WO2000017166A1 - 4-carboxyamino-2-methyl-1,2,3,4-tetrahydroquinolines as cetp inhibitors - Google Patents

4-carboxyamino-2-methyl-1,2,3,4-tetrahydroquinolines as cetp inhibitors Download PDF

Info

Publication number
WO2000017166A1
WO2000017166A1 PCT/IB1999/001529 IB9901529W WO0017166A1 WO 2000017166 A1 WO2000017166 A1 WO 2000017166A1 IB 9901529 W IB9901529 W IB 9901529W WO 0017166 A1 WO0017166 A1 WO 0017166A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
substituted
optionally
alkyl
methyl
Prior art date
Application number
PCT/IB1999/001529
Other languages
French (fr)
Inventor
Michael Paul Deninno
Christian James Mularski
Roger Benjamin Ruggeri
Ronald Thure Wester
Original Assignee
Pfizer Products 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
Application filed by Pfizer Products Inc. filed Critical Pfizer Products Inc.
Priority to CA002344556A priority Critical patent/CA2344556A1/en
Priority to AU54398/99A priority patent/AU5439899A/en
Priority to EP99940421A priority patent/EP1114033B1/en
Priority to SI9930672T priority patent/SI1114033T1/en
Priority to DE69920727T priority patent/DE69920727T2/en
Priority to AT99940421T priority patent/ATE277906T1/en
Priority to BR9913842-5A priority patent/BR9913842A/en
Priority to JP2000574076A priority patent/JP3655193B2/en
Publication of WO2000017166A1 publication Critical patent/WO2000017166A1/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
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/04Anorexiants; Antiobesity agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/06Antihyperlipidemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/12Antihypertensives
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D215/00Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems
    • C07D215/02Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom
    • C07D215/16Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D215/38Nitrogen atoms
    • C07D215/42Nitrogen atoms attached in position 4
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D215/00Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems
    • C07D215/02Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom
    • C07D215/16Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D215/48Carbon atoms having three bonds to hetero atoms with at the most one bond to halogen
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/12Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings linked by a chain containing hetero atoms as chain links

