WO1997018813A1 - Inhibitors of farnesyl-protein transferase - Google Patents

Inhibitors of farnesyl-protein transferase Download PDF

Info

Publication number
WO1997018813A1
WO1997018813A1 PCT/US1996/018811 US9618811W WO9718813A1 WO 1997018813 A1 WO1997018813 A1 WO 1997018813A1 US 9618811 W US9618811 W US 9618811W WO 9718813 A1 WO9718813 A1 WO 9718813A1
Authority
WO
WIPO (PCT)
Prior art keywords
substituted
piperidine
cyanobenzyl
imidazol
ylethyl
Prior art date
Application number
PCT/US1996/018811
Other languages
French (fr)
Inventor
Byeong M. Kim
Anthony W. Shaw
Samuel L. Graham
S. Jane Desolms
Terrence M. Ciccarone
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 GBGB9604311.2A external-priority patent/GB9604311D0/en
Application filed by Merck & Co., Inc. filed Critical Merck & Co., Inc.
Priority to JP9519941A priority Critical patent/JP2000500502A/en
Priority to EP96942798A priority patent/EP0862435A4/en
Priority to AU11626/97A priority patent/AU704139B2/en
Publication of WO1997018813A1 publication Critical patent/WO1997018813A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D233/00Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings
    • C07D233/04Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings having one double bond between ring members or between a ring member and a non-ring member
    • C07D233/20Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings having one double bond between ring members or between a ring member and a non-ring member with substituted hydrocarbon radicals, directly attached to ring carbon atoms
    • C07D233/24Radicals substituted by nitrogen atoms not forming part of a nitro radical

Definitions

  • Ras protein is part of a signalling pathway that links cell surface growth factor receptors to nuclear signals initiating cellular proliferation. Biological and biochemical studies of Ras action indicate that Ras functions like a G-regulatory protein. 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
  • ras genes are found in many human cancers, including colorectal carcinoma, exocrine pancreatic carcinoma, and myeloid leukemias. The protein products of these genes are defective in their GTPase activity and constitutively transmit a growth stimulatory signal.
  • Ras 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 al, Nature 570:583-586 (1984)).
  • this motif serves as a signal sequence for the enzymes farnesyl-protein transferase or
  • the Ras protein is one of several proteins that are known to undergo post-translational farnesylation. 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.
  • HMG-CoA reductase the rate limiting enzyme for the production of polyisoprenoids
  • farnesyl pyrophosphate the rate limiting enzyme for the production of polyisoprenoids
  • Farnesyl-protein transferase utilizes farnesyl pyrophosphate to covalently modify the Cys thiol group of the Ras CAAX box with a farnesyl group (Reiss et al., Cell, 62:81-88 (1990); Schaber et al, J. Biol. Chem., 265: 14701-14704 (1990); Schafer et al., Science, 249: 1133-1139 (1990); Manne et al., Proc. Natl Acad.
  • FPTase farnesyl-protein transferase
  • the peptide derived inhibitors that have been described are generally cysteine containing molecules that are related to the CAAX motif that is the signal for protein prenylation. (Schaber et al., ibid; Reiss et. al., ibid; Reiss et al., PNAS, 88:732-736 (1991)). Such inhibitors may inhibit protein prenylation while serving as alternate substrates for the farnesyl-protein transferase enzyme, or may be purely competitive inhibitors (U.S.
  • farnesyl-protein transferase inhibitors are inhibitors of proliferation of vascular smooth muscle cells and are therefore useful in the prevention and therapy of
  • the present invention comprises peptidomimetic piperidine, 1,4-dihydropyridine and 1,2,3,4-tetrahydropyridine compounds which inhibit the farnesyl-protein transferase.
  • 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: wherein:
  • R 1 a , R 1 b and R 1 c are independently selected from:
  • R 2 is selected from: H; unsubstituted or substituted C 1 -8 alkyl,
  • substituted group is substituted with one or more of:
  • R 3 is selected from: H;
  • R 4 is independently selected from:
  • R 5 is independently selected from:
  • perfluoroalkyl F, Cl, Br, R 8 O-, R 9 S(O) m -, R 8 C(O)NR 8 -, CN, NO 2 , (R 8 ) 2 N-C-(NR 8 )-, R 8 C(O)-, R 8 OC(O)-, N 3 , -N(R 8 ) 2 , or R 9 OC(O)NR 8 -, and
  • R 6 , R 7 and R 7a are independently selected from: H; C 1-4 alkyl, C 3 -6 cycloalkyl, heterocycle, aryl, C 1 -4 perfluoroalkyl, unsubstituted or substituted with one or two substituents selected from:
  • R 6 and R 7 may be joined in a ring
  • R 7 and R 7a may be joined in a ring;
  • R 8 is independently selected from hydrogen, C 1 -C 6 alkyl, benzyl,
  • R 9 is independently selected from C 1 -C 6 alkyl and aryl;
  • R 10 is selected from: H; R 8 C(O)-; R 9 S(O) m -; unsubstituted or substituted C 1 -4 alkyl, unsubstituted or substituted C 3 -6 cycloalkyl, unsubstituted or substituted heterocycle, unsubstituted or substituted aryl, substituted aroyl, unsubstituted or substituted heteroaroyl, substituted arylsulfonyl, unsubstituted or substituted heteroarylsulfonyl, wherein the substituted group is substituted with one or two substituents selected from:
  • 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 ;
  • W is a heterocycle
  • n 0, 1 , 2, 3 or 4;
  • r is 0 to 5, provided that r is 0 when V is hydrogen;
  • s 1 or 2;
  • t is 0 or 1 ; and the dashed lines represent optional double bonds; or an optical isomer or a pharmaceutically acceptable salt thereof.
  • a preferred embodiment of the compounds of this invention is illustrated by the following formula:
  • R 1 a and R 1 c are independently selected from: hydrogen, C 3 -C 10 cycloalkyl, R 8 O-, -N(R 8 ) 2 , F or C 1 -C 6 alkyl;
  • R 1 b is independently selected from:
  • R 2 is selected from:
  • perfluoroalkyl F, Cl, R 8 O-, R 8 C(O)NR 8 -, CN, NO 2 , (R 8 ) 2 N-C(NR 8 )-, R 8 C(O)-, R 8 OC(O)-, -N(R 8 ) 2 , or R 9 OC(O)NR 8 -, and
  • R 5 is selected from:
  • R 6 , R 7 and R 7a are independently selected from:
  • R 8 is independently selected from hydrogen, C 1 -C 6 alkyl, benzyl,
  • R 9 is independently selected from C 1 -C 6 alkyl and aryl
  • R 10 is selected from: H; R 8 C(O)-; R 9 S(O) m -; unsubstituted or substituted C 1 -4 alkyl, unsubstituted or substituted C 3-6 cycloalkyl, unsubstituted or substituted heterocycle, unsubstituted or substituted aryl, substituted aroyl, unsubstituted or substituted heteroaroyl, substituted arylsulfonyl, unsubstituted or substituted heteroarylsulfonyl, wherein the substituted group is substituted with one or two substituents selected from:
  • V is selected from:
  • heterocycle selected from pyrrolidinyl, imidazolyl,
  • W is a heterocycle selected from pyrrolidinyl, imidazolyl, pyridinyl, thiazolyl, pyridonyl, 2-oxopiperidinyl, indolyl, quinolinyl, or isoquinolinyl;
  • n 0, 1, 2, 3 or 4;
  • P is 1, 2 or 3;
  • q is 0 or 1 ;
  • r is 0 to 5, provided that r is 0 when V is hydrogen;
  • s is 1 or 2;
  • t is 1 ; or an optical isomer or a pharmaceutically acceptable salt thereof.
  • R 1 a and R 1 c are independently selected from: hydrogen, C 3 -C 10 cycloalkyl, R 8 O-, -N(R 8 ) 2 , F or C 1 -C 6 alkyl;
  • R 1b 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 8 O- and -N(R 8 ) 2 ;
  • R 2 is selected from: H; unsubstituted or substituted C 1 -8 alkyl,
  • substituted group is substituted with one or more of:
  • R 3 is selected from: H; ,
  • R 4 is independently selected from:
  • R 8 is independently selected from hydrogen, C 1 -C 6 alkyl, 2,2,2- trifluoroethyl, benzyl and aryl;
  • R 9 is independently selected from C 1 -C 6 alkyl and aryl;
  • R 10 is selected from: H; R 8 C(O)-; R 9 S(O) m -; unsubstituted or substituted C 1 -4 alkyl, unsubstituted or substituted C 3-6 cycloalkyl, unsubstituted or substituted heterocycle, unsubstituted or substituted aryl, substituted aroyl, unsubstituted or substituted heteroaroyl, substituted arylsulfonyl, unsubstituted or substituted heteroarylsulfonyl, wherein the substituted group is substituted with one or two substituents selected from:
  • 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 ;
  • n 0, 1, 2, 3 or 4;
  • p 0, 1, 2, 3 or 4;
  • q is 0 or 1 ; and r is 0 to 5, provided that r is 0 when V is hydrogen; or an optical isomer or pharmaceutically acceptable salt thereof.
  • Another preferred embodiment of the compounds of this invention are illustrated by the formula C:
  • R 1 a and R 1c are independently selected from: hydrogen, C 3 -C 10 cycloalkyl, R 8 O-, -N(R 8 ) 2 , F or C 1 -C 6 alkyl;
  • R 1 b 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 8 O- and -N(R 8 ) 2 ;
  • R 2 is selected from: H; unsubstituted or substituted C 1 -8 alkyl, unsubstituted or substituted C 2-8 alkenyl, unsubstituted or substituted aryl, unsubstituted or substituted heterocycle, ,
  • substituted group is substituted with one or more of: or ,
  • R 3 is selected from: H;
  • R 4 is independently selected from:
  • aryl substituted aryl, heterocycle, substituted heterocycle, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 1 -C 6 perfluoroalkyl, F, Cl, R 8 O-, R 8 C(O)NR 8 -, CN, NO 2 , (R 8 ) 2 N-C(NR 8 )-, R 8 C(O)-, -N(R 8 ) 2 , or R 9 OC(O)NR 8 -, and
  • R 5a and R 5b are independently hydrogen, C 1 -C 6 alkyl, cyclopropyl, trifluoromethyl and halogen;
  • R 6 , R 7 and R 7a are independently selected from:
  • R 8 is independently selected from hydrogen, C 1 -C 6 alkyl, 2,2,2- trifluoroethyl, benzyl and aryl;
  • R 9 is independently selected from C 1 -C 6 alkyl and aryl;
  • R 10 is selected from: H; R 8 C(O)-; R 9 S(O) m -; unsubstituted or substituted C 1 -4 alkyl, unsubstituted or substituted C 3-6 cycloalkyl, unsubstituted or substituted heterocycle, unsubstituted or substituted aryl, substituted aroyl, unsubstituted or substituted heteroaroyl, substituted arylsulfonyl, unsubstituted or substituted heteroarylsulfonyl, wherein the substituted group is substituted with one or two substituents selected from:
  • V is selected from:
  • a) hyddogen b) heterocycle selected from pyrrolidinyl, imidazolyl, imidazolinyl, pyridinyl, thiazolyl, pyridonyl, 2- oxopiperidinyl, oxazolyl, indolyl, quinolinyl, isoquinolinyl, triazolyl and thienyl,
  • 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 ;
  • n 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
  • q is 0 or 1 ;
  • r is 0 to 5, provided that r is 0 when V is hydrogen; or an optical isomer or pharmaceutically acceptable salt thereof.
  • R 1 a and R 1 c are independently selected from: hydrogen, C 3 -C 10
  • R 1 b is independently selected from:
  • R 2 is selected from: H; unsubstituted or substituted C 1 -8 alkyl, unsubstituted or substituted aryl, , and -S(O) 2 R 6 , wherein the substituted group is substituted with one or more of:
  • R 3 is selected from: H;
  • R 4 is independently selected from:
  • aryl substituted aryl, heterocycle, substituted heterocycle, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 1 -C 6 perfluoroalkyl, F, Cl, R 8 O-, R 8 C(O)NR 8 -, CN, NO 2 , (R 8 ) 2 N-C(NR 8 )-, R 8 C(O)-, -N(R 8 ) 2 , or R 9 OC(O)NR 8 -, and
  • R 5a and R 5b are independently hydrogen, ethyl, cyclopropyl or methyl;
  • R 6 , R 7 and R 7a are independently selected from:
  • R 8 is independently selected from hydrogen, C 1 -C 6 alkyl, 2,2,2- trifluoroethyl, benzyl and aryl;
  • R 9 is independently selected from C 1 -C 6 alkyl and aryl;
  • R 10 is selected from: H; R 8 C(O)-; R 9 S(O) m -; unsubstituted or substituted C 1 -4 alkyl, wherein the substituted alkyl group is substituted with one or two substituents selected from:
  • a 1 is selected from: a bond, -C(O)-, O, -N(R 8 )-, or S(O) m ;
  • n is 0 or 1 ; provided that n is not 0 if A 1 is a bond, O, -N(R 8 )-, or S(O) m ;
  • n 0, 1 or 2;
  • p 0, 1 , 2, 3 or 4;
  • q is 0 or 1 ; or an optical isomer or pharmaceutically acceptable salt thereof.
  • the inhibitors of farnesyl-protein transferase are illustrated by the formula E:
  • R 1 a and R 1 c are independently selected from: hydrogen, R 8 O-,
  • R 1b is independently selected from:
  • R 2 is selected from: H; unsubstituted or substituted C 1 -8 alkyl, unsubstituted or substituted aryl, , and -S(O) 2 R 6 , wherein the substituted group is substituted with one or more of:
  • R 3 is selected from: H;
  • R 4 is independently selected from:
  • aryl substituted aryl, heterocycle, substituted heterocycle, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 1 -C 6 perfluoroalkyl, F, Cl, R 8 O-, R 8 C(O)NR 8 -, CN, NO 2 , (R 8 ) 2 N-C(NR 8 )-, R 8 C(O)-, -N(R 8 ) 2 , or R 9 OC(O)NR 8 -, and
  • R 5a and R 5b are independently hydrogen, ethyl, cyclopropyl or methyl;
  • R 6 , R 7 and R 7a are independently selected from:
  • R 8 is independently selected from hydrogen, C 1 -C 6 alkyl, 2,2,2- trifluoroethyl, benzyl and aryl;
  • R 9 is independently selected from C 1 -C 6 alkyl and aryl;
  • R 10 is selected from: H; R 8 C(O)-; R 9 S(O) m -; unsubstituted or substituted C 1 -4 alkyl, wherein the substituted alkyl group is substituted with one or two substituents selected from:
  • n 0, 1 or 2;
  • p is 0, 1, 2, 3 or 4, provided that p is not 0 if X is a bond
  • q is 0 or 1 ; or an optical isomer or pharmaceutically acceptable salt thereof.
  • R 1 a and R 1 c are independently selected from: hydrogen, C 3 -C 10
  • R 1b is independently selected from:
  • R 2 is selected from: H; unsubstituted or substituted C 1 -8 alkyl, unsubstituted or substituted aryl, , and -S(O) 2 R 6 , wherein the substituted rou is substituted with one or more of:
  • R 3 is selected from: H;
  • R 5a and R 5b are independently hydrogen, ethyl, cyclopropyl or methyl;
  • R 6 , R 7 and R 7a are independently selected from:
  • R 8 is independently selected from hydrogen, C 1 -C 6 alkyl, 2,2,2- trifluoroethyl, benzyl and aryl;
  • R 9 is independently selected from C 1 -C 6 alkyl and aryl;
  • R 10 is selected from: H; R 8 C(O)-; R 9 S(O) m -; unsubstituted or substituted C 1 -4 alkyl, wherein the substituted alkyl group is substituted with one or two substituents selected from:
  • p 0, 1 , 2, 3 or 4;
  • q is 0 or 1 ; or an optical isomer or pharmaceutically acceptable salt thereof.
  • R 1 a and R 1 c are independently selected from: hydrogen, R 8 O-,
  • R 1 b is independently selected from:
  • R 2 is selected from: H; unsubstituted or substituted C 1 -8 alkyl, unsubstituted or substituted aryl, , and -S(O) 2 R 6 , wherein the substituted group is substituted with one or more of:
  • R 3 is selected from: H;
  • R 5a and R5b are independently hydrogen, ethyl, cyclopropyl or methyl;
  • R 6 , R 7 and R 7a are independently selected from:
  • R 8 is independently selected from hydrogen, C 1 -C 6 alkyl, 2,2,2- trifluoroethyl, benzyl and aryl;
  • R 9 is independently selected from C 1 -C 6 alkyl and aryl;
  • R 10 is selected from: H; R 8 C(O)-; R 9 S(O) m -; unsubstituted or substituted C 1 -4 alkyl, wherein the substituted alkyl group is substituted with one or two substituents selected from:
  • a 1 is selected from: a bond, -C(O)-, O, -N(R 8 )-, or S(O) m ;
  • n 0 or 1 ;
  • the compounds of the present invention may have asymmetric centers and occur as racemates, racemic mixtures, and as individual diastereomers, with all possible isomers, including optical isomers, being included in the present invention.
  • any variable e.g. aryl, heterocycle, R 1 a , R 4 etc.
  • its definition on each occurence is independent at every other occurence.
  • combinations of substituents/or variables are permissible only if such combinations result in stable compounds.
  • alkyl is intended to include both branched and straight-chain saturated aliphatic hydrocarbon groups having the specified number of carbon atoms; “alkoxy” represents an alkyl group of indicated number of carbon atoms attached through an oxygen bridge.
  • Halogen or “halo” as used herein means fluoro, chloro, bromo and iodo.
  • aryl is intended to mean any stable monocyclic, bicyclic or tricyclic carbon ring of up to 7 members in each ring, wherein at least one ring is aromatic.
  • monocyclic and bicyclic aryl elements include phenyl, naphthyl, tetrahydronaphthyl, indanyl, biphenyl, phenanthryl, anthryl or
  • tricyclic aryl elements include 10,11-dihydro-5H-dibenzo[a,d]cyclohepten-5-yl (which is also known as dibenzylsuberyl), 9-fluorenyl and 9,10-dihydroanthracen-9-yl.
  • aryl is a monocyclic or bicyclic carbon ring.
  • heterocycle or heterocyclic represents a stable 5- to 7-membered monocyclic or stable 8- to 11-membered bicyclic heterocyclic ring or stable 13- to 15-membered tricyclic heterocyclic ring, which is either saturated or unsaturated, and which consists of carbon atoms and from one to four heteroatoms selected from the group consisting of N, O, and S, and including any bicyclic group in which any of the above-defined heterocyclic rings is fused to a benzene ring.
  • the heterocyclic ring may be attached at any heteroatom or carbon atom which results in the creation of a stable structure.
  • monocyclic and bicyclic heterocyclic elements include, but are not limited to, azepinyl, benzimidazolyl, benzisoxazolyl, benzofurazanyl, benzopyranyl, benzothiopyranyl, benzofuryl,
  • heterocyclic elements include, but are not limited to, 6,11-dihydro-5H-benzo[5,6]cyclohepta[1,2-b]pyridine, 9,10-dihydro-4H-3-thia-benzo[f]azulen-4-yl and 9-xanthenyl.
  • the 6,11-dihydro-5H-benzo[5,6]cyclohepta[1,2-b]pyridine moiety has the following structure:
  • heterocyclic is a monocyclic or bicyclic moiety.
  • heteroaryl is intended to mean any stable monocyclic, bicyclic or tricyclic carbon ring of up to 7 members in each ring, wherein at least one ring is aromatic and wherein from one to four carbon atoms are replaced by heteroatoms selected from the group consisting of N, O, and S.
  • monocyclic and bicyclic heteroaryl elements include, but are not limited to, benzimidazolyl, benzisoxazolyl, benzofurazanyl, benzopyranyl, benzothiopyranyl, benzofuryl, benzothiazolyl, benzothienyl, benzoxazolyl, chromanyl, cinnolinyl, dihydrobenzofuryl, dihydrobenzothienyl,
  • tricyclic heteroaryl elements include, but are not limited to, 6,1 1-dihydro-5H-benzo[5,6]cyclohepta[1,2-b]pyridine.
  • heteroaryl is a monocyclic or bicyclic moiety.
  • substituted aryl As used herein, the terms “substituted aryl”, “substituted heterocycle” and “substituted cycloalkyl” are intended to include the cyclic group containing from 1 to 3 substitutents in addition to the point of attachment to the rest of the compound.
  • Such substitutents are preferably selected from the group which includes but is not limited to F, 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 alkyl)C(O)-, (C 1 -C 6 alkyl)OC(O)-, N 3 ,(C 1 -C 6 alkyl)OC(O)NH-and C 1 -C 20 alkyl.
  • cyclic amine moieties are formed.
  • examples of such cyclic moieties include, but are not limited to:
  • Such cyclic moieties may optionally include another heteroatom(s).
  • heteroatom-containing cyclic amine moieties include, but are not limited to:
  • Lines drawn into the ring systems from substituents indicate that the indicated bond may be attached to any of the substitutable ring carbon atoms.
  • R 1 a and R 1 b are independently selected from: hydrogen, -N(R 8 ) 2 , R 8 C(O)NR 8 - or C 1 -C 6 alkyl which is
  • R 2 is selected from:
  • R 2 comprises at least one unsubstituted or substituted phenyl.
  • R 4 is selected from: hydrogen, perfluoroalkyl, F, Cl, Br, R 8 O-, R 9 S(O) m -, CN, NO 2 , R 8 2 N-C(NR 8 )-, R 8 C(O)-, N 3 , -N(R 8 ) 2 , R 9 OC(O)NR 8 - and C 1 -C 6 alkyl.
  • R 5 is hydrogen
  • R 7b is C 1 -C 6 alkyl substituted with hydrogen or an unsubstituted or substituted aryl group.
  • R 8 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 8 -, -NR 8 C(O)-, O, -N(R 8 )-, -S(O) 2 N(R 8 )- and- N(R 8 )S(O) 2 -.
  • V is selected from hydrogen, heterocycle and aryl.
  • W is imidazolyl.
  • n, p and r are independently 0, 1 , or 2.
  • t is 1.
  • the pharmaceutically acceptable salts of the compounds of this invention include the conventional non-toxic salts of the compounds of this invention as formed, e.g., from non-toxic inorganic or organic acids.
  • such conventional non-toxic salts include those derived from inorganic acids such as hydrochloric, hydrobromic, sulfuric, sulfamic, phosphoric, nitric and the like: and the salts prepared from organic acids such as acetic, propionic, succinic, glycolic, stearic, lactic, malic, tartaric, citric, ascorbic, pamoic, maleic, hydroxymaleic, phenylacetic, glutamic, benzoic, salicylic, sulfanilic, 2-acetoxy-benzoic, fumaric, toluenesulfonic, methanesulfonic, ethane disulfonic, oxalic, isethionic, trifluoroacetic and the like.
  • -N(R 8 ) 2 represents -NH 2 , -NHCH 3 , -NHC 2 H 5 , etc. It is understood that substituents and substitution patterns on the compounds of the instant invention can be selected by one of ordinary skill in the art to provide compounds that are chemically stable and that can be readily synthesized by techniques known in the art, as well as those methods set forth below, from readily available starting materials.
  • the pharmaceutically acceptable salts of the compounds of this invention 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
  • the compounds of this invention are prepared by:
  • the monocarboxylic acid 2 can be treated with an appropriately substituted amine in the presence of a suitable coupling reagent, such as EDC/HOBT, and the like, to provide the 5-carboxynipecotamide 3.
  • a suitable coupling reagent such as EDC/HOBT, and the like.
  • the suitably substituted 5-carboxynipecotamide is then deprotected and the piperidine nitrogen can then be reductively alkylated to provide intermediate 4.
  • the remaining ester moiety is saponified and then similarly functionalized with another suitably substituted amine to provide the bisamidopiperidine 5.
  • the monocarboxylic acid 2 can undergo a Curtius rearrangment to provide the piperidine 6 after catalytic reduction. Subsequent amide formation provides intermediate 7, which is then subjected to the reactions illustrated in Scheme 1 to provide compound 8 of the instant invention.
  • the instant invention also includes 1,4-dihydropyridine and 1,2,3,4-tetrahydropyridine analogs of the piperidine compounds whose syntheses are described above.
  • Scheme 4 illustrates the synthetic route to the intermediates 11 and 13 which correspond to the saturated ring intermedate 2 illustrated in Scheme 1.
  • the appropriately substituted pyridine may be N-alkylated to provide the quaternary intermediate 9.
  • Subsequent reduction of this intermediate provides the 1 ,4-dihydropyridine 10, which can be selectively hydrolized to the key intermediate 11.
  • the 1 ,4-dihydropyridine 10 can be further reduced to provide the enantiomeric mixture of
  • Schemes 5-7 illustrate the syntheses of 1,3-disubstituted piperidines of the instant invention wherein the "X" moiety is other than an amido moiety.
  • the reactions illustrated therein may be modified by using appropriate protecting groups and reagents well known to one skilled in the art to provide 1,3,5-trisubstituted piperidines of the instant invention.
  • Scheme 5 illustrates the syntheses of compounds of the instant invention wherein "X" is -S- or -SO 2 -.
  • a racemic nipecotate 14 can be resolved by the selective crystallization of chiral tartrate salts and is then reductively alkylated to provide the ester 15.
  • Intermediate 15 is reduced to the alcohol 16, activated and treated with a suitable solvent
  • the thioacetate to provide the thioester 17.
  • the thiol is then generated and may be alkylated and optionally oxidized to provide compounds 18 and 19 of the instant invention.
  • the intermediate 16 may be selectively oxidized back to an aldehyde, which can then be utilized to reductively alkylate a suitably substituted amine to provide the instant compound 20.
  • the secondary amine of 20 can be further functionalized as illustrated.
  • the activated alcohol can also be reacted with a suitably substituted imidazolyl to provide compounds of the instant invention wherein "X" is a bond, as shown in Scheme 7.
  • Scheme 8 illustrates the syntheses of compounds of the instant invention wherein R 2 is an aryl moiety.
  • R Sc CH 2 - is R 2 or a protected precursor thereof; and R Sb - is R 6 or a protected precusor thereof; and
  • R- is a "substituent" or a protected precusor thereof.
  • the selectively protected intermediate 20 utilized in the synthesis illustrated in Scheme 9 can be reductively alkylated with a variety of aldehydes, such as 21.
  • the aldehydes can be prepared by standard procedures, such as that described by O. P. Goel, U. Krolls, M. Stier and S. Kesten in Organic Syntheses, 1988, 67, 69-75.
  • the reductive alkylation can be accomplished at pH 5-7 with a variety of reducing agents, such as sodium triacetoxyborohydride or sodium cyanoborohydride in a solvent such as dichloroethane, methanol or dimethylformamide.
  • the ester product 22 can be deprotected with trifluoroacetic acid in methylene chloride to give the substituted diamine 23.
  • That diamine may be isolated in the salt form, for example, as a trifluoroacetate, hydrochloride or acetate salt, among others.
  • the product diamine 23 can be further selectively protected and reductively alkylated with a second aldehyde to obtain an analogous tertiary amine.
  • the diamine 23 can be cyclized to obtain intermediates such as the dihydroimidazole 24 by procedures known in the literature.
  • the ester 24 can then be utilized in a reaction such as illustrated in Scheme 3 hereinabove or can be converted to the amine 26, via the azido intermediate 25. That amine can then be utilized in reactions such as illustrated in Scheme 1.
  • Scheme 10 illustrates preparation of aralkyl imidazolyl intermediates 31 that can be utilized in reactions such as illustrated in Scheme 3.
  • imidazole acetic acid 27 can be converted to the protected acetate 29 by standard procedures, and 29 can be first reacted with an alkyl halide, then treated with refluxing methanol to provide the regiospecifically alkylated imidazole acetic acid ester 30. Hydrolysis provides the acetic acid 31.
  • intermediate 31 can be converted into the homologous amine 34 via the azido intermediate 33, as shown in
  • the Boc protected phthalimidyl alcohol 39 can also be utilized to synthesize 2-aziridinylmethylamines such as 42 (Scheme 13). Treating 39 with 1,1'-sulfonyldiimidazole and sodium hydride in a solvent such as dimethylformamide led to the formation of aziridine 42. The aziridine may then be reacted in the presence of a nucleophile, such as a thiol, in the presence of base to yield, after deprotection, the ring- opened intermediate amine 43.
  • a nucleophile such as a thiol
  • amines such as 48 derived from amino acids such as O-alkylated tyrosines can be prepared according to standard procedures as shown in Scheme 14. Illustrated is a procedure where the amine moiety is derived from the azide of an intermediate such as 47.
  • Schemes 15-18 illustrate syntheses of suitably substituted alkanols useful in the syntheses of the instant compounds wherein the variable W is present as a pyridyl moiety.
  • the hydroxyl moiety of such intermediates may be converted into the corresponding amine, as illustrated in Scheme 15 or may be converted to a suitable leaving group, as illustrated in Scheme 17.
  • Similar synthetic strategies for preparing alkanols that incorporate other heterocyclic moieties for variable W are also well known in the art.
  • Scheme 20 illustrates synthesis of an instant compound wherein a non-hydrogen R 5 b is incorporated in the instant compound.
  • a readily available 4-substituted imidazole 53 may be selectively iodinated to provide the 5-iodoimidazole 54. That imidazole may then be protected and coupled to a suitably substituted benzyl moiety to provide intermediate 55. Intermediate 55 can then undergo the alkylation reactions that were described hereinabove.
  • R' is R 1 a or a protected precursor thereof
  • R' is (R 4 ) r -V- or a protected precursor thereol
  • R'CH 2 - is R 8 or a protected precursor thereof
  • the instant compounds are useful as pharmaceutical agents for mammals, especially for humans. These compounds may be administered to patients for use in the treatment of cancer. Examples of the type of cancer which may be treated with the compounds of this invention include, but are not limited to, colorectal carcinoma, exocrine pancreatic carcinoma, myeloid leukemias and neurological tumors.
  • Such tumors may arise by mutations in the ras genes themselves, mutations in the proteins that can regulate Ras activity (i.e.,
  • NF-1 neurofibromin
  • neu neu
  • ser ser
  • ab1 ser
  • ab1 ser
  • lck ab1
  • fyn fyn
  • 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)).
  • the compounds of this invention are also useful for inhibiting other proliferative diseases, both benign and malignant, wherein Ras proteins are aberrantly activated as a result of oncogenic mutation in other genes (i.e., the Ras gene itself is not activated by mutation to an oncogenic form) with said inhibition being accomplished by the administration of an effective amount of the compounds of the invention to a mammal in need of such treatment.
  • a component of NF-1 is a benign proliferative disorder.
  • the instant compounds may also be useful in the treatment of certain viral infections, in particular in the treatment of hepatitis delta and related viruses (J.S. Glenn et al. Science, 256: 1331-1333 (1992).
  • the compounds of the instant invention are also useful in the prevention of restenosis after percutaneous transluminal coronary angioplasty by inhibiting neointimal formation (C. Indolfi et al. Nature medicine, 1 :541-545(1995).
  • the instant compounds may also be useful in the treatment and prevention of poly cystic kidney disease (D.L. Schaffner et al. American Journal of Pathology, 142:1051-1060 (1993) and B. Cowley, Jr. et 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
  • the compounds can be administered orally or parenterally, including the intravenous, intramuscular, intraperitoneal, subcutaneous, rectal and topical routes of administration.
  • the selected compound may be administered, for example, in the form of tablets or capsules, or as an aqueous solution or suspension.
  • carriers which are commonly used include lactose and com starch, and lubricating agents, such as magnesium stearate, are commonly added.
  • useful diluents include lactose and dried com starch.
  • the active ingredient is combined with emulsifying and suspending agents. If desired, certain sweetening and/or flavoring agents may be added.
  • sterile solutions of the active ingredient are usually prepared, and the pH of the solutions should be suitably adjusted and buffered.
  • the total concentration of solutes should be controlled in order to render the preparation isotonic.
  • the present invention also encompasses a pharmaceutical composition useful in the treatment of cancer, comprising the
  • compositions of this invention include aqueous solutions comprising compounds of this invention and pharmacologically acceptable carriers, e.g., saline, at a pH level, e.g., 7.4.
  • pharmacologically acceptable carriers e.g., saline
  • the solutions may be introduced into a patient's intramuscular blood-stream by local bolus injection.
  • composition is intended to encompass a product comprising the specified ingredients in the specific amounts, as well as any product which results, directly or indirectly, from combination of the specific ingredients in the specified amounts.
  • the daily dosage will normally be determined by the prescribing physician with the dosage generally varying according to the age, weight, and response of the individual patient, as well as the severity of the patient's symptoms.
  • a suitable amount of compound is administered to a mammal undergoing treatment for cancer.
  • Administration occurs in an amount between about 0.1 mg/kg of body weight to about 60 mg/kg of body weight per day, preferably of between 0.5 mg/kg of body weight to about 40 mg/kg of body weight per day.
  • the compounds of the instant invention are also useful as a component in an assay to rapidly determine the presence and quantity of farnesyl-protein transferase (FPTase) in a composition.
  • FPTase farnesyl-protein transferase
  • composition to be tested may be divided and the two
  • 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.
  • 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.
  • Step B Preparation of Piperidine-cis, trans-3,5-dicarboxylic acid methyl ester hydrochloride
  • Piperidine-3,5-dicarboxylic acid methyl ester hydrochloride (7.11 g, 29.9 mmol) was dissolved in THF (60 mL) and H 2 O (60 mL).
  • Sodium bicarbonate (13.81 g, 0.164 mol) was added followed by di-tert-butyl dicarbonate (9.79 g, 44.9 mmol).
  • the mixture was stirred at ambient temperature for 5 hrs.
  • the THF was removed under reduced pressure, and the solution was extracted with CH 2 Cl 2 (3 ⁇ 100 mL). The combined CH 2 Cl 2 layers were washed with brine and dried (MgSO 4 ).
  • Step D Preparation of 1-(t-Butoxycarbonyl)-cis-3- methoxycarbonyl-piperidine-5-carboxylic acid
  • NY-Pivaloyloxymethyl-N ⁇ -phthaloylhistamine (4.55 g, 12.8 mmol) was prepared as previously described (J. C. Emmett, F. H.
  • the volume of the filtrate was reduced to 10 mL, the solution was heated at 55°C for 1 hr, then cooled to room temperature, diluted with EtOAc (25 mL) and filtered to obtain additional white solid. The solids were combined, dried, and used without further purification.
  • Step F Preparation of 1-(t-Butoxycarbonyl)-cis-3-methoxycarbonyl-5-[N-(1-(4-cyanobenzyl)-1H-imidazol-5- ylethyl)carbonyl]-piperidine
  • Step D Following the procedure of Steps D-F but substituting the 1-(t-butoxycarbonyl)piperidine-trans-3,5-dicarboxylic acid methyl ester prepared as described in Step C for the 1-(t-Butoxycarbonyl)piperidine-cis-3,5-dicarboxylic acid methyl ester utilized in Step D provided 1-(t- butoxycarbonyl)-trans-3-methoxy-carbonyl-5-[N-(1 -(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl]-piperidine.
  • Step A Preparation of cis-3-Methoxycarbonyl-5-[N-(1-(4- cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
  • Step B Preparation of 1-Phenethyl-cis-3-methoxycarbonyl-5-[N- (1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
  • N-[1-Phenethyl-cis-5-(N'-(4-cyanobenzyl-1 -imidazol-5-ylethyl)carbamoyl) piperidine-3-carbonyl] methionine methyl ester (19 mg, 0.030 mmol) was dissolved in THF (2 mL) and H 2 O (1 mL). A IN solution of LiOH•H 2 O (30.1 ⁇ L, 0.030 mmol) was added and the solution was stirred overnight at ambient temperature.
  • the solution was purified on a RP HPLC VYDAC column (0.1% TFA in CH 3 CN: 0.1 % TFA in H 2 O, 5:95 to 95:5 gradient) and lyophilized to give the title compound as cis diastereomers.
  • FAB MS 617 (M+1)
  • the solution was heated at 90°C for 4 h.
  • the solution was diluted with EtOAc and was washed with Sat. NaHCO3 solution, water, and brine.
  • the organics were dried (MgSO 4 ), filtered, and concentrated to give the title compound without further purification.
  • Step C Preparation of 1H-Imidazole-4- acetic acid methyl ester hydrochloride
  • Step D Preparation of 1-(Triphenylmethyl)-1H-imidazol-4-ylacetic acid methyl ester
  • Step E Preparation of [1-(4-Cyanobenzyl)-1H-imidazol-5-yl]acetic acid methyl ester
  • Step F Preparation of [1-(4-cyanobenzyl)-1H-imidazol-5-yl]acetic acid A solution of [1-(4-cyanobenzyl)-1H-imidazol-5-yl]acetic acid methyl ester (4.44g, 17.4mmol ) in THF (100ml) and 1 M lithium hydroxide (17.4 ml, 17.4 mmol) was stirred at RT for 18 hr. 1 M HCl (17.4 ml) was added and the THF was removed by evaporation in vacuo. The aqueous solution was lyophilized to afford the title
  • Step G Preparation of 1-(t-Butoxycarbonyl)-cis-3-methoxycarbonyl-5-[N-(1-(4-cyanobenzyl)-1H-imidazol-5- yl)acetylamino]-piperidine
  • Step B Preparation of 1-Phenethyl-cis-3-methoxycarbonyl-5-[N- (1-(4-cyanobenzyl)-1H-imidazol-5-yl)acetylamino] piperidine
  • Step A Preparation of 1 -(2,2-Diphenylethyl)-3-carboxy piperidine
  • Nipecotic acid 300 mg, 2.38 mmol
  • diphenylacetaldehyde (1.26 mL, 7.13 mmol)
  • sodium cyanoborohydride 448 mg, 7.13 mmol
  • HOAc 204 uL, 3.57 mmol
  • MeOH MeOH (20 mL)
  • the solution was concentrated under reduced pressure, take up in ether and 1N NaOH, extract with ether (3X), acidify the aqueous layer with 1N HCl, and extract with EtOAc (3X).
  • the EtOAc layers were dried (MgSO 4 ) and concentrated to give the title compound without further purification.
  • Step B Preparation of 1 -(2,2-Diphenylethyl)-3-[N-(1-(4- cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine 1-(2,2-Diphenylethyl)-3-carboxy piperidine(472 mg, 1,52 mmol),3-(4-cyanobenzyl) histamine (456 mg, 1.52 mmol) (EXAMPLE 1, Step E) HOBT (216 mg, 1.60 mmol), EDC (307 mg, 1.60 mmol), and Et3N (637 uL, 4.57 mmol) were dissolved in DMF (10 mL) and was stirred overnight at ambient temperature.
  • Step A Preparation of S-(-)-Ethyl nipecotate
  • Step D Preparation of 1-(tert-Butyloxycarbonyl)-3(S)-[N- 1- (4- cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
  • 1-(tert-Butoxycarbonyl)piperidine-3(S)-carboxylic acid 5.06 g, 0.022 mol
  • 3-(4-cyanobenzyl)histamine Example 1 , Step E
  • Step E Preparation of 3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazole-5- ethyl)carbamoyl] piperidine dihydrochloride
  • Step F Preparation of 2-(3-Chlorophenyl)-2-phenyl oxirane
  • Step H Preparation of 1-(2-(3-Chlorophenyl)-2-phenylethyl)-3- (S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5- ylethyl)carbamoyl] piperidine
  • Step A Preparation of Ethyl 1-[2-(2-pyridyl)-2-phenyl-2- hydroxyethyl] piperidine-3(S)- carboxylate
  • Step B Preparation of 1-[2-(2-Pyridyl)-2-phenyl-2-hydroxyethyl] piperidine-3(S)- carboxylic acid
  • Step C Preparation of 1-[2-(2-Pyridyl)-2-phenyl-2-hydroxyethyl]- 3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5- ylethy l)carbamoyl] piperidine
  • Step B Preparation of 1-(2-Pyridylethyl) piperidine-3(S)- carboxylic acid
  • Step C Preparation of 1-(2-Pyridylethyl)-3(S)-[N-(1-(4- cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine Following the procedure of Example 15, Step C, the title compound was prepared. FAB MS (M+1) 413.
  • Step B Preparation of 1-Phenyl-(S)- piperidine carboxylic acid
  • Step C Preparation of 1-Phenyl-3(S)-[N-(1-(4-cyanobenzyl)-1H- imidazol-5-ylethyl)carbamoyl] piperidine
  • Step C Preparation of 1-(2,2-Diphenylethyl)-3(S)-tosyloxymethyl- piperidine
  • 1-(2,2-Diphenylethyl)-3(S)-hydroxymethyl-piperidine (1.01 g, 3.41 mmol) was dissolved in dry pyridine (25 mL) and tosyl chloride (0.684 g, 3.58 mmol) was added to the solution After stirring at ambient temperature for 18 hr, the solution was concentrated, the residue taken up in EtOAc, washed with sat. NaHCO 3 solution, H 2 O, brine, and dried (MgSO 4 ). Filtration and concentration gave the title compound without further purification.
  • Step D Preparation of 1 -(2,2-Diphenylethyl)-3(S)- acetylthiomethyl-piperidine
  • Step D Preparation of the disulfide of 1-(2,2-Diphenylethyl)-3(S)- mercaptomethyl-piperidine
  • Step E Preparation of 1-(2,2-Diphenylethyl)-3(S)- mercaptomethyl-piperidine
  • the filtrate was concentrated in vacuo to a volume (100 mL), reheated at 60 °C for another 2hrs, cooled to room temperature, and concentrated in vacuo to provide a pale yellow solid. All of the solid material was combined, dissolved in methanol (500mL), and warmed to 60 °C. After 2hrs, the solution was concentrated in vacuo to provide a white solid which was triturated with hexane to remove soluble materials. Removal of residual solvents in vacuo provided the titled product hydrobromide as a white solid which was used in the next step without further purification.
  • the material was sufficiently pure to be used without further
  • Step K Preparation of 1-(2,2-Diphenylethyl)-3(S)-[N-(1-(4- cyanobenzyl)- H-imidazol-5-ylethylthiomethyl] piperidine 1-(2,2-Diphenylethyl)-3(S)-mercaptomethyl-piperidine (0.322 mmol), 1-(4-cyanobenzyl)-5-(chloromethyl)-imidazole (0.1 17 g, 0.386 mmol), and diisopropylethylamine (0.168 mL, 0.966 mmol) were dissolved in CH 2 Cl 2 (10mL) and refluxed overnight.
  • Step A Preparation of 1-(tert-Butyloxycarbonyl)-3(S)-[N-(1-(4- cyanobenzyl)-1H-imidazol-5-ylethyl)-N-methylcarbamoyl] piperidine
  • Step B Preparation of 1-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazole- 5-ethyl)-N-methylcarbamoyl] piperidine
  • Step C Preparation of 1-(2,2-Diphenylethyl)-3(S)-[N-1-(4- cyanobenzyl)-1H-imidazol-5-ylethyl)-N-methylcarbamoyl] piperidine
  • Step B Preparation of 1-(2,2-Diphenylethyl)-3(S)-[N-(1- (cyanobenzy1)-1H-imidazol-5-ylethyl)aminomethyl]- piperidine
  • Step B Preparation of 1-(2,2-Diphenylethyl)-3(S)-[5-(4- cyanobenzyl)-1H-imidazol-1-ylmethyl] piperidine bis trifluoroacetate bistrifluoroacetate
  • Step C Preparation of 1-(2,2-Diphenylethyl)-3(S)-[5-(4- cyanobenzyl)-1H-imidazol-1-ylethylcarbamoyl] piperdine
  • 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 [3H]-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 [ 3 H]-FPP was utilized during the reaction period.
  • Purified compounds were dissolved in 100% dimethyl sulfoxide (DMSO) and were diluted 20-fold into the assay. Percentage inhibition is measured by the amount of
  • 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%
  • the cells are labelled in 3 ml methionine-free DMEM supple-meted with 10% regular DMEM, 2% fetal bovine serum and 400 mCi[ 35 S]methionine (1000 Ci/mmol).
  • 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 ⁇ 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 Y13-259 (Furth, M.E. et al., J. Virol.
  • Rat 1 cells transformed with either v-ras, v-raf, or v-mos are seeded at a density of 1 ⁇ 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.

