09-05 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US9828377B2 (en) 2013-10-04 2017-11-28 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US10329299B2 (en) 2013-10-04 2019-06-25 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US11541059B2 (en) 2014-03-19 2023-01-03 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US9775844B2 (en) 2014-03-19 2017-10-03 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US10675286B2 (en) 2014-03-19 2020-06-09 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
WO2016018702A1 (en) * 2014-07-28 2016-02-04 Merck Sharp & Dohme Corp. FACTOR XIa INHIBITORS
US9975874B2 (en) 2014-07-28 2018-05-22 Merck Sharp & Dohme Corp. Factor XIa inhibitors
US10253047B2 (en) 2014-10-03 2019-04-09 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US9708348B2 (en) 2014-10-03 2017-07-18 Infinity Pharmaceuticals, Inc. Trisubstituted bicyclic heterocyclic compounds with kinase activities and uses thereof
US10941162B2 (en) 2014-10-03 2021-03-09 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US10919902B2 (en) 2015-07-06 2021-02-16 Alkermes, Inc. Hetero-halo inhibitors of histone deacetylase
US10421756B2 (en) 2015-07-06 2019-09-24 Rodin Therapeutics, Inc. Heterobicyclic N-aminophenyl-amides as inhibitors of histone deacetylase
US11858939B2 (en) 2015-07-06 2024-01-02 Alkermes, Inc. Hetero-halo inhibitors of histone deacetylase
US11247995B2 (en) 2015-09-14 2022-02-15 Infinity Pharmaceuticals, Inc. Solid forms of isoquinolinones, and process of making, composition comprising, and methods of using the same
US10160761B2 (en) 2015-09-14 2018-12-25 Infinity Pharmaceuticals, Inc. Solid forms of isoquinolinones, and process of making, composition comprising, and methods of using the same
US11939333B2 (en) 2015-09-14 2024-03-26 Infinity Pharmaceuticals, Inc. Solid forms of isoquinolinones, and process of making, composition comprising, and methods of using the same
US10759806B2 (en) 2016-03-17 2020-09-01 Infinity Pharmaceuticals, Inc. Isotopologues of isoquinolinone and quinazolinone compounds and uses thereof as PI3K kinase inhibitors
US10919914B2 (en) 2016-06-08 2021-02-16 Infinity Pharmaceuticals, Inc. Heterocyclic compounds and uses thereof
US10696673B2 (en) 2017-01-11 2020-06-30 Rodin Therapeutics, Inc. Bicyclic inhibitors of histone deacetylase
US11225479B2 (en) 2017-01-11 2022-01-18 Alkermes, Inc. Bicyclic inhibitors of histone deacetylase
US11286256B2 (en) 2017-01-11 2022-03-29 Alkermes, Inc. Bicyclic inhibitors of histone deacetylase
US9951069B1 (en) 2017-01-11 2018-04-24 Rodin Therapeutics, Inc. Bicyclic inhibitors of histone deacetylase
US10519149B2 (en) 2017-01-11 2019-12-31 Rodin Therapeutics, Inc. Bicyclic inhibitors of histone deacetylase
US10793567B2 (en) 2017-01-11 2020-10-06 Rodin Therapeutics, Inc. Bicyclic inhibitors of histone deacetylase
US11987580B2 (en) 2017-01-11 2024-05-21 Alkermes, Inc. Bicyclic inhibitors of histone deacetylase
US11225475B2 (en) 2017-08-07 2022-01-18 Alkermes, Inc. Substituted pyridines as inhibitors of histone deacetylase
US11912702B2 (en) 2017-08-07 2024-02-27 Alkermes, Inc. Substituted pyridines as inhibitors of histone deacetylase

Also Published As

Publication number Publication date
AU2430197A (en) 1997-10-22
CA2249607A1 (en) 1997-10-09
JP2000507590A (en) 2000-06-20
EP0891361A1 (en) 1999-01-20
AU706150B2 (en) 1999-06-10

Similar Documents

Publication Publication Date Title
US5872136A (en) Arylheteroaryl inhibitors of farnesyl-protein transferase
US5854264A (en) Inhibitors of farnesyl-protein transferase
US5883105A (en) Inhibitors of farnesyl-protein transferase
US5854265A (en) Biheteroaryl inhibitors of farnesyl-protein transferase
WO1997036901A1 (en) Inhibitors of farnesyl-protein transferase
US6080870A (en) Biaryl substituted imidazole compounds useful as farnesyl-protein transferase inhibitors
AU714851B2 (en) Inhibitors of farnesyl-protein transferase
US5919785A (en) Inhibitors of farnesyl-protein transferase
US5939557A (en) Inhibitors of farnesyl-protein transferase
US5859035A (en) Arylheteroaryl inhibitors of farnesyl-protein transferase
US5874452A (en) Biheteroaryl inhibitors of farnesyl-protein transferase
US6093737A (en) Inhibitors of farnesyl-protein transferase
US5880140A (en) Biheteroaryl inhibitors of farnesyl-protein transferase
US5925651A (en) Inhibitors of farnesyl-protein transferase
WO1997036881A1 (en) Inhibitors of farnesyl-protein transferase
AU715606B2 (en) Inhibitors of farnesyl-protein transferase
AU2542597A (en) Inhibitors of farnesyl-protein transferase
WO1997036900A1 (en) Inhibitors of farnesyl-protein transferase
WO1997036585A1 (en) Inhibitors of farnesyl-protein transferase
WO1997036888A1 (en) Inhibitors of farnesyl-protein transferase
AU721952B2 (en) Inhibitors of farnesyl-protein transferase
AU704792B2 (en) Inhibitors of farnesyl-protein transferase
AU706314B2 (en) Inhibitors of farnesyl-protein transferase
US6001835A (en) Inhibitors of farnesyl-protein transferase
WO1997036591A1 (en) Inhibitors of farnesyl-protein transferase

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

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

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): GH KE LS MW SD SZ UG AT BE CH DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN ML MR NE SN TD TG

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

Ref document number: 2249607

Country of ref document: CA

Ref country code: CA

Ref document number: 2249607

Kind code of ref document: A

Format of ref document f/p: F

WWE Wipo information: entry into national phase

Ref document number: 1997920000

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 1997920000

Country of ref document: EP

WWW Wipo information: withdrawn in national office

Ref document number: 1997920000

Country of ref document: EP