Definitions

  • This invention relates to cholesteryl ester transfer protein (CETP) inhibitors, pharmaceutical compositions containing such inhibitors and the use of such inhibitors to elevate certain plasma lipid levels, including high density lipoprotein (HDL)-cholesterol and to lower certain other plasma lipid levels, such as low density lipoprotein (LD )-cholesteroi and triglycerides and accordingly to treat diseases which are affected by low levels of HDL cholesterol and/or high levels of LDL-cholesterol and triglycerides, such as atherosclerosis and cardiovascular diseases in certain mammals (i.e., those which have CETP in their plasma), including humans.
  • CETP cholesteryl ester transfer protein
  • Atherosclerosis and its associated coronary artery disease is the leading cause of mortality in the industrialized world.
  • CAD coronary artery disease
  • CHD coronary heart disease
  • dyslipidemia is not a unitary risk profile for CHD but may be comprised of one or more lipid aberrations.
  • cholesteryl ester transfer protein activity affects all three.
  • the net result of CETP activity is a lowering of HDL cholesterol and an increase in LDL cholesterol. This effect on lipoprotein profile is believed to be pro-atherogenic, especially in subjects whose lipid profile constitutes an increased risk for CHD.
  • EP0818448 discloses the preparation of certain 5,6,7,8 substituted tetrahydroquinolines and analogs as cholesteryl ester transfer protein inhibitors.
  • U.S. Pat. No. 5,231 ,102 discloses a class of 4-substituted 1 ,2,3,4- tetrahydroquinolines that possess an acidic group (or group convertible thereto in vivo) at the 2-position that are specific antagonists of N-methyl-D-aspartate (NMDA) receptors and are therefore useful in the treatment and/or prevention of neurodegenerative disorders.
  • NMDA N-methyl-D-aspartate
  • This invention is directed to compounds of Formula I
  • R 1 is hydrogen, Y, W-X, W-Y; wherein W is a carbonyl, thiocarbonyl, sulfinyl or sulfonyl; X is -ON, -S-Y, - ⁇ (H)-Y or -N-(Y) 2 ; wherein Y for each occurrence is independently Z or a fully saturated, partially unsaturated or fully unsaturated one to ten membered straight or branched carbon chain wherein the carbons, other than the connecting carbon, may optionally be replaced with one or two heteroatoms selected independently from oxygen, sulfur and nitrogen and said carbon is optionally mono-, di- or tri-substituted independently with halo, said carbon is optionally mono-substituted with hydroxy, said carbon is optionally mono-substituted with oxo, said sulfur is optionally mono- or di-substituted with oxo,
  • Z is a partially saturated, fully saturated or fully unsaturated three to twelve membered ring optionally having one to four heteroatoms selected independently from oxygen, sulfur and nitrogen, or a bicyclic ring consisting of two fused partially saturated, fully saturated or fully unsaturated three to six membered rings, taken independently, optionally having one to four heteroatoms selected independently from nitrogen, sulfur and oxygen; wherein said Z substituent is optionally mono-, di- or tri-substituted independently with halo, (C 2 -C 6 )alkenyl, (C C 6 ) alkyl, hydroxy, (C 1 -C 6 )alkoxy, (C,-C )alkylt.hio, amino, nitro, cyano, oxo, carboxy, (C C 6 )alkyloxycarbonyl, mono-N- or di-N,N-(C C 6 )alkylamino wherein said (C 1 -C 6 )alkyl substituent is optionally mono-, di
  • (C C 6 )alkyl optionally having from one to nine fluorines; wherein either R 3 must contain V or R 4 must contain V 1 ; and R 5 , R 6 , R 7 and R 8 are each independently hydrogen, a bond, nitro or halo wherein said bond is substituted with T or a partially saturated, fully saturated or fully unsaturated (C C ⁇ 2 ) straight or branched carbon chain wherein carbon may optionally be replaced with one or two heteroatoms selected independently from oxygen, sulfur and nitrogen wherein said carbon atoms are optionally mono-, di- or tri-substituted independently with halo, said carbon is optionally mono-substituted with hydroxy, said carbon is optionally mono-substituted with oxo, said sulfur is optionally mono- or di-substituted with oxo, said nitrogen is optionally mono- or di-substituted with oxo, and said carbon is optionally mono-substituted with T; wherein T is a partially saturated
  • a preferred group of compounds designated the A Group, contains those compounds having the Formula I as shown above wherein the C 2 methyl is beta; the C 4 nitrogen is beta:
  • R 1 is W-X ;
  • W is carbonyl, thiocarbonyl or sulfonyl
  • X is -O-Y-, S-Y-, N(H)-Y- or -N-(Y) 2 -;
  • Y for each occurrence is independently Z or (C C 4 )alkyl, said (C C 4 )alkyl optionally having hydroxy or from one to nine fluorines or said (C C 4 )alkyl optionally mono-substituted with Z wherein Z is a partially saturated, fully saturated or fully unsaturated three to six membered ring optionally having one to two heteroatoms selected independently from oxygen, suifur and nitrogen; wherein said Z substituent is optionally mono-, di- or tri-substituted independently with halo, (C C 4 )alkyl, (C 1 -C )alkoxy, (C C 4 )alkylthio, nitro, cyano, oxo, or (C C 6 )alkyioxycarbonyl, said (C r C 4 )alkyl optionally substituted with from one to
  • R 4 is (C C 4 )alkyl
  • R 6 and R 7 are each independently hydrogen, halo, T, (C C 6 )alkyl or (C
  • T is a partially saturated, fully saturated or fully unsaturated five to six membered ring optionally having one to two heteroatoms selected independently from oxygen, sulfur and nitrogen; wherein said T substituent is optionally mono-, di- or tri-substituted independently with halo, (C r C 6 )alkyl, hydroxy, (C C 6 )alkoxy, (C C 4 )alkylthio, amino, oxo, carboxy, (C C 6 )alkyloxycarbonyl, mono-N- or di-N,N-(C
  • R 5 and R 8 are H; and pharmaceutically acceptable salts thereof.
  • a group of compounds which is preferred among the A Group of compounds, designated the B Group, contains those compounds wherein W is carbonyl;
  • X is O-Y wherein Y is (C C 4 )alkyl, said (C C 4 )alkyl optionally having one hydroxy or from one to nine fluorines;
  • Q is (C C 4 )alkyl and V is phenyl, pyridinyl, or py ⁇ midinyl; wherein said V ring is optionally mono-, di- or tri-substituted independently with halo, (C C 6 )alkyl, hydroxy, (C C 6 )alkoxy, nitro, cyano or oxo wherein said (C C 6 )alkyl substituent optionally has from one to nine fluorines;
  • R 6 and R 7 are each independently hydrogen, halo or (C r C 3 )alkyl, said (C r C 3 )alkyl
  • C 3 )alkyl optionally having from one to seven fluorines; and pharmaceutically acceptable salts thereof.
  • a group of compounds which is preferred among the B Group of compounds, designated the C Group, contains those compounds wherein
  • Q is methyiene and V is phenyl or pyridinyl; wherein said V ring is optionally mono-, di- or tri-substituted independently with halo, (C C 2 )alkyl, or nitro wherein said (C C 2 )alkyl optionally has from one to five fluorines; and pharmaceutically acceptable salts thereof.
  • Especially preferred compounds of Formula I are the compounds:
  • [2R,4S] 4-[(3,5-bis-trifluoromethyl-benzyl)-methoxycarbonyl-amino]-2,6,7- trimethyl-3,4-dihydro-2H-quinoiine-1-carboxylic acid ethyl ester; and pharmaceutically acceptable salts of said compounds.
  • [2R,4S] 4-[(3,5-bis-trifluoromethyl-benzyl)-methoxycarbonyl-amino]-6J- diethyl-2-methyl-3,4-dihydro-2H-quinoline-1-carboxyiic acid ethyl ester; [2R,4S] 4-[(3,5-bis-trifiuoromethyl-benzyl)-methoxycarbonyl-amino]-6-ethyl-2- methyl-3,4-dihydro-2H-quinoline-1-carboxylic acid ethyi ester;
  • Y is ethyl; R 3 is 3,5-bis-trifluoromethyiphenylmethyl; R 4 is methyl; R 6 is chloro; and R 7 is hydrogen; d.
  • Y is ethyl;
  • R 3 is 3,5-bis-trifluoromethylphenylmethyl;
  • R 4 is methyl;
  • R 6 is methyl; and
  • R 7 is methyl;
  • e. Y is ethyl;
  • R 3 is 3,5-bis-trifluoromethylphenylmethyl;
  • R 4 is methyl;
  • R 6 is ethyl; and
  • R 7 is ethyl;
  • f. Y is ethyl;
  • R 3 is 3,5-bis-trifluoromethylphenylmethyl;
  • R 4 is methyl;
  • R 6 is ethyl; and
  • R 7 is hydrogen;
  • g. Y is ethyl;
  • R 3 is 3,5-bis-trifluoromethylphenyimethyl
  • R 4 is methyl; R 6 is trifluoromethyl; and
  • R 7 is hydrogen; and h. Y is isopropyi;
  • R 3 is 3,5-bis-trifluoromethylphenylmethyl
  • R 4 is methyl; R 6 is trifluoromethyl; and
  • R 7 is hydrogen, and pharmaceutically acceptable salts of said compounds.
  • a preferred group of compounds designated the D Group, contains those compounds having Formula I as shown above wherein the C 2 methyl is beta; the C 4 nitrogen is beta;
  • R 1 is W-X ;
  • W is carbonyl, thiocarbonyl or sulfonyl;
  • X is -O-Y-, S-Y-, N(H)-Y- or -N-(Y) 2 -;
  • Y for each occurrence is independently (C r C 4 )alkyl, said (C r C 4 )alkyl optionally having from one to nine fluorines or said (C C 4 )alkyl optionally mono-substituted with Z wherein Z is a partially saturated, fully saturated or fully unsaturated three to six membered ring optionally having one to two heteroatoms selected independently from oxygen, sulfur and nitrogen; wherein said Z substituent is optionally mono-, di- or tri-substituted independently with halo, (C 1 -C 4 )alkyl, (C C 4 )alkoxy, (C 1 -C 4 )alkyithio, nitro, cyano, oxo, or (C C 6 )alkyloxycarbonyl said (C C 4 )alkyl optionally having from one to nine fluorines;
  • R 3 is Q-V wherein Q is (C r C )alkyl and V is a five or six membered partially saturated, fully saturated or fully unsaturated ring optionally having one to three heteroatoms selected independently from oxygen, sulfur and nitrogen; wherein said V ring is optionally mono-, di-, tri- or tetra-substituted independently with halo, (C r C 6 )alkyl, hydroxy, (C 1 -C 6 )alkoxy, nitro, cyano or oxo wherein said (C C 6 )alkyl substituent is optionally mono-, di- or tri- substituted independently with (C C 6 )alkoxy or (C C 4 )alkylthio or said (C C 6 )alkyi optionally having from one to nine fluorines; R 4 is (C C 4 )alkyl;
  • R 6 and R 7 are each independently H, carbamoyl, oxycarbonyl, oxy or halo, or said carbamoyl, oxycarbonyl or oxy are substituted with (C r C 4 )alkyl and said (C C )alkyl is optionally mono-, di-, or tri-substituted with halo or hydroxy or said (C C 4 )alkyl is optionally mono-substituted with (C C 4 )alkoxy, carboxy, (C C 4 )alkylthio, (C 1 -C 4 )alkoxycarbonyi, amino or mono-N- or di-N,N-(C C 4 )alkylamino or said (C 1 -C 4 )alkyl optionally having from one to nine fluorines; or said (C C 4 )aikyl, carbamoyl, oxycarbonyl or oxy are optionally mono- substituted with T; wherein T is a partially saturated, fully saturated
  • R 5 and R 8 are H; or a pharmaceutically acceptable salt thereof.
  • a group of compounds which is preferred among the D Group of compounds, designated the E Group, contains those compounds wherein W is carbonyl;
  • X is O-Y wherein Y is (C 1 -C 4 ), said (C C 4 )alkyl optionally having from one to nine fluorines;
  • Q is (C r C 4 )alkylene and V is phenyl, pyridinyl, or pyrimidinyl; wherein said V ring is optionally mono-, di- or tri-substituted independently with halo, (C C 6 )alkyl, hydroxy, (C C 6 )alkoxy, nitro, cyano or oxo wherein said (C-,-C 6 )alkyl substituent optionally has from one to nine fluorines;
  • R 6 is H, carbamoyl, oxycarbonyl, oxy or halo, or said carbamoyl, oxycarbonyl or oxy are substituted with (C C 2 )alkyl and said (C C 2 )alkyl is optionally mono-, di-, or tri-substituted with halo or hydroxy or said (C C 4 )alkyl optionally having from one to nine fluorines;
  • R 7 is H, carbamoyl, oxycarbonyl, oxy or halo, or said carbamoyl, oxycarbonyl or oxy are optionally substituted with (C C 4 )alkyl and said (C C 4 )alkyi is optionally mono-, di-, or tri-substituted with halo or hydroxy or said (C C 4 )alkyl optionally having from one to nine fluorines; and pharmaceutically acceptable saits thereof.
  • Yet another aspect of this invention is directed to methods for treating atherosclerosis, peripheral vascular disease, dyslipidemia, hyperbetalipoproteinemia, hypoalphaiipoproteinemia, hypercholesterolemia, hypertrigiyceridemia, familial- hypercholesterolemia, cardiovascular disorders, angina, ischemia, cardiac ischemia, stroke, myocardial infarction, reperfusion injury, angioplastic restenosis, hypertension, vascular complications of diabetes, obesity or endotoxemia in a mammal (including a human being either male or female) by administering to a mammal in need of such treatment an atherosclerosis, peripheral vascular disease, dyslipidemia, hyperbetalipoproteinemia, hypoalphalipoproteinemia, hypercholesterolemia, hypertrigiyceridemia, familial-hypercholesterolemia, cardiovascular disorders, angina, ischemia, cardiac ischemia, stroke, myocardial infarction, reperfusion injury, angioplastic restenosis, hypertension, vascular complications of
  • Yet another aspect of this invention is directed to a method for treating atherosclerosis in a mammal (including a human being) by administering to a mammal in need of such treatment an atherosclerotic treating amount of a Formula I compound, a prodrug thereof, or a pharmaceutically acceptable salt of said compound or of said prodrug.
  • Yet another aspect of this invention is directed to a method for treating peripheral vascular disease in a mammal (including a human being) by administering to a mammal in need of such treatment a peripheral vascular disease treating amount of a Formula I compound, a prodrug thereof, or a pharmaceutically acceptable salt of said compound or of said prodrug.
  • Yet another aspect of this invention is directed to a method for treating dyslipidemia in a mammai (including a human being) by administering to a mammal in need of such treatment a dyslipidemia treating amount of a Formula I compound, a prodrug thereof, or a pharmaceutically acceptable salt of said compound or of said prodrug.
  • Yet another aspect of this invention is directed to a method for treating hyperbetalipoproteinemia in a mammal (including a human being) by administering to a mammal in need of such treatment a hyperbetalipoproteinemia treating amount of a mammal (including a human being) by administering to a mammal in need of such treatment a hyperbetalipoproteinemia treating amount of a mammal (including a human being) by administering to a mammal in need of such treatment a hyperbetalipoproteinemia treating amount of a
  • Yet another aspect of this invention is directed to a method for treating hypoalphalipoproteinemia in a mammal (including a human being) by administering to a mammal in need of such treatment a hypoalphalipoproteinemia treating amount of a Formula i compound, a prodrug thereof, or a pharmaceutically acceptable salt of said compound or of said prodrug.
  • Yet another aspect of this invention is directed to a method for treating hypercholesterolemia in a mammal (including a human being) by administering to a mammal in need of such treatment a hypercholesterolemia treating amount of a Formula I compound, a prodrug thereof, or a pharmaceutically acceptable salt of said compound or of said prodrug.
  • Yet another aspect of this invention is directed to a method for treating hypertrigiyceridemia in a mammal (including a human being) by administering to a mammal in need of such treatment a hypertrigiyceridemia treating amount of a Formula I compound, a prodrug thereof, or a pharmaceutically acceptable salt of said compound or of said prodrug.
  • Yet another aspect of this invention is directed to a method for treating familial-hypercholesterolemia in a mammal (including a human being) by administering to a mammal in need of such treatment a familial- hypercholesterolemia treating amount of a Formula I compound, a prodrug thereof, or a pharmaceutically acceptable salt of said compound or of said prodrug.
  • Yet another aspect of this invention is directed to a method for treating cardiovascular disorders in a mammal (including a human being) by administering to a mammal in need of such treatment a cardiovascular disorder treating amount of a Formula I compound, a prodrug thereof, or a pharmaceutically acceptable salt of said compound or of said prodrug.
  • Yet another aspect of this invention is directed to a method for treating angina in a mammal (including a human being) by administering to a mammal in need of such treatment an angina treating amount of a Formula I compound, a prodrug thereof, or a pharmaceutically acceptable salt of said compound or of said prodrug.
  • Yet another aspect of this invention is directed to a method for treating ischemia in a mammal (including a human being) by administering to a mammal in need of such treatment an ischemic disease treating amount of a Formula I compound, a prodrug thereof, or a pharmaceutically acceptable salt of said compound or of said prodrug.
  • Yet another aspect of this invention is directed to a method for treating cardiac ischemia in a mammal (including a human being) by administering to a mammal in need of such treatment a cardiac ischemic treating amount of a Formula I compound, a prodrug thereof, or a pharmaceutically acceptable salt of said compound or of said prodrug.
  • Yet another aspect of this invention is directed to a method for treating stroke in a mammal (including a human being) by administering to a mammal in need of such treatment a stroke treating amount of a Formula I compound, a prodrug thereof, or a pharmaceutically acceptable salt of said compound or of said prodrug.
  • Yet another aspect of this invention is directed to a method for treating a myocardial infarction in a mammal (including a human being) by administering to a mammal in need of such treatment a myocardial infarction treating amount of a Formula I compound, a prodrug thereof, or a pharmaceutically acceptable salt of said compound or of said prodrug.
  • Yet another aspect of this invention is directed to a method for treating reperfusion injury in a mammal (including a human being) by administering to a mammal in need of such treatment a reperfusion injury treating amount of a Formula I compound, a prodrug thereof, or a pharmaceutically acceptable salt of said compound or of said prodrug.
  • Yet another aspect of this invention is directed to a method for treating angioplastic restenosis in a mammal (including a human being) by administering to a mammal in need of such treatment an angioplastic restenosis treating amount of a Formula I compound, a prodrug thereof, or a pharmaceutically acceptable sait of said compound or of said prodrug.
  • Yet another aspect of this invention is directed to a method for treating hypertension in a mammal (including a human being) by administering to a mammal in need of such treatment a hypertension treating amount of a Formula I compound, a prodrug thereof or a pharmaceutically acceptable salt of said compound or of said prodrug.
  • Yet another aspect of this invention is directed to a method for treating the vascular complications of diabetes in a mammal (including a human being) by administering to a mammal in need of such treatment a vascular complications of diabetes treating amount of a Formula I compound, a prodrug thereof or a pharmaceutically acceptable salt of said compound or of said prodrug.
  • Yet another aspect of this invention is directed to a method for treating obesity in a mammal (including a human being) by administering to a mammal in need of such treatment an obesity treating amount of a Formula I compound, a prodrug thereof, or a pharmaceutically acceptable salt of said compound or of said prodrug.
  • Yet another aspect of this invention is directed to a method for treating endotoxemia in a mammal (including a human being) by administering to a mammal in need of such treatment an endotoxemia treating amount of a Formula I compoundT a prodrug thereof, or a pharmaceutically acceptable salt of said compound or of said prodrug.
  • a preferred dosage is about 0.001 to 100 mg/kg/day of a Formula I compound, a prodrug thereof, or a pharmaceutically acceptable salt of said compound or of said prodrug.
  • An especially preferred dosage is about 0.01 to 10 mg/kg/day of a Formula I compound, a prodrug thereof, or a pharmaceutically acceptable salt of said compound or of said prodrug.
  • compositions which comprise a therapeutically effective amount of a compound of Formula I, a prodrug thereof, or a pharmaceutically acceptable salt of said compound or of said prodrug and a pharmaceutically acceptable carrier.
  • This invention is also directed to pharmaceutical compositions for the treatment of atherosclerosis, peripheral vascular disease, dyslipidemia, hyperbetalipoproteinemia, hypoalphalipoproteinemia, hypercholesterolemia, hypertrigiyceridemia, familial-hypercholesterolemia, cardiovascular disorders, angina, ischemia, cardiac ischemia, stroke, myocardial infarction, reperfusion injury, angioplastic restenosis, hypertension, vascular complications of diabetes, obesity or endotoxemia in a mammal (including a human being) which comprise a therapeutically effective amount of a compound of Formula I, a prodrug thereof, or a pharmaceutically acceptable salt of said compound or of said prodrug and a pharmaceutically acceptable carrier.
  • compositions for the treatment of atherosclerosis in a mammal which comprise an atherosclerosis treating amount of a compound of Formula I, a prodrug thereof, or a pharmaceutically acceptable salt of said compound or of said prodrug and a pharmaceutically acceptable carrier.
  • compositions for the treatment of peripheral vascular disease in a mammal which comprise a peripheral vascular disease treating amount of a compound of Formula I, a prodrug thereof, or a pharmaceutically acceptable salt of said compound or of said prodrug and a pharmaceutically acceptable carrier.
  • compositions for the treatment of dyslipidemia in a mammal which comprise a ⁇ dyslipidemia treating amount of a compound of Formula I, a prodrug thereof, or a pharmaceutically acceptable salt of said compound or of said prodrug and a pharmaceutically acceptable carrier.
  • compositions for the treatment of hyperbetalipoproteinemia in a mammal which comprise a hyperbetalipoproteinemia treating amount of a compound of Formula I, a prodrug thereof, or a pharmaceutically acceptable salt of said compound or of said prodrug and a pharmaceutically acceptable carrier.
  • This invention is also directed to pharmaceutical compositions for the treatment of hypoalphalipoproteinemia in a mammal (including a human being) which comprise a hypoalphalipoproteinemia treating amount of a compound of Formula I, a prodrug thereof, or a pharmaceutically acceptable salt of said compound or of said prodrug and a pharmaceutically acceptable carrier.
  • compositions for the treatment of hypercholesterolemia in a mammal which comprise a hypercholesterolemia treating amount of a compound of Formula I, a prodrug thereof, or a pharmaceutically acceptable salt of said compound or of said prodrug and a pharmaceutically acceptable carrier.
  • compositions for the treatment of hypertrigiyceridemia in a mammal which comprise a hypertrigiyceridemia treating amount of a compound of Formula I, a prodrug thereof, or a pharmaceutically acceptable salt of said compound or of said prodrug and a pharmaceutically acceptable carrier.
  • This invention is also directed to pharmaceutical compositions for the treatment of familial-hypercholesterolemia in a mammal (including a human being) which comprise a familial-hypercholesterolemia treating amount of a compound of Formula I, a prodrug thereof, or a pharmaceutically acceptable salt of said compound or of said prodrug and a pharmaceutically acceptable carrier.
  • This invention is also directed to pharmaceutical compositions for the treatment of angina in a mammal (including a human being) which comprise an angina treating amount of a compound of Formula I, a prodrug thereof, or a pharmaceutically acceptable salt of said compound or of said prodrug and a pharmaceutically acceptable carrier.
  • compositions for the treatment of ischemia in a mammal which comprise an ischemic treating amount of a compound of Formula I, a prodrug thereof, or a pha ⁇ naceutically acceptable salt of said compound or of said prodrug and a pharmaceutically acceptable carrier.
  • compositions for the treatment of cardiac ischemia in a mammal which comprise a cardiac ischemic treating amount of a compound of Formula I, a prodrug thereof, or a pharmaceutically acceptable salt of said compound or of said prodrug and a pharmaceutically acceptable earner.
  • This invention is also directed to pharmaceutical compositions for the treatment of stroke in a mammal (including a human being) which comprise a stroke treating amount of a compound of Formula I, a prodrug thereof, or a pharmaceutically acceptable salt of said compound or of said prodrug and a pharmaceutically acceptable earner.
  • compositions for the treatment of a myocardial infarction in a mammal which comprise a myocardial infarction treating amount of a compound of Formula I, a prodrug thereof, or a pharmaceutically acceptable salt of said compound or of said prodrug and a pharmaceutically acceptable carrier.
  • compositions for the treatment of reperfusion injury in a mammal which comprise a reperfusion injury treating amount of a compound of Formula I, a prodrug thereof, or a pharmaceutically acceptable salt of said compound or of said prodrug and a pharmaceutically acceptable earner.
  • compositions for the treatment of angioplastic restenosis in a mammal which comprise an angioplastic restenosis treating amount of a compound of Formula I, a prodrug thereof, or a pharmaceutically acceptable salt of said compound or of said prodrug and a pharmaceutically acceptable carrier.
  • compositions for the treatment of hypertension in a mammal which comprise a hypertension treating amount of a compound of Formula I, a prodrug thereof or a ⁇ pharmaceutically acceptable salt of said compound or of said prodrug and a pharmaceutically acceptable carrier.
  • compositions for the treatment of the vascular complications of diabetes in a mammal which comprise a vascular complications of diabetes treating amount of a compound of Formula I, a prodrug thereof, or a pharmaceutically acceptable salt of said compound or of said prodrug and a pharmaceutically acceptable carrier.
  • compositions for the treatment of obesity in a mammal which comprise an obesity treating amount of a compound of Formula I, a prodrug thereof, or a pharmaceutically acceptable salt of said compound or of said prodrug and a pharmaceutically acceptable carrier.
  • compositions for the treatment of endotoxemia in a mammal which comprise an endotoxemia treating amount of a compound of Formula I, a prodrug thereof, or a pharmaceutically acceptable salt of said compound or of said prodrug and a pharmaceutically acceptable carrier.
  • This invention is also directed to a pharmaceutical combination composition
  • a pharmaceutical combination composition comprising: a therapeutically effective amount of a composition comprising a first compound, said first compound being a Fo ⁇ nula I compound, a prodrug thereof, or a pharmaceutically acceptable salt of said compound or of said prodrug; a second compound, said second compound being an HMG-CoA reductase inhibitor, an microsomal triglyceride transfer protein (MTP)/Apo B secretion inhibitor, a PPAR activator, a bile acid reuptake inhibitor, a cholesterol absorption inhibitor, a cholesterol synthesis inhibitor, a fibrate, niacin, an ion-exchange resin, an antioxidant, an ACAT inhibitor or a bile acid sequestrant; and/or optionally a pharmaceutical carrier.
  • MTP microsomal triglyceride transfer protein
  • PPAR activator a bile acid reuptake inhibitor
  • a cholesterol absorption inhibitor a cholesterol synthesis inhibitor
  • a particularly preferred HMG-CoA reductase inhibitor is lovastatin, simvastatin, pravastatin, fluvastatin, atorvastatin or rivastatin.
  • Another aspect of this invention is a method for treating atherosclerosis in a mammai comprising administering to a mammal suffering from atherosclerosis; a first compound, said first compound being a Formula I compound a prodrug thereof, or a pharmaceutically acceptable salt of said compound or of said prodrug; and a second compound, said second compound being an HMG-CoA reductase inhibitor, an MTP/Apo B secretion inhibitor, a cholesterol absorption inhibitor, a cholesterol synthesis inhibitor, a fibrate, niacin, an ion-exchange resin, an antioxidant, an ACAT inhibitor or a bile acid sequestrant wherein the amounts of the first and second compounds result in a therapeutic effect.
  • a preferred aspect of the above method is wherein the second compound is an HMG-CoA
  • a particularly preferred aspect of the above method is wherein the HMG-CoA reductase inhibitor is lovastatin, simvastatin, pravastatin, fluvastatin, atorvastatin or rivastatin.
  • a kit comprising: a. a first compound, said first compound being a Formula I compound, a prodrug thereof, or a pharmaceutically acceptable salt of said compound or of said prodrug and a pharmaceutically acceptable carrier in a first unit dosage form; b.
  • a second compound being an HMG CoA reductase inhibitor, an MTP/Apo B secretion inhibitor, a cholesterol absorption inhibitor, a cholesterol synthesis inhibitor, a fibrate, niacin, an ion-exchange resin, an antioxidant, an ACAT inhibitor or a bile acid sequestrant and a pharmaceutically acceptable carrier in a second unit dosage form; and c. means for containing said first and second dosage forms wherein the amounts of the first and second compounds result in a therapeutic effect.
  • a preferred second compound is an HMG-CoA reductase inhibitor or an MTP/Apo B secretion inhibitor.
  • a particularly preferred HMG-CoA reductase inhibitor is lovastatin, simvastatin, pravastatin, fluvastatin, atorvastatin or rivastatin.
  • mammals is meant to refer to all mammals which contain CETP in their plasma, for example, rabbits and primates such as monkeys and humans. Certain other mammals e.g., dogs, cats, cattle, goats, sheep and ⁇ horses do not contain CETP in their plasma and so are not included herein.
  • treating includes preventative (e.g., prophylactic) and palliative treatment.
  • pharmaceutically acceptable is meant the carrier, diluent, excipients, and/or salt must be compatible with the other ingredients of the formulation, and not deleterious to the recipient thereof.
  • prodrug refers to compounds that are drug precursors which following administration, release the drug in vivo via some chemical or physiological process (e.g., a prodrug on being brought to the physiological pH or through enzyme action is converted to the desired drug form).
  • exemplary prodrugs upon cleavage release the corresponding free acid, and such hydrolyzable ester-forming residues of the Formula I compounds include but are not limited to those having a carboxyl moiety wherein the free hydrogen is replaced by (C 1 -C 4 )alkyl, (C 2 - C 7 )alkanoyloxymethyl, 1-(alkanoyloxy)ethyl having from 4 to 9 carbon atoms, 1- methyl-1-(alkanoyioxy)-ethyl having from 5 to 10 carbon atoms, alkoxycarbonyloxymethyl having from 3 to 6 carbon atoms, 1- (alkoxycarbonyloxy)ethyl having from 4 to 7 carbon atoms, 1-methyl-1- (alkoxycarbonyloxy
  • Exemplary five to six membered aromatic rings optionally having one or two heteroatoms selected independently from oxygen, nitrogen and sulfur include phenyl, furyl, thienyl, pyrrolyl, oxazolyl, thiazolyl, imidazolyl, pyrazolyi, isoxazolyl, isothiazolyl, pyridinyl, pyridiazinyl, pyrimidinyl and pyrazinyl.
  • Exemplary partially saturated, fully saturated or fully unsaturated five to eight membered rings optionally having one to four heteroatoms selected independently from oxygen, sulfur and nitrogen include cyciopentyl, cyclohexyl, cycloheptyl, ⁇ cyclooctyl and phenyl.
  • Further exemplary five membered rings include 2H-pyrrolyl, 3H-pyrrolyl, 2-pyrrolinyl, 3-pyrrolinyl, pyrrolidinyl, 1 ,3-dioxolanyl, oxazolyl, thiazolyl, imidazolyl, 2H-imidazolyl, 2-imidazoiinyl, imidazolidinyi, pyrazolyi, 2-pyrazoiinyl, pyrazolidinyl, isoxazolyl, isothiazolyl, 1 ,2-dithioiyi, 1 ,3-dithiolyl, 3H-1 ,2-oxathiolyl, 1,2,3-oxadiazolyl, 1 ,2,4-oxadiazolyl, 1,2,5-oxadiazolyl, 1 ,3,4-oxadiazolyl, 1 ,2,3- triazolyl, 1 ,2,4-triazolyl, 1 ,3,
  • FIG. 1 For exemplary six membered rings, include 2H-pyranyi, 4H-pyranyl, pyridinyl, piperidinyl, 1,2-dioxinyl, 1 ,3-dioxinyl, 1,4-dioxanyl, mo ⁇ holinyl, 1 ,4-dithianyl, thiomo ⁇ holinyi, pyridazinyl, pyrimidinyi, pyrazinyl, piperazinyi, 1,3,5-triazinyl, 1,2,4- triazinyl, 1 ,2,3-triazinyl, 1,3,5-trithianyl, 4H-1 ,2-oxazinyl, 2H-1 ,3-oxazinyl, 6H-1.3- oxazinyi, 6H-1 ,2-oxazinyl, 1,4-oxazinyl, 2H-1 ,2-oxazinyl, 4H-1 ,4-o
  • FIG. 1 Further exemplary seven membered rings include azepinyl, oxepinyl, and thiepinyl.
  • FIG. 1 Further exemplary eight membered rings include cyclooctyl, cyclooctenyl and cyclooctadienyl.
  • Exemplary bicyclic rings consisting of two fused partially saturated, fully saturated or fully unsaturated five or six membered rings, taken independently, optionally having one to four heteroatoms selected independently from nitrogen, sulfur and oxygen include indolizinyl, indolyl, isoindolyl, 3H-indolyl, 1 H-isoindolyl, indolinyi, cyclopenta(b)pyridinyl, pyrano(3,4-b)pyrroiyl, benzofuryl, isobenzofuryl, benzo(b)thienyl, benzo(c)thienyl, 1 H-indazoiyl, indoxazinyl, benzoxazolyl, benzimidazolyl, benzthiazolyl, purinyl, 4H-quinoiizinyl, quinolinyl, isoquinolinyl, cinnolinyl, phthaiazinyl, quinazolin
  • alkylene saturated hydrocarbon (straight chain or branched ) wherein a hydrogen atom is removed from each of the terminal carbons.
  • exemplary of such groups are methylene, ethylene, propylene, butylene, pentylene, hexylene, heptylene).
  • halo is meant chloro, bromo, iodo, or fluoro.
  • alkyl straight chain saturated hydrocarbon or branched chain saturated hydrocarbon.
  • alkyl groups (assuming the designated length encompasses the particular example) are methyl, ethyl, propyl, isopropyl, butyl, sec-butyl, tertiary butyl, pentyl, isopentyl, neopentyl, tertiary pentyl, 1- methylbutyl, 2-methylbutyl, 3-methylbutyl, hexyl, isohexyl, heptyi and octyl.
  • alkoxy is meant straight chain saturated alkyl or branched chain saturated alkyl bonded through an oxy.
  • alkoxy groups (assuming the designated length encompasses the particular example) are methoxy, ethoxy, propoxy, isopropoxy, butoxy, isobutoxy, tertiary butoxy, pentoxy, isopentoxy, neopentoxy, tertiary pentoxy, hexoxy, isohexoxy, heptoxy and octoxy .
  • mono-N- or di-N,N-(C C x )alkyl refers to the (C C x )alkyl moiety taken independently when it is di-N,N-(C C x )alkyi...(x refers to integers).
  • a carbocyciic or heterocyclic moiety may be bonded or otherwise attached to a designated substrate through differing ring atoms without denoting a specific point of attachment, then all possible points are intended, whether through a carbon atom or, for example, a trivalent nitrogen atom.
  • pyridyl means 2-, 3-, or 4-pyridyl
  • thienyl means 2-, or 3-thienyl, and so forth.
  • pharmaceutically-acceptabie salt refers to nontoxic anionic salts containing anions such as (but not limited to) chloride, bromide, iodide, sulfate, bisulfate, phosphate, acetate, maleate, fumarate, oxalate, lactate, tartrate, citrate, gluconate, methanesulfonate and 4-toluene-sulfonate.
  • anions such as (but not limited to) chloride, bromide, iodide, sulfate, bisulfate, phosphate, acetate, maleate, fumarate, oxalate, lactate, tartrate, citrate, gluconate, methanesulfonate and 4-toluene-sulfonate.
  • nontoxic cationic salts such as (but not limited to) sodium, potassium, calcium, magnesium, ammonium or protonated benzathine (N,N'-dibenzylethylenediamine), choline, ethanolamine, diethanolamine, ethylenediamine, meglamine (N-methyl- glucamine), benethamine (N-benzylphenethylamine), piperazine or tromethamine (2- amino-2-hydroxymethyl-1 ,3-propanediol).
  • nontoxic cationic salts such as (but not limited to) sodium, potassium, calcium, magnesium, ammonium or protonated benzathine (N,N'-dibenzylethylenediamine), choline, ethanolamine, diethanolamine, ethylenediamine, meglamine (N-methyl- glucamine), benethamine (N-benzylphenethylamine), piperazine or tromethamine (2- amino-2-hydroxymethyl-1 ,3-propaned
  • reaction-inert solvent and “inert solvent” refers to a solvent or a mixture thereof which does not interact with starting materials, reagents, intermediates or products in a manner which adversely affects the yield of the desired product.
  • Beta refers to the orientation of a substituent with reference to the plane of the ring (i.e., page). Beta is above the plane of the ring (i.e., page) and Alpha is below the plane of the ring (i.e., page).
  • the compounds of this invention can exist in radioiabelled form, i.e., said compounds may contain one or more atoms containing an atomic mass or mass number different from the atomic mass or mass number usually found in nature.
  • Radioisotopes of hydrogen, carbon, phosphorous, fluorine and chlorine include 3 H, 14 C, 32 P, 35 S, 18 F and ⁇ Cl, respectively.
  • Compounds of this invention, a prodrug thereof, or a pharmaceutically acceptable salt of said compound or of said prodrug which contain those radioisotopes and/or other radioisotopes of other atoms are within the scope of this invention.
  • Radiolabelled compounds of Formula I of this invention and prodrugs thereof can generally be prepared by methods well known to those skilled in the art. Conveniently, such radiolabelled compounds can be prepared by carrying out the procedures disclosed in the Schemes and/or in the Examples and Preparations below by substituting a readily available radiolabelled reagent for a non-radiolabelled reagent.
  • DTT means dithiothreitol.
  • DMSO means dimethyl sulfoxide.
  • EDTA means ethylenediamine tetraacetic acid.
  • certain Formula I compounds contain primary amines or carboxylic acid functionalities which may interfere with reactions at other sites of the molecule if left unprotected. Accordingly, such functionalities may be protected by an appropriate protecting group which may be removed in a subsequent step.
  • Suitable protecting groups for amine and carboxylic acid protection include those protecting groups commonly used in peptide synthesis (such as N-t-butoxycarbonyl, benzyloxycarbonyl, and 9-fluorenylmethylenoxycarbonyl for amines and iower alkyl or benzyl esters for carboxylic acids) which are generally not chemically reactive under the reaction conditions described and can typically be removed without chemically altering other functionality in the Formula I compound.
  • the Formula III compounds wherein R 5 , R D , R 7 , and R ⁇ are as described above and P 2 is an appropriate protecting group may be prepared from the appropriate Formula II aromatic amine wherein R 5 , R 6 , R 7 and R 8 are as described above.
  • the Formula III tetrahydroquinoiine is prepared by treating the appropriate
  • Formula II aromatic amine with acetaldehyde in an inert solvent such as a hydrocarbon (e.g., hexanes, pentanes or cyciohexane), an aromatic hydrocarbon (e.g., benzene, toluene or xylene), a halocarbon (e.g., dichloromethane, chloroform, carbon tetrachloride or dichloroethane), an ether (e.g., diethyi ether, diisopropyl ether, tetrahydrofuran, tetrahydropyran, dioxane, dimethoxyethane, methyl tert-butyl ether, etc.), a nitrile (e.g., acetonitrile or propionitrile), a nitroalkane (e.g., nitromethane or nitrobenzene), preferably dichloromethane with a dehydrating agent (e
  • a suitably substituted e.g., benzyloxycarbonyl, t- butoxycarbonyi, methoxycarbonyl, formyl-, acetyl-, diallyl- or dibenzyl-
  • carboxybenzyloxy-, N-vinyl species e.g., boron trifluoride, ⁇ boron trifluoride etherate, zinc chloride, titanium tetrachloride, iron trichloride, aluminum trichloride, alkyl aluminum dichloride, dialkyl aluminum chloride or ytterbium (III) trifiate; preferably boron trifluoride etherate
  • a protic acid such as a hydrohalogenic acid (e.g., fluoro, chloro, bromo or iodo), an aikyl sulfonic acid (e.g., p- toluene, methane or trif
  • the Formula II amine and acetaldehyde may be condensed by treating a solution of the amine and an alkyl amine base (preferably triethylamine) in a polar aprotic solvent (preferably dichloromethane) with titanium tetrachloride in a polar aprotic solvent (preferably in dichloromethane) at a temperature between about -78°C to about 40°C (preferably 0°C) followed by treatment with acetaldehyde at a temperature between about -78°C to about 40°C (preferably 0°C).
  • the reaction is allowed to proceed for about 0.1 to about 10 hours (preferably 1 hour) at a temperature between about 0°C to about 40°C (preferably room temperature) yielding the imine which is reacted with the N-vinyi species as above.
  • the compounds of Formula IV wherein R 1 , R 5 , R 6 , R 7 and R 8 are as described above and P 1 and P 2 are protecting groups may be prepared from the corresponding Formula III amine by various amine reaction routes known to those skilled in the art.
  • the Formula IV compounds wherein R 1 , R 5 , R 6 , R 7 , and R 8 are as described above and P 1 and P 2 are appropriately differentiated protecting groups for the amine moieties are prepared from the corresponding Formula III tetrahydroquinoline employing standard methods for derivatizing amines into the functional groups described for R 1 above, see Richard Larock, Comprehensive Organic Transformations. VCH Publishers Inc., New York, 1989 and Jerry March, Advanced Organic Chemistry. John Wiley & Sons, New York, 1985.
  • a Formula ill compound is treated with the appropriate carbonyl chloride, sulfonyl chloride, or sulf ⁇ nyl chloride, isocyanate or thioisocyanate in a polar aprotic solvent (preferably dichloromethane) in the presence of a base (preferably pyridine) at a temperature of from about -78°C to about 100°C (preferably starting at 0°C and letting warm to room temperature) for a period of 1 to 24 hours (preferably 12 hours).
  • a polar aprotic solvent preferably dichloromethane
  • Formula IV carbamate and urea compounds may be prepared from the Formula III amines via the corresponding carbamoyl chlorides by treating the Formula III amine with a phosgene solution in a hydrocarbon solvent (preferably toluene) at a temperature between about 0°C and about 200°C (preferably at reflux) for between 0.1 and 24 hours (preferably 2 hours).
  • a hydrocarbon solvent preferably toluene
  • the corresponding ureas may be prepared by treating a solution of the carbamoyl chlorides (prepared as described above) with the appropriate amine in a polar solvent (preferably dichloromethane) at a temperature between about -78°C and about 100°C (preferably ambient temperature) for between 1 and 24 hours (preferably 12 hours).
  • a polar solvent preferably dichloromethane
  • the corresponding carbamate may be prepared by treating a solution of the carbamoyl chlorides (prepared as described above) with the appropriate alcohol and a suitable base (preferably sodium hydride) in a polar solvent (preferably dioxane) at a temperature between about -78°C and about 100°C (preferably ambient temperature) for between 1 and 24 hours (preferably 12 hours).
  • a suitable base preferably sodium hydride
  • a polar solvent preferably dioxane
  • the corresponding carbamate may be prepared by treating a solution of the carbamoyl chlorides at a temperature between about 0°C and about 200°C in the appropriate alcohol for between 1 and 240 hours (preferably 24 hours).
  • the Formula IV compound wherein R 1 is Y may be prepared using methods known to those skilled in the art to introduce Y substituents such as an alkyl or alkyl linked substituent. Methods include, for example, formation of the amide from the amine of Formula III and an activated carboxylic acid followed by reduction of the amide with borane in an etheral solvent such as tetrahydrofuran.
  • the alkyl or alkyl linked substituent may be appended by reduction after condensing the amine of Formula III with the required carbonyl containing reactant.
  • the amine of Formula III may be reacted with the appropriate alkyl or aryl halide according to methods known to those skilled in the art.
  • the Formula III amine and an acid are treated with the appropriate carbonyl containing reactant in a polar solvent (preferably ethanol) at a temperature of about 0°C to about 100°C (preferably room temperature) for about 0.1 to 24 hours (preferably 1 hour) followed by treatment with a hydride source (e.g., sodium borohydride, sodium cyanoborohydride, preferably sodium triacetoxyborohydride) at a temperature of about 0°C to about 100°C (preferably ambient temperature) for 0.1 to 100 hours (preferably 5 hours).
  • a polar solvent preferably ethanol
  • a hydride source e.g., sodium borohydride, sodium cyanoborohydride, preferably sodium triacetoxyborohydride
  • the Formula V amine wherein R 1 , R 5 , R 6 , R 7 , and R ⁇ are as described above and P ' is a protecting group may be prepared from the corresponding Formula IV compound by deprotection (P 2 ) using methods known to those skilled in the art, including hydrogenolysis, treatment with an acid (e.g., trifiuoroacetic acid, hydrobromic), a base (sodium hydroxide), or reaction with a nucleophile (e.g. sodium methylthiolate, sodium cyanide, etc.) and for the trialkylsilylethoxy carbonyl group a fluoride is used (e.g., tetrabutyl ammonium fluoride).
  • an acid e.g., trifiuoroacetic acid, hydrobromic
  • a base sodium hydroxide
  • a nucleophile e.g. sodium methylthiolate, sodium cyanide, etc.
  • a fluoride e.g.,
  • hydrogenolysis is performed by treating the Formula IV compound with a hydride source (e.g., 1 to 10 atmospheres of hydrogen gas, cyclohexene or ammonium formate) in the presence of a suitable catalyst (e.g., 5- 20% palladium on carbon, palladium hydroxide; preferably 10% palladium on carbon) in a polar solvent (e.g., methanol, ethanol or ethyl acetate; preferably ethanol) at a temperature between about -78°C and about 100°C, preferably ambient temperature, for 0.1 to 24 hours, preferably 1 hour.
  • a hydride source e.g., 1 to 10 atmospheres of hydrogen gas, cyclohexene or ammonium formate
  • a suitable catalyst e.g., 5- 20% palladium on carbon, palladium hydroxide; preferably 10% palladium on carbon
  • a polar solvent e.g., methanol, ethanol or ethyl acetate; preferably
  • the Formula VI secondary amine wherein R 3 is as described above may be prepared using methods known to those skilled in the art to introduce R 3 substituents such as an alkyl or alkyl linked substituent.
  • Methods include, for example, formation of an amide from the Formula V amine and an activated carboxylic acid followed by reduction of the amide with borane in an etheral solvent such as tetrahydrofuran.
  • an alkyl or alkyl linked substituent may be appended by reduction of the appropriate imine, the imine being formed by condensing the Formula V amine with the required carbonyl containing reactant.
  • the Formula V amine may be reacted with the appropriate alkyl halide according to methods known to those skilled in the art.
  • the Formula V amine and an acid are treated with the appropriate carbonyl containing reactant in a polar solvent (preferably dichloromethane) at a temperature of about 0°C to about 100°C (preferably room temperature) for about 0.1 to 24 hours (preferably 1 hour) followed by treatment with a hydride source (e.g., sodium borohydride or sodium cyanoborohydride; preferably sodium triacetoxyborohydride) at a temperature of about 0°C to about 100°C (preferably ambient temperature) for 0.1 to 100 hours (preferably 5 hours).
  • a polar solvent preferably dichloromethane
  • a hydride source e.g., sodium borohydride or sodium cyanoborohydride; preferably sodium triacetoxyborohydride
  • the Formula VII compound wherein R 1 , R 3 , R 5 , R 6 , R 7 and R 8 are as described above and P 1 and P 2 are protecting groups may be prepared from the corresponding Fo ⁇ nula IV compound by methods known to those skilled in the art; for example, the methods described for the introduction of the R 3 substituent above in the transformation of the Formula V compound to the Formula VI compound. Following this, the corresponding Formula VI compound may be prepared from the Formula VII compound by appropriate deprotection such as the methods described above for the transformation of the Formula IV compound to the Formula V compound.
  • R 4 may be represented by R 3 in the Formulas VI and VII in Scheme I, thus providing a synthetic scheme for such compounds.
  • the Formula XI dihydroquinolone compounds wherein R 5 , R 6 , R 7 , R 8 and Y are as described above, and P 1 is a protecting group, may be prepared from the corresponding Formula X quinoiines by treatment with a metallo- methyl species and a chloroformate followed by hydrolysis.
  • a mixture of the Formula X quinoline and an excess (preferably 1.5 equivalents) of a methyl magnesium species (Grignard reagent) in a polar aprotic sumble (e.g., diethyi ether or dichloromethane; preferably tetrahydrofuran) is treated with an excess (preferably 1.5 equivalents) of a Y- or P 1 -chloroformate at a temperature between about -100°C and about 70°C (preferably -78°C) followed by warming to a temperature between about 0°C and about 70°C (preferably ambient temperature) for between 0.1 and 24 hours (preferably 1 hour).
  • a methyl magnesium species e.g., diethyi ether or dichloromethane; preferably tetrahydrofuran
  • the Formula XI compounds are the Formula XVI compounds wherein R 1 is -C(O)OY or P is -C(0)OP 1 without further transformation.
  • Formula XV compounds wherein R 5 , R 6 , R 7 and R 8 are as described above may be prepared from the corresponding Formula XI dihydroquinolone (wherein Formula XI compound contains P 1 ) by appropriate deprotection (including spontaneous decarboxylation) as described for the transformation of the Formula IV compound to the Formula V compound.
  • the Formula XVI compounds wherein R 1 , R 5 , R 6 , R 7 and R 8 are as described above and P 1 is a protecting group may be prepared from the corresponding Formula XV dihydroquinolone as described for the transformation of the Formula III compound to the Formula IV compound.
  • the substituent may be conveniently removed by treatment with acid (e.g., aqueous HCI) or base (e.g., aqueous sodium hydroxide).
  • the resulting Formula XII imine is reduced by treatment with a reducing agent (preferably sodium borohydride) in an appropriate polar solvent (preferably ethanol) at a temperature between about 0°C and about 80°C (preferably room temperature) for between 1 and 24 hours (preferably 12 hours) resulting in a mixture of diastereomeric Formula VI amines, generally favoring the trans isomer.
  • a reducing agent preferably sodium borohydride
  • an appropriate polar solvent preferably ethanol
  • the reduction may be performed by treating the Formula XII imine directly with an excess (preferably 5 equivalents) of zinc borohydride as a solution in ether (preferably 0.2 molar) at a temperature between about 0°C and about 40°C (preferably ambient temperature) for between 1 and 24 hours (preferably 12 hours) resulting in a mixture of diastereomeric Formula VI amines, generally favoring the cis isomer.
  • an excess preferably 5 equivalents
  • zinc borohydride as a solution in ether (preferably 0.2 molar) at a temperature between about 0°C and about 40°C (preferably ambient temperature) for between 1 and 24 hours (preferably 12 hours) resulting in a mixture of diastereomeric Formula VI amines, generally favoring the cis isomer.
  • the Formula VI amine wherein R 1 , R 3 , R 5 , R 6 , R 7 and R 8 are as described above and P is a protecting group may be prepared from the corresponding Formula XVI dihydroquinolones by formation of an oxime, reduction and substitution of the amine.
  • the Formula XVI dihydroquinolone, excess (preferably 3 equivalents) hydroxylamine hydrochloride and an excess (preferably 2.5 equivalents) of base (preferably sodium acetate) are reacted at a temperature between about 0°C and about 100°C (preferably at reflux) for between 1 and 24 hours (preferably 2 hours) in a polar solvent (preferably ethanol).
  • the resulting Formula XIII oxime is treated with excess (preferably 6 equivalents) aqueous base (preferably 2N potassium hydroxide) in a polar solvent (preferably ethanol) and an excess (preferably 4 equivalents) of a nickel-aluminum alloy (preferably 1:1 by weight) at a temperature between about 0°C and about 100°C (preferably ambient temperature) for between 0.25 and 24 hours (preferably 1 hour).
  • aqueous base preferably 2N potassium hydroxide
  • a polar solvent preferably ethanol
  • an excess (preferably 4 equivalents) of a nickel-aluminum alloy preferably 1:1 by weight
  • the Formula VI secondary amine wherein R 1 , R 3 , R 5 , R 6 , R 7 and R 8 are as described above and P 1 is a protecting group may be prepared from the appropriate Formula V amine as described in Scheme I for the transformation of the Formula V compound to the Formula VI compound.
  • the Formula I compounds as described above may be prepared from the appropriate Formula VI compounds by conversion to the desired carbamate.
  • the Formula VI amine is treated with the appropriate activated carbonate (e.g., chloroformate, dicarbonate or carbonyl diimidazole followed by the appropriate alcohol) in a polar solvent (preferably dichloromethane) in the presence of an excess of amine base (preferably pyridine) at a temperature between about -20°C and about 40°C (preferably ambient temperature) for between 1 and 24 hours (preferably 12 hours) to yield the Formula I compound.
  • a polar solvent preferably dichloromethane
  • an excess of amine base preferably pyridine
  • the P 1 protected Formula VI compound may be transformed to the Formula I compound through protection/deprotection sequences and introduction of the desired substituents.
  • the Formula VI amine is treated with the appropriate reagent (e.g., protecting group precursor, activated carbonate (e.g., chloroformate, dicartDonate or carbonyl imidazole)) in a polar solvent (preferably dichloromethane) in the presence of an excess of amine base (preferably pyridine) at a temperature between about -20°C and about 40 °C (preferably ambient temperature) for between 1 and 24 hours (preferably 12 hours) to yield the Formula XX compound .
  • the appropriate reagent e.g., protecting group precursor, activated carbonate (e.g., chloroformate, dicartDonate or carbonyl imidazole)
  • a polar solvent preferably dichloromethane
  • an excess of amine base preferably pyridine
  • the Formula XXI compound is conveniently prepared by treatment with an acid (preferably trifluoroacetic acid) at a temperature between about 0°C and about 100°C (preferably room temperature) for 0.1 to 24 hours (preferably 1 hour).
  • an acid preferably trifluoroacetic acid