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
INHIBITORS OF FARNESYL-PROTEIN TRANSFERASE
BACKGROUND OF THE INVENTION
The Ras protein is part of a signalling pathway that links cell surface growth factor receptors to nuclear signals initiating cellular proliferation. Biological and biochemical studies of Ras action indicate that Ras functions like a G-regulatory protein. 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. (52:851 - 891 (1993)). Mutated ras genes are found in many human cancers, including colorectal carcinoma, exocrine pancreatic carcinoma, and myeloid leukemias. The protein products of these genes are defective in their GTPase activity and constitutively transmit a growth stimulatory signal.
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 al, Nature 570:583-586 (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 farnesylation. 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., 91:9141-9145 (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. Farnesyl-protein transferase utilizes farnesyl pyrophosphate to covalently modify the Cys thiol group of the Ras CAAX box with a farnesyl group (Reiss et al., Cell, 62:81-88 (1990); Schaber et al, J. Biol. Chem., 265: 14701-14704 (1990); Schafer et al., Science, 249: 1133-1139 (1990); Manne et al., Proc. Natl Acad. Sci USA, 87:7541-7545 (1990)). 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 two general classes. The first are analogs of farnesyl diphosphate (FPP), while the second class of inhibitors is related to the protein substrates (e.g., Ras) for the enzyme. The peptide derived inhibitors that have been described are generally cysteine containing molecules that are related to the CAAX motif that is the signal for protein prenylation. (Schaber et al., ibid; Reiss et. al., ibid; Reiss et al., PNAS, 88:732-736 (1991)). Such inhibitors may inhibit protein prenylation while serving as alternate substrates for the farnesyl-protein transferase enzyme, or may be purely competitive inhibitors (U.S.
Patent 5, 141,851 , University of Texas; N.E. Kohl et al., Science, 260:1934-1937 (1993); Graham, et al., J. Med. Chem., 37, 725 (1994)). 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 shown 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 H7-112930). It has also recently been disclosed that certain tricyclic compounds which optionally incorporate a piperidine moiety are inhibitors of FPTase (WO 95/10514, WO 95/10515 and WO 95/10516).
It is, therefore, an object of this invention to develop novel peptidomimetic compounds that do not have a thiol moiety, and that will inhibit 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 peptidomimetic piperidine, 1,4-dihydropyridine and 1,2,3,4-tetrahydropyridine compounds which inhibit the farnesyl-protein transferase.
Furthermore, these compounds differ from such heterocyclic
compounds previously described as inhibitors of farnesyl-protein transferase with respect to the position of substituents about the nitrogen containing ring. 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 formulae 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:
R1 a, R1 b and R1 c are independently selected from:
a) hydrogen,
b) unsubstituted or substituted aryl, unsubstituted or
substituted heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, R8O-, R9S(O)m-, R8C(O)NR8-, CN, NO2, (R8)2N-C(NR8)-, R8C(O)-, R8OC(O)-, N3, -N(R8)2, or R9OC(O)NR8-,
c) C1-C6 alkyl unsubstituted or substituted by unsubstituted or substituted aryl, heterocyclic, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, R8O-, R9S(O)m-, R8C(O)NR8-, CN, (R8)2N-C(NR8)-, R8C(O)-, R8OC(O)-, N3, -N(R8)2, or R9OC(O)-NR8-;
R2 is selected from: H; unsubstituted or substituted C1 -8 alkyl,
unsubstituted or substituted C2-8 alkenyl, unsubstituted or substituted aryl, ,
unsubstituted or substituted heterocycle,
Figure imgf000007_0002
Figure imgf000007_0003
and -S(O)2R6,
wherein the substituted group is substituted with one or more of:
1) aryl or heterocycle, unsubstituted or substituted with one or two groups selected from:
Figure imgf000007_0004
Figure imgf000008_0001
Figure imgf000009_0003
or ,
R3 is selected from: H;
Figure imgf000009_0001
Figure imgf000009_0002
R4 is independently selected from:
a) hydrogen,
b) unsubstituted or substituted aryl, unsubstituted or
substituted heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, perfluoroalkyl, F, Cl, Br, R8O-, R9S(O)m- , R8C(O)NR8-, CN, NO2, R8 2N-C(NR8)-, R8C(O)-, R8OC(O)-, N3, -N(R8)2, or R9OC(O)NR8-, and c) C1-C6 alkyl unsubstituted or substituted by aryl,
heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, perfluoroalkyl, F, CI, Br, R8O-, R9S(O)m-, R8C(O)NH-, CN, H2N-C(NH)-, R8C(O)-, R8OC(O)-, N3, -N(R8)2, or R8OC(O)NH-;
R5 is independently selected from:
a) hydrogen,
b) C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl,
perfluoroalkyl, F, Cl, Br, R8O-, R9S(O)m-, R8C(O)NR8-, CN, NO2, (R8)2N-C-(NR8)-, R8C(O)-, R8OC(O)-, N3, -N(R8)2, or R9OC(O)NR8-, and
c) C1-C6 alkyl, unsubstituted or substituted by perfluoroalkyl,
F, Cl, Br, R8O-, R9S(O)m-, R8C(O)NR8-, CN, (R8)2N- C(NR8)-, R8C(O)-, R8OC(O)-, N3, -N(R8)2, or R9OC(O)NR8-; R6, R7 and R7a are independently selected from: H; C 1-4 alkyl, C3 -6 cycloalkyl, heterocycle, aryl, C1 -4 perfluoroalkyl, unsubstituted or substituted with one or two substituents selected from:
Figure imgf000009_0004
Figure imgf000010_0001
R6 and R7 may be joined in a ring;
R7 and R7a may be joined in a ring; R8 is independently selected from hydrogen, C1 -C6 alkyl, benzyl,
2,2,2-trifluoroethyl and aryl; R9 is independently selected from C1-C6 alkyl and aryl;
R10 is selected from: H; R8C(O)-; R9S(O)m-; unsubstituted or substituted C1 -4 alkyl, unsubstituted or substituted C3 -6 cycloalkyl, unsubstituted or substituted heterocycle, unsubstituted or substituted aryl, substituted aroyl, unsubstituted or substituted heteroaroyl, substituted arylsulfonyl, unsubstituted or substituted heteroarylsulfonyl, wherein the substituted group is substituted with one or two substituents selected from:
Figure imgf000010_0002
Figure imgf000011_0001
A1 and A2 are independently selected from: a bond, -CH=CH-, -C≡C-,
-C(O)-, -C(O)NR8-, -NR8C(O)-, O, -N(R8)-,
-S(O)2N(R8)-, -N(R8)S(O)2-, or S(O)m; V is selected from:
a) hydrogen,
b) heterocycle,
c) aryl,
d) C1-C20 alkyl wherein from 0 to 4 carbon atoms are
replaced with a a heteroatom selected from O, S, and N, and
e) C2-C20 alkenyl,
provided that V is not hydrogen if A1 is S(O)m and V is not hydrogen if A1 is a bond, n is 0 and A2 is S(O)m;
W is a heterocycle;
X is a bond, -C(=O)NR10-, -NR10C(=O)-, -S(O)m-, -NR10-, O or -C(=O)-; m is 0, 1 or 2;
n is 0, 1 , 2, 3 or 4;
p is 0, 1, 2, 3 or 4; q is 0, 1 , 2, 3 or 4;
r is 0 to 5, provided that r is 0 when V is hydrogen;
s is 1 or 2;
t is 0 or 1 ; and the dashed lines represent optional double bonds; or an optical isomer or a pharmaceutically acceptable salt thereof. A preferred embodiment of the compounds of this invention is illustrated by the following formula:
Figure imgf000012_0001
wherein: R1 a and R1 c are independently selected from: hydrogen, C3-C10 cycloalkyl, R8O-, -N(R8)2, F or C1-C6 alkyl;
R1 b is independently selected from:
a) hydrogen,
b) unsubstituted or substituted aryl, unsubstituted or
substituted heterocycle, C3-C6 cycloalkyl, R8O-, -N(R8)2 or C2-C6 alkenyl,
c) C1-C6 alkyl unsubstituted or substituted by unsubstituted or substituted aryl, heterocycle, C3-C6 cycloalkyl, C2-C6 alkenyl, R8O-, or -N(R8)2;
R2 is selected from:
Figure imgf000013_0003
or ,
R3 is selected from: H;
Figure imgf000013_0001
Figure imgf000013_0002
R4 is independently selected from:
a) hydrogen,
b) C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6
perfluoroalkyl, F, Cl, R8O-, R8C(O)NR8-, CN, NO2, (R8)2N-C(NR8)-, R8C(O)-, R8OC(O)-, -N(R8)2, or R9OC(O)NR8-, and
c) C1-C6 alkyl substituted by C1-C6 perfluoroalkyl, R8O-, R8C(O)NR8-, (R8)2N-C(NR8)-, R8C(O)-, R8OC(O)-, -N(R8)2, or R9OC(O)NR8-;
R5 is selected from:
a) hydrogen,
b) C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, C1 -C6 perfluoroalkyl, F, Cl, R8O-, R9S(O)m-, R8C(O)NR8-, CN, NO2, (R8)2N-C(NR8)-, R8C(O)-, R8OC(O)-, -N(R8)2, or R9OC(O)NR8-, and
c) C1-C6 alkyl unsubstituted or substituted by C1-C6
perfluoroalkyl, F, Cl, R8O-, R9S(O)m-, R8C(O)NR8-, CN, (R8)2N-C(NR8)-, R8C(O)-, R8OC(O)-, -N(R8)2, or R9OC(O)NR8-; R6, R7 and R7a are independently selected from:
H; C1 -4 alkyl, C3-6 cycloalkyl, aryl, heterocycle,
unsubstituted or substituted with:
a) C1 -4 alkoxy,
b) halogen, or
c) substituted or unsubstituted aryl or substituted or unsubstituted heterocycle, R8 is independently selected from hydrogen, C1-C6 alkyl, benzyl,
2,2,2-trifluoroethyl and aryl; R9 is independently selected from C1 -C6 alkyl and aryl; R10 is selected from: H; R8C(O)-; R9S(O)m-; unsubstituted or substituted C1 -4 alkyl, unsubstituted or substituted C3-6 cycloalkyl, unsubstituted or substituted heterocycle, unsubstituted or substituted aryl, substituted aroyl, unsubstituted or substituted heteroaroyl, substituted arylsulfonyl, unsubstituted or substituted heteroarylsulfonyl, wherein the substituted group is substituted with one or two substituents selected from:
Figure imgf000015_0001
A1 and A2 are independently selected from: a bond, -CH=CH-, -CHC-,
-C(O)-, -C(O)NR8-, O, -N(R8)-, or S(O)m; V is selected from:
a) heterocycle selected from pyrrolidinyl, imidazolyl,
pyridinyl, thiazolyl, pyridonyl, 2-oxopiperidinyl, indolyl, quinolinyl, isoquinolinyl, and thienyl, and
b) aryl; W is a heterocycle selected from pyrrolidinyl, imidazolyl, pyridinyl, thiazolyl, pyridonyl, 2-oxopiperidinyl, indolyl, quinolinyl, or isoquinolinyl; X is a bond, -C(=O)NR10-, -NR10C(=O)-, -S(O)m- or -NR10-; m is 0, 1 or 2;
n is 0, 1, 2, 3 or 4;
P is 1, 2 or 3;
q is 0 or 1 ;
r is 0 to 5, provided that r is 0 when V is hydrogen;
s is 1 or 2; and
t is 1 ; or an optical isomer or a pharmaceutically acceptable salt thereof.
Another preferred embodiment of the compounds of this invention are illustrated by the formula B:
Figure imgf000016_0001
wherein:
R1 a and R1 c are independently selected from: hydrogen, C3-C10 cycloalkyl, R8O-, -N(R8)2, F or C1 -C6 alkyl;
R1b is independently selected from:
a) hydrogen, b) aryl, heterocycle, C3-C10 cycloalkyl, R8O-, -N(R8)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, R8O- and -N(R8)2;
R2 is selected from: H; unsubstituted or substituted C1 -8 alkyl,
unsubstituted or substituted C2-8 alkenyl, unsubstituted or substituted aryl, unsubstituted or substituted heterocycle,
Figure imgf000017_0001
,
Figure imgf000017_0002
and
-S(O)2R6,
wherein the substituted group is substituted with one or more of:
Figure imgf000017_0003
Figure imgf000018_0003
or
R3 is selected from: H;
Figure imgf000018_0001
Figure imgf000018_0002
,
R4 is independently selected from:
a) hydrogen,
b) aryl, substituted aryl, heterocycle, substituted heterocycle, C1 -C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1 -C6 perfluoroalkyl, F, Cl, R8O-, R8C(O)NR8-, CN, NO2, (R8)2N-C(NR8)-, R8C(O)-, -N(R8)2, or R9OC(O)NR8-, and c) C1 -C6 alkyl substituted by C1 -C6 perfluoroalkyl, R8O-, R8C(O)NR8-, (R8)2N-C(NR8)-, R8C(O)-, -N(R8)2, or R9OC(O)NR8-; R5a and R5b are independently hydrogen, C1-C6 alkyl, cyclopropyl, trifluoromethyl and halogen; R6, R7 and R7a are independently selected from:
H; C1 -4 alkyl, C3 -6 cycloalkyl, aryl, heterocycle, unsubstituted or substituted with:
a) C1 -4 alkoxy,
b) halogen, or
c) substituted or unsubstituted aryl or substituted or unsubstituted heterocycle; R8 is independently selected from hydrogen, C1 -C6 alkyl, 2,2,2- trifluoroethyl, benzyl and aryl; R9 is independently selected from C1 -C6 alkyl and aryl;
R10 is selected from: H; R8C(O)-; R9S(O)m-; unsubstituted or substituted C1 -4 alkyl, unsubstituted or substituted C3-6 cycloalkyl, unsubstituted or substituted heterocycle, unsubstituted or substituted aryl, substituted aroyl, unsubstituted or substituted heteroaroyl, substituted arylsulfonyl, unsubstituted or substituted heteroarylsulfonyl, wherein the substituted group is substituted with one or two substituents selected from:
Figure imgf000019_0001
A1 and A2 are independently selected from: a bond, -CH=CH-, -C≡C-,
-C(O)-, -C(O)NR8-, O, -N(R8)-, or S(O)m;
V is selected from:
a) hydrogen,
b) heterocycle selected from pyrrolidinyl, imidazolyl,
imidazolinyl, pyridinyl, thiazolyl, pyridonyl, 2- oxopiperidinyl, oxazolyl, indolyl, quinolinyl, isoquinolinyl, triazolyl and thienyl,
c) aryl,
d) C 1-C20 alkyl wherein from 0 to 4 carbon atoms are
replaced with a a heteroatom selected from O, S, and N, and
e) C2-C20 alkenyl, and
provided that V is not hydrogen if A1 is S(O)m and V is not hydrogen if A1 is a bond, n is 0 and A2 is S(O)m;
X is a bond, -C(=O)NR10-, -NR10C(=O)-, -S(O)m- or -NR10-; m is 0, 1 or 2;
n is 0, 1, 2, 3 or 4;
p is 0, 1, 2, 3 or 4;
q is 0 or 1 ; and r is 0 to 5, provided that r is 0 when V is hydrogen; or an optical isomer or pharmaceutically acceptable salt thereof. Another preferred embodiment of the compounds of this invention are illustrated by the formula C:
Figure imgf000021_0001
wherein: R1 a and R1c are independently selected from: hydrogen, C3-C10 cycloalkyl, R8O-, -N(R8)2, F or C1 -C6 alkyl;
R1 b is independently selected from:
a) hydrogen,
b) aryl, heterocycle, C3-C10 cycloalkyl, R8O-, -N(R8)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, R8O- and -N(R8)2;
R2 is selected from: H; unsubstituted or substituted C1 -8 alkyl, unsubstituted or substituted C2-8 alkenyl, unsubstituted or substituted aryl, unsubstituted or substituted heterocycle,
Figure imgf000021_0002
,
Figure imgf000021_0003
and -S(O)2R6,
wherein the substituted group is substituted with one or more of:
Figure imgf000022_0001
Figure imgf000023_0003
or ,
R3 is selected from: H;
Figure imgf000023_0001
Figure imgf000023_0002
R4 is independently selected from:
a) hydrogen,
b) aryl, substituted aryl, heterocycle, substituted heterocycle, C1 -C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1 -C6 perfluoroalkyl, F, Cl, R8O-, R8C(O)NR8-, CN, NO2, (R8)2N-C(NR8)-, R8C(O)-, -N(R8)2, or R9OC(O)NR8-, and
c) C1 -C6 alkyl substituted by C1-C6 perfluoroalkyl, R8O-, R8C(O)NR8-, (R8)2N-C(NR8)-, R8C(O)-, -N(R8)2, or R9OC(O)NR8-;
R5a and R5b are independently hydrogen, C1 -C6 alkyl, cyclopropyl, trifluoromethyl and halogen; R6, R7 and R7a are independently selected from:
H; C1 -4 alkyl, C3-6 cycloalkyl, aryl, heterocycle, unsubstituted or substituted with:
a) C1 -4 alkoxy,
b) halogen, or
c) substituted or unsubstituted aryl or substituted or unsubstituted heterocycle; R8 is independently selected from hydrogen, C1 -C6 alkyl, 2,2,2- trifluoroethyl, benzyl and aryl; R9 is independently selected from C1 -C6 alkyl and aryl;
R10 is selected from: H; R8C(O)-; R9S(O)m-; unsubstituted or substituted C1 -4 alkyl, unsubstituted or substituted C3-6 cycloalkyl, unsubstituted or substituted heterocycle, unsubstituted or substituted aryl, substituted aroyl, unsubstituted or substituted heteroaroyl, substituted arylsulfonyl, unsubstituted or substituted heteroarylsulfonyl, wherein the substituted group is substituted with one or two substituents selected from:
Figure imgf000024_0001
A1 and A2 are independently selected from: a bond, -CH=CH-, -C≡C-,
-C(O)-, -C(O)NR8-, O, -N(R8)-, or S(O)m; V is selected from:
a) hyddogen, b) heterocycle selected from pyrrolidinyl, imidazolyl, imidazolinyl, pyridinyl, thiazolyl, pyridonyl, 2- oxopiperidinyl, oxazolyl, indolyl, quinolinyl, isoquinolinyl, triazolyl and thienyl,
c) aryl,
d) C1-C20 alkyl wherein from 0 to 4 carbon atoms are
replaced with a a heteroatom selected from O, S, and N, and
e) C2-C20 alkenyl, and
provided that V is not hydrogen if A1 is S(O)m and V is not hydrogen if A1 is a bond, n is 0 and A2 is S(O)m;
X is a bond, -C(=O)NR10-, -NR10C(=O)-, -S(O)m- or -NR10-; m is 0, 1 or 2;
n is 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,
-NR8- or O;
q is 0 or 1 ; and
r is 0 to 5, provided that r is 0 when V is hydrogen; or an optical isomer or pharmaceutically acceptable salt thereof.
In a more preferred embodiment of this invention, the inhibitors of farnesyl-protein transferase are illustrated by the formula D:
Figure imgf000025_0001
wherein: R1 a and R1 c are independently selected from: hydrogen, C3-C10
cycloalkyl or C1 -C6 alkyl; R1 b is independently selected from:
a) hydrogen,
b) aryl, heterocycle, C3-C10 cycloalkyl, R8O-, -N(R8)2, F or C2-C6 alkenyl,
c) C1 -C6 alkyl unsubstituted or substituted by aryl,
heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, R8O-, or
-N(R8)2;
R2 is selected from: H; unsubstituted or substituted C1 -8 alkyl, unsubstituted or substituted aryl,
Figure imgf000026_0001
,
Figure imgf000026_0002
and -S(O)2R6, wherein the substituted group is substituted with one or more of:
Figure imgf000026_0003
Figure imgf000027_0001
or ,
R3 is selected from: H;
Figure imgf000028_0001
Figure imgf000028_0002
R4 is independently selected from:
a) hydrogen,
b) aryl, substituted aryl, heterocycle, substituted heterocycle, C1 -C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1 -C6 perfluoroalkyl, F, Cl, R8O-, R8C(O)NR8-, CN, NO2, (R8)2N-C(NR8)-, R8C(O)-, -N(R8)2, or R9OC(O)NR8-, and
c) C1 -C6 alkyl substituted by C1 -C6 perfluoroalkyl, R8O-, R8C(O)NR8-, (R8)2N-C(NR8)-, R8C(O)-, -N(R8)2, or R9OC(O)NR8-;
R5a and R5b are independently hydrogen, ethyl, cyclopropyl or methyl; R6, R7 and R7a are independently selected from:
H; C1 -4 alkyl, C3-6 cycloalkyl, aryl, heterocycle,
unsubstituted or substituted with:
a) C1 -4 alkoxy,
b) halogen, or
c) substituted or unsubstituted aryl or substituted or unsubstituted heterocycle; R8 is independently selected from hydrogen, C1 -C6 alkyl, 2,2,2- trifluoroethyl, benzyl and aryl; R9 is independently selected from C1 -C6 alkyl and aryl;
R10 is selected from: H; R8C(O)-; R9S(O)m-; unsubstituted or substituted C1 -4 alkyl, wherein the substituted alkyl group is substituted with one or two substituents selected from:
Figure imgf000028_0003
Figure imgf000029_0001
A 1 is selected from: a bond, -C(O)-, O, -N(R8)-, or S(O)m; X is a bond, -C(=O)NR10-, -NR10C(=O)-, -S(O)m- or -NR10-; n is 0 or 1 ; provided that n is not 0 if A1 is a bond, O, -N(R8)-, or S(O)m;
m is 0, 1 or 2;
p is 0, 1 , 2, 3 or 4; and
q is 0 or 1 ; or an optical isomer or pharmaceutically acceptable salt thereof. In another more preferred embodiment of this invention, the inhibitors of farnesyl-protein transferase are illustrated by the formula E:
Figure imgf000030_0001
wherein:
R1 a and R1 c are independently selected from: hydrogen, R8O-,
-N(R8)2, F, C3-C10 cycloalkyl or C1 -C6 alkyl;
R1b is independently selected from:
a) hydrogen,
b) aryl, heterocycle, C3-C10 cycloalkyl, R8O-, -N(R8)2, F or C2-C6 alkenyl,
c) C1 -C6 alkyl unsubstituted or substituted by aryl,
heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, R8O-, or -N(R8)2; R2 is selected from: H; unsubstituted or substituted C1 -8 alkyl, unsubstituted or substituted aryl,
Figure imgf000030_0002
,
Figure imgf000030_0003
and -S(O)2R6, wherein the substituted group is substituted with one or more of:
Figure imgf000030_0004
Figure imgf000031_0001
or ,
R3 is selected from: H;
Figure imgf000032_0001
Figure imgf000032_0002
R4 is independently selected from:
a) hydrogen,
b) aryl, substituted aryl, heterocycle, substituted heterocycle, C1 -C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 perfluoroalkyl, F, Cl, R8O-, R8C(O)NR8-, CN, NO2, (R8)2N-C(NR8)-, R8C(O)-, -N(R8)2, or R9OC(O)NR8-, and
c) C 1-C6 alkyl substituted by C1 -C6 perfluoroalkyl, R8O-, R8C(O)NR8-, (R8)2N-C(NR8)-, R8C(O)-, -N(R8)2, or R9OC(O)NR8-;
R5a and R5b are independently hydrogen, ethyl, cyclopropyl or methyl; R6, R7 and R7a are independently selected from:
H; C1 -4 alkyl, C3-6 cycloalkyl, aryl, heterocycle,
unsubstituted or substituted with:
a) C1 -4 alkoxy,
b) halogen, or
c) substituted or unsubstituted aryl or substituted or unsubstituted heterocycle; R8 is independently selected from hydrogen, C1 -C6 alkyl, 2,2,2- trifluoroethyl, benzyl and aryl; R9 is independently selected from C1 -C6 alkyl and aryl;
R10 is selected from: H; R8C(O)-; R9S(O)m-; unsubstituted or substituted C1 -4 alkyl, wherein the substituted alkyl group is substituted with one or two substituents selected from:
Figure imgf000032_0003
Figure imgf000033_0002
X is a bond, -C(=O)NR10-, -NR10C(=O)-, -S(O)m- or -NR10-; n is 0 or 1 ;
m is 0, 1 or 2;
p is 0, 1, 2, 3 or 4, provided that p is not 0 if X is a bond,
-NR8- or O; and
q is 0 or 1 ; or an optical isomer or pharmaceutically acceptable salt thereof.
In a further embodiment of this invention, the inhibitors of farnesyl-protein transferase are illustrated by the formula F:
Figure imgf000033_0001
wherein:
R1 a and R1 c are independently selected from: hydrogen, C3-C10
cycloalkyl or C1 -C6 alkyl;
R1b is independently selected from:
a) hydrogen,
b) aryl, heterocycle, C3-C10 cycloalkyl, R8O-, -N(R8)2 or F, c) C1 -C6 alkyl unsubstituted or substituted by aryl,
heterocycle, C3-C10 cycloalkyl, R8O-, or -N(R8)2;
R2 is selected from: H; unsubstituted or substituted C1 -8 alkyl, unsubstituted or substituted aryl,
Figure imgf000034_0001
,
Figure imgf000034_0002
and -S(O)2R6, wherein the substituted rou is substituted with one or more of:
Figure imgf000034_0003
Figure imgf000035_0001
or ,
R3 is selected from: H;
Figure imgf000036_0001
Figure imgf000036_0002
R5a and R5b are independently hydrogen, ethyl, cyclopropyl or methyl; R6, R7 and R7a are independently selected from:
H; C1 -4 alkyl, C3-6 cycloalkyl, aryl, heterocycle,
unsubstituted or substituted with:
a) C 1 -4 alkoxy,
b) halogen, or
c) substituted or unsubstituted aryl or substituted or unsubstituted heterocycle; R8 is independently selected from hydrogen, C1 -C6 alkyl, 2,2,2- trifluoroethyl, benzyl and aryl; R9 is independently selected from C1 -C6 alkyl and aryl;
R10 is selected from: H; R8C(O)-; R9S(O)m-; unsubstituted or substituted C1 -4 alkyl, wherein the substituted alkyl group is substituted with one or two substituents selected from:
Figure imgf000036_0003
Figure imgf000037_0002
X is a bond, -C(=O)NR10-, -NR10C(=O)-, -S(O)m- or -NR10-; m is 0, 1 or 2;
p is 0, 1 , 2, 3 or 4; and
q is 0 or 1 ; or an optical isomer or pharmaceutically acceptable salt thereof.
In a further embodiment of this invention, the inhibitors of farnesyl-protein transferase are illustrated by the formula G:
Figure imgf000037_0001
wherein:
R1 a and R1 c are independently selected from: hydrogen, R8O-,
-N(R8)2, F, C3-C10 cycloalkyl or C1 -C6 alkyl;
R1 b 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, R8O-, or -N(R8)2; R2 is selected from: H; unsubstituted or substituted C1 -8 alkyl, unsubstituted or substituted aryl,
Figure imgf000038_0001
,
Figure imgf000038_0002
and -S(O)2R6, wherein the substituted group is substituted with one or more of:
Figure imgf000038_0003
Figure imgf000039_0003
or ,
R3 is selected from: H;
Figure imgf000039_0001
Figure imgf000039_0002
R5a and R5b are independently hydrogen, ethyl, cyclopropyl or methyl; R6, R7 and R7a are independently selected from:
H; C1 -4 alkyl, C3-6 cycloalkyl, aryl, heterocycle,
unsubstituted or substituted with:
a) C1 -4 alkoxy,
b) halogen, or c) substituted or unsubstituted aryl or substituted or unsubstituted heterocycle; R8 is independently selected from hydrogen, C1 -C6 alkyl, 2,2,2- trifluoroethyl, benzyl and aryl; R9 is independently selected from C1 -C6 alkyl and aryl;
R10 is selected from: H; R8C(O)-; R9S(O)m-; unsubstituted or substituted C1 -4 alkyl, wherein the substituted alkyl group is substituted with one or two substituents selected from:
Figure imgf000040_0001
A1 is selected from: a bond, -C(O)-, O, -N(R8)-, or S(O)m;
X is a bond, -C(=O)NR10-, -NR10C(=O)-, -S(O)m- or -NR10-; m is 0, 1 or 2;
n is 0 or 1 ;
p is 1, 2 or 3; and q is 0 or 1 ; or an optical isomer or pharmaceutically acceptable salt thereof. The preferred compounds of this invention are as follows:
1-(t-Butoxycarbonyl)-cis-3-methoxycarbonyl-5-[N-(1 -(4-cyanobenzyl-1H-imidazol-5-ylethyl)carbamoyl] piperidine 1-Phenethyl-cis-3-methoxycarbonyl-5-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
1-(1 -Naphthylmethyl)-cis-3-methoxycarbonyl-5-[N-(1 -(4-cyanobenzyl- 1H-imidazol-5-ylethyl)carbamoyl] piperidine
1-Benzyl-cis-3-methoxycarbonyl-5-[N-(1-(4-cyanobenzyl-1H-imidazol- 5-ylethyl)carbamoyl] piperidine
1-Methyl-cis-3-methoxycarbonyl-5-[N-(1-(4-cyanobenzyl- 1H-imidazol-5-ylethyl)carbamoyl] piperidine
1-(2-Indanyl)-cis-3-methoxycarbonyl-5-[N-(1 -(4-cyanobenzyI-1H-imidazol-5-ylethyl)carbamoyl] piperidine 1-(2-Diphenylethyl)-cis-3-methoxycarbonyl-5-[N-(1-(4-cyanobenzyl-1H-imidazol-5-ylethyl)carbamoyl] piperidine 1-(3-Phenylpropyl)-cis-3-methoxycarbonyl-5-[N-(1-(4-cyanobenzyl-1H-imidazol-5-ylethyl)carbamoyl] piperidine
1-(2-Methylpropyl)-cis-3-methoxycarbonyl-5-[N-(1 -(4-cyanobenzyl-1H-imidazol-5-ylethyl)carbamoyl] piperidine
1-Phenethyl-cis-3-carboxyl-5-[N-(1-(4-cyanobenzyl-1H-imidazol-5-ylethyl)carbamoyl] piperidine 1-Phenethyl-cis-3-[N-(1-morpholinyl)carbamyl]-5-[N-(1-(4-cyanobenzyl-1H-imidazol-5-ylethyl)carbamoyl]piperidine
1-Phenethyl-cis-3-[N-(benzyl)carbamyl]-5-[N-(1-(4-cyanobenzyl-1H-imidazol-5-ylethyl)carbamoyl] piperidine
1-Phenethyl-cis-3-[N-(cyclopropyl)carbamyl]-5-[N-(1-(4-cyanobenzyl-1H-imidazol-5-ylethyl)carbamoyl] piperidine 1-Phenethyl-cis-3-[N-(t-butyl)carbamyl]-5-[N-(1 -(4-cyanobenzyl-1H-imidazol-5-ylethyl)carbamoyl] piperidine
1-(2,2-Diphenylethyl)-cis-3-[N-( 1-morpholinyl)carbamyl]-5-[N-(1-(4-cyanobenzyl-1H-imidazol-5-ylethyl)carbamoyl] piperidine
1-(2,2-Diphenylethyl)-cis-3-[N-(t-butyl)carbamyl]-5-[N-(1-(4-cyanobenzyl- 1H-imidazol-5-ylethyl)carbamoyl] piperidine
N-[1-Phenethyl-cis-5-(N'-(4-cyanobenzyl-1-imidazol-5-ylethyl)carbamyl) piperidine-3-carbonyl] methionine methyl ester
N-[1-Phenethyl-cis-5-(N'-(4-cyanobenzyl-1-imidazol-5-ylethyl)carbamyl) piperidine-3-carbonyl] methionine 1-(t-Butoxycarbonyl)-cis-3-methoxycarbonyl-5-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylacetyl)amino] piperidine