  • the compounds of Formula I or compounds of Formula XXII may be prepared from the corresponding Formula XXI amine (wherein R 4 or P 2 is present respectively) by various amine reaction routes known to those skilled in the art, for example, those described in Scheme I for the transformation of the Formula III compound to the Formula IV compound.
  • the Formula XXIII amines may be prepared from the Formula XXII compounds by suitable deprotection.
  • the Formula XXIII compound is prepared by treatment with an excess of a hydride source (e.g., cyclohexene, hydrogen gas or preferably ammonium formate) in the presence of 0.01 to 2 equivalents (preferably 0.1 equivalent) of a suitable catalyst (preferably 10% palladium on carbon) in a polar solvent (preferably ethanol) at a temperature between about 0°C and about 100°C (preferably room temperature) for 0.1 to 24 hours (preferably 1 hour).
  • a hydride source e.g., cyclohexene, hydrogen gas or preferably ammonium formate
  • a suitable catalyst preferably 10% palladium on carbon
  • a polar solvent preferably ethanol
  • the Formula I compound wherein R 4 is as described above may be prepared using the methods described for the conversion of the Formula VI compound to the Formula I compound in Scheme III above.
  • the Formula XXXI oxime is treated with an excess (preferably six equivalents) of an aqueous base (preferably 2N potassium hydroxide) and an excess (preferably four equivalents) of a nickel-aluminum alloy (preferably 1 :1 by weight) in a polar solvent (preferably ethanoi) at a temperature between about 0°C and about 100°C (preferably ambient temperature) for between 0.25 and 24 hours (preferably 2 hours) to prepare the corresponding Formula XXXII amine.
  • a polar solvent preferably ethanoi
  • the P 3 protecting group may be removed using standard methods if the oxime transformation does not result in such cleavage.
  • the Formula XXX compound may be deprotected (removal of the P 3 ) by methods known to those skilled in the art prior to formation of the Formula XXXI oxime (wherein P 3 is H) which can then be reduced to form the Formula XXXII amine.
  • the Formula V compound wherein R 6 is an oxy-linked moiety may be prepared by treating the Formula XXXII alcohol under, for example, Mitsunobu conditions.
  • Formula XXXII phenol is treated with a phosphine (preferably triphenylphosphine) and an azodicarboxylate (preferably bis-(N-methylpiperazinyl)- azodicarboxamide) and the required alcohol in a polar solvent (preferably benzene).
  • a phosphine preferably triphenylphosphine
  • an azodicarboxylate preferably bis-(N-methylpiperazinyl)- azodicarboxamide
  • a polar solvent preferably benzene
  • the Formula XX compound wherein R 6 is an ether linked moiety and wherein R 1 , R 3 and R 4 are as described above and P 1 and P 2 are protecting groups may be prepared from the Formula XXXII alcohols as described below.
  • such processes may be used to prepare the corresponding compounds wherein R 5 , R 7 , or R 8 are an ether iinked moiety starting from the corresponding Formula XXXII compound and thus ultimately the Formula XXX compound (i.e., the Formula XXX compound having a P 3 0- at either the R 5 , R 7 , or R 8 positions).
  • the Formula XXXIII secondary amine wherein R 3 is as described above may be prepared from the corresponding Formula XXXII compound according to methods in Scheme I described above for the conversion of the Formula V compound to the Formula VI compound.
  • the Formula XXXIV compounds wherein R 4 is as described above may be prepared from Formula XXXIII amines by methods analogous to that described in Scheme III for the transformation of the Formula VI compound to the Formula I compound.
  • the Formula XXXV phenol may be selectively deprotected for example when R 4 0 2 CO- is present by treating the Formula XXXIV carbonate with potassium carbonate in a polar solvent (preferably methanol) at a temperature between about 0°C and about 100°C (preferably ambient temperature) for between 1 and 24 hours (preferably 12 hours).
  • a polar solvent preferably methanol
  • the corresponding XX ethers may be prepared from the Formula XXXV phenol using for example, the Mitsunobu conditions described above for the conversion of the Formula XXXII compounds to the Formula V compounds.
  • the phenol may be derivatized to a variety of functional groups using standard methods, for example, as described in March or Larock, or by conversion to the corresponding triflate for use in a variety of reactions involving transition metal catalysis.
  • R 7 and R 8 are as described above, P 1 and P 2 are protecting groups, and X 1 is a linking group wherein a carbon (e.g., methylene) is directly linked to the carbonyl moiety may be prepared from the corresponding ester (wherein R 12 is a convenient alkyl moiety) by reduction.
  • the Formula L ester is treated with sodium borohydride/methanoi or a borane-dimethylsuifide complex in a polar solvent (preferably tetrahydrofuran) at a temperature between about 0°C and about 100°C (preferably at reflux) for between 1 and 24 hours (preferably 3 hours).
  • the Formula Lll compounds wherein R 1 , R 3 , R 4 , R 5 , R 7 and R 8 are as described above, P 1 and P 2 are protecting groups and wherein the R 6 position includes an alkyl haiide functionality may be prepared from the corresponding Formula LI alcohol by treatment with a trialkylphosphine (preferably triphenylphosphine) and a dihalogen (e.g., bromine) in a polar solvent (preferably dichloromethane) at a temperature between about -78°C and about 100°C (preferably 0°C) for between 0.1 and 10 hours (preferably 0.5 hours) followed by warming to room temperature for between 0.1 and 10 hours (preferably 3 hours).
  • a trialkylphosphine preferably triphenylphosphine
  • a dihalogen e.g., bromine
  • a polar solvent preferably dichloromethane
  • the Formula Llll compounds wherein R 1 , R 3 , R 4 , R 5 , R 7 and R 8 are as described above, P 1 and P 2 are protecting groups, the R 6 position includes ether or thioether moieties (i.e., Y 1 is S or O) and R 13 is a carbon linked substituent may be prepared by treating the Formula LI I alkyl halide in a polar solvent (preferably N,N-dimethylformamide) with the requisite alkoxide or thioalkoxide at a temperature between about 0°C and about 100°C (preferably at room temperature) for between 1 and 24 hours (preferably 6 hours).
  • a polar solvent preferably N,N-dimethylformamide
  • the Formula Llll ethers and thioethers may be prepared by treating the corresponding Formula LIV alcohols and thiols (i.e., Y 1 is S or O), wherein X 1 is a substituent linked directly through carbon to the methyiene moiety, with a base (preferably sodium hydride) and the requisite alkylating agent in a polar solvent (preferably N,N-dimethylformamide) at a temperature between about 0°C and about 100 C C (preferably at room temperature) for between 1 and 50 hours (preferably 18 hours).
  • a base preferably sodium hydride
  • a polar solvent preferably N,N-dimethylformamide
  • the Formula LV compounds wherein R 1 , R 3 , R 4 , R 5 , R 7 and R 8 are as described above, P 1 and P 2 are protecting groups, the R 6 position includes alkyl halides (e.g., fluorides) and X 1 is a substituent that is carbon linked directly to the methyiene moiety may be prepared by treating the corresponding Formula LI alcohol with a halogenating agent.
  • a fluorinating agent preferably diethylaminosuifur trifluoride
  • a polar solvent preferably 1 ,2-dichioroethane
  • R 8 are as described above, P 1 and P 2 are protecting groups and wherein R 6 includes an amide functionality (such that X is a substituent that is carbon linked directly to the carbonyl moiety and R 10 and R 11 are substituents selected to yield the desired R 6 substituent defined above) may be prepared from the corresponding Formula LVI carboxylic acid which may in turn be prepared from the corresponding Fo ⁇ nula L carboxylic ester.
  • the Fo ⁇ nula L ester is treated with an aqueous hydroxide
  • a polar solvent preferably tetrahydrofuran and/or methanol
  • the Formula LVII amide may be prepared from the corresponding Formula LVI acid by standard methods. Prefered is conversion of the carboxylic acid to the acid chloride by dissolving the acid in thionyl chloride and holding the solution at a temperature between about 0°C and about 80°C (preferably at reflux) for between 0.1 and 24 hours (preferably 1 hour) before evaporation of the excess thionyl chloride.
  • This step is followed by treating the resulting acid chloride residue in a polar solvent (preferably dichloromethane) with the appropriate amine, selected to yield the amide functionality, and optionally an amine base (preferably triethylamine) at a temperature between about -78°C and about 100°C (preferably room temperature) for between 0.1 and 100 hours (preferably 1 hour).
  • a polar solvent preferably dichloromethane
  • an amine base preferably triethylamine
  • R 4 , R 5 , R 6 , and R 7 are as described above and P 1 and P 2 are protecting groups may be prepared from the corresponding Formula LX compound by nitration.
  • the Formula LX compound is treated with nitrosyitriflate in a halogenated solvent, such as dichloromethane at a temperature of about -78°C to about 0°C for about 0.5 hour to about 3 hours followed by warming to ambient temperature.
  • a halogenated solvent such as dichloromethane
  • the Formula LXl I amine wherein R 1 , R 3 , R 4 , R 5 , R 6 , and R 7 are as described above and P 1 and P 2 are protecting groups may be prepared from the corresponding Formula LXl compound by reduction.
  • the Formula LXl compound is hydrogenated by treatment with hydrogen gas in the presence of a noble metal catalyst (e.g., palladium on carbon) in a polar solvent such as ethanol at a temperature of about 0°C to about 100°C for about 1 to 24 hours at elevated pressure (e.g., 1 to 3 atmospheres).
  • a noble metal catalyst e.g., palladium on carbon
  • the Formula LXl 11 compound wherein R 1 , R 3 , R 4 , R 5 , R 6 , and R 7 are as described above, P 1 and P 2 are protecting groups and R 8 is an amine linked functionality may be prepared from the corresponding Formula LXII.
  • the Formula LXII amine is derivatized following procedures analogous to those described in Scheme I for the conversion of the Formula III compound to the Formula IV compound.
  • the Formula LXIV compound wherein R 1 , R 3 , R 4 , R 5 , R 6 , and R 7 are as described above and P 1 and P 2 are protecting groups may be prepared from the corresponding Formula LXII compound.
  • the Formula LXII amine is treated with t-butyl nitrate and anhydrous cupric halide in a polar solvent at a temperature of about 30°C to about 100°C for about 1 hour to about 24 hours.
  • halide may be derivatized to a variety of functional groups using standard methods for example as described in Larock or March.
  • Prodrugs of the compounds of Formula I may be prepared according to methods known to those skilled in the art. Exemplary processes are described below.
  • Prodrugs of this invention where a carboxyl group in a carboxylic acid of Formula I is replaced by an ester may be prepared by combining the carboxylic acid with the appropriate alkyl halide in the presence of a base such as potassium carbonate in an inert solvent such as dimethylformamide at a temperature of about 0 to 100°C for about 1 to about 24 hours.
  • the acid is combined with appropriate alcohol as solvent in the presence of a catalytic amount of acid such as concentrated sulfuric acid at a temperature of about 20 to 100°C, preferably at a reflux, for about 1 hour to about 24 hours.
  • Another method is the reaction of the acid with a stoichiometric amount of the alcohol in the presence of a catalytic amount of acid in an inert solvent such as toluene or tetrahydrofuran, with concomitant removal of the water being produced by physical (e.g., Dean-Stark trap) or chemical (e.g., molecular sieves) means.
  • Prodrugs of this invention where an alcohol function has been derivatized as an ether may be prepared by combining the alcohol with the appropriate alkyl bromide or iodide in the presence of a base such as potassium carbonate in an inert solvent such as dimethyiformamide at a temperature of about 0 to 100°C for about 1 to about 24 hours.
  • Alkanoylaminomethyl ethers may be obtained by reaction of the alcohol with a bis-(alkanoylamino)methane in the presence of a catalytic amount of acid in an inert solvent such as tetrahydrofuran, according to a method described in US 4,997,984.
  • these compounds may be prepared by the methods described by Hoffman et al. in J. Org. Chem. 1994, 59, 3530.
  • Glycosides are prepared by reaction of the alcohol and a carbohydrate in an inert solvent such as toluene in the presence of acid. Typically the water formed in the reaction is removed as it is being formed as described above.
  • An alternate procedure is the reaction of the alcohol with a suitably protected glycosyl halide in the presence of base followed by deprotection.
  • N-(l-hydroxyalkyl) amides, N-(1-hydroxy-1-(alkoxycarbonyl)methyl) amides may be prepared by the reaction of the parent amide with the appropriate aldehyde under neutral or basic conditions (e.g., sodium ethoxide in ethanol) at temperatures between 25 and 70°C.
  • N-alkoxymethyl or N-1- (alkoxy)alkyl derivatives can be obtained by reaction of the N-unsubstituted compound with the necessary alkyl halide in the presence of a base in an inert solvent.
  • the compounds of this invention may also be used in conjunction with other pharmaceutical agents (e.g., LDL-cholesterol lowering agents, triglyceride lowering agents) for the treatment of the disease/conditions described herein.
  • other pharmaceutical agents e.g., LDL-cholesterol lowering agents, triglyceride lowering agents
  • they may be used in combination with cholesterol synthesis inhibitors, cholesterol abso ⁇ tion inhibitors, MTP/Apo B secretion inhibitors, and other cholesterol lowering agents such as fibrates, niacin, ion-exchange resins, antioxidants, ACAT inhibitors and bile acid sequestrants.
  • both the compounds of this invention and the other drug therapies are administered to mammals (e.g., humans, male or female) by conventional methods.
  • HMG-CoA reductase inhibitor refers to compounds which inhibit the byconversion of hydroxymethylglutaryl- coenzyme A to mevalonic acid catalyzed by the enzyme HMG-CoA reductase. Such inhibition is readily determined by those skilled in the art according to standard assays (e.g., Meth. Enzymol. 1981 ; 71:455-509 and references cited therein). A variety of these compounds are described and referenced below however other HMG-CoA reductase inhibitors will be known to those skilled in the art.
  • 4,346,227 discloses ML-236B derivatives, such as pravastatin.
  • EP-491226A discloses certain pyridyldihydroxyheptenoic acids, such as rivastatin.
  • U.S. Pat. No. 5,273,995 discloses certain 6- [2-(substituted-pyrrol-1-yl)alkyl]pyran-2-ones such as atorvastatin.
  • MTP/Apo B secretion inhibitor refers to compounds which inhibit the secretion of triglycerides, cholesteryl ester, and phosphoiipids. Such inhibition is readily determined by those skilled in the art according to standard assays (e.g., Wetterau, J. R. 1992; Science 258:999). A variety of these compounds are described and referenced vide however other MTP/Apo B secretion inhibitors will be known to those skilled in the art.
  • WO 96/40640 and WO 98/23593 are two exemplary publications.
  • MTP/Apo B secretion inhibitors are particularly useful: 4'-trifluoromethyl-biphenyl-2-carboxyiic acid [2-(1 H-[1 ,2,4,]triazol-3-yimethyl)-1 ,2,3,4- tetrahydro-isoquinolin-6-y -amide;
  • HMG-CoA synthase inhibitor refers to compounds which inhibit the biosynthesis of hydroxymethylglutaryi-coenzyme A from acetyl-coenzyme A and acetoacetyl-coenzyme A, catalyzed by the enzyme HMG-CoA synthase. Such inhibition is readily determined by those skilled in the art according to standard assays (Meth Enzymol. 1975; 35:155-160: Meth. Enzymol. 1985; 110:19-26 and references cited therein).
  • 4,847,271 discloses certain oxetane compounds such as 11-(3-hydroxymethyl-4-oxo-2-oxetayl)- 3,5,7-trimethyl-2,4-undeca-dienoic acid derivatives.
  • Any compound that decreases HMG-CoA reductase gene expression may be used as the second compound in the combination aspect of this invention.
  • These agents may be HMG-CoA reductase transcription inhibitors that block the transcription of DNA or translation inhibitors that prevent translation of mRNA coding for HMG-CoA reductase into protein.
  • Such compounds may either affect transcription or translation directly, or may be biotransformed to compounds that have the aforementioned activities by one or more enzymes in the cholesterol biosynthetic cascade or may lead to the accumulation of an isoprene metabolite that has the aforementioned activities.
  • Such regulation is readily determined by those skilled in the art according to standard assays (Meth. Enzymol. 1985; 110:9-19).
  • squalene synthetase inhibitor refers to compounds which inhibit the condensation of 2 molecules of famesylpyrophosphate to form squalene, catalyzed by the enzyme squalene synthetase. Such inhibition is readily determined by those skilled in the art according to standard assays (Meth. Enzymol. 1969; 15: 393-454 and Meth. Enzymol. 1985; 110:359-373 and references contained therein). A variety of these compounds are described in and referenced below however other squalene synthetase inhibitors will be known to those skilled in the art. U.S. Pat. No.
  • 5,026,554 discloses fermentation products of the microorganism MF5465 (ATCC 74011) including zaragozic acid.
  • a summary of other patented squalene synthetase inhibitors has been compiled (Curr. Op. Ther. Patents (1993) 861-4).
  • squalene epoxidase inhibitor refers to compounds which inhibit the bioconversion of squalene and molecular oxygen into squalene-2,3-epoxide, catalyzed by the enzyme squalene epoxidase. Such inhibition is readily determined by those skilled in the art according to standard assays (Biochim. Biophys. Acta 1984; 794:466-471 ). A variety of these compounds are described and referenced below, however other squalene epoxidase inhibitors will be known to those skilled in the art. U.S. Pat. Nos.
  • squalene cyciase inhibitor refers to compounds which inhibit the bioconversion of squalene-2,3-epoxide to lanosterol, catalyzed by the enzyme squalene cyclase. Such inhibition is readily determined by those skilled in the art according to standard assays (FEBS Lett. 1989;244:347-350.).
  • the compounds described and referenced below are squalene cyciase inhibitors, however other squalene cyclase inhibitors will also be known to those skilled in the art.
  • PCT publication WO9410150 discloses certain 1 ,2,3,5,6,7,8,8 ⁇ -octahydro-5,5,8 ⁇ (beta)- trimethyl-6-isoquinolineamine derivatives, such as N-trifluoroacetyl-1 ,2,3,5,6,7,8,8 ⁇ - octahydro-2-allyl-5,5,8 ⁇ (beta)-trimethyl-6(beta)-isoquinolineamine.
  • French patent publication 2697250 discloses certain beta, beta-dimethyl-4-piperidine ethanol derivatives such as 1- (1,5,9-trimethyldecyl)-beta,beta-dimethyl-4-piperidineethanol.
  • Any combined squalene epoxidase/squalene cyciase inhibitor may be used as the second component in the combination aspect of this invention.
  • the term combined squalene epoxidase/squalene cyclase inhibitor refers to compounds that inhibit the bioconversion of squalene to lanosterol via a squalene-2,3-epoxide intermediate.
  • EP publication 468,434 discloses certain piperidyl ether and thio-ether derivatives such as 2-(1-piperidyl)pentyl isopentyl sulfoxide and 2-(1-piperidyl)ethyl ethyl sulflde.
  • PCT publication WO 9401404 discloses certain acyl-piperidines such as 1-(1-oxopentyl-5-phenylthio)-4-(2-hydroxy-1 -methyl)- ethy piperidine.
  • U.S. Pat. No. 5,102,915 discloses certain cyclopropyloxy-squalene derivatives.
  • Diasteromeric mixtures can be separated into their individual diastereomers on the basis of their physical chemical differences by methods known per se ⁇ , for example, by chromatography and/or fractional crystallization.
  • Enantiomers can be separated by, for example, chiral HPLC methods or converting the enantiomeric mixture into a diasteromeric mixture by reaction with an appropriate optically active compound (e.g., alcohol), separating the diastereomers and converting (e.g., hydrolyzing) the individual diastereomers to the corresponding pure enantiomers.
  • an appropriate optically active compound e.g., alcohol
  • an enantiomeric mixture of the Formula I compounds or an intermediate in their synthesis which contain an acidic or basic moiety may be separated into their corresponding pure enantiomers by forming a diastereomeric salt with an optically pure chiral base or acid (e.g., 1 -phenyl-ethyl amine or tahtaric acid) and separating the diasteromers by fractional crystallization followed by neutralization to break the salt, thus providing the corresponding pure enantiomers. All such isomers, including diastereomers, enantiomers and mixtures thereof are considered as part of this invention.
  • the compounds of this invention are atropisomers (e.g., substituted biaryls) and are considered as part of this invention.
  • the Formula I compounds of this invention may be obtained in enantiomerically enriched form by resolving the racemate of the final compound or an intermediate in its synthesis (preferably the final compound) employing chromatography (preferably high pressure liquid chromatography [HPLC]) on an asymmetric resin (preferably ChiralcelTM AD or OD [obtained from Chiral Technologies, Exton, Pennsylvania]) with a mobile phase consisting of a hydrocarbon (preferably heptane or hexane) containing between 0 and 50% isopropanol (preferably between 2 and 20 %) and between 0 and 5% of an alkyl amine (preferably 0.1 % of diethylamine). Concentration of the product containing fractions affords the desired materials. ⁇
  • Some of the Formula I compounds of this invention are acidic and they form a salt with a pharmaceutically acceptable cation.
  • Some of the Formula I compounds of this invention are basic and they form a salt with a pharmaceutically acceptable anion. All such salts are within the scope of this invention and they can be prepared by conventional methods such as combining the acidic and basic entities, usually in a stoichiometric ratio, in either an aqueous, non-aqueous or partially aqueous medium, as appropriate.
  • the salts are recovered either by filtration, by precipitation with a non- solvent followed by filtration, by evaporation of the solvent, or, in the case of aqueous solutions, by lyophilization, as appropriate.
  • the compounds can be obtained in crystalline form by dissolution in an appropriate solvent(s) such as ethanol, hexanes or water/ethanol mixtures.
  • the Formula I compounds of this invention are all adapted to therapeutic use as agents that inhibit cholesterol ester transfer protein activity in mammals, particularly humans.
  • the compounds of this invention elevate plasma HDL cholesterol, its associated components, and the functions performed by them in mammals, particularly humans. By virtue of their activity, these agents also reduce plasma levels of triglycerides, LDL cholesterol, VLDL cholesterol and their associated components in mammals, particularly humans.
  • these compounds are useful for the treatment and correction of the various dyslipidemias observed to be associated with the development and incidence of atherosclerosis and cardiovascular disease, including hypoalphalipoproteinemia, hyperbetalipoproteinemia, hypertrigiyceridemia, and familial-hypercholesterolemia.
  • introduction of a functional CETP gene into an animal lacking CETP results in reduced HDL levels (Agellon, L.B., et al: J. Biol. Chem. (1991) 266: 10796-10801.) increased susceptibility to atherosclerosis.(Marotti, K.R., et al: Nature (1993) 364: 73-75.).
  • the Formula I compounds of this invention are useful for the prevention, arrestment and/or regression of atherosclerosis and its associated disease states.
  • cardiovascular disorders e.g., angina, cardiac ischemia and myocardial infarction
  • complications due to cardiovascular disease therapies e.g., reperfusion injury and angioplastic restenosis
  • hypertension stroke, and atherosclerosis associated with organ transplantation.
  • an agent which inhibits CETP activity in humans by virtue of its HDL increasing ability, also provides valuable avenues for therapy in a number of other disease areas as well.
  • mR ⁇ A for CETP is expressed at high levels in adipose tissue.
  • the adipose message increases with fat feeding (Martin, L. J., Connelly, P.
  • HDL cholesteryl ester The uptake of HDL cholesteryl ester is dependent in large part on CETP (Benoist, F., Lau, P., McDonnell, M., Doelle, H., Milne, R. and McPherson, R., 1997. Journal of Biological Chemistry. 272 (38):23572-7).
  • This ability of CETP to stimulate HDL cholesteryl uptake, coupled with the enhanced binding of HDL to adipocytes in obese subjects Jimenez, J. G., Fong, B., Julien, P., Despres, J. P., Rotstein, L, and Angel, A., 1989. International Journal of Obesity.
  • CETP inhibitors are useful in the treatment of inflammation due to Gram- negative sepsis and septic shock.
  • the systemic toxicity of Gram- negative sepsis is in large part due to endotoxin, a lipopoiysaccharide (LPS) released from the outer surface of the bacteria, which causes an extensive inflammatory response.
  • LPS lipopoiysaccharide
  • Lipopoiysaccharide can form complexes with lipoproteins (Ulevitch, R.J., Johhston, A.R., and Weinstein, D.B., 1981. J. Clin. Invest. 67, 827-37).
  • LPS lipoproteins
  • In vitro studies have demonstrated that binding of LPS to HDL substantially reduces the production and release of mediators of inflammation (Ulevitch, R.J., Johhston, A.R., 1978. J. Clin. Invest. 62, 1313-24).
  • Such assays also provide a means whereby the activities of the Formula I compounds of this invention, their prodrugs and the salts of such compounds and prodrugs (or the other agents described herein) can be compared to each other and with the activities of other known compounds. The results of these comparisons are useful for determining dosage levels in mammals, including humans, for the treatment of such diseases.
  • the following protocols can of course be varied by those skilled in the art.
  • the hyperalphachoiesteroiemic activity of the Formula I compounds can be determined by assessing the effect of these compounds on the action of cholesteryl ester transfer protein by measuring the relative transfer ratio of radiolabeled lipids between lipoprotein fractions, essentially as previously described by Morton in J. Biol. Chem. 256, 11992, 1981 and by Dias in Clin. Chem. 34, 2322, 1988.
  • CETP IN VITRO ASSSAY The following is a brief description of the assay of cholesteryl ester transfer in human plasma (in vitro) and animal plasma (ex vivo): CETP activity in the presence or absence of drug is assayed by determining the transfer of 3 H-labeled cholesteryl oleate (CO) from exogenous tracer HDL to the nonHDL lipoprotein fraction in human piasma, or from 3 H-labeled LDL to the HDL fraction in transgenic mouse plasma. Labeled human lipoprotein substrates are prepared similarly to the method described by Morton in which the endogenous CETP activity in plasma is employed to transfer 3 H-CO from phospholipid liposomes to all the lipoprotein fractions in plasma.
  • CO 3 H-labeled cholesteryl oleate
  • 3 H- labeled LDL and HDL are subsequently isolated by sequential ultracentrifugation at the density cuts of 1.019-1.063 and 1.10-1.21 g/ml, respectively.
  • 3 H-labeled lipoprotein is added to plasma at 10-25 nmoles CO/ml and the samples incubated at 37° C for 2.5-3 hrs.
  • Non-HDL lipoproteins are then precipitated by the addition of an equal volume of 20% (wt/vol) polyethylene glycol 8000 (Dias).
  • the samples are centrifuged 750 g x 20 minutes and the radioactivity contained in the HDL containing supernatant determined by liquid scintillation.
  • CETP IN VIVO ASSSAY Activity of these compounds in vivo can be determined by the amount of agent required to be administered, relative to control, to inhibit cholesteryl ester transfer activity by 50% at various time points ex vivo or to elevate HDL cholesterol by a given percentage in a CETP-containing animal species.
  • Transgenic mice expressing both human CETP and human apolipoprotein Al may be used to assess compounds in vivo.
  • the compounds to be examined are administered by oral gavage in an emulsion vehicle containing olive oil and sodium taurocholate. Blood is taken from mice retroorbitally before dosing.
  • CETP activity is determined by a method similar to that described above except that 3 H-cholesteryl oleate containing LDL is used as the donor source as opposed to HDL. The values obtained for iipids and transfer activity are compared to those obtained prior to dosing and/or to those from mice receiving vehicle alone.
  • the activity of these compounds may also be demonstrated by determining the amount of agent required to alter plasma iipid levels, for example HDL cholesterol levels, LDL cholesterol levels, VLDL cholesterol levels or triglycerides, in the plasma of certain mammals, for example marmosets that possess CETP activity and a plasma lipoprotein profile similar to that of humans (Crook et al. Arteriosclerosis 10, 625, 1990).
  • Adult marmosets are assigned to treatment groups so that each group has a similar mean ⁇ SD for total, HDL, and/or LDL plasma cholesterol concentrations. After group assignment, marmosets are dosed daily with compound as a dietary admix or by intragastric intubation for from one to eight days.
  • Plasma total, LDL, VLDL and HDL cholesterol values can be determined at any point during the study by obtaining blood from an antecubital vein and separating plasma lipoproteins into their individual subclasses by density gradient centrifugation, and by measuring cholesterol concentration as previously described (Crook et al. Arteriosclerosis 10, 625, 1990).
  • IN VIVO ATHEROSCLEROSIS ASSAY
  • Anti-atherosclerotic effects of the compounds can be determined by the amount of compound required to reduce the iipid deposition in rabbit aorta.
  • Male New Zealand White rabbits are fed a diet containing 0.2% cholesterol and 10% coconut oil for 4 days (meal-fed once per day). Rabbits are bled from the marginal ear vein and total plasma cholesterol values are determined from these samples. The rabbits are then assigned to treatment groups so that each group has a similar mean ⁇ SD for total plasma cholesterol concentration, HDL cholesterol concentration, triglyceride concentration and/or cholesteryl ester transfer protein activity. After group assignment, rabbits are dosed daily with compound given as a dietary admix or on a small piece of gelatin based confection.
  • Control rabbits receive only the dosing vehicle, be it the food or the gelatin confection.
  • the cholesterol/coconut oil diet is continued along with the compound administration throughout the study.
  • Plasma cholesterol values and cholesteryl ester transfer protein activity can be determined at any point during the study by obtaining blood from the marginal ear vein.
  • the rabbits are sacrificed and the aortae are removed from the thoracic arch to the branch of the iliac arteries.
  • the aortae are cleaned of adventitia, opened longitudinally and then stained with Sudan IV as described by Holman et. al. (Lab. Invest. 1958, 7, 42-47).
  • the percent of the surface area stained is quantitated by densitometry using an Optimas Image Analyzing System (Image Processing Systems). Reduced lipid deposition is indicated by a reduction in the percent surface area stained in the compound-receiving group in comparison with the control rabbits.
  • ANTIOBESITY PROTOCOL The ability of CETP inhibitors to cause weight loss can be assessed in obese human subjects with body mass index (BMI) > 30 kg/m 2 . Doses of inhibitor are administered sufficient to result in an increase of > 25% in HDL cholesterol levels. BMI and body fat distribution, defined as waist (W) to hip (H) ratio (WHR), are monitored during the course of the 3-6 month studies, and the results for treatment groups compared to those receiving placebo.
  • the number of surviving mice is determined at times up to 48h after LPS injection and compared to those mice administered vehicle (minus CETP inhibitor) only.
  • Administration of the compounds of this invention can be via any method which delivers a compound of this invention systemically and/or locally. These methods include oral routes, parenteral, intraduodenal routes, etc.
  • parenteral administration e.g., intravenous, intramuscular, subcutaneous or intramedullary
  • parenteral administration may be utilized, for example, where oral administration is inappropriate for the target or where the patient is unable to ingest the drug.
  • an amount of a compound of this invention is used that is sufficient to achieve the therapeutic effect desired (e.g., HDL elevation).
  • an effective dosage for the Formula I compounds of this invention, their prodrugs and the salts of such compounds and prodrugs is in the range of 0.01 to 10 mg/kg/day, preferably 0.1 to 5 mg/kg/day.
  • a dosage of the combination pharmaceutical agents to be used in conjuction with the CETP inhibitors is used that is effective for the indication being treated.
  • typically an effective dosage for HMG-CoA reductase inhibitors is in the range of 0.01 to 100 mg/kg/day.
  • an effect dosage for the MTP/Apo B secretion inhibitors is in the range of 0.01 to 100 mg/kg/day.
  • the compounds of the present invention are generally administered in the form of a pharmaceutical composition comprising at least one of the compounds of this invention together with a pharmaceutically acceptable vehicle, diluent or carrier.
  • a pharmaceutically acceptable vehicle diluent or carrier.
  • the compounds of this invention can be administered individually or together in any conventional oral, parenteral, rectal or transdermal dosage form.
  • a pharmaceutical composition can take the form of solutions, suspensions, tablets, pills, capsules, powders, and the like.
  • Tablets containing various excipients such as sodium citrate, calcium carbonate and calcium phosphate are employed along with various disintegrants such as starch and preferably potato or tapioca starch and certain complex silicates, together with binding agents such as polyvinylpyrrolidone, sucrose, gelatin and acacia.
  • binding agents such as polyvinylpyrrolidone, sucrose, gelatin and acacia.
  • lubricating agents such as magnesium stearate, sodium lauryl sulfate and talc are often very useful for tabletting pu ⁇ oses.
  • Solid compositions of a similar type are also employed as fillers in soft and hard-filled gelatin capsules; preferred materials in this connection also include lactose or milk sugar as well as high molecular weight polyethylene glycols.
  • a preferred formulation is a solution or suspension in an oil, for example olive oil, MiglyolTM or CapmulTM, in a soft gelatin capsule.
  • Antioxidants may be added to prevent long term degradation as appropriate.
  • the compounds of this invention can be combined with various sweetening agents, flavoring agents, coloring agents, emulsifying agents and/or suspending agents, as well as such diiuents as water, ethanol, propylene glycol, glycerin and various like combinations thereof.
  • solutions in sesame or peanut oil or in aqueous propylene giycol can be employed, as well as sterile aqueous solutions of the corresponding water-soluble salts.
  • aqueous solutions may be suitably buffered, if necessary, and the liquid diluent first rendered isotonic with sufficient saline or glucose.
  • aqueous solutions are especially suitable for intravenous, intramuscular, subcutaneous and intraperitoneal injection pu ⁇ oses.
  • the sterile aqueous media employed are all readily obtainable by standard techniques well-known to those skilled in the art.
  • aqueous or partially aqueous solutions are prepared.
  • compositions according to the invention may contain 0.1%- 95% of the compound(s) of this invention, preferably 1 %-70%.
  • the composition or formulation to be administered will contain a quantity of a compound(s) according to the invention in an amount effective to treat the disease/condition of the subject being treated, e.g., atherosclerosis.
  • kits comprises two separate pharmaceutical compositions: a compound of Formula I a prodrug thereof or a salt of such compound or prodrug and a second compound as described above.
  • the kit comprises means for containing the separate compositions such as a container, a divided bottle or a divided foil packet.
  • the kit comprises directions for the administration of the separate components.
  • the kit form is particularly advantageous when the separate components are preferably administered in different dosage forms (e.g., orai and parenteral), are administered at different dosage intervals, or when titration of the individual components of the combination is desired by the prescribing physician.
  • Blister packs are well known in the packaging industry and are being widely used for the packaging of pharmaceutical unit dosage forms (tablets, capsules, and the like). Blister packs generally consist of a sheet of relatively stiff material covered with a foil of a preferably transparent plastic material. During the packaging process recesses are formed in the plastic foil. The recesses have the size and shape of the tablets or capsules to be packed. Next, the tablets or capsules are placed in the recesses and the sheet of relatively stiff material is sealed against the plastic foil at the face of the foil which is opposite from the direction in which the recesses were formed. As a result, the tablets or capsules are sealed in the recesses between the plastic foil and the sheet. Preferably the strength of the sheet is such that the tablets or capsules can be removed from the blister pack by manually applying pressure on the recesses whereby an opening is formed in the sheet at the place of the recess. The tablet or capsule can then be removed via said opening.
  • a memory aid on the kit, e.g., in the form of numbers next to the tablets or capsules whereby the numbers correspond with the days of the regimen which the tablets or capsules so specified should be ingested.
  • a memory aid is a calendar printed on the card, e.g., as follows "First Week, Monday, Tuesday, ...etc.... Second Week, Monday, Tuesday, etc.
  • a “daily dose” can be a single tablet or capsule or several pills or capsules to be taken on a given day.
  • a daily dose of Formula I compound can consist of one tablet or capsule whiie a daily dose of the second compound can consist of several tablets or capsules and vice versa.
  • a dispenser designed to dispense the daily doses one at a time in the order of their intended use is provided.
  • the dispenser is equipped with a memory-aid, so as to further facilitate compliance with the regimen.
  • a memory-aid is a mechanical counter which indicates the number of daily doses that has been dispensed.
  • a battery-powered micro-chip memory coupied with a liquid crystal readout, or audible reminder signal which, for example, reads out the date that the last daily dose has been taken and/or reminds one when the next dose is to be taken.
  • the compounds of this invention either alone or in combination with each other or other compounds generally will be administered in a convenient formulation. The following formulation examples only are illustrative and are not intended to limit the scope of the present invention.
  • active ingredient means a compound of this invention.
  • Hard gelatin capsules are prepared using the following: Ingredient Quantity (mg/capsule)
  • a tablet formulation is prepared using the ingredients below: Formulation 2: Tablets
  • Stearate acid 5-15 The components are blended and compressed to form tablets.
  • tablets each containing 0.25-100 mg of active ingredients are made up as follows: Formulation 3: Tablets
  • the active ingredients, starch, and cellulose are passed through a No. 45 mesh U.S. sieve and mixed thoroughly.
  • the solution of polyvinylpyrroiidone is mixed with the resultant powders which are then passed through a No. 14 mesh U.S. sieve.
  • the granules so produced are dried at 50° - 60°C and passed through a No. 18 mesh U.S. sieve.
  • the sodium carboxymethyl starch, magnesium stearate, and talc previously passed through a No. 60 U.S. sieve, are then added to the granules which, after mixing, are compressed on a tablet machine to yield tablets.
  • Aerosol solution is prepared containing the following ingredients: Formulation 5: Aerosol
  • Propellant 22 (Chlorodifluoromethane) 70.00
  • the active ingredient is mixed with ethanol and the mixture added to a portion of the propellant 22, cooled to 30°C, and transferred to a filling device. The required amount is then fed to a stainless steel container and diluted with the remaining propellant. The valve units are then fitted to the container.
  • Suppositories are prepared as follows: Formulation 6: Suppositories
  • the active ingredient is passed through a No. 60 mesh U.S. sieve and suspended in the saturated fatty acid glycerides previously melted using the minimal necessary heat. The mixture is then poured into a suppository mold of nominal 2 g capacity and allowed to cool.
  • An intravenous formulation is prepared as follows: Formulation 7: Intravenous Solution
  • IntraiipidTM emulsion 1 ,000 mL The solution of the above ingredients is intravenously administered to a patient at a rate of about 1 mL per minute.
  • Soft gelatin capsules are prepared using the following: Formulation 8: Soft Gelatin Capsule with Oil Formulation
  • the active ingredient above may also be a combination of agents.
  • the expected intensity ratio was observed (approximately 3:1 for 35 CI/ 37 CI-containing ions) and 1 :1 for ⁇ Br/ ⁇ Br-containing ions) and the intensity of only the lower mass ion is given.
  • concentrate and evaporated refer to removal of solvent at water aspirator pressure on a rotary evaporator with a bath temperature of less than 45°C. Reactions conducted at “0-20°C” or “0-25°C” were conducted with initial cooling of the vessel in an insulated ice bath which was allowed to warm to room temperature over several hours. The abbreviation “min” and “h” stand for “minutes” and “hours” respectively.
  • Example 1 c/s-4-Benzyloxycarbonylamino-6-methoxy-2-methyl-1.2.3.4-tetrahvdro-quinoline-7- carboxylic acid methyl ester: To a solution of 4.26g methyl 2-methoxy-5- aminobenzoate (23.5 mmol) in 65 ml anhydrous dichloromethane was added 3.34g anhydrous sodium sulfate, and the resulting mixture was cooled to -25°C. Freshly distilled acetaldehyde (1.04g, 23.5 mmol) was added and the mixture was stirred at - 25°C for 2h.
  • Example 3 c/ ' s-4-Amino-6-methoxy-2-methyl-3.4-dihydro-2H-quinoline-1.7-dicarboxylic acid 1- ethyl ester 7-methyl ester: cs-4-Benzyioxycarbonyiamino-6-methoxy-2-methyl-3,4- dihydro-2H-quinoline-1 , 7-dicarboxylic acid 1 -ethyl ester 7-methyi ester (322 mg, 0.71 mmol), 10% palladium on carbon (32 mg) and cyclohexene (9ml) in 16 ml ethanol was heated to 80 °C for 1.25 h.
  • Example 4 c/s-4-(3.5-Bis-trifluoromethyl-benzylaminoV6-methoxy-2-methyl-3.4-dihydro-2H- quinoiine-1.7-dicarboxylic acid 1-ethyl ester 7-methyl ester: To a solution of 4-amino- 6-methoxy-2-methyl-3,4-dihydro-2H-quinoline-1 ,7-dicarboxyiic acid 1 -ethyl ester 7- methyl ester (208 mg, 0.64 mmol) in anhydrous 1 ,2-dichloroethane (5 mL) was added acetic acid (0.041 mL, 0.71 mmol), followed by 3,5-bis(trifluoromethyl)benzaldehyde (172 mg, 0.71 mmol) and sodium triacetoxyborohydride (0.205 g, 0.97 mmol).
  • Example 5 2-Methyl-4-oxo-6-trifluoromethyl-3.4-dihydro-2H-quinoiine-1 -carboxylic acid benzyl ester.
  • 4-Methoxy-6-trifluoromethylquinoline (5.0 g, 22.0 mmol) was dissolved in anhydrous tetrahydrofuran (20 mL). The mixture was cooled to -78 °C, and methyl magnesium chloride (73 mL of a 3.0M solution in tetrahydrofuran, 220 mmol) was added to the resulting slurry. After addition, benzyl chloroformate (41 mL, 286 mmol) was added to the slurry.
  • Example 6 8-Chloro-2-methyl-4-oxo-3.4-dihydro-2H-quinoline-1 -carboxylic acid ethyl ester.
  • 8- Chloro-4-methoxyquinoline (1.0 g, 5.2 mmol, dissolved in 6 mL of anhydrous tetrahydrofuran) was added dropwise via cannula to a solution of methyllithium (18.4 mL of a 1.4 M solution in diethyl ether) cooled with an ice/water bath. After 45 min, ethyl chloroformate (4.9 mL, 51.7 mmol) was added to the reaction mixture via syringe.
  • reaction mixture was quenched with 60 mL of a 1 N aqueous HCI solution, 100 mL of THF and 20 mL of methanol. After 3 days, the tetrahydrofuran was removed in vacuo , and the remaining aqueous phase was extracted with ethyl acetate (3 x 100 mL). The organic phases were combined and washed with brine, dried over sodium sulfate, filtered, and concentrated in vacuo to give 1.5 g of crude product. Purification by silica gel chromatography using 0-20% ethyl acetate/hexanes as eiuent afforded 0.79 g of the title product (57%).
  • 6-Fluoro-2- methyl-4-oxo-7-trifluoromethyl-3,4-dihydro-2H-quinoline-1 -carboxylic acid benzyl ester (3.0 g, 7.9 mmol) was combined with 250 mg of 10% Pd/C in 50 mL of ethanol and agitated under a hydrogen atmosphere (40 psi) for 1 hour before being filtered through Celite®, rinsing with ethyl acetate, and evaporating the volatiies under reduced pressure to afford 1.34 (69%) of the title compound as a colorless oil.
  • 6-Fluoro-2-methyl-4-oxo-7-trifluoromethyl-3,4-dihydro-2H-quinoline (1.01 g, 4.08 mmol) was dissolved in 10 mL of dichloromethane and 5 mL of pyridine, and stirred as isopropyl chloroformate (48 mL of a 1 molar solution in toluene, 48 mmol) was added slowly via syringe.
  • 6-Fluoro-4-hvdroxyimino-2-methyl-7-trifluoromethyl-3.4-dihydro-2H-quinoline-1- carboxylic acid isopropyl ester A stirred solution of 6-fluoro-2-methyl-4-oxo-7- trifluoromethyl-3,4-dihydro-2H-quinoline-1 -carboxylic acid isopropyl ester (940 mg, 2.8 mmol), hydroxylamine hydrochloride (215 mg, 3.1 mmoi), and sodium acetate (254 mg, 3.1 mmol) in ethanol (20 mL) was heated at reflux for 3 h. Water (25 mL) was added, and the volatiies were removed in vacuo.
  • Example 12 cis and frans-4-Amino-6-fluoro-2-methyl-7-trifluoromethyl-3.4-dihydro-2H-quinoline-1 - carboxylic acid isopropyl ester.
  • 6-fluoro-4-hydroxyimino-2- methyl-7-trifluoromethyl-3,4-dihydro-2H-quinoline-1 -carboxylic acid isopropyl ester 937 mg, 2.82 mmoi
  • ethanol 40 mL
  • aqueous 2N KOH 40 mL, 80 mmol
  • aluminum-nickel alloy (1.57 g, 1 :1 by weight, 18 mmol
  • Example 15 c/s-4-[(3.5-Bis-trifluoromethyl-benzy ⁇ -methoxycarbonyl-amino]-7- ethoxycarbonylmethyl-6-methoxy-2-methyl-3.4-dihvdro-2H-quinoline-1 -carboxylic acid ethyl ester. MS m/z 634.5 (M + ); 1 H NMR (CDCI 3 ) ⁇ 3.6 (AB. 2H), 3.8 (s, 3H), 6.4
  • Example 21 c/s-4-[(3.5-Bis-trifluoromethyl-benzy ⁇ -methoxycarbonyl-amino]-6- methanesulfonylmethyl-7-methoxy-2-methyl-3.4-dihydro-2H-quinoline-1-carboxylic acid ethyl ester.
  • MS m/z 641 (M + + 1 ); 1 H NMR (CDCI 3 ) ⁇ 6.88 (s, 1 H), 3.81 (s, 3H).
  • Example 23 c/s-4-[(3.5-Bis-trifluoromethyl-benzy ⁇ -methoxycarbonyl-amino]-6-(methoxycarbonyl- methyl-amino)-2-methyl-3.4-dihvdro-2H-quinoline-1 -carboxylic acid ethyl ester.
  • Example 26 c/s-4-[(3.5-Bis-trifluoromethyl-ben -- yl)-meth ⁇ xycarbonyl-amino]-6-fluoro-7-methoxy-2- methyl-3.4-dihvdro-2H-quinoline-1 -carboxylic acid ethyl ester. MS m/z 567 (M + + 1 ),
  • Example 27 c/s-4-[(3.5-Bis-trifluoromethyl-ben--y ⁇ -methoxycarbonyl-amino]-6-methoxy-7-(1- methoxycarbonyl-cvclopentyl)-2-methyl-3.4-dihvdro-2H-quinoline-1-carboxylic acid ethyl ester.
  • Example 33 c/s-4-[(3.5-Bis-trifluoromethyl-benzvh-methoxycarbonyl-amino1-8-chloro-2-methyl-3.4- dihvdro-2H-quinoline-1 -carboxylic acid ethyl ester. MS m/z 552 (M + ), 569 (M + + 17);
  • Example 34 c/s-4-[(3.5-Bis-trifluoromethyl-benzv ⁇ -methoxycarbonyl-amino]-6.7-bis-tert- butoxycarbonylamino-2-methyl-3.4-dihvdro-2H-quinoline-1 -carboxylic acid ethyl ester. MS m/z 618 (M + - 130); 1 H NMR (CDCI 3 ) ⁇ 3.8 (s, 3H), 7.4 (s, 1H).
  • Example 35 cs-4-[(3.5-Bis-trifluoromethyl-benzvi)-methoxycarbonyl-amino]-2.6-dimethyl-3.4- dihvdro-2H-quinoline-1 -carboxyiic acid ethyl ester. MS m/z 532 (M + ), 533 (M + + 1 );
  • Example 38 cs-4-[(3.5-Bis-trifluoromethyl-benzv ⁇ -methoxycarbonyl-amino]-6-(2-ethoxycarbonyl- ethylV2-methyl-3.4-dihydro-2H-quinoline-1 -carboxylic acid ethyl ester.
  • Example 50 c/s-4-[(3.5-Bis-trifluoromethyl-benzyh-methoxycarbonyl-amino]-2.6.8-trimethyl-3.4- dihydro-2H-quinoline-1 -carboxylic acid ethyl ester.
  • Example 51 Example 51.
  • Example 13 Prepared from c/s-4- [(3,5-Bis-trifluoromethyl-benzyl)-methoxycarbonyl-amino]-6-methoxy-2-methyl-3,4- dihydro-2H-quinoline-1 , 7-dicarboxylic acid 1 -ethyl ester 7-methyl ester (Example 13) in a manner analogous to Example 60.
  • Example 62 Example 62.
  • Examples 63-69 were prepared in an analogous manner to Example 62 substituting with the appropriate amine from either Example 60 or Example 61.
  • Example 63. c/s-4-[(3.5-Bis-trifluoromethyl-benzylVmethoxycarbonyl-amino]-6-[2-(3H-imidazol-4- v ⁇ -ethvicarbamoyl]-7-methoxy-2-methyl-3.4-dihydro-2H-quinoline-1-carboxylic acid ethyl ester.
  • MS m z 686 (M + + 1 ); 1 H NMR (CDCI 3 ) ⁇ 3.74 (s, 3H), 7.18 (s, 1H).
  • Example 56 (2.8 g, 4.3 mmol), ethanol (50 mL), and 10% palladium on carbon (280 mg) were combined in a Parr bottle and agitated under 50 psi of hydrogen gas on a Parr shaker for 0.5 h. The mixture was then filtered through a bed of Celite®, eluting with ethyl acetate, and the filtrate concentrated in vacuo .
  • Examples 71 and 72 were prepared from the indicated starting compounds in an analogous manner to Example 70.
  • Example 71. c/s-(3.5-Bis-trifluoromethvi-benzv ⁇ - (6-chloro-7-trifluoromethyl -2-methyt-1.2.3.4- tetrahydro-quinolin-4-ylVcarbamic acid methyl ester. Prepared from c/s-4-[(3,5-bis- trifluoromethyl-benzyl)-methoxycarbonyl-amino]-6-chloro-2-methyl-7-trifluoromethyl- 3,4-dihydro-2H-quinoline-1 -carboxylic acid benzyl ester.
  • Example 72 c/s-(3.5-Bis-trifluoromethyl-benzylV (7-trifluoromethyl -2-methyl-1.2.3.4-tetrahydro- quinolin-4-y0-carbamic acid methyl ester.
  • Examples 74-84 were prepared in an analogous manner to Example 73 from the appropriate amine from Examples 70-72.
  • Example 74. c/s-4-[(3.5-Bis-trifluoromethyl-benzy ⁇ -methoxycarbonyl-amino]-2-methyl-6- trifluoromethyl-3.4-dihydro-2H- ⁇ uinoline-1 -carboxyiic acid tert-butyl ester.
  • Example 83 c/s-(3.5-Bis-trifluoromethyl-benzv ⁇ -(1-isopropylcarbamoy ⁇ -2-methyl-6-trifluoromethyl- 1.2.3.4-tetrahvdro- ⁇ uinolin-4-y ⁇ -carbamic acid methyl ester. MS m/z 600 (M + + H); 1 H NMR (CDCI 3 ) ⁇ 7.82 (bs, 1H), 7.8 (bs, 1H), 7.7 (bs, 1 H), 7.2 (bs, 1H), 6.7 (bs, 1H), 3.8 (s, 3H), 3.75 (bs, 3H).
  • Example 84 c/s-(3.5-Bis-trifluoromethyl-benzv ⁇ -(1-isopropylcarbamoy ⁇ -2-methyl-6-trifluoromethyl- 1.2.3.4-tetrahvdro- ⁇ uinolin-4-y ⁇ -carbamic acid methyl ester. MS m/
  • Examples 87-89 were prepared in an analogous manner to Example 86 from the indicated starting esters.
  • Example 87. c/s-4-[(3.5-Bis-trifluoromethyl-benzy ⁇ -methoxycarbonyl-am ⁇ no]-7-(2-hydroxy-ethv ⁇ -6- methoxy-2-methyl-3.4-dihydro-2H-quinoline-1 -carboxyiic acid ethyl ester.
  • Example 90 c/s-4-[(3.5-Bis-trifluoromethyl-benzyl)-methoxycarbonvi-amino]-7-bromomethyl-6- methoxy-2-methyi-3.4-dihydro-2H-quinoline-1 -carboxylic acid ethyl ester.
  • triphenylphosphine 0.57 g, 1.74 mmol
  • bromine 84 ⁇ L, 1.6 mmoi
  • Examples 93-102 were prepared in an analogous manner to Example 92 employing the requisite bromide (Example 90 or 91) and the appropriate alkoxide or thioalkoxide. -
  • Example 93 c/s-4-[(3.5-Bis-trifluoromethyl-benzy ⁇ -methoxycarbonyl-amino]-7-ethyisulfanylmethyl- 6-methoxy-2-methyl-3.4-dihydro-2H-quinoline-1 -carboxyiic acid ethyl ester.
  • 1 H NMR (CDCI 3 ) ⁇ 1.1 (d, 3H), 2.55 (q, 2H), 3.7 (s, 3H), 6.4 (s, 1 H), 7.3 (s, 1 H), 7.7 (d, 2H), 7.8 (s, 1 H).
  • Example 94 Example 94.
  • Example 99 c/s-4-[(3.5-Bis-trifluoromethyl-benzyl)-methoxycarbonyl-amino]-7-methoxy-6-(2- methoxy-ethoxymethv ⁇ -2-methyl-3.4-dihydro-2H-quinoline-1 -carboxylic acid ethyl ester.
  • Example 100 c/s-4-[(3.5-Bis-trifluoromethyl-benzyl)-methoxycarbonyl-amino]-7-methoxy-2-methyl- 6-(1-methyl-1H-tetrazol-5-ylsulfanylmethyl)-3.4-dihydro-2H-quinoline-1 -carboxylic acid ethyl ester.
  • MS m/z 694 (M + + NH4 + ); 1 H NMR (CDCI 3 ) ⁇ 3.76 (s, 3H), 6.99 (s,
  • Example 101 c/s-4-[(3.5-Bis-trifluoromethyl-benzy ⁇ -methoxycarbonyl-amino]-6-methoxy-7-(2- methoxy-ethoxymethyl)-2-methyl-3.4-dihydro-2H-quinoline-1 -carboxylic acid ethyl ester.
  • MS m z 561 (M + -75); 1 H NMR (CDCI 3 ) ⁇ 1.1 (d, 3H), 1.3 (t, 3H), 3.6 (s, 3H),
  • Example 102 c/s-4-[(3.5-Bis-trifluoromethyl-benzyl -methoxycarbonyl-amino]-7- propylsulfanylmethvi-6-methoxy-2-methyl-3.4-dihydro-2H-quinoiine-1-carboxylic acid ethyl ester.
  • 1 H NMR (CDCI 3 ) ⁇ 1.0 (t, 3H), 1.1 , (d, 3H), 1.3 (t, 3H), 2.5 (t, 2H), 3.7 (s,
  • Example 103 c/s-4-[(3.5-Bis-trifluoromethyl-benzy ⁇ -methoxycarbonyl-amino]-6-methoxy-7- methoxymethyl-2-methyl-3.4-dihvdro-2H-quinoline-1 -carboxyiic acid ethyl ester.
  • Examples 104 and 105 were prepared in analogous manner to Example 103 from the indicated starting alcohol.
  • Example 104. cs-4-[(3.5-Bis-trifluoromethyl-benzyl)-methoxycarbonyl-amino]-7-methoxy-6- methoxymethyl-2-methyl-3.4-dihydro-2H-quinoline-1 -carboxyiic acid ethyl ester.
  • Example 107 c/s-4-[(3,5-Bis-trifluoromethyl-benzyl)- methoxycarbonyi-amino]-6-chloro-2-methyl-8-nitro-3,4-dihydro-2H-quinoline-1- carboxylic acid ethyl ester (Example 107) (0.747 g, 1.25 mmol), ethanol (20 mL), and 10% palladium on carbon (0.40 g) were combined and agitated under 36 psi of H 2 on a Parr shaker for 2 h and 15 min. The mixture was then filtered through a bed of Celite®. The Celite® was washed with ethanol, and the filtrate was concentrated in vacuo.
  • Example 110 cis- 8-Amino-4-[(3.5-bis-trifluoromethyl-benzv ⁇ -methoxycarbonyl-amino] -2-methyl -6- trifluoromethyl-3.4-dihvdro-2H-quinoline-1 -carboxylic acid isopropyl ester.
  • Example 111 Prepared as in Example 109 from c/s-4-[(3,5-bis-trifiuoromethyl-benzyl)-methoxycarbonyl- amino]-6-trifluoromethyl-2-methyl-8-nitro-3,4-dihydro-2H-quinoline-1-carboxylic acid isopropyl ester (Example 108) MS m/z 616 (M + + H); 1 H NMR (CDCI 3 ) ⁇ 7.8 (bs, 1 H), 7.65 (bs, 2H), 7.62 (bs, 1 H), 6.8 (bs, 1 H), 3.8 (s, 3H).
  • Example 111 Example 111.
  • Example 116 [2R.4S] 4-[(3.5-Bis-trifluoromethyl-benzy ⁇ -methoxycarbonyl-amino]-2-methyl-6- trifluoromethyl-3.4-dihydro-2H-quinoline-1 -carboxylic acid propyl ester.
  • Example 75