1-Phenethyl-cis-3-methoxycarbonyl-5-[N-(1 -(4-cyanobenzyl)-1H-imidazol-5-ylacetyl)amino] piperidine
1-Diphenylacetyl-cis-3-methoxycarbonyl-5-[N-(1-(4-cyanobenzyl-1H-imidazol-5-ylethyl)carbamoyl] piperidine
1-(t-Butoxycarbonyl)-trans-3-methoxycarbonyl-5-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl]-piperidine 1-(2,2-Diphenylethyl)-3-[N-1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
1-(2,2-Diphenylethyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylacetyl)amino] piperidine
1-(2,2-Diphenylethyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylpropionyl)amino] piperidine 1 -(2,2-Diphenylethyl)-3(S)-[N-(1 -(4-cyanobenzyl)- 1H-imidazol-5-ylcarbonyl)amino] piperidine
1-(Phenylacetyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
1-(Diphenylacetyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
1-(3-Chlorobenzoyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
1-(2-(3-Chlorophenyl)-2-phenylethyl)-3(S)-[N-( 1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine 1-(Dibenzylsuberylmethyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
1-(2-(3-Methylphenyl)-2-phenylethyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
1-(2-(3-Trifluoromethylphenyl)-2-phenylethyl)-3(S)-[N-( 1 -(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
1-(2-(2-Chlorophenyl)-2-phenylethyl)-3(S)-[N-(1 -(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine 1 -(2-(4-Chlorophenyl)-2-phenylethyl)-3(S)-[N-(1 -(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
1-(2-(3-Aminomethylphenyl)-2-phenylethyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
1-(2-Phenethyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine 1-(2-Phenethyl)-3-(R)-[N-(1-(4-cyanobenzyl)- 1 H-imidazol-5-ylethyl)carbamoyl] piperidine
1-(3-Phenylpropyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
1-(2-Benzyl)-3-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
1-(2-Chlorobenzyl)-3-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
1-(3-Chlorobenzyl)-3-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine 1-(3-Chlorobenzyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
1 -(2,2-Diphenyl-2-hydroxyethyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
1-(3-Methoxybenzyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
1-(3,5-Dichlorobenzyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine 1-(3-Trifluoromethoxybenzyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
1 -(2,5-Dimethylbenzyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
1-(3-Trifluoromethylbenzyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine 1 -(3-Bromobenzyl)-3(S)-[N-(1-(4-cyanobenzyl)-l H-imidazol-5-ylethyl)carbamoyl] piperidine
1-(3-Methylbenzyl)-3(S)-[N-(1-(4-cyanobenzyl)-1-H-imidazol-5-ylethyl)carbamoyl] piperidine
1-Isobutyl-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
1-(2-Methyl-2-phenylethyl)-3-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
1-(2-(1-Morpholinyl)-2-phenylethyl)-3(S)-[N-(1 -(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine 1-(2-(1-Piperidinyl)-2-phenylethyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
1-(2,2-Diphenylethyl)-3(S)-[N-(1-(4-cyanobenzyl)-2-methyl-1H-imidazol-5-ylethyl)carbamoyl] piperidine
1-(2,2-Diphenylethyl)-3(S)-[N-(1-(4-methoxybenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
1-(Diphenylmethyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine 1-(3-Methoxyphenethyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
1-(1-Naphthylethyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
1-(3-Chlorophenethyl)-3(S)-[N-1-(4- cyanobenzy)l-1H-imidazol-5-ylethyl)carbamoyl] piperidine 1-(α-Methylbenzyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
1-(Diphenylmethyl)-3(S)-[N-(1-(4-cyanobenzyl)-2-methyl-1H-imidazol-5-ylethyl)carbamoyl] piperidine
1-(α-Toluenesulfonyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
1-(Benzenesulfonyl)-3(S)-[N-(1-(4-cyanobenzyl)- 1H-imidazol-5-ylethyl)carbamoyl] piperidine
1-(1-Naphthylenesulfonyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine 1-(3-Chlorobenzenesulfonyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
1 -(3,5-Dichlorobenzenesulfonyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
1-(α-Toluenesulfonyl)-3-(R)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
1-(α-Toluenesulfonyl)-cis-3-methoxycarbonyl-5-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine 1-(Methanesulfonyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
1-(Diphenylcarbamoyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
1-(Phenylcarbamoyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine 1-[2-(2-Pyridyl)-2-phenyl-2-hydroxyethyl]-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
1-(2-Pyridylethyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
1-Phenyl-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl]piperidine
1 -(3-Methylphenyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
1-(2,2-Diphenylethyl)-3(S)-[2-(1-(4-cyanobenzyl)-1H-imidazol-5-yl)ethylthiomethyl] piperidine 1-(2,2-Diphenylethyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-yl)ethylsulfonylmethyl] piperidine
1-(2,2-Diphenylethyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)-N-methyl-carbamoyl] piperidine
1-(3-Bromobenzyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)N-methyl-carbamoyl] piperidine
1-(2,2-Diphenylethyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)aminomethyl] piperidine
1-(2,2-Diphenylethyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)-N-acetyl-aminomethyl] piperidine 1-(2,2-Diphenylethyl)-3(S)-[N-(1-(4-cyanobenzyl)-2-methyl-1H-imidazol-5-ylethyl)-N-acetyl-aminomethyl] piperidine
1-(2,2-Diphenylethyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)-N-cyclopropylmethyl-aminomethyl] piperidine
1-(2,2-Diphenylethyl)-3(S)-[N-(2-methyl-1H-imidazol-4-ylethyl)-N-(4-cyanobenzoyl)aminomethyl] piperidine
1-(2,2-Diphenylethyl)-3(S)-[5-(4-cyanobenzyl)-1H-imidazol- 1-ylmethyl]piperidine
1-(2,2-Diphenylethyl)-3(S)-[5-(4-cyanobenzyl)-1 H-imidazol-1 -ylethylcarbamoyl] piperdine or an optical isomer or a pharmaceutically acceptable salt thereof.
Specific examples of the compounds of the invention are: 1-Phenethyl-cis-3-[N-(1-morpholinyl)carbamyl]-5-[N-(1-(4-cyanobenzyl-1H-imidazol-5-ylethyl)carbamoyl]piperidine
Figure imgf000048_0001
1-(2-Diphenylethyl)-cis-3-[N-(1-morpholinyl)carbamyl]-5-[N-(1-(4-cyanobenzyl-1H-imidazol-5-ylethyl)carbamoyl] piperidine
Figure imgf000049_0001
N-[1-Phenethyl-5-(N'-(4-cyanobenzyl-1-imidazol-5-ylethyl)carbamyl) piperidine-cis-3-carbonyl] methionine
Figure imgf000049_0002
1-(2-Diphenylethyl)-3-methoxycarbonyl-5-[N-(1-(4-cyanobenzyl-1H-imidazole-cis-5-ethyl)carbamoyl] piperidine
Figure imgf000050_0001
1-(2,2-Diphenylethyl)-cis-3-[N-(1-(4-cyanobenzyl)- 1H-imidazol-5-ylethyl)carbamoyl] piperidine
Figure imgf000050_0002
1-(3-Chlorobenzyl)-3-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
Figure imgf000051_0001
1-(2-(1-Morpholinyl)-2-phenylethyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
Figure imgf000051_0002
1-(Diphenylmethyl)-3(S)-[N-(1-(4-cyanobenzyl)-2-methyl-1H-imidazol-5-ylethyl)carbamoyl] piperidine
Figure imgf000052_0001
1-(3-Methylphenyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
Figure imgf000052_0002
1-(2,2-Diphenylethyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)-N-acetyl-aminomethyl] piperidine
Figure imgf000053_0001
1-(Benzenesulfonyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
Figure imgf000053_0002
or an optical isomer or a pharmaceutically acceptable salt 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 a, R4 etc.) occurs more than one time in any constituent, its definition on each occurence is independent at every other occurence. Also, combinations of substituents/or variables are permissible only if such combinations result in stable compounds.
As used herein, "alkyl" is intended to include both branched and straight-chain saturated aliphatic hydrocarbon groups having the specified number of carbon atoms; "alkoxy" represents an alkyl group of indicated number of carbon atoms attached through an oxygen bridge. "Halogen" or "halo" as used herein means fluoro, chloro, bromo and iodo.
As used herein, "aryl" is intended to mean any stable monocyclic, bicyclic or tricyclic carbon ring of up to 7 members in each ring, wherein at least one ring is aromatic. Examples of
monocyclic and bicyclic aryl elements include phenyl, naphthyl, tetrahydronaphthyl, indanyl, biphenyl, phenanthryl, anthryl or
acenaphthyl. Examples of tricyclic aryl elements include 10,11-dihydro-5H-dibenzo[a,d]cyclohepten-5-yl (which is also known as dibenzylsuberyl), 9-fluorenyl and 9,10-dihydroanthracen-9-yl.
Preferably, "aryl" is a monocyclic or bicyclic carbon ring.
The term heterocycle or heterocyclic, as used herein, represents a stable 5- to 7-membered monocyclic or stable 8- to 11-membered bicyclic heterocyclic ring or stable 13- to 15-membered tricyclic heterocyclic ring, which is either saturated or unsaturated, and which consists of carbon atoms and from one to four heteroatoms selected from the group consisting of N, O, and S, and including any bicyclic group in which any of the above-defined heterocyclic rings is fused to a benzene ring. The heterocyclic ring may be attached at any heteroatom or carbon atom which results in the creation of a stable structure. Examples of monocyclic and bicyclic heterocyclic elements include, but are not limited to, azepinyl, benzimidazolyl, benzisoxazolyl, benzofurazanyl, benzopyranyl, benzothiopyranyl, benzofuryl,
benzothiazolyl, benzothienyl, benzoxazolyl, chromanyl, cinnolinyl, dihydrobenzofuryl, dihydrobenzothienyl, dihydrobenzothiopyranyl, dihydrobenzothiopyranyl sulfone, furyl, imidazolidinyl, imidazolinyl, imidazolyl, indolinyl, indolyl, isochromanyl, isoindolinyl, isoquinolinyl, isothiazolidinyl, isothiazolyl, isothiazolidinyl, morpholinyl,
naphthyridinyl, oxadiazolyl, 2-oxoazepinyl, 2-oxopiperazinyl, 2-oxopiperdinyl, 2-oxopyrrolidinyl, piperidyl, piperazinyl, pyridyl, pyrazinyl, pyrazolidinyl, pyrazolyl, pyridazinyl, pyrimidinyl,
pyrrolidinyl, pyrrolyl, quinazolinyl, quinolinyl, quinoxalinyl,
tetrahydrofuryl, tetrahydroisoquinolinyl, tetrahydroquinolinyl, thiamorpholinyl, thiamorpholinyl sulfoxide, thiazolyl, thiazolinyl, thienofuryl, thienothienyl, and thienyl. Examples of tricyclic
heterocyclic elements include, but are not limited to, 6,11-dihydro-5H-benzo[5,6]cyclohepta[1,2-b]pyridine, 9,10-dihydro-4H-3-thia-benzo[f]azulen-4-yl and 9-xanthenyl. The 6,11-dihydro-5H-benzo[5,6]cyclohepta[1,2-b]pyridine moiety has the following structure:
Figure imgf000055_0001
Preferably, "heterocyclic" is a monocyclic or bicyclic moiety.
As used herein, "heteroaryl" is intended to mean any stable monocyclic, bicyclic or tricyclic carbon ring of up to 7 members in each ring, wherein at least one ring is aromatic and wherein from one to four carbon atoms are replaced by heteroatoms selected from the group consisting of N, O, and S. Examples of monocyclic and bicyclic heteroaryl elements include, but are not limited to, benzimidazolyl, benzisoxazolyl, benzofurazanyl, benzopyranyl, benzothiopyranyl, benzofuryl, benzothiazolyl, benzothienyl, benzoxazolyl, chromanyl, cinnolinyl, dihydrobenzofuryl, dihydrobenzothienyl,
dihydrobenzothiopyranyl, dihydrobenzothiopyranyl sulfone, furyl, imidazolyl, indolinyl, indolyl, isochromanyl, isoindolinyl, isoquinolinyl, isothiazolyl, naphthyridinyl, oxadiazolyl, pyridyl, pyrazinyl, pyrazolyl, pyridazinyl, pyrimidinyl, pyrrolyl, quinazolinyl, quinolinyl, quinoxalinyl, tetrahydroisoquinolinyl, tetrahydroquinolinyl, thiazolyl, thienofuryl, thienothienyl, and thienyl. Examples of tricyclic heteroaryl elements include, but are not limited to, 6,1 1-dihydro-5H-benzo[5,6]cyclohepta[1,2-b]pyridine. Preferably, "heteroaryl" is a monocyclic or bicyclic moiety.
As used herein, the terms "substituted aryl", "substituted heterocycle" and "substituted cycloalkyl" are intended to include the cyclic group containing from 1 to 3 substitutents in addition to the point of attachment to the rest of the compound. Such substitutents are preferably selected from the group which includes but is not limited to F, 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-and C1-C20 alkyl.
When R6 and R7 or R7 and R7a are combined to form a ring, cyclic amine moieties are formed. Examples of such cyclic moieties include, but are not limited to:
Figure imgf000056_0001
In addition, such cyclic moieties may optionally include another heteroatom(s). Examples of such heteroatom-containing cyclic amine moieties include, but are not limited to:
Figure imgf000057_0001
Lines drawn into the ring systems from substituents (such as from R2, R3, R4 etc.) indicate that the indicated bond may be attached to any of the substitutable ring carbon atoms.
Preferably, R1 a and R1 b are independently selected from: hydrogen, -N(R8)2, R8C(O)NR8- or C1-C6 alkyl which is
unsubstituted or substituted by -N(R8)2, R8O- or RSC(O)NR8-.
Preferably, R2 is selected from:
Figure imgf000057_0002
Figure imgf000058_0001
Preferably, R2 comprises at least one unsubstituted or substituted phenyl.
Preferably, R4 is selected from: hydrogen, perfluoroalkyl, F, Cl, Br, R8O-, R9S(O)m-, CN, NO2, R8 2N-C(NR8)-, R8C(O)-, N3, -N(R8)2, R9OC(O)NR8- and C1 -C6 alkyl.
Preferably, R5 is hydrogen.
Preferably, R7b is C1 -C6 alkyl substituted with hydrogen or an unsubstituted or substituted aryl group.
Preferably, R8 is selected from H, C1-C6 alkyl and benzyl. Preferably, A1 and A2 are independently selected from: a bond, -C(O)NR8-, -NR8C(O)-, O, -N(R8)-, -S(O)2N(R8)- and- N(R8)S(O)2-.
Preferably, V is selected from hydrogen, heterocycle and aryl.
Preferably, W is imidazolyl.
Preferably, X is a bond, -C(=O)NR10-, -NR10C(=O)- or -NR10-
Preferably, n, p and r are independently 0, 1 , or 2.
Preferably t is 1.
The pharmaceutically acceptable salts of the compounds of this invention include the conventional non-toxic salts of the compounds of this invention as formed, e.g., from non-toxic inorganic or organic acids. 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.
It is intended that the definition of any substituent or variable (e.g., R1 a, Z, n, etc.) at a particular location in a molecule be independent of its definitions elsewhere in that molecule. Thus,
-N(R8)2 represents -NH2, -NHCH3, -NHC2H5, etc. It is understood that substituents and substitution patterns on the compounds of the instant invention can be selected by one of ordinary skill in the art to provide compounds that are chemically stable and that can be readily synthesized by techniques known in the art, as well as those methods set forth below, from readily available starting materials.
The pharmaceutically acceptable salts of the compounds of this invention 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.
Abbreviations used in the description of the chemistry and in the Examples that follow are:
Figure imgf000059_0001
Figure imgf000060_0001
The compounds of this invention are prepared by
employing reactions as shown in the Schemes 1-21, in addition to other standard manipulations such as ester hydrolysis, cleavage of protecting groups, etc., as may be known in the literature or exemplified in the experimental procedures. While stereochemistry is shown in the Schemes, a person of ordinary skill in the art would understand that the illustrated compounds represent racemic mixtures which may be separated at a subsequent purification step or may be utilized as the racemic mixture.
These reactions may be employed in a linear sequence to provide the compounds of the invention or they may be used to synthesize fragments which are subsequently joined by the reductive alkylation or acylation reactions described in the Schemes.
Synopsis of Schemes 1-8:
The requisite intermediates are in some cases commercially available, or can be prepared according to literature procedures, for the most part. In Schemes 1-3, for example, the syntheses of 1,3,5- trisubstituted piperidines are outlined. The reactions described therein may be similarly applied to suitably protected commercially available nipecotic acid or nipecotamide to provide compounds of the instant invention wherein R3 is hydrogen. As shown in Scheme 1, the pyridinedicarboxylic acid diester may be catalytically hydrogenated and then N-protected to provide a mixture of piperidine diesters 1. The protected piperidine can then be partially hydrolyzed to provide a racemic mixture of 3,5-cis- and trans-isomers, that can be separated by chromatography. The remainder of Scheme 1 and Schemes 2 and 3 illustrate manipulation of the racemic mixture of the cis-isomers. It is well understood by one of ordinary skill in the art that such chemical manipulations can also be applied to the racemic mixture of the trans-isomers to obtain other compounds of the instant invention. Furthermore, such manipulations can also be applied to enantiomerically pure isomers (i.e., the (-f)-cis isomer or the (-)-cis isomer). The trans-isomer may also be epimerized to the cis-isomer by treatment with a base, such as sodium carbonate. Racemic final compounds may be separated on a chiral preparative HPLC column to give their respective diastereomers.
The monocarboxylic acid 2 can be treated with an appropriately substituted amine in the presence of a suitable coupling reagent, such as EDC/HOBT, and the like, to provide the 5-carboxynipecotamide 3. The suitably substituted 5-carboxynipecotamide is then deprotected and the piperidine nitrogen can then be reductively alkylated to provide intermediate 4. The remaining ester moiety is saponified and then similarly functionalized with another suitably substituted amine to provide the bisamidopiperidine 5.
An alternative synthetic route to compound 5, starting with the carboxynipecotamide 3, is illustrated in Scheme 2.
As shown in Scheme 3, the monocarboxylic acid 2 can undergo a Curtius rearrangment to provide the piperidine 6 after catalytic reduction. Subsequent amide formation provides intermediate 7, which is then subjected to the reactions illustrated in Scheme 1 to provide compound 8 of the instant invention.
The instant invention also includes 1,4-dihydropyridine and 1,2,3,4-tetrahydropyridine analogs of the piperidine compounds whose syntheses are described above. Scheme 4 illustrates the synthetic route to the intermediates 11 and 13 which correspond to the saturated ring intermedate 2 illustrated in Scheme 1. Thus, the appropriately substituted pyridine may be N-alkylated to provide the quaternary intermediate 9. Subsequent reduction of this intermediate provides the 1 ,4-dihydropyridine 10, which can be selectively hydrolized to the key intermediate 11. Alternatively, the 1 ,4-dihydropyridine 10 can be further reduced to provide the enantiomeric mixture of
tetrahydropyridines 12, which can be hydrolized and resolved by chromatography to provide the key intermediate 13 (and the
enantiomer which is not illustrated). Intermediates 11 and 13 can then undergo synthetic modifications as described hereinabove in Schemes 1-3.
Schemes 5-7 illustrate the syntheses of 1,3-disubstituted piperidines of the instant invention wherein the "X" moiety is other than an amido moiety. The reactions illustrated therein may be modified by using appropriate protecting groups and reagents well known to one skilled in the art to provide 1,3,5-trisubstituted piperidines of the instant invention.
Scheme 5 illustrates the syntheses of compounds of the instant invention wherein "X" is -S- or -SO2-. A racemic nipecotate 14 can be resolved by the selective crystallization of chiral tartrate salts and is then reductively alkylated to provide the ester 15. Intermediate 15 is reduced to the alcohol 16, activated and treated with a suitable
thioacetate to provide the thioester 17. The thiol is then generated and may be alkylated and optionally oxidized to provide compounds 18 and 19 of the instant invention.
The intermediate 16 may be selectively oxidized back to an aldehyde, which can then be utilized to reductively alkylate a suitably substituted amine to provide the instant compound 20. The secondary amine of 20 can be further functionalized as illustrated.
The activated alcohol can also be reacted with a suitably substituted imidazolyl to provide compounds of the instant invention wherein "X" is a bond, as shown in Scheme 7.
Scheme 8 illustrates the syntheses of compounds of the instant invention wherein R2 is an aryl moiety.
Figure imgf000063_0001
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
In the above Schemes it is understood that
Figure imgf000073_0001
or a protected precursor thereof;
RScCH2- is R2 or a protected precursor thereof; and RSb- is R6 or a protected precusor thereof; and
R- is a "substituent" or a protected precusor thereof.
It is understood that a variety of amines and acids, either commercially available or readily synthesized by reactions well known in the art, which contain the side-chain moieties RSa and RSd(C=O) may be utilized in the reactions described hereinabove. Schemes 9-21 illustrate specific reactions wherein such intermediates containing the side-chain moieties RSa and RSd(C=O) may be prepared. It is understood that while Schemes 9-21 illustrate preparation of both protected and unprotected intermediates, a person of ordinary skill would appreciate that subsequent reactions which utilize those
intermediates, such as those described in Schemes 1-8, may require protection and eventual deprotection of certain intermediate moieties..
The selectively protected intermediate 20 utilized in the synthesis illustrated in Scheme 9 can be reductively alkylated with a variety of aldehydes, such as 21. The aldehydes can be prepared by standard procedures, such as that described by O. P. Goel, U. Krolls, M. Stier and S. Kesten in Organic Syntheses, 1988, 67, 69-75. The reductive alkylation can be accomplished at pH 5-7 with a variety of reducing agents, such as sodium triacetoxyborohydride or sodium cyanoborohydride in a solvent such as dichloroethane, methanol or dimethylformamide. The ester product 22 can be deprotected with trifluoroacetic acid in methylene chloride to give the substituted diamine 23. That diamine may be isolated in the salt form, for example, as a trifluoroacetate, hydrochloride or acetate salt, among others. The product diamine 23 can be further selectively protected and reductively alkylated with a second aldehyde to obtain an analogous tertiary amine. Alternatively, the diamine 23 can be cyclized to obtain intermediates such as the dihydroimidazole 24 by procedures known in the literature. The ester 24 can then be utilized in a reaction such as illustrated in Scheme 3 hereinabove or can be converted to the amine 26, via the azido intermediate 25. That amine can then be utilized in reactions such as illustrated in Scheme 1.
Scheme 10 illustrates preparation of aralkyl imidazolyl intermediates 31 that can be utilized in reactions such as illustrated in Scheme 3. Thus imidazole acetic acid 27 can be converted to the protected acetate 29 by standard procedures, and 29 can be first reacted with an alkyl halide, then treated with refluxing methanol to provide the regiospecifically alkylated imidazole acetic acid ester 30. Hydrolysis provides the acetic acid 31.
Alternatively, intermediate 31 can be converted into the homologous amine 34 via the azido intermediate 33, as shown in
Scheme 1 1. This amine can then be utilized in reactions such as illustrated in Scheme 1.
Preparation of amine intermediates having mixed heteroatom substitution is illustrated in Schemes 12 and 13. Thus the protected serine 35 can be reduced to the alcohol 36, which can then undergo a Mitsunobu reaction to provide the phthalimidyl intermediate 37. Deprotection and selective reprotection give the alcohol 39, which can be oxidized to the aldehyde 40. The aldehyde 40 can be
subsequently alkylated and finally deprotected to provide the amine intermediate 41.
The Boc protected phthalimidyl alcohol 39 can also be utilized to synthesize 2-aziridinylmethylamines such as 42 (Scheme 13). Treating 39 with 1,1'-sulfonyldiimidazole and sodium hydride in a solvent such as dimethylformamide led to the formation of aziridine 42. The aziridine may then be reacted in the presence of a nucleophile, such as a thiol, in the presence of base to yield, after deprotection, the ring- opened intermediate amine 43.
In addition, amines such as 48 derived from amino acids such as O-alkylated tyrosines can be prepared according to standard procedures as shown in Scheme 14. Illustrated is a procedure where the amine moiety is derived from the azide of an intermediate such as 47.
Schemes 15-18 illustrate syntheses of suitably substituted alkanols useful in the syntheses of the instant compounds wherein the variable W is present as a pyridyl moiety. The hydroxyl moiety of such intermediates may be converted into the corresponding amine, as illustrated in Scheme 15 or may be converted to a suitable leaving group, as illustrated in Scheme 17. Similar synthetic strategies for preparing alkanols that incorporate other heterocyclic moieties for variable W are also well known in the art.
Compounds of the instant invention wherein the A1 (CR1 a2)nA2(CR1 a2)n linker is a substituted methylene may be synthesized by the methods shown in Scheme 19. Thus, the N-protected imidazolyl iodide 50 is reacted, under Grignard conditions with a suitably protected benzaldehyde to provide the alcohol 51. Acylation, followed by the alkylation procedure illustrated in the Schemes above (in particular, Scheme 7) provides the instant compound 52. If other R1 substituents are desired, the acetyl moiety can be manipulated as illustrated in the Scheme.
Scheme 20 illustrates synthesis of an instant compound wherein a non-hydrogen R5 b is incorporated in the instant compound. Thus, a readily available 4-substituted imidazole 53 may be selectively iodinated to provide the 5-iodoimidazole 54. That imidazole may then be protected and coupled to a suitably substituted benzyl moiety to provide intermediate 55. Intermediate 55 can then undergo the alkylation reactions that were described hereinabove.
Compounds of the instant invention wherein the A1(CR12)nA2(CR12)n linker is oxygen may be synthesized by methods known in the art, for example as shown in Scheme 21. The suitably substituted phenol 56 may be reacted with methyl N- (cyano)methanimidate to provide the 4-phenoxyimidazole 57. After selective protection of one of the imidazolyl nitrogens, the intermediate 58 can undergo alkylation reactions as described for the
benzylimidazoles hereinabove.
Figure imgf000077_0001
Figure imgf000078_0001
Figure imgf000079_0001
Figure imgf000080_0001
Figure imgf000081_0001
Figure imgf000082_0001
wherein R' is R1 a or a protected precursor thereof
Figure imgf000083_0001
wherein R' is (R4)r-V- or a protected precursor thereol
Figure imgf000084_0001
Figure imgf000085_0001
wherein R'CH2- is R8 or a protected precursor thereof
Figure imgf000086_0001
Figure imgf000087_0001
Figure imgf000088_0001
Figure imgf000089_0001
Figure imgf000090_0001
Figure imgf000091_0001
Figure imgf000092_0001
Figure imgf000093_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, ser, ab1, 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 poly cystic kidney disease (D.L. Schaffner et al. American Journal of Pathology, 142:1051-1060 (1993) and B. Cowley, Jr. et al.FASEB Journal, 2:A3160 (1988)).
The instant compounds may also be useful for the treatment of fungal infections.
The 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 com 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 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 pharmacologically acceptable carriers, e.g., saline, at a pH level, e.g., 7.4. The solutions may be introduced into a patient's intramuscular blood-stream by local bolus injection.
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
quantitating the enzyme. Thus, potent inhibitor compounds of the instant invention may be used in an active site titration assay to determine the quantity of enzyme in the sample. A series of samples composed of aliquots of a tissue extract containing an unknown amount of 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. Purification by HPLC or column chromatography was utilized for each of the Examples 1-24 as set forth below. EXAMPLE 1
Preparation of 1-(t-Butoxycarbonyl)-cis-3-methoxycarbonyl-5-[N-(1- (4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl]piperidine
Step A: Preparation of Pyridine-3,5-dicarboxylic acid methyl ester hydrochloride
Pyridine-3,5-dicarboxylic acid (5.00 g, 29.9 mmol) was stirred in CH3OH (100 mL), cooled in an ice bath, and treated dropwise with thionyl chloride (17.45 mL, 239.2 mmol). After 0.5 hr, the reaction mixture was refluxed for 48 hrs. Concentration to dryness gave the title compound as an off-white solid.
Step B: Preparation of Piperidine-cis, trans-3,5-dicarboxylic acid methyl ester hydrochloride
Pyridine-3,5-dicarboxylic acid methyl ester hydrochloride (7.07 g, 30.5 mmol) was dissolved in acetic acid(75 mL), treated with platinum(IV) oxide (100 mg, 0.44 mmol) and shaken on a Parr apparatus at 50 psi overnight. The reaction mixture was filtered through celite and concentrated to dryness to give the title compound.
Step C: Preparation of 1-(t-Butoxycarbonyl)piperidine-cis and
trans-3,5-dicarboxylic acid methyl ester
Piperidine-3,5-dicarboxylic acid methyl ester hydrochloride (7.11 g, 29.9 mmol) was dissolved in THF (60 mL) and H2O (60 mL). Sodium bicarbonate (13.81 g, 0.164 mol) was added followed by di-tert-butyl dicarbonate (9.79 g, 44.9 mmol). The mixture was stirred at ambient temperature for 5 hrs. The THF was removed under reduced pressure, and the solution was extracted with CH2Cl2 (3×100 mL). The combined CH2Cl2 layers were washed with brine and dried (MgSO4). Filtration and concentration to dryness gave, after chromatography (silica gel, 10% ethyl acetate/hexane), racemic 1 -(t-Butoxycarbonyl)piperidine-cis-3,5-dicarboxylic acid methyl ester and racemic 1-(t-Butoxycarbonyl)piperidine-trans-3,5-dicarboxylic acid methyl ester.
Step D: Preparation of 1-(t-Butoxycarbonyl)-cis-3- methoxycarbonyl-piperidine-5-carboxylic acid
1-(t-Butoxycarbonyl)piperidine-cis-3,5-dicarboxylic acid methyl ester(0.761 g, 2.52 mmol) was dissolved in DME (6 mL) and H2O (6 mL) followed by addition of LiOH H2O (0.106 g, 2.52 mmol). The mixture was stirred at ambient temperature overnight. The DME was removed under reduced pressure, taken up in EtOAc and water, acidified to pH 3, and extracted with EtOAc (10X). The EtOAc layers were combined, dried (MgSO4), filtered, and concentrated to dryness to give the title compound after chromatography (silica gel, 1-3%
MeOH/CH2Cl2).
Step E: Preparation of 3-(4-cyanobenzyl) histamine
NY-Pivaloyloxymethyl-Nα-phthaloylhistamine (4.55 g, 12.8 mmol) was prepared as previously described (J. C. Emmett, F. H.
Holloway, and J. L. Turner, J. Chem. Soc, Perkin Trans. I, 1341, (1979)). α-Bromo-p-tolunitrile (3.77 g, 19.2 mmol) was dissolved in acetonitrile (70 mL). The solution was heated at 55°C for 4 h, cooled to room temperature, and filtered to remove the white solid. The acetonitrile (30 mL) was concentrated to 1/2 its volume under reduced pressure and the solution was heated at 55°C overnight. The solution was cooled and filtered to give a white solid. The volume of the filtrate was reduced to 10 mL, the solution was heated at 55°C for 1 hr, then cooled to room temperature, diluted with EtOAc (25 mL) and filtered to obtain additional white solid. The solids were combined, dried, and used without further purification.
1-Pivaloyloxymethyl-3-(4-cyanobenzyl)-4-(2-phthalimidoethyl)imidazolium bromide (6.13 g, 11.1 mmol) in methanol (100 mL) was saturated with ammonia gas while the temperature was maintained below 30°C. The solution was stirred for 1 hr, concentrated to dryness, and extracted with CH2Cl2 (3×200 mL), dried (MgSO4), concentrated, and chromatographed (silica gel, 10:90: 1 MeOH/CH2O2/NH4OH) to give 4-cyanobenzyl-Nα-phthaloylhistamine.
3-(4-Cyanobenzyl)-Nα-phthaloylhistamine (1.64 g, 4.61 mmol), and hydrazine (1.46 mL, 46.1 mmol) were dissolved in absolute EtOH (70 mL). The solution was concentrated after 1 hr and filtered to remove a white precipitate which was washed several times with EtOH. The filtrate was concentrated and the residue was chromatographed (silica gel, 10:90: 1 MeOH/CH2Cl2/NH4OH) to give the title compound.
Step F: Preparation of 1-(t-Butoxycarbonyl)-cis-3-methoxycarbonyl-5-[N-(1-(4-cyanobenzyl)-1H-imidazol-5- ylethyl)carbonyl]-piperidine
1-t-Butoxycarbonyl-cis-3-methoxycarbonyl-piperidine-5-carboxylic acid (1.45 g, 5.06 mmol), 3-(4-cyanobenzyl) histamine ( 1.14 g, 5.06 mmol), HOBT (0.72 g, 5.31 mmol), EDC (1.02 g, 5.31 mmol), Et3N (0.63 mL, 4.55 mmol) were dissolved in DMF (20 mL). The solution was stirred overnight, concentrated, and partitioned between EtOAc (300 mL) and sat NaHCO3 solution (200 mL). The organics were washed with brine, dried (MgSO4), filtered and concentration to dryness gave the title compound after chromatography (silica gel, 1-2% MeOH/CHCl2). 1H NMR (CDCI3); δ 7.64 (d, 2H, J=8 Hz), 7.52 (s, 1H), 7.14 (d, 2H, J=8 Hz), 6.90 (s, 1H), 6.12 (br s, 1H), 5.21 (s, 2H), 4.10-4.33 (m, 2H), 3.69 (s, 3H), 3.32-3.46 (m, 2H), 2.71-2.92 (m, 2H), 2.54-2.68 (m, 2H) 2.39-2.50 (m, 1H), 2.12-2.28 (m, 2H), 1.78-1.92 (m, 1H), 1.45 (s, 9H). FAB MS 496 (M+1)
Anal. calcd for C26H33N5O5•0.3 H2O: C, 62.34; H, 6.76; N, 13.98;
Found: C, 62.32; H, 6.61 ; N, 13.89.
Following the procedure of Steps D-F but substituting the 1-(t-butoxycarbonyl)piperidine-trans-3,5-dicarboxylic acid methyl ester prepared as described in Step C for the 1-(t-Butoxycarbonyl)piperidine-cis-3,5-dicarboxylic acid methyl ester utilized in Step D provided 1-(t- butoxycarbonyl)-trans-3-methoxy-carbonyl-5-[N-(1 -(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl]-piperidine.
EXAMPLE 2
Preparation of 1-Phenethyl-cis-3-methoxycarbonyl-5-[N-(4-cyanobenzyl-1-imidazol-5-ylethyl)carbonyl] piperidine
Step A: Preparation of cis-3-Methoxycarbonyl-5-[N-(1-(4- cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
1-(t-Butoxycarbonyl)-cis-3-methoxycarbonyl-5-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine (1.14 g, 2.30 mmol) was dissolved in CH2Cl2 (12 mL). Trifluoroacetic acid (6 mL) was added and the solution was stirred at ambient temperature for 3 h. The solution was concentrated to dryness to give the title
compound.
Step B: Preparation of 1-Phenethyl-cis-3-methoxycarbonyl-5-[N- (1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
cis-3-Methoxycarbonyl-5-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine (59.0 mg, 0.149 mmol) was dissolved in MeOH (2 mL). Phenylacetaldehyde (52.3 ul, 0.447 mmol) was added followed by sodium cyanoborohydride (28.1 mg, 0.447 mmol). The solution was stirred overnight at ambient temperature. The MeOH was removed under reduced pressure and chromatography (silica gel, 1-2% MeOH/CH2Cl2/NH4OH) gave the title compound as a white solid. 1H NMR (CDCI3); δ 7.62 (d, 2H, J=8 Hz), 7.50 (s, 1H), 7.08-7.37 (m, 7H), 6.88 (s, 1 H), 6.46 (br s, 1H), 5.22 (s, 2H), 3.64 (s, 3H), 3.27-3.19 (m, 1H), 3.03-3.20 (m, 1H), 2.86-3.00 (m, 1H),2.73- 2.85 (m, 3H), 2.44-2.70 (m, 5H), 2.26-2.41 (m, 2H), 1.98-2.10 (m, 2H), 1.80-1.95 (m, 1H). FAB MS 500 (M+1) Using the methods described in Example 2, but substituting the requisite aldehyde for phenylacetaldehyde in Step B, the following compounds were prepared: 1-(1-Naphthylmethyl)-cis-3-methoxycarbonyl-5-[N-(1-(4-cyanobenzy 1H-imidazol-5-ylethyl)carbamoyl] piperidine
FAB MS 536 (M + 1)
1-Benzyl-cis-3-methoxycarbonyl-5-[N-(1 -(4-cyanobenzyl)-1H-imidazol; 5-ylethyl)carbamoyl] piperidine
FAB MS 486 (M + 1)
1-Methyl-cis-3-methoxycarbonyl-5-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
FAB MS 410 (M + 1)
1-(2-Indanyl)-cis-3-methoxycarbonyl-5-[N-( 1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
FAB MS 512 (M + 1)
Anal. calcd for C30H33N5O3•0.15 H2O• 0.5 CHCl3:
C, 63.82; H, 5.94; N, 12.20;
Found: C, 63.83; H, 5.95; N, 12.16.
1-(2,2-Diphenylethyl)-cis-3-methoxycarbonyl-5-[N-(1-(4-cyanobenzy1)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
FAB MS 576 (M + 1)
Anal. calcd for C35H37N5O3• 0.9 H2O: C, 71.02; H, 6.61; N, 11.83;
Found: C, 71.08; H, 6.35; N, 1 1.71. Separation of the diastereomers of this compound on a Chiralcel OD HPLC column eluting with hexane/0.2% DEA: 1-propanol, 55:45 provided the following:
1-(2,2-Diphenylethyl)-cis-3(S)-methoxycarbonyl-5-(R)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine FAB MS 576 (M + 1)
Anal. calcd for C35H37N5O3• 0.9 H2O: C, 71.02; H, 6.61 ; N, 11.83;
Found: C, 71.08; H, 6.35; N, 11.71.
1-(2,2-Diphenylethyl)-cis-3-(R)-methoxycarbonyl-5-(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
FAB MS 576 (M + 1)
Anal. calcd for C35H37N5O3•0.9 H2O: C, 71.02; H, 6.61 ; N, 1 1.83;
Found: C, 71.08; H, 6.35; N, 11.71.
1-(3-Phenylpropyl)-cis-3-methoxycarbonyl-5-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
FAB MS 514 (M + 1)
Anal. calcd for C30H35N5O3•0.8 H2O: C, 68.24; H, 6.99; N, 13.26;
Found: C, 68.20; H, 6.69; N, 13.14.
1-(2-Methylpropyl)-cis-3-methoxycarbonyl-5-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
FAB MS 452 (M + 1)
Anal. calcd for C25H33N5O3• 0.75 H2O: C, 64.57; H, 7.48; N, 15.06;
Found: C, 64.61; H, 7.19; N, 14.68.
EXAMPLE 3
Preparation of 1-Phenethyl-cis-3-carboxyl-5-[N-(1-(4-cyanobenzyl)- 1H-imidazol-5-ylethyl)carbamoyl] piperidine
1-Phenethyl-cis-3-methoxycarbonyl-5-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyI)carbamoyl] piperidine (43.1 mg, 0.086 mmol) was dissolved in THF (4 mL) and H2O (1 mL). A IN solution of LiOH•H2O (86.2 uL, 0.086 mmol) was added and the solution was stirred overnight at ambient temperature. The solution was purified on a WATERS PrepPak column (0.1% TFA in CH3CN: 0.1% TFA in H2O, 5:95 to 95:5 gradient) to give the title compound. 1H NMR (CD3OD) δ 8.72 (s, 1H), 7.78 (d, 2H, J=8 Hz), 7.43 (d, 2H, J=8 Hz), 7.20-7.40 (m, 6H), 5.55 (s, 2H), 3.75 (d,1H, J=l 1 Hz), 3.58 (d, 1H, J=l l Hz), 3.30-3.46 (m, 3H), 2.83-3.16 (m, 6H), 2.77 (t, 2H, J=7 Hz), 2.30 (d, 1H, J=13 Hz), 1.65-1.80 (m, 1H), 1.37 (d, 1H, J=6 Hz). FAB MS 486 (M+1).
EXAMPLE 4
Preparation of 1-Phenethyl-cis-3-[N-( 1-morpholinyl)carbamoyl]-5-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl]piperidine
The lithium salt of 1-phenethyl-cis-3-carboxy-5-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine (55.2 mg, 0.112 mmol), morpholine (1 1.7 μL, 0.134 mmol), HOOBT (21.9 mg, 0.134 mmol), EDC (25.8 mg, 0.134 mmol), and Et3N (14.0 μL, 0.101 mmol) were dissolved in DMF (2 mL). The solution was stirred overnight at ambient temperature. The solution was concentrated under reduced pressure and the residue chromatographed (silica gel, 1-3% MeOH/CH2Cl2with NH4OH) to give the title compound. 1H NMR (CDCI3) δ 7.63 (d, 2H, J=8 Hz), 7.52 ( s, 1H), 7.06-7.33 (m, 7H), 6.90 (s, 1H), 5.89 (br s, 1H), 5.21 (s, 2H), 3.55-3.76 (m, 6H), 3.43-3.53 (m, 2H), 3.28-3.40 (m, 2H), 3.09 (d, 1H, J=1 1 Hz), 2.91 (d, 1H, J=1 1 Hz), 2.71-2.86 (m, 3H), 2.53-2.70 (m, 4H), 2.36-2.46 (m, 1H), 2.25 (t, 1H, J=11 Hz), 2.15 (t, 1H, J=11 Hz), 2.71-2.93 (m, 2H). FAB MS 555 (M+1)
Anal. calcd for C32H38N6O3•0.30 H2O•0.30 CHCl3:
C, 65.10; H, 6.58; N, 14.10;
Found C, 65.16; H, 6.59; N, 13.86.
Using the methods described in Examples 2 and 4, the following compounds were prepared:
1-Phenethyl-cis-3-[N-(benzyl)carbamoyl]-5-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
FAB MS 575 (M + 1) Anal. calcd for C35H38N6O2•0.2 H2O• 0.35 CH2Cl2:
C, 69.83; H, 6.48; N, 13.82;
Found: C, 69.82; H, 6.46; N, 13.53. 1-Phenethyl-cis-3-[N-(cyclopropyl)carbamoyl]-5-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
FAB MS 524 (M + 1)
Anal. calcd for C3 1H36N6O2•0.5 H2O• 0.25 EtOAc:
C, 69.17; H, 7.07; N, 15.12; Found: C, 69.22; H, 6.86; N, 15.12.
1-Phenethyl-cis-3-[N-(t-butyl)carbamoyl]-5-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
FAB MS 541 (M + 1)
Anal. calcd for C32H40N6O2•0.2 H2O• 0.25 CH2Cl2:
C, 68.49; H, 7.29; N, 14.86;
Found: C, 68.48; H, 7.27; N, 14.51.
1-(2,2-Diphenylethyl)-cis-3-[N-(t-butyl)carbamoyl]-5-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
FAB MS 617 (M + 1)
Anal. calcd for C38H44N6O2• 1.05 H2O: C, 71.80; H, 7.31 ; N, 13.22;
Found: C, 71.76; H, 7.34; N, 12.83. 1-(2,2-Diphenylethyl)-cis-3-[N-(1-morpholinyl)carbamoyl]-5-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
FAB MS 631 (M + 1)
Anal. calcd for C38H42N6O3•0.4 H2O• 0.4 EtOAc:
C, 70.65; H, 6.89; N, 12.48;
Found: C, 70.63; H, 6.63; N, 12.46.
Separation of the diastereomers of this compound on a Chiralcel OD HPLC column eluting with hexane/0.1% DEA: ethanol, 55:45 provided the following: 1-(2,2-Diphenylethyl)-cis-3-(R)-[N-(1-morpholinyl)carbamoyl]-5-(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine FAB MS 576 (M + 1)
Anal. calcd for C35H37N5O3•0.9 H2O: C, 71.02; H, 6.61 ; N, 1 1.83;
Found: C, 71.08; H, 6.35; N, 11.71.
1-(2,2-Diphenylethyl)-cis-3(S)-[N-(1-morpholinyl)carbamoyl]-5-(R)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine FAB MS 576 (M + 1)
Anal. calcd for C35H37N5O3•0.9 H2O: C, 71.02; H, 6.61 ; N, 11.83;
Found: C, 71.08; H, 6.35; N, 1 1.71.
EXAMPLE 5
Preparation of N-[1-Phenethyl-cis-5-(N'-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl) piperidine-3-carbonyl] methionine methyl ester
The lithium salt of 1-phenethyl-cis-3-carboxy-5-[N-(1 -(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine (90.0 mg, 0.183 mmol), methionine (43.8 mg, 0.219 mmol), HOOBT (35.8 mg, 0.219 mmol), EDC (42.1 mg, 0.219 mmol), and Et3N (60 uL, 0.430 mL) were dissolved in DMF (2 mL). The above solution was stirred overnight at ambient temperature. The solution was concentrated under reduced pressure and the residue chromatographed (silica gel, 1-2.5% MeOH/CH2Cl2 with NH4OH) to give the title compound. 1H NMR (CDCI3) δ 7.63(d, 2H, J=8 Hz), 7.51 (s, 1H), 7.10-7.33 (m, 7H), 6.90 (s, 1H), 6.50-6.63 (m, 1H), 6.1 1-6.36 (m, 1H), 5.21 (s, 2H), 4.65-4.75 (m, 1H), 3.75 (s, 3H),3.25-3.48 (m, 2H), 2.95-3.11 (m, 2H), 2.73-2.83 (m, 2H), 2.53-2.71 (m, 4H), 2.45-2.52 (m, 3H), 2.35-2.44 (m, 1H), 1.93-2.33 (m, 5H), 2.09 (s, 3H,), 1.67-1.85 (m, 1H). FAB MS
631(M+1)
Anal. calculated for C34H42N6O4S•0.30 CH2Cl2: C, 62.46; H, 6.52; N, 12.72;
Found C, 62.45; H, 6.53; N, 12.53.
EXAMPLE 6
Preparation of N-[1-Phenethyl-cis-5-(N'-(1-(4-cyanobenzyl-1-imidazol- 5-ylethyl)carbamoyl) piperidine-3-carbonyl] methionine
N-[1-Phenethyl-cis-5-(N'-(4-cyanobenzyl-1 -imidazol-5-ylethyl)carbamoyl) piperidine-3-carbonyl] methionine methyl ester (19 mg, 0.030 mmol) was dissolved in THF (2 mL) and H2O (1 mL). A IN solution of LiOH•H2O (30.1μL, 0.030 mmol) was added and the solution was stirred overnight at ambient temperature. The solution was purified on a RP HPLC VYDAC column (0.1% TFA in CH3CN: 0.1 % TFA in H2O, 5:95 to 95:5 gradient) and lyophilized to give the title compound as cis diastereomers.
Diastereomer A: 1H NMR (CD3OD) δ 8.99 (s, 1H), 7.80 (d, 2H, J=8 Hz), 7.51 (s, 1H), 7.47 (d, 2H, J=8 Hz), 7.20-7.37 (m, 5H), 5.61 (s, 2H), 4.26-4.57 (m, 1H), 3.60-3.75 (m, 2H), 3.44-3.55 (m, 1H), 3.30-3.43 (m, 3H), 2.90-3.15 (m, 6H), 2.75-2.85 (m, 2H), 2.48-2.70 (m, 2H), 2.09-2.28 (m, 2H), 2.09 (s, 3H), 1.90-2.08 (m,1H), 1.61-1.75 (m, 1H). FAB MS 617 (M+1)
Diastereomer B: 1H NMR (CD3OD) δ 9.02 (s, 1H), 7.81 (d, 2H, J=8 Hz), 7.53 (s, 1H), 7.51 (d, 2H, J=8 Hz), 7.22-7.39 (m, 5H), 5.62 (s, 2H), 4.29-4.59 (m, 1H), 3.76 (d, 1H, J=8 Hz), 3.68 (d,1H, J=10 Hz) 3.33- 3.52 (m, 4H), 2.95-3.20 (m, 6H), 2.79-2.87 (m, 2H), 2.50-2.68 (m, 2H), 2.05-2.13 (m, 2H), 2.09 (s, 3H), 1.93-2.08 (m,1H), 1.70- 1.82 (m, 1H). FAB MS = 617 (M+1)
EXAMPLE 7
Preparation of 1-(t-Butoxycarbonyl)-cis-3-methoxycarbonyl-5-[N-(1 - (4-cyanobenzyl)-1H-imidazol-5-yl)acetylamino] piperidine Step A: Preparation of 1-(t-Butoxycarbonyl)-cis-3- methoxycarbonyl-5-(benzyloxycarbonyl)amino piperidine 1-(t-Butoxycarbonyl)-cis-3-methoxycarbonyl-piperidine-5-carboxylic acid (1.87 g, 6.51 mmol), benzyl alcohol (1.68 mL, 16,3 mmol), diphenyl phosphorylazide (1.47 mL, 6.83 mmol), and Et3N (0.95 mL, 6.83 mmol) were disolved in toluene (50 mL). The solution was heated at 90°C for 4 h. The solution was diluted with EtOAc and was washed with Sat. NaHCO3 solution, water, and brine. The organics were dried (MgSO4), filtered, and concentrated to give the title compound without further purification.
Step B: Preparation of 1-(t-Butoxycarbonyl)-cis-3-methoxycarbonyl-5-amino piperidine
1-(t-Butoxycarbonyl)-cis-3-methoxycarbonyl-5-(benzyloxycarbonyl)amino piperidine (2.55 g, 6.51 mmol) was dissolved in EtOAc (75 mL), treated with Pd/C (510 mg) and shaken on a Parr apparatus at 45 psi overnight. The reaction mixture was filtered through celite, concentrated, and chromatographed (silica gel, 2%
MeOH/CH2Cl2 with NH4OH) to give the title compound.
Step C: Preparation of 1H-Imidazole-4- acetic acid methyl ester hydrochloride
A solution of 1H-imidazole-4-acetic acid hydrochloride
(4.00g, 24.6 mmol) in methanol (100 ml) was saturated with gaseous hydrogen chloride. The resulting solution was allowed to stand at room temperature (RT) for 18hr. The solvent was evaporated in vacuo to afford the title compound as a white solid.
1H NMR(CDCl3, 400 MHz) δ 8.85(1H, s),7.45(1H, s), 3.89(2H, s) and
3.75(3H, s) ppm.
Step D: Preparation of 1-(Triphenylmethyl)-1H-imidazol-4-ylacetic acid methyl ester
To a solution of 1H-imidazole-4- acetic acid methyl ester hydrochloride (24.85g, 0.141mol) in dimethyl formamide (DMF) (115ml) was added triethylamine (57.2 ml, 0.412mol) and
triphenylmethyl bromide(55.3g, 0.171mol) and the suspension was stirred for 24hr. After this time, the reaction mixture was diluted with ethyl acetate (EtOAc) (1 1) and water (350 ml). The organic phase was washed with sat. aq. NaHCO3 (350 ml), dried (Na2SO4) and
evaporated in vacuo. The residue was purified by flash chromatography (SiO2, 0-100% ethyl acetate in hexanes; gradient elution) to provide the title compound as a white solid.
1H NMR (CDCI3, 400 MHz) δ 7.35(1H, s), 7.31(9H, m), 7.22(6H, m), 6.76(1H, s), 3.68(3H, s) and 3.60(2H, s) ppm.
Step E: Preparation of [1-(4-Cyanobenzyl)-1H-imidazol-5-yl]acetic acid methyl ester
To a solution of 1-(triphenylmethyl)-1H-imidazol-4-ylacetic acid methyl ester (8.00g, 20.9mmol) in acetonitrile (70 ml) was added bromo-p-toluonitrile (4.10g, 20.92 mmol) and heated at 55°C for 3 hr. After this time, the reaction was cooled to room temperature and the resulting imidazolium salt (white precipitate) was collected by filtration. The filtrate was heated at 55°C for 18hr. The reaction mixture was cooled to room temperature and evaporated in vacuo. To the residue was added EtOAc (70 ml) and the resulting white precipitate collected by filtration. The precipitated imidazolium salts were combined, suspended in methanol (100 ml) and heated to reflux for 30min. After this time, the solvent was removed in vacuo, the resulting residue was suspended in EtOAc (75ml) and the solid isolated by filtration and washed (EtOAc). The solid was treated with sat aq
NaHCO3 (300ml) and CH2Cl2 (300ml) and stirred at room
temperature for 2 hr. The organic layer was separated, dried (MgSO4) and evaporated in vacuo to afford the title compound as a white solid : 1HNMR(CDCl3, 400 MHz) δ 7.65(1H, d, J=8Hz), 7.53(1H, s), 7.15(1H, d, J=8Hz), 7.04(1H, s), 5.24(2H, s), 3.62(3H, s) and 3.45(2H, s) ppm.
Step F: Preparation of [1-(4-cyanobenzyl)-1H-imidazol-5-yl]acetic acid A solution of [1-(4-cyanobenzyl)-1H-imidazol-5-yl]acetic acid methyl ester (4.44g, 17.4mmol ) in THF (100ml) and 1 M lithium hydroxide (17.4 ml, 17.4 mmol) was stirred at RT for 18 hr. 1 M HCl (17.4 ml) was added and the THF was removed by evaporation in vacuo. The aqueous solution was lyophilized to afford the title
compound containing lithium chloride as a white solid.
1H NMR(CD3OD, 400 MHz) δ 8.22(1H, s), 7.74(1H, d, J=8.4Hz), 7.36(1H, d, J=8.4Hz), 7.15(1H, s), 5.43(2H, s) and 3.49(2H, s) ppm.
Step G: Preparation of 1-(t-Butoxycarbonyl)-cis-3-methoxycarbonyl-5-[N-(1-(4-cyanobenzyl)-1H-imidazol-5- yl)acetylamino]-piperidine
1-(t-Butoxycarbonyl)-cis-3-methoxycarbonyl-5-amino piperidine (134 mg, 0.520 mmol), [1-(4-cyanobenzyl)-1H-imidazol-5-yl]acetic acid (147 mg, 0.520 mmol), HOBT (73,8 mg, 0.546 mmol), EDC (104 mg, 0.546 mmol), and Et3N (65.2 uL, 0.468 mmol) were dissolved in DMF (4mL) and stirred at ambient temperature overnight. The solution was concentrated under reduced pressure and the residue was chromatographed ( silica gel, 0.5-2% MeOH/CH2Cl2 with NH4OH) to give the title compound. 1H NMR (CDCI3) d 7.64 (d, 2H, J=8 Hz), 7.52 (s, 1H), 7.19 (d, 2H, J=8 Hz), 6.90-7.05 (m, 1H), 6.98 (s, 1H), 5.22-5.35 (m, 2H), 3.78-3.91 (m, 2H), 3.69 (s, 3H), 3,57-3.64 (m, 1H), 3.45-3.56 (m, 1H), 3.34 (s, 2H), 3.05-3.30 (m, 1H), 2.55-2.68 (m, 1H), 2.05-2.13 (m, 1H), 1.55-1.70 (m, 1H), 1.42 (s, 9H). FAB MS 482 (M+1)
Anal. calculated for C25H3 1N5O5•0.90 H2O:
C, 60.32; H, 6.64; N, 14.07;
Found C, 60.38; H, 6.36; N, 13.78.
EXAMPLE 8
Preparation of 1-Phenethyl-cis-3-methoxycarbonyl-5-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-yl)acetylamino] piperedine Step A: Preparation of cis-3-methoxycarbonyl-5-[N-(1-(4- cyanobenzyl)-1H-imidazol-5-yl)acetylamino] piperidine 1-(t-Butoxycarbonyl)-3-methoxycarbonyl-5-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-yl)acetylamino] piperidine (44.6 mg, 0.093 mmol) was dissolved in CH2Cl2 (6 mL). Trifluoroacetic acid (3 mL) was added and the solution was stirred at ambient temperature for 4 h. The solution was concentrated under reduced pressure to give the title compound without futher purification.
Step B: Preparation of 1-Phenethyl-cis-3-methoxycarbonyl-5-[N- (1-(4-cyanobenzyl)-1H-imidazol-5-yl)acetylamino] piperidine
cis-3-Methoxycarbonyl-5-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-yl)acetylamino] piperidine (35.3 mg, 0.093 mmol), benzaldehyde (32.5 uL, 0.278 mmol), and sodium cyanoborohydride (17.5 mg, 0.278 mmol) was dissolved in MeOH (2 mL) and stirred overnight at ambient temperature. The solution was concentrated under reduced pressure and chromatographed (silica gel, 0.5-2%
MeOH/CH2Cl2 with NH4OH) to give the title compound. 1 H NMR (CDCI3) δ 7.62 (d, 2H, J=8 Hz), 7.52 (s, 1H), 7.11-7.31 (m, 7H), 6.99 (s, 1H), 6.25 (br s, 1H), 5.24 (s, 2H), 3.86-3.96 (m, 1H), 3.65 (s, 3H), 3.29 (s, 2H), 2.48-2.83 (m, 8H), 2.13-2.24 (m, 1H), 1.83-1.95 (m, 1H), 1.50-1.65 (m, 1H). FAB MS 486 (M+1)
Anal. calculated for C28H31N5O3• 0.30 H2O•0.25 CHCl3:
C, 65.15; H, 6.16; N, 13.45;
Found C, 65.18; H, 6.15; N, 13.46.
Using the methods described in Examples 7 and 8, but substituting 1-(t-butoxycarbonyl-piperidine-3(S)-carboxylic acid for 1-(t-butoxycarbonyl)-cis-3-methoxycarbonyl-piperidine-5- carboxylic acid in Ex. 7, Step A, and diphenylacetaldehyde for phenylacetaldehyde in Ex. 8, Step B, the following compound was prepared: 1-(2,2-Diphenylethyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazoI-5-ylacetyl)amino] piperidine
Anal. calculated for C32H33N5O• 0.30 H2O:
C, 75.50; H, 6.65; N, 13.62;
Found C, 75.51 ; H, 6.79; N, 13.76.
FAB MS (M+1) 504
Using the methods described for Examples 7 and 8, but substituting 1H-imidazole-4-propionic acid for 1H-imidazole-4-acetic acid in Ex. 7, Step C, the following compound was prepared:
1-(2,2-Diphenylethyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylpropionyl)amino] piperidine
Anal. calculated for C33H35N5O• 0.55 H2O:
C, 75.13; H, 6.90; N, 13.72;
Found C, 75.15; H, 6.89; N, 13.40.
FAB MS (M+1) 518
Using the methods described for Examples 7 and 8, but substituting 1H-imidazole-4-carboxylic acid for 1H-imidazole-4-acetic acid in Ex. 7, Step C, the following compound was prepared:
1-(2,2-Diphenylethyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylcarbonyPamino] piperidine
FAB MS (M+1) 490
EXAMPLE 9 Preparation of 1-(Diphenylacetyl)-cis-3-methoxycarbonyl-5-[N-(1 -(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