Abstract

Cholesteryl ester transfer protein inhibitors of formula (I), pharmaceutical compositions containing such inhibitors and the use of such inhibitors to elevate certain plasma lipid levels, including high density lipoprotein-cholesterol and to lower certain other plasma lipid levels, such as LDL-cholesterol and triglycerides and accordingly to treat diseases which are exacerbated by low levels of HDL cholesterol and/or high levels of LDL-cholesterol and triglycerides, such as atherosclerosis and cardiovascular diseases in some mammals, including humans.

Description

-CARB0XYAMIN0-2-METHYL-l,2,3,4-TETRAHYDR0QUIN0LINES AS CETP INHIBITORS
BACKGROUND OF INVENTION This invention relates to cholesteryl ester transfer protein (CETP) inhibitors, pharmaceutical compositions containing such inhibitors and the use of such inhibitors to elevate certain plasma lipid levels, including high density lipoprotein (HDL)-cholesterol and to lower certain other plasma lipid levels, such as low density lipoprotein (LD )-cholesteroi and triglycerides and accordingly to treat diseases which are affected by low levels of HDL cholesterol and/or high levels of LDL-cholesterol and triglycerides, such as atherosclerosis and cardiovascular diseases in certain mammals (i.e., those which have CETP in their plasma), including humans.
Atherosclerosis and its associated coronary artery disease (CAD) is the leading cause of mortality in the industrialized world. Despite attempts to modify secondary risk factors (smoking, obesity, lack of exercise) and treatment of dyslipidemia with dietary modification and drug therapy, coronary heart disease (CHD) remains the most common cause of death in the U.S., where cardiovascular disease accounts for 44% of all deaths, with 53% of these associated with atherosclerotic coronary heart disease.
Risk for development of this condition has been shown to be strongly correlated with certain plasma lipid levels. While elevated LDL-C may be the most recognized form of dyslipidemia, it is by no means the only significant lipid associated contributor to CHD. Low HDL-C is also a known risk factor for CHD (Gordon, D.J., et al.,: "High-density Lipoprotein Cholesterol and Cardiovascular Disease", Circulation, (1989), 79: 8-15). High LDL-cholesterol and triglyceride levels are positively correlated, while high levels of HDL-cholesterol are negatively correlated with the risk for developing cardiovascular diseases. Thus, dyslipidemia is not a unitary risk profile for CHD but may be comprised of one or more lipid aberrations.
Among the many factors controlling plasma levels of these disease dependent principles, cholesteryl ester transfer protein (CETP) activity affects all three. The role of this 70,000 dalton plasma glycoprotein found in a number of animal species, including humans, is to transfer cholesteryl ester and triglyceride between lipoprotein particles, including high density lipoproteins (HDL), low density lipoproteins (LDL), very low density iipoproteins (VLDL), and chylomicrons. The net result of CETP activity is a lowering of HDL cholesterol and an increase in LDL cholesterol. This effect on lipoprotein profile is believed to be pro-atherogenic, especially in subjects whose lipid profile constitutes an increased risk for CHD.
No wholly satisfactory HDL-eievating therapies exist. Niacin can significantly increase HDL, but has serious toleration issues which reduce compliance. Fibrates and the HMG CoA reductase inhibitors raise HDL-C only modestly (-10-12%). As a result, there is a significant unmet medical need for a well-tolerated agent which can significantly elevate plasma HDL levels, thereby reversing or slowing the progression of atherosclerosis.
Thus, although there are a variety of anti-atherosclerosis therapies, there is a continuing need and a continuing search in this field of art for alternative therapies.
EP0818448 (970624) discloses the preparation of certain 5,6,7,8 substituted tetrahydroquinolines and analogs as cholesteryl ester transfer protein inhibitors.
U.S. Pat. No. 5,231 ,102 discloses a class of 4-substituted 1 ,2,3,4- tetrahydroquinolines that possess an acidic group (or group convertible thereto in vivo) at the 2-position that are specific antagonists of N-methyl-D-aspartate (NMDA) receptors and are therefore useful in the treatment and/or prevention of neurodegenerative disorders.
U.S. Pat. No. 5,288,725 discloses pyrroloquinoline bradykinin antagonists. SUMMARY OF THE INVENTION
This invention is directed to compounds of Formula I
Figure imgf000004_0001
prodrugs thereof, and pharmaceutically acceptable salts of said compounds and said prodrugs; wherein R1 is hydrogen, Y, W-X, W-Y; wherein W is a carbonyl, thiocarbonyl, sulfinyl or sulfonyl; X is -ON, -S-Y, -Ν(H)-Y or -N-(Y)2; wherein Y for each occurrence is independently Z or a fully saturated, partially unsaturated or fully unsaturated one to ten membered straight or branched carbon chain wherein the carbons, other than the connecting carbon, may optionally be replaced with one or two heteroatoms selected independently from oxygen, sulfur and nitrogen and said carbon is optionally mono-, di- or tri-substituted independently with halo, said carbon is optionally mono-substituted with hydroxy, said carbon is optionally mono-substituted with oxo, said sulfur is optionally mono- or di-substituted with oxo, said nitrogen is optionally mono-, or di-substituted with oxo, and said carbon chain is optionally mono-substituted with Z;
Z is a partially saturated, fully saturated or fully unsaturated three to twelve membered ring optionally having one to four heteroatoms selected independently from oxygen, sulfur and nitrogen, or a bicyclic ring consisting of two fused partially saturated, fully saturated or fully unsaturated three to six membered rings, taken independently, optionally having one to four heteroatoms selected independently from nitrogen, sulfur and oxygen; wherein said Z substituent is optionally mono-, di- or tri-substituted independently with halo, (C2-C6)alkenyl, (C C6) alkyl, hydroxy, (C1-C6)alkoxy, (C,-C )alkylt.hio, amino, nitro, cyano, oxo, carboxy, (C C6)alkyloxycarbonyl, mono-N- or di-N,N-(C C6)alkylamino wherein said (C1-C6)alkyl substituent is optionally mono-, di- or tri-substituted independently with halo, hydroxy, (C C6)alkoxy, (C C4)alkylthio, amino, nitro, cyano, oxo, carboxy, (C C6)alkyioxycarbonyl, mono-N- or di-N,N-(C C6)alkylamino, said (C C6)alkyl optionally substituted with from one to nine fluorines; R3 is hydrogen or Q; wherein Q is a fully saturated, partially unsaturated or fully unsaturated one to six membered straight or branched carbon chain wherein the carbons, other than the connecting carbon, may optionally be replaced with one heteroatom selected from oxygen, sulfur and nitrogen and said carbon is optionally mono-, di- or tri-substituted independently with halo, said carbon is optionally mono-substituted with hydroxy, said carbon is optionally mono- substituted with oxo, said sulfur is optionally mono- or di-substituted with oxo, said nitrogen is optionally mono- or di-substituted with oxo, and said carbon chain is optionally mono-substituted with V; wherein V is a partially saturated, fully saturated or fully unsaturated three to twelve membered ring optionally having one to four heteroatoms selected independently from oxygen, sulfur and nitrogen, or a bicyciic ring consisting of two fused partially saturated, fully saturated or fully unsaturated three to six membered rings, taken independently, optionally having one to four heteroatoms selected independently from nitrogen, sulfur and oxygen; wherein said V substituent is optionally mono-, di-, tri-, or tetra- substituted independently with halo, (C C6)alkyl, (C2-C6)alkenyl, hydroxy, (C C6)alkoxy, (C C4)alkylthio, amino, nitro, cyano, oxo, carboxamoyl, mono-N- or di-N,N-(C C6) alkylcarboxamoyl, carboxy, (C1-C6)alkyioxycarbonyl, mono- N- or di-N,N-(C C6)alkylamino wherein said (C C6)alkyl or (C2-C6)alkenyl substituent is optionally mono-, di- or tri-substituted independently with hydroxy, (C C6)alkoxy, (C C4)alkylthio, amino, nitro, cyano, oxo, carboxy, (C-rC^alkyloxycarbonyl, mono-N- or di-N,N-(C1-C6)alkylamino or said (C C6)alkyi or (C2-C6)alkenyl optionally substituted with from one to nine fluorines; R4 is Q1 or V1 wherein Q1 a fully saturated, partially unsaturated or fully unsaturated one to six membered straight or branched carbon chain wherein the carbons, other than the connecting carbon, may optionally be replaced with one heteroatom selected from oxygen, sulfur and nitrogen and said carbon is optionally mono-, di- or tri-substituted independently with halo, said carbon is optionally mono-substituted with hydroxy, said carbon is optionally mono- substituted with oxo, said sulfur is optionally mono- or di-substituted with oxo, said nitrogen is optionally mono- or di-substituted with oxo, and said carbon chain is optionally mono-substituted with V1; wherein V1 is is a partially saturated, fully saturated or fully unsaturated three to six membered ring optionally having one to two heteroatoms selected independently from oxygen, sulfur and nitrogen; wherein said V1 substituent is optionally mono-, di-, tri-, or tetra- substituted independently with halo, (C C6)alkyl, (C1-C6)alkoxy, amino, nitro, cyano, (C1-C6)alkyloxycarbonyl, mono-N- or di-N,N-(C -C6)alkyiamino wherein said (C1-C6)alkyl substituent is optionally mono-substituted with oxo, or said
(C C6)alkyl optionally having from one to nine fluorines; wherein either R3 must contain V or R4 must contain V1; and R5 , R6 , R7 and R8 are each independently hydrogen, a bond, nitro or halo wherein said bond is substituted with T or a partially saturated, fully saturated or fully unsaturated (C Cι2) straight or branched carbon chain wherein carbon may optionally be replaced with one or two heteroatoms selected independently from oxygen, sulfur and nitrogen wherein said carbon atoms are optionally mono-, di- or tri-substituted independently with halo, said carbon is optionally mono-substituted with hydroxy, said carbon is optionally mono-substituted with oxo, said sulfur is optionally mono- or di-substituted with oxo, said nitrogen is optionally mono- or di-substituted with oxo, and said carbon is optionally mono-substituted with T; wherein T is a partially saturated, fully saturated or fully unsaturated three to twelve membered ring optionally having one to four heteroatoms selected independently from oxygen, sulfur and nitrogen, or a bicyclic ring consisting of two fused partially saturated, fully saturated or fully unsaturated three to six membered rings, taken independently, optionally having one to four heteroatoms selected independently from nitrogen, sulfur and oxygen; wherein said T substituent is optionally mono-, di- or tri-substituted independently with halo, (Cι-C6)alkyl, (C2-C6)alkenyl, hydroxy, (C C6)alkoxy, (C C4)alkylthio, amino, nitro, cyano, oxo, carboxy, (C C6)alkyloxycarbonyl, mono-N- or di-N,N-(C C6)alkylamino wherein said (C C6)alkyl substituent is optionally mono-, di- or tri-substituted independently with hydroxy, (C C6)alkoxy, (C C4)alkylthio, amino, nitro, cyano, oxo, carboxy, (Cr C6)alkyloxycarbonyl, mono-N- or di-N,N-(C C6)alkylamino or said (C C6)alkyl optionally having from one to nine fluorines; provided that at least one of substituents R5, R6, R7 and Rδ is not hydrogen and is not linked to the quinoline moiety through oxy.
A preferred group of compounds, designated the A Group, contains those compounds having the Formula I as shown above wherein the C2 methyl is beta; the C4 nitrogen is beta:
R1 is W-X ;
W is carbonyl, thiocarbonyl or sulfonyl;
X is -O-Y-, S-Y-, N(H)-Y- or -N-(Y)2-; Y for each occurrence is independently Z or (C C4)alkyl, said (C C4)alkyl optionally having hydroxy or from one to nine fluorines or said (C C4)alkyl optionally mono-substituted with Z wherein Z is a partially saturated, fully saturated or fully unsaturated three to six membered ring optionally having one to two heteroatoms selected independently from oxygen, suifur and nitrogen; wherein said Z substituent is optionally mono-, di- or tri-substituted independently with halo, (C C4)alkyl, (C1-C )alkoxy, (C C4)alkylthio, nitro, cyano, oxo, or (C C6)alkyioxycarbonyl, said (CrC4)alkyl optionally substituted with from one to nine fluorines; R3 is Q-V wherein Q is (C C )alkyl and V is a five or six membered partially saturated, fully saturated or fully unsaturated ring optionally having one to three heteroatoms selected independently from oxygen, sulfur and nitrogen; wherein said V ring is optionally mono-, di-, tri- or tetra-substituted independently with halo, (C C6)alkyl, hydroxy, (C C6)alkoxy, nitro, cyano or oxo wherein said (C C6)alkyl optionally having from one to nine fluorines;
R4 is (C C4)alkyl;
R6 and R7 are each independently hydrogen, halo, T, (C C6)alkyl or (C
C6)alkoxy, said (CrC6)alkyl or (C C6)alkoxy optionally having from one to nine fluorines or said (C C6)alkoxy or (C C6)alkyl optionally moπo- substituted with T, wherein T is a partially saturated, fully saturated or fully unsaturated five to six membered ring optionally having one to two heteroatoms selected independently from oxygen, sulfur and nitrogen; wherein said T substituent is optionally mono-, di- or tri-substituted independently with halo, (CrC6)alkyl, hydroxy, (C C6)alkoxy, (C C4)alkylthio, amino, oxo, carboxy, (C C6)alkyloxycarbonyl, mono-N- or di-N,N-(C
C6)alkylamino wherein said (C C6)alkyl substituent optionally has from one to nine fluorines;
R5 and R8 are H; and pharmaceutically acceptable salts thereof.
A group of compounds which is preferred among the A Group of compounds, designated the B Group, contains those compounds wherein W is carbonyl;
X is O-Y wherein Y is (C C4)alkyl, said (C C4)alkyl optionally having one hydroxy or from one to nine fluorines;
Q is (C C4)alkyl and V is phenyl, pyridinyl, or pyπmidinyl; wherein said V ring is optionally mono-, di- or tri-substituted independently with halo, (C C6)alkyl, hydroxy, (C C6)alkoxy, nitro, cyano or oxo wherein said (C C6)alkyl substituent optionally has from one to nine fluorines;
R6 and R7 are each independently hydrogen, halo or (CrC3)alkyl, said (C
C3)alkyl optionally having from one to seven fluorines; and pharmaceutically acceptable salts thereof.
A group of compounds which is preferred among the B Group of compounds, designated the C Group, contains those compounds wherein
Q is methyiene and V is phenyl or pyridinyl; wherein said V ring is optionally mono-, di- or tri-substituted independently with halo, (C C2)alkyl, or nitro wherein said (C C2)alkyl optionally has from one to five fluorines; and pharmaceutically acceptable salts thereof.
Especially preferred compounds of Formula I are the compounds:
[2R,4S] 4-[(3,5-bis-trifluoromethyl-benzyl)-methoxycarbonyl-amino]-2-methyl- 7-trifluoromethyl-3,4-dihydro-2H-quinoline-1-carboxylic acid ethyl ester;
[2R,4S] 4-[(3,5-bis-trifluoromethyl-benzyl)-methoxycarbonyl-amino]-7-chloro-
2-methyl-3,4-dihydro-2H-quinoline-1-carboxylic acid ethyl ester; [2R,4S] 4-[(3,5-bis-trifiuoromethyl-benzyl)-methoxycarbonyl-amino]-6-chloro-
2-methyl-3,4-dihydro-2H-quinoline-1-carboxylic acid ethyl ester;
[2R,4S] 4-[(3,5-bis-trifluoromethyl-benzyl)-methoxycarbonyl-amino]-2,6,7- trimethyl-3,4-dihydro-2H-quinoiine-1-carboxylic acid ethyl ester; and pharmaceutically acceptable salts of said compounds.
Other especially preferred compounds of Formula I are the compounds:
[2R,4S] 4-[(3,5-bis-trifluoromethyl-benzyl)-methoxycarbonyl-amino]-6J- diethyl-2-methyl-3,4-dihydro-2H-quinoline-1-carboxyiic acid ethyl ester; [2R,4S] 4-[(3,5-bis-trifiuoromethyl-benzyl)-methoxycarbonyl-amino]-6-ethyl-2- methyl-3,4-dihydro-2H-quinoline-1-carboxylic acid ethyi ester;
[2R,4S] 4-[(3,5-bis-trifluoromethyl-benzyl)-methoxycarbonyl-amino]-2-methyl-
6-trifluoromethyl-3,4-dihydro-2H-quinoiine-1-carboxylic acid ethyl ester;
[2R,4S] 4-[(3,5-bis-trifluoromethyl-benzyl)-methoxycarbonyl-amino]-2-methyl- 6-trifluoromethyl-3,4-dihydro-2H-quinoline-1-carboxylic acid isopropyl ester; and pharmaceutically acceptable salts of said compounds.
Especially preferred compounds within the C Group of compounds are compounds wherein a. Y is ethyl; R3 is 3,5-bis-trifluoromethylphenylmethyl; R4 is methyl; R6 is hydrogen; and R7 is trifluoromethyl; b. Y is ethyl; R3 is 3,5-bis-trifluoromethylphenylmethyl; R4 is methyl; R6 is hydrogen; and R7 is chloro; c. Y is ethyl; R3 is 3,5-bis-trifluoromethyiphenylmethyl; R4 is methyl; R6 is chloro; and R7 is hydrogen; d. Y is ethyl;
R3 is 3,5-bis-trifluoromethylphenylmethyl; R4 is methyl; R6 is methyl; and R7 is methyl; e. Y is ethyl;
R3 is 3,5-bis-trifluoromethylphenylmethyl; R4 is methyl; R6 is ethyl; and R7 is ethyl; f. Y is ethyl;
R3 is 3,5-bis-trifluoromethylphenylmethyl; R4 is methyl; R6 is ethyl; and R7 is hydrogen; g. Y is ethyl;
R3 is 3,5-bis-trifluoromethylphenyimethyl;
R4 is methyl; R6 is trifluoromethyl; and
R7 is hydrogen; and h. Y is isopropyi;
R3 is 3,5-bis-trifluoromethylphenylmethyl;
R4 is methyl; R6 is trifluoromethyl; and
R7 is hydrogen, and pharmaceutically acceptable salts of said compounds.
A preferred group of compounds, designated the D Group, contains those compounds having Formula I as shown above wherein the C2 methyl is beta; the C4 nitrogen is beta;
R1 is W-X ; W is carbonyl, thiocarbonyl or sulfonyl; X is -O-Y-, S-Y-, N(H)-Y- or -N-(Y)2-;
Y for each occurrence is independently (CrC4)alkyl, said (CrC4)alkyl optionally having from one to nine fluorines or said (C C4)alkyl optionally mono-substituted with Z wherein Z is a partially saturated, fully saturated or fully unsaturated three to six membered ring optionally having one to two heteroatoms selected independently from oxygen, sulfur and nitrogen; wherein said Z substituent is optionally mono-, di- or tri-substituted independently with halo, (C1-C4)alkyl, (C C4)alkoxy, (C1-C4)alkyithio, nitro, cyano, oxo, or (C C6)alkyloxycarbonyl said (C C4)alkyl optionally having from one to nine fluorines;
R3 is Q-V wherein Q is (CrC )alkyl and V is a five or six membered partially saturated, fully saturated or fully unsaturated ring optionally having one to three heteroatoms selected independently from oxygen, sulfur and nitrogen; wherein said V ring is optionally mono-, di-, tri- or tetra-substituted independently with halo, (CrC6)alkyl, hydroxy, (C1-C6)alkoxy, nitro, cyano or oxo wherein said (C C6)alkyl substituent is optionally mono-, di- or tri- substituted independently with (C C6)alkoxy or (C C4)alkylthio or said (C C6)alkyi optionally having from one to nine fluorines; R4 is (C C4)alkyl;
R6 and R7 are each independently H, carbamoyl, oxycarbonyl, oxy or halo, or said carbamoyl, oxycarbonyl or oxy are substituted with (CrC4)alkyl and said (C C )alkyl is optionally mono-, di-, or tri-substituted with halo or hydroxy or said (C C4)alkyl is optionally mono-substituted with (C C4)alkoxy, carboxy, (C C4)alkylthio, (C1-C4)alkoxycarbonyi, amino or mono-N- or di-N,N-(C C4)alkylamino or said (C1-C4)alkyl optionally having from one to nine fluorines; or said (C C4)aikyl, carbamoyl, oxycarbonyl or oxy are optionally mono- substituted with T; wherein T is a partially saturated, fully saturated or fully unsaturated three to six membered ring optionally having one to two heteroatoms selected independently from oxygen, sulfur and nitrogen; wherein said T ring is optionally mono-, di-, tri- or tetra-substituted independently with halo, (C1-C6)alkyl, hydroxy, (C1-C6)alkoxy, nitro, cyano or oxo wherein said (C C6)alkyl substituent is optionally mono-, di- or tri- substituted independently with (C C6)alkoxy or (C C )alkylthio or said (Cr C6)alkyl optionally having from one to nine fluorines; ~
R5 and R8 are H; or a pharmaceutically acceptable salt thereof.
A group of compounds which is preferred among the D Group of compounds, designated the E Group, contains those compounds wherein W is carbonyl;
X is O-Y wherein Y is (C1-C4), said (C C4)alkyl optionally having from one to nine fluorines;
Q is (CrC4)alkylene and V is phenyl, pyridinyl, or pyrimidinyl; wherein said V ring is optionally mono-, di- or tri-substituted independently with halo, (C C6)alkyl, hydroxy, (C C6)alkoxy, nitro, cyano or oxo wherein said (C-,-C6)alkyl substituent optionally has from one to nine fluorines;
R6 is H, carbamoyl, oxycarbonyl, oxy or halo, or said carbamoyl, oxycarbonyl or oxy are substituted with (C C2)alkyl and said (C C2)alkyl is optionally mono-, di-, or tri-substituted with halo or hydroxy or said (C C4)alkyl optionally having from one to nine fluorines;
R7 is H, carbamoyl, oxycarbonyl, oxy or halo, or said carbamoyl, oxycarbonyl or oxy are optionally substituted with (C C4)alkyl and said (C C4)alkyi is optionally mono-, di-, or tri-substituted with halo or hydroxy or said (C C4)alkyl optionally having from one to nine fluorines; and pharmaceutically acceptable saits thereof.
Yet another aspect of this invention is directed to methods for treating atherosclerosis, peripheral vascular disease, dyslipidemia, hyperbetalipoproteinemia, hypoalphaiipoproteinemia, hypercholesterolemia, hypertrigiyceridemia, familial- hypercholesterolemia, cardiovascular disorders, angina, ischemia, cardiac ischemia, stroke, myocardial infarction, reperfusion injury, angioplastic restenosis, hypertension, vascular complications of diabetes, obesity or endotoxemia in a mammal (including a human being either male or female) by administering to a mammal in need of such treatment an atherosclerosis, peripheral vascular disease, dyslipidemia, hyperbetalipoproteinemia, hypoalphalipoproteinemia, hypercholesterolemia, hypertrigiyceridemia, familial-hypercholesterolemia, cardiovascular disorders, angina, ischemia, cardiac ischemia, stroke, myocardial infarction, reperfusion injury, angioplastic restenosis, hypertension, vascular complications of diabetes, obesity or endotoxemia treating amount of a Formula I compound, a prodrug thereof, or a pharmaceutically acceptable salt of said compound or of said prodrug.
Yet another aspect of this invention is directed to a method for treating atherosclerosis in a mammal (including a human being) by administering to a mammal in need of such treatment an atherosclerotic treating amount of a Formula I compound, a prodrug thereof, or a pharmaceutically acceptable salt of said compound or of said prodrug.
Yet another aspect of this invention is directed to a method for treating peripheral vascular disease in a mammal (including a human being) by administering to a mammal in need of such treatment a peripheral vascular disease treating amount of a Formula I compound, a prodrug thereof, or a pharmaceutically acceptable salt of said compound or of said prodrug.
Yet another aspect of this invention is directed to a method for treating dyslipidemia in a mammai (including a human being) by administering to a mammal in need of such treatment a dyslipidemia treating amount of a Formula I compound, a prodrug thereof, or a pharmaceutically acceptable salt of said compound or of said prodrug.
Yet another aspect of this invention is directed to a method for treating hyperbetalipoproteinemia in a mammal (including a human being) by administering to a mammal in need of such treatment a hyperbetalipoproteinemia treating amount of a
Formula I compound, a prodrug thereof, or a pharmaceutically acceptable salt of said compound or of said prodrug.
Yet another aspect of this invention is directed to a method for treating hypoalphalipoproteinemia in a mammal (including a human being) by administering to a mammal in need of such treatment a hypoalphalipoproteinemia treating amount of a Formula i compound, a prodrug thereof, or a pharmaceutically acceptable salt of said compound or of said prodrug. Yet another aspect of this invention is directed to a method for treating hypercholesterolemia in a mammal (including a human being) by administering to a mammal in need of such treatment a hypercholesterolemia treating amount of a Formula I compound, a prodrug thereof, or a pharmaceutically acceptable salt of said compound or of said prodrug.
Yet another aspect of this invention is directed to a method for treating hypertrigiyceridemia in a mammal (including a human being) by administering to a mammal in need of such treatment a hypertrigiyceridemia treating amount of a Formula I compound, a prodrug thereof, or a pharmaceutically acceptable salt of said compound or of said prodrug.
Yet another aspect of this invention is directed to a method for treating familial-hypercholesterolemia in a mammal (including a human being) by administering to a mammal in need of such treatment a familial- hypercholesterolemia treating amount of a Formula I compound, a prodrug thereof, or a pharmaceutically acceptable salt of said compound or of said prodrug.
Yet another aspect of this invention is directed to a method for treating cardiovascular disorders in a mammal (including a human being) by administering to a mammal in need of such treatment a cardiovascular disorder treating amount of a Formula I compound, a prodrug thereof, or a pharmaceutically acceptable salt of said compound or of said prodrug.
Yet another aspect of this invention is directed to a method for treating angina in a mammal (including a human being) by administering to a mammal in need of such treatment an angina treating amount of a Formula I compound, a prodrug thereof, or a pharmaceutically acceptable salt of said compound or of said prodrug. Yet another aspect of this invention is directed to a method for treating ischemia in a mammal (including a human being) by administering to a mammal in need of such treatment an ischemic disease treating amount of a Formula I compound, a prodrug thereof, or a pharmaceutically acceptable salt of said compound or of said prodrug. Yet another aspect of this invention is directed to a method for treating cardiac ischemia in a mammal (including a human being) by administering to a mammal in need of such treatment a cardiac ischemic treating amount of a Formula I compound, a prodrug thereof, or a pharmaceutically acceptable salt of said compound or of said prodrug.
Yet another aspect of this invention is directed to a method for treating stroke in a mammal (including a human being) by administering to a mammal in need of such treatment a stroke treating amount of a Formula I compound, a prodrug thereof, or a pharmaceutically acceptable salt of said compound or of said prodrug.
Yet another aspect of this invention is directed to a method for treating a myocardial infarction in a mammal (including a human being) by administering to a mammal in need of such treatment a myocardial infarction treating amount of a Formula I compound, a prodrug thereof, or a pharmaceutically acceptable salt of said compound or of said prodrug.
Yet another aspect of this invention is directed to a method for treating reperfusion injury in a mammal (including a human being) by administering to a mammal in need of such treatment a reperfusion injury treating amount of a Formula I compound, a prodrug thereof, or a pharmaceutically acceptable salt of said compound or of said prodrug.
Yet another aspect of this invention is directed to a method for treating angioplastic restenosis in a mammal (including a human being) by administering to a mammal in need of such treatment an angioplastic restenosis treating amount of a Formula I compound, a prodrug thereof, or a pharmaceutically acceptable sait of said compound or of said prodrug.
Yet another aspect of this invention is directed to a method for treating hypertension in a mammal (including a human being) by administering to a mammal in need of such treatment a hypertension treating amount of a Formula I compound, a prodrug thereof or a pharmaceutically acceptable salt of said compound or of said prodrug.
Yet another aspect of this invention is directed to a method for treating the vascular complications of diabetes in a mammal (including a human being) by administering to a mammal in need of such treatment a vascular complications of diabetes treating amount of a Formula I compound, a prodrug thereof or a pharmaceutically acceptable salt of said compound or of said prodrug.
Yet another aspect of this invention is directed to a method for treating obesity in a mammal (including a human being) by administering to a mammal in need of such treatment an obesity treating amount of a Formula I compound, a prodrug thereof, or a pharmaceutically acceptable salt of said compound or of said prodrug.
Yet another aspect of this invention is directed to a method for treating endotoxemia in a mammal (including a human being) by administering to a mammal in need of such treatment an endotoxemia treating amount of a Formula I compoundT a prodrug thereof, or a pharmaceutically acceptable salt of said compound or of said prodrug.
A preferred dosage is about 0.001 to 100 mg/kg/day of a Formula I compound, a prodrug thereof, or a pharmaceutically acceptable salt of said compound or of said prodrug. An especially preferred dosage is about 0.01 to 10 mg/kg/day of a Formula I compound, a prodrug thereof, or a pharmaceutically acceptable salt of said compound or of said prodrug.
This invention is also directed to pharmaceutical compositions which comprise a therapeutically effective amount of a compound of Formula I, a prodrug thereof, or a pharmaceutically acceptable salt of said compound or of said prodrug and a pharmaceutically acceptable carrier.
This invention is also directed to pharmaceutical compositions for the treatment of atherosclerosis, peripheral vascular disease, dyslipidemia, hyperbetalipoproteinemia, hypoalphalipoproteinemia, hypercholesterolemia, hypertrigiyceridemia, familial-hypercholesterolemia, cardiovascular disorders, angina, ischemia, cardiac ischemia, stroke, myocardial infarction, reperfusion injury, angioplastic restenosis, hypertension, vascular complications of diabetes, obesity or endotoxemia in a mammal (including a human being) which comprise a therapeutically effective amount of a compound of Formula I, a prodrug thereof, or a pharmaceutically acceptable salt of said compound or of said prodrug and a pharmaceutically acceptable carrier.
This invention is also directed to pharmaceutical compositions for the treatment of atherosclerosis in a mammal (including a human being) which comprise an atherosclerosis treating amount of a compound of Formula I, a prodrug thereof, or a pharmaceutically acceptable salt of said compound or of said prodrug and a pharmaceutically acceptable carrier.
This invention is also directed to pharmaceutical compositions for the treatment of peripheral vascular disease in a mammal (including a human being) which comprise a peripheral vascular disease treating amount of a compound of Formula I, a prodrug thereof, or a pharmaceutically acceptable salt of said compound or of said prodrug and a pharmaceutically acceptable carrier.
This invention is also directed to pharmaceutical compositions for the treatment of dyslipidemia in a mammal (including a human being) which comprise a ~ dyslipidemia treating amount of a compound of Formula I, a prodrug thereof, or a pharmaceutically acceptable salt of said compound or of said prodrug and a pharmaceutically acceptable carrier.
This invention is also directed to pharmaceutical compositions for the treatment of hyperbetalipoproteinemia in a mammal (including a human being) which comprise a hyperbetalipoproteinemia treating amount of a compound of Formula I, a prodrug thereof, or a pharmaceutically acceptable salt of said compound or of said prodrug and a pharmaceutically acceptable carrier.
This invention is also directed to pharmaceutical compositions for the treatment of hypoalphalipoproteinemia in a mammal (including a human being) which comprise a hypoalphalipoproteinemia treating amount of a compound of Formula I, a prodrug thereof, or a pharmaceutically acceptable salt of said compound or of said prodrug and a pharmaceutically acceptable carrier.
This invention is also directed to pharmaceutical compositions for the treatment of hypercholesterolemia in a mammal (including a human being) which comprise a hypercholesterolemia treating amount of a compound of Formula I, a prodrug thereof, or a pharmaceutically acceptable salt of said compound or of said prodrug and a pharmaceutically acceptable carrier.
This invention is also directed to pharmaceutical compositions for the treatment of hypertrigiyceridemia in a mammal (including a human being) which comprise a hypertrigiyceridemia treating amount of a compound of Formula I, a prodrug thereof, or a pharmaceutically acceptable salt of said compound or of said prodrug and a pharmaceutically acceptable carrier.
This invention is also directed to pharmaceutical compositions for the treatment of familial-hypercholesterolemia in a mammal (including a human being) which comprise a familial-hypercholesterolemia treating amount of a compound of Formula I, a prodrug thereof, or a pharmaceutically acceptable salt of said compound or of said prodrug and a pharmaceutically acceptable carrier. This invention is also directed to pharmaceutical compositions for the treatment of angina in a mammal (including a human being) which comprise an angina treating amount of a compound of Formula I, a prodrug thereof, or a pharmaceutically acceptable salt of said compound or of said prodrug and a pharmaceutically acceptable carrier. ~
This invention is also directed to pharmaceutical compositions for the treatment of ischemia in a mammal (including a human being) which comprise an ischemic treating amount of a compound of Formula I, a prodrug thereof, or a phaπnaceutically acceptable salt of said compound or of said prodrug and a pharmaceutically acceptable carrier.
This invention is also directed to pharmaceutical compositions for the treatment of cardiac ischemia in a mammal (including a human being) which comprise a cardiac ischemic treating amount of a compound of Formula I, a prodrug thereof, or a pharmaceutically acceptable salt of said compound or of said prodrug and a pharmaceutically acceptable earner.
This invention is also directed to pharmaceutical compositions for the treatment of stroke in a mammal (including a human being) which comprise a stroke treating amount of a compound of Formula I, a prodrug thereof, or a pharmaceutically acceptable salt of said compound or of said prodrug and a pharmaceutically acceptable earner.
This invention is also directed to pharmaceutical compositions for the treatment of a myocardial infarction in a mammal (including a human being) which comprise a myocardial infarction treating amount of a compound of Formula I, a prodrug thereof, or a pharmaceutically acceptable salt of said compound or of said prodrug and a pharmaceutically acceptable carrier.
This invention is also directed to pharmaceutical compositions for the treatment of reperfusion injury in a mammal (including a human being) which comprise a reperfusion injury treating amount of a compound of Formula I, a prodrug thereof, or a pharmaceutically acceptable salt of said compound or of said prodrug and a pharmaceutically acceptable earner.
This invention is also directed to pharmaceutical compositions for the treatment of angioplastic restenosis in a mammal (including a human being) which comprise an angioplastic restenosis treating amount of a compound of Formula I, a prodrug thereof, or a pharmaceutically acceptable salt of said compound or of said prodrug and a pharmaceutically acceptable carrier.
This invention is also directed to pharmaceutical compositions for the treatment of hypertension in a mammal (including a human being) which comprise a hypertension treating amount of a compound of Formula I, a prodrug thereof or a ~ pharmaceutically acceptable salt of said compound or of said prodrug and a pharmaceutically acceptable carrier.
This invention is also directed to pharmaceutical compositions for the treatment of the vascular complications of diabetes in a mammal (including a human being) which comprise a vascular complications of diabetes treating amount of a compound of Formula I, a prodrug thereof, or a pharmaceutically acceptable salt of said compound or of said prodrug and a pharmaceutically acceptable carrier.
This invention is also directed to pharmaceutical compositions for the treatment of obesity in a mammal (including a human being) which comprise an obesity treating amount of a compound of Formula I, a prodrug thereof, or a pharmaceutically acceptable salt of said compound or of said prodrug and a pharmaceutically acceptable carrier.
This invention is also directed to pharmaceutical compositions for the treatment of endotoxemia in a mammal (including a human being) which comprise an endotoxemia treating amount of a compound of Formula I, a prodrug thereof, or a pharmaceutically acceptable salt of said compound or of said prodrug and a pharmaceutically acceptable carrier.
This invention is also directed to a pharmaceutical combination composition comprising: a therapeutically effective amount of a composition comprising a first compound, said first compound being a Foπnula I compound, a prodrug thereof, or a pharmaceutically acceptable salt of said compound or of said prodrug; a second compound, said second compound being an HMG-CoA reductase inhibitor, an microsomal triglyceride transfer protein (MTP)/Apo B secretion inhibitor, a PPAR activator, a bile acid reuptake inhibitor, a cholesterol absorption inhibitor, a cholesterol synthesis inhibitor, a fibrate, niacin, an ion-exchange resin, an antioxidant, an ACAT inhibitor or a bile acid sequestrant; and/or optionally a pharmaceutical carrier. Preferred among the second compounds are an HMG-CoA reductase inhibitor and a MTP/Apo B secretion inhibitor.
A particularly preferred HMG-CoA reductase inhibitor is lovastatin, simvastatin, pravastatin, fluvastatin, atorvastatin or rivastatin. Another aspect of this invention is a method for treating atherosclerosis in a mammai comprising administering to a mammal suffering from atherosclerosis; a first compound, said first compound being a Formula I compound a prodrug thereof, or a pharmaceutically acceptable salt of said compound or of said prodrug; and a second compound, said second compound being an HMG-CoA reductase inhibitor, an MTP/Apo B secretion inhibitor, a cholesterol absorption inhibitor, a cholesterol synthesis inhibitor, a fibrate, niacin, an ion-exchange resin, an antioxidant, an ACAT inhibitor or a bile acid sequestrant wherein the amounts of the first and second compounds result in a therapeutic effect. A preferred aspect of the above method is wherein the second compound is an HMG-CoA reductase inhibitor or an MTP/Apo B secretion inhibitor.
A particularly preferred aspect of the above method is wherein the HMG-CoA reductase inhibitor is lovastatin, simvastatin, pravastatin, fluvastatin, atorvastatin or rivastatin. Yet another aspect of this invention is a kit comprising: a. a first compound, said first compound being a Formula I compound, a prodrug thereof, or a pharmaceutically acceptable salt of said compound or of said prodrug and a pharmaceutically acceptable carrier in a first unit dosage form; b. of a second compound, said second compound being an HMG CoA reductase inhibitor, an MTP/Apo B secretion inhibitor, a cholesterol absorption inhibitor, a cholesterol synthesis inhibitor, a fibrate, niacin, an ion-exchange resin, an antioxidant, an ACAT inhibitor or a bile acid sequestrant and a pharmaceutically acceptable carrier in a second unit dosage form; and c. means for containing said first and second dosage forms wherein the amounts of the first and second compounds result in a therapeutic effect.
A preferred second compound is an HMG-CoA reductase inhibitor or an MTP/Apo B secretion inhibitor. A particularly preferred HMG-CoA reductase inhibitor is lovastatin, simvastatin, pravastatin, fluvastatin, atorvastatin or rivastatin.
As used herein the term mammals is meant to refer to all mammals which contain CETP in their plasma, for example, rabbits and primates such as monkeys and humans. Certain other mammals e.g., dogs, cats, cattle, goats, sheep and ~ horses do not contain CETP in their plasma and so are not included herein.
The term "treating", "treat" or "treatment" as used herein includes preventative (e.g., prophylactic) and palliative treatment.
By "pharmaceutically acceptable" is meant the carrier, diluent, excipients, and/or salt must be compatible with the other ingredients of the formulation, and not deleterious to the recipient thereof.
The expression "prodrug" refers to compounds that are drug precursors which following administration, release the drug in vivo via some chemical or physiological process (e.g., a prodrug on being brought to the physiological pH or through enzyme action is converted to the desired drug form). Exemplary prodrugs upon cleavage release the corresponding free acid, and such hydrolyzable ester-forming residues of the Formula I compounds include but are not limited to those having a carboxyl moiety wherein the free hydrogen is replaced by (C1-C4)alkyl, (C2- C7)alkanoyloxymethyl, 1-(alkanoyloxy)ethyl having from 4 to 9 carbon atoms, 1- methyl-1-(alkanoyioxy)-ethyl having from 5 to 10 carbon atoms, alkoxycarbonyloxymethyl having from 3 to 6 carbon atoms, 1- (alkoxycarbonyloxy)ethyl having from 4 to 7 carbon atoms, 1-methyl-1- (alkoxycarbonyloxy)ethyl having from 5 to 8 carbon atoms, N- (alkoxycarbonyl)aminomethyl having from 3 to 9 carbon atoms, 1-(N- (alkoxycarbonyl)amino)ethyl having from 4 to 10 carbon atoms, 3-phthalidyl, 4- crotonolactonyl, gamma-butyrolacton-4-yl, di-N,N-(C C2)alkylamino(C2-C3)alkyl (such as b-dimethylaminoethyl), carbamoyl-(C C2)alkyl, N,N-di(Cr C2)alkylcarbamoyi-(C1-C2)alkyl and piperidino-, pyrrolidino- or moφholino(C2-C3)alkyl. The following paragraphs describe exemplary ring(s) for the generic ring descriptions contained herein.
Exemplary five to six membered aromatic rings optionally having one or two heteroatoms selected independently from oxygen, nitrogen and sulfur include phenyl, furyl, thienyl, pyrrolyl, oxazolyl, thiazolyl, imidazolyl, pyrazolyi, isoxazolyl, isothiazolyl, pyridinyl, pyridiazinyl, pyrimidinyl and pyrazinyl.
Exemplary partially saturated, fully saturated or fully unsaturated five to eight membered rings optionally having one to four heteroatoms selected independently from oxygen, sulfur and nitrogen include cyciopentyl, cyclohexyl, cycloheptyl, ~ cyclooctyl and phenyl. Further exemplary five membered rings include 2H-pyrrolyl, 3H-pyrrolyl, 2-pyrrolinyl, 3-pyrrolinyl, pyrrolidinyl, 1 ,3-dioxolanyl, oxazolyl, thiazolyl, imidazolyl, 2H-imidazolyl, 2-imidazoiinyl, imidazolidinyi, pyrazolyi, 2-pyrazoiinyl, pyrazolidinyl, isoxazolyl, isothiazolyl, 1 ,2-dithioiyi, 1 ,3-dithiolyl, 3H-1 ,2-oxathiolyl, 1,2,3-oxadiazolyl, 1 ,2,4-oxadiazolyl, 1,2,5-oxadiazolyl, 1 ,3,4-oxadiazolyl, 1 ,2,3- triazolyl, 1 ,2,4-triazolyl, 1 ,3,4-thiadiazolyl, 1,2,3,4-oxatriazolyl, 1 ,2,3,5-oxatriazolyl, 3H-1 ,2,3-dioxazolyl, 1 ,2,4-dioxazoiyl, 1 ,3,2-dioxazoiyl, 1 ,3,4-dioxazolyl, 5H-1 ,2,5- oxathiazolyl and 1 ,3-oxathiolyl.
Further exemplary six membered rings include 2H-pyranyi, 4H-pyranyl, pyridinyl, piperidinyl, 1,2-dioxinyl, 1 ,3-dioxinyl, 1,4-dioxanyl, moφholinyl, 1 ,4-dithianyl, thiomoφholinyi, pyridazinyl, pyrimidinyi, pyrazinyl, piperazinyi, 1,3,5-triazinyl, 1,2,4- triazinyl, 1 ,2,3-triazinyl, 1,3,5-trithianyl, 4H-1 ,2-oxazinyl, 2H-1 ,3-oxazinyl, 6H-1.3- oxazinyi, 6H-1 ,2-oxazinyl, 1,4-oxazinyl, 2H-1 ,2-oxazinyl, 4H-1 ,4-oxazinyl, 1 ,2,5- oxathiazinyl, 1 ,4-oxazinyl, o-isoxazinyl, p-isoxazinyl, 1,2,5-oxathiazinyl, 1 ,2,6- oxathiazinyl, 1 ,4,2-oxadiazinyl and 1 ,3,5,2-oxadiazinyl.
Further exemplary seven membered rings include azepinyl, oxepinyl, and thiepinyl.
Further exemplary eight membered rings include cyclooctyl, cyclooctenyl and cyclooctadienyl.
Exemplary bicyclic rings consisting of two fused partially saturated, fully saturated or fully unsaturated five or six membered rings, taken independently, optionally having one to four heteroatoms selected independently from nitrogen, sulfur and oxygen include indolizinyl, indolyl, isoindolyl, 3H-indolyl, 1 H-isoindolyl, indolinyi, cyclopenta(b)pyridinyl, pyrano(3,4-b)pyrroiyl, benzofuryl, isobenzofuryl, benzo(b)thienyl, benzo(c)thienyl, 1 H-indazoiyl, indoxazinyl, benzoxazolyl, benzimidazolyl, benzthiazolyl, purinyl, 4H-quinoiizinyl, quinolinyl, isoquinolinyl, cinnolinyl, phthaiazinyl, quinazolinyl, quinoxalinyl, 1 ,8-naphthyridinyi, pteridinyl, indenyl, isoindenyl, naphthyl, tetralinyl, decaiinyl, 2H-1-benzopyranyl, pyrido(3,4-b)- pyridinyl, pyrido(3,2-b)-pyridinyl, pyrido(4,3-b)-pyridinyl, 2H-1 ,3-benzoxazinyl, 2H-1.4- benzoxazinyl, 1 H-2,3-benzoxazinyl, 4H-3,1-benzoxazinyl, 2H-1 ,2-benzoxazinyi and 4H-1 ,4-benzoxazinyl.
By alkylene is meant saturated hydrocarbon (straight chain or branched ) wherein a hydrogen atom is removed from each of the terminal carbons. Exemplary of such groups (assuming the designated length encompasses the particular ~" example) are methylene, ethylene, propylene, butylene, pentylene, hexylene, heptylene).
By halo is meant chloro, bromo, iodo, or fluoro.
By alkyl is meant straight chain saturated hydrocarbon or branched chain saturated hydrocarbon. Exemplary of such alkyl groups (assuming the designated length encompasses the particular example) are methyl, ethyl, propyl, isopropyl, butyl, sec-butyl, tertiary butyl, pentyl, isopentyl, neopentyl, tertiary pentyl, 1- methylbutyl, 2-methylbutyl, 3-methylbutyl, hexyl, isohexyl, heptyi and octyl.
By alkoxy is meant straight chain saturated alkyl or branched chain saturated alkyl bonded through an oxy. Exemplary of such alkoxy groups (assuming the designated length encompasses the particular example) are methoxy, ethoxy, propoxy, isopropoxy, butoxy, isobutoxy, tertiary butoxy, pentoxy, isopentoxy, neopentoxy, tertiary pentoxy, hexoxy, isohexoxy, heptoxy and octoxy .
As used herein the term mono-N- or di-N,N-(C Cx)alkyl... refers to the (C Cx)alkyl moiety taken independently when it is di-N,N-(C Cx)alkyi...(x refers to integers).
References (e.g., claim 1) to "said carbon" in the phrase "said carbon is optionally mono-, di- or tri-substituted independently with halo, said carbon is optionally mono-substituted with hydroxy, said carbon is optionally mono-substituted with oxo" refers to each of the carbons in the carbon chain including the connecting carbon.
References to "Nitrogen... di-substituted with oxo" herein (e.g., claim 1 ) refer to a terminal nitrogen which constitutes a nitro functionality.
It is to be understood that if a carbocyciic or heterocyclic moiety may be bonded or otherwise attached to a designated substrate through differing ring atoms without denoting a specific point of attachment, then all possible points are intended, whether through a carbon atom or, for example, a trivalent nitrogen atom. For example, the term "pyridyl" means 2-, 3-, or 4-pyridyl, the term "thienyl" means 2-, or 3-thienyl, and so forth.
The expression "pharmaceutically-acceptabie salt" refers to nontoxic anionic salts containing anions such as (but not limited to) chloride, bromide, iodide, sulfate, bisulfate, phosphate, acetate, maleate, fumarate, oxalate, lactate, tartrate, citrate, gluconate, methanesulfonate and 4-toluene-sulfonate. The expression also refers to nontoxic cationic salts such as (but not limited to) sodium, potassium, calcium, magnesium, ammonium or protonated benzathine (N,N'-dibenzylethylenediamine), choline, ethanolamine, diethanolamine, ethylenediamine, meglamine (N-methyl- glucamine), benethamine (N-benzylphenethylamine), piperazine or tromethamine (2- amino-2-hydroxymethyl-1 ,3-propanediol).
As used herein, the expressions "reaction-inert solvent" and "inert solvent" refers to a solvent or a mixture thereof which does not interact with starting materials, reagents, intermediates or products in a manner which adversely affects the yield of the desired product.
The term "cis" refers to the orientation of two substituents with reference to each other and the plane of the ring (either both "up" or both "down"). Analogously, the term "trans" refers to the orientation of two substituents with reference to each other and the plane of the ring (the substituents being on opposite sides of the ring). Alpha and Beta refer to the orientation of a substituent with reference to the plane of the ring (i.e., page). Beta is above the plane of the ring (i.e., page) and Alpha is below the plane of the ring (i.e., page).
The chemist of ordinary skill will recognize that certain compounds of this invention will contain one or more atoms which may be in a particular stereochemical or geometric configuration, giving rise to stereoisomers and configurational isomers. All such isomers and mixtures thereof are included in this invention. Hydrates and solvates of the compounds of this invention are also included.
It will be recognized that the compounds of this invention can exist in radioiabelled form, i.e., said compounds may contain one or more atoms containing an atomic mass or mass number different from the atomic mass or mass number usually found in nature. Radioisotopes of hydrogen, carbon, phosphorous, fluorine and chlorine include 3H, 14C, 32P, 35S, 18F and ^Cl, respectively. Compounds of this invention, a prodrug thereof, or a pharmaceutically acceptable salt of said compound or of said prodrug which contain those radioisotopes and/or other radioisotopes of other atoms are within the scope of this invention. Tritiated, i.e., 3H, and carbon-14, i.e., 14C, radioisotopes are particularly preferred for their ease of preparation and detectability. Radiolabelled compounds of Formula I of this invention and prodrugs thereof can generally be prepared by methods well known to those skilled in the art. Conveniently, such radiolabelled compounds can be prepared by carrying out the procedures disclosed in the Schemes and/or in the Examples and Preparations below by substituting a readily available radiolabelled reagent for a non-radiolabelled reagent. DTT means dithiothreitol. DMSO means dimethyl sulfoxide. EDTA means ethylenediamine tetraacetic acid.
Other features and advantages of this invention will be apparent from this specification and the appendant claims which describe the invention. DETAILED DESCRIPTION OF THE INVENTION In general the compounds of this invention can be made by processes which include processes analogous to those known in the chemical arts, particularly in light of the description contained herein. Certain processes for the manufacture of the compounds of this invention are provided as further features of the invention and are illustrated by the following reaction schemes. Other processes may be described in the experimental section.
SCHEMEI
Figure imgf000027_0001
VII VI SCHEME II
Figure imgf000028_0001
VI SCHEME El
Figure imgf000029_0001
Figure imgf000030_0001
XX SCHEME V
Figure imgf000031_0001
V
SCHEME VI
Figure imgf000032_0001
LXII LXIII
Figure imgf000032_0002
LXIV As an initial note, in the preparation of the Formula I compounds it is noted that some of the preparation methods useful for the preparation of the compounds described herein may require protection of remote functionality (e.g., primary amine, secondary amine, carboxyl in Formula I precursors). The need for such protection will vary depending on the nature of the remote functionality and the conditions of the preparation methods. The need for such protection is readily determined by one skilled in the art. The use of such protection/deprotection methods is also within the skill in the art. For a general description of protecting groups and their use, see T.W. Greene, Protective Groups in Organic Synthesis. John Wiley & Sons, New York, 1991.
For example, in Reaction Schemes I and II certain Formula I compounds contain primary amines or carboxylic acid functionalities which may interfere with reactions at other sites of the molecule if left unprotected. Accordingly, such functionalities may be protected by an appropriate protecting group which may be removed in a subsequent step. Suitable protecting groups for amine and carboxylic acid protection include those protecting groups commonly used in peptide synthesis (such as N-t-butoxycarbonyl, benzyloxycarbonyl, and 9-fluorenylmethylenoxycarbonyl for amines and iower alkyl or benzyl esters for carboxylic acids) which are generally not chemically reactive under the reaction conditions described and can typically be removed without chemically altering other functionality in the Formula I compound.
According to Reaction Scheme I, the Formula III compounds wherein R5 , RD , R7, and Rδ are as described above and P2 is an appropriate protecting group may be prepared from the appropriate Formula II aromatic amine wherein R5 , R6 , R7 and R8 are as described above. The Formula III tetrahydroquinoiine is prepared by treating the appropriate
Formula II aromatic amine with acetaldehyde in an inert solvent such as a hydrocarbon (e.g., hexanes, pentanes or cyciohexane), an aromatic hydrocarbon (e.g., benzene, toluene or xylene), a halocarbon (e.g., dichloromethane, chloroform, carbon tetrachloride or dichloroethane), an ether (e.g., diethyi ether, diisopropyl ether, tetrahydrofuran, tetrahydropyran, dioxane, dimethoxyethane, methyl tert-butyl ether, etc.), a nitrile (e.g., acetonitrile or propionitrile), a nitroalkane (e.g., nitromethane or nitrobenzene), preferably dichloromethane with a dehydrating agent (e.g., sodium sulfate or magnesium suifate) at a temperature of about 0°C to about 100°C (preferably ambient temperature) for 1-24 hours (preferably 1 hour). The resulting solution is treated with a suitably substituted (e.g., benzyloxycarbonyl, t- butoxycarbonyi, methoxycarbonyl, formyl-, acetyl-, diallyl- or dibenzyl-), preferably carboxybenzyloxy-, N-vinyl species and with a Lewis acid (e.g., boron trifluoride, ~ boron trifluoride etherate, zinc chloride, titanium tetrachloride, iron trichloride, aluminum trichloride, alkyl aluminum dichloride, dialkyl aluminum chloride or ytterbium (III) trifiate; preferably boron trifluoride etherate) or a protic acid such as a hydrohalogenic acid (e.g., fluoro, chloro, bromo or iodo), an aikyl sulfonic acid (e.g., p- toluene, methane or trifioromethane) or carboxylic acid (e.g., formic, acetic, trifluoroacetic or beπzoic) at a temperature of from about
-78°C to about 50°C (preferably ambient temperature) for 0.1 to 24 hours (preferably 1 hour).
Alternatively, the Formula II amine and acetaldehyde may be condensed by treating a solution of the amine and an alkyl amine base (preferably triethylamine) in a polar aprotic solvent (preferably dichloromethane) with titanium tetrachloride in a polar aprotic solvent (preferably in dichloromethane) at a temperature between about -78°C to about 40°C (preferably 0°C) followed by treatment with acetaldehyde at a temperature between about -78°C to about 40°C (preferably 0°C). The reaction is allowed to proceed for about 0.1 to about 10 hours (preferably 1 hour) at a temperature between about 0°C to about 40°C (preferably room temperature) yielding the imine which is reacted with the N-vinyi species as above.
The compounds of Formula IV wherein R1, R5, R6, R7 and R8are as described above and P1 and P2 are protecting groups may be prepared from the corresponding Formula III amine by various amine reaction routes known to those skilled in the art. Thus, the Formula IV compounds wherein R1, R5 , R6 , R7, and R8are as described above and P1 and P2 are appropriately differentiated protecting groups for the amine moieties are prepared from the corresponding Formula III tetrahydroquinoline employing standard methods for derivatizing amines into the functional groups described for R1 above, see Richard Larock, Comprehensive Organic Transformations. VCH Publishers Inc., New York, 1989 and Jerry March, Advanced Organic Chemistry. John Wiley & Sons, New York, 1985. For example, a Formula ill compound is treated with the appropriate carbonyl chloride, sulfonyl chloride, or sulfϊnyl chloride, isocyanate or thioisocyanate in a polar aprotic solvent (preferably dichloromethane) in the presence of a base (preferably pyridine) at a temperature of from about -78°C to about 100°C (preferably starting at 0°C and letting warm to room temperature) for a period of 1 to 24 hours (preferably 12 hours).
Formula IV carbamate and urea compounds (wherein R1 is W=C(0), X=0-Y, S-Y, N(H)-Y, or NY2) may be prepared from the Formula III amines via the corresponding carbamoyl chlorides by treating the Formula III amine with a phosgene solution in a hydrocarbon solvent (preferably toluene) at a temperature between about 0°C and about 200°C (preferably at reflux) for between 0.1 and 24 hours (preferably 2 hours).
The corresponding ureas may be prepared by treating a solution of the carbamoyl chlorides (prepared as described above) with the appropriate amine in a polar solvent (preferably dichloromethane) at a temperature between about -78°C and about 100°C (preferably ambient temperature) for between 1 and 24 hours (preferably 12 hours).
The corresponding carbamate may be prepared by treating a solution of the carbamoyl chlorides (prepared as described above) with the appropriate alcohol and a suitable base (preferably sodium hydride) in a polar solvent (preferably dioxane) at a temperature between about -78°C and about 100°C (preferably ambient temperature) for between 1 and 24 hours (preferably 12 hours).
Alternatively, the corresponding carbamate may be prepared by treating a solution of the carbamoyl chlorides at a temperature between about 0°C and about 200°C in the appropriate alcohol for between 1 and 240 hours (preferably 24 hours). The Formula IV compound wherein R1 is Y may be prepared using methods known to those skilled in the art to introduce Y substituents such as an alkyl or alkyl linked substituent. Methods include, for example, formation of the amide from the amine of Formula III and an activated carboxylic acid followed by reduction of the amide with borane in an etheral solvent such as tetrahydrofuran. Alternatively, the alkyl or alkyl linked substituent may be appended by reduction after condensing the amine of Formula III with the required carbonyl containing reactant. Also, the amine of Formula III may be reacted with the appropriate alkyl or aryl halide according to methods known to those skilled in the art.
Thus, the Formula III amine and an acid (e.g., halogenic, sulfuric, sulfonic or carboxylic, preferably acetic) are treated with the appropriate carbonyl containing reactant in a polar solvent (preferably ethanol) at a temperature of about 0°C to about 100°C (preferably room temperature) for about 0.1 to 24 hours (preferably 1 hour) followed by treatment with a hydride source (e.g., sodium borohydride, sodium cyanoborohydride, preferably sodium triacetoxyborohydride) at a temperature of about 0°C to about 100°C (preferably ambient temperature) for 0.1 to 100 hours (preferably 5 hours).
The Formula V amine wherein R1, R5 , R6 , R7, and Rδare as described above and P ' is a protecting group may be prepared from the corresponding Formula IV compound by deprotection (P2) using methods known to those skilled in the art, including hydrogenolysis, treatment with an acid (e.g., trifiuoroacetic acid, hydrobromic), a base (sodium hydroxide), or reaction with a nucleophile (e.g. sodium methylthiolate, sodium cyanide, etc.) and for the trialkylsilylethoxy carbonyl group a fluoride is used (e.g., tetrabutyl ammonium fluoride). For removal of a benzyloxycarbonyl group, hydrogenolysis is performed by treating the Formula IV compound with a hydride source (e.g., 1 to 10 atmospheres of hydrogen gas, cyclohexene or ammonium formate) in the presence of a suitable catalyst (e.g., 5- 20% palladium on carbon, palladium hydroxide; preferably 10% palladium on carbon) in a polar solvent (e.g., methanol, ethanol or ethyl acetate; preferably ethanol) at a temperature between about -78°C and about 100°C, preferably ambient temperature, for 0.1 to 24 hours, preferably 1 hour. The compounds of Formula VI wherein R1, R3 , R5 , R6 , R7 and R8 are as described above and P1 is a protecting group as described above may be prepared from the corresponding Formula V amine by various amine reaction routes known to those skilled in the art.
The Formula VI secondary amine wherein R3 is as described above may be prepared using methods known to those skilled in the art to introduce R3 substituents such as an alkyl or alkyl linked substituent. Methods include, for example, formation of an amide from the Formula V amine and an activated carboxylic acid followed by reduction of the amide with borane in an etheral solvent such as tetrahydrofuran. Alternatively, an alkyl or alkyl linked substituent may be appended by reduction of the appropriate imine, the imine being formed by condensing the Formula V amine with the required carbonyl containing reactant. Also, the Formula V amine may be reacted with the appropriate alkyl halide according to methods known to those skilled in the art.
Thus, the Formula V amine and an acid (e.g., halogenic, sulfuric, sulfonic or carboxylic, preferably hydrochloric) are treated with the appropriate carbonyl containing reactant in a polar solvent (preferably dichloromethane) at a temperature of about 0°C to about 100°C (preferably room temperature) for about 0.1 to 24 hours (preferably 1 hour) followed by treatment with a hydride source (e.g., sodium borohydride or sodium cyanoborohydride; preferably sodium triacetoxyborohydride) at a temperature of about 0°C to about 100°C (preferably ambient temperature) for 0.1 to 100 hours (preferably 5 hours). The Formula VII compound wherein R1, R3 , R5 , R6 , R7 and R8are as described above and P1 and P2 are protecting groups may be prepared from the corresponding Foπnula IV compound by methods known to those skilled in the art; for example, the methods described for the introduction of the R3 substituent above in the transformation of the Formula V compound to the Formula VI compound. Following this, the corresponding Formula VI compound may be prepared from the Formula VII compound by appropriate deprotection such as the methods described above for the transformation of the Formula IV compound to the Formula V compound.
When R3 is H and R4 is as described above R4 may be represented by R3 in the Formulas VI and VII in Scheme I, thus providing a synthetic scheme for such compounds.
According to Scheme II, the Formula XI dihydroquinolone compounds wherein R5 , R6 , R7, R8 and Y are as described above, and P1 is a protecting group, may be prepared from the corresponding Formula X quinoiines by treatment with a metallo- methyl species and a chloroformate followed by hydrolysis.
Thus, a mixture of the Formula X quinoline and an excess (preferably 1.5 equivalents) of a methyl magnesium species (Grignard reagent) in a polar aprotic soivent (e.g., diethyi ether or dichloromethane; preferably tetrahydrofuran) is treated with an excess (preferably 1.5 equivalents) of a Y- or P1-chloroformate at a temperature between about -100°C and about 70°C (preferably -78°C) followed by warming to a temperature between about 0°C and about 70°C (preferably ambient temperature) for between 0.1 and 24 hours (preferably 1 hour). The resulting mixture is combined with an excess (preferably 2 equivalents) of an aqueous acid (preferably 1 molar hydrochloric acid) and mixed vigorously for between 0.1 and 24 hours (preferably 1 hour, or until hydrolysis of the intermediate enol ether is determined to be complete). Of course, the Formula XI compounds are the Formula XVI compounds wherein R1 is -C(O)OY or P is -C(0)OP1 without further transformation.
The Formula XV compounds wherein R5 , R6 , R7 and R8 are as described above may be prepared from the corresponding Formula XI dihydroquinolone (wherein Formula XI compound contains P1) by appropriate deprotection (including spontaneous decarboxylation) as described for the transformation of the Formula IV compound to the Formula V compound.
The Formula XVI compounds wherein R1, R5 , R6 , R7 and R8 are as described above and P1 is a protecting group may be prepared from the corresponding Formula XV dihydroquinolone as described for the transformation of the Formula III compound to the Formula IV compound. In certain cases where the reagent has also reacted on the 4-position carbonyl oxygen, the substituent may be conveniently removed by treatment with acid (e.g., aqueous HCI) or base (e.g., aqueous sodium hydroxide).
Again, for those Formula XVI compounds wherein R1 or P1 is the same as for the Formula XI compound such transformation as described above is not needed. The Formula VI amine compounds wherein R1, R3, R5 , R6 , R7 and R8 are as described above and P1 is a protecting group may be prepared from the corresponding Formula XVI dihydroquinolone by a reductive amination sequence. The Formula XVI dihydroquinolone, an excess (preferably 1.1 equivalents) of an R3- amine and an excess (preferably 7 equivalents) of an amine base (preferably triethylamine) in a polar solvent (preferably dichloromethane) are treated with 0.5 to 1.0 equivalents (preferably 0.55 equivalents) of titanium tetrachloride as a solution in a suitable polar solvent (preferably dichloromethane) at a temperature between about 0°C and about 40°C (preferably ambient temperature) for between 1 to 24 hours (preferably 12 hours). The resulting Formula XII imine is reduced by treatment with a reducing agent (preferably sodium borohydride) in an appropriate polar solvent (preferably ethanol) at a temperature between about 0°C and about 80°C (preferably room temperature) for between 1 and 24 hours (preferably 12 hours) resulting in a mixture of diastereomeric Formula VI amines, generally favoring the trans isomer. Alternatively, the reduction may be performed by treating the Formula XII imine directly with an excess (preferably 5 equivalents) of zinc borohydride as a solution in ether (preferably 0.2 molar) at a temperature between about 0°C and about 40°C (preferably ambient temperature) for between 1 and 24 hours (preferably 12 hours) resulting in a mixture of diastereomeric Formula VI amines, generally favoring the cis isomer.
Alternatively, the Formula VI amine wherein R1, R3, R5 , R6 , R7 and R8 are as described above and P is a protecting group may be prepared from the corresponding Formula XVI dihydroquinolones by formation of an oxime, reduction and substitution of the amine. Thus, the Formula XVI dihydroquinolone, excess (preferably 3 equivalents) hydroxylamine hydrochloride and an excess (preferably 2.5 equivalents) of base (preferably sodium acetate) are reacted at a temperature between about 0°C and about 100°C (preferably at reflux) for between 1 and 24 hours (preferably 2 hours) in a polar solvent (preferably ethanol). The resulting Formula XIII oxime is treated with excess (preferably 6 equivalents) aqueous base (preferably 2N potassium hydroxide) in a polar solvent (preferably ethanol) and an excess (preferably 4 equivalents) of a nickel-aluminum alloy (preferably 1:1 by weight) at a temperature between about 0°C and about 100°C (preferably ambient temperature) for between 0.25 and 24 hours (preferably 1 hour). The resulting Formula V amine is obtained as a diastereomeric mixture (generally the cis isomer). The Formula VI secondary amine wherein R1, R3, R5 , R6 , R7 and R8 are as described above and P1 is a protecting group may be prepared from the appropriate Formula V amine as described in Scheme I for the transformation of the Formula V compound to the Formula VI compound. According to Scheme III the Formula I compounds as described above may be prepared from the appropriate Formula VI compounds by conversion to the desired carbamate. Thus, the Formula VI amine is treated with the appropriate activated carbonate (e.g., chloroformate, dicarbonate or carbonyl diimidazole followed by the appropriate alcohol) in a polar solvent (preferably dichloromethane) in the presence of an excess of amine base (preferably pyridine) at a temperature between about -20°C and about 40°C (preferably ambient temperature) for between 1 and 24 hours (preferably 12 hours) to yield the Formula I compound.
Alternatively, according to Scheme III, where appropriate, if the functionality at R1 is incompatible with the reaction to form the Formula I compound, then the P1 protected Formula VI compound may be transformed to the Formula I compound through protection/deprotection sequences and introduction of the desired substituents. Thus, the Formula VI amine is treated with the appropriate reagent (e.g., protecting group precursor, activated carbonate (e.g., chloroformate, dicartDonate or carbonyl imidazole)) in a polar solvent (preferably dichloromethane) in the presence of an excess of amine base (preferably pyridine) at a temperature between about -20°C and about 40 °C (preferably ambient temperature) for between 1 and 24 hours (preferably 12 hours) to yield the Formula XX compound .
Also, the Formula XX compounds, wherein P2 is present may be obtained as shown in Scheme I for the Formula VII compounds (having P ).
The Formula XXI amines wherein R3, R5 , R6 , R7, R8 and R4 are as described above and P2 is a protecting group may be prepared from the Formula XX compound by selective deprotection.
When P1 is, for example, t-butoxycarbonyl, the Formula XXI compound is conveniently prepared by treatment with an acid (preferably trifluoroacetic acid) at a temperature between about 0°C and about 100°C (preferably room temperature) for 0.1 to 24 hours (preferably 1 hour).
The compounds of Formula I or compounds of Formula XXII (wherein R1 is as described above) may be prepared from the corresponding Formula XXI amine (wherein R4 or P2 is present respectively) by various amine reaction routes known to those skilled in the art, for example, those described in Scheme I for the transformation of the Formula III compound to the Formula IV compound. The Formula XXIII amines may be prepared from the Formula XXII compounds by suitable deprotection. When P2 is, for example, benzyloxycarbonyl, the Formula XXIII compound is prepared by treatment with an excess of a hydride source (e.g., cyclohexene, hydrogen gas or preferably ammonium formate) in the presence of 0.01 to 2 equivalents (preferably 0.1 equivalent) of a suitable catalyst (preferably 10% palladium on carbon) in a polar solvent (preferably ethanol) at a temperature between about 0°C and about 100°C (preferably room temperature) for 0.1 to 24 hours (preferably 1 hour).
The Formula I compound wherein R4 is as described above may be prepared using the methods described for the conversion of the Formula VI compound to the Formula I compound in Scheme III above.
According to Scheme IV the Formula V compounds wherein R1 , R5 , R7 and R8 are as described above, and R6 is an ether linked moiety can be obtained from the Formula XXX quinolones having a OP3 moiety, wherein P3 is a protecting group, at the R6 position employing the following methods. In addition, in an analogous manner such processes may be used to prepare the corresponding compounds wherein R5, R7, or R8 are an ether linked moiety starting from the corresponding Foπnula XXX compound having an OP3 moiety at either the R5, R', or R8 positions.
Thus, the Formula XXX quinolone is combined with hydroxylamine hydrochloride and a mineral base (preferably sodium acetate) in a polar solvent
(preferably ethanoi) at a temperature between about 0°C and about 100°C (preferably at reflux) for between 1 and 24 hours (preferably 2 hours) to yield the Formula XXXI oxime.
The Formula XXXI oxime is treated with an excess (preferably six equivalents) of an aqueous base (preferably 2N potassium hydroxide) and an excess (preferably four equivalents) of a nickel-aluminum alloy (preferably 1 :1 by weight) in a polar solvent (preferably ethanoi) at a temperature between about 0°C and about 100°C (preferably ambient temperature) for between 0.25 and 24 hours (preferably 2 hours) to prepare the corresponding Formula XXXII amine. If necessary, the P3 protecting group may be removed using standard methods if the oxime transformation does not result in such cleavage. Altematively, the Formula XXX compound may be deprotected (removal of the P3) by methods known to those skilled in the art prior to formation of the Formula XXXI oxime (wherein P3 is H) which can then be reduced to form the Formula XXXII amine. The Formula V compound wherein R6 is an oxy-linked moiety may be prepared by treating the Formula XXXII alcohol under, for example, Mitsunobu conditions. Thus, the Formula XXXII phenol is treated with a phosphine (preferably triphenylphosphine) and an azodicarboxylate (preferably bis-(N-methylpiperazinyl)- azodicarboxamide) and the required alcohol in a polar solvent (preferably benzene). Of course, via Schemes I and II the resulting Formula V compound may be transformed into the Formula VI precursors for the Formula I compounds of this invention.
Alternatively, the Formula XX compound wherein R6 is an ether linked moiety and wherein R1, R3 and R4 are as described above and P1 and P2 are protecting groups may be prepared from the Formula XXXII alcohols as described below. In addition, in an analogous manner such processes may be used to prepare the corresponding compounds wherein R5, R7, or R8 are an ether iinked moiety starting from the corresponding Formula XXXII compound and thus ultimately the Formula XXX compound (i.e., the Formula XXX compound having a P30- at either the R5, R7, or R8 positions).
The Formula XXXIII secondary amine wherein R3 is as described above may be prepared from the corresponding Formula XXXII compound according to methods in Scheme I described above for the conversion of the Formula V compound to the Formula VI compound. The Formula XXXIV compounds wherein R4 is as described above may be prepared from Formula XXXIII amines by methods analogous to that described in Scheme III for the transformation of the Formula VI compound to the Formula I compound.
The Formula XXXV phenol may be selectively deprotected for example when R402CO- is present by treating the Formula XXXIV carbonate with potassium carbonate in a polar solvent (preferably methanol) at a temperature between about 0°C and about 100°C (preferably ambient temperature) for between 1 and 24 hours (preferably 12 hours).
The corresponding XX ethers may be prepared from the Formula XXXV phenol using for example, the Mitsunobu conditions described above for the conversion of the Formula XXXII compounds to the Formula V compounds. Of course, one skilled in the art will appreciate that the phenol may be derivatized to a variety of functional groups using standard methods, for example, as described in March or Larock, or by conversion to the corresponding triflate for use in a variety of reactions involving transition metal catalysis.
Although the following description of Scheme V is directed to modifications of the R6 position (the R6 position described in Formula I above) those skilled in the art will appreciate that analogous methods may be applied to the R5 , R7 and R8 positions. According to Scheme V the Formula LI alcohol wherein R1, R3, R4, R5,
R7 and R8 are as described above, P1 and P2 are protecting groups, and X1 is a linking group wherein a carbon (e.g., methylene) is directly linked to the carbonyl moiety may be prepared from the corresponding ester (wherein R12 is a convenient alkyl moiety) by reduction. Thus, the Formula L ester is treated with sodium borohydride/methanoi or a borane-dimethylsuifide complex in a polar solvent (preferably tetrahydrofuran) at a temperature between about 0°C and about 100°C (preferably at reflux) for between 1 and 24 hours (preferably 3 hours). The Formula Lll compounds wherein R1, R3, R4, R5 , R7 and R8 are as described above, P1 and P2 are protecting groups and wherein the R6 position includes an alkyl haiide functionality may be prepared from the corresponding Formula LI alcohol by treatment with a trialkylphosphine (preferably triphenylphosphine) and a dihalogen (e.g., bromine) in a polar solvent (preferably dichloromethane) at a temperature between about -78°C and about 100°C (preferably 0°C) for between 0.1 and 10 hours (preferably 0.5 hours) followed by warming to room temperature for between 0.1 and 10 hours (preferably 3 hours). The Formula Llll compounds wherein R1, R3, R4, R5 , R7 and R8are as described above, P1 and P2 are protecting groups, the R6 position includes ether or thioether moieties (i.e., Y1 is S or O) and R13 is a carbon linked substituent may be prepared by treating the Formula LI I alkyl halide in a polar solvent (preferably N,N-dimethylformamide) with the requisite alkoxide or thioalkoxide at a temperature between about 0°C and about 100°C (preferably at room temperature) for between 1 and 24 hours (preferably 6 hours).
Alternatively, the Formula Llll ethers and thioethers may be prepared by treating the corresponding Formula LIV alcohols and thiols (i.e., Y1 is S or O), wherein X1 is a substituent linked directly through carbon to the methyiene moiety, with a base (preferably sodium hydride) and the requisite alkylating agent in a polar solvent (preferably N,N-dimethylformamide) at a temperature between about 0°C and about 100CC (preferably at room temperature) for between 1 and 50 hours (preferably 18 hours).
The Formula LV compounds wherein R1, R3, R4, R5 , R7 and R8are as described above, P1 and P2 are protecting groups, the R6 position includes alkyl halides (e.g., fluorides) and X1 is a substituent that is carbon linked directly to the methyiene moiety may be prepared by treating the corresponding Formula LI alcohol with a halogenating agent. For example, the alcohol is treated with a fluorinating agent (preferably diethylaminosuifur trifluoride) in a polar solvent (preferably 1 ,2-dichioroethane) at a temperature between about 0°C and about 100°C (preferably 80 °C) for between 0.1 and 10 hours (preferably 0.75 hours). The Formula LVII amide compounds wherein R1, R3, R4, R5 , R7 and
R8are as described above, P1 and P2 are protecting groups and wherein R6 includes an amide functionality (such that X is a substituent that is carbon linked directly to the carbonyl moiety and R10 and R11 are substituents selected to yield the desired R6 substituent defined above) may be prepared from the corresponding Formula LVI carboxylic acid which may in turn be prepared from the corresponding Foπnula L carboxylic ester. Thus, the Foπnula L ester is treated with an aqueous hydroxide
(preferably lithium, sodium or potassium) in a polar solvent (preferably tetrahydrofuran and/or methanol) at a temperature between about 0°C and about 100°C (preferably room temperature) for between 0.1 and 100 hours (preferably 1 hour).
The Formula LVII amide may be prepared from the corresponding Formula LVI acid by standard methods. Prefered is conversion of the carboxylic acid to the acid chloride by dissolving the acid in thionyl chloride and holding the solution at a temperature between about 0°C and about 80°C (preferably at reflux) for between 0.1 and 24 hours (preferably 1 hour) before evaporation of the excess thionyl chloride. This step is followed by treating the resulting acid chloride residue in a polar solvent (preferably dichloromethane) with the appropriate amine, selected to yield the amide functionality, and optionally an amine base (preferably triethylamine) at a temperature between about -78°C and about 100°C (preferably room temperature) for between 0.1 and 100 hours (preferably 1 hour).
Although the following Scheme VI is directed to modifications of the R8 position those skilled in the art will appreciate that analogous methods may be applied to the R5 , R6 and R7 positions. According to Scheme VI the Formula LXl compound wherein R1, R3,
R4, R5 , R6, and R7 are as described above and P1 and P2 are protecting groups may be prepared from the corresponding Formula LX compound by nitration. The Formula LX compound is treated with nitrosyitriflate in a halogenated solvent, such as dichloromethane at a temperature of about -78°C to about 0°C for about 0.5 hour to about 3 hours followed by warming to ambient temperature.
The Formula LXl I amine wherein R1, R3, R4, R5 , R6, and R7 are as described above and P1 and P2 are protecting groups may be prepared from the corresponding Formula LXl compound by reduction. The Formula LXl compound is hydrogenated by treatment with hydrogen gas in the presence of a noble metal catalyst (e.g., palladium on carbon) in a polar solvent such as ethanol at a temperature of about 0°C to about 100°C for about 1 to 24 hours at elevated pressure (e.g., 1 to 3 atmospheres).
The Formula LXl 11 compound wherein R1, R3, R4, R5 , R6, and R7 are as described above, P1 and P2 are protecting groups and R8 is an amine linked functionality may be prepared from the corresponding Formula LXII. The Formula LXII amine is derivatized following procedures analogous to those described in Scheme I for the conversion of the Formula III compound to the Formula IV compound.
The Formula LXIV compound wherein R1, R3, R4, R5 , R6, and R7 are as described above and P1 and P2 are protecting groups may be prepared from the corresponding Formula LXII compound. The Formula LXII amine is treated with t-butyl nitrate and anhydrous cupric halide in a polar solvent at a temperature of about 30°C to about 100°C for about 1 hour to about 24 hours.
Of course, one skilled in the art will understand that the halide may be derivatized to a variety of functional groups using standard methods for example as described in Larock or March.
Prodrugs of the compounds of Formula I may be prepared according to methods known to those skilled in the art. Exemplary processes are described below. Prodrugs of this invention where a carboxyl group in a carboxylic acid of Formula I is replaced by an ester may be prepared by combining the carboxylic acid with the appropriate alkyl halide in the presence of a base such as potassium carbonate in an inert solvent such as dimethylformamide at a temperature of about 0 to 100°C for about 1 to about 24 hours. Alternatively the acid is combined with appropriate alcohol as solvent in the presence of a catalytic amount of acid such as concentrated sulfuric acid at a temperature of about 20 to 100°C, preferably at a reflux, for about 1 hour to about 24 hours. Another method is the reaction of the acid with a stoichiometric amount of the alcohol in the presence of a catalytic amount of acid in an inert solvent such as toluene or tetrahydrofuran, with concomitant removal of the water being produced by physical (e.g., Dean-Stark trap) or chemical (e.g., molecular sieves) means. Prodrugs of this invention where an alcohol function has been derivatized as an ether may be prepared by combining the alcohol with the appropriate alkyl bromide or iodide in the presence of a base such as potassium carbonate in an inert solvent such as dimethyiformamide at a temperature of about 0 to 100°C for about 1 to about 24 hours.
Alkanoylaminomethyl ethers may be obtained by reaction of the alcohol with a bis-(alkanoylamino)methane in the presence of a catalytic amount of acid in an inert solvent such as tetrahydrofuran, according to a method described in US 4,997,984. Alternatively, these compounds may be prepared by the methods described by Hoffman et al. in J. Org. Chem. 1994, 59, 3530.
Glycosides are prepared by reaction of the alcohol and a carbohydrate in an inert solvent such as toluene in the presence of acid. Typically the water formed in the reaction is removed as it is being formed as described above. An alternate procedure is the reaction of the alcohol with a suitably protected glycosyl halide in the presence of base followed by deprotection.
N-(l-hydroxyalkyl) amides, N-(1-hydroxy-1-(alkoxycarbonyl)methyl) amides may be prepared by the reaction of the parent amide with the appropriate aldehyde under neutral or basic conditions (e.g., sodium ethoxide in ethanol) at temperatures between 25 and 70°C. N-alkoxymethyl or N-1- (alkoxy)alkyl derivatives can be obtained by reaction of the N-unsubstituted compound with the necessary alkyl halide in the presence of a base in an inert solvent.
The compounds of this invention may also be used in conjunction with other pharmaceutical agents (e.g., LDL-cholesterol lowering agents, triglyceride lowering agents) for the treatment of the disease/conditions described herein. For example, they may be used in combination with cholesterol synthesis inhibitors, cholesterol absoφtion inhibitors, MTP/Apo B secretion inhibitors, and other cholesterol lowering agents such as fibrates, niacin, ion-exchange resins, antioxidants, ACAT inhibitors and bile acid sequestrants. In combination therapy treatment, both the compounds of this invention and the other drug therapies are administered to mammals (e.g., humans, male or female) by conventional methods.
Any HMG-CoA reductase inhibitor may be used as the second compound in the combination aspect of this invention. The term HMG-CoA reductase inhibitor refers to compounds which inhibit the byconversion of hydroxymethylglutaryl- coenzyme A to mevalonic acid catalyzed by the enzyme HMG-CoA reductase. Such inhibition is readily determined by those skilled in the art according to standard assays (e.g., Meth. Enzymol. 1981 ; 71:455-509 and references cited therein). A variety of these compounds are described and referenced below however other HMG-CoA reductase inhibitors will be known to those skilled in the art. U.S. Pat. No. 4,231 ,938 (the disclosure of which is hereby incoφorated by reference) discloses certain compounds isolated after cultivation of a microorganism belonging to the genus Aspergillus, such as lovastatin. Also, U.S. Pat. No. 4,444,784 (the disclosure of which is hereby incoφorated by reference) discloses synthetic derivatives of the aforementioned compounds, such as simvastatin. Also, U.S. Pat. No. 4,739,073 (the disclosure of which is incoφorated by reference) discloses certain substituted indoles, such as fluvastatin. Also, U.S. Pat. No. 4,346,227 (the disclosure of which is incoφorated by reference) discloses ML-236B derivatives, such as pravastatin. Also, EP-491226A (the disclosure of which is incoφorated by reference) discloses certain pyridyldihydroxyheptenoic acids, such as rivastatin. In addition, U.S. Pat. No. 5,273,995 (the disclosure of which is incoφorated by reference) discloses certain 6- [2-(substituted-pyrrol-1-yl)alkyl]pyran-2-ones such as atorvastatin.
Any MTP/Apo B secretion (microsomal triglyceride transfer protein and or apolipoprotein B) inhibitor may be used as the second compound in the combination aspect of this invention. The term MTP/Apo B secretion inhibitor refers to compounds which inhibit the secretion of triglycerides, cholesteryl ester, and phosphoiipids. Such inhibition is readily determined by those skilled in the art according to standard assays (e.g., Wetterau, J. R. 1992; Science 258:999). A variety of these compounds are described and referenced beiow however other MTP/Apo B secretion inhibitors will be known to those skilled in the art.
WO 96/40640 and WO 98/23593 are two exemplary publications. For example, the following MTP/Apo B secretion inhibitors are particularly useful: 4'-trifluoromethyl-biphenyl-2-carboxyiic acid [2-(1 H-[1 ,2,4,]triazol-3-yimethyl)-1 ,2,3,4- tetrahydro-isoquinolin-6-y -amide;
4'-trifluoromethyl-biphenyl-2-carboxylic acid [2-(2-acetylamino-ethyl)-1 ,2,3,4- tetrahydro-isoquinolin-6-y -amide; (2-{6-[(4'-trifluoromethyl-biphenyl-2-carbonyl)-amino]-3,4-dihydro-1H-isoquinolin-2-yl}- ethyl)-carbamic acid methyl ester;
4'-trifluoromethyl-biphenyl-2-carboxyiic acid [2-(1 H-imidazoi-2-ylmethyl)-1 ,2,3,4- tetrahydro-isoquinolin-6-yl]-amide; 4'-trifluoromethyl-biphenyl-2-carboxylic acid [2-(2,2-diphenyl-ethyl)-1 ,2,3,4-tetrahydro- isoquinolin-6-yl]-amide; and
4'-trifluoromethyl-biphenyl-2-carboxylic acid [2-(2-ethoxy-ethyl)-1 ,2,3,4-tetrahydro- isoquinolin-6-yl]-amide.
Any HMG-CoA synthase inhibitor may be used as the second compound in the combination aspect of this invention. The term HMG-CoA synthase inhibitor refers to compounds which inhibit the biosynthesis of hydroxymethylglutaryi-coenzyme A from acetyl-coenzyme A and acetoacetyl-coenzyme A, catalyzed by the enzyme HMG-CoA synthase. Such inhibition is readily determined by those skilled in the art according to standard assays (Meth Enzymol. 1975; 35:155-160: Meth. Enzymol. 1985; 110:19-26 and references cited therein). A variety of these compounds are described and referenced below, however other HMG-CoA synthase inhibitors will be known to those skilled in the art. U.S. Pat. No. 5,120,729 (the disclosure of which is hereby incoφorated by reference) discloses certain beta-lactam derivatives. U.S. Pat. No. 5,064,856 (the disclosure of which is hereby incorporated by reference) discloses certain spiro-lactone derivatives prepared by culturing a microorganism (MF5253). U.S. Pat. No. 4,847,271 (the disclosure of which is hereby incoφorated by reference) discloses certain oxetane compounds such as 11-(3-hydroxymethyl-4-oxo-2-oxetayl)- 3,5,7-trimethyl-2,4-undeca-dienoic acid derivatives.
Any compound that decreases HMG-CoA reductase gene expression may be used as the second compound in the combination aspect of this invention. These agents may be HMG-CoA reductase transcription inhibitors that block the transcription of DNA or translation inhibitors that prevent translation of mRNA coding for HMG-CoA reductase into protein. Such compounds may either affect transcription or translation directly, or may be biotransformed to compounds that have the aforementioned activities by one or more enzymes in the cholesterol biosynthetic cascade or may lead to the accumulation of an isoprene metabolite that has the aforementioned activities. Such regulation is readily determined by those skilled in the art according to standard assays (Meth. Enzymol. 1985; 110:9-19). Several compounds are described and referenced below, however other inhibitors of HMG- CoA reductase gene expression will be known to those skilled in the art. U.S. Pat. No. 5,041 ,432 (the disclosure of which is incoφorated by reference) discloses certain 15- substituted ianosterol derivatives. Other oxygenated sterols that suppress synthesis of HMG-CoA reductase are discussed by E.I. Mercer (Prog.Lip. Res. 1993;32:357- 416).
Any squalene synthetase inhibitor may be used as the second compound of this invention. The term squalene synthetase inhibitor refers to compounds which inhibit the condensation of 2 molecules of famesylpyrophosphate to form squalene, catalyzed by the enzyme squalene synthetase. Such inhibition is readily determined by those skilled in the art according to standard assays (Meth. Enzymol. 1969; 15: 393-454 and Meth. Enzymol. 1985; 110:359-373 and references contained therein). A variety of these compounds are described in and referenced below however other squalene synthetase inhibitors will be known to those skilled in the art. U.S. Pat. No. 5,026,554 (the disclosure of which is incoφorated by reference) discloses fermentation products of the microorganism MF5465 (ATCC 74011) including zaragozic acid. A summary of other patented squalene synthetase inhibitors has been compiled (Curr. Op. Ther. Patents (1993) 861-4).