CM-3-Methoxycarbonyl-5-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine (86.8 mg, 0.219 mmol) (EXAMPLE 2, Step A) diphenyl acetic acid (302 mg,1.42 mmol), HOOBT (54.3 mg, 0.328 mmol), EDC (63 mg, 0.328 mmol), and Et3N (392 uL, 5.34 mmol) were dissolved in DMF (4 mL) and stirred at ambient temperature for 5 days. The solution was concentrated, the residue was taken up in EtOAc, washed with sat. NaHCO3
solution,water, and brine. The organics were dried (MgSO4), concentrated, and chromatographed (silica gel, 0.5-2% MeOH/CH2Cl2 with NH4OH) to give the title compound. FAB MS = 590 (M+l) Anal. calculated for C35H35N5O4• 0.35 CHCl3•0.20 H2O:
C, 66.86; H, 5.67; N, 11.03; Found C, 66.86; H, 5.67; N, 1 1.01.
Using the methods described above but substituting 3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine (Example 1 1, Step E) as the starting material and the requisite acid, the following compounds were prepared:
1-(Phenylacetyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
FAB MS (M+1) 456
1-(Diphenylacetyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
Anal. calculated for C33H33N5O2• 0.45 CH2Cl2•0.10 H2O:
C, 70.28; H, 6.01 ; N, 12.25; Found C, 70.24; H, 5.91; N, 12.09.
1-(3-Chlorobenzoyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
Anal. calculated for C26H26N5O2Cl: C, 64.03; H, 5.64; N, 14.36;
Found C, 64.09; H, 5.39; N, 14.12.
FAB MS (M+1) 476 EXAMPLE 10
Preparation of 1-(2,2-Diphenylethyl)-3-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
Step A: Preparation of 1 -(2,2-Diphenylethyl)-3-carboxy piperidine
Nipecotic acid (300 mg, 2.38 mmol), diphenylacetaldehyde (1.26 mL, 7.13 mmol), sodium cyanoborohydride (448 mg, 7.13 mmol), and HOAc (204 uL, 3.57 mmol) were dissolved in MeOH (20 mL) and stirred at ambient temperature overnight. The solution was concentrated under reduced pressure, take up in ether and 1N NaOH, extract with ether (3X), acidify the aqueous layer with 1N HCl, and extract with EtOAc (3X). The EtOAc layers were dried (MgSO4) and concentrated to give the title compound without further purification.
Step B: Preparation of 1 -(2,2-Diphenylethyl)-3-[N-(1-(4- cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine 1-(2,2-Diphenylethyl)-3-carboxy piperidine(472 mg, 1,52 mmol),3-(4-cyanobenzyl) histamine (456 mg, 1.52 mmol) (EXAMPLE 1, Step E) HOBT (216 mg, 1.60 mmol), EDC (307 mg, 1.60 mmol), and Et3N (637 uL, 4.57 mmol) were dissolved in DMF (10 mL) and was stirred overnight at ambient temperature. The solution was concentrated under reduced pressure and chromatographed (silica gel, 0.5-2% MeOH/CH2Cl2 with NH4OH) to give the title compound 1H NMR (CDCI3) δ 7.96 (br s, 1H), 7.60 d, 2H, J=8 Hz), 7.46 (s, 1H), 7.09-7.37 (m, 12H), 6,74 ( s, 1H), 5.20 (s, 2H), 4.26 ( t, 1H, J=8 Hz), 3.05-3.17 (m, 2H), 2.93-3.04 (m,1H), 2.78-2.91 (m,2H), 2.49-2,61 (m 1H), 22.40-2.47 (m, 1H), 2.15-2.30 (m, 2H), 1.95-2.14 (m, 2H), 1.86 (d, 1H, J=12 Hz), 1.29-1.55 (m, 3H). FAB MS 518 (M+1)
Anal. calculated for C33H35N5O1• 0.35 CHCl3• 0.10 H2O:
C, 71.37; H, 6.38; N, 12.48; Found C, 71.41 ; H, 6.32; N, 12.36. EXAMPLE 11
Preparation of 1-(2-(3-Chlorophenyl)-2-phenylethyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
Step A: Preparation of S-(-)-Ethyl nipecotate
Racemic ethyl nipecotate (122.5 g, 0.78 mol) was resolved with D-tartaric acid (117 g, 0.78 mol) in 95% EtOH (611 mL)
following the procedure described by P. Magnus et al. (J. Org. Chem. 1991, 56, 1166-1170) to give S-(-)-ethyl nipecotate.
Step B: Preparation of Ethyl 1-(t-butoxycarbonyl)piperidine-3(S)- carboxylate
S-(-)-Ethyl nipecotate (20.0 g, 0.127 mol) was dissolved in THF (250 mL)- H2O (250 mL) at ambient temperature and treated with NaHCO3 (32.0 g, 0.381 mol) and di-tert-butyl-dicarbonate (43.8 mL, 0.190 mol). After stirring for 16 h, the reaction mixture was
concentrated to remove THF and extracted with EtOAc (2 × 200 mL). The organics were combined, washed with brine, dried (MgSO4), filtered and concentrated to give the title compound.
Step C: Preparation of 1-(tert-Butoxycarbonyl)piperidine-3(S)- carboxylic acid
Ethyl 1-(t-butoxycarbonyl)piperidine-3(S)-carboxylate (35.9 g, 0.099 mol) was dissolved in abs EtOH (70 mL), treated with 0.5N NaOH (418 mL, 0.209 mol), and heated at reflux for 0.75 h. The reaction mixture was cooled and extracted with EtOAc. The aqueous basic layer was cooled with ice, carefully acidified (pH 3) with 3N HCl and extracted with CH2Cl2 (3 × 100 mL). The organics were combined, dried (MgSO4), filtered, and concentrated to give the title compound.
Step D: Preparation of 1-(tert-Butyloxycarbonyl)-3(S)-[N- 1- (4- cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine 1-(tert-Butoxycarbonyl)piperidine-3(S)-carboxylic acid (5.06 g, 0.022 mol) and 3-(4-cyanobenzyl)histamine (Example 1 , Step E) (6.6 g, 0.022 mol) were dissolved in DMF (30 mL) at ambient
temperature and treated with EDC ( 5.07 g, 0.026 mol), HOBT ( 3.58 g, 0.26 mol), and N-methylmorpholine (12.12 mL, 0.11 mol). After stirring for 18 hr, the reaction mixture was partitioned between EtOAc(500 mL)-aq satd NaHCO3 soln, the organic layer separated, washed with satd
NaHCO3 soln, H2O, brine, dried (MgSO4), filtered and concentrated to give the title compound which was used without further purification.
Step E: Preparation of 3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazole-5- ethyl)carbamoyl] piperidine dihydrochloride
1-(tert-Butyloxycarbonyl)-3(S)-[N-1- (4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine (9.25 g, 0.021 mol) was dissolved in EtOAc (500 mL) with stirring at 0°C. in an ice-water bath. HCl gas was bubbled through the solution for 5 min., the flask
stoppered, and the solution stirred for 1 hr. The solution was purged with N2 then concentrated to dryness to give the title compound. 1H
NMR (CD3OD); δ 9.02 (s, 1H), 7.80 (d, 2H, J= 8 Hz), 7.52 (s, 1H), 7.48 (d, 2 H, J = 8 Hz),5.62 (s, 2H), 3.42 (td, 2H, J = 3,7 Hz), 3.00-3.26 (m, 4H), 2.80 (t, 2H, J = 7 Hz), 2.67-2.78 (m, 1H), 1.65 - 2.0 (m, 4H).
FAB MS 338 (M+1).
Step F: Preparation of 2-(3-Chlorophenyl)-2-phenyl oxirane
A 250 mL-round bottom flask was charged with NaH (60% dispersion in mineral oil) (1.92 g, 0.048 mol), washed with petroleum ether, then treated with dry DMSO (40 mL) under N2. To this reaction mixture was added trimethylsulfoxonium iodide (10.56 g, 0.048 mol) through a solid addition funnel over 15 min. After stirring for 0.5 hr, a solution of 3-chlorobenzophenone (8.66 g, 0.04 mol) in DMSO (15 mL) was added dropwise, and the mixture was heated at 55°C. for 2 hr. The mixture was added to ice-water, extracted with ether (3 × 100 mL), the organics combined, dried (MgSO4), filtered, and concentrated to give the title compound. Step G: Preparation of 2-(3-Chlorophenyl)-2-phenyl
carboxaldehyde
2-(3-Chlorophenyl)-2-phenyl oxirane (10.19 g, 0.04 mol) was dissolved in dry benzene (250 mL), treated with BF3 . etherate (0.2 mL) and stirred at ambient temperature for 1hr. The benzene was extracted with H2O until the aqueous layer was no longer acidic, then concentrated to dryness to give the title compound after
chromatography (5% EtOAc/hexane on SiO2). 1H NMR (CDCl3) δ 9.95 (s, 1H), 7.1 - 7.5 (m, 9H), 4.85 (s, 1H).
Step H: Preparation of 1-(2-(3-Chlorophenyl)-2-phenylethyl)-3- (S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5- ylethyl)carbamoyl] piperidine
3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5- ylethyl)carbamoyl] piperidine dihydrochloride (0.256 g, 0.623 mmol) was dissolved in MeOH (10 mL), the pH adjusted to 5 with Et3N, and 2-(3-chlorophenyl)-2-phenylcarboxaldehyde (0.427 g, 1.87 mmol) and NaCNBH3 (0.078 g, 1.25 mmol) were added. After stirring for 18 hr at ambient temperature, the reaction mixture was partitioned between EtOAc and satd NaHCO3 soln, the organic layer separated, washed with H2O, brine, dried (MgSO4), filtered and concentrated to dryness to give the title compound after chromatography on SiO2 eluting with 3% MeOH/ CH2Cl2 w/ NH4OH. 1H NMR (CD3OD); δ 7.88 (br s, 1H), 7.60 (d, 2H, J= 8 Hz), 7.47 (s, 1H), 7.07 - 7.36 (m, 11H), 6.76 (d, 1H, J = 2.7 Hz), 5.21 (s, 2H), 4.24 (td, 1H, J= 2, 8 Hz), 2.80-3.13 (m, 5H), 2.50 - 2.65 (m, 1H), 2.44 (s, 1H), 2.0 - 2.3 (m, 4H), 1.86 (d, 1H, J = 12 Hz), 1.3 - 1.55 (m, 3H). FAB MS 552 (M+1).
Anal. calculated for C33H34N5OCI• 0.40 H2O:
C, 70.87; H, 6.27; N, 12.52;
Found C, 70.84; H, 6.31; N, 12.39.
Using the methods described above, but substituting the requisite ketone in Step F, the following compounds were prepared: 1-(Dibenzylsuberylmethyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
Anal. calculated for C35H37N5O• 0.45 H2O:
C, 76.18; H, 6.92; N, 12.69;
Found C, 76.15; H, 6.84; N, 12.49.
1-(2-(3-Methylphenyl)-2-phenylethyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
Anal. calculated for C34H37N5O• 0.40 H2O:
C, 75.78; H, 7.07; N, 13.00;
Found C, 75.81; H, 7.01; N, 13.20
1-(2-(3-Trifluoromethylphenyl)-2-phenylethyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
Anal. calculated for C34H34N5OF3• 0.20 H2O:
C, 69.30; H, 5.88; N, 1 1.88;
Found C, 69.32; H, 5.84; N, 12.04. 1-(2-(2-Chlorophenyl)-2-phenylethyl)-3(S)-[N-(1 -(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
Anal. calculated for C33H34N5OCI• 0.50 H2O:
C, 70.64; H, 6.29; N, 12.48;
Found C, 70.67; H, 6.16; N, 12.50.
1-(2-(4-Chlorophenyl)-2-phenylethyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
Anal. calculated for C33H34N5OCI: C, 70.98; H, 6.26; N, 12.54;
Found C, 71.02; H, 6.22; N, 12.40.
1-(2-(3-Aminomethylphenyl)-2-phenylethyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
Anal. calculated for C34H38N6O• 4.25 CF3CO2H• 0.45 H2O:
C, 49.11 ; H, 4.18; N, 8.09; Found C, 49.11; H, 4.16; N, 8.15.
Using the methods described above, but substituting commercially available aldehydes for 2-(3-chlorophenyl)-2-phenylcarboxaldehyde in Step H, the following compounds were prepared:
1-(2-Phenethyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
FAB MS (M+1) 442
1-(2-Phenethyl)-3-(R)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
Anal. calculated for C27H31N5O• 2.0 HCl• 1.60 H2O:
C, 59.69; H, 6.72; N, 12.89; Found C, 59.72; H, 7.18; N, 11.91.
FAB MS (M+1) 442
1-(3-Phenylpropyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
Anal. calculated for C28H33N5O• 2.75 CF3CO2H• 0.75 H2O:
C, 51.14; H, 4.80; N, 8.95;
Found C, 51.42; H, 4.78; N, 9.00.
FAB MS (M+1) 456 1-(2-Benzyl)-3-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
Anal. calculated for C26H29N5O• 2.80 CF3CO2H• 0.70 H2O:
C, 49.98; H, 4.41; N, 9.22;
Found C, 49.98; H, 4.39; N, 9.67.
FAB MS (M+1) 428
1-(2-Chlorobenzyl)-3-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
Anal. calculated for C26H28N5OCI• 3.25 CF3CO2H• 0.50 H2O: C, 46.38; H, 3.86; N, 8.32;
Found C, 46.39; H, 3.82; N, 8.54.
FAB MS (M+1) 462 1-(3-Chlorobenzyl)-3-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
Anal. calculated for C26H28N5OCI• 2.75 CF3CO2H• 0.40 H2O:
C, 48.33; H, 4.06; N, 8.95; Found C, 48.30; H, 4.06; N, 9.16. FAB MS (M+1) 462
1-(3-Chlorobenzyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
Anal. calculated for C26H28N5OCI• 0.45 H2O:
C, 66.43; H, 6.20; N, 14.90;
Found C, 66.43; H, 6.07; N, 14.97.
1-(2,2-Diphenyl-2-hydroxyethyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
Anal. calculated for C33H35N5O2• 0.25 H2O:
C, 73.65; H, 6.65; N, 13.01 ; Found C, 73.69; H, 6.79; N, 12.84. FAB MS (M+1) 534 1-(3-Methoxybenzyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
FAB MS (M+1) 458
1-(3,5-Dichlorobenzyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
Anal. calculated for C26H27N5OCl2• 0.40 H2O:
C, 62.01; H, 5.56; N, 13.91; Found C, 61.98; H, 5.55; N, 13.57. 1-(3-Trifluoromethoxybenzyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
Anal. calculated for C27H28N5O2F3• 2.0 HCl• 1.85 H2O:
C, 52.49; H, 5.50; N, 11.34; Found C, 52.50; H, 5.72; N, 11.34.
1-(2,5-Dimethylbenzyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
Anal. calculated for C28H338N5O• 0.40 H2O:
C, 72.67; H, 7.36; N, 15.13;
Found C, 72.67; H, 7.27; N, 14.77.
1-(3-Trifluoromethylbenzyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
Anal. calculated for C27H28N5OF3• 0.15 H2O:
C, 65.09; H, 5.73; N, 14.06;
Found C, 65.14; H, 5.83; N, 14.01.
1-(3-Bromobenzyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
Anal. calculated for C26H28N5OBr• 2.0 HCl• 1.0 H2O:
C, 52.28; H, 5.40; N, 11.72;
Found C, 52.33; H, 5.51; N, 11.26. 1-(3-Methylbenzyl)-3(S)-[N-(1-(4-cyanobenzyl)-1-H-imidazol-5-ylethyl)carbamoyl] piperidine
Anal. calculated for C27H31N5O• 2.0 HCl• 1.30 H2O:
C, 60.54; H, 6.70; N, 13.07;
Found C, 60.58; H, 6.68; N, 12.22.
1-Isobutyl-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
Anal. calculated for C23H31N5O• 2.3 HCl• 0.95 H2O:
C, 55.87; H, 7.17; N, 14.16; Found C, 55.91; H, 7.38; N, 14.01.
FAB MS (M+1) 394
1-(2-Methyl-2-phenylethyl)-3-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
Anal. calculated for C34H37N5O• 0.40 H2O:
C, 75.78; H, 7.07; N, 13.00;
Found C, 75.81; H, 7.01; N, 13.20.
FAB MS (M+1) 532
Using the methods described above, but substituting 1-morpholinyl-α-phenylacetaldehyde or 1-piperidinyl-α-phenylacetaldehyde (prepared following the procedure described by L. Duhamel, P. Duhamel, P. Siret, Bull Soc. Chim. Fr., [7-8], 2460-2466 (1973)) in Step H, the following compounds were prepared:
1-(2-(1-Morpholinyl)-2-phenylethyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl ] piperidine-Diastereomer A
Anal. calculated for C3 1H38N6O2• 0.80 H2O• 0.25 EtOAc:
C, 68.25; H, 7.45; N, 14.92;
Found C, 68.27; H, 7.13; N, 14.92.
1-(2-(1-Morpholinyl)-2-phenylethyl)-3(S)-[N-(1-(4-cyanobenzyl)- 1H-imidazol-5-ylethyl)carbamoyl] piperidine-Diastereomer B
Anal. calculated for C3 1H38N6O2• 0.65 H2O:
C, 69.16; H, 7.36; N, 15.61 ;
Found C, 69.15; H, 7.31 ; N, 15.48.
1-(2-(1-Piperidinyl)-2-phenylethyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
FAB MS (M+1) 525 Using the methods described in Examples 11 and 1 , but substituting 2-methylhistamine for the starting material in Ex.l, Step E, the following compound was prepared:
1-(2,2-Diphenylethyl)-3(S)-[N-(1-(4-cyanobenzyl)-2-methyl-1H-imidazol-5-ylethyl)carbamoyl] piperidine
Anal. calculated for C34H37N5O• 0.45 H2O:
C, 75.65; H, 7.08; N, 12.97;
Found C, 75.65; H, 6.93; N, 12.68.
Using the methods described in Examples 11 and 1 , but substituting 4-methoxybenzyl bromide for 4-cyanobenzyl bromide in Ex.l, Step E, the following compound was prepared: 1-(2,2-Diphenylethyl)-3(S)-[N-(1-(4-methoxybenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
Anal. calculated for C33H38N4O2• 2.0 HCl• 1.40 H2O:
C, 63.84; H, 6.95; N, 9.02;
Found C, 63.91 ; H, 7.33; N, 9.45.
EXAMPLE 12
Preparation of 1-(Diphenylmethyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
3(S)-[N-1-(4-Cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine dihydrochloride (Example 11, Step E)
(0.203 g, 0.494 mmol) was dissolved in DMF (10 mL), treated with
K2CO3 (0.503 g, 1.976 mmol) and bromodiphenylmethane (0.134 g, 0.543 mmol), and stirred for 48 hr at ambient temperature. The reaction mixture was concentrated, dissolved in EtOAc, washed with aq satd NaHCO3 soln, H2O, brine, and dried (MgSO4), filtered and concentrated to give the title compound after SiO2 chromatography eluting with 0-3% MeOH: CH2Cl2 with NH4OH. Anal. calculated for C32H33N5O• 0.75 H2O:
C, 74.31 ; H, 6.72; N, 13.54; Found C, 74.37; H, 6.48; N, 13.37. FAB MS (M+1) 504.
Using the method described above, but substituting the requisite bromide or mesylate for bromodiphenylmethane, the following compounds were prepared:
1-(3-Methoxyphenethyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
Anal. calculated for C28H33N5O2• 2 HCl• 1.95 H2O:
C, 58.02; H, 6.76; N, 12.08; Found C, 58.01 ; H, 7.03; N, 1 1.99.
FAB MS (M+1) 472.
1-(1-Naphthylethyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
Anal. calculated for C31H33N5O• 0.90 H2O:
C, 73.32; H, 6.91 ; N, 13.79; Found C, 73.32; H, 7.04; N, 13.46.
1-(3-Chlorophenethyl)-3(S)-[N-1-(4- cyanobenzy)1-1H-imidazol-5-ylethyl)carbamoyl] piperidine
Anal. calculated for C27H30N5OCI• 2 HCl• 1.65 H2O:
C, 56.04; H, 6.15; N, 12.10; Found C, 56.09; H, 6.43; N, 11.77. 1-(α-Methylbenzyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
Anal. calculated for C27H3 1N5O• 0.45 H2O:
C, 72.12; H, 7.15; N, 15.57; Found C, 71.80; H, 6.81 ; N, 15.96. FAB MS (M+1) 442.
Using the methods described in Examples 12 and 1, but substituting 2-methylhistamine for the starting material in Ex. l, Step E, the following compound was prepared:
1-(2-Diphenylmethyl)-3(S)-[N-(1-(4-cyanobenzyl)-2-methyl-1H-imidazol-5-ylethyl)carbamoyl] piperidine
Anal. calculated for C33H35N5O• 0.40 EtOAc :
C, 75.11; H, 6.96; N, 12.66;
Found C, 75.15; H, 7.24; N, 12.60.
FAB MS (M+1) 518.
EXAMPLE 13
Preparation of 1-(α-Toluenesulfonyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
3(S)-[N-1-(4-Cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine dihydrochloride (Example 11 , Step E) (0.210 g, 0.512 mmol) was dissolved in CH2Cl2 (6 mL), treated with Et3N (0.285 mL, 2.046 mmol) and α-toluenesulfonyl chloride (0.195 g, 1.023 mmol) and stirred at ambient temperature for 2 hr. The reaction mixture was concentrated to dryness, partitioned between EtOAc and satd NaHCO3 soln, the organic layer separated, washed with brine, and dried (MgSO4). Filtration and concentration to dryness gave the title compound after purification on a Waters Prep Pak eluting with
0.1 %TFA/H2O: 0.1 %TFA/CH3CN, 95:5 to 5:95. 1H NMR (CD3OD); δ 8.80 (s, 1H), 8.02 - 8.12 (m, 1H), 7.78 (d, 2H, J= 8 Hz), 7.34 - 7.5 (m, 7H), 5.54 (s, 2H), 4.32 (s, 2H), 3.2-3.56 (m, 4H), 2.65 - 2.95 (m, 4H), 2.25-2.4 (m, 1H), 1.4 - 1.8 (m, 4H). FAB MS 492 (M+1).
Using this procedure, but substituting the sulfonyl chloride, the following compounds were prepared: 1 -(Benzenesulfonyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
FAB MS (M+1) 478.
1-(1-Naphthylenesulfonyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
Anal. calculated for C29H29N5O3S• 1.50 CF3CO2H• 0.10 H2O:
C, 54.87; H, 4.42; N, 10.00;
Found C, 54.84; H, 4.31 ; N, 10.19.
1-(3-Chlorobenzenesulfonyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
Anal. calculated for C25H26N5O3SCl• 0.55 H2O:
C, 57.53; H, 5.23; N, 13.42;
Found C, 57.51 ; H, 5.20; N, 13.28.
1-(3,5-Dichlorobenzenesulfonyl)-3(S)-[N-(1 -(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
Anal. calculated for C25H25N5O3SCl• 0.30 H2O:
C, 54.41 ; H, 4.68; N, 12.69;
Found C, 54.43; H, 4.82; N, 12.49.
1-(α-Toluenesulfonyl)-3-(R)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
Anal. calculated for C26H29N5O3S• 0.35 H2O:
C, 62.72; H, 6.01 ; N, 14.07;
Found C, 62.73; H, 5.85; N, 13.84. 1-(α-Toluenesulfonyl)-cis-3-methoxycarbonyl-5-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
FAB MS (M+1) 550 1-(Methanesulfonyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
Anal. calculated for C20H25N5O3S• 0.25 CH2Cl2:
C, 55.68; H, 5.89; N, 16.04;
Found C, 56.04; H, 5.89; N, 15.70.
FAB MS (M+1 ) 416
EXAMPLE 14 Preparation of 1-(Diphenylcarbamoyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine dihydrochloride (Example 11, Step E) (0.346 g, 0.843mmol) was dissolved in CH2Cl2 (10 mL), treated with Et3N (0.587 mL, 4.2 mmol) and diphenylcarbamoyl chloride (0.390 g, 1.686 mmol) and stirred at ambient temperature for 1 hr. The reaction mixture was concentrated to dryness, partitioned between EtOAc and satd NaHCO3 soln, the organic layer separated, washed with brine, and dried (MgSO4). Filtration and concentration to dryness gave the title compound after purification on a silica gel column eluting with 0 -2% MeOH/CH2Cl2. Anal. calculated for C32H32N6O2• 0.95 H2O:
C, 69.91 ; H, 6.22; N, 15.29;
Found C, 69.97; H, 6.15; N, 14.80.
FAB MS (M+1) 533.
Using phenylisocyanate in place of diphenylcarbamoyl chloride provided:
1-(Phenylcarbamoyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
Anal. calculated for C26H28N6O2• 0.45 H2O• 0.45 EtOAc:
C, 66.21 ; H, 6.50; N, 16.66;
Found C, 66.18; H, 6.56; N, 16.71.
FAB MS (M+1) 457. EXAMPLE 15
Preparation of 1-[2-(2-Pyridyl)-2-phenyl-2-hydroxyethyl]-3(S)-[N-(1 -(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
Step A: Preparation of Ethyl 1-[2-(2-pyridyl)-2-phenyl-2- hydroxyethyl] piperidine-3(S)- carboxylate
2-(2-pyridyl)-2-phenyl oxirane (prepared following the procedure of Example 11 , Step F) (0.106 g, 0.537 mmol) and S-(-)-ethyl nipecotate (0.093 g, 0.591 mmol) in EtOH (0.90 mL) were heated at 50°C. in a sealed tube with stirring for 24 hr. The reaction mixture was dissolved in EtOAc, washed with brine, dried (Na2SO4), filtered, and concentrated to dryness to give the title compound.
Step B: Preparation of 1-[2-(2-Pyridyl)-2-phenyl-2-hydroxyethyl] piperidine-3(S)- carboxylic acid
Ethyl 1-[2-(2-pyridyl)-2-phenyl-2-hydroxyethyl] piperidine-3(S)- carboxylate (0.187 g, 0.527 mmol) was dissolved in EtOH (5 mL), 1N NaOH (0.791 mL, 0.791 mmol) and H2O (5 mL) added, and the reaction mixture heated at reflux for 3 hr.
Neutralization with 1N HCl (to a pH of 6) and concentration to dryness gave the title compound which was used without further purification. Step C: Preparation of 1-[2-(2-Pyridyl)-2-phenyl-2-hydroxyethyl]- 3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5- ylethy l)carbamoyl] piperidine
1-[2-(2-Pyridyl)-2-phenyl-2-hydroxyethyl] piperidine-3(S)-carboxylic acid (0.172 g, 0.527 mmol) was suspended in DMF ( 5 mL) and treated with EDC (0.106 g, 0.553 mmol), HOBT (0.068 g, 0.501 mmol), followed by N-methylmorpholine to adjust the pH to 7, and 4-(4-Cyanobenzyl) histamine dihydrochloride (0.158 g, 0.527 mmol). stirring at ambient temperature for 18 hr under N2, the reaction mixture was concentrated, partitioned between CH2Cl2 and H2O, the organic phase washed with aq satd NaHCO3 soln, brine, and dried (Na2SO4). Filtration and concentration to dryness gave the title compound after purification by RP HPLC on a Waters Prep Pak eluting with 0.1%TFA/H2O: 0.1%TFA/CH3CN, 95:5 to 5:95. FAB MS (M+1) 535.
Anal. calculated for C32H34N6O2• 3.0 HCl• 2.10 H2O :
C, 56.37; H, 6.09; N, 12.33;
Found C, 56.36; H, 6.14; N, 11.54. EXAMPLE 16
Preparation of 1-(2-Pyridylethyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine Step A: Preparation of Ethyl 1-(2-pyridylethyl) piperidine-3(S)- carboxylate
S-(-)-Ethyl nipecotate (0.650 g, 4.14 mmol) and 2-vinylpyridine (0.670 mL, 6.21 mmol) were dissolved in n-butanol (50 mL) with stirring under Ar and heated at reflux for 18 hr. The reaction mixture was concentrated, and the residue chromatographed on SiO2 eluting with CH2Cl2:MeOH, 95:5 to 9: 1 to give the title compound. 1H NMR (CDCl3); δ 8.52 (dd, 1H, J = 1, 4 Hz), 7.58 (td, 1H, J = 1.8, 8 Hz), 7.26 (s, 1H), 7.18 (d, 1H, J = 8 Hz), 7.106 (dd, 1H, J = 4, 6 Hz), 4.12 (q, 2H, J = 7 Hz), 3.07 (d, 1H, J = 10 Hz), 2.95 - 3.03 (m, 2H), 2.737-2.88 (m, 3H), 2.52 - 2.62 (m, 1H), 2.28 (t, 1H, J = 10 Hz), 2.11 (td, 1H, J = 3, 10 Hz), 1.9 - 2.0 (m, 1H), 1.4 - 1.8 (m, 4H), 1.25 (t, 3H, J = 7 Hz).
Step B: Preparation of 1-(2-Pyridylethyl) piperidine-3(S)- carboxylic acid
Following the procedure of Example 15, Step B, the title compound was prepared. Step C: Preparation of 1-(2-Pyridylethyl)-3(S)-[N-(1-(4- cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine Following the procedure of Example 15, Step C, the title compound was prepared. FAB MS (M+1) 413.
EXAMPLE 17
Preparation of 1-Phenyl-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
Step A: Preparation of Ethyl 1-phenyl-(S)-piperidine carboxylate
To a solution of S-ethyl nipecotate (0.3 g, 1.91 mmol) in CH2Cl2 (20 mL) was added triphenylbismuth (1.68 g, 3.82 mmol), copper acetate (0.52 g, 2.86 mmol), and Et3N (0.39 mL, 2.86 mmol). The resulting mixture was stirred at 25 °C for 18 hr. The reaction was partitioned with CH2Cl2 (50ml) and satd NaHCO3 (30 mL), the organic layer washe with brine (30ml) and dried (MgSO4). Filtration and concentration in vacuo gave the title compound after SiO2
chromatography eluting with hexane: ethyl acetate 95:5.
Step B: Preparation of 1-Phenyl-(S)- piperidine carboxylic acid
Following the procedure of Example 15, Step B, the title compound was prepared. Step C: Preparation of 1-Phenyl-3(S)-[N-(1-(4-cyanobenzyl)-1H- imidazol-5-ylethyl)carbamoyl] piperidine
Following the procedure of Example 15, Step C, the title compound was prepared. Purification was carried out on RP HPLC Waters Prep Pak eluting with 0.1%TFA/H2O: 0.1 %TFA/CH3CN, 95:5 to 5:95.
Anal. calculated for C25H27N5O• 3.1 CF3CO2H• 0.8 H2O :
C, 47.96; H, 4.09; N, 8.96;
Found C, 47.94; H, 4.12; N, 8.97. Using the methods above, but substituting tri-3-methylphenylbismuth for triphenylbismuth, the following compound was prepared:
1-(3-Methylphenyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
Anal. calculated for C26H29N5O• 2.9 CF3CO2H• 0.8 H2O :
C, 49.44; H, 4.37; N, 9.06;
Found C, 49.43; H, 4.35; N, 9.15. EXAMPLE 18
Preparation of 1-(2,2-Diphenylethyl)-3(S)-[2-(1-(4-cyanobenzyl)-1H- imidazol-5yl)ethylthiomethyl] piperidine Step A: Preparation of Ethyl 1-(2,2-Diphenylethyl)-3(S)- piperidine carboxylate
Following the procedure outlined in Example 10, Step A, but using ethyl 3(S)-piperidine carboxylate instead of nipecotic acid, the title compound was prepared.
Step B: Preparation of 1-(2,2-Diphenylethyl)-3(S)-hydroxymethyl- piperidine
Ethyl 1-(2,2-Diphenylethyl)-3(S)-piperidine carboxylate (4.90 g, 0.014 mol) dissolved in dry ether (40mL) was added to a suspension of lithium aluminum hydride (1.93 g, 0.051 mol) in dry ether (40mL). The solution was refluxed until the starting material was consumed and then was quenched with saturated potassium sodium tartrate solution (100mL) and stirred for 2 hr. The layers were separated and the aqueous layer extracted with ether (2X). The combined organic extracts were dried (MgSO4) and concentrated to yield the title compound without further purification.
Step C: Preparation of 1-(2,2-Diphenylethyl)-3(S)-tosyloxymethyl- piperidine 1-(2,2-Diphenylethyl)-3(S)-hydroxymethyl-piperidine (1.01 g, 3.41 mmol) was dissolved in dry pyridine (25 mL) and tosyl chloride (0.684 g, 3.58 mmol) was added to the solution After stirring at ambient temperature for 18 hr, the solution was concentrated, the residue taken up in EtOAc, washed with sat. NaHCO3 solution, H2O, brine, and dried (MgSO4). Filtration and concentration gave the title compound without further purification.
Step D: Preparation of 1 -(2,2-Diphenylethyl)-3(S)- acetylthiomethyl-piperidine
1-(2,2-Diphenylethyl)-3(S)-tosyloxymethyl-piperidine (1.35 g, 3.23 mmol), potassium thiol acetate (1.47g, 12.9 mmol) and DMF (30mL) were heated at 100°C for 4 hr. The solution was poured into ice and the resulting green precipitate was dissolved in ETOAc. The organics were washed with H2O (2X), sat. NaHCO3 solution, brine, dried (MgSO4) and concentrated to give the title compound without further purification.
Step D: Preparation of the disulfide of 1-(2,2-Diphenylethyl)-3(S)- mercaptomethyl-piperidine
1-(2,2-Diphenylethyl)-3(S)-acetylthiomethyl-piperidine (0.523 g, 1.47 mmol), EtOH (20 mL), and NaOH (0.5N, 20mL) was refluxed 3 hr. The solution was concentrated to remove ethanol and the remaining aqueous solution was decanted from the green oil. The oil was washed with water (2X) and decanted. The oil was dried under reduced pressure to give the title compound without further
purification.
Step E: Preparation of 1-(2,2-Diphenylethyl)-3(S)- mercaptomethyl-piperidine
The disulfide (0.10 g, 0.161 mmol) was dissolved in acetone (15mL) and 10% aq. MeOH (10mL). Tributyl phosphine (0.261 mL, 0.322 mmol) was . The solution was stirred for 2 hr,
concentrated, and the residue was taken up in EtOAc, washed with satd NaHCO3 and dried (MgSO4). Filtration and concentration gave the title compound without further purification.
Step F: Preparation of 1-Triphenylmethyl-4-(hydroxymethyl)- imidazole
To a solution of 4-(hydroxymethyl)imidazole
hydrochloride (35.0 g, 260 mmol) in dry DMF (250 ml) at ambient temperature was added Et3N (90.6 mL, 650 mmol). A white solid precipitated from the solution. Chlorotriphenylmethane (76.1 g, 273 mmol) in of DMF (500 mL) was added dropwise. The reaction mixture was stirred for 20 hrs, poured over ice, filtered, and washed with ice water. The resulting product was slurried with cold dioxane, filtered, and dried in vacuo to provide the title compound as a white solid which was sufficiently pure for use in the next step.
Step G: Preparation of 1-Triphenylmethyl-4-(acetoxymethyl)- imidazole
1 -Triphenylmethyl-4-(hydroxymethyl)-imidazole (260 mmol) was suspended in pyridine (500 mL). Acetic anhydride (74 mL, 780 mmol) was added dropwise, and the reaction was stirred for 48 hr during which it became homogeneous. The solution was poured into EtOAc, washed sequentially with water, 5% aqueous HCl solution, satd aqueous NaHCO3, solution, and brine. The organic extracts were dried (Na2SO4), and concentrated in vacuo to provide the product as a white powder, which was sufficiently pure for use in the next reaction.
Step H: Preparation of 1-(4-Cyanobenzyl)-5-(acetoxymethyl)- imidazole hydrobromide
1-Triphenylmethyl-4-(acetoxymethyl)-imidazole (85.8 g, 225 mmol) and 4-cyanobenzyl bromide (50.1 g, 232 mmol) in EtOAc (500mL) were stirred at 60 °C for 20 hr, during which a pale yellow precipitate formed. The reaction was cooled to room temperature and filtered to provide the solid imidazolium bromide salt. The filtrate was concentrated in vacuo to a volume (200 mL), reheated at 60 °C for 2 hrs, cooled to room temperature, and filtered again. The filtrate was concentrated in vacuo to a volume (100 mL), reheated at 60 °C for another 2hrs, cooled to room temperature, and concentrated in vacuo to provide a pale yellow solid. All of the solid material was combined, dissolved in methanol (500mL), and warmed to 60 °C. After 2hrs, the solution was concentrated in vacuo to provide a white solid which was triturated with hexane to remove soluble materials. Removal of residual solvents in vacuo provided the titled product hydrobromide as a white solid which was used in the next step without further purification.
Step I: Preparation of 1-(4-Cyanobenzyl)-5-(hydroxymethyl)- imidazole
1-(4-Cyanobenzyl)-5-(acetoxymethyl)-imidazole hydrobromide (50.4 g, 150 mmol) in 3: 1 THF/water (1.5 L) at 0 °C was added lithium hydroxide monohydrate (18.9 g, 450 mmol). After 1hr, the reaction was concentrated in vacuo, diluted with EtOAc (3 L), and washed with water, sat. aq. NaHCO3 and brine. The solution was then dried (Na2SO4), filtered, and concentrated in vacuo to provide the crude product as a pale yellow fluffy solid which was sufficiently pure for use in the next step without further purification.
Step J: Preparation of 1-(4-Cyanobenzyl)-5-(chloromethyl)- imidazole
A solution of 1-(4-cyanobenzyl)-5-(hydroxymethyl)-imidazole (1.0g, 4.70mmol), in thionyl chloride (5ml), was stirred at
70°C for 16hrs. The solvent was evaporated in vacuo and the resulting solid suspended in CH2Cl2, collected by filtration and dried in vacuo.
The material was sufficiently pure to be used without further
purification.
1H NMR CD3OD d 9.06 (1H, s), 7.83(2H, d, J=8.0Hz), 7.77(1H, s),
7.55(2H, d, J=8.0Hz), 5.67(2H, s) and 4.78(2H, s) ppm.
Step K: Preparation of 1-(2,2-Diphenylethyl)-3(S)-[N-(1-(4- cyanobenzyl)- H-imidazol-5-ylethylthiomethyl] piperidine 1-(2,2-Diphenylethyl)-3(S)-mercaptomethyl-piperidine (0.322 mmol), 1-(4-cyanobenzyl)-5-(chloromethyl)-imidazole (0.1 17 g, 0.386 mmol), and diisopropylethylamine (0.168 mL, 0.966 mmol) were dissolved in CH2Cl2 (10mL) and refluxed overnight. The solution was concentrated and the residue was taken up in EtOAc, washed with sat. NaHCO3, water and brine. The organics were dried (MgSO4), concentrated, chromatographed (0-3% MeOH,CH2Cl2,NH4OH), Prep HPLC (100:0 - 5:95 H2O: CH3CN w/ 0.1% TFA), free based, and acidified with 1N HCl solution in ether to yield the title compound. H NMR (CD3OD); δ 9.03 (s, 1H), 7.80 (D, 2H, J=8Hz), 7.65 (s, 1H), 7.50-7.33 (m, 10H), 7.26 (D, 2H, J=8Hz), 5.64 (s, 2H), 4.68 (t, 1H, J=7Hz), 3.98-3.88 (m, 2H), 3.72 (s, 2H), 3.59-3.56 (m, 1H), 3.51-3.47 (m, 1H), 2.87-2.83 (m, 1H), 2.64 (t, 1H, J=12Hz), 2.49-2.42 (m, 1H), 2.35-2.27 (m, 1H), 2.02 (br s, 1H), 1.90-1.77 (m, 3H), 1.19-1.12 (m,1H). FAB MS 507 (M+1)
Anal. calculated for C32H34N4S• 2.5 HCl• 1.65 H2O :
C, 61.24; H, 6.69; N, 8.93;
Found C, 61.20; H, 5.93; N, 8.72. EXAMPLE 19
Preparation of 1-(2,2-Diphenylethyl)-3(S)-[2-(1-(4-cyanobenzyl)-1H-imidazol-5-yl)ethylsulfonylmethyl] piperidine
1-(2,2-Diphenylethyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethylthiomethyl] piperidine (0.050 g, 0.087 mmol) was dissolved in MeOH (lmL) and H2O (lmL), treated with Oxone (0.106 g, 0.173 mmol), and stirred at ambient temperature for 1hr. The solution was concentrated and the residue was taken up in EtOAc, washed with water and brine. The organics were dried (MgSO4) and chromatographed (RP HPLC Waters Prep Pak, 100:0 - 5:95
H2O:CH3CN w/ 0.1% TFA) to give the title compound as a TFA salt. FAB MS 539 (M+1)
Anal. calculated for C32H34N4O2S• 3.70 CF3CO2H• 0.90 H2O :
C, 48.44; H, 4.08; N, 5.74; Found C, 48.43; H, 4.06; N, 5.96.
EXAMPLE 20 Preparation of 1-(2,2-Diphenylethyl)-3(S)-[N-( 1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)-N-methyl-carbamoyl] piperidine
Step A: Preparation of 1-(tert-Butyloxycarbonyl)-3(S)-[N-(1-(4- cyanobenzyl)-1H-imidazol-5-ylethyl)-N-methylcarbamoyl] piperidine
1-(tert-Butyloxycarbonyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine (Example 11, Step D)(0.100 g, 0.228 mmol) was dissolved in dry DMF (5mL) and cooled in a ice bath. NaH (0.01 1 g, 0.274 mmol) and CH3I (0.023 mL, 0.365mmol) were added, and the mixture was stirred at 0°C. for 1.5 hr. The reaction was quenched with water, extracted with EtOAc, the organics washed with H2O (3X), dried (MgSO4), and concentrated to give the title compound. Step B: Preparation of 1-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazole- 5-ethyl)-N-methylcarbamoyl] piperidine
The title compound was prepared following the procedure outlined in Example 11, Step E. Step C: Preparation of 1-(2,2-Diphenylethyl)-3(S)-[N-1-(4- cyanobenzyl)-1H-imidazol-5-ylethyl)-N-methylcarbamoyl] piperidine
The title compound was prepared following the procedure outlined in Example 11, Step H. FAB MS (M+l) 532.
Anal. calculated for C34H37N5O• 2.70 CF3CO2H• 1.60 H2O :
C, 54.50; H, 4.98; N, 8.06;
Found C, 54.49; H, 4.97; N, 7.99. Using the methods described above the following compound was prepared:
1-(3-Bromobenzyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)-N-methyl-carbamoyl] piperidine
FAB MS (M + 1) 520
EXAMPLE 21 Preparation of 1-(2,2-Diphenylethyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)aminomethyl] piperidine
Step A: Preparation of 1-(2,2-Diphenylethyl)-3(S)-piperidine
carboxaldehyde
1-(2,2-Diphenylethyl)-3(S)-hydroxymethyl-piperidine
(Example 18, Step B)_(2.10 g, 7.10mmol) and Et3N (2.97 mL, 21.3 mmol) were dissolved in dry DMSO (40mL) and cooled in an ice bath. Pyridine SO3 complex (3.39 g, 21.3 mmol) was added slowly, and the solution was stirred for 1hr while allowing to warm to room
temperature. The solution was poured into ice water, extracted with CH2CL2 (2X), the combined organics washed with satd NaHCO3 solution, H2O, brine, and dried (MgSO4). Filtration and concentration gave the title compound without further purification. Step B: Preparation of 1-(2,2-Diphenylethyl)-3(S)-[N-(1- (cyanobenzy1)-1H-imidazol-5-ylethyl)aminomethyl]- piperidine
1-(2,2-Diphenylethyl)-3(S)-piperidine
carboxaldehyde (0.575 g, 1.96 mmol),3-(4-cyanobenzyl) histamine dihydrochloride (0.388 g, 1.30 mmol), and NaCNBH3 (0.123 g,
1.96mmol) were dissolved in MeOH (15mL) and stirred at ambient temperature overnight. The solution was concentrated and the residue taken up in EtOAc, washed with sat. NaHCO3 solution, H2O, and brine. The organics were dried, concentrated, and chromatographed (0-4% MeOH/CH2Cl2/NH4OH) to give the title compound which was isolated as a tris HCl salt . 1H NMR (CD3OD); δ 9.05 (s, 1H), 7.81 (d, 2H, J=8Hz), 7.68 (s, 1H), 7.54 (d, 2H, J=8Hz), 7.49-7.25 (m, 10H), 5.67 (s, 2H), 4.76-7.74 (m, 1H), 4.05-3.88 (m, 3H), 3.54-3.47 (m, 1H), 3.33-3.30 (m, 1H), 3.19-3.15 (m, 2H), 3.02-3.00 (m, 2H), 2.92-2.90 (m, 2H), 2.42 (br s, 1H), 1.99-1.91 (m, 2H), 1.82-1.78 (m, 1H), 1.31-1.28 (m, 2H). FAB MS 504 (M+1).
Anal. calculated for C33H37N5• 3.0 HCl• 0.90 H2O :
C, 62.99; H, 6.70; N, 1 1.13;
Found C, 63.04; H, 6.83; N, 11.05.
EXAMPLE 22
Preparation of 1-(2,2-Diphenylethyl)-3(S)-[N-(1-(4-cyanobenzyl)- 1H-imidazol-5-ylethyl)-N-acetyl-aminomethyl] piperidine
1-Diphenylethyl-3(S)-[N-1(4-cyanobenzyl)-1H-imidazol-5-ylethylaminomethyl]piperidine (0.151 g , 0.246 mmol) and Et3N (0.154 mL, 1.10 mmol) were dissolved in dry CH2Cl2 (15mL) and cooled in an ice bath. A solution of acetyl chloride (0.026 mL, 0.369 mmol) in CH2Cl2 (1mL) was added and the reaction was stirred for 18 hr while allowing to warm to room temperature. The solution was diluted with EtOAc, washed with sat. NaHCO3 solution, H2O, and brine. The organics were dried (MgSO4) and concentrated to yield the title compound. 1H NMR (CDCl3); δ 7.59 (d, 2H, J=8Hz), 7.52 (s, 1 H), 7.49-7.04 (m, 12H), 6.87 (s, 1H), 5.29 (s, 2H), 4.17 (t, 1H, J=7Hz), 3.32-3.25 (m, 1H), 3.21-3.13 (m, 1H), 3.04-2.96 (m, 4H), 2.68-2.49 (m, 4H), 2.24-2.20 (m, 1H), 1.91-1.73 (m, 6H), 1.56-1.50 (m, 1H), 1.46-1.43 (m, 1H), 0.96-0.92 (m, 1H). FAB MS 546 (M+1 ).
Anal. calculated for C35H39N5O• 0.70 H2O :
C, 75.29; H, 7.29; N, 12.54;
Found C, 75.25; H, 7.26; N, 12.06. 1 Using the methods described in Examples 21 and 22, but substituting the requisite amine in Ex. 21, Step B, and the requisite acylating or alkylating agent in Ex. 22, the following compounds were prepared: 1-(2,2-Diphenylethyl)-3(S)-[N-(1-(4-cyanobenzyl)-2-methyl-1H-imidazol-5-ylethyl)-N-acetyl-aminomethyl] piperidine
Anal. calculated for C36H41N5O• 0.35 H2O :
C, 76.39; H, 7.43; N, 12.37;
Found C, 76.39; H, 7.13; N, 12.32.
FAB MS (M+1) 560
1-(2,2-Diphenylethyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)-N-cyclopropylmethyl-aminomethyl] piperidine
Anal. calculated for C37H43N5• 3.0 HCl• 0.95 H2O• 0.60 CH2Cl2 :
C, 62.41; H, 6.73; N, 9.53;
Found C, 62.42; H, 7.02; N, 9.43.
FAB MS (M+1) 558
1-(2,2-Diphenylethyl)-3(S)-[N-(2-methyl-1H-imidazol-4-ylethyl)-N-(4-cyanobenzoyl)aminomethyl] piperidine
Anal. calculated for C34H37N5O• 2.95 CF3CO2H• 1.00 H2O :
C, 54.08; H, 4.77; N, 7.90;
Found C, 54.06; H, 4.74; N, 7.91.
FAB MS (M+1) 532
EXAMPLE 23
Preparation of 1-(2,2-Diphenylethyl)-3(S)-[5-(4-cyanobenzyl)-1H-imidazol-1 -ylmethyl] piperidine bistrifluoroacetate
Step A: Preparation of 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 for 45 minutes, at 20°C. The suspension was cooled to 0°C and α-bromo-p-toluinitrile (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 (2.40g, 3.64 mmol) and 5-iodotrityl imidazole (15.95g, 36.6 mmol) were added in one
portion.The resulting mixture was stirred 16 hours at 20°C and then quenched by addition of saturated 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 × 250 mL), dried (MgSO4) and the solvent evaporated in vacuo. The residue was chromatographed (Silica gel, 0-20% EtOAc inCH2Cl2) to afford the title compound as a white solid.
1H NMR (CDCI3, 400Mz) δ (7.54 (2H, d, J=7.9Hz), 7.38(1H, s), 7.36-7.29 (1 1H, m), 7.15-7.09(6H, m), 6.58(1H, s) and 3.93(2H, s) ppm.
Step B: Preparation of 1-(2,2-Diphenylethyl)-3(S)-[5-(4- cyanobenzyl)-1H-imidazol-1-ylmethyl] piperidine bis trifluoroacetate bistrifluoroacetate
To a solution of 1-(2,2-Diphenylethyl)-3(S)-hydroxymethylpiperdine (Example 18, Step B) (0.271 g, 0.917 mmol) and 1 -trityl-4-(4-cyanobenzyl)imidazole (0.390 g,0.917 mmol) in CH2Cl2 (7 mL) was added diisopropylethylamine (0.639 mL, 3.67 mmol) under N2. The mixture was cooled to -78 °C and
trifluoromethanesulfonic anhydride (0.154 mL, 0.917mmol) was added dropwise via syringe. The cooling bath was removed and the reaction was stirred at 25 °C for 18hr. The reaction was evaporated in vacuo and the residue was dissolved in methanol (20mL) and heated to reflux for 1hr. After cooling the mixture was evaporated in vacuo and the residue was partitioned with EtOAc (50mL) and satd NaHCO3(30mL), the organic layer separated, washed with brine (30mL) and dried
(MgSO4). Filtration and evaporation in vacuo gave the title compound which was purified by chromatography on silica gel using
CH2Cl2:MeOH, 98:2, followed by preparative HPLC on a Waters C-18 Delta-pak column. Anal. calculated for C3 1H32N4•3.10 CF3CO2H• 0.75 H2O :
C, 53.98; H, 4.46; N, 6.77;
Found C, 54.02; H, 4.47; N, 6.75. EXAMPLE 24
Preparation of 1-(2,2-Diphenylethyl)-3(S)-[5-(4-cyanobenzyl)-1H-imidazol- 1-ylethylcarbamoyl] piperdine Step A: Preparation of 5-(4-cyanobenzyl)-1H-imidazol-1- ylethylphthalimide
2-Hydroxyethylphthalimide (3.23 g, 16.9 mmol) was dissolved in CH2Cl2 (50ml) and Et3N(3.92 mL, 28.2 mmol) under N2. The reaction was cooled to -78°C and trifluoromethanesulfonic anhydride (2.85 mL, 16.9 mmol) was added dropwise via syringe.
After stirring for 0.5 hr at 0°C the reaction was cooled to -20°C and 1-trityl-4-(4-cyanobenzyl)imidazole (2.4 g, 5.64 mmol) in CH2Cl2 (20mL) was added to the mixture which was stirred at 25°C for 18 hr. The reaction was concentrated in vacuo to dryness, and the residue was dissolved in MeOH (75mL) and heated to reflux for 2 hr. After cooling and evaporation in vacuo the residue was partitioned with EtOAc (100 mL) and satd NaHCO3 (30 mL). The organic layer was washed with brine (30 mL) and dried (MgSO4). Evaporation in vacuo gave the title compound after chromatography on silica gel eluting with
CH2Cl2:MeOH:NH4OH 95:5:0.5 (3 L), 90: 10: 1.0 (2 L), 85: 15: 1.5 (3 L).
Step B: Preparation of 5-(4-cyanobenzyl)-1-(2-aminoethyl)- 1H- imidazole
To a solution of 5-(4-cyanobenzyl)-1H-imidazol-1-ylethylphthalimide (3.2 g, 9.2 mmol) in absolute EtOH (75 mL) was added hydrazine (0.72 mL, 23.0 mmol), and the mixture was refluxed for 18 hr. Dimethyl phthalate (7.59 mL, 46 mmol) was added to the mixture and refluxing was continued for 4 hr. The reaction was cooled in a freezer for 18 hr. The solids that formed were filtered and washed with water. Evaporation in vacuo afforded the title compound after chromatography on silica gel eluting with CH2Cl2:MeOH:NH4OH
95:5:0.5 (2 L), 90:10: 1.0 (2 L), 85: 15: 1.5 (2 L).
Step C: Preparation of 1-(2,2-Diphenylethyl)-3(S)-[5-(4- cyanobenzyl)-1H-imidazol-1-ylethylcarbamoyl] piperdine
Following the procedure outlined in Example 10, Step B using 1-(2,2-Diphenylethyl)-3(S)-carboxy piperidine and the amine from Step B above, the title compound was prepared.
Anal. calculated for C33H35N5O• 0.60 H2O :
C, 74.99; H, 6.90; N, 13.25;
Found C, 74.99; H, 6.98; N, 13.19.
EXAMPLE 25 In vitro inhibition of ras farnesyl transferase
Assays of farnesyl-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
incoφoration of radioactivity in the presence of the test compound when compared to the amount of incoφoration 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 compounds of the instant invention that are described in Example 1-24 were tested for inhibitory activity against human FPTase by the assay described above and were found to have IC50 of < 10 μM. EXAMPLE 26
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[35S]methionine (1000 Ci/mmol). After an additional 20 hours, the cells are lysed in 1 ml lysis buffer (1% NP40/20 mM HEPES, pH 7.5/5 mM MgCl2/1mM 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 × 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 Y13-259 (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 27
In vivo growth inhibition assay
To determine the biological consequences of FPTase inhibition, the effect of the compounds of the instant invention on the anchorage-independent growth of Ratl cells transformed with either a v-ras, v-raf, or v-mos oncogene is tested. Cells transformed by v-Raf and v-Mos maybe included in the analysis to evaluate the specificity of instant compounds for Ras-induced cell transformation.
Rat 1 cells transformed with either v-ras, v-raf, or v-mos are seeded at a density of 1 × 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 imgf000145_0001
wherein:
R1 a, R1b and R1 c are independently selected from:
a) hydrogen,
b) unsubstituted or substituted aryl, unsubstituted or
substituted heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, R8O-, R9S(O)m-, R8C(O)NR8-, CN, NO2, (R8)2N-C(NR8)-, R8C(O)-, R8OC(O)-, N3, -N(R8)2, or R9OC(O)NR8-,
c) C1-C6 alkyl unsubstituted or substituted by unsubstituted or substituted aryl, heterocyclic, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, R8O-, R9S(O)m-, R8C(O)NR8-, CN, (R8)2N-C(NR8)-, R8C(O)-, R8OC(O)-, N3, -N(R8)2, or R9OC(O)-NR8-;
R2 is selected from: H; unsubstituted or substituted C1 -8 alkyl,
unsubstituted or substituted C2-8 alkenyl, unsubstituted or substituted aryl, ,
unsubstituted or substituted heterocycle,
Figure imgf000145_0003
Figure imgf000145_0002
and -S(O)2R6,
wherein the substituted group is substituted with one or more of
Figure imgf000146_0001
Figure imgf000147_0003
or ,
R3 is selected from: H;
Figure imgf000147_0001
Figure imgf000147_0002
R4 is independently selected from:
a) hydrogen,
b) unsubstituted or substituted aryl, unsubstituted or
substituted heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, perfluoroalkyl, F, Cl, Br, R8O-, R9S(O)m- , R8C(O)NR8-, CN, NO2, R8 2N-C(NR8)-, R8C(O)-, R8OC(O)-, N3, -N(R8)2, or R9OC(O)NR8-, and c) C 1-C6 alkyl unsubstituted or substituted by aryl,
heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, perfluoroalkyl, F, CI, Br, R8O-, R9S(O)m-, R8C(O)NH-, CN, H2N-C(NH)-, R8C(O)-, R8OC(O)-, N3, -N(R8)2, or R8OC(O)NH-;
R5 is independently selected from:
a) hydrogen,
b) C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl,
perfluoroalkyl, F, CI, Br, R8O-, R9S(O)m-, R8C(O)NR8-, CN, NO2, (R8)2N-C-(NR8)-, R8C(O)-, R8OC(O)-, N3,
-N(R8)2, or R9OC(O)NR8-, and
c) C1-C6 alkyl, unsubstituted or substituted by perfluoroalkyl, F, Cl, Br, R8O-, R9S(O)m-, R8C(O)NR8-, CN, (R8)2N- C(NR8)-, R8C(O)-, R8OC(O)-, N3, -N(R8)2, or
R9OC(O)NR8-;
R6, R7 and R7a are independently selected from: H; C 1-4 alkyl, C3-6 cycloalkyl, heterocycle, aryl, C1 -4 perfluoroalkyl, unsubstituted or substituted with one or two substituents selected from:
Figure imgf000148_0001
R6 and R7 may be joined in a ring;
R7 and R7a may be joined in a ring; R8 is independently selected from hydrogen, C1-C6 alkyl, benzyl,
2,2,2-trifluoroethyl and aryl; R9 is independently selected from C1-C6 alkyl and aryl;
R10 is selected from: H; R8C(O)-; R9S(O)m-; unsubstituted or substituted C1 -4 alkyl, unsubstituted or substituted C3-6 cycloalkyl, unsubstituted or substituted heterocycle, unsubstituted or substituted aryl, substituted aroyl, unsubstituted or substituted heteroaroyl, substituted arylsulfonyl, unsubstituted or substituted heteroarylsulfonyl, wherein the substituted group is substituted with one or two substituents selected from:
Figure imgf000149_0001
A1 and A2 are independently selected from: a bond, -CH=CH-, -C≡C-,
-C(O)-, -C(O)NR8-, -NR8C(O)-, O, -N(R8)-, -S(O)2N(R8)-, -N(R8)S(O)2-, or S(O)m;
V is selected from:
a) hydrogen,
b) heterocycle,
c) aryl,
d) C 1-C20 alkyl wherein from 0 to 4 carbon atoms are
replaced with a a heteroatom selected from O, S, and N, and
e) C2-C20 alkenyl,
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; W is a heterocycle;
X is a bond, -C(=O)NR10-, -NR10C(=O)-, -S(O)m-, -NR10-, O or -C(=O)-; m is 0, 1 or 2;
n is 0, 1 , 2, 3 or 4;
p is 0, 1, 2, 3 or 4;
q is 0, 1, 2, 3 or 4;
r is 0 to 5, provided that r is 0 when V is hydrogen;
s is 1 or 2;
t is 0 or 1 ; and the dashed lines represent optional double bonds; or an optical isomer or pharmaceutically acceptable salt thereof.
2. The compound according to Claim 1, which inhibits farnesyl-protein transferase, of the formula A:
Figure imgf000150_0001
wherein:
R1 a and R1 c are independently selected from: hydrogen, C3-C10 cycloalkyl, R8O-, -N(R8)2, F or C1-C6 alkyl;
R1b is independently selected from:
a) hydrogen, b) unsubstituted or substituted aryl, unsubstituted or
substituted heterocycle, C3-C6 cycloalkyl, R8O-, -N(R8)2 or C2-C6 alkenyl,
c) C 1-C6 alkyl unsubstituted or substituted by unsubstituted or substituted aryl, heterocycle, C3-C6 cycloalkyl, C2-C6 alkenyl, R8O-, or -N(R8)2;
R2 is selected from:
Figure imgf000151_0001
Figure imgf000152_0003
or
R3 is selected from: H;
Figure imgf000152_0001
Figure imgf000152_0002
R4 is independently selected from:
a) hydrogen,
b) C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6
perfluoroalkyl, F, Cl, R8O-, R8C(O)NR8-, CN, NO2, (R8)2N-C(NR8)-, R8C(O)-, R8OC(O)-, -N(R8)2, or R9OC(O)NR8-, and
c) C1-C6 alkyl substituted by C1 -C6 perfluoroalkyl, R8O-, R8C(O)NR8-, (R8)2N-C(NR8)-, R8C(O)-, R8OC(O)-, -N(R8)2, or R9OC(O)NR8-; R5 is selected from:
a) hydrogen,
b) C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, C1-C6 perfluoroalkyl, F, Cl, R8O-, R9S(O)m-, R8C(O)NR8-, CN, NO2, (R8)2N-C(NR8)-, R8C(O)-, R8OC(O)-, -N(R8)2, or R9OC(O)NR8-, and
c) C1-C6 alkyl unsubstituted or substituted by C 1-C6
perfluoroalkyl, F, Cl, R8O-, R9S(O)m-, R8C(O)NR8-, CN, (R8)2N-C(NR8)-, R8C(O)-, R8OC(O)-, -N(R8)2, or R9OC(O)NR8-; R6, R7 and R7a are independently selected from:
H; C1 -4 alkyl, C3-6 cycloalkyl, aryl, heterocycle,
unsubstituted or substituted with:
a) C1 -4 alkoxy,
b) halogen, or
c) substituted or unsubstituted aryl or substituted or unsubstituted heterocycle; R8 is independently selected from hydrogen, C1-C6 alkyl, benzyl, 2,2,2-trifluoroethyl and aryl; R9 is independently selected from C1-C6 alkyl and aryl;
R10 is selected from: H; R8C(O)-; R9S(O)m-; unsubstituted or substituted C1 -4 alkyl, unsubstituted or substituted C3-6 cycloalkyl, unsubstituted or substituted heterocycle, unsubstituted or substituted aryl, substituted aroyl, unsubstituted or substituted heteroaroyl, substituted arylsulfonyl, unsubstituted or substituted heteroarylsulfonyl, wherein the substituted group is substituted with one or two substituents selected from:
Figure imgf000153_0001
A1 and A2 are independently selected from: a bond, -CH=CH-, -C≡C-,
-C(O)-, -C(O)NR8-, O, -N(R8)-, or S(O)m; V is selected from: a) heterocycle selected from pyrrolidinyl, imidazolyl, pyridinyl, thiazolyl, pyridonyl, 2-oxopiperidinyl, indolyl, quinolinyl, isoquinolinyl, and thienyl, and
b) aryl;
W is a heterocycle selected from pyrrolidinyl, imidazolyl, pyridinyl, thiazolyl, pyridonyl, 2-oxopiperidinyl, indolyl, quinolinyl, or isoquinolinyl; X is a bond, -C(=O)NR10-, -NR10C(=O)-, -S(O)m- or -NR10-; m is 0, 1 or 2;
n is 0, 1, 2, 3 or 4;
p is 1, 2 or 3;
q is 0 or 1 ;
r is 0 to 5, provided that r is 0 when V is hydrogen;
s is 1 or 2; and
t is 1; or an optical isomer or pharmaceutically acceptable salt thereof.
3. The compound according to Claim 1, which inhibits farnesyl-protein transferase, of the formula B:
Figure imgf000154_0001
wherein: R1 a and R1c are independently selected from: hydrogen, C3-C10
cycloalkyl, R8O-, -N(R8)2, F or C1-C6 alkyl;
R1 b is independently selected from:
a) hydrogen,
b) aryl, heterocycle, C3-C10 cycloalkyl, R8O-, -N(R8)2, F or C2-C6 alkenyl,
c) unsubstituted or substituted C1-C6 alkyl wherein the
substituent on the substituted C 1-C6 alkyl is selected from unsubstituted or substituted aryl, heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, R8O- and -N(R8)2;
R2 is selected from: H; unsubstituted or substituted C1-8 alkyl,
unsubstituted or substituted C2-8 alkenyl, unsubstituted or substituted aryl, unsubstituted or substituted heterocycle,
Figure imgf000155_0001
,
Figure imgf000155_0002
and
-S(O)2R6,
wherein the substituted group is substituted with one or more of:
Figure imgf000155_0003
Figure imgf000156_0001
or
R3 is selected from: H;
Figure imgf000157_0001
Figure imgf000157_0002
R4 is independently selected from:
a) hydrogen,
b) aryl, substituted aryl, heterocycle, substituted heterocycle,
C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1 -C6 perfluoroalkyl, F, Cl, R8O-, R8C(O)NR8-, CN, NO2, (R8)2N-C(NR8)-, R8C(O)-, -N(R8)2, or R9OC(O)NR8-, and