Any squalene epoxidase inhibitor may be used as the second compound in the combination aspect of this invention. The term squalene epoxidase inhibitor refers to compounds which inhibit the bioconversion of squalene and molecular oxygen into squalene-2,3-epoxide, catalyzed by the enzyme squalene epoxidase. Such inhibition is readily determined by those skilled in the art according to standard assays (Biochim. Biophys. Acta 1984; 794:466-471 ). A variety of these compounds are described and referenced below, however other squalene epoxidase inhibitors will be known to those skilled in the art. U.S. Pat. Nos. 5,011 ,859 and 5,064,864 (the disclosures of which are incoφorated by reference) disclose certain fluoro analogs of squalene. EP publication 395,768 A (the disclosure of which is incoφorated by reference) discloses certain substituted allylamine derivatives. PCT publication WO 9312069 A (the disclosure of which is hereby incoφorated by reference) discloses certain amino alcohol derivatives. U.S. Pat. No. 5,051 ,534 (the disclosure of which is hereby incoφorated by reference) discloses certain cyclopropyloxy-squalene derivatives. Any squalene cyclase inhibitor may be used as the second component in the combination aspect of this invention. The term squalene cyciase inhibitor refers to compounds which inhibit the bioconversion of squalene-2,3-epoxide to lanosterol, catalyzed by the enzyme squalene cyclase. Such inhibition is readily determined by those skilled in the art according to standard assays (FEBS Lett. 1989;244:347-350.). In addition, the compounds described and referenced below are squalene cyciase inhibitors, however other squalene cyclase inhibitors will also be known to those skilled in the art. PCT publication WO9410150 (the disclosure of which is hereby incoφorated by reference) discloses certain 1 ,2,3,5,6,7,8,8α-octahydro-5,5,8α(beta)- trimethyl-6-isoquinolineamine derivatives, such as N-trifluoroacetyl-1 ,2,3,5,6,7,8,8α- octahydro-2-allyl-5,5,8α(beta)-trimethyl-6(beta)-isoquinolineamine. French patent publication 2697250 (the disclosure of which is hereby incorporated by reference) discloses certain beta, beta-dimethyl-4-piperidine ethanol derivatives such as 1- (1,5,9-trimethyldecyl)-beta,beta-dimethyl-4-piperidineethanol. Any combined squalene epoxidase/squalene cyciase inhibitor may be used as the second component in the combination aspect of this invention. The term combined squalene epoxidase/squalene cyclase inhibitor refers to compounds that inhibit the bioconversion of squalene to lanosterol via a squalene-2,3-epoxide intermediate. In some assays it is not possible to distinguish between squalene epoxidase inhibitors and squalene cyclase inhibitors, however, these assays are recognized by those skilled in the art. Thus, inhibition by combined squalene epoxidase/squalene cyclase inhibitors is readily determined by those skilled in art according to the aforementioned standard assays for squalene cyclase or squalene epoxidase inhibitors. A variety of these compounds are described and referenced below, however other squalene epoxidase/squalene cyclase inhibitors will be known to those skilled in the art. U.S. Pat. Nos. 5,084,461 and 5,278,171 (the disclosures of which are incoφorated by reference) disclose certain azadecalin derivatives. EP publication 468,434 (the disclosure of which is incoφorated by reference) discloses certain piperidyl ether and thio-ether derivatives such as 2-(1-piperidyl)pentyl isopentyl sulfoxide and 2-(1-piperidyl)ethyl ethyl sulflde. PCT publication WO 9401404 (the disclosure of which is hereby incoφorated by reference) discloses certain acyl-piperidines such as 1-(1-oxopentyl-5-phenylthio)-4-(2-hydroxy-1 -methyl)- ethy piperidine. U.S. Pat. No. 5,102,915 (the disclosure of which is hereby incoφorated by reference) discloses certain cyclopropyloxy-squalene derivatives.
The starting materials and reagents for the above described Formula I compounds, are also readily available or can be easily synthesized by those skilled in the art using conventional methods of organic synthesis. For example, many of the compounds used herein, are related to, or are derived from compounds in which there is a large scientific interest and commercial need, and accordingly many such compounds are commercially available or are reported in the literature or are easiiy prepared from other commonly available substances by methods which are reported in the literature.
Some of the Formula I compounds of this invention, or intermediates in their synthesis, have asymmetric carbon atoms and therefore are enantiomers or diastereomers. Diasteromeric mixtures can be separated into their individual diastereomers on the basis of their physical chemical differences by methods known per se^, for example, by chromatography and/or fractional crystallization. Enantiomers can be separated by, for example, chiral HPLC methods or converting the enantiomeric mixture into a diasteromeric mixture by reaction with an appropriate optically active compound (e.g., alcohol), separating the diastereomers and converting (e.g., hydrolyzing) the individual diastereomers to the corresponding pure enantiomers. Also, an enantiomeric mixture of the Formula I compounds or an intermediate in their synthesis which contain an acidic or basic moiety may be separated into their corresponding pure enantiomers by forming a diastereomeric salt with an optically pure chiral base or acid (e.g., 1 -phenyl-ethyl amine or tahtaric acid) and separating the diasteromers by fractional crystallization followed by neutralization to break the salt, thus providing the corresponding pure enantiomers. All such isomers, including diastereomers, enantiomers and mixtures thereof are considered as part of this invention. Also, some of the compounds of this invention are atropisomers (e.g., substituted biaryls) and are considered as part of this invention. More specifically, the Formula I compounds of this invention may be obtained in enantiomerically enriched form by resolving the racemate of the final compound or an intermediate in its synthesis (preferably the final compound) employing chromatography (preferably high pressure liquid chromatography [HPLC]) on an asymmetric resin (preferably Chiralcel™ AD or OD [obtained from Chiral Technologies, Exton, Pennsylvania]) with a mobile phase consisting of a hydrocarbon (preferably heptane or hexane) containing between 0 and 50% isopropanol (preferably between 2 and 20 %) and between 0 and 5% of an alkyl amine (preferably 0.1 % of diethylamine). Concentration of the product containing fractions affords the desired materials. ~~
Some of the Formula I compounds of this invention are acidic and they form a salt with a pharmaceutically acceptable cation. Some of the Formula I compounds of this invention are basic and they form a salt with a pharmaceutically acceptable anion. All such salts are within the scope of this invention and they can be prepared by conventional methods such as combining the acidic and basic entities, usually in a stoichiometric ratio, in either an aqueous, non-aqueous or partially aqueous medium, as appropriate. The salts are recovered either by filtration, by precipitation with a non- solvent followed by filtration, by evaporation of the solvent, or, in the case of aqueous solutions, by lyophilization, as appropriate. The compounds can be obtained in crystalline form by dissolution in an appropriate solvent(s) such as ethanol, hexanes or water/ethanol mixtures.
In addition, when the Formula I compounds of this invention form hydrates or solvates they are also within the scope of the invention.
The Formula I compounds of this invention, their prodrugs and the salts of such compounds and prodrugs are all adapted to therapeutic use as agents that inhibit cholesterol ester transfer protein activity in mammals, particularly humans. Thus, the compounds of this invention elevate plasma HDL cholesterol, its associated components, and the functions performed by them in mammals, particularly humans. By virtue of their activity, these agents also reduce plasma levels of triglycerides, LDL cholesterol, VLDL cholesterol and their associated components in mammals, particularly humans.
Hence, these compounds are useful for the treatment and correction of the various dyslipidemias observed to be associated with the development and incidence of atherosclerosis and cardiovascular disease, including hypoalphalipoproteinemia, hyperbetalipoproteinemia, hypertrigiyceridemia, and familial-hypercholesterolemia. Further, introduction of a functional CETP gene into an animal lacking CETP (mouse) results in reduced HDL levels (Agellon, L.B., et al: J. Biol. Chem. (1991) 266: 10796-10801.) increased susceptibility to atherosclerosis.(Marotti, K.R., et al: Nature (1993) 364: 73-75.). Also, inhibition of CETP activity with an inhibitory antibody raises HDL-cholesterol in hamster (Evans, G.F., et al: J. of Lipid Research (1994) 35: 1634- 1645.) and rabbit (Whitlock, M.E., et al: J. Clin. Invest. (1989) 84: 129-137). Suppression of increased plasma CETP by intravenous injection with antisense oligodeoxynucleotides against CETP mRNA reduced atherosclerosis in cholesterol- fed rabbits (Sugano, M., et al: J. ofBiol. Chem. (1998) 273: 5033-5036.) Importantly, human subjects deficient in plasma CETP, due to a genetic mutation possess markedly elevated plasma HDL-cholesterol levels and apolipoprotein A-l, the major apoprotein component of HDL. In addition, most demonstrate markedly decreased plasma LDL cholesterol and apolipoprotein B (the major apolipoprotein component of LDL. (Inazu, A., Brown, M.L., Hesier, C.B., et al.: N. Engl. J. Med. (1990) 323: 1234- 1238.)
Given the negative correlation between the levels of HDL cholesterol and HDL associated lipoproteins, and the positive correlation between triglycerides, LDL cholesterol, and their associated apolipoproteins in blood with the development of cardiovascular, cerebral vascular and peripheral vascular diseases, the Formula I compounds of this invention, their prodrugs and the salts of such compounds and prodrugs, by virtue of their pharmacologic action, are useful for the prevention, arrestment and/or regression of atherosclerosis and its associated disease states. These include cardiovascular disorders (e.g., angina, cardiac ischemia and myocardial infarction), complications due to cardiovascular disease therapies (e.g., reperfusion injury and angioplastic restenosis), hypertension, stroke, and atherosclerosis associated with organ transplantation.
Because of the beneficial effects widely associated with elevated HDL levels, an agent which inhibits CETP activity in humans, by virtue of its HDL increasing ability, also provides valuable avenues for therapy in a number of other disease areas as well.
Thus, given the ability of the Formula I compounds of this invention, their prodrugs and the salts of such compounds and prodrugs to alter lipoprotein composition via inhibition of cholesterol ester transfer, they are of use in the treatment of vascular complications associated with diabetes. Hyperlipidemia is present in most subjects with diabetes mellitus (Howard, B.V. 1987. J. Lipid Res. 28, 613). Even in the presence of normal lipid levels, diabetic subjects experience a greater risk of cardiovascular disease (Kannel, W.B. and McGee, D.L. 1979. Diabetes Care 2, 120). CETP-mediated cholesteryl ester transfer is known to be abnormally increased in both insulin-dependent (Bagdade, J.D., Subbaiah, PN. and Ritter, M.C. 1991. Eur. J. Clin. Invest. 21, 161) and non-insulin dependent diabetes (Bagdade. J.D., Ritter, M.C, Lane, J. and Subbaiah. 1993. Atherosclerosis 104, 69). It has been suggested that the abnormal increase in cholesterol transfer results in changes in lipoprotein composition, particularly for VLDL and LDL, that are more atherogenic (Bagdade, J.D., Wagner, J.D., Rudel, L.L, and Clarkson, T.B. 1995. J. Lipid Res. 36, 759). These changes would not necessarily be observed during routine lipid screening. Thus the present invention will be useful in reducing the risk of vascular complications as a result of the diabetic condition.
The described agents are useful in the treatment of obesity. In both humans (Radeau, T., Lau, P., Robb, M., McDonnell, M., Ailhaud, G. and McPherson, R., 1995. Journal of Lipid Research. 36 (12):2552-61) and nonhuman primates (Quinet, E., Tall, A., Ramakrishnan, R. and Rudel, L., 1991. Journal of Clinical Investigation. 87 (5): 1559-66) mRΝA for CETP is expressed at high levels in adipose tissue. The adipose message increases with fat feeding (Martin, L. J., Connelly, P. W., Νancoo, D., Wood, Ν., Zhang, Z. J., Maguire, G., Quinet, E., Tall, A. R., Marcel, Y. L. and McPherson, R., 1993. Journal of Lipid Research. 34 (3):437-46), and is translated into functional transfer protein and through secretion contributes significantly to plasma CETP levels. In human adipocytes the bulk of cholesterol is provided by plasma LDL and HDL (Fong, B. S., and Angel, A., 1989. Biochimica etBiophysica Ada. 1004 (1):53-60). The uptake of HDL cholesteryl ester is dependent in large part on CETP (Benoist, F., Lau, P., McDonnell, M., Doelle, H., Milne, R. and McPherson, R., 1997. Journal of Biological Chemistry. 272 (38):23572-7). This ability of CETP to stimulate HDL cholesteryl uptake, coupled with the enhanced binding of HDL to adipocytes in obese subjects (Jimenez, J. G., Fong, B., Julien, P., Despres, J. P., Rotstein, L, and Angel, A., 1989. International Journal of Obesity. 13 (5):699-709), suggests a role for CETP, not only in generating the low HDL phenotype for these subjects, but in the development of obesity itself by promoting cholesterol accumulation. Inhibitors of CETP activity that block this process therefore serve as useful adjuvants to dietary therapy in causing weight reduction. CETP inhibitors are useful in the treatment of inflammation due to Gram- negative sepsis and septic shock. For example, the systemic toxicity of Gram- negative sepsis is in large part due to endotoxin, a lipopoiysaccharide (LPS) released from the outer surface of the bacteria, which causes an extensive inflammatory response. Lipopoiysaccharide can form complexes with lipoproteins (Ulevitch, R.J., Johhston, A.R., and Weinstein, D.B., 1981. J. Clin. Invest. 67, 827-37). In vitro studies have demonstrated that binding of LPS to HDL substantially reduces the production and release of mediators of inflammation (Ulevitch, R.J., Johhston, A.R., 1978. J. Clin. Invest. 62, 1313-24). In vivo studies show that transgenic mice expressing human apo-AI and elevated HDL levels are protected from septic shock (Levine, D.M., Parker, T.S., Donnelly, T.M., Walsh, A.M., and Rubin, A.L. 1993. Proc. Natl. Acad. Sci. 90, 12040-44). Importantly, administration of reconstituted HDL to humans challenged with endotoxin resulted in a decreased inflammatory response (Pajkrt, D., Doran, J.E., Koster, F., Lerch, P.G., Amet, B., van der Poll, T., ten Cate, J.W., and van Deventer, S.J.H. 1996. J. Exp. Med. 184, 1601-08). The CETP inhibitors, by virtue of the fact that they raise HDL levels, attenuate the development of inflammation and septic shock.
The utility of the Formula I compounds of the invention, their prodrugs and the salts of such compounds and prodrugs as medical agents in the treatment of the above described disease/conditions in mammals (e.g. humans, male or female) is demonstrated by the activity of the compounds of this invention in conventional assays and the in vivo assay described beiow. The in vivo assay (with appropriate modifications within the skill in the art) may be used to determine the activity of other lipid or triglyceride controlling agents as well as the compounds of this invention. The combination protocol described beiow is useful for demonstrating the utility of the combinations of the lipid and triglyceride agents (e.g., the compounds of this invention) described herein. Such assays also provide a means whereby the activities of the Formula I compounds of this invention, their prodrugs and the salts of such compounds and prodrugs (or the other agents described herein) can be compared to each other and with the activities of other known compounds. The results of these comparisons are useful for determining dosage levels in mammals, including humans, for the treatment of such diseases. The following protocols can of course be varied by those skilled in the art. The hyperalphachoiesteroiemic activity of the Formula I compounds can be determined by assessing the effect of these compounds on the action of cholesteryl ester transfer protein by measuring the relative transfer ratio of radiolabeled lipids between lipoprotein fractions, essentially as previously described by Morton in J. Biol. Chem. 256, 11992, 1981 and by Dias in Clin. Chem. 34, 2322, 1988.
CETP IN VITRO ASSSAY The following is a brief description of the assay of cholesteryl ester transfer in human plasma (in vitro) and animal plasma (ex vivo): CETP activity in the presence or absence of drug is assayed by determining the transfer of 3H-labeled cholesteryl oleate (CO) from exogenous tracer HDL to the nonHDL lipoprotein fraction in human piasma, or from 3H-labeled LDL to the HDL fraction in transgenic mouse plasma. Labeled human lipoprotein substrates are prepared similarly to the method described by Morton in which the endogenous CETP activity in plasma is employed to transfer 3H-CO from phospholipid liposomes to all the lipoprotein fractions in plasma. 3H- labeled LDL and HDL are subsequently isolated by sequential ultracentrifugation at the density cuts of 1.019-1.063 and 1.10-1.21 g/ml, respectively. For the activity assay, 3H-labeled lipoprotein is added to plasma at 10-25 nmoles CO/ml and the samples incubated at 37° C for 2.5-3 hrs. Non-HDL lipoproteins are then precipitated by the addition of an equal volume of 20% (wt/vol) polyethylene glycol 8000 (Dias). The samples are centrifuged 750 g x 20 minutes and the radioactivity contained in the HDL containing supernatant determined by liquid scintillation. Introducing varying quantities of the compounds of this invention as a solution in dimethylsuifoxide to human plasma, before addition of the radiolabeled cholesteryl oleate, and comparing the relative amounts of radiolabel transferred allows relative cholesteryl ester transfer inhibitory activities to be determined.
CETP IN VIVO ASSSAY Activity of these compounds in vivo can be determined by the amount of agent required to be administered, relative to control, to inhibit cholesteryl ester transfer activity by 50% at various time points ex vivo or to elevate HDL cholesterol by a given percentage in a CETP-containing animal species. Transgenic mice expressing both human CETP and human apolipoprotein Al (Charles River, Boston, MA) may be used to assess compounds in vivo. The compounds to be examined are administered by oral gavage in an emulsion vehicle containing olive oil and sodium taurocholate. Blood is taken from mice retroorbitally before dosing. At various times after dosing, ranging from 4h to 24h, the animals are sacrificed, blood obtained by heart puncture, and lipid parameters measured, including total cholesterol, HDL and LDL cholesterol, and triglycerides. CETP activity is determined by a method similar to that described above except that 3H-cholesteryl oleate containing LDL is used as the donor source as opposed to HDL. The values obtained for iipids and transfer activity are compared to those obtained prior to dosing and/or to those from mice receiving vehicle alone. PLASMA LIPIDS ASSAY
The activity of these compounds may also be demonstrated by determining the amount of agent required to alter plasma iipid levels, for example HDL cholesterol levels, LDL cholesterol levels, VLDL cholesterol levels or triglycerides, in the plasma of certain mammals, for example marmosets that possess CETP activity and a plasma lipoprotein profile similar to that of humans (Crook et al. Arteriosclerosis 10, 625, 1990). Adult marmosets are assigned to treatment groups so that each group has a similar mean ±SD for total, HDL, and/or LDL plasma cholesterol concentrations. After group assignment, marmosets are dosed daily with compound as a dietary admix or by intragastric intubation for from one to eight days. Control marmosets receive only the dosing vehicle. Plasma total, LDL, VLDL and HDL cholesterol values can be determined at any point during the study by obtaining blood from an antecubital vein and separating plasma lipoproteins into their individual subclasses by density gradient centrifugation, and by measuring cholesterol concentration as previously described (Crook et al. Arteriosclerosis 10, 625, 1990). IN VIVO ATHEROSCLEROSIS ASSAY
Anti-atherosclerotic effects of the compounds can be determined by the amount of compound required to reduce the iipid deposition in rabbit aorta. Male New Zealand White rabbits are fed a diet containing 0.2% cholesterol and 10% coconut oil for 4 days (meal-fed once per day). Rabbits are bled from the marginal ear vein and total plasma cholesterol values are determined from these samples. The rabbits are then assigned to treatment groups so that each group has a similar mean ±SD for total plasma cholesterol concentration, HDL cholesterol concentration, triglyceride concentration and/or cholesteryl ester transfer protein activity. After group assignment, rabbits are dosed daily with compound given as a dietary admix or on a small piece of gelatin based confection. Control rabbits receive only the dosing vehicle, be it the food or the gelatin confection. The cholesterol/coconut oil diet is continued along with the compound administration throughout the study. Plasma cholesterol values and cholesteryl ester transfer protein activity can be determined at any point during the study by obtaining blood from the marginal ear vein. After 3-5 months, the rabbits are sacrificed and the aortae are removed from the thoracic arch to the branch of the iliac arteries. The aortae are cleaned of adventitia, opened longitudinally and then stained with Sudan IV as described by Holman et. al. (Lab. Invest. 1958, 7, 42-47). The percent of the surface area stained is quantitated by densitometry using an Optimas Image Analyzing System (Image Processing Systems). Reduced lipid deposition is indicated by a reduction in the percent surface area stained in the compound-receiving group in comparison with the control rabbits.
ANTIOBESITY PROTOCOL The ability of CETP inhibitors to cause weight loss can be assessed in obese human subjects with body mass index (BMI) > 30 kg/m2. Doses of inhibitor are administered sufficient to result in an increase of > 25% in HDL cholesterol levels. BMI and body fat distribution, defined as waist (W) to hip (H) ratio (WHR), are monitored during the course of the 3-6 month studies, and the results for treatment groups compared to those receiving placebo.
IN VIVO SEPSIS ASSAY
In vivo studies show that transgenic mice expressing human apo-AI and elevated HDL levels are protected from septic shock. Thus the ability of CETP inhibitors to protect from septic shock can be demonstrated in transgenic mice expressing both human apo-AI and human CETP transgenes (Levine, D. M., Parker, T.S., Donnelly, T. M., Walsh, A. M. and Rubin, A.L., 1993. Proc. Natl. Acad. Sci. 90, 12040-44). LPS derived from E. coli is administered at 30mg/kg by i.p. injection to animals which have been administered a CETP inhibitor at an appropriate dose to result in elevation of HDL. The number of surviving mice is determined at times up to 48h after LPS injection and compared to those mice administered vehicle (minus CETP inhibitor) only. Administration of the compounds of this invention can be via any method which delivers a compound of this invention systemically and/or locally. These methods include oral routes, parenteral, intraduodenal routes, etc. Generally, the compounds of this invention are administered orally, but parenteral administration (e.g., intravenous, intramuscular, subcutaneous or intramedullary) may be utilized, for example, where oral administration is inappropriate for the target or where the patient is unable to ingest the drug.
In general an amount of a compound of this invention is used that is sufficient to achieve the therapeutic effect desired (e.g., HDL elevation). In general an effective dosage for the Formula I compounds of this invention, their prodrugs and the salts of such compounds and prodrugs is in the range of 0.01 to 10 mg/kg/day, preferably 0.1 to 5 mg/kg/day.
A dosage of the combination pharmaceutical agents to be used in conjuction with the CETP inhibitors is used that is effective for the indication being treated. For example, typically an effective dosage for HMG-CoA reductase inhibitors is in the range of 0.01 to 100 mg/kg/day. In general an effect dosage for the MTP/Apo B secretion inhibitors is in the range of 0.01 to 100 mg/kg/day.
The compounds of the present invention are generally administered in the form of a pharmaceutical composition comprising at least one of the compounds of this invention together with a pharmaceutically acceptable vehicle, diluent or carrier. Thus, the compounds of this invention can be administered individually or together in any conventional oral, parenteral, rectal or transdermal dosage form.
For oral administration a pharmaceutical composition can take the form of solutions, suspensions, tablets, pills, capsules, powders, and the like. Tablets containing various excipients such as sodium citrate, calcium carbonate and calcium phosphate are employed along with various disintegrants such as starch and preferably potato or tapioca starch and certain complex silicates, together with binding agents such as polyvinylpyrrolidone, sucrose, gelatin and acacia. Additionally, lubricating agents such as magnesium stearate, sodium lauryl sulfate and talc are often very useful for tabletting puφoses. Solid compositions of a similar type are also employed as fillers in soft and hard-filled gelatin capsules; preferred materials in this connection also include lactose or milk sugar as well as high molecular weight polyethylene glycols. A preferred formulation is a solution or suspension in an oil, for example olive oil, Miglyol™ or Capmul™, in a soft gelatin capsule. Antioxidants may be added to prevent long term degradation as appropriate. When aqueous suspensions and/or elixirs are desired for oral administration, the compounds of this invention can be combined with various sweetening agents, flavoring agents, coloring agents, emulsifying agents and/or suspending agents, as well as such diiuents as water, ethanol, propylene glycol, glycerin and various like combinations thereof.
For puφoses of parenteral administration, solutions in sesame or peanut oil or in aqueous propylene giycol can be employed, as well as sterile aqueous solutions of the corresponding water-soluble salts. Such aqueous solutions may be suitably buffered, if necessary, and the liquid diluent first rendered isotonic with sufficient saline or glucose. These aqueous solutions are especially suitable for intravenous, intramuscular, subcutaneous and intraperitoneal injection puφoses. in this connection, the sterile aqueous media employed are all readily obtainable by standard techniques well-known to those skilled in the art.
For puφoses of transdermal (e.g..topical) administration, dilute sterile, aqueous or partially aqueous solutions (usually in about 0.1% to 5% concentration), otherwise similar to the above parenteral solutions, are prepared.
Methods of preparing various pharmaceutical compositions with a certain amount of active ingredient are known, or will be apparent in light of this disclosure, to those skilled in this art. For examples of methods of preparing pharmaceutical compositions, see Remington's Pharmaceutical Sciences. Mack Publishing Company, Easter, Pa., 15th Edition (1975).
Pharmaceutical compositions according to the invention may contain 0.1%- 95% of the compound(s) of this invention, preferably 1 %-70%. In any event, the composition or formulation to be administered will contain a quantity of a compound(s) according to the invention in an amount effective to treat the disease/condition of the subject being treated, e.g., atherosclerosis.
Since the present invention has an aspect that relates to the treatment of the disease/conditions described herein with a combination of active ingredients which may be administered separately, the invention also relates to combining separate pharmaceutical compositions in kit form. The kit comprises two separate pharmaceutical compositions: a compound of Formula I a prodrug thereof or a salt of such compound or prodrug and a second compound as described above. The kit comprises means for containing the separate compositions such as a container, a divided bottle or a divided foil packet. Typically the kit comprises directions for the administration of the separate components. The kit form is particularly advantageous when the separate components are preferably administered in different dosage forms (e.g., orai and parenteral), are administered at different dosage intervals, or when titration of the individual components of the combination is desired by the prescribing physician.
An example of such a kit is a so-called biister pack. Blister packs are well known in the packaging industry and are being widely used for the packaging of pharmaceutical unit dosage forms (tablets, capsules, and the like). Blister packs generally consist of a sheet of relatively stiff material covered with a foil of a preferably transparent plastic material. During the packaging process recesses are formed in the plastic foil. The recesses have the size and shape of the tablets or capsules to be packed. Next, the tablets or capsules are placed in the recesses and the sheet of relatively stiff material is sealed against the plastic foil at the face of the foil which is opposite from the direction in which the recesses were formed. As a result, the tablets or capsules are sealed in the recesses between the plastic foil and the sheet. Preferably the strength of the sheet is such that the tablets or capsules can be removed from the blister pack by manually applying pressure on the recesses whereby an opening is formed in the sheet at the place of the recess. The tablet or capsule can then be removed via said opening.
It may be desirable to provide a memory aid on the kit, e.g., in the form of numbers next to the tablets or capsules whereby the numbers correspond with the days of the regimen which the tablets or capsules so specified should be ingested. Another example of such a memory aid is a calendar printed on the card, e.g., as follows "First Week, Monday, Tuesday, ...etc.... Second Week, Monday, Tuesday,..." etc. Other variations of memory aids will be readily apparent. A "daily dose" can be a single tablet or capsule or several pills or capsules to be taken on a given day. Also, a daily dose of Formula I compound can consist of one tablet or capsule whiie a daily dose of the second compound can consist of several tablets or capsules and vice versa. The memory aid should reflect this. In another specific embodiment of the invention, a dispenser designed to dispense the daily doses one at a time in the order of their intended use is provided. Preferably, the dispenser is equipped with a memory-aid, so as to further facilitate compliance with the regimen. An example of such a memory-aid is a mechanical counter which indicates the number of daily doses that has been dispensed. Another example of such a memory-aid is a battery-powered micro-chip memory coupied with a liquid crystal readout, or audible reminder signal which, for example, reads out the date that the last daily dose has been taken and/or reminds one when the next dose is to be taken. The compounds of this invention either alone or in combination with each other or other compounds generally will be administered in a convenient formulation. The following formulation examples only are illustrative and are not intended to limit the scope of the present invention.
In the formulations which follow, "active ingredient" means a compound of this invention.
Formulation 1 : Gelatin Capsules
Hard gelatin capsules are prepared using the following: Ingredient Quantity (mg/capsule)
Active ingredient 0.25-100
Starch, NF 0-650
Starch flowabie powder 0-50
Silicone fluid 350 centistokes 0-15
A tablet formulation is prepared using the ingredients below: Formulation 2: Tablets
Ingredient Quantity (mg/tablet)
Active ingredient 0.25-100
Cellulose, microcrystalline 200-650
Silicon dioxide, fumed 10-650
Stearate acid 5-15 The components are blended and compressed to form tablets.
Alternatively, tablets each containing 0.25-100 mg of active ingredients are made up as follows: Formulation 3: Tablets
Ingredient Quantity (mg/tablet)
Active ingredient 0.25-100
Starch 45
Cellulose, microcrystalline 35 ~
Polyvinylpyrroiidone (as 10% solution in water) 4
Sodium carboxymethyl cellulose 4.5
Magnesium stearate 0.5
Talc 1
The active ingredients, starch, and cellulose are passed through a No. 45 mesh U.S. sieve and mixed thoroughly. The solution of polyvinylpyrroiidone is mixed with the resultant powders which are then passed through a No. 14 mesh U.S. sieve. The granules so produced are dried at 50° - 60°C and passed through a No. 18 mesh U.S. sieve. The sodium carboxymethyl starch, magnesium stearate, and talc, previously passed through a No. 60 U.S. sieve, are then added to the granules which, after mixing, are compressed on a tablet machine to yield tablets.
Suspensions each containing 0.25-100 mg of active ingredient per 5 ml dose are made as follows:
Formulation 4: Suspensions
Ingredient Quantity (mg/5 ml)
Active ingredient 0.25-100 mg
Sodium carboxymethyl cellulose 50 mg
Syrup 1.25 mg
Benzoic acid solution 0.10 mL
Flavor q.v.
Color q.v.
Purified Water to 5 mL
The active ingredient is passed through a No. 45 mesh U.S. sieve and mixed with the sodium carboxymethyl cellulose and syrup to form smooth paste. The benzoic acid solution, flavor, and color are diluted with some of the water and added, with stirring. Sufficient water is then added to produce the required volume. An aerosol solution is prepared containing the following ingredients: Formulation 5: Aerosol
Ingredient Quantity (% by weight)
Active ingredient 0.25
Ethanol 25.75
Propellant 22 (Chlorodifluoromethane) 70.00
The active ingredient is mixed with ethanol and the mixture added to a portion of the propellant 22, cooled to 30°C, and transferred to a filling device. The required amount is then fed to a stainless steel container and diluted with the remaining propellant. The valve units are then fitted to the container.
Suppositories are prepared as follows: Formulation 6: Suppositories
Ingredient Quantity (mg/suppository)
Active ingredient 250
Saturated fatty acid glycerides 2,000
The active ingredient is passed through a No. 60 mesh U.S. sieve and suspended in the saturated fatty acid glycerides previously melted using the minimal necessary heat. The mixture is then poured into a suppository mold of nominal 2 g capacity and allowed to cool.
An intravenous formulation is prepared as follows: Formulation 7: Intravenous Solution
Ingredient Quantity
Active ingredient dissolved in ethanol 1% 20 mg
Intraiipid™ emulsion 1 ,000 mL The solution of the above ingredients is intravenously administered to a patient at a rate of about 1 mL per minute.
Soft gelatin capsules are prepared using the following: Formulation 8: Soft Gelatin Capsule with Oil Formulation
Ingredient Quantity (mg/capsule)
Active ingredient 10-500
Olive Oil or Miglyol™ Oil 500-1000 The active ingredient above may also be a combination of agents.
GENERAL EXPERIMENTAL PROCEDURES NMR spectra were recorded on a Varian XL-300 (Varian Co., Palo Alto, California), a Bruker AM-300 spectrometer (Bruker Co., Billerica, Massachusetts) or a Varian Unity 400 at about 23°C at 300 MHz for proton and 75.4 mHz for carbon nuciei. Chemical shifts are expressed in parts per million downfield from tetramethylsiiane. The peak shapes are denoted as follows: s, singlet; d, doublet; t, triplet, q, quartet; m, multiple.; bs=broad singlet. Resonances designated as exchangeable did not appear in a separate NMR experiment where the sample was shaken with several drops of D20 in the same solvent. Atmospheric pressure chemical ionization (APCI) mass spectra were obtained on a Fisons Platform II Spectrometer. Chemical ionization mass spectra were obtained on a Hewlett-Packard 5989 instrument (Hewlett-Packard Co., Palo Alto, California) (ammonia ionization, PBMS). Where the intensity of chlorine or bromine-containing ions are described, the expected intensity ratio was observed (approximately 3:1 for 35CI/37CI-containing ions) and 1 :1 for ^Br/^Br-containing ions) and the intensity of only the lower mass ion is given.
Column chromatography was performed with either Baker Silica Gel (40 μm) (J.T. Baker, Phillipsburg, N.J.) or Silica Gel 60 (EM Sciences, Gibbstown, N.J.) in glass columns under low nitrogen pressure. Radial Chromatography was performed using a Chromatron (model 7924T, Harrison Research). Unless otherwise specified, reagents were used as obtained from commercial sources. Dimethylformamide, 2- propanol, tetrahydrofuran, and dichloromethane used as reaction solvents were the anhydrous grade supplied by Aldrich Chemical Company (Milwaukee, Wisconsin). Microanaiyses were performed by Schwarzkopf Microanalyticai Laboratory, Woodside, NY. The terms "concentrated" and "evaporated" refer to removal of solvent at water aspirator pressure on a rotary evaporator with a bath temperature of less than 45°C. Reactions conducted at "0-20°C" or "0-25°C" were conducted with initial cooling of the vessel in an insulated ice bath which was allowed to warm to room temperature over several hours. The abbreviation "min" and "h" stand for "minutes" and "hours" respectively. Examples Example 1 c/s-4-Benzyloxycarbonylamino-6-methoxy-2-methyl-1.2.3.4-tetrahvdro-quinoline-7- carboxylic acid methyl ester: To a solution of 4.26g methyl 2-methoxy-5- aminobenzoate (23.5 mmol) in 65 ml anhydrous dichloromethane was added 3.34g anhydrous sodium sulfate, and the resulting mixture was cooled to -25°C. Freshly distilled acetaldehyde (1.04g, 23.5 mmol) was added and the mixture was stirred at - 25°C for 2h. The supernatant was cannulated into a -25°C solution of N-vinyl-O- benzyl carbamate (1.4 g, 7.8 mmol) in 10 ml dichloromethane. BF3-etherate (111mg, 0.78 mmol) was added over 10 min, and the the reaction was stirred at -25°C for 1.5 h. The reaction was quenched cold with the addition of an aqueous 10% sodium sulfate solution. The aqueous phase was separated and was extracted with dichloromethane (100mL). The combined organic layers were dried (MgS04), filtered and concentrated, and the crude material was purified by silica gel chromotography using dichloromethane to afford the title product (0.99g). 1H NMR (DMSO-d6) δ 1.13 (d, 3H), 6.58 (s, 1H). Example 2 c/s-4-Benzyloxycarbonylamino-6-methoxy-2-methyl-3.4-dihydro-2H-quinoiine-1.7- dicarboxylic acid 1 -ethyl ester 7-methyl ester: To a solution of c/s-4- benzyloxycarbonylamino-6-methoxy-2-methyl-1 ,2,3,4-tetrahydro-quinoline-7- carboxyiic acid methyl ester (0.99 g, 2.6 mmol) in anhydrous dichloromethane (30 mL) was added pyridine (0.27 mL, 3.35 mmol). The mixture was cooled to 0 °C, and ethyl chloroformate (0.29 mL, 3.1 mmol) was slowly added. The reaction was stirred at 0 °C for 30 min, then at room temperature for 2.5 h. Additional aliquots of pyridine and methyl chloroformate were added to drive the reaction to completion. The reaction mixture was evaporated to dryness, and the residue was partitioned between ethyl acetate (75ml) and water (25ml). The organic layer was washed with water (25mL), dried over magnesium sulfate, filtered and concentrated in vacuo to give the crude product. Purification by silica gel chromatography using 25% ethyl acetate/hexanes as eluent afforded the title product (1.06 g). MS m/z 457 (M+ + 1 ); 1H NMR (DMSO-d6) δ 1.45 (d, 3H), 6.68 (s, 1H). Example 3 c/'s-4-Amino-6-methoxy-2-methyl-3.4-dihydro-2H-quinoline-1.7-dicarboxylic acid 1- ethyl ester 7-methyl ester: cs-4-Benzyioxycarbonyiamino-6-methoxy-2-methyl-3,4- dihydro-2H-quinoline-1 , 7-dicarboxylic acid 1 -ethyl ester 7-methyi ester (322 mg, 0.71 mmol), 10% palladium on carbon (32 mg) and cyclohexene (9ml) in 16 ml ethanol was heated to 80 °C for 1.25 h. The reaction mixture was cooled to room temperature, filtered through Celite®, and concentrated in vacuo. Purification by silica gel chromatography using 1% isopropanol-dichloromethane afforded the title product (208 mg, 91%). 1H NMR (DMSO-d6) δ 1.02 (d, 3H), 6.68 (s, 1H). Example 4 c/s-4-(3.5-Bis-trifluoromethyl-benzylaminoV6-methoxy-2-methyl-3.4-dihydro-2H- quinoiine-1.7-dicarboxylic acid 1-ethyl ester 7-methyl ester: To a solution of 4-amino- 6-methoxy-2-methyl-3,4-dihydro-2H-quinoline-1 ,7-dicarboxyiic acid 1 -ethyl ester 7- methyl ester (208 mg, 0.64 mmol) in anhydrous 1 ,2-dichloroethane (5 mL) was added acetic acid (0.041 mL, 0.71 mmol), followed by 3,5-bis(trifluoromethyl)benzaldehyde (172 mg, 0.71 mmol) and sodium triacetoxyborohydride (0.205 g, 0.97 mmol). The reaction was stirred at room temperature for 2 h. The reaction mixture was partitioned between dichloromethane (45ml) and water (100 ml) and the aqueous phase was adjusted to pH 9. The aqueous phase was extracted with dichloromethane (3 x 10 mL). The combined extracts were dried over magnesium sulfate, filtered and concentrated in vacuo . Purification by silica gel chromatography using 20% acetone/hexanes as eiuent afforded the title product (337 mg, 95%). MS m/z 567 (M+ + NH4); 1H NMR (CDCI3) δ 1.40 (d, 3H), 7.14 (s, 1H). Example 5 2-Methyl-4-oxo-6-trifluoromethyl-3.4-dihydro-2H-quinoiine-1 -carboxylic acid benzyl ester. 4-Methoxy-6-trifluoromethylquinoline (5.0 g, 22.0 mmol) was dissolved in anhydrous tetrahydrofuran (20 mL). The mixture was cooled to -78 °C, and methyl magnesium chloride (73 mL of a 3.0M solution in tetrahydrofuran, 220 mmol) was added to the resulting slurry. After addition, benzyl chloroformate (41 mL, 286 mmol) was added to the slurry. After stirring at room temperature overnight, 75 mL of methanol was added followed by 75 mL of a 1 N aqueous HCI solution. After hydrolysis of the intermediate enol ether was judged to be complete by thin layer chromatography, the voiatiies were removed in vacuo , and the remaining aqueous phase was extracted with ethyl acetate (3 x 150 mL). The organic phases were combined and washed with a saturated sodium bicarbonate solution (2 x 75 mL), brine (100 mL), dried over sodium sulfate, filtered, and concentrated in vacuo to give 47 g of crude product. Purification by silica gel chromatography using 10% ethyl acetate/hexanes as eiuent afforded 5.54 g of the title product (69%). 1H NMR (CDCI3) δ 1.25 (d, 3H, J = 7 Hz), 2.62 (dd, 1H, J = 17, 2 Hz), 3.05 (dd, 1 H, J = 17, 6 Hz), 5.7- 5.2 (m, 1H), 5.28 (d, 1 H, J = 12 Hz), 5.35 (d, 1H, J = 12 Hz), 7.3-7.5 (m, 5H), 7.71 (dd, 1 H, J = 9, 2 Hz), 8.05 (d, 1H, J = 9 Hz), 8.27 (d, 1 H, J = 2 Hz). Example 6 8-Chloro-2-methyl-4-oxo-3.4-dihydro-2H-quinoline-1 -carboxylic acid ethyl ester. 8- Chloro-4-methoxyquinoline (1.0 g, 5.2 mmol, dissolved in 6 mL of anhydrous tetrahydrofuran) was added dropwise via cannula to a solution of methyllithium (18.4 mL of a 1.4 M solution in diethyl ether) cooled with an ice/water bath. After 45 min, ethyl chloroformate (4.9 mL, 51.7 mmol) was added to the reaction mixture via syringe. After 1.5 hours, the reaction mixture was quenched with 60 mL of a 1 N aqueous HCI solution, 100 mL of THF and 20 mL of methanol. After 3 days, the tetrahydrofuran was removed in vacuo , and the remaining aqueous phase was extracted with ethyl acetate (3 x 100 mL). The organic phases were combined and washed with brine, dried over sodium sulfate, filtered, and concentrated in vacuo to give 1.5 g of crude product. Purification by silica gel chromatography using 0-20% ethyl acetate/hexanes as eiuent afforded 0.79 g of the title product (57%). 1H NMR (CDCI3) δ 1.2 (d, 3H), 1.3 (t, 3H), 2.6 (d, 1H), 3.1 (dd, 1 H), 4.2-4.4 (m, 2H), 5.1-5.2 (m, 1H), 7.2 (d, 1H), 7.6 (d, 1H), 7.9 (d, 1H). Example 7 6-Fluoro-2-methyl-4-oxo-7-trifluoromethyl-3.4-dihydro-2H-quinoline. 6-Fluoro-2- methyl-4-oxo-7-trifluoromethyl-3,4-dihydro-2H-quinoline-1 -carboxylic acid benzyl ester (3.0 g, 7.9 mmol) was combined with 250 mg of 10% Pd/C in 50 mL of ethanol and agitated under a hydrogen atmosphere (40 psi) for 1 hour before being filtered through Celite®, rinsing with ethyl acetate, and evaporating the volatiies under reduced pressure to afford 1.34 (69%) of the title compound as a colorless oil. 1H NMR (CDCI3) δ 1.4 (d, 3H), 2.5 (dd, 1 H), 2.7 (dd, 1 H), 3.7-3.9 (m, 1 H), 4.4 (bs, 1H), 6.9 (d, 1 H), 7.6 (d, 1H). Example 8 6-Fluoro-2-methyl-4-oxo-7-trifluoromethyl-3.4-dihydro-2H-quinoiine-1-carboxylic acid isopropyi ester. 6-Fluoro-2-methyl-4-oxo-7-trifluoromethyl-3,4-dihydro-2H-quinoline (1.01 g, 4.08 mmol) was dissolved in 10 mL of dichloromethane and 5 mL of pyridine, and stirred as isopropyl chloroformate (48 mL of a 1 molar solution in toluene, 48 mmol) was added slowly via syringe. After stirring overnight, 100 ml of a 1 M aqueous potassium hydroxide solution was added and the aqueous phase was extracted with ethyl acetate (3 x 75 mL), the combined organics were washed with 1 M HCI (2 x 50 mL), 100 mL each of a saturated sodium bicarbonate solution and brine, before being dried over sodium sulfate, filtered and the filtrate concentrated under reduced pressure to afford 1.1 g of an oii, which was purified by chromatography on silica gel eluting with 10% ethyl acetate in hexanes to afford 940 mg (69%) of the title compound as an oil. 1H NMR (CDCI3) δ 1.24 (d, 3H), 1.34 (d, 3H), 1.38 (d, 3H), 2.65 (dd, 1 H), 3.05 (dd, 1H), 5.1-5.3 (m, 2H), Example 9 4-(3.5-Bis-trifluoromethyl-benzylimino)-2-methyl-6-trifluoromethyl-3.4-dihydro-2H- quinoline-1 -carboxylic acid benzyl ester. 2-Methyl-4-oxo-6-trifluoromethyl-3,4-dihydro- 2H-quinoline-1 -carboxylic acid benzyl ester (5.5 g, 15.1 mmol) was combined with triethylamine (15 mL, 106 mmol) and 3,5-bis-trifiuoromethyl-benzylamine (5.5 g, 22.7 mmol) in anhydrous dichloromethane (80 mL). This solution was cooled in a room temperature water bath as 7.6 mL of 1 M solution of titanium tetrachloride (TiC^) in dichloromethane (7.6 mmol) was slowly added. The reaction was stirred at room temperature overnight before the mixture was poured into a stirred solution of water (125 mL) and potassium carbonate (15 g). After filtration of the resulting emulsion, the filtrate was extracted with ethyl acetate (1 x 200 mL, then 2 x 75 mL), the combined organic phases were washed with water (100 mL), brine (50 mL), dried over sodium sulfate, filtered, and concentrated in vacuo to give the title product (10.63 g, > theory). 1H NMR (CDCI3) δ 1.2 (d, 3H), 2.7-2.9 (m, 2H), 4.0-4.1 (m, 1H), 4.65 (d, 1 H), 4.75 (d, 1 H), 5.1-5.4 (m, 2H), 7.3-7.5 (m, 5H). Example 10 cis- and trans-4-(3.5-Bis-trifluoromethyl-benzylamino -2-methyl-6-trifluoromethyl-3.4- dihydro-2H-quinoline-1 -carboxylic acid benzyl ester. 4-(3,5-Bis-trifIuoromethyl-benzylimino)-2-methyl-6-trifluoromethyl-3,4-dihydro-2H- quinoiine-1-carboxyiic acid benzyl ester (-15 mmol) was dissolved in 230 mL of a 0.2M solution of zinc borohydride in diethyl ether (46 mmol). After the reaction was stirred overnight, methanol was added to react excess reagent followed by 400 mL of water. The mixture was treated with potassium carbonate until it became biphasic (pH = 10), followed by extraction with ethyl acetate (1 x 300, then 2 x 100 mL). The organic phases were combined and washed with water (100 mL), brine (75 mL), dried over sodium sulfate, filtered, and concentrated in vacuo to give 10.6 g of a crude mixture of product amines. Purification by silica gel chromatography using 0- 50% ethyl acetate/hexanes as eiuent afforded the cis title product (2.93 g, 33%). 1H NMR (CDCI3) δ 1.2-1.3 (m, 1H), 1.25 (d, 1H, J = 6 Hz), 2.7 (ddd, 1 H), 3.6 (dd, 1H), 4.1 (d, 1 H), 4.2 (d, 1H), 4.45 (ddq, 1H), 5.17 (d, 1H), 5.27 (d, 1 H), 7.35 (bs, 5H), 7.5 (d, 1H), 7.6 (d, 1H), 7.80 (s, 1H), 7.82 (s, 1H), 7.95 (s, 2H). Continued elutions using increasing concentrations of ethyl acetate provide the trans title product. Example 11