c) C1 -C6 alkyl substituted by C1 -C6 perfluoroalkyl, R8O-, R8C(O)NR8-, (R8)2N-C(NR8)-, R8C(O)-, -N(R8)2, or R9OC(O)NR8-;
R5a and R5b are independently hydrogen, C1-C6 alkyl, cyclopropyl, trifluoromethyl and halogen;
R6, R7 and R7a are independently selected from:
H; C1 -4 alkyl, C3-6 cycloalkyl, aryl, heterocycle, unsubstituted or substituted with:
a) C1 -4 alkoxy,
b) halogen, or
c) substituted or unsubstituted aryl or substituted or unsubstituted heterocycle; R8 is independently selected from hydrogen, C1 -C6 alkyl, 2,2,2- trifluoroethyl, benzyl and aryl; R9 is independently selected from C1-C6 alkyl and aryl; R!0 is selected from: H; R8C(O)-; R9S(O)m-; unsubstituted or substituted Cι_4 alkyl, unsubstituted or substituted C3-6 cycloalkyl, unsubstituted or substituted heterocycle, unsubstituted or substituted aryl, substituted aroyl, unsubstituted or substituted heteroaroyl, substituted arylsulfonyl, unsubstituted or substituted heteroarylsulfonyl, wherein the substituted group is substituted with one or two substituents selected from:
Figure imgf000158_0001
A1 and A2 are independently selected from: a bond, -CH=CH-, -C≡C-,
-C(O)-, -C(O)NR8-, O, -N(R8)-, or S(O)m;
V is selected from:
a) hydrogen,
b) heterocycle selected from pyrrolidinyl, imidazolyl,
imidazolinyl, pyridinyl, thiazolyl, pyridonyl, 2- oxopiperidinyl, oxazolyl, indolyl, quinolinyl, isoquinolinyl, triazolyl and thienyl,
c) aryl,
d) C1 -C20 alkyl wherein from 0 to 4 carbon atoms are
replaced with a a heteroatom selected from O, S, and N, and
e) C2-C20 alkenyl, and provided that V is not hydrogen if A1 is S(O)m and V is not hydrogen if A1 is a bond, n is 0 and A2 is S(O)m;
X is a bond, -C(=O)NR10-, -NR10C(=O)-, -S(O)m- or -NR10-; m is 0, 1 or 2;
n is 0, 1, 2, 3 or 4;
p is 0, 1, 2, 3 or 4;
q is 0 or 1; and
r is 0 to 5, provided that r is 0 when V is hydrogen; or an optical isomer or pharmaceutically acceptable salt thereof.
4. The compound according to Claim 1, which inhibits farnesyl-protein transferase, of the formula C:
Figure imgf000159_0001
wherein: R1a and R1c are independently selected from: hydrogen, C3-C10 cycloalkyl, R8O-, -N(R8)2, F or C1 -C6 alkyl;
R1b is independently selected from:
a) hydrogen,
b) aryl, heterocycle, C3-C10 cycloalkyl, R8O-, -N(R8)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, R8O- and -N(R8)2;
R2 is selected from: H; unsubstituted or substituted C1 -8 alkyl,
unsubstituted or substituted C2-8 alkenyl, unsubstituted or substituted aryl, unsubstituted or substituted heterocycle,
Figure imgf000160_0001
,
Figure imgf000160_0002
and
-S(O)2R6,
wherein the substituted grou is substituted with one or more of:
Figure imgf000160_0003
Figure imgf000161_0003
or ,
R3 is selected from: H;
Figure imgf000161_0001
Figure imgf000161_0002
R4 is independently selected from:
a) hydrogen,
b) aryl, substituted aryl, heterocycle, substituted heterocycle, C1 -C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 perfluoroalkyl, F, Cl, R8O-, R8C(O)NR8-, CN, NO2, (R8)2N-C(NR8)-, R8C(O)-, -N(R8)2, or R9OC(O)NR8-, and c) C1 -C6 alkyl substituted by C1 -C6 perfluoroalkyl, R8O-, R8C(O)NR8-, (R8)2N-C(NR8)-, R8C(O)-, -N(R8)2, or R9OC(O)NR8-; R5a and R5b are independently hydrogen, C1 -C6 alkyl, cyclopropyl, trifluoromethyl and halogen;
R6, R7 and R7a are independently selected from:
H; C1 -4 alkyl, C3-6 cycloalkyl, aryl, heterocycle, unsubstituted or substituted with:
a) C1 -4 alkoxy,
b) halogen, or
c) substituted or unsubstituted aryl or substituted or unsubstituted heterocycle; R8 is independently selected from hydrogen, C1 -C6 alkyl, 2,2,2- trifluoroethyl, benzyl and aryl; R9 is independently selected from C1 -C6 alkyl and aryl;
R10 is selected from: H; R8C(O)-; R9S(O)m-; unsubstituted or substituted C1 -4 alkyl, unsubstituted or substituted C3-6 cycloalkyl, unsubstituted or substituted heterocycle, unsubstituted or substituted aryl, substituted aroyl, unsubstituted or substituted heteroaroyl, substituted arylsulfonyl, unsubstituted or substituted heteroarylsulfonyl, wherein the substituted group is substituted with one or two substituents selected from:
Figure imgf000162_0001
Figure imgf000163_0001
A1 and A2 are independently selected from: a bond, -CH=CH-, -C≡C-,
-C(O)-, -C(O)NR8-, O, -N(R8)-, or S(O)m;
V is selected from:
a) hydrogen,
b) heterocycle selected from pyrrolidinyl, imidazolyl,
imidazolinyl, pyridinyl, thiazolyl, pyridonyl, 2- oxopiperidinyl, oxazolyl, indolyl, quinolinyl, isoquinolinyl, triazolyl and thienyl,
c) aryl,
d) C1-C20 alkyl wherein from 0 to 4 carbon atoms are
replaced with a a heteroatom selected from O, S, and N, and
e) C2-C20 alkenyl, and
provided that V is not hydrogen if A1 is S(O)m and V is not hydrogen if A1 is a bond, n is 0 and A2 is S(O)m;
X is a bond, -C(=O)NR10-, -NR10C(=O)-, -S(O)m- or -NR10-; m is 0, 1 or 2;
n is 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,
-NR8- or O; q is 0 or 1 ; and
r is 0 to 5, provided that r is 0 when V is hydrogen; or an optical isomer or pharmaceutically acceptable salt thereof.
5. The compound according to Claim 3, which inhibits farnesyl-protein transferase, of the formula D:
Figure imgf000164_0001
wherein:
R1 a and R1 c are independently selected from: hydrogen, C3-C10
cycloalkyl or C1 -C6 alkyl; R1 b is independently selected from:
a) hydrogen,
b) aryl, heterocycle, C3-C10 cycloalkyl, R8O-, -N(R8)2, F or C2-C6 alkenyl,
c) C1 -C6 alkyl unsubstituted or substituted by aryl,
heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, R8O-, or
-N(R8)2;
R2 is selected from: H; unsubstituted or substituted C1 -8 alkyl, unsubstituted or substituted aryl,
Figure imgf000164_0002
,
Figure imgf000164_0003
and -S(O)2R6, wherein the substituted group is substituted with one or more of:
Figure imgf000165_0001
Figure imgf000166_0004
Figure imgf000166_0003
or ,
R3 is selected from: H;
Figure imgf000166_0001
Figure imgf000166_0002
R4 is independently selected from:
a) hydrogen,
b) aryl, substituted aryl, heterocycle, substituted heterocycle, C1 -C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1 -C6 perfluoroalkyl, F, Cl, R8O-, R8C(O)NR8-, CN, NO2, (R8)2N-C(NR8)-, R8C(O)-, -N(R8)2, or R9OC(O)NR8-, and
c) C1 -C6 alkyl substituted by C1 -C6 perfluoroalkyl, R8O-, R8C(O)NR8-, (R8)2N-C(NR8)-, R8C(O)-, -N(R8)2, or R9OC(O)NR8-;
R5a and R5b are independently hydrogen, ethyl, cyclopropyl or methyl;
R6, R7 and R7a are independently selected from:
H; C1 -4 alkyl, C3-6 cycloalkyl, aryl, heterocycle,
unsubstituted or substituted with:
a) C1 -4 alkoxy,
b) halogen, or
c) substituted or unsubstituted aryl or substituted or unsubstituted heterocycle; R8 is independently selected from hydrogen, C1 -C6 alkyl, 2,2,2- trifluoroethyl, benzyl and aryl; R9 is independently selected from C1 -C6 alkyl and aryl;
R10 is selected from: H; R8C(O)-; R9S(O)m-; unsubstituted or substituted C1 -4 alkyl, wherein the substituted alkyl group is substituted with one or two substituents selected from:
Figure imgf000167_0001
A1 is selected from: a bond, -C(O)-, O, -N(R8)-, or S(O)m; X is a bond, -C(=O)NR10-, -NR10C(=O)-, -S(O)m- or -NR10-; n is 0 or 1 ; provided that n is not 0 if A1 is a bond, O, -N(R8)-, or S(O)m;
m is 0, 1 or 2;
p is 0, 1, 2, 3 or 4; and
q is 0 or 1 ; or an optical isomer or pharmaceutically acceptable salt thereof.
6. The compound according to Claim 4, which inhibits farnesyl-protein transferase, of the formula E:
Figure imgf000168_0001
wherein:
R1 a and R1 c are independently selected from: hydrogen, R8O-,
-N(R8)2, F, C3-C10 cycloalkyl or C 1 -C6 alkyl;
R1b is independently selected from:
a) hydrogen,
b) aryl, heterocycle, C3-C10 cycloalkyl, R8O-, -N(R8)2, F or C2-C6 alkenyl,
c) C1-C6 alkyl unsubstituted or substituted by aryl,
heterocycle, C3-C10 cycloalkyl, C2-C6 alkenyl, R8O-, or -N(R8)2; R2 is selected from: H; unsubstituted or substituted C1 -8 alkyl, unsubstituted or substituted aryl,
Figure imgf000168_0002
,
Figure imgf000168_0003
and -S(O)2R6, wherein the substituted group is substituted with one or more of:
Figure imgf000168_0004
Figure imgf000169_0001
Figure imgf000170_0003
or
R 3 is selected from: H:
Figure imgf000170_0001
Figure imgf000170_0002
,
R4 is independently selected from:
a) hydrogen,
b) aryl, substituted aryl, heterocycle, substituted heterocycle, C1 -C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 perfluoroalkyl, F, Cl, R8O-, R8C(O)NR8-, CN, NO2, (R8)2N-C(NR8)-, R8C(O)-, -N(R8)2, or R9OC(O)NR8-, and
c) C1 -C6 alkyl substituted by C1-C6 perfluoroalkyl, R8O-, R8C(O)NR8-, (R8)2N-C(NR8)-, R8C(O)-, -N(R8)2, or R9OC(O)NR8-;
R5a and R5b are independently hydrogen, ethyl, cyclopropyl or methyl; R6, R7 and R7a are independently selected from:
H; C1 -4 alkyl, C3-6 cycloalkyl, aryl, heterocycle,
unsubstituted or substituted with:
a) C1 -4 alkoxy,
b) halogen, or
c) substituted or unsubstituted aryl or substituted or unsubstituted heterocycle; R8 is independently selected from hydrogen, C1 -C6 alkyl, 2,2,2- trifluoroethyl, benzyl and aryl; R9 is independently selected from C1 -C6 alkyl and aryl;
R10 is selected from: H; R8C(O)-; R9S(O)m-; unsubstituted or substituted C1 -4 alkyl, wherein the substituted alkyl group is substituted with one or two substituents selected from:
Figure imgf000171_0001
X is a bond, -C(=O)NR10-, -NR10C(=O)-, -S(O)m- or -NR10-; n is 0 or 1;
m is 0, 1 or 2;
p is 0, 1, 2, 3 or 4, provided that p is not 0 if X is a bond,
-NR8- or O; and
q is 0 or 1; or an optical isomer or pharmaceutically acceptable salt thereof.
7. The compound according to Claim 5, which inhibits farnesyl-protein transferase, of the formula F:
Figure imgf000172_0001
wherein: R1 a and R1 c are independently selected from: hydrogen, C3-C10
cycloalkyl or C1 -C6 alkyl;
R1 b is independently selected from:
a) hydrogen,
b) aryl, heterocycle, C3-C10 cycloalkyl, R8O-, -N(R8)2 or F, c) C1 -C6 alkyl unsubstituted or substituted by aryl,
heterocycle, C3-C10 cycloalkyl, R8O-, or -N(R8)2;
R2 is selected from: H; unsubstituted or substituted C1 -8 alkyl, unsubstituted or substituted aryl,
Figure imgf000172_0002
,
Figure imgf000172_0003
and -S(O)2R6, wherein the substituted group is substituted with one or more of:
Figure imgf000172_0004
Figure imgf000173_0001
or ,
R3 is selected from: H;
Figure imgf000174_0001
Figure imgf000174_0002
R5a and R5b are independently hydrogen, ethyl, cyclopropyl or methyl; R6, R7 and R7a are independently selected from:
H; C1 -4 alkyl, C3 -6 cycloalkyl, aryl, heterocycle,
unsubstituted or substituted with:
a) C1 -4 alkoxy,
b) halogen, or
c) substituted or unsubstituted aryl or substituted or unsubstituted heterocycle; R8 is independently selected from hydrogen, C1 -C6 alkyl, 2,2,2- trifluoroethyl, benzyl and aryl; R9 is independently selected from C1 -C6 alkyl and aryl;
R10 is selected from: H; R8C(O)-; R9S(O)m-; unsubstituted or substituted C1 -4 alkyl, wherein the substituted alkyl group is substituted with one or two substituents selected from:
Figure imgf000174_0003
Figure imgf000175_0002
X is a bond, -C(=O)NR10-, -NR10C(=O)-, -S(O)m- or -NR10-; m is 0, 1 or 2;
p is 0, 1, 2, 3 or 4; and
q is 0 or 1 ; or an optical isomer or pharmaceutically acceptable salt thereof.
8. The compound according to Claim 6, which inhibits farnesyl-protein transferase, of the formula G:
Figure imgf000175_0001
wherein:
R1 a and R1 c are independently selected from: hydrogen, R8O-,
-N(R8)2, F, C3-C10 cycloalkyl or C1 -C6 alkyl;
R1b 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, R8O-, or -N(R8)2; R2 is selected from: H; unsubstituted or substituted C1 -8 alkyl, unsubstituted or substituted aryl,
Figure imgf000176_0001
,
Figure imgf000176_0002
and -S(O)2R6, wherein the substituted group is substituted with one or more of:
Figure imgf000176_0003
Figure imgf000177_0003
or
R3 is selecte d from: H;
Figure imgf000177_0001
Figure imgf000177_0002
,
R5a and R5a are independently hydrogen, ethyl, cyclopropyl or methyl;
R6, R7 and R7a are independently selected from:
H; C1 -4 alkyl, C3-6 cycloalkyl, aryl, heterocycle,
unsubstituted or substituted with:
a) C1 -4 alkoxy,
b) halogen, or c) substituted or unsubstituted aryl or substituted or unsubstituted heterocycle; R8 is independently selected from hydrogen, C1 -C6 alkyl, 2,2,2- trifluoroethyl, benzyl and aryl; R9 is independently selected from C1 -C6 alkyl and aryl;
R10 is selected from: H; R8C(O)-; R9S(O)m-; unsubstituted or substituted C1 -4 alkyl, wherein the substituted alkyl group is substituted with one or two substituents selected from:
Figure imgf000178_0001
A 1 is selected from: a bond, -C(O)-, O, -N(R8)-, or S(O)m;
X is a bond, -C(=O)NR10-, -NR10C(=O)-, -S(O)m- or -NR10-; m is 0, 1 or 2;
n is 0 or 1 ;
p is 1, 2 or 3; and q is 0 or 1 ; or an optical isomer or pharmaceutically acceptable salt thereof.
9. A compound which inhibits farnesyl-protein transferase which is:
1-(t-Butoxycarbonyl)-cis-3-methoxycarbonyl-5-[N-(1-(4-cyanobenzyl-1H-imidazol-5-ylethyl)carbamoyl] piperidine
1-Phenethyl-cis-3-methoxycarbonyl-5-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
1-(1 -Naphthylmethyl)-cis-3-methoxycarbonyl-5-[N-(1-(4-cyanobenzyl-1H-imidazol-5-ylethyl)carbamoyl] piperidine
1-Benzyl-cis-3-methoxycarbonyl-5-[N-(1-(4-cyanobenzyl-1H-imidazol-5-ylethyl)carbamoyl] piperidine 1-Methyl-cis-3-methoxycarbonyl-5-[N-(1 -(4-cyanobenzyl-1H-imidazol-5-ylethyl)carbamoyl] piperidine
1-(2-Indanyl)-cis-3-methoxycarbonyl-5-[N-(1-(4-cyanobenzyl-1H-imidazol-5-ylethyl)carbamoyl] piperidine
1-(2-Diphenylethyl)-cis-3-methoxycarbonyl-5-[N-(1-(4-cyanobenzyl-1H-imidazol-5-ylethyl)carbamoyl] piperidine
1-(3-Phenylpropyl)-cis-3-methoxycarbonyl-5-[N-(1-(4-cyanobenzyl-1H-imidazol-5-ylethyl)carbamoyl] piperidine
1-(2-Methylpropyl)-cis-3-methoxycarbonyl-5-[N-(1-(4-cyanobenzyl-1H-imidazol-5-ylethyl)carbamoyl] piperidine 1-Phenethyl-cis-3-carboxyl-5-[N-(1-(4-cyanobenzyl-1H-imidazol-5-ylethyl)carbamoyl] piperidine 1-Phenethyl-cis-3-[N-(1-morpholinyl)carbamyl]-5-[N-(1-(4-cyanobenzyl-1H-imidazol-5-ylethyl)carbamoyl]piperidine 1-Phenethyl-cis-3-[N-(benzyl)carbamyl]-5-[N-(1-(4-cyanobenzyl-1H-imidazol-5-ylethyl)carbamoyl] piperidine
1-Phenethyl-cis-3-[N-(cyclopropyl)carbamyl]-5-[N-(1-(4-cyanobenzyl-1H-imidazol-5-ylethyl)carbamoyl] piperidine
1-Phenethyl-cis-3-[N-(t-butyl)carbamyl]-5-[N-(1-(4-cyanobenzyl-1 H-imidazol-5-ylethyl)carbamoyl] piperidine
1-(2,2-Diphenylethyl)-cis-3-[N-(1-morpholinyl)carbamyl]-5-[N-(1-(4-cyanobenzyl-1H-imidazol-5-ylethyl)carbamoyl] piperidine
1 -(2,2-Diphenylethyl)-cis-3-[N-(t-butyl)carbamyl]-5-[N-(1-(4-cyanobenzyl-1H-imidazol-5-ylethyl)carbamoyl] piperidine N-[1 -Phenethyl-cis-5-(N'-(4-cyanobenzyl- 1-imidazol-5-ylethyl)carbamyl) piperidine-3-carbonyl] methionine methyl ester
N-[1-Phenethyl-cis-5-(N'-(4-cyanobenzyl- 1-imidazol-5-ylethyl)carbamyl) piperidine-3-carbonyl] methionine
1-(t-Butoxycarbonyl)-cis-3-methoxycarbonyl-5-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylacetyl)amino] piperidine
1-Phenethyl-cis-3-methoxycarbonyl-5-[N-( 1 -(4-cyanobenzyl)-1H-imidazol-5-ylacetyl)amino] piperidine
1-Diphenylacetyl-cis-3-methoxycarbonyl-5-[N-(1-(4-cyanobenzyl-1H-imidazol-5-ylethyl)carbamoyl] piperidine 1-(t-Butoxycarbonyl)-trans-3-methoxycarbonyl-5-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl]-piperidine 1-(2,2-Diphenylethyl)-3-[N-1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
1-(2,2-Diphenylethyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylacetyl)amino] piperidine
1-(2,2-Diphenylethyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylpropionyl)amino] piperidine 1-(2,2-Diphenylethyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylcarbonyl)amino] piperidine
1-(Phenylacetyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
1-(Diphenylacetyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
1-(3-Chlorobenzoyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
1-(2-(3-Chlorophenyl)-2-phenylethyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine 1-(Dibenzylsuberylmethyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
1-(2-(3-Methylphenyl)-2-phenylethyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
1-(2-(3-Trifluoromethylphenyl)-2-phenylethyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
1-(2-(2-Chlorophenyl)-2-phenylethyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine 1-(2-(4-Chlorophenyl)-2-phenylethyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
1-(2-(3-Aminomethylphenyl)-2-phenylethyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
1-(2-Phenethyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine 1 -(2-Phenethyl)-3-(R)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
1-(3-Phenylpropyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
1-(2-Benzyl)-3-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
1-(2-Chlorobenzyl)-3-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
1-(3-Chlorobenzyl)-3-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine 1-(3-Chlorobenzyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
1-(2,2-Diphenyl-2-hydroxyethyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
1-(3-Methoxybenzyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
1-(3,5-Dichlorobenzyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine 1-(3-Trifluoromethoxybenzyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
1-(2,5-Dimethylbenzyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
1-(3-Trifluoromethylbenzyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine 1-(3-Bromobenzyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
1-(3-Methylbenzyl)-3(S)-[N-(1-(4-cyanobenzyl)-1-H-imidazol-5-ylethyl)carbamoyl] piperidine
1-Isobutyl-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
1-(2-Methyl-2-phenylethyl)-3-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
1-(2-(1-Morpholinyl)-2-phenylethyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine 1-(2-(1-Piperidinyl)-2-phenylethyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
1-(2,2-Diphenylethyl)-3(S)-[N-(1-(4-cyanobenzyl)-2-methyl-1H-imidazol-5-ylethyl)carbamoyl] piperidine
1-(2,2-Diphenylethyl)-3(S)-[N-(1-(4-methoxybenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
1-(Diphenylmethyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine 1-(3-Methoxyphenethyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
1-(1-Naphthylethyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
1-(3-Chlorophenethyl)-3(S)-[N-1-(4- cyanobenzy)1-1H-imidazol-5-ylethyl)carbamoyl] piperidine 1-(α-Methylbenzyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
1-(Diphenylmethyl)-3(S)-[N-(1-(4-cyanobenzyl)-2-methyl-1H-imidazol-5-ylethyl)carbamoyl] piperidine
1-(α-Toluenesulfonyl)-3(S)-[N-(1-(4-cyanobenzyl)- 1H-imidazol-5-ylethyl)carbamoyl] piperidine
1-(Benzenesulfonyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
1-(1-Naphthylenesulfonyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine 1-(3-Chlorobenzenesulfonyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
1-(3,5-Dichlorobenzenesulfonyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
1-(α-Toluenesulfonyl)-3-(R)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
1-(α-Toluenesulfonyl)-cis-3-methoxycarbonyl-5-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine 1-(Methanesulfonyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
1-(Diphenylcarbamoyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
1-(Phenylcarbamoyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine 1-[2-(2-Pyridyl)-2-phenyl-2-hydroxyethyl]-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
1-(2-Pyridylethyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
1-Phenyl-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl]piperidine
1-(3-Methylphenyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
1-(2,2-Diphenylethyl)-3(S)-[2-(1-(4-cyanobenzyl)-1H-imidazol-5-yl)ethylthiomethyl] piperidine 1-(2,2-Diphenylethyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-yl)ethylsulfonylmethyl] piperidine
1-(2,2-Diphenylethyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)-N-methyl-carbamoyl] piperidine
1-(3-Bromobenzyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)N-methyl-carbamoyl] piperidine
1 -(2,2-Diphenylethyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)aminomethyl] piperidine
1-(2,2-Diphenylethyl)-3(S)-[N-(1-(--cyanobenzyl)-1H-imidazol-5-ylethyl)-N-acetyI-aminomethyl] piperidine 1-(2,2-Diphenylethyl)-3(S)-[N-(1-(4-cyanobenzyl)-2-methyl-1H-imidazol-5-ylethyl)-N-acetyl-aminomethyl] piperidine
1-(2,2-DiphenylethyI)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)-N-cyclopropylmethyl-aminomethyl] piperidine
1-(2,2-Diphenylethyl)-3(S)-[N-(2-methyl-1H-imidazol-4-ylethyl)-N-(4-cyanobenzoyl)aminomethyl] piperidine
1-(2,2-Diphenylethyl)-3(S)-[5-(4-cyanobenzyl)-1H-imidazoi- 1-ylmethyl] piperidine
1-(2,2-Diphenylethyl)-3(S)-[5-(4-cyanobenzyl)-1H-imidazol-1-ylethylcarbamoyl] piperdine or an optical isomer or a pharmaceutically acceptable salt thereof.
10. The compound according to Claim 9 which is: 1-Phenethyl-cis-3-[N-(1-morpholinyl)carbamyl]-5-[N-(4-cyanobenzyl-1 -imidazole-5-ethyl)carbamyl]piperidine
or an optical isomer or a pharmaceutically acceptable salt thereof.
1 1. The compound according to Claim 9 which is: 1-(2-Diphenylethyl)-cis-3-[N-(1-morpholinyl)carbamyl]-5-[N-(4-cyanobenzyl-1-imidazole-5-ethyl)carbamyl] piperidine
Figure imgf000187_0001
or an optical isomer or a pharmaceutically acceptable salt thereof.
12. The compound according to Claim 9 which is:
N-[1-Phenethyl-5-(N'-(4-cyanobenzyl-1-imidazole-5-ethyl)carbamyl) piperidine-3-carbonyl] methionine
Figure imgf000188_0001
or an optical isomer or a pharmaceutically acceptable salt thereof.
13. The compound according to Claim 9 which is:
1-(2-Diphenylethyl)-3-methoxycarbonyl-5-[N-(4-cyanobenzyl-1-imidazole-5-ethyl)carbamyl] piperidine
Figure imgf000188_0002
or an optical isomer or a pharmaceutically acceptable salt thereof.
14. The compound according to Claim 9 which is:
1-(2,2-Diphenylethyl)-cis-3-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
Figure imgf000189_0001
or an optical isomer or a pharmaceutically acceptable salt thereof.
15. The compound according to Claim 9 which is:
1-(3-Chlorobenzyl)-3-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
Figure imgf000190_0001
or an optical isomer or a pharmaceutically acceptable salt thereof.
16. The compound according to Claim 9 which is:
1-(2-(1-Morpholinyl)-2-phenylethyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
Figure imgf000190_0002
or an optical isomer or a pharmaceutically acceptable salt thereof.
17. The compound according to Claim 9 which is:
1-(Diphenylmethyl)-3(S)-[N-(1-(4-cyanobenzyl)-2-methyl-1H-imidazol-5-ylethyl)carbamoyl] piperidine
Figure imgf000191_0001
or an optical isomer or a pharmaceutically acceptable salt thereof.
18. The compound according to Claim 9 which is:
1-(3-Methylphenyl)-3(S)-[N-(1-(--cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
Figure imgf000192_0002
or an optical isomer or a pharmaceutically acceptable salt thereof.
19. The compound according to Claim 9 which is:
1-(2,2-Diphenylethyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)-N-acetyl-aminomethyl] piperidine
Figure imgf000192_0001
or an optical isomer or a pharmaceutically acceptable salt thereof.
20. The compound according to Claim 9 which is: 1-(Benzenesulfonyl)-3(S)-[N-(1-(4-cyanobenzyl)-1H-imidazol-5-ylethyl)carbamoyl] piperidine
Figure imgf000193_0001
or an optical isomer or a pharmaceutically acceptable salt thereof.
21. A pharmaceutical composition comprising a pharmaceutical carrier, and dispersed therein, a therapeutically effective amount of a compound of Claim 1.
22. A pharmaceutical composition comprising a pharmaceutical carrier, and dispersed therein, a therapeutically effective amount of a compound of Claim 5.
23. A pharmaceutical composition comprising a pharmaceutical carrier, and dispersed therein, a therapeutically effective amount of a compound of Claim 6.
24. A pharmaceutical composition comprising a
pharmaceutical carrier, and dispersed therein, a therapeutically effective amount of a compound of Claim 9.
25. 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 21.
26. 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 22.
27. 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 23.
28. 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 24.
29. A method for treating cancer which comprises administering to a mammal in need thereof a therapeutically effective amount of a composition of Claim 21.
30. A method for treating cancer which comprises administering to a mammal in need thereof a therapeutically effective amount of a composition of Claim 22.
31. A method for treating cancer which comprises administering to a mammal in need thereof a therapeutically effective amount of a composition of Claim 23.
32. A method for treating cancer which comprises administering to a mammal in need thereof a therapeutically effective amount of a composition of Claim 24.
33. 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 21.
34. 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 21.
35. 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 21.
36. A method for preventing restenosis which comprises administering to a mammal in need thereof a therapeutically effective amount of a composition of Claim 21.
37. 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 21.
38. A pharmaceutical composition made by combining the compound of Claim 1 and a pharmaceutically acceptable carrier.
39. A process for making a pharmaceutical composition comprising combining a compound of Claim 1 and a pharmaceutically acceptable carrier.
PCT/US1996/018811 1995-11-22 1996-11-18 Inhibitors of farnesyl-protein transferase WO1997018813A1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
JP9519941A JP2000500502A (en) 1995-11-22 1996-11-18 Farnesyl-protein transferase inhibitor
EP96942798A EP0862435A4 (en) 1995-11-22 1996-11-18 Inhibitors of farnesyl-protein transferase
AU11626/97A AU704139B2 (en) 1995-11-22 1996-11-18 Inhibitors of farnesyl-protein transferase

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US749895P 1995-11-22 1995-11-22
US60/007,498 1995-11-22
GBGB9604311.2A GB9604311D0 (en) 1996-02-29 1996-02-29 Inhibitors of farnesyl-protein transferase
GB9604311.2 1996-02-29

Publications (1)

Publication Number Publication Date
WO1997018813A1 true WO1997018813A1 (en) 1997-05-29

Family

ID=26308829

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1996/018811 WO1997018813A1 (en) 1995-11-22 1996-11-18 Inhibitors of farnesyl-protein transferase

Country Status (5)

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

Cited By (86)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1997038665A2 (en) * 1996-04-03 1997-10-23 Merck & Co., Inc. Inhibitors of farnesyl-protein transferase
EP0880320A1 (en) * 1996-01-30 1998-12-02 Merck & Co., Inc. Inhibitors of farnesyl-protein transferase
US5891889A (en) * 1996-04-03 1999-04-06 Merck & Co., Inc. Inhibitors of farnesyl-protein transferase
WO1999027933A1 (en) * 1997-12-04 1999-06-10 Merck & Co., Inc. Inhibitors of farnesyl-protein transferase
US5932590A (en) * 1996-12-05 1999-08-03 Merck & Co., Inc. Inhibitors of farnesyl-protein transferase
US5958940A (en) * 1997-09-11 1999-09-28 Schering Corporation Tricyclic compounds useful as inhibitors of farnesyl-protein transferase
US5965570A (en) * 1996-09-13 1999-10-12 Schering Corporation Tricyclic piperidinyl compounds useful as inhibitors of farnesyl-protein transferase
US5977134A (en) * 1996-12-05 1999-11-02 Merck & Co., Inc. Inhibitors of farnesyl-protein transferase
FR2780892A1 (en) * 1998-07-08 2000-01-14 Sod Conseils Rech Applic USE OF PRENYLTRANSFERASE INHIBITORS FOR THE PREPARATION OF A MEDICINAL PRODUCT FOR TREATING CONDITIONS RESULTING FROM MEMBRANE FIXATION OF HETEROTRIMERIC PROTEIN
US6015817A (en) * 1996-12-05 2000-01-18 Merck & Co., Inc. Inhibitors of farnesyl-protein transferase
EP1035849A1 (en) * 1997-12-04 2000-09-20 Merck & Co., Inc. Inhibitors of farnesyl-protein transferase
US6271197B1 (en) 1996-04-11 2001-08-07 Gpc-Biotech Inc. Assays and reagents for identifying anti-fungal agents, and uses related thereto
WO2001092226A1 (en) * 2000-05-25 2001-12-06 Sepracor, Inc. Heterocyclic analgesic compounds and method of use thereof
US6432959B1 (en) 1998-12-23 2002-08-13 Schering Corporation Inhibitors of farnesyl-protein transferase
US6455281B1 (en) 1996-04-11 2002-09-24 Gpc Biotech Inc. Nucleic acids for identifying anti-fungal agents, and uses related thereto
WO2003066600A1 (en) * 2002-02-08 2003-08-14 Yuhan Corporation Processes for preparing imidazole derivatives and salts thereof
US6627629B2 (en) 2000-06-30 2003-09-30 Bristol-Myers Squibb Pharma N-ureidoheterocycloalkyl-piperidines as modulators of chemokine receptor activity
US6635661B2 (en) 2000-05-25 2003-10-21 Sepracor Inc. Heterocyclic analgesic compounds and methods of use thereof
US6696280B2 (en) 1996-04-11 2004-02-24 Gpc Biotech, Inc. Candida geranylgeranyl-protein transferase polypetide, compositions and methods related thereto
US6818637B2 (en) * 1999-01-11 2004-11-16 Aventis Pharma Sa Polyhydroxypyrazine derivatives, their preparation and the pharmaceutical compositions which comprise them
US6946477B2 (en) 2000-03-14 2005-09-20 Sepracor Inc. 3-Substituted piperidines comprising urea functionality, and methods of use thereof
US7129228B2 (en) 1999-05-25 2006-10-31 Sepracor Inc. Heterocyclic analgesic compounds and methods of use thereof
WO2006123182A2 (en) 2005-05-17 2006-11-23 Merck Sharp & Dohme Limited Cyclohexyl sulphones for treatment of cancer
US7148230B2 (en) 2003-07-29 2006-12-12 Astrazeneca Ab Quinazoline derivatives
WO2007077005A1 (en) 2005-12-30 2007-07-12 Novartis Ag 3 , 5-substitγued piperidine compounds as renin inhibitors
WO2007093827A1 (en) 2006-02-15 2007-08-23 Istituto Di Ricerche Di Biologia Molecolare P. Angeletti Spa Thiophene and thiazole substituted trifluoroethanone derivatives as histone deacetylase (hdac) inhibitors
US7361666B2 (en) 1999-05-25 2008-04-22 Sepracor, Inc. Heterocyclic analgesic compounds and methods of use thereof
US7378436B2 (en) 2003-06-17 2008-05-27 Pfizer Inc. Compounds
WO2008093737A1 (en) 2007-01-31 2008-08-07 Dainippon Sumitomo Pharma Co., Ltd. Amide derivative
WO2008106692A1 (en) 2007-03-01 2008-09-04 Novartis Vaccines And Diagnostics, Inc. Pim kinase inhibitors and methods of their use
WO2008144062A1 (en) 2007-05-21 2008-11-27 Novartis Ag Csf-1r inhibitors, compositions, and methods of use
WO2009002495A1 (en) 2007-06-27 2008-12-31 Merck & Co., Inc. 4-carboxybenzylamino derivatives as histone deacetylase inhibitors
US7569577B2 (en) 2003-09-16 2009-08-04 Astrazeneca Ab Quinazoline derivatives as tyrosine kinase inhibitors
WO2010114780A1 (en) 2009-04-01 2010-10-07 Merck Sharp & Dohme Corp. Inhibitors of akt activity
WO2010150840A1 (en) 2009-06-24 2010-12-29 大日本住友製薬株式会社 N-substituted-cyclic amino derivative
US7910731B2 (en) 2002-03-30 2011-03-22 Boehringer Ingelheim Pharma Gmbh & Co. Kg Bicyclic heterocyclic compounds, pharmaceutical compositions containing these compounds, their use and process for preparing them
WO2011046771A1 (en) 2009-10-14 2011-04-21 Schering Corporation SUBSTITUTED PIPERIDINES THAT INCREASE p53 ACTIVITY AND THE USES THEREOF
EP2336120A1 (en) 2007-01-10 2011-06-22 Istituto di ricerche di Biologia Molecolare P. Angeletti S.R.L. Combinations containing amide substituted indazoles as poly(ADP-ribose)polymerase (PARP) inhibitors
US7998949B2 (en) 2007-02-06 2011-08-16 Boehringer Ingelheim International Gmbh Bicyclic heterocycles, drugs containing said compounds, use thereof, and method for production thereof
WO2011115725A2 (en) 2010-03-16 2011-09-22 Dana-Farber Cancer Institute, Inc. Indazole compounds and their uses
WO2011163330A1 (en) 2010-06-24 2011-12-29 Merck Sharp & Dohme Corp. Novel heterocyclic compounds as erk inhibitors
US8088782B2 (en) 2008-05-13 2012-01-03 Astrazeneca Ab Crystalline 4-(3-chloro-2-fluoroanilino)-7 methoxy-6-{[1-(N-methylcarbamoylmethyl)piperidin-4-yl]oxy}quinazoline difumarate form A
WO2012018754A2 (en) 2010-08-02 2012-02-09 Merck Sharp & Dohme Corp. RNA INTERFERENCE MEDIATED INHIBITION OF CATENIN (CADHERIN-ASSOCIATED PROTEIN), BETA 1 (CTNNB1) GENE EXPRESSION USING SHORT INTERFERING NUCLEIC ACID (siNA)
WO2012024170A2 (en) 2010-08-17 2012-02-23 Merck Sharp & Dohme Corp. RNA INTERFERENCE MEDIATED INHIBITION OF HEPATITIS B VIRUS (HBV) GENE EXPRESSION USING SHORT INTERFERING NUCLEIC ACID (siNA)
WO2012027236A1 (en) 2010-08-23 2012-03-01 Schering Corporation NOVEL PYRAZOLO[1,5-a]PYRIMIDINE DERIVATIVES AS mTOR INHIBITORS
WO2012030685A2 (en) 2010-09-01 2012-03-08 Schering Corporation Indazole derivatives useful as erk inhibitors
WO2012036997A1 (en) 2010-09-16 2012-03-22 Schering Corporation Fused pyrazole derivatives as novel erk inhibitors
WO2012058210A1 (en) 2010-10-29 2012-05-03 Merck Sharp & Dohme Corp. RNA INTERFERENCE MEDIATED INHIBITION OF GENE EXPRESSION USING SHORT INTERFERING NUCLEIC ACIDS (siNA)
WO2012087772A1 (en) 2010-12-21 2012-06-28 Schering Corporation Indazole derivatives useful as erk inhibitors
WO2012145471A1 (en) 2011-04-21 2012-10-26 Merck Sharp & Dohme Corp. Insulin-like growth factor-1 receptor inhibitors
US8383650B2 (en) 2007-06-25 2013-02-26 Novartis Ag Organic compounds
US8399461B2 (en) 2006-11-10 2013-03-19 Boehringer Ingelheim International Gmbh Bicyclic heterocycles, medicaments containing said compounds, use thereof, and method for production of same
WO2013063214A1 (en) 2011-10-27 2013-05-02 Merck Sharp & Dohme Corp. Novel compounds that are erk inhibitors
US8497369B2 (en) 2008-02-07 2013-07-30 Boehringer Ingelheim International Gmbh Spirocyclic heterocycles medicaments containing said compounds, use thereof and method for their production
WO2013165816A2 (en) 2012-05-02 2013-11-07 Merck Sharp & Dohme Corp. SHORT INTERFERING NUCLEIC ACID (siNA) COMPOSITIONS
US8648191B2 (en) 2008-08-08 2014-02-11 Boehringer Ingelheim International Gmbh Cyclohexyloxy substituted heterocycles, pharmaceutical compositions containing these compounds and processes for preparing them
EP2698157A1 (en) 2006-09-22 2014-02-19 Merck Sharp & Dohme Corp. Method of treatment using fatty acid synthesis inhibitors
WO2014052563A2 (en) 2012-09-28 2014-04-03 Merck Sharp & Dohme Corp. Novel compounds that are erk inhibitors
WO2014085216A1 (en) 2012-11-28 2014-06-05 Merck Sharp & Dohme Corp. Compositions and methods for treating cancer
WO2014100065A1 (en) 2012-12-20 2014-06-26 Merck Sharp & Dohme Corp. Substituted imidazopyridines as hdm2 inhibitors
US8765747B2 (en) 2009-06-12 2014-07-01 Dana-Farber Cancer Institute, Inc. Fused 2-aminothiazole compounds
WO2014120748A1 (en) 2013-01-30 2014-08-07 Merck Sharp & Dohme Corp. 2,6,7,8 substituted purines as hdm2 inhibitors
WO2015034925A1 (en) 2013-09-03 2015-03-12 Moderna Therapeutics, Inc. Circular polynucleotides
WO2016020864A1 (en) 2014-08-06 2016-02-11 Novartis Ag Protein kinase c inhibitors and methods of their use
US9758522B2 (en) 2012-10-19 2017-09-12 Dana-Farber Cancer Institute, Inc. Hydrophobically tagged small molecules as inducers of protein degradation
US9862688B2 (en) 2014-04-23 2018-01-09 Dana-Farber Cancer Institute, Inc. Hydrophobically tagged janus kinase inhibitors and uses thereof
WO2018058022A1 (en) 2016-09-26 2018-03-29 Merck Sharp & Dohme Corp. Anti-cd27 antibodies
US10000483B2 (en) 2012-10-19 2018-06-19 Dana-Farber Cancer Institute, Inc. Bone marrow on X chromosome kinase (BMX) inhibitors and uses thereof
US10017477B2 (en) 2014-04-23 2018-07-10 Dana-Farber Cancer Institute, Inc. Janus kinase inhibitors and uses thereof
WO2018190719A2 (en) 2017-04-13 2018-10-18 Aduro Biotech Holdings, Europe B.V. Anti-sirp alpha antibodies
US10112927B2 (en) 2012-10-18 2018-10-30 Dana-Farber Cancer Institute, Inc. Inhibitors of cyclin-dependent kinase 7 (CDK7)
US10144730B2 (en) 2011-11-17 2018-12-04 Dana-Farber Cancer Institute, Inc. Inhibitors of c-Jun-N-terminal kinase (JNK)
WO2019094311A1 (en) 2017-11-08 2019-05-16 Merck Sharp & Dohme Corp. Prmt5 inhibitors
WO2019152642A1 (en) 2018-02-01 2019-08-08 Merck Sharp & Dohme Corp. Anti-pd-1/lag3 bispecific antibodies
US10450269B1 (en) 2013-11-18 2019-10-22 Global Blood Therapeutics, Inc. Compounds and uses thereof for the modulation of hemoglobin
US10550121B2 (en) 2015-03-27 2020-02-04 Dana-Farber Cancer Institute, Inc. Inhibitors of cyclin-dependent kinases
WO2020033282A1 (en) 2018-08-07 2020-02-13 Merck Sharp & Dohme Corp. Prmt5 inhibitors
WO2020033284A1 (en) 2018-08-07 2020-02-13 Merck Sharp & Dohme Corp. Prmt5 inhibitors
US10702527B2 (en) 2015-06-12 2020-07-07 Dana-Farber Cancer Institute, Inc. Combination therapy of transcription inhibitors and kinase inhibitors
US10870651B2 (en) 2014-12-23 2020-12-22 Dana-Farber Cancer Institute, Inc. Inhibitors of cyclin-dependent kinase 7 (CDK7)
US10906889B2 (en) 2013-10-18 2021-02-02 Dana-Farber Cancer Institute, Inc. Polycyclic inhibitors of cyclin-dependent kinase 7 (CDK7)
US11040957B2 (en) 2013-10-18 2021-06-22 Dana-Farber Cancer Institute, Inc. Heteroaromatic compounds useful for the treatment of proliferative diseases
US11053195B2 (en) 2013-03-15 2021-07-06 Global Blood Therapeutics, Inc. Compounds and uses thereof for the modulation of hemoglobin
US11096950B2 (en) 2006-11-01 2021-08-24 Barbara Brooke Jennings Compounds, methods, and treatments for abnormal signaling pathways for prenatal and postnatal development
US11142507B2 (en) 2015-09-09 2021-10-12 Dana-Farber Cancer Institute, Inc. Inhibitors of cyclin-dependent kinases
US11826365B2 (en) 2009-12-29 2023-11-28 Dana-Farber Cancer Institute, Inc. Type II raf kinase inhibitors

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3038835A (en) * 1959-07-01 1962-06-12 Bofors Ab Method for the production of lowtoxic surface anaesthetics
US5476942A (en) * 1993-09-14 1995-12-19 Synthelabo 1-[2-amino-5-[1-(triphenylmethyl)-1H-imidazol-4-YL]-1-oxopentyl] piperidine derivatives, their preparation and their use as synthetic intermediates

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR2579596B1 (en) * 1985-03-26 1987-11-20 Inst Nat Sante Rech Med (IMIDAZOLYL-4) PIPERIDINES, THEIR PREPARATION AND THEIR THERAPEUTIC APPLICATION
WO1991009030A1 (en) * 1989-12-12 1991-06-27 Rhone-Poulenc Sante 2-substituted 4,5-diphenyl-imidazoles
DK0619818T3 (en) * 1991-12-18 1996-11-25 Schering Corp Imidazoylalkyl substituted with a six-membered nitrogen-containing heterocyclic ring
GB9312806D0 (en) * 1993-06-22 1993-08-04 Boots Co Plc Therapeutic agents
RU95104898A (en) * 1994-03-31 1996-12-27 Бристоль-Мейерз Сквибб Компани (US) Imedazole containing inhibitors of ferneside proteintansferase, and method of treatment diseases related therewith

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3038835A (en) * 1959-07-01 1962-06-12 Bofors Ab Method for the production of lowtoxic surface anaesthetics
US5476942A (en) * 1993-09-14 1995-12-19 Synthelabo 1-[2-amino-5-[1-(triphenylmethyl)-1H-imidazol-4-YL]-1-oxopentyl] piperidine derivatives, their preparation and their use as synthetic intermediates

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See also references of EP0862435A4 *

Cited By (115)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0880320A1 (en) * 1996-01-30 1998-12-02 Merck & Co., Inc. Inhibitors of farnesyl-protein transferase
EP0880320A4 (en) * 1996-01-30 1999-06-16 Merck & Co Inc Inhibitors of farnesyl-protein transferase
WO1997038665A3 (en) * 1996-04-03 1997-11-27 Merck & Co Inc Inhibitors of farnesyl-protein transferase
US5891889A (en) * 1996-04-03 1999-04-06 Merck & Co., Inc. Inhibitors of farnesyl-protein transferase
WO1997038665A2 (en) * 1996-04-03 1997-10-23 Merck & Co., Inc. Inhibitors of farnesyl-protein transferase
US6727082B1 (en) 1996-04-11 2004-04-27 Gpc Biotech Inc. Assays and reagents for identifying anti-fungal agents, and uses related thereto
US6271197B1 (en) 1996-04-11 2001-08-07 Gpc-Biotech Inc. Assays and reagents for identifying anti-fungal agents, and uses related thereto
US6455281B1 (en) 1996-04-11 2002-09-24 Gpc Biotech Inc. Nucleic acids for identifying anti-fungal agents, and uses related thereto
US6277564B1 (en) 1996-04-11 2001-08-21 Gpc Biotech Inc. Assays and reagents for identifying anti-fungal agents, and uses related thereto
US6696280B2 (en) 1996-04-11 2004-02-24 Gpc Biotech, Inc. Candida geranylgeranyl-protein transferase polypetide, compositions and methods related thereto
US5965570A (en) * 1996-09-13 1999-10-12 Schering Corporation Tricyclic piperidinyl compounds useful as inhibitors of farnesyl-protein transferase
US5977134A (en) * 1996-12-05 1999-11-02 Merck & Co., Inc. Inhibitors of farnesyl-protein transferase
US6015817A (en) * 1996-12-05 2000-01-18 Merck & Co., Inc. Inhibitors of farnesyl-protein transferase
US5972966A (en) * 1996-12-05 1999-10-26 Merck & Co., Inc. Inhibitors of farnesyl-protein transferase
US5932590A (en) * 1996-12-05 1999-08-03 Merck & Co., Inc. Inhibitors of farnesyl-protein transferase
US5958940A (en) * 1997-09-11 1999-09-28 Schering Corporation Tricyclic compounds useful as inhibitors of farnesyl-protein transferase
EP1045844A4 (en) * 1997-12-04 2002-08-21 Merck & Co Inc Inhibitors of farnesyl-protein transferase
EP1035849A1 (en) * 1997-12-04 2000-09-20 Merck & Co., Inc. Inhibitors of farnesyl-protein transferase
EP1045844A1 (en) * 1997-12-04 2000-10-25 Merck & Co., Inc. Inhibitors of farnesyl-protein transferase
EP1035849A4 (en) * 1997-12-04 2001-09-12 Merck & Co Inc Inhibitors of farnesyl-protein transferase
WO1999027933A1 (en) * 1997-12-04 1999-06-10 Merck & Co., Inc. Inhibitors of farnesyl-protein transferase
FR2780892A1 (en) * 1998-07-08 2000-01-14 Sod Conseils Rech Applic USE OF PRENYLTRANSFERASE INHIBITORS FOR THE PREPARATION OF A MEDICINAL PRODUCT FOR TREATING CONDITIONS RESULTING FROM MEMBRANE FIXATION OF HETEROTRIMERIC PROTEIN
US6432959B1 (en) 1998-12-23 2002-08-13 Schering Corporation Inhibitors of farnesyl-protein transferase
US6818637B2 (en) * 1999-01-11 2004-11-16 Aventis Pharma Sa Polyhydroxypyrazine derivatives, their preparation and the pharmaceutical compositions which comprise them
US7361666B2 (en) 1999-05-25 2008-04-22 Sepracor, Inc. Heterocyclic analgesic compounds and methods of use thereof
US7129228B2 (en) 1999-05-25 2006-10-31 Sepracor Inc. Heterocyclic analgesic compounds and methods of use thereof
US6946477B2 (en) 2000-03-14 2005-09-20 Sepracor Inc. 3-Substituted piperidines comprising urea functionality, and methods of use thereof
US6645980B1 (en) 2000-05-25 2003-11-11 Sepracor Inc. Heterocyclic analgesic compounds and methods of use thereof
US6635661B2 (en) 2000-05-25 2003-10-21 Sepracor Inc. Heterocyclic analgesic compounds and methods of use thereof
WO2001092226A1 (en) * 2000-05-25 2001-12-06 Sepracor, Inc. Heterocyclic analgesic compounds and method of use thereof
US6627629B2 (en) 2000-06-30 2003-09-30 Bristol-Myers Squibb Pharma N-ureidoheterocycloalkyl-piperidines as modulators of chemokine receptor activity
US6949546B2 (en) 2000-06-30 2005-09-27 Bristol-Myers Squibb Pharma Company N-ureidoheterocycloalkyl-piperidines as modulators of chemokine receptor activity
WO2003066600A1 (en) * 2002-02-08 2003-08-14 Yuhan Corporation Processes for preparing imidazole derivatives and salts thereof
US6706888B2 (en) 2002-02-08 2004-03-16 Yuhan Corporation Processes for preparing imidazole derivatives and salts thereof
US8343982B2 (en) 2002-03-30 2013-01-01 Boehringer Ingelheim Pharma Gmbh & Co. Kg Bicyclic heterocyclic compounds pharmaceutical compositions containing these compounds, their use and process for preparing the same
US7910731B2 (en) 2002-03-30 2011-03-22 Boehringer Ingelheim Pharma Gmbh & Co. Kg Bicyclic heterocyclic compounds, pharmaceutical compositions containing these compounds, their use and process for preparing them
US7378436B2 (en) 2003-06-17 2008-05-27 Pfizer Inc. Compounds
US7148230B2 (en) 2003-07-29 2006-12-12 Astrazeneca Ab Quinazoline derivatives
US7569577B2 (en) 2003-09-16 2009-08-04 Astrazeneca Ab Quinazoline derivatives as tyrosine kinase inhibitors
WO2006123182A2 (en) 2005-05-17 2006-11-23 Merck Sharp & Dohme Limited Cyclohexyl sulphones for treatment of cancer
WO2007077005A1 (en) 2005-12-30 2007-07-12 Novartis Ag 3 , 5-substitγued piperidine compounds as renin inhibitors
US8129411B2 (en) 2005-12-30 2012-03-06 Novartis Ag Organic compounds
WO2007093827A1 (en) 2006-02-15 2007-08-23 Istituto Di Ricerche Di Biologia Molecolare P. Angeletti Spa Thiophene and thiazole substituted trifluoroethanone derivatives as histone deacetylase (hdac) inhibitors
EP2698157A1 (en) 2006-09-22 2014-02-19 Merck Sharp & Dohme Corp. Method of treatment using fatty acid synthesis inhibitors
EP2946778A1 (en) 2006-09-22 2015-11-25 Merck Sharp & Dohme Corp. Method of treatment using fatty acid synthesis inhibitors
US11096950B2 (en) 2006-11-01 2021-08-24 Barbara Brooke Jennings Compounds, methods, and treatments for abnormal signaling pathways for prenatal and postnatal development
US8399461B2 (en) 2006-11-10 2013-03-19 Boehringer Ingelheim International Gmbh Bicyclic heterocycles, medicaments containing said compounds, use thereof, and method for production of same
EP2805945A1 (en) 2007-01-10 2014-11-26 MSD Italia S.r.l. Amide substituted indazoles as poly(ADP-ribose)polymerase (PARP) inhibitors
EP2336120A1 (en) 2007-01-10 2011-06-22 Istituto di ricerche di Biologia Molecolare P. Angeletti S.R.L. Combinations containing amide substituted indazoles as poly(ADP-ribose)polymerase (PARP) inhibitors
WO2008093737A1 (en) 2007-01-31 2008-08-07 Dainippon Sumitomo Pharma Co., Ltd. Amide derivative
US7998949B2 (en) 2007-02-06 2011-08-16 Boehringer Ingelheim International Gmbh Bicyclic heterocycles, drugs containing said compounds, use thereof, and method for production thereof
WO2008106692A1 (en) 2007-03-01 2008-09-04 Novartis Vaccines And Diagnostics, Inc. Pim kinase inhibitors and methods of their use
WO2008144062A1 (en) 2007-05-21 2008-11-27 Novartis Ag Csf-1r inhibitors, compositions, and methods of use
US8383650B2 (en) 2007-06-25 2013-02-26 Novartis Ag Organic compounds
US8497286B2 (en) 2007-06-25 2013-07-30 Novartis Ag Organic compounds
WO2009002495A1 (en) 2007-06-27 2008-12-31 Merck & Co., Inc. 4-carboxybenzylamino derivatives as histone deacetylase inhibitors
EP3103791A1 (en) 2007-06-27 2016-12-14 Merck Sharp & Dohme Corp. 4-carboxybenzylamino derivatives as histone deacetylase inhibitors
US8497369B2 (en) 2008-02-07 2013-07-30 Boehringer Ingelheim International Gmbh Spirocyclic heterocycles medicaments containing said compounds, use thereof and method for their production
US8772298B2 (en) 2008-02-07 2014-07-08 Boehringer Ingelheim International Gmbh Spirocyclic heterocycles medicaments containing said compounds, use thereof and method for their production
US8088782B2 (en) 2008-05-13 2012-01-03 Astrazeneca Ab Crystalline 4-(3-chloro-2-fluoroanilino)-7 methoxy-6-{[1-(N-methylcarbamoylmethyl)piperidin-4-yl]oxy}quinazoline difumarate form A
US8648191B2 (en) 2008-08-08 2014-02-11 Boehringer Ingelheim International Gmbh Cyclohexyloxy substituted heterocycles, pharmaceutical compositions containing these compounds and processes for preparing them
WO2010114780A1 (en) 2009-04-01 2010-10-07 Merck Sharp & Dohme Corp. Inhibitors of akt activity
US9505784B2 (en) 2009-06-12 2016-11-29 Dana-Farber Cancer Institute, Inc. Fused 2-aminothiazole compounds
US8765747B2 (en) 2009-06-12 2014-07-01 Dana-Farber Cancer Institute, Inc. Fused 2-aminothiazole compounds
WO2010150840A1 (en) 2009-06-24 2010-12-29 大日本住友製薬株式会社 N-substituted-cyclic amino derivative
WO2011046771A1 (en) 2009-10-14 2011-04-21 Schering Corporation SUBSTITUTED PIPERIDINES THAT INCREASE p53 ACTIVITY AND THE USES THEREOF
US11826365B2 (en) 2009-12-29 2023-11-28 Dana-Farber Cancer Institute, Inc. Type II raf kinase inhibitors
WO2011115725A2 (en) 2010-03-16 2011-09-22 Dana-Farber Cancer Institute, Inc. Indazole compounds and their uses
US8987275B2 (en) 2010-03-16 2015-03-24 Dana-Farber Cancer Institute, Inc. Indazole compounds and their uses
WO2011163330A1 (en) 2010-06-24 2011-12-29 Merck Sharp & Dohme Corp. Novel heterocyclic compounds as erk inhibitors
EP3330377A1 (en) 2010-08-02 2018-06-06 Sirna Therapeutics, Inc. Rna interference mediated inhibition of catenin (cadherin-associated protein), beta 1 (ctnnb1) gene expression using short interfering nucleic acid (sina)
WO2012018754A2 (en) 2010-08-02 2012-02-09 Merck Sharp & Dohme Corp. RNA INTERFERENCE MEDIATED INHIBITION OF CATENIN (CADHERIN-ASSOCIATED PROTEIN), BETA 1 (CTNNB1) GENE EXPRESSION USING SHORT INTERFERING NUCLEIC ACID (siNA)
EP4079856A1 (en) 2010-08-17 2022-10-26 Sirna Therapeutics, Inc. Rna interference mediated inhibition of hepatitis b virus (hbv) gene expression using short interfering nucleic acid (sina)
WO2012024170A2 (en) 2010-08-17 2012-02-23 Merck Sharp & Dohme Corp. RNA INTERFERENCE MEDIATED INHIBITION OF HEPATITIS B VIRUS (HBV) GENE EXPRESSION USING SHORT INTERFERING NUCLEIC ACID (siNA)
WO2012027236A1 (en) 2010-08-23 2012-03-01 Schering Corporation NOVEL PYRAZOLO[1,5-a]PYRIMIDINE DERIVATIVES AS mTOR INHIBITORS
WO2012030685A2 (en) 2010-09-01 2012-03-08 Schering Corporation Indazole derivatives useful as erk inhibitors
WO2012036997A1 (en) 2010-09-16 2012-03-22 Schering Corporation Fused pyrazole derivatives as novel erk inhibitors
EP3327125A1 (en) 2010-10-29 2018-05-30 Sirna Therapeutics, Inc. Rna interference mediated inhibition of gene expression using short interfering nucleic acids (sina)
EP3766975A1 (en) 2010-10-29 2021-01-20 Sirna Therapeutics, Inc. Rna interference mediated inhibition of gene expression using short interfering nucleic acid (sina)
WO2012058210A1 (en) 2010-10-29 2012-05-03 Merck Sharp & Dohme Corp. RNA INTERFERENCE MEDIATED INHIBITION OF GENE EXPRESSION USING SHORT INTERFERING NUCLEIC ACIDS (siNA)
WO2012087772A1 (en) 2010-12-21 2012-06-28 Schering Corporation Indazole derivatives useful as erk inhibitors
WO2012145471A1 (en) 2011-04-21 2012-10-26 Merck Sharp & Dohme Corp. Insulin-like growth factor-1 receptor inhibitors
WO2013063214A1 (en) 2011-10-27 2013-05-02 Merck Sharp & Dohme Corp. Novel compounds that are erk inhibitors
US10144730B2 (en) 2011-11-17 2018-12-04 Dana-Farber Cancer Institute, Inc. Inhibitors of c-Jun-N-terminal kinase (JNK)
WO2013165816A2 (en) 2012-05-02 2013-11-07 Merck Sharp & Dohme Corp. SHORT INTERFERING NUCLEIC ACID (siNA) COMPOSITIONS
EP3919620A1 (en) 2012-05-02 2021-12-08 Sirna Therapeutics, Inc. Short interfering nucleic acid (sina) compositions
WO2014052563A2 (en) 2012-09-28 2014-04-03 Merck Sharp & Dohme Corp. Novel compounds that are erk inhibitors
US10112927B2 (en) 2012-10-18 2018-10-30 Dana-Farber Cancer Institute, Inc. Inhibitors of cyclin-dependent kinase 7 (CDK7)
US10787436B2 (en) 2012-10-18 2020-09-29 Dana-Farber Cancer Institute, Inc. Inhibitors of cyclin-dependent kinase 7 (CDK7)
US10000483B2 (en) 2012-10-19 2018-06-19 Dana-Farber Cancer Institute, Inc. Bone marrow on X chromosome kinase (BMX) inhibitors and uses thereof
US9758522B2 (en) 2012-10-19 2017-09-12 Dana-Farber Cancer Institute, Inc. Hydrophobically tagged small molecules as inducers of protein degradation
USRE48175E1 (en) 2012-10-19 2020-08-25 Dana-Farber Cancer Institute, Inc. Hydrophobically tagged small molecules as inducers of protein degradation
WO2014085216A1 (en) 2012-11-28 2014-06-05 Merck Sharp & Dohme Corp. Compositions and methods for treating cancer
WO2014100065A1 (en) 2012-12-20 2014-06-26 Merck Sharp & Dohme Corp. Substituted imidazopyridines as hdm2 inhibitors
WO2014120748A1 (en) 2013-01-30 2014-08-07 Merck Sharp & Dohme Corp. 2,6,7,8 substituted purines as hdm2 inhibitors
US11053195B2 (en) 2013-03-15 2021-07-06 Global Blood Therapeutics, Inc. Compounds and uses thereof for the modulation of hemoglobin
WO2015034925A1 (en) 2013-09-03 2015-03-12 Moderna Therapeutics, Inc. Circular polynucleotides
US11040957B2 (en) 2013-10-18 2021-06-22 Dana-Farber Cancer Institute, Inc. Heteroaromatic compounds useful for the treatment of proliferative diseases
US10906889B2 (en) 2013-10-18 2021-02-02 Dana-Farber Cancer Institute, Inc. Polycyclic inhibitors of cyclin-dependent kinase 7 (CDK7)
US10450269B1 (en) 2013-11-18 2019-10-22 Global Blood Therapeutics, Inc. Compounds and uses thereof for the modulation of hemoglobin
US9862688B2 (en) 2014-04-23 2018-01-09 Dana-Farber Cancer Institute, Inc. Hydrophobically tagged janus kinase inhibitors and uses thereof
US10017477B2 (en) 2014-04-23 2018-07-10 Dana-Farber Cancer Institute, Inc. Janus kinase inhibitors and uses thereof
EP3514151A1 (en) 2014-08-06 2019-07-24 Novartis AG Protein kinase c inhibitors and methods of their use
WO2016020864A1 (en) 2014-08-06 2016-02-11 Novartis Ag Protein kinase c inhibitors and methods of their use
US10870651B2 (en) 2014-12-23 2020-12-22 Dana-Farber Cancer Institute, Inc. Inhibitors of cyclin-dependent kinase 7 (CDK7)
US10550121B2 (en) 2015-03-27 2020-02-04 Dana-Farber Cancer Institute, Inc. Inhibitors of cyclin-dependent kinases
US11325910B2 (en) 2015-03-27 2022-05-10 Dana-Farber Cancer Institute, Inc. Inhibitors of cyclin-dependent kinases
US10702527B2 (en) 2015-06-12 2020-07-07 Dana-Farber Cancer Institute, Inc. Combination therapy of transcription inhibitors and kinase inhibitors
US11142507B2 (en) 2015-09-09 2021-10-12 Dana-Farber Cancer Institute, Inc. Inhibitors of cyclin-dependent kinases
WO2018058022A1 (en) 2016-09-26 2018-03-29 Merck Sharp & Dohme Corp. Anti-cd27 antibodies
WO2018190719A2 (en) 2017-04-13 2018-10-18 Aduro Biotech Holdings, Europe B.V. Anti-sirp alpha antibodies
WO2019094311A1 (en) 2017-11-08 2019-05-16 Merck Sharp & Dohme Corp. Prmt5 inhibitors
WO2019152642A1 (en) 2018-02-01 2019-08-08 Merck Sharp & Dohme Corp. Anti-pd-1/lag3 bispecific antibodies
WO2020033282A1 (en) 2018-08-07 2020-02-13 Merck Sharp & Dohme Corp. Prmt5 inhibitors
WO2020033284A1 (en) 2018-08-07 2020-02-13 Merck Sharp & Dohme Corp. Prmt5 inhibitors

Also Published As

Publication number Publication date
AU704139B2 (en) 1999-04-15
AU1162697A (en) 1997-06-11
EP0862435A4 (en) 1999-02-03
CA2238081A1 (en) 1997-05-29
JP2000500502A (en) 2000-01-18
EP0862435A1 (en) 1998-09-09

Similar Documents

Publication Publication Date Title
US6127366A (en) Inhibitors of farnesyl-protein transferase
US5817678A (en) Inhibitors of farnesyl-protein transferase
AU704139B2 (en) Inhibitors of farnesyl-protein transferase
US5756528A (en) Inhibitors of farnesyl-protein transferase
US6066738A (en) Inhibitors of farnesyl-protein transferase
US5914341A (en) Inhibitors of farnesyl-protein transferase
US5968965A (en) Inhibitors of farnesyl-protein transferase
US5872135A (en) Inhibitors of farnesyl-protein transferase
US5919785A (en) Inhibitors of farnesyl-protein transferase
US5869682A (en) Inhibitors of farnesyl-protein transferase
WO1997036889A1 (en) Inhibitors of farnesyl-protein transferase
US5885995A (en) Inhibitors of farnesyl-protein transferase
WO1996030343A1 (en) Inhibitors of farnesyl-protein transferase
WO1997036897A1 (en) Inhibitors of farnesyl-protein transferase
WO1997038665A2 (en) Inhibitors of farnesyl-protein transferase
EP0891360A1 (en) Inhibitors of farnesyl-protein transferase
US5780492A (en) Inhibitors of farnesyl-protein transferase
WO1997036605A1 (en) Inhibitors of farnesyl-protein transferase
AU715667B2 (en) Inhibitors of farnesyl-protein transferase
US5624936A (en) Inhibitors of farnesyl-protein transferase
WO1997027752A1 (en) Inhibitors of farnesyl-protein transferase
AU2660797A (en) Inhibitors of farnesyl-protein transferase
WO1997036593A1 (en) Inhibitors of farnesyl-protein transferase
WO1996034010A2 (en) Inhibitors of farnesyl-protein transferase
WO1997036892A1 (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 AM AZ BY KG KZ MD RU TJ TM

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): 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

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

Ref document number: 1996942798

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2238081

Country of ref document: CA

Ref country code: CA

Ref document number: 2238081

Kind code of ref document: A

Format of ref document f/p: F

ENP Entry into the national phase

Ref country code: JP

Ref document number: 1997 519941

Kind code of ref document: A

Format of ref document f/p: F

WWP Wipo information: published in national office

Ref document number: 1996942798

Country of ref document: EP

WWW Wipo information: withdrawn in national office

Ref document number: 1996942798

Country of ref document: EP