6-Fluoro-4-hvdroxyimino-2-methyl-7-trifluoromethyl-3.4-dihydro-2H-quinoline-1- carboxylic acid isopropyl ester. A stirred solution of 6-fluoro-2-methyl-4-oxo-7- trifluoromethyl-3,4-dihydro-2H-quinoline-1 -carboxylic acid isopropyl ester (940 mg, 2.8 mmol), hydroxylamine hydrochloride (215 mg, 3.1 mmoi), and sodium acetate (254 mg, 3.1 mmol) in ethanol (20 mL) was heated at reflux for 3 h. Water (25 mL) was added, and the volatiies were removed in vacuo. Ethyl acetate (100 mL) was added, and the mixture was stirred vigorously for 10 min. The aqueous phase was separated and extracted with ethyl acetate (2 x 100 mL). The combined organic layers were washed with brine (100 mL), dried over sodium sulfate, filtered and concentrated in vacuo to give the title compound as a white foam (937 mg, ca. 100%): 1H NMR (CDCI3) δ 1.1 (d, 3H), 1.28 (d, 3H), 1.34 (d, 3H), 2.73 (dd, 1 H), 3.09 (d, 1H), 5.0-5.1 (m, 2H), 7.68 (d, 1H), 7.88 (s, 1 H). Example 12 cis and frans-4-Amino-6-fluoro-2-methyl-7-trifluoromethyl-3.4-dihydro-2H-quinoline-1 - carboxylic acid isopropyl ester. To a stirred solution of 6-fluoro-4-hydroxyimino-2- methyl-7-trifluoromethyl-3,4-dihydro-2H-quinoline-1 -carboxylic acid isopropyl ester (937 mg, 2.82 mmoi) in ethanol (40 mL) and aqueous 2N KOH (40 mL, 80 mmol) was added aluminum-nickel alloy (1.57 g, 1 :1 by weight, 18 mmol) in portions over 15 min (gas evolution). After stirring for 3 hours, water (25 mL) was added then the slurry filtered through a nylon filter, rinsing with ethyl acetate. The voiatiies were removed in vacuo, and the resulting aqueous phase was extracted with ethyl acetate (3 x 100 mL). The combined organic phases were washed with brine (100 mL), dried over sodium sulfate, filtered, and concentrated in vacuo to give 946 mg of an oil containing the title compound as an approximately 3:1 mixture of cis and trans diastereomers (94%). 1H NMR -Major (cis) product (CDCI3) δ 1.2 (d, 3H), 1.25 (d, 3H), 1.3 (d, 3H), 2.5 (ddd, 1 H), 3.8 (bd, 1H), 4.4-4.6 (m, 1H), 5.0 (septet, 1H) 7.3 (d, 1 H), 7.65 (d, 1 H). Example 13. c/s-4-[(3.5-Bis-trifluoromethyl-benzvπ-methoxycarbonyl-amino]-6-methoxy-2-methyl- 3.4-dihydro-2H-quinoline-1.7-dicarboxylic acid 1 -ethyl ester 7-methvi ester. c/s-4-(3,5- Bis-trifluoromethyl-benzylamino)-6-methoxy-2-methyl-3,4-dihydro-2H-quinoline-1,7- dicarboxylic acid 1-ethyl ester 7-methyl ester (335 mg, 0.61 mmol) was dissolved in anhydrous dichloromethane (5 mL), and pyridine (0.20 mL, 2.5 mmol) was added. Methyl chloroformate (0.057 mL, 0.73 mmol) was slowly added and the reaction was stirred at room temperature for 3 h. Additional aliquots of reagents were added to drive the reaction to completion. The reaction mixture was then diluted with 35 ml dichloromethane, washed with an aqueous 1 N HCI solution (2 x 7mL) and with a saturated sodium bicarbonate solution (2 x 7mL) saturated NaHC03. The organic layer was dried over magnesium suifate, filtered and concentrated in vacuo. Purification by silica gel chromatography using 20% acetone/hexanes as eiuent afforded the title product (0.50 g, 90%). MS mfz 607 (M+ + 1 ); 1H NMR (CDCI3) δ 1.5- 1.75 (br, 1 H), 6.42 (s, 1 H). Using the appropriate starting materials, Examples 14-16, 19-21, 27, 34, 38,
42, 47 and 58 were prepared in an analogous manner to the sequence of reactions described for Examples 1 , 2, 3, 4, and 13; and Examples 22, 24-26, 29, 31 , 32, 43, 46, 54, 56 and 59 were prepared in an analogous manner to the sequence of reactions described for Examples 5, 9, 10 and 13; and Examples 17, 18, 23, 28, 30, 35-37, 39-41 , 44, 45 and 48 were prepared in an analogous manner to the sequence of reactions described for Examples 5, 11 , 12, 4 and 13; and Examples 33, 49-52 and 57 were prepared in an analogous manner to the sequence of reactions described for Examples 6, 9, 10, and 13; and Examples 53 and 55 were prepared in an analogous manner to the sequence of reactions described for Examples 5, 7, 8,
11 , 12, 4 and 13.
Example 14. c/s-4-[(3.5-Bis-trifluoromethyl-benzvh-methoxycarbonyl-amino]-7-methoxy-2-methyl- 3.4-dihvdro-2H-quinoline-1.6-dicarboxylic acid 1 -ethyl ester 6-methyl ester. MS m/z
607 (M+ + 1 ); 1H NMR (DMSO-d6) δ 7.08 (s, 1H), 1.34-1.6 (br, 1 H).
Example 15. c/s-4-[(3.5-Bis-trifluoromethyl-benzyπ-methoxycarbonyl-amino]-7- ethoxycarbonylmethyl-6-methoxy-2-methyl-3.4-dihvdro-2H-quinoline-1 -carboxylic acid ethyl ester. MS m/z 634.5 (M+); 1H NMR (CDCI3) δ 3.6 (AB. 2H), 3.8 (s, 3H), 6.4
(s, 1H).
Example 16. c/s-4-[(3.5-Bis-trifluoromethyl-benzyl)-methoxycarbonyl-amino]-2-methyl-3.4-dihvdro-
2H-quinoline-1.6.7-tricarboxylic acid 1 -ethyl ester 6.7-dimethyl ester. MS m/z 652 (M+ + NH4 + ); 1H NMR (CDCI3) δ 7.22 (s, 1H), 3.84 (s, 3H).
Example 17. c/s-4-[(3.5-Bis-trifluoromethyl-benzyl)-methoxycarbonyi-amino]-2-methyl-7- trifluoromethyl-3.4-dihvdro-2H-quinoline-1 -carboxylic acid ethyl ester. MS m/z 587
(M+ + 1 ), 604 (M+ + 18); 1H NMR (CDCI3) δ 7.82 (C8, s, 1 H), 6.40 (C5, s, 1 H), 1.20 (C2-Me, d, 3H, J = 6.1 Hz).
Example 18. c/'s-4-[(3.5-Bis-trifluoromethyl-benzyπ-methoxycarbonyl-amino -7-methoxy-2.6- dimethyl-3.4-dihydro-2H-quinoline-1 -carboxylic acid ethyl ester. MS m/z 563 (M+ + 1 );
1H NMR (CDCI3) δ 6.39 (s, 1 H), 1.44-1.64 (aliph., 6H). Example 19. c/s-4-[(3.5-Bis-trifluoromethyl-benzvπ-methoxycarbonyl-amino]-5- methanesulfonylmethyl-6-methoxy-2-methyl-3.4-dihydro-2H-quinoline-1-carboxylic acid ethyl ester. MS m/z 640.3 (M+);1H NMR (CDCI3) δ 1.6 (d, 3H), 2.6 (s, 3H), 3.5 (s,
3H), 6.7 (d, 1 H), 7.4 (d, 1 H), 7.6 (d, 1H). Example 20. c/s-4-[(3.5-Bis-trifluoromethyl-benzyπ-methoxycarbonyl-amino]-7- methanesulfonylmethyl-6-methoxy-2-methyl-3.4-dihydro-2H-quinoline-1 -carboxylic acid ethyl ester. MS m/z 640.3 (M+); 1H NMR (CDCI3) δ 3.8 (m, 3H), 6.4 (s, 1H).
Example 21. c/s-4-[(3.5-Bis-trifluoromethyl-benzyπ-methoxycarbonyl-amino]-6- methanesulfonylmethyl-7-methoxy-2-methyl-3.4-dihydro-2H-quinoline-1-carboxylic acid ethyl ester. MS m/z 641 (M+ + 1 ); 1H NMR (CDCI3) δ 6.88 (s, 1 H), 3.81 (s, 3H).
Example 22. c/s-4-[(3.5-Bis-trifluoromethyl-benzyπ-ethoxycarbonyl-amino]-2-methyl-6- trifluoromethyl-3.4-dihydro-2H-quinoline-1 -carboxylic acid ethyl ester. MS m/z 601
(M+ + 1 ), 618 (M+ + 18); 1H NMR (CDCI3) δ 7.12 (C5, s, 1 H), 1.10 (C2-Me, d, 3H).
Example 23. c/s-4-[(3.5-Bis-trifluoromethyl-benzyπ-methoxycarbonyl-amino]-6-(methoxycarbonyl- methyl-amino)-2-methyl-3.4-dihvdro-2H-quinoline-1 -carboxylic acid ethyl ester. MS m/z 605 (M+ ), 623 (M+ + 18); 1H NMR (CDCI3) δ 6.83 (s, 1H), 3.81 (s, 3H).
Example 24. c/'s-4-[(3.5-Bis-trifluoromethyl-benzyh-methoxycarbonyl-amino]-7-chloro-2-methyl-3.4- dihydro-2H-quinoline-1 -carboxylic acid ethyl ester. MS m/z 553 (M+ + 1 ), 570 (M+ +
18); 1H NMR (CDCI3) δ 7.81 (s, 1H), 3.83 (s, 3H). Example 25. c/s-4-[(3.5-Bis-trifluoromethyl-benzyl)-methoxycaπ3θnyl-amino]-6-chloro-7-methoxy-2- methyl-3.4-dihvdro-2H-quinoline-1 -carboxylic acid ethyl ester. MS m/z 583 (M+ + 1 ),
600 (M+ + 18); 1H NMR (CDCI3) δ 7.79 (s, 1 H), 3.81 (s, 3H).
Example 26. c/s-4-[(3.5-Bis-trifluoromethyl-ben--yl)-methσxycarbonyl-amino]-6-fluoro-7-methoxy-2- methyl-3.4-dihvdro-2H-quinoline-1 -carboxylic acid ethyl ester. MS m/z 567 (M+ + 1 ),
584 (M+ + 18); 1H NMR (CDCI3) δ 6.65 (d, 1H, J = 11.1 Hz), 3.87 (s, 3H).
Example 27. c/s-4-[(3.5-Bis-trifluoromethyl-ben--yπ-methoxycarbonyl-amino]-6-methoxy-7-(1- methoxycarbonyl-cvclopentyl)-2-methyl-3.4-dihvdro-2H-quinoline-1-carboxylic acid ethyl ester. MS mfr. 674.3 (M+ ); 1H NMR (CDCI3) δ 1.2 (d, 3H), 3.6 (s, 3H), 3.7 (s,
3H), 3.8 (s, 3H), 6.4 (s, 1 H), 7.4 (s, 1H), 7.7 (s, 2H), 7.8 (s, 1 H). Example 28. c/s-4-[(3.5-Bis-trifluoromethyl-benzvπ-isopropoxycarbonyl-amino]-2-methyl-7- trifluoromethyl-3.4-dihvdro-2H-quinoline-1 -carboxylic acid ethyl ester. MS m/z 615
(M+ + 1), 632 (M+ + 18); 1H NMR (CDCI3) δ 7.75 (s, 1H), 2.25-2.10 (m, 1H). Example 29. c/s-4-[(3.5-Bis-trifluoromethyl-benzyπ-methoxycarbonyl-amino]-6-chloro-2-methyl-7- trifluoromethyl-3.4-dihvdro-2H-quinoline-1 -carboxylic acid ethyl ester. MS m/z 621
(M+ + 1 ), 638 (M+ + 18); 1H NMR (CDCI3) δ 7.71 (m, 1H), 3.84 (s, 3H).
Example 30. c/'s-4-[('3.5-Bis-trifluoromethyl-benzvh-methoxycarbonyl-amino]-2-methyl-6- trifluoromethyl-3.4-dihvdro-2H-quinoline-1 -carboxylic acid ethyl ester. MS m/z 588
(M+ + 2), 605 (M+ + 19); 1H NMR (CDCI3) δ 7.12 (C5, s, 1H), 3.83 (OMe, s, 3H), 1.16
(Me, d, 3H, J = 6.0 Hz).
Example 31. c/s-4-[(3.5-Bis-trifluoromethyl-benzvπ-methoxycarbonyl-amino]-6.7-dichloro-2-methyl-
3.4-dihydro-2H-quinoline-1 -carboxylic acid ethyl ester. MS m/z 587 (M+ + 1 ), 604 (M+
+ 18); 1H NMR (CDCI3) δ 7.71-7.65 (m, 2H), 3.83 (s, 3H).
Example 32. c/s-4-[(3.5-Bis-trifluoromethyl-benzyπ-methoxycarbonyl-amino]-6-chloro-2-methyl-3.4- dihvdro-2H-quinoline-1 -carboxylic acid ethyl ester. MS m/z 553 (M+ + 1 ), 570 (M+ +
18); H NMR (CDCI3) δ 6.88 (C5, s, 1 H), 3.81 (OMe, s, 3H), 1.16 (Me, d, 3H, J = 5.6
Hz).
Example 33. c/s-4-[(3.5-Bis-trifluoromethyl-benzvh-methoxycarbonyl-amino1-8-chloro-2-methyl-3.4- dihvdro-2H-quinoline-1 -carboxylic acid ethyl ester. MS m/z 552 (M+), 569 (M+ + 17);
1H NMR (CDCI3) δ 3.80 (OMe, s, 3H), 1.14 (Me, d, 3H, J = 6.2 Hz).
Example 34. c/s-4-[(3.5-Bis-trifluoromethyl-benzvπ-methoxycarbonyl-amino]-6.7-bis-tert- butoxycarbonylamino-2-methyl-3.4-dihvdro-2H-quinoline-1 -carboxylic acid ethyl ester. MS m/z 618 (M+- 130); 1H NMR (CDCI3) δ 3.8 (s, 3H), 7.4 (s, 1H).
Example 35. cs-4-[(3.5-Bis-trifluoromethyl-benzvi)-methoxycarbonyl-amino]-2.6-dimethyl-3.4- dihvdro-2H-quinoline-1 -carboxyiic acid ethyl ester. MS m/z 532 (M+ ), 533 (M+ + 1 );
1H NMR (CDCI3) δ 7.78 (s, 1H), 3.81 (s, 3H), 3.87 (s, 6H).
Example 36. c/s-4-[(3.5-Bis-trifluoromethyl-benzvh-methoxycarbonyl-amino]-6-methoxy-2-methyl-
7-trifluoromethyl-3.4-dihydro-2H-quinoline-1 -carboxylic acid ethyl ester. MS m/z 616.5
(M+ ); 1H NMR (CDCI3) δ 3.8 (s, 3H), 6.5 (s, 1H).
Example 37. c/s-4-[(3.5-Bis-trifluoromethyl-benzyh-methoxycarbonyl-amino]-2.6.7-trimethyl-3.4- dihydro-2H-quinoline-1 -carboxylic acid ethyl ester. MS m/z 547 (M+ + 1), 565 (M+ +
19); 1H NMR (CDCI3) δ 7.78 (s, 1H), 3.81 (s, 3H).
Example 38. cs-4-[(3.5-Bis-trifluoromethyl-benzvπ-methoxycarbonyl-amino]-6-(2-ethoxycarbonyl- ethylV2-methyl-3.4-dihydro-2H-quinoline-1 -carboxylic acid ethyl ester. MS m/z 618.2 (M+); 1H NMR (CDCI3) δ 2.6 (t, 2H), 2.9 (t, 2H), 3.8 (s, 3H), 6.7 (s, 1H), 7.8 (s, 1H).
Example 39. c/s-4-[(3.5-Bis-trifluoromethyl-benzyπ-methoxycarbonyl-amino]-2.5.6-trimethyl-3.4- dihydro-2H-quinoline-1 -carboxyiic acid ethyl ester. MS m/z 547 (M+ + 1), 565 (M+ +
19); 1H NMR (CDCI3) δ 7.64 (s, 1H), 1.65-1.50 (m, 6H). Example 40. c/s-4-[(3.5-Bis-trifluoromethyl-benzvh-methoxycarbonyl-amino]-6-tert-butyl-2-methyl-
3.4-dihydro-2H-quinoline-1 -carboxylic acid ethyl ester. MS m/z 575 (M+ + 1 ), 593 (M+
+ 19); 1H NMR (CDCI3) δ 6.89 (s, 1 H), 3.80 (s, 3H).
Example 41. c s-4-[(3.5-Bis-trifluoromethyl-benzyh-methoxycarbonyi-amino]-6-fluoro-2-methyl-3.4- dihydro-2H-quinoline-1 -carboxyiic acid ethyl ester. MS m/z 537 (M+ + 1 ); 1H NMR
(CDCI3) δ 3.80 (OMe, s, 3H), 1.18 (Me, d, 3H, J = 6.0 Hz).
Example 42. cs-4-[(3.5-Bis-trifluoromethyl-benzvπ-methoxycarbonyl-amino]-2.8-dimethyl-6- trifluoromethyl-3.4-dihvdro-2H-quinoline-1 -carboxylic acid ethyl ester. MS m/z 601.6
(M+ + 1), 617.5 (M+ + 17).
Example 43. c/s-4-[(3.5-Bis-trifluoromethyl-benzyh-methoxycarbonyl-amino]-6-bromo-2-methyl-3.4- dihydro-2H-quinoline-1 -carboxylic acid ethyl ester. MS m/z 597 (M+), 614 (M+ + 17),
616 (M+ + 19); 1H NMR (CDCI3) δ 7.03 (C5, s, 1 H), 3.82 (OMe, s, 3H), 1.16 (Me, d,
3H, J = 6.0 Hz). Example 44. - c/s-4-[(3.5-Bis-trifluoromethyl-benzyπ-methoxycarbonyl-amino]-6J-diethyl-2-methyl-
3.4-dihydro-2H-quinoline-1 -carboxyiic acid ethyl ester. MS /z 575 (M" + 1), 592 (M+
+ 18); 1H NMR (CDCI3) δ 7.30 (s, 1H), 3.81 (s, 3H).
Example 45. c/s-4-[(3.5-Bis-trifluoromethyl-benzyl)-methoxycarbonyl-amino]-6-ethyl-2-methyl-3.4- dihydro-2H-quinoline-1 -carboxylic acid ethyl ester. MS m/z 565 (M+ + 19); 1H NMR
(CDCI3) δ 6.72 (C5, s, 1 H), 3.80 (OMe, s, 3H).
Example 46. c/s-4-[(3.5-Bis-trifluoromethyl-benzyl)-methoxycarbonyi-amino]-6-bromo-2-methyl-7- trifluoromethyl-3.4-dihydro-2H-quinoline-1 -carboxyiic acid ethyl ester. MS m/z 666
(M+ + 1 ), 1H NMR (CDCI3) δ 7.71 (s, 2H), 3.84 (s, 3H).
Example 47. c/s-4-[(3.5-Bis-trifluoromethyl-benzyh-methoxycarbonyl-amino]-8-fluoro-2.6-dimethyl-
3.4-dihydro-2H-quinoline-1 -carboxylic acid ethyl ester. MS m/z 551 (M+ + H); 1H NMR (CDCI3) δ 7.80 (bs, 1 H), 7.7 (bs, 1H), 7.65 (bs, 1 H), 6.85 (bs, 1 H), 6.5 (bs,
1 H), 3.8 (s, 3H), 2.32 (s, 3H), 1.18 (d, 3H, J = 6.0 Hz).
Example 48. c/s-4-[(3.5-Bis-trifluoromethyl-benzvπ-methoxycarbonyl-amino]-6-isopropyl-2-methvi-
3.4-dihvdro-2H-guinoline-1 -carboxyiic acid ethyl ester. H NMR (CDCI3) δ 6.73 (C5, s, 1 H), 3.80 (OMe, s, 3H), 1.16 (Me, d, 3H, J = 6.0 Hz).
Example 49. c/s-4-[(3.5-Bis-trifluoromethyl-benzyl)-methoxycarbonyl-amino]-6.8-dichloro-2-methyl-
3.4-dihydro-2H-quinoline-1 -carboxylic acid ethyl ester. MS m/z 587 (M+ + H); 1H
NMR (CDCI3) δ 7.8 (bs, 1H), 7.7 (bs, 1 H), 7.65 (bs, 1 H), 7.4 (bs, 1H), 6.85 (bs, 1H), 3.8 (bs, 3H).
Example 50. c/s-4-[(3.5-Bis-trifluoromethyl-benzyh-methoxycarbonyl-amino]-2.6.8-trimethyl-3.4- dihydro-2H-quinoline-1 -carboxylic acid ethyl ester. MS m/z 547 (M+ + H); 1H NMR (CDCI3) δ 7.8 (bs, 1H), 7.7 (bs, 1H), 7.65 (bs, 1H), 6.69 (bs, 1H), 6.5 (bs, 1H), 3.78 (s, 3H), 2.3 (s, 3H), 2.18 (s, 3H). Example 51. c/s- -[(3.5-Bis-trifluoromethyl-benzyl)-ethoxycarbonyi-amino1-2.6.8-trimethyl-3.4- dihydro-2H-quinoline-1 -carboxylic acid ethyl ester. MS m/z 561 (M+ + H); 1H NMR (CDCI3) δ 7.8 (bs, 1H), 7.7 (bs, 2H), 6.95 (bs, 1H), 6.6 (bs, 1H), 2.3 (s, 3H), 2.18 (s, 3H). Example 52. c/s-4-[(3.5-Bis-trifluoromethyl-benzyl)-methoxycarbonyl-amino]-7.8-dichloro-2-methyl- 3.4-dihydro-2H-quinoline-1 -carboxylic acid ethyl ester. MS m/z 587 (Mτ + H); 1H NMR (CDCI3) δ 7.8 (bs, 1 H), 7.7 (bs, 1H), 7.65 (bs, 1H), 7.35 (bd, 1H), 6.8 (bd, 1H), 3.8 (s, 3H). Example 53. c/s-4-[(3.5-Bis-trifluoromethyl-benzyπ-methoxycarbonyl-aminol-6-fluoro-2-methyl-7- trifluoromethyl-3.4-dihvdro-2H-quinoline-1 -carboxylic acid tert-butyl ester. MS m/z 633 (M+ + 1), 650 (M+ + 18); 1H NMR (CDCI3) δ 6.76 (d, 1H, J = 10.4 Hz), 3.83 (s, 3H). Example 54. c/s-4-[(3.5-Bis-trifluoromethyl-benzyh-methoxycarbonyl-amino]-6-chloro-2-methyl-7- trifluoromethyl-3.4-dihvdro-2H-quinoline-1 -carboxylic acid benzyl ester. MS m/z 683 (M+ + 1), 700 (M+ + 18); H NMR (CDCI3) δ 7.02 (C8, s, 1 H), 3.83 (OMe aliph., s, 3H). Example 55. c/s-4-[(3.5-Bis-trifluoromethyl-benzylVmethoxycarbonyl-amino]-6-fluoro-2-methyl-7- trifluoromethyl-3.4-dihvdro-2H-quinoline-1 -carboxylic acid isopropyl ester. MS m/z 636 (M+ + 18); 1H NMR (CDCI3) δ 6.77 (d, 1H, J = 9.90 Hz), 3.83 (s, 3H). Example 56. c/s-4-[(3.5-Bis-trifluoromethyl-benzyl)-methoxycarbonyl-amino]-2-methyl-6- trifluoromethyl-3.4-dihvdro-2H-quinoline-1 -carboxyiic acid benzyl ester. 1H NMR (CDCI3) δ 1.2 (d, 2H), 3.8 (s, 3H), 5.2 (d, 1H), 5.3 (d, 1 H), 7.1 (s, 1H), 7.4 (s, 5H). Example 57. c/s-4-[(3.5-Bis-trifluoromethyl-benzyl)-methoxycarbonyl-amino]-7.8-dichloro-2-methyl- 3.4-dihvdro-2H-quinoline-1 -carboxyiic acid t-butyl ester. MS m/z 615 (M+ + H); 1H NMR (CDCI3) δ 7.8 (bs, 1 H), 7.65 (bs, 2H), 7.3 (bd, 1 H), 6.8 (bd, 1 H), 3.8 (s, 3H). Example 58. ~ c/s-4-[(3.5-Bis-trifluoromethyl-benzvπ-methoxycarbonyl-amino]-6-chloro-2-methyl-3.4- dihydro-2H-quinoline-1 -carboxyiic acid isopropyl ester. MS m/z 567 (M+ + H); 1H NMR (CDCI3) δ 7.8 (bs, 1H), 7.7 (bs, 1H), 7.65 (bs, 1H), 7.45 (m, 1H), 7.2 (m, 1H), 6.5 (d, 1H), 3.8 (s, 3H), 3.82 (s, 3H). Example 59. c/s-4-[(3.5-Bis-trifluoromethyl-benzyl)-methoxycarbonyi-amino1-2-methyl-7- trifluoromethyl-3.4-dihydro-2H-quinoline-1 -carboxylic acid benzyl ester. 1H NMR (CDCI3) δ 1.20 (d, 3H), 3.81 s, 3H), 7.04 (d, 1H), 7.3-7.4 (m, 6H), 7.6-7.8 (m, 4H). Example 60. c/s^-[(3.5-Bis-trifluoromethyl-benzyl)-methoxycarbonyl-amino]-7-methoxy-2-methyl- 3.4-dihydro-2H-quinoline-1.6-dicarboxylic acid 1 -ethyl ester. A mixture of c/s-4-[(3,5- bis-trifluoromethyl-benzyl)-methoxycarbonyi-amino]-7-methoxy-2-methyl-3,4-dihydro- 2H-quinoline-1 ,6-dicarboxylic acid 1 -ethyl ester 6-methyl ester (Example 14) (500mg, 0.82 mmol) and 0.41 ml 2N NaOH in 7.5 ml THF was heated to 70°C. After 1.5 hours, the cooled mixture was evaporated to dryness and the sodium salt was isolated by tituration with diisopropyi ether. The salt was suspended in 10 ml water, acidified with 0.1N HCI and extracted 3 x 25 ml ethyl acetate. The combined extracts were dried (MgS04), filtered and concentrated to provide the title compound (325 mg). MS m/z 593 (M+ + 1); 1H NMR (CDCI3) δ 3.62 (s, 3H), 6.76 (s, 1 H). Example 61. c/s-4-[(3.5-Bis-trifluoromethyl-benzvπ-methoxycarbonyl-amino]-6-methoxy-2-methyl- 3.4-dihydro-2H-quinoline-1 , 7-dicarboxylic acid 1 -ethyl ester. Prepared from c/s-4- [(3,5-Bis-trifluoromethyl-benzyl)-methoxycarbonyl-amino]-6-methoxy-2-methyl-3,4- dihydro-2H-quinoline-1 , 7-dicarboxylic acid 1 -ethyl ester 7-methyl ester (Example 13) in a manner analogous to Example 60. MS m/z 593 (Mτ ); H NMR (CDCI3) δ 3.80 (s, 3H), 6.42 (s, 1H). Example 62. c/s-4-[(3.5-Bis-trifluoromethyl-benzyl)-methoxycarbonyl-amino]-6-(2-dimethylamino- ethylcarbamoyl)-7-methoxy-2-methyl-3.4-dihydro-2H-quinoline-1 -carboxyiic acid ethyl ester. A mixture of c/s-4-[(3,5-bis-trifluoromethyl-benzyl)-methoxycarbonyl-amino]-7- methoxy-2-methyl-3,4-dihydro-2H-quinoline-1 ,6-dicarboxylic acid 1 -ethyl ester (Example 60) (75 mg, 1.26 mmol) and 1.5 ml SOCI2 was heated at reflux for 1.5 h7 The cooled solution was evaporated to dryness and the residue dried under high vacuum for 30 min. The residue was diluted with 0.75 ml dichloromethane, and triethylamine (0.021 ml) and N,N-dimethyl ethyienediamine (11 mg, 1.26 mmol) were added sequentially. After 1.5 h, the reaction mixture was concentrated and the residue was chromatographed (5% MeOH-dichloromethane) to afford the desired product (72 mg (86%). MS m/z 663 (M+ + 1 ); 1H NMR (CDCI3) δ 3.75 (s, 3H), 7.22 (s, 1 H).
Examples 63-69 were prepared in an analogous manner to Example 62 substituting with the appropriate amine from either Example 60 or Example 61. Example 63. c/s-4-[(3.5-Bis-trifluoromethyl-benzylVmethoxycarbonyl-amino]-6-[2-(3H-imidazol-4- vπ-ethvicarbamoyl]-7-methoxy-2-methyl-3.4-dihydro-2H-quinoline-1-carboxylic acid ethyl ester. MS m z 686 (M+ + 1 ); 1H NMR (CDCI3) δ 3.74 (s, 3H), 7.18 (s, 1H). Example 64. c/s-4-[(3.5-Bis-trifluoromethyl-benzyl)-methoxycarbonyl-amino]-7-carbamoyl-6- methoxy-2-methyl-3.4-dihvdro-2H-quinoiine-1 -carboxylic acid ethyl ester. MS m/z 593 (M+ + 1 ); 1H NMR (CDCI3) δ 3.81 (s, 3H), 6.42 (s, 1 H). Example 65. c/s- -[(3.5-Bis-trifluoromethyl-benzvπ-methoxycarbonyl-amino]-6-methoxy-2-methyl- 7-methylcarbamoyl-3.4-dihydro-2H-quinoline-1 -carboxylic acid ethyl ester. MS m/z 606 (M+ + 1); 1H NMR (CDCI3) δ 3.80 (s, 3H), 6.39 (s, 1 H). Example 66. c/s-4-[(3.5-Bis-trifluoromethyl-benzvπ-methoxycarbonyl-amino]-7-[2-(1 H-imidazol-4- vπ-ethylcarbamoyl]-6-methoxy-2-methyl-3.4-dihydro-2H-quinoline-1 -carboxyiic acid ethyl ester. MS m/z 686 (M+ + 1 ); 1H NMR (CDCI3) δ 3.74 (s, 3H), 6.38 (s, 1 H). Example 67. c/s^-ffS.S-Bis-trifluoromethyl-benzyπ-methoxycarbonyl-aminol^-methoxy^-methyl- 6-methylcarbamoyl-3.4-dihydro-2H-quinoline-1 -carboxyiic acid ethyl ester. MS m/z 606 (M+ + 1 ); 1H NMR (CDCI3) δ 3.90 (s, 3H), 7.18 (s, 1 H). Example 68. c/s^-[(3.5-Bis-trifluoromethyl-benzyh-methoxycarbonyl-amino]-7-(2-dimethylamino- ethylcarbamoyπ-6-methoxy-2-methyl-3.4-dihydro-2H-quinoline-1 -carboxylic acid ethyl ester. MS m/z 663 (M+ + 1 ); 1H NMR (CDCI3) δ 3.78 (s, 3H), 6.39 (s, 1 H). Example 69. c/s-4-[(3.5-Bis-trifluoromethyl-benzyh-methoxycarbonyl-amino]-6-methoxy-2-methyl- 7-(2-moφholin-4-yl-ethvicart>amoylV3.4-dihydro-2H-quinoline- -carboxylic acid ethyl ester. MS m/z 705 (M+ + 1 ); 1H NMR (CDCI3) δ 3.81 (s, 3H), 6.39 (s, 1 H). Example 70. c/s-(3.5-Bis-trifluoromethyl-benzyπ-(2-methyl-6-trifluoromethyl-1.2.3.4-tetrahvdro- quinolin-4-ylVcarbamic acid methyl ester. c/s-4-[(3,5-Bis-trifluoromethyl-benzyl)- methoxycarbonyl-amino]-2-methyl-6-trifluoromethyl-3,4-dihydro-2H-quinoline-1 - carboxylic acid benzyl ester. (Example 56) (2.8 g, 4.3 mmol), ethanol (50 mL), and 10% palladium on carbon (280 mg) were combined in a Parr bottle and agitated under 50 psi of hydrogen gas on a Parr shaker for 0.5 h. The mixture was then filtered through a bed of Celite®, eluting with ethyl acetate, and the filtrate concentrated in vacuo . The residue was purified by silica gel chromatography using 5% ethyl acetate/hexanes as eiuent to afford 2.0 g of the title product (90%): MS m/z 516 (M+ + 2); 1H NMR (CDCI3) δ 1.18 (C2-Me, d, 3H, J = 5.8 Hz), 6.50 (C8, d, 1H, J = 8.3 Hz), 7.21 (C7, d, 1 H, J = 8.5).
Examples 71 and 72 were prepared from the indicated starting compounds in an analogous manner to Example 70. Example 71. c/s-(3.5-Bis-trifluoromethvi-benzvπ- (6-chloro-7-trifluoromethyl -2-methyt-1.2.3.4- tetrahydro-quinolin-4-ylVcarbamic acid methyl ester. Prepared from c/s-4-[(3,5-bis- trifluoromethyl-benzyl)-methoxycarbonyl-amino]-6-chloro-2-methyl-7-trifluoromethyl- 3,4-dihydro-2H-quinoline-1 -carboxylic acid benzyl ester. (Example 54) MS m/z 549 (M+ + 1); 1H NMR (CDCI3) δ 1.17 (C2-Me, d, 3H, J = 6 Hz), 6.79 (s, 1H), 6.85-6.95 (m, 1 H), 7.55-7.65 (m, 2H), 7.75 (s, 1H). Example 72. c/s-(3.5-Bis-trifluoromethyl-benzylV (7-trifluoromethyl -2-methyl-1.2.3.4-tetrahydro- quinolin-4-y0-carbamic acid methyl ester. Prepared from c/s-4-[(3,5-bis- trifluoromethyl-benzyl)-methoxycarbonyi-amino]-2-methyl-7-trifluoromethyl-3,4- dihydro-2H-quinoiine-1-carboxylic acid benzyl ester. (Example 59). MS m/z 515 (M + 1); 1H NMR (CDCI3) δ 1.17 (C2-Me, d, 3H), 6.7 (s, 1 H), 7.75 (s, 1 H). Example 73. c/s-4-[(3.5-Bis-trifluoromethyl-benzyh-methoxycarbonyl-amino]-2-methyl-6- trifluoromethyl-3.4-dihydro-2H-quinoline-1 -carboxylic acid isopropyl ester. To a solution of c/s-(3,5-bis-trifluoromethyl-benzyl)-(2-methyl-6-trifluoromethyl-1 ,2,3,4- tetrahydro-quinoiin-4-yl)-carbamic acid methyl ester (Example 70) (95 mg, 0.185 mmol) and anhydrous pyridine (0.5 mL) in anhydrous dichloromethane (2 mL) was added isopropyl chloroformate (1.9 mL of a 1M solution in toluene, 1.85 mmol). After stirring at room temperature overnight, water (5 mL) and an aqueous 10% KOH solution (5 mL) were added, and the mixture was extracted with ethyl acetate (3 x 10 mL). The combined organic phases were then washed with 1 N HCI (3 x 10 mL), then a saturated sodium bicarbonate solution, and then brine (10 mL of each). The organic layer was dried over sodium sulfate, filtered and concentrated in vacuo to give 117 mg crude product. Purification by silica gel chromatography using 0-10% ethyl acetate/hexanes as eiuent afforded 90 mg of the title compound (81%): MS m/z 602 (M+ + 2), 619 (M+ + 19); 1H NMR (CDCI3) δ 1.20 (Me, d, 3H, J = 6.1 Hz), 1.28 (d, 3H), 1.31 (d, 3H), 3.83 (OMe, s, 3H), 5.04 (septet, 1 H), 7.12 (C5, s, 1H).
Examples 74-84 were prepared in an analogous manner to Example 73 from the appropriate amine from Examples 70-72. Example 74. c/s-4-[(3.5-Bis-trifluoromethyl-benzyπ-methoxycarbonyl-amino]-2-methyl-6- trifluoromethyl-3.4-dihydro-2H-αuinoline-1 -carboxyiic acid tert-butyl ester. MS m/z 615 (M+ + 1 ), 632 (M+ + 18); 1H NMR (CDCI3) δ 7.10 (C5, s, 1H), 3.81 (OMe, s, 3H). Example 75. c/s-4-[(3.5-Bis-trifluoromethyl-benzylVmethoxycarbonyl-amino]-2-methyl-6- trifluoromethyl-3.4-dihvdro-2H-quinoline-1 -carboxylic acid propyl ester. MS m/z 619 (M+ + 19); 1H NMR (CDCI3) δ 7.12 (C5, s, 1 H), 3.82 (OMe, s, 3H), 1.21 (C2-Me, d,
3H, J = 6.1 Hz).
Example 76. c/s- -[(3.5-Bis-trifluoromethyl-benzy -methoxycarbonyl-amiπo]-2-methyl-6- trifluoromethyl-3.4-dihydro-2H-quinoline-1 -carboxyiic acid isobutyl ester. MS m/z
633 (M+ + 19); 1H NMR (CDCI3) δ 7.12 (C8, s, 1H), 3.82 (OMe, s, 3H), 1.21 (C2-Me, d, 3H, J = 5.8 Hz).
Example 77. cis- 4-[(3.5-Bis-trifluoromethyl-benzyi)-methoxycarbonyl-amino]-2-methyl-6- trifluoromethyl-3.4-dihydro-2H-quinoline-1 -carboxyiic acid cyclopentvi ester. 1H NMR
(CDCI3) δ 7.12 (C5 arom., s, 1H), 3.83 (OMe, s, 3H), 1.24 (C2-Me, d, 3H, J = 15.5
Hz).
Example 78. c/s-4-[(3.5-Bis-trifluoromethyl-benzyh-methoxycarbonyl-amino]-2-methyl-7- trifluoromethyl-3.4-dihydro-2H-quinoline-1 -carboxyiic acid isopropyl ester. MS m/z
601 (M+ + 1), 618 (M+ + 18); 1H NMR (CDCI3) δ 7.04 (C5 arom., d, 1H, J = 8.0 Hz),
3.82 (OMe, s, 3H).
Example 79. c/s-4-[(3.5-Bis-trifluoromethyl-benzy -methoxycarbonyl-amino]-2-methyl-7- trifluoromethyl-3.4-dihydro-2H-quinoline-1 -carboxyiic acid propyl ester. MS m/z 601
(M+ + 1 ), 618 (M+ + 18); 1H NMR (CDCI3) δ 7.04 (C5 arom., d, 1H). 3.82 (OMe, s,
3H).
Example 80. c/'s- -[(3.5-Bis-trifluoromethyl-benzyπ-methoxycarbonyl-amino]-2-methyl-7- trifluoromethyl-3.4-dihydro-2H-quinoline-1 -carboxyiic acid tert-butyl ester. MS m/z
615 (M+ + 1 ), 632 (M+ + 18) 1H NMR (CDCI3) δ 7.01 (C5 arom., d, 1H), 3.82 (OMe aliph., s, 3H).
Example 81. c/s-4-[(3.5-Bis-trifluoromethyl-benzvπ-methoxycarbonyl-amino]-6-chloro-2-methyl-7- trifluoromethyl-3.4-dihydro-2H-quinoline-1 -carboxyiic acid isopropyl ester. MS m/z
636 (M+ + 2), 653 (M+ + 19); 1H NMR (CDCI3) δ 7.02 (C8 arom., s, 1 H), 3.84 (OMe aliph., s, 3H). Example 82. c/s-4-[(3.5-Bis-trifluoromethyl-benzyh-methoxycarbonyl-amino]-6-chloro-2-methyl-7- trifluoromethyl-3.4-dihydro-2H-quinoline-1 -carboxyiic acid tert-butyl ester H NMR (CDCI3) δ 1.18 (d,3H), 1.50 (s, 9H), 3.84 (OMe aliph., s, 3H). 7.0 (s, 1H). Example 83. c/s-(3.5-Bis-trifluoromethyl-benzvπ-(1-isopropylcarbamoyπ-2-methyl-6-trifluoromethyl- 1.2.3.4-tetrahvdro-αuinolin-4-yπ-carbamic acid methyl ester. MS m/z 600 (M+ + H); 1H NMR (CDCI3) δ 7.82 (bs, 1H), 7.8 (bs, 1H), 7.7 (bs, 1 H), 7.2 (bs, 1H), 6.7 (bs, 1H), 3.8 (s, 3H), 3.75 (bs, 3H). Example 84. cs-4-[(3.5-Bis-trifluoromethyl-benzyl)-methoxycarbonyl-aminol-2-methyl-6- trifluoromethyl-3.4-dihvdro-2H-quinoiine-1 -carboxyiic acid 2.2-dimethyl-propyi ester. MS m/z 628 (M+), 646 (M+ + 18); 1H NMR (CDCI3) δ 7.12 (C5, s, 1H), 3.83 (OMe, s, 3H), 1.23 (Me, d, 3H, J = 6.1 Hz). Example 85. c/s-4-[(3.5-Bis-trifluoromethyl-benzvi)-methoxycarbonyl-amino1-7-hydroxymethyl-6- methoxy-2-methyl-3.4-dihydro-2H-quinoline-1 -carboxyiic acid ethyl ester. To a solution of c/s-4-[(3,5-bis-trifluoromethyl-benzyl)-methoxycarbonyl-amino]-6-methoxy- 2-methyl-3,4-dihydro-2H-quinoline-1, 7-dicarboxylic acid 1 -ethyl ester 7-methyl ester (Example 13) (100mg; 0.16 mmol) in 20 mL tetrahydrofuran was added sodium borohydride (62 mg; 1.6 mmol). The reaction was heated to reflux and methanol (40 mL) was added slowly. After an additional 30 min at reflux the reaction was concentrated and the residue was diluted with 20 ml H20 and extracted 3 x 50 mL of ethyl acetate. The combined extracts were dried (MgS04), filtered, concentrated, and the residue was purified by chromatography (30-40% ethyl acetate:hexane) to afford 100 mg of the title product. 1H NMR (CDCI3) δ 1.1 (d, 3H), 1.3 (t, 3H), 3.75 (s, 3H), 6.4 (s, 1H), 7.4 (s, 1 H), 7.7 (s, 2H), 7.8 (s, 1H). -Example 86. c/s^-[(3.5-Bis-trifluoromethyl-benzyπ-methoxycarbonyl-amino]-7-(1-hvdroxymethyl- cvclopentyl)-6-methoxy-2-methyl-3.4-dihydro-2H-quinoline-1 -carboxyiic acid ethyl ester. To a solution of c/s-4-[(3,5-bis-trifluoromethyl-benzyl)-methoxycarbonyl- amino]-6-methoxy-7-(1-methoxycarbonyi-cyclopentyl)-2-methyl-3,4-dihydro-2H- quinoline-1 -carboxylic acid ethyl ester (Example 27) (700mg; 1.04mmol) in 20 mL anhydrous tetrahydrofuran was added borane-dimethylsulfide complex (1M THF; 2.28 mL) and the reaction was heated to reflux for 3 hours. The reaction was quenched with H20 and extracted with 50 mL of ethyl acetate. The organic phase was dried (MgS04), filtered and concentrated. The residue was chromatographed (25% ethyl acetate-hexane) to afford the title product (20mg). MS m/z 664 (M+ + 18); 1H NMR (CDCI3) δ 1.1 (d, 3H), 3.6 (s, 2H), 3.7 (s, 3H), 3.8 (s, 3H), 6.3 (S, 1H), 7.3 (S, 1 H), 7.7 (S, 2H), 7.8 (S, 1H).
Examples 87-89 were prepared in an analogous manner to Example 86 from the indicated starting esters. Example 87. c/s-4-[(3.5-Bis-trifluoromethyl-benzyπ-methoxycarbonyl-amιno]-7-(2-hydroxy-ethvπ-6- methoxy-2-methyl-3.4-dihydro-2H-quinoline-1 -carboxyiic acid ethyl ester. Prepared from c/s-4-[(3,5-bis-trifluoromethyl-benzyl)-methoxycarbonyl-amιno]-7- ethoxycarbonylmethyl-6-methoxy-2-methyl-3,4-dihydro-2H-quιnolιne-1 -carboxylic acid ethyl ester (Example 15) MS m/z 592.2 (M+); 1H NMR (CDCI3) δ 1.1 (d, 3H), 1.3 (t, 3H), 6.4 (S, 1H), 7.4 (S, 1H), 7.7 (S, 2H), 7.8 (S, 1 H). Example 88. c/s-4-[(3.5-Bis-trifluoromethyl-benzyπ-methoxycarbonyl-amιno]-6-hvdroxymethyl-7- methoxy-2-methyl-3.4-dihydro-2H-quinoline-1 -carboxyiic acid ethyl ester. Prepared from c/'s-4-[(3,5-bis-trifluoromethyl-benzyl)-methoxycarbonyl-amιno]-7-methoxy-2- methyl-3,4-dihydro-2H-quinoline-1 ,6-dicarboxylιc acid 1 -ethyl ester 6-methyl ester (Example 14) MS m/z 578 (M+ + 1); 1H NMR (CDCI3) δ 6.73 (s, 1 H), 3.8 (s, 3H). Example 89. c/s-4-[(3.5-Bis-trifluoromethyl-benzy -methoxycarbonyl-amιno]-6.7-bis- hydroxymethyl-2-methyl-3.4-dihydro-2H-quinoline-1 -carboxylic acid ethyl ester. Prepared from c/s-4-[(3,5-bis-trifluoromethyl-benzyl)-methoxycarbonyl-amino]-2- methyl-3,4-dihydro-2H-quinoiine-1,6,7-tricarboxylic acid 1 -ethyl ester 6,7-dimethyl ester (Example 16) MS m/z 596 (M+ + NH4 +); 1H NMR (CDCI3) δ 6.95 (s, 1 H), 3.8 (s, 3H).
Example 90. c/s-4-[(3.5-Bis-trifluoromethyl-benzyl)-methoxycarbonvi-amino]-7-bromomethyl-6- methoxy-2-methyi-3.4-dihydro-2H-quinoline-1 -carboxylic acid ethyl ester. To a solution of triphenylphosphine (0.457 g, 1.74 mmol) in anhydrous dichloromethane (15 mL) at 0 °C was slowly added bromine (84 μL, 1.6 mmoi). After the reaction was stirred at 0 °C for 20 min, a solution of c/s-4-[(3,5-bis-trifluoromethyl-benzyl)- methoxycarbonyl-amino]-7-hydroxymethyl-6-methoxy-2-methyl-3,4-dihydro-2H- quinoline-1 -carboxylic acid ethyl ester (Example 85) (0.630 g, 1.09 mmol) in dichloromethane (15ml) was added. The reaction was stirred at 0 °C for 20 min, then at room temperature for 3 h. The reaction mixture was then diluted with chloroform and washed with brine. The organic layer was dried over magnesium sulfate, filtered and concentrated in vacuo. Purification by silica gel chromatography using 10-15% ethyl acetate/hexanes as eiuent afforded the title product (0.500 g, 72%): MS m/z 642.1 (M+ +1); 1H NMR (CDCI3) δ 1.15 (d, 3H), 1.3 (t, 3H), 3.8 (s, 3H), 3.85 (s, 3H), 6.4 (s, 1H), 7.4 (s, 1 H), 7.7 br, 2H), 7.8 (s, 1 H). Example 91. c/s^-^S.δ-Bis-trifluoromethyl-benzyll-methoxycarbonyl-aminol-e-bromomethyl^- methoxy-2-methyl-3.4-dihydro-2H-quinoline-1 -carboxylic acid ethyl ester. Prepared as in Example 90 from c/s-4-[(3,5-bis-trifluoromethyl-benzyl)-methoxycarbonyl- amino]-6-hydroxymethyl-7-methoxy-2-methyl-3,4-dihydro-2H-quinoline-1 -carboxylic acid ethyl ester (Example 88). MS m/z 642 (M+ + 1 ); 1H NMR (CDCI3) δ 6.8 (s, 1 H), 3.8 (s, 3H). Example 92. c/s-4-[(3.5-Bis-trifluoromethyl-benzyl)-methoxycarbonyl-amino]-6-methoxy-2-methyl- 7-methylsulfanylmethyl-3.4-dihvdro-2H-quinoline-1 -carboxylic acid ethyl ester. To a suspension of sodium thiomethoxide (11 mg, 0.156 mmol) in anhydrous N,N- dimethylformamide (5 mL) was added c/'s-4-[(3,5-bis-trifluoromethyl-benzyi)- methoxycarbonyl-amino]-7-bromomethyl-6-methoxy-2-methyl-3,4-dihydro-2H- quinoline-1 -carboxylic acid ethyl ester (Example 90) (100 mg, 0.156 mmol), and the reaction was stirred for 6 h. The reaction mixture was then poured into water and extracted with ethyl acetate (2 x 20 mL). The combined organic extracts were dried over magnesium sulfate, filtered and concentrated in vacuo . The excess N,N- dimethylformamide was azeotroped off with heptanes. Purification by silica gel chromatography using 10-15% ethyl acetate/hexanes as eiuent afforded the title product (0.040 g, 42%); 1H NMR (CDCI3) δ 1.1 (d, 3H), 2.1 (s, 3H), 3.8 (m, 6H), 6.4 (s, 1H), 7.3 (s, 1H), 7.1 (s, 2H), 7.8 (s, 1 H).
Examples 93-102 were prepared in an analogous manner to Example 92 employing the requisite bromide (Example 90 or 91) and the appropriate alkoxide or thioalkoxide. -
Example 93. c/s-4-[(3.5-Bis-trifluoromethyl-benzyπ-methoxycarbonyl-amino]-7-ethyisulfanylmethyl- 6-methoxy-2-methyl-3.4-dihydro-2H-quinoline-1 -carboxyiic acid ethyl ester. 1H NMR (CDCI3) δ 1.1 (d, 3H), 2.55 (q, 2H), 3.7 (s, 3H), 6.4 (s, 1 H), 7.3 (s, 1 H), 7.7 (d, 2H), 7.8 (s, 1 H). Example 94. c/s-4-[(3.5-Bis-trifiuoromethyl-benzyl)-methoxycarbonyl-amino]-7-(5-ethoxycarbonyl- 4-methyl-thiazol-2-ylsulfanylmethyl)-6-methoxy-2-methyl-3.4-dihydro-2H-quinoline-1- carboxylic acid ethyl ester. MS m/z 764.2 (M+ ); 1H NMR (CDCI3) δ 1.1 (d, 3H), 1.3 (t, 3H), 2.7 (s, 3H), 6.4 (s, 1 H), 7.4 (s, 1 H), 7.7 (s, 2H), 7.8 (s, 1 H). Example 95. c/s^-fO.S-Bis-trifluoromethyl-benzyh-methoxycarbonyl-aminoj^-p-OH-imidazoM- yπ-ethoxymethyl]-6-methoxy-2-methyl-3.4-dihydro-2H-quinoline-1-carboxylic acid ethyl ester. MS m/z 673.2 (M+ + 1); 1H NMR (CDCI3) δ 1.15 (d, 3H), 1.25 (t, 3H), 2.1 (t, 1 H), 3.8 (s, 3H), 3.85 (s, 3H), 6.4 (s, 1H), 6.8 (s, 1 H), 7.2 (s, 1H), 7.5 (s, 1H), 1.1 (s, 2H), 7.8 (s, 1 H). Example 96. c/s-4-[(3.5-Bis-trifluoromethyl-benzyπ-methoxycarbonyl-amino]-7-(5-ethoxycarbonyl- thiazol-2-ylsulfanylmethvπ-6-methoxy-2-methyl-3.4-dihydro-2H-quinoline-1-carboxylic acid ethyl ester. MS nVz 749.9 (M+ ); 1H NMR (CDCI3) δ 1.1 (d, 3H), 1.4 (t, 3H), 3.8 (s, 6H), 4.4 (q, 2H), 6.4 (s, 1 H), 7.4 (s, 1 H), 7.7 (s, 2H), 7.8 (s, 1H), 8.0 (s, 1 H). Example 97. c/'s-4-[(3.5-Bis-trifluoromethyl-benzylVmethoxycarbonyl-amino]-6-ethylsulfanylmethyl- 7-methoxy-2-methyl-3.4-dihydro-2H-quinoline-1 -carboxylic acid ethyl ester. MS m/z 623 (M+ + 1 ); 1H NMR (CDCI3) δ 6.68 (s, 1 H), 3.76 (s, 3H). Example 98. c/'s-4-[(3.5-Bis-trifluoromethyl-benzyl)-methoxycarbonyl-amino]-7-methoxy-2-methyl-
6-methylsulfanylmethyl-3.4-dihydro-2H-quinoline-1 -carboxylic acid ethyl ester. MS m/z 609 (M+ + 1); 1H NMR (CDCI3) δ 6.66 (s, 1 H), 3.76 (s, 3H).
Example 99. c/s-4-[(3.5-Bis-trifluoromethyl-benzyl)-methoxycarbonyl-amino]-7-methoxy-6-(2- methoxy-ethoxymethvπ-2-methyl-3.4-dihydro-2H-quinoline-1 -carboxylic acid ethyl ester. MS m/z 637 (M+ + 1 ); 1H NMR (CDCI3) δ 6.82 (s, 1 H), 3.78 (s, 3H).
Example 100. c/s-4-[(3.5-Bis-trifluoromethyl-benzyl)-methoxycarbonyl-amino]-7-methoxy-2-methyl- 6-(1-methyl-1H-tetrazol-5-ylsulfanylmethyl)-3.4-dihydro-2H-quinoline-1 -carboxylic acid ethyl ester. MS m/z 694 (M+ + NH4+); 1H NMR (CDCI3) δ 3.76 (s, 3H), 6.99 (s,
1 H).
Example 101. c/s-4-[(3.5-Bis-trifluoromethyl-benzyπ-methoxycarbonyl-amino]-6-methoxy-7-(2- methoxy-ethoxymethyl)-2-methyl-3.4-dihydro-2H-quinoline-1 -carboxylic acid ethyl ester. MS m z 561 (M+ -75); 1H NMR (CDCI3) δ 1.1 (d, 3H), 1.3 (t, 3H), 3.6 (s, 3H),
6.2 (s, 1 H), 7.8 (s, 1H).
Example 102. c/s-4-[(3.5-Bis-trifluoromethyl-benzyl -methoxycarbonyl-amino]-7- propylsulfanylmethvi-6-methoxy-2-methyl-3.4-dihydro-2H-quinoiine-1-carboxylic acid ethyl ester. 1H NMR (CDCI3) δ 1.0 (t, 3H), 1.1 , (d, 3H), 1.3 (t, 3H), 2.5 (t, 2H), 3.7 (s,
3H), 6.4 (s, 1 H), 7.3 (s, 1 H), 7.7 (m, 2H), 7.8 (s, 1H).
Example 103. c/s-4-[(3.5-Bis-trifluoromethyl-benzyπ-methoxycarbonyl-amino]-6-methoxy-7- methoxymethyl-2-methyl-3.4-dihvdro-2H-quinoline-1 -carboxyiic acid ethyl ester. To a suspension of sodium hydride (4.2 mg, 60% dispersion in mineral oil, 0.10 mmol) in
N,N-dimethylforτnamide (2.5 mL) was added c/s-4-[(3,5-bis-trifluoromethyl-benzyl)- methoxycarbonyl-amino]-7-hydroxymethyl-6-methoxy-2-methyl-3,4-dihydro-2H- quinoline-1 -carboxylic acid ethyl ester (Example 85) (50 mg, 0.087 mmol). After the reaction was stirred at room temperature for 30 min, iodomethane (60 μL, 0.96 mmol) was added, and the reaction was stirred for 18 h. The reaction mixture was then poured into water and extracted with ethyl acetate (2 x 20 mL). The combined organic extracts were dried over magnesium sulfate, filtered and concentrated in vacuo. The excess N,N-dimethylformamide was azeotroped off with heptanes. Purification by silica gel chromatography using 15-25% ethyi acetate/hexanes as eiuent afforded the title product (30 mg, 59%): 1H NMR (CDCI3) δ 1.1 (d, 3H), 1.3 (t, 3H), 3.4 (s, 3H), 3.75 (s, 3H), 3.8 (s, 3H), 6.4 (s, 1 H), 7.4 (s, 1 H), 7.75 (s, 2H), 7.8 (s, 1 H).
Examples 104 and 105 were prepared in analogous manner to Example 103 from the indicated starting alcohol. Example 104. cs-4-[(3.5-Bis-trifluoromethyl-benzyl)-methoxycarbonyl-amino]-7-methoxy-6- methoxymethyl-2-methyl-3.4-dihydro-2H-quinoline-1 -carboxyiic acid ethyl ester. Prepared from c/s-4-[(3,5-bis-trifluoromethyl-benzyl)-methoxycarbonyl-amino]-6- hydroxymethyi-7-methoxy-2-methyl-3,4-dihydro-2H-quinoline-1 -carboxylic acid ethyl ester (Example 88) MS m/z 610 (M+ + NH4 +); 1H NMR (CDCI3) δ 6.8 (s, 1 H), 3.76 (s, 3H). Example 105. c/s-4-[(3.5-Bis-trifluoromethyl-benzylVmethoxycarbonyl-amino]-6.7-bis- methoxymethyl-2-methyl-3.4-dihvdro-2H-quinoline-1 -carboxyiic acid ethyl ester. Prepared from c/s^-[(3,5-bis-trifluoromethyl-benzyl)-methoxycarbonyl-amino]-6J-bis- hydroxymethyl-2-methyl-3,4-dihydro-2H-quinoline-1 -carboxylic acid ethyl ester (Example 89) MS m/z 607 (M+ + 1); 1H NMR (CDCI3) δ 6.95 (s, 1 H), 3.82 (s, 3H). Example 106. c/s-4-[(3.5-Bis-trifluoromethyl-benzylVmethoxycarbonyl-amino]-6.7-bis-fluoromethyl- 2-methyl-3.4-dihydro-2H-quinoline-1 -carboxylic acid ethyl ester. To a solution of c/s- 4-[(3,5-bis-trifluoromethyl-benzyl)-methoxycarbonyl-amino]-6J-bis-hydroxymethyl-2- methyl-3,4-dihydro-2H-quinoline-1 -carboxylic acid ethyi ester (Example 89) in 15 mL 1 ,2-dichloroethane was added diethyiaminosulfur trifluoride (0.5 mL) and the reaction was heated to 80°C for 45 min. The reaction mixture was cooled, adsorbed onto Si02 and chromatographed (20% ethyl acetate-hexane) to afford the title product (60mg). MS m/z 582.3 (M+); 1H NMR (CDCI3) δ 1.1 (d, 3H), 3.8 (s, 3H), 5.35 (s, 2H), 5.5 (s, 2H), 6.9 (s, 1 H), 7.65 (m, 3 H), 7.8 (s, 1 H). Example 107. c/s~4-[(3.5-Bis-trifluoromethyl-benzyπ-methoxycarbonyl-amino]-6-chloro-2-methyl-8- nitro-3.4-dihvdro-2H-quinoline-1 -carboxylic acid ethyl ester. To a pre-dried flask under nitrogen atmosphere was prepared a slurry of F3CS03N02 (2.6 mmol) in 9 mL of dichloromethane [ref. J. Org. Chem. 1973, 38 (25), 4243]. The solution was cooled to -78 °C and a solution of c/s-4-[(3,5-bis-trifluoromethyl-benzyl)-methoxycarbonyl- amino]-6-chloro-2-methyl-3,4-dihydro-2H-quinoline-1 -carboxylic acid ethyl ester (Example 32) (0.72 g, 1.3 mmol) in anhydrous dichloromethane (5 mL) was added. The reaction was stirred at -78 °C for 1.5 h, then warmed to 0 °C and stirred for 1 h and 40 min. Water (30 mL) was then added, and the mixture was extracted three times with dichloromethane. The combined organic layers were dried over magnesium sulfate, filtered and concentrated in vacuo to give the title product (0.75 g, 96%). MS m/z 598 (M+ + H); 1H NMR (CDCI3) δ 7.85 (bs, 1H), 7.8 (bs, 1 H), 7.7 (bs, 1 H), 7.65 (bs, 1H), 7.1 (bs, 1H), 3.80 (s, 3H). Example 108. c s-4-[(3.5-Bis-trifluoromethyl-benzyπ-methoxycarbonyl-amino]-6-trifluoromethyl-2- methyl-8-nitro-3.4-dihvdro-2H-quinoline-1 -carboxylic acid isopropyl ester. Prepared in an analogous manner to Example 107 from c/s-4-[(3,5-bis-trifluoromethyl-benzyl)- methoxycarbonyl-amino]-2-methyl-6-trifluoromethyl-3,4-dihydro-2H-quinoline-1- carboxylic acid isopropyl ester (Example 73). MS m/z 646 (M+ + H); 1H NMR (CDCI3) δ 8.1 (bs, 1 H), 7.8 (bs, 1 H), 7.7 (bs, 1 H), 7.65 (bs, 1 H), 7.35 (bs, 1 H), 3.82 (s, 3H). Examples 109 c/s-8-Amino-4-[(3.5-bis-trifluoromethyl-benzyπ-methoxycarbonyl-amino]-6-chloro-2- methyl-3.4-dihydro-2H-quinoline-1 -carboxylic acid ethyl ester and: c/s-8-Aminc-4- [(3.5-bis-trifluoromethyl-benzyπ-methoxycarbonyl-amiπo'|-2-methyl-3.4-dihydro-2H- quinoline-1 -carboxylic acid ethyl ester. c/s-4-[(3,5-Bis-trifluoromethyl-benzyl)- methoxycarbonyi-amino]-6-chloro-2-methyl-8-nitro-3,4-dihydro-2H-quinoline-1- carboxylic acid ethyl ester (Example 107) (0.747 g, 1.25 mmol), ethanol (20 mL), and 10% palladium on carbon (0.40 g) were combined and agitated under 36 psi of H2 on a Parr shaker for 2 h and 15 min. The mixture was then filtered through a bed of Celite®. The Celite® was washed with ethanol, and the filtrate was concentrated in vacuo. Purification by silica gel chromatography using 30-100% ethyl acetate/hexanes as eiuent afforded 0.386 g (54%) of c/s-8-amino-4-[(3,5-bis- trifluoromethyl-benzyl)-methoxycarbonyl-amino]-6-chloro-2-methyl-3,4-dihydro-2H- quinoline-1 -carboxylic acid ethyl ester MS m/z 568 (M+ + H); 1H NMR (CDCI3) δ 7.8 (bs, 1 H), 7.7 (bs, 1H), 7.65 (bs, 1H), 6.7 (bs, 1 H), 6.38 (bs, 1H), 3.78 (s, 3H), and 0.161 g (24%) of c/s-8-amino-4-[(3,5-bis-trifluoromethyl-benzyl)-methoxycarbonyl- amino]-2-methyl-3,4-dihydro-2H-quinoiine-1 -carboxylic acid ethyl ester MS m/z 534 (M+ + H); 1H NMR (CDCI3) δ 7.79 (bs, 1H), 7.7 (bs, 1 H), 7.65 (bs, 1H), 7.02 (bt, 1H), 6.72 (bs, 1H), 6.4 (bd, 1H), 3.78 (s, 3H). Example 110. cis- 8-Amino-4-[(3.5-bis-trifluoromethyl-benzvπ-methoxycarbonyl-amino] -2-methyl -6- trifluoromethyl-3.4-dihvdro-2H-quinoline-1 -carboxylic acid isopropyl ester. Prepared as in Example 109 from c/s-4-[(3,5-bis-trifiuoromethyl-benzyl)-methoxycarbonyl- amino]-6-trifluoromethyl-2-methyl-8-nitro-3,4-dihydro-2H-quinoline-1-carboxylic acid isopropyl ester (Example 108) MS m/z 616 (M+ + H); 1H NMR (CDCI3) δ 7.8 (bs, 1 H), 7.65 (bs, 2H), 7.62 (bs, 1 H), 6.8 (bs, 1 H), 3.8 (s, 3H). Example 111. c/s-8-Acetylamino-4-[(3.5-bis-trifluoromethyl-benzv -methoxycarbonyl-amino]-6- chloro-2-methyl-3.4-dihydro-2H-quinoline-1 -carboxyiic acid ethyl ester. To a pre-dried flask under nitrogen atmosphere was added c/s-8-amino-4-[(3,5-bis-trifluoromethyl- benzyl)-methoxycarbonyi-amino]-6-chloro-2-methyl-3,4-dihydro-2H-quinoline-1- carboxylic acid ethyl ester (Example 109) (30 mg, 0.053 mmol), followed by anhydrous dichloromethane (2 mL), pyridine (0.5 mL), and acetyl chloride (0.015 mL, 0.21 mmol). The reaction was stirred overnight at room temperature before ethyl acetate was added, and the mixture was extracted three times with 1N HCI, followed by brine. The organic layer was dried over sodium sulfate, filtered and concentrated in vacuo to give the title product (33 mg, 100%) MS m/z 610 (M+ + H); 1H NMR (CDCI3) δ 7.8 (bs, 2H), 7.7 (bs, 1H), 7.65 (bs, 1H), 6. 8 (bs, 1 H), 3.8 (s, 3H), 2.1 (s, 3H). Example 112. c/s^-fO.δ-Bis-trifluoromethyl-benzyπ-methoxycarbonyl-aminol-δ-chloro-δ- methoxycarbonylamino-2-methyl-3.4-dihydro-2H-quinoliπe-1 -carboxylic acid ethyl ester. To a pre-dried flask under nitrogen atmosphere was added c/s-8-amino-4- [(3,5-bis-trifluoromethyl-benzyl)-methoxycarbonyl-amino]-6-chloro-2-methyl-3,4- dihydro-2H-quinoiine-1-carboxylic acid ethyl ester (Example 109) (30 mg, 0.053 mmol), followed by anhydrous dichloromethane (2 mL), pyridine (0.5 mL), and methyl chloroformate (0.016 mL, 0.21 mmol). The reaction was stirred overnight at room temperature before ethyl acetate was added, and the mixture was extracted three times with 1 N HCI, followed by brine. The organic layer was dried over sodium sulfate, filtered and concentrated in vacuo to give the desired product (29 mg, 87%).
MS m/z 626 (M+ + H); 1H NMR (CDCI3) δ 7.82 (bs, 1 H), 7.8 (bs, 1 H), 1.1 (bs, 1H),
6.68 (bs, 1H), 6.7 (bs, 1H), 3.8 (s, 3H), 3.75 (s, 3H). The following examples were prepared in optically enriched form by resoluton of the corresponding racemate indicated, or an intermediate in its synthesis, using the methods described in the specification.
Example 113.
[2R.4S] 4-[(3.5-Bis-trifluoromethyl-benzyl)-methoxycarbonyl-amino]-2-methyl-6- trifluoromethyl-3.4-dihydro-2H-quinoline-1 -carboxylic acid ethyl ester. Example 30.
Example 114.
[2R.4S] 4-[(3.5-bis-trifluoromethyl-benzyl)-methoxycarbonyl-amino]-2-methyl-6- trifluoromethyl-3.4-dihydro-2H-quinoiine-1 -carboxylic acid isopropyl ester. Example
73. Example 115.
[2R.4S] 4-[(3.5-Bis-trifluoromethyl-benzyπ-methoxycarbonyl-amino]-2-methyi-6- trifluoromethyl-3.4-dihydro-2H-αuinoiine-1 -carboxylic acid tert-butyl ester. Example
74.
Example 116. [2R.4S] 4-[(3.5-Bis-trifluoromethyl-benzyπ-methoxycarbonyl-amino]-2-methyl-6- trifluoromethyl-3.4-dihydro-2H-quinoline-1 -carboxylic acid propyl ester. Example 75.

Claims

1. A compound of the Formula
Figure imgf000093_0001
a prodrug thereof, or a pharmaceutically acceptable salt of said compound or of said prodrug; wherein R1 is hydrogen, Y, W-X, W-Y; wherein W is a carbonyl, thiocarbonyl, sulfinyl or sulfonyl; X is -ON, -S-Y, -Ν(H)-Y or -N-(Y)2; wherein Y for each occurrence is independently Z or a fully saturated, partially unsaturated or fully unsaturated one to ten membered straight or branched carbon chain wherein the carbons, other than the connecting carbon, may optionally be replaced with one or two heteroatoms selected independently from oxygen, sulfur and nitrogen and said carbon is optionally mono-, di- or tri-substituted independently with halo, said carbon is optionally mono-substituted with hydroxy, said carbon is optionally mono-substituted with oxo, said sulfur is optionally mono- or di-substituted with oxo, said nitrogen is optionally mono-, or di-substituted with oxo, and said carbon chain is optionally mono-substituted with Z; Z is a partially saturated, fully saturated or fully unsaturated three to twelve membered ring optionally having one to four heteroatoms selected independently from oxygen, sulfur and nitrogen, or a bicyciic ring consisting of two fused partially saturated, fully saturated or fully unsaturated three to six membered rings, taken independently, optionally having one to four heteroatoms selected independently from nitrogen, sulfur and oxygen; wherein said Z substituent is optionally mono-, di- or tri-substituted independently with halo, (C2-C6)alkenyl, (C C6) alkyl, hydroxy, (CrC6)alkoxy, (C C4)alkylthio, amino, nitro, cyano, oxo, carboxy, (C1-C6)alkyloxycarbonyl, mono-N- or di-N,N-(C C6)alkylamino wherein said (C C6)alkyl substituent is optionally mono-, di- or tri-substituted independently with halo, hydroxy, (C-p C6)alkoxy, (CrC4)alkylthio, amino, nitro, cyano, oxo, carboxy, (C C6)alkyloxycarbonyl, mono-N- or di-N,N-(C C6)alkylamino, said (C C6)alkyl is also optionally substituted with from one to nine fluorines; R3 is hydrogen or Q; wherein Q is a fully saturated, partially unsaturated or fully unsaturated one to six membered straight or branched carbon chain wherein the carbons, other than the connecting carbon, may optionally be replaced with one heteroatom selected from oxygen, sulfur and nitrogen and said carbon is optionally mono-, di- or tri-substituted independently with halo, said carbon is optionally mono-substituted with hydroxy, said carbon is optionally mono- substituted with oxo, said sulfur is optionally mono- or di-substituted with oxo, said nitrogen is optionally mono- or di-substituted with oxo, and said carbon chain is optionally mono-substituted with V; wherein V is a partially saturated, fully saturated or fully unsaturated three to twelve membered ring optionally having one to four heteroatoms selected independently from oxygen, sulfur and nitrogen, or, a bicyclic ring consisting of two fused partially saturated, fully saturated or fully unsaturated three to six membered rings, taken independently, optionally having one to four heteroatoms selected independently from nitrogen, sulfur and oxygen; wherein said V substituent is optionally mono-, di-, tri-, or tetra- substituted independently with halo, (C-,-C6)alkyl, (C2-C6)alkenyl, hydroxy, (C C6)alkoxy, (C C )alkylthio, amino, nitro, cyano, oxo, carboxamoyl, mono-N- or di-N,N-(C C6) alkyicarboxamoyl, carboxy, (C C6)alkyloxycarbonyl, mono- N- or di-N,N-(CrC6)alkylamino wherein said (CrC6)alkyl or (C2-C6)alkenyl substituent is optionally mono-, di- or tri-substituted independently with hydroxy, (C C6)aikoxy, (C C4)alkylthio, amino, nitro, cyano, oxo, carboxy, (C C6)alkyloxycarbonyl, mono-N- or di-N,N-(C1-C6)alkylamino or said (C C6)alkyl or (C2-C6)alkenyl substituents are optionally substituted with from one to nine fluorines;
R4 is Q1 or V1 wherein Q1 a fully saturated, partially unsaturated or fully unsaturated one to six membered straight or branched carbon chain wherein the carbons, other than the connecting carbon, may optionally be replaced with one heteroatom selected from oxygen, sulfur and nitrogen and said carbon is optionally mono-, di- or tri-substituted independently with halo, said carbon is optionally mono-substituted with hydroxy, said carbon is optionally mono- substituted with oxo, said sulfur is optionally mono- or di-substituted with oxo, said nitrogen is optionally mono- or di-substituted with oxo, and said carbon chain is optionally mono-substituted with V1; wherein V1 is is a partially saturated, fully saturated or fully unsaturated three to six membered ring optionally having one to two heteroatoms selected independently from oxygen, sulfur and nitrogen; wherein said V1 substituent is optionally mono-, di-, tri-, or tetra- substituted independently with halo, (CrC6)alkyl, (C CgJalkoxy, amino, nitro, cyano, (C C6)alkyloxycarbonyl, mono-N- or di-N,N-(CrC6)alkylamino wherein said (C C6)alkyl substituent is optionally mono-substituted with oxo, said (Cr C6)alkyl substituent is optionally substituted with from one to nine fluorines; wherein either R3 must contain V or R4 must contain V1; and R5 , R6 , R7and R8 are each independently hydrogen, a bond, nitro or halo wherein said bond is substituted with T or a partially saturated, fully saturated or fully unsaturated (C C12) straight or branched carbon chain wherein carbon may optionally be replaced with one or two heteroatoms selected independently from oxygen, sulfur and nitrogen wherein said carbon atoms are optionally mono-, di- or tri-substituted independently with halo, said carbon is optionally mono-substituted with hydroxy, said carbon is optionally mono-substituted with oxo, said sulfur is optionally mono- or di-substituted with oxo, said nitrogen is optionally mono- or di-substituted with oxo, and said carbon is optionally mono-substituted with T; wherein T is a partially saturated, fully saturated or fully unsaturated three to twelve membered ring optionally having one to four heteroatoms selected independently from oxygen, sulfur and nitrogen, or, a bicyciic ring consisting of two fused partially saturated, fully saturated or fully unsaturated three to six membered rings, taken independently, optionally having one to four heteroatoms selected independently from nitrogen, sulfur and oxygen; wherein said T substituent is optionally mono-, di- or tri-substituted independently with halo, (C1-C6)alkyl, (C2-C6)alkenyl, hydroxy, (C C6)alkoxy, (C C4)alkylthio, amino, nitro, cyano, oxo, carboxy, (C C6)alkyloxycarbonyl, mono-N- or di-N,N-(C1-C6)alkylamino wherein said (C C6)alkyl substituent is optionally mono-, di- or tri-substituted independently with hydroxy, (C C6)alkoxy, (CrC4)alkylthio, amino, nitro, cyano, oxo, carboxy, (C C5)alkyloxycarbonyl, mono-N- or di-N,N-(C1-C6)alkylamino, said (C C6)alkyl substituent is also optionally substituted with from one to nine fluorines; provided that at least one of substituents R5, R6, R7 and R8 is not hydrogen and is not linked to the quinoiine moiety through oxy. 2. A compound as recited in claim 1 wherein the C2 methyl is beta; the C4 nitrogen is beta: R1 is W-X ; W is carbonyl, thiocarbonyl or sulfonyl; X is -0-Y-, S-Y-, N(H)-Y- or -N-(Y)2-;
Y for each occurrence is independently Z or (CrC4)alkyl, said (C C4)aikyi optionally having hydroxy or from one to nine fluorines or said (C C4)alkyl optionally mono-substituted with Z wherein Z is a partially saturated, fully saturated or fully unsaturated three to six membered ring optionally having one to two heteroatoms selected independently from oxygen, sulfur and nitrogen; wherein said Z substituent is optionally mono-, di- or tri-substituted independently with halo, (CrC4)alkyl, (C C4)alkoxy, (C C4)alkylthio, nitro, cyano, oxo, or (C C6)alkyloxycarbonyl, said (C C4)alkyl substituent is also optionally substituted with from one to nine fluorines;
R3 is Q-V wherein Q is (C C4)alkyl and V is a five or six membered partially saturated, fully saturated or fully unsaturated ring optionally having one to three heteroatoms selected independently from oxygen, sulfur and nitrogen; wherein said V ring is optionally mono-, di-, tri- or tetra-substituted independently with halo, (CrC^alkyl, hydroxy, (C C6)alkoxy, nitro, cyano or oxo wherein said (C C6)alkyl substituent is optionally substituted with from one to nine fluorines; R4 is (C C4)alkyl;
R6 and R7 are each independently hydrogen, halo, T, (C C6)alkyl or (Cr C6)alkoxy, said (C C6)alkyl or (C C6)alkoxy optionally having from one to nine fluorines or said (C C6)alkoxy or (C C6)alkyl optionally mono- substituted with T, wherein T is a partially saturated, fully saturated or fully unsaturated five to six membered ring optionally having one to two heteroatoms selected independently from oxygen, sulfur and nitrogen; wherein said T substituent is optionally mono-, di- or tri-substituted independently with halo, (C C6)alkyl, hydroxy, (C C6)alkoxy, (CrC4)alkylthio, amino, oxo, carboxy, (C-|-C6)alkyloxycarbonyl, mono-N- or di-N,N-(C C6)alkylamino wherein said (C C6)alkyl substituent optionally has from one to nine fluorines; and R5 and R8 are H; or a pharmaceutically acceptable salt thereof.
3. A compound as recited in claim 2 wherein W is carbonyl;
X is O-Y wherein Y is (C C4)alkyl, said (C C4)alkyl optionally having hydroxy or from one to nine fluorines;
Q is (C C4)alkyl and V is phenyl, pyridinyl, or pyrimidinyl; wherein said V ring is optionally mono-, di- or tri-substituted independently with halo, (C C6)alkyi, hydroxy, (CrC6)alkoxy, nitro, cyano or oxo wherein said (C -C6)alkyl substituent optionally has from one to nine fluorines; and
R6 and R7 are each independently hydrogen, halo or (C C3)alkyl, said (C
C3)alkyl optionally having from one to seven fluorines; or a pharmaceutically acceptable salt thereof.
4. A compound as recited in claim 3 wherein Q is methyiene and V is phenyl or pyridinyl; wherein said V ring is optionally mono-, di- or tri-substituted independently with haio, (C C2)alkyl, or nitro wherein said (C C2)alkyl optionally has from one to five fluorines; or a pharmaceutically acceptable salt thereof. 5. A compound as recited in claim 1 wherein said compound is
[2R,4S] 4-[(3,5-Bis-trifluoromethyl-benzyl)-methoxycarbonyl-amino]-2-methyl-
7-trifluoromethyl-3,4-dihydro-2H-quinoline-1 -carboxylic acid ethyl ester;
[2R.4S] 4-[(3,5-Bis-trifluoromethyl-benzyl)-methoxycarbonyl-amino]-7-chloro-
2-methyl-3,4-dihydro-2H-quinoline-1 -carboxylic acid ethyl ester; [2R,4S] 4-[(3,5-Bis-trifluoromethyl-benzyl)-methoxycarbonyl-amino]-6-chloro-
2-methyl-3,4-dihydro-2H-quinoline-1-carboxyiic acid ethyl ester;
[2R,4S] 4-[(3,5-Bis-trifluoromethyl-benzyl)-methoxycarbonyl-amino]-2,6,7- trimethyl-3,4-dihydro-2H-quinoline-1 -carboxylic acid ethyl ester or a pharmaceutically acceptable salt of said compounds. 6. A compound as recited in claim 1 wherein said compound is
[2R,4S] 4-[(3,5-Bis-trifluoromethyl-benzyl)-methoxycarbonyl-amino]-6,7- diethyl-2-methyl-3,4-dihydro-2H-quinoline-1 -carboxylic acid ethyl ester;
[2R.4S] 4-[(3,5-Bis-trifluoromethyl-benzyl)-methoxycarbonyl-amino]-6-ethyl-2- methyl-3,4-dihydro-2H-quinoline-1 -carboxylic acid ethyl ester; [2R,4S] 4-[(3,5-Bis-trifluoromethyl-benzyl)-methoxycarbonyl-amino]-2-methyl-
6-trifluoromethyl-3,4-dihydro-2H-quinoiine-1 -carboxylic acid ethyl ester;
[2R,4S] 4-[(3,5-bis-trifluoromethyl-benzyl)-methoxycarbonyl-amino]-2-methyl-
6-trifiuoromethyl-3,4-dihydro-2H-quinoline-1 -carboxylic acid isopropyl ester; or a pharmaceutically acceptable salt of said compounds. 7. A compound as recited in claim 4 wherein
Y is ethyl;
R3 is 3,5-bis-trifluoromethylphenylmethyl;
R4 is methyl;
R6 is hydrogen; and R7 is trifluoromethyl; or a phaπnaceutically acceptable salt thereof.
8. A compound as recited in claim 4 wherein Y is ethyl;
R3 is 3,5-bis-trifluoromethylphenyimethyl; R4 is methyl; R6 is hydrogen; and R7 is chloro; or a pharmaceutically acceptable salt thereof.
9. A compound as recited in claim 4 wherein
Y is ethyl;
R3 is 3,5-bis-trifluoromethylphenylmethyl; R4 is methyl; R6 is chloro; and R7 is hydrogen; or a pharmaceutically acceptable salt thereof.
10. A compound as recited in claim 4 wherein Y is ethyl;
R3 is 3,5-bis-trifluoromethylphenylmethyl; R4 is methyl; R6 is methyl; and R7 is methyl; or a pharmaceutically acceptable salt thereof.
11. A compound as recited in claim 4 wherein
Y is ethyl;
R3 is 3,5-bis-trifluoromethylphenylmethyl; R4 is methyl; R6 is ethyl; and R7 is ethyl; or a pharmaceutically acceptable salt thereof.
12. A compound as recited in claim 4 wherein
Y is ethyl; R3 is 3,5-bis-trifluoromethylphenylmethyl;
R4 is methyl; R6 is ethyi; and R7 is hydrogen; or a pharmaceutically acceptable salt thereof.
13. A compound as recited in claim 4 wherein
Y is ethyl; R3 is 3,5-bis-trifluoromethylphenyimethyl; R4 is methyl; R6 is trifluoromethyl; and R7 is hydrogen; or a pharmaceutically acceptable salt thereof. 14. A compound as recited in claim 4 wherein
Y is isopropyl;
R3 is 3,5-bis-trifluoromethylphenylmethyi;
R4 is methyl;
R6 is trifluoromethyl; and R7 is hydrogen; or a pharmaceutically acceptable salt of said compound.
15. A compound as recited in claim 1 wherein the C2 methyl is beta; the C4 nitrogen is beta: R1 is W-X ;
W is carbonyl, thiocarbonyi or sulfonyl;
X is -0-Y-, S-Y-, N(H)-Y- or -N-(Y)2-;
Y for each occurrence is independently (C C4)alkyi, said (C C4)alkyl optionally having from one to nine fluorines or said (CrC4)alkyl optionally mono-substituted with Z wherein Z is a partially saturated, fully saturated or fully unsaturated three to six membered ring optionally having one to two heteroatoms selected independently from oxygen, sulfur and nitrogen; wherein said Z substituent is optionally mono-, di- or tri-substituted independently with halo, (C C4)alkyl, (C C4)alkoxy, (C C4)alkylthio, nitro, cyano, oxo, or (C-pCeJalkyloxycarbonyl, said (C C4)alkyl substituent is also optionally substituted with from one to nine fluorines; R3 is Q-V wherein Q is (C C4)alkyl and V is a five or six membered partially saturated, fully saturated or fully unsaturated ring optionally having one to three heteroatoms selected independently from oxygen, sulfur and nitrogen; wherein said V ring is optionally mono-, di-, tri- or tetra-substituted independently with halo, (Ct-C^alkyl, hydroxy, (C C6)alkoxy, nitro, cyano or oxo wherein said (C C6)a!kyl substituent is optionally mono-, di- or tri- substituted independently with (C C6)alkoxy or (C C4)alkylthio, said (C C6)alkyl substitutent is also optionally substituted with from one to nine fluorines; R4 is (CrC4)alkyl;
R6 and R7 are each independently H, carbamoyl, oxycarbonyl, oxy or halo, or said carbamoyl, oxycarbonyl or oxy are substituted with (C C4)alkyi and said (C C4)alkyi is optionally mono-, di-, or tri-substituted with halo or hydroxy or said (C C4)alkyl is optionally mono-substituted with (C-|-C4)alkoxy, carboxy, (C1-C4)alkylthio, (C1-C4)alkoxycarbonyl, amino or mono-N- or di-N,N-(C1-C4)alkylamino or said (C1-C )alkyl optionally having from one to nine fluorines; or said (C1-C4)alkyl, carbamoyl, oxycarbonyl or oxy are optionally mono- substituted with T; wherein T is a partially saturated, fully saturated or fully unsaturated three to six membered ring optionally having one to two heteroatoms selected independently from oxygen, sulfur and nitrogen; wherein said T ring is optionally mono-, di-, tri- or tetra-substituted independently with halo, (C C6)alkyl, hydroxy, (C C6)alkoxy, nitro, cyano or oxo wherein said (C1-C6)alkyl substituent is optionally mono-, di- or tri- substituted independently with (C C6)alkoxy or (C C4)alkylthio wherein said (CrC6)alkyl substituent optionally has from one to nine fluorines; and R5 and R8 are H; or a pharmaceutically acceptable salt thereof. 16. A compound as recited in claim 15 wherein W is carbonyl;
X is O-Y wherein Y is (C C4), said (C C4)alkyl optionally having from one to nine fluorines; Q is (C C4)alkyiene and V is phenyl, pyridinyl, or pyrimidinyl; wherein said V ring is optionally mono-, di- or tri-substituted independently with halo, (C C6)alkyl, hydroxy, (C C6)alkoxy, nitro, cyano or oxo wherein said (C C6)alkyl substituent optionally has from one to nine fluorines; -
R6 is H, carbamoyl, oxycarbonyl, oxy or halo, or said carbamoyl, oxycarbonyl or oxy are substituted with (C C^alkyl and said (C-ι-C2)alkyl is optionally mono-, di-, or tri-substituted with halo or hydroxy, said (C C2)alkyl substituent optionally has from one to five fluorines; and R7 is H, carbamoyl, oxycarbonyl, oxy or halo, or said carbamoyl, oxycarbonyl or oxy are optionally substituted with (C C4)aikyi and said (C1-C4)alkyl is optionally mono-, di-, or tri-substituted with halo or hydroxy, said (C C4)alkyl substituent optionally has from one to nine fluorines; or a pharmaceutically acceptable salt thereof. 17. A method for treating atherosclerosis, peripheral vascular disease, dyslipidemia, hyperbetalipoproteinemia, hypoalphalipoproteinemia, hypercholesterolemia, hypertrigiyceridemia, familial-hypercholesterolemia, cardiovascular disorders, angina, ischemia, cardiac ischemia, stroke, myocardial infarction, reperfusion injury, angioplastic restenosis, hypertension, vascular complications of diabetes, obesity or endotoxemia in a mammal (including a human being either male or female) by administering to a mammal in need of such treatment an atherosclerosis, peripheral vascular disease, dyslipidemia, hyperbetalipoproteinemia, hypoalphalipoproteinemia, hypercholesterolemia, hypertrigiyceridemia, familial-hypercholesterolemia, cardiovascular disorders, angina, ischemia, cardiac ischemia, stroke, myocardial infarction, reperfusion injury, angioplastic restenosis, hypertension, reperfusion injury, vascular complications of diabetes, obesity or endotoxemia treating amount of a compound of claim 1 , a prodrug thereof, or a pharmaceutically acceptable salt of said compound or of said prodrug. 18. A method as recited in claim 17 wherein atherosclerosis is treated.
19. A method as recited in claim 17 wherein peripheral vascular disease is treated.
20. A method as recited in claim 17 wherein dyslipidemia is treated.
21. A method as recited in claim 17 wherein hyperbetalipoproteinemia is treated.
22. A method as recited in claim 17 wherein hypoalphalipoproteinemia is treated.
23. A method as recited in claim 17 wherein hypercholesterolemia is treated. 24. A method as recited in claim 17 wherein hypertrigiyceridemia is treated.
25. A method as recited in claim 17 wherein cardiovascular disorders are treated.
26. A pharmaceutical composition which comprises a therapeutically effective amount of a compound of claim 1 , a prodrug thereof, or a pharmaceutically acceptable salt of said compound or of said prodrug and a pharmaceutically acceptable carrier.
27. A pharmaceutical composition for the treatment of atherosclerosis, peripheral vascular disease, dyslipidemia, hyperbetalipoproteinemia, hypoalphalipoproteinemia, hypercholesterolemia, hypertrigiyceridemia, familial-hypercholesterolemia, cardiovascular disorders, angina, ischemia, cardiac ischemia, stroke, myocardial infarction, reperfusion injury, angioplastic restenosis, hypertension, vascular complications of diabetes, obesity or endotoxemia in a mammal which comprise a therapeutically effective amount of a compound of claim 1 , a prodrug thereof, or a pharmaceutically acceptable salt of said compound or of said prodrug and a pharmaceutically acceptable carrier. 28. A pharmaceutical composition for the treatment of atherosclerosis in a mammal which comprises an atherosclerosis treating amount of a compound of claim 1 , a prodrug thereof, or a pharmaceutically acceptable salt of said compound or of said prodrug and a pharmaceutically acceptable carrier. 29. A pharmaceutical combination composition comprising: a therapeutically effective amount of a composition comprising a first compound, said first compound being a compound of claim 1 , a prodrug thereof, or a pharmaceutically acceptable salt of said compound or of said prodrug; a second compound, said second compound being an HMG CoA reductase inhibitor, an MTP/Apo B secretion inhibitor, a PPAR activator, a bile acid reuptake inhibitor, a cholesterol absoφtion inhibitor, a cholesterol synthesis inhibitor, a fibrate, niacin, an ion-exchange resin, an antioxidant, an ACAT inhibitor or a bile acid sequestrant; and a pharmaceutical earner. 30. A pharmaceutical combination composition as recited in claim 29 wherein the second compound is an HMG-CoA reductase inhibitor or a MTP/Apo B secretion inhibitor. 31. A pharmaceutical combination composition as recited in claim 29 wherein the second compound is lovastatin, simvastatin, pravastatin, fluvastatin, atorvastatin or rivastatin.
32. A method for treating atherosclerosis in a mammal comprising administering to a mammal in need of treatment thereof; a first compound, said first compound being a compound of claim 1 , a prodrug thereof, or a pharmaceutically acceptable salt of said compound or of said prodrug; and a second compound, said second compound being an HMG CoA reductase inhibitor, an MTP/Apo B secretion inhibitor, a cholesterol absoφtion inhibitor, a cholesterol synthesis inhibitor, a fibrate, niacin, an ion-exchange resin, an antioxidant, an ACAT inhibitor or a bile acid sequestrant wherein the amounts of first and second compounds result in a therapeutic effect.
33. A method for treating atherosclerosis as recited in claim 32 wherein the second compound is an HMG-CoA reductase inhibitor or a MTP/Apo B secretion inhibitor.
34. A method for treating atherosclerosis as recited in ciaim 32 wherein the second compound is lovastatin, simvastatin, pravastatin, fluvastatin, atorvastatin or rivastatin. 35. A kit comprising: a. a first compound, said first compound being a compound of claim 1 , a prodrug thereof or a pharmaceutically acceptable salt of said compound or of said prodrug and a pharmaceutically acceptable earner in a first unit dosage form; b. a second compound, said second compound being an HMG CoA reductase inhibitor, an MTP/Apo B secretion inhibitor, a cholesterol absoφtion inhibitor, a cholesterol synthesis inhibitor, a fibrate, niacin, an ion-exchange resin, an antioxidant, an ACAT inhibitor or a bile acid sequestrant and a pharmaceutically acceptable carrier in a second unit dosage form; and c. means for containing said first and second dosage forms wherein the amounts of first and second compounds result in a therapeutic effect.
36. A kit as recited in claim 35 wherein said second compound is an HMG-CoA reductase inhibitor or an MTP/Apo B secretion inhibitor.
37. A kit as recited in claim 35 wherein said second compound is lovastatin, simvastatin, pravastatin, fluvastatin, atorvastatin or rivastatin.
PCT/IB1999/001529 1998-09-17 1999-09-10 4-carboxyamino-2-methyl-1,2,3,4-tetrahydroquinolines as cetp inhibitors WO2000017166A1 (en)

Priority Applications (8)

Application Number Priority Date Filing Date Title
CA002344556A CA2344556A1 (en) 1998-09-17 1999-09-10 4-carboxyamino-2-methyl-1,2,3,4-tetrahydroquinolines
AU54398/99A AU5439899A (en) 1998-09-17 1999-09-10 4-carboxyamino-2-methyl-1,2,3,4-tetrahydroquinolines as cetp inhibitors
EP99940421A EP1114033B1 (en) 1998-09-17 1999-09-10 4-carboxyamino-2-methyl-1,2,3,4-tetrahydroquinolines as cetp inhibitors
SI9930672T SI1114033T1 (en) 1998-09-17 1999-09-10 4-carboxyamino-2-methyl-1,2,3,4-tetrahydroquinolines as cetp inhibitors
DE69920727T DE69920727T2 (en) 1998-09-17 1999-09-10 4-CARBOXAMINO-2-METHYL-1,2,3,4-TETRAHYDROCHINOLINE AS CETP INHIBITORS
AT99940421T ATE277906T1 (en) 1998-09-17 1999-09-10 4-CARBOXAMINO-2-METHYL-1,2,3,4-TETRAHYDROCHINOLINES AS CETP INHIBITORS
BR9913842-5A BR9913842A (en) 1998-09-17 1999-09-10 4-carboxyamino-2-methyl-1,2,3,4-tetrahydroquinolines as cetp inhibitors
JP2000574076A JP3655193B2 (en) 1998-09-17 1999-09-10 4-Carboxyamino-2-methyl-1,2,3,4-tetrahydroquinolines as CETP inhibitors

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US10092998P 1998-09-17 1998-09-17
US60/100,929 1998-09-17

Publications (1)

Publication Number Publication Date
WO2000017166A1 true WO2000017166A1 (en) 2000-03-30

Family

ID=22282259

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB1999/001529 WO2000017166A1 (en) 1998-09-17 1999-09-10 4-carboxyamino-2-methyl-1,2,3,4-tetrahydroquinolines as cetp inhibitors

Country Status (25)

Country Link
US (2) US6147090A (en)
EP (1) EP1114033B1 (en)
JP (1) JP3655193B2 (en)
AR (1) AR021482A1 (en)
AT (1) ATE277906T1 (en)
AU (1) AU5439899A (en)
BR (1) BR9913842A (en)
CA (1) CA2344556A1 (en)
CO (1) CO5150220A1 (en)
DE (1) DE69920727T2 (en)
DK (1) DK1114033T3 (en)
DZ (1) DZ2889A1 (en)
ES (1) ES2228086T3 (en)
GT (1) GT199900149A (en)
HN (1) HN1999000155A (en)
MA (1) MA26687A1 (en)
MY (1) MY121838A (en)
PA (1) PA8481501A1 (en)
PE (1) PE20001048A1 (en)
PT (1) PT1114033E (en)
SI (1) SI1114033T1 (en)
SV (1) SV1999000149A (en)
TN (1) TNSN99172A1 (en)
TW (1) TWI232859B (en)
WO (1) WO2000017166A1 (en)

Cited By (29)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002079165A1 (en) * 2001-03-30 2002-10-10 Astrazeneca Ab Tetrahydroquinoline derivatives as stat6-modulators, preparation and use thereof
WO2003000235A1 (en) * 2001-06-22 2003-01-03 Pfizer Products Inc. Pharmaceutical compositions of dispersions of drugs and neutral polymers
WO2005095409A2 (en) * 2004-04-02 2005-10-13 Tanabe Seiyaku Co., Ltd. Tetrahydroquinoline derivatives and a process for preparing the same
WO2005097806A1 (en) 2004-03-26 2005-10-20 Eli Lilly And Company Compounds and methods for treating dyslipidemia
US6962931B2 (en) 2001-06-21 2005-11-08 Pfizer Inc. Self-emulsifying formulations of cholesteryl ester transfer protein inhibitors
WO2005095395A3 (en) * 2004-04-02 2006-06-01 Tanabe Seiyaku Co Tetrahydronaphthyridine derivatives as cholesteryl ester transfer protein inhibitors
US7094801B2 (en) 2001-12-19 2006-08-22 Atherogenics, Inc. Chalcone derivatives and their use to treat diseases
JP2007508301A (en) * 2003-10-08 2007-04-05 イーライ リリー アンド カンパニー Compounds and methods for the treatment of dyslipidemia
US7202247B2 (en) 2001-12-19 2007-04-10 Atherogenics, Inc. 1,3-bis-(substituted-phenyl)-2-propyn-1-ones and their use to treat disorders
US7211672B2 (en) 2002-10-04 2007-05-01 Millennium Pharmaceuticals, Inc. PGD2 receptor antagonists for the treatment of inflammatory diseases
WO2007088999A1 (en) 2006-01-31 2007-08-09 Mitsubishi Tanabe Pharma Corporation Trisubstituted amine compound
WO2007090749A2 (en) * 2006-02-07 2007-08-16 F. Hoffmann-La Roche Ag Benzamide and heteroarene derivatives as cetp inhibitors
WO2007107843A1 (en) * 2006-03-22 2007-09-27 Pfizer Products Inc. Methods of treatment with cetp inhibitors
WO2007116922A1 (en) 2006-03-30 2007-10-18 Mitsubishi Tanabe Pharma Corporation A process for preparing tetrahydroquinoline derivatives
US7317025B2 (en) 2003-09-24 2008-01-08 Johnson & Johnson Pharmaceutical Research & Development, Llc Non-peptidic NPY Y2 receptor inhibitors
WO2008018529A1 (en) 2006-08-11 2008-02-14 Kowa Company, Ltd. Novel pyrimidine compound having benzyl(pyridylmethyl)amine structure and pharmaceutical comprising the compound
US7332514B2 (en) 2002-08-30 2008-02-19 Japan Tobacco Inc. Dibenzylamine compound and medicinal use thereof
WO2008060476A2 (en) 2006-11-15 2008-05-22 Schering Corporation Nitrogen-containing heterocyclic compounds and methods of use thereof
WO2008111604A1 (en) 2007-03-13 2008-09-18 Kowa Company, Ltd. Novel pyrimidine compound having benzyl(heterocyclicmethyl)amine structure and pharmaceutical product containing the same
WO2008129951A1 (en) 2007-04-13 2008-10-30 Kowa Company, Ltd. Novel pyrimidine compound having dibenzylamine structure, and medicine comprising the compound
US7504508B2 (en) 2002-10-04 2009-03-17 Millennium Pharmaceuticals, Inc. PGD2 receptor antagonists for the treatment of inflammatory diseases
WO2010075068A1 (en) 2008-12-16 2010-07-01 Schering Corporation Pyridopyrimidine derivatives and methods of use thereof
WO2010075069A1 (en) 2008-12-16 2010-07-01 Schering Corporation Bicyclic pyranone derivatives as nicotinic acid receptor agonists
EP2275091A1 (en) 2001-06-22 2011-01-19 Pfizer Products Inc. Pharmaceutical compositions comprising adsorbates of an amorphous drug
EP2283822A2 (en) 2003-08-04 2011-02-16 Bend Research, Inc. Spray drying processes for forming solid amorphous dispersions of drugs and polymers
US7906517B2 (en) 2006-01-31 2011-03-15 Mitsubishi Tanabe Pharma Corporation Trisubstituted amine compound
US7951950B2 (en) 2005-02-24 2011-05-31 Millennium Pharmaceuticals, Inc. PGD2 receptor antagonists for the treatment of inflammatory diseases
EP2548894A1 (en) 2005-02-03 2013-01-23 Bend Research, Inc. Pharmaceutical compositions with enhanced performance
US20210393614A1 (en) * 2018-10-17 2021-12-23 Duke University Quinone reductase 2 inhibitors for use as neuroprotective agents

Families Citing this family (58)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB9602166D0 (en) * 1996-02-02 1996-04-03 Zeneca Ltd Aminoheterocyclic derivatives
US6147089A (en) * 1998-09-17 2000-11-14 Pfizer Inc. Annulated 4-carboxyamino-2-methyl-1,2,3,4,-tetrahydroquinolines
US6197786B1 (en) * 1998-09-17 2001-03-06 Pfizer Inc 4-Carboxyamino-2-substituted-1,2,3,4-tetrahydroquinolines
ATE524163T1 (en) 2000-04-10 2011-09-15 Teva Pharma STABLE PHARMACEUTICAL COMPOSITIONS CONTAINING 7-SUBSTITUTED-3,5-DIHYDROXYHEPTENOIC ACIDS OR 7-SUBSTITUTED-3,5-DIHYDROXYHEPTENOIC ACIDS
USRE44578E1 (en) 2000-04-10 2013-11-05 Teva Pharmaceutical Industries, Ltd. Stable pharmaceutical compositions containing 7-substituted-3,5-dihydroxyheptanoic acids or 7-substituted-3,5-dihydroxyheptenoic acids
US7115279B2 (en) * 2000-08-03 2006-10-03 Curatolo William J Pharmaceutical compositions of cholesteryl ester transfer protein inhibitors
US6982251B2 (en) * 2000-12-20 2006-01-03 Schering Corporation Substituted 2-azetidinones useful as hypocholesterolemic agents
US20060287254A1 (en) * 2001-01-26 2006-12-21 Schering Corporation Use of substituted azetidinone compounds for the treatment of sitosterolemia
EP1911462A3 (en) 2001-01-26 2011-11-30 Schering Corporation Compositions comprising a sterol absorption inhibitor
WO2002078625A2 (en) * 2001-03-28 2002-10-10 Pharmacia Corporation Therapeutic combinations for cardiovascular and inflammatory indications
EP1269994A3 (en) 2001-06-22 2003-02-12 Pfizer Products Inc. Pharmaceutical compositions comprising drug and concentration-enhancing polymers
AR038375A1 (en) 2002-02-01 2005-01-12 Pfizer Prod Inc PHARMACEUTICAL COMPOSITIONS OF INHIBITORS OF THE PROTEIN OF TRANSFER OF ESTERES DE COLESTERILO
EP1469832B2 (en) * 2002-02-01 2016-10-26 Bend Research, Inc. Pharmaceutical compositions of amorphous dispersions of drugs and lipophilic microphase-forming materials
RU2004123637A (en) * 2002-02-01 2005-04-20 Пфайзер Продактс Инк. (Us) PHARMACEUTICAL MEDICINAL FORMS OF CONTROLLED RELEASE OF THE CHOLESTEROL PROTEIN PROTEIN INHIBITOR
US20040053842A1 (en) * 2002-07-02 2004-03-18 Pfizer Inc. Methods of treatment with CETP inhibitors and antihypertensive agents
US7071210B2 (en) * 2002-07-02 2006-07-04 Pfizer Inc. CETP inhibitors in combination with antihypertensive agents and uses thereof
US20090181966A1 (en) * 2002-10-04 2009-07-16 Millennium Pharmaceuticals, Inc. PGD2 receptor antagonists for the treatment of inflammatory diseases
BR0315547A (en) * 2002-10-21 2005-09-20 Warner Lambert Co Quinoline derivatives as crth2 antagonists
US20040181075A1 (en) * 2002-12-19 2004-09-16 Weingarten M. David Process of making chalcone derivatives
BR0317521A (en) 2002-12-20 2005-11-16 Pfizer Prod Inc Dosage forms comprising a cetp inhibitor and an hmg-coa reductase inhibitor
US20040132771A1 (en) * 2002-12-20 2004-07-08 Pfizer Inc Compositions of choleseteryl ester transfer protein inhibitors and HMG-CoA reductase inhibitors
DE602004018617D1 (en) 2003-03-07 2009-02-05 Schering Corp SUBSTITUTED AZETIDINONE DERIVATIVES, THEIR PHARMACEUTICAL FORMULATIONS AND THEIR USE FOR THE TREATMENT OF HYPERCHOLESTEROLMIA
ATE406364T1 (en) 2003-03-07 2008-09-15 Schering Corp SUBSTITUTED AZETIDINONE DERIVATIVES, THEIR PHARMACEUTICAL FORMULATIONS AND THEIR USE IN THE TREATMENT OF HYPERCHOLESTEROLEMIA
WO2004082675A1 (en) * 2003-03-17 2004-09-30 Japan Tobacco Inc. Method for increasing the oral bioavailability of s-[2- ([[1- (2-ethylbutyl) cyclohexyl] carbonyl] amino) phenyl] 2-methylpropanethioate
SI1603553T1 (en) * 2003-03-17 2012-03-30 Japan Tobacco Inc Pharmaceutical compositions of cetp inhibitors
US20040204450A1 (en) * 2003-03-28 2004-10-14 Pfizer Inc Quinoline and quinoxaline compounds
TWI494102B (en) * 2003-05-02 2015-08-01 Japan Tobacco Inc Combination comprising s-(2-(((1-(2-ethylbutyl)cyclohexyl)carbonyl)amino)phenyl)2-methylpropanethioate and an hmg coa reductase inhibitor
BRPI0410840A (en) * 2003-05-30 2006-07-04 Ranbaxy Lab Ltd substituted pyrrol derivatives, pharmaceutical composition containing them and process for their preparation
ATE540671T1 (en) 2003-08-04 2012-01-15 Bend Res Inc PHARMACEUTICAL COMPOSITIONS OF AMORPHOUS DRUG ADSORBATES AND LIPOPHILIC MICROPHASE-FORMING MATERIALS
CA2554982A1 (en) 2003-09-26 2005-04-07 Japan Tobacco Inc. Method of inhibiting remnant lipoprotein production
EP1918000A2 (en) 2003-11-05 2008-05-07 Schering Corporation Combinations of lipid modulating agents and substituted azetidinones and treatments for vascular conditions
US20060063828A1 (en) * 2004-06-28 2006-03-23 Weingarten M D 1,2-Bis-(substituted-phenyl)-2-propen-1-ones and pharmaceutical compositions thereof
US20060069080A1 (en) * 2004-09-29 2006-03-30 Veltri Enrico P Combinations of substituted azetidinones and CB1 antagonists
NZ555320A (en) 2004-12-03 2010-11-26 Schering Corp Substituted piperazines as CB1 antagonists
WO2006069162A1 (en) * 2004-12-20 2006-06-29 Reddy Us Therapeutics, Inc. Novel heterocyclic compounds and their pharmaceutical compositions
WO2006098394A1 (en) * 2005-03-14 2006-09-21 Japan Tobacco Inc. Method for inhibiting lipid absorption and lipid absorption inhibitor
JP4681526B2 (en) * 2005-09-29 2011-05-11 田辺三菱製薬株式会社 Pharmaceutical composition
BRPI0618379A2 (en) 2005-11-08 2011-08-30 Ranbaxy Lab Ltd (3r, 5r) -7- [2- (4-fluorophenyl) -5-isopropyl-3-phenyl-4 - [(4-hydroxymethylphenylamino) carbonyl] -pyrroleic acid 1-yl] -3,5-dihydroxy heptanoic
JP2009528266A (en) 2006-01-18 2009-08-06 シェーリング コーポレイション Cannabinoid receptor modifier
US20070238716A1 (en) * 2006-03-14 2007-10-11 Murthy Ayanampudi S R Statin stabilizing dosage formulations
CL2007002044A1 (en) * 2006-07-14 2008-06-13 Ranbaxy Lab Ltd CRYSTALLINE POLYMORPH OF HEMICALCIC ACID SALT (3R, 5R) -7- [2- (4-FLUOROPHENYL) -5-ISOPROPIL-3-PHENYL-4 - [(4-HYDROXIMETHYLPHENYLAMINE) CARBON]] -PIRROL-1-IL] -3,5-DIHYDROXIHEPTANOIC; PHARMACEUTICAL COMPOSITION; AND USE FOR THE TREATMENT OF DIABETES, ENF
WO2008130616A2 (en) * 2007-04-19 2008-10-30 Schering Corporation Diaryl morpholines as cb1 modulators
KR20100051625A (en) 2007-06-28 2010-05-17 인터벳 인터내셔널 비.브이. Substituted piperazines as cb1 antagonists
CN101790521A (en) * 2007-06-28 2010-07-28 英特维特国际股份有限公司 Substituted-piperazinyl as the CB1 antagonist
AU2008317057B8 (en) 2007-10-22 2014-02-13 Merck Sharp & Dohme Corp. Bicyclic Heterocycle Derivatives and their use as modulators of the activity of GPR119
EP2297117B1 (en) 2008-05-19 2012-10-31 Merck Sharp & Dohme Corp. Bicyclic heterocycle derivatives and use thereof as gpr119 modulators
EP2318404B1 (en) 2008-07-16 2013-08-21 Merck Sharp & Dohme Corp. Bicyclic heterocycle derivatives and use thereof as gpr119 modulators
WO2010075273A1 (en) 2008-12-23 2010-07-01 Schering Corporation Bicyclic heterocycle derivatives and methods of use thereof
CA2747809A1 (en) 2008-12-23 2010-07-01 Joel M. Harris Bicyclic heterocycle derivatives and methods of use thereof
US8722882B2 (en) 2008-12-23 2014-05-13 Merck Sharp & Dohme Corp. Pyrimidine derivatives as GPCR modulators for use in the treatment of obesity and diabetes
WO2010114957A1 (en) 2009-04-03 2010-10-07 Schering Corporation Bicyclic piperidine and piperazine derivatives as gpcr modulators for the treatment of obesity, diabetes and other metabolic disorders
AR076024A1 (en) 2009-04-03 2011-05-11 Schering Corp DERIVATIVES OF BRIDGED BICYCLIC HETEROCICLES AND METHODS OF USE OF THE SAME
EP2493307B1 (en) 2009-10-29 2016-04-27 Merck Sharp & Dohme Corp. Bridged bicyclic piperidine derivatives and methods of use thereof
EP2503891B1 (en) 2009-11-23 2016-08-03 Merck Sharp & Dohme Corp. Pyrimidine ether derivatives and methods of use thereof
US20120232073A1 (en) 2009-11-23 2012-09-13 Santhosh Francis Neelamkavil Fused bicyclic pyrimidine derivatives and methods of use thereof
EP2503887B1 (en) 2009-11-24 2016-01-06 Merck Sharp & Dohme Corp. Substituted biaryl derivatives and methods of use thereof
US20130156720A1 (en) 2010-08-27 2013-06-20 Ironwood Pharmaceuticals, Inc. Compositions and methods for treating or preventing metabolic syndrome and related diseases and disorders
BR112017003745A2 (en) 2014-08-29 2017-12-05 Tes Pharma S R L alpha-amino-beta-carboximuconic acid semialdehyde decarboxylase inhibitors

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0818448A1 (en) * 1996-07-08 1998-01-14 Bayer Ag Cycloalkano-pyridine as CETP inhibitors

Family Cites Families (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5231102A (en) * 1989-03-08 1993-07-27 Merck Sharp & Dohme, Ltd. Tetrahydroquinoline derivatives useful for neurodegenerative disorders
US5276168A (en) * 1990-06-18 1994-01-04 E. R. Squibb & Sons, Inc. Benzopyran derivatives and heterocyclic analogs thereof as antiischemic agents
US5401848A (en) * 1990-11-26 1995-03-28 E. R. Squibb & Sons, Inc. Indane and quinoline derivatives
KR920014799A (en) * 1991-01-18 1992-08-25 나오가따 다이도 Novel benzo [5,6] cyclohepta [1,2-b] pyridine derivatives and anti-allergic agents containing them
US5288725A (en) * 1992-10-15 1994-02-22 Merck & Co., Inc. Pyrroloquinoline Bradykinin antagonist
DE19627431A1 (en) 1996-07-08 1998-01-15 Bayer Ag Heterocyclically fused pyridines
US6147089A (en) * 1998-09-17 2000-11-14 Pfizer Inc. Annulated 4-carboxyamino-2-methyl-1,2,3,4,-tetrahydroquinolines
US6197786B1 (en) * 1998-09-17 2001-03-06 Pfizer Inc 4-Carboxyamino-2-substituted-1,2,3,4-tetrahydroquinolines
AU2157400A (en) 1998-12-23 2000-07-31 G.D. Searle & Co. Combinations of cholesteryl ester transfer protein inhibitors and hmg coa reductase inhibitors for cardiovascular indications

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0818448A1 (en) * 1996-07-08 1998-01-14 Bayer Ag Cycloalkano-pyridine as CETP inhibitors

Cited By (53)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002079165A1 (en) * 2001-03-30 2002-10-10 Astrazeneca Ab Tetrahydroquinoline derivatives as stat6-modulators, preparation and use thereof
US7030246B2 (en) 2001-03-30 2006-04-18 Astrazeneca Ab Tetrahydroquinoline derivatives as stat6-modulators, preparation and use thereof
US6962931B2 (en) 2001-06-21 2005-11-08 Pfizer Inc. Self-emulsifying formulations of cholesteryl ester transfer protein inhibitors
EP2275091A1 (en) 2001-06-22 2011-01-19 Pfizer Products Inc. Pharmaceutical compositions comprising adsorbates of an amorphous drug
US8389006B2 (en) 2001-06-22 2013-03-05 Bend Research, Inc. Pharmaceutical compositions of adsorbates of amorphous drug
US9468604B2 (en) 2001-06-22 2016-10-18 Bend Research, Inc. Pharmaceutical compositions of dispersions of drug and neutral polymers
WO2003000235A1 (en) * 2001-06-22 2003-01-03 Pfizer Products Inc. Pharmaceutical compositions of dispersions of drugs and neutral polymers
US8703199B2 (en) 2001-06-22 2014-04-22 Bend Research, Inc. Pharmaceutical compositions of adsorbates of amorphous drug
US7202247B2 (en) 2001-12-19 2007-04-10 Atherogenics, Inc. 1,3-bis-(substituted-phenyl)-2-propyn-1-ones and their use to treat disorders
US7094801B2 (en) 2001-12-19 2006-08-22 Atherogenics, Inc. Chalcone derivatives and their use to treat diseases
US7807701B2 (en) 2002-08-30 2010-10-05 Japan Tobacco Inc. Dibenzylamine compounds and pharmaceutical use thereof
US7332514B2 (en) 2002-08-30 2008-02-19 Japan Tobacco Inc. Dibenzylamine compound and medicinal use thereof
US7211672B2 (en) 2002-10-04 2007-05-01 Millennium Pharmaceuticals, Inc. PGD2 receptor antagonists for the treatment of inflammatory diseases
US7504508B2 (en) 2002-10-04 2009-03-17 Millennium Pharmaceuticals, Inc. PGD2 receptor antagonists for the treatment of inflammatory diseases
EP2283822A2 (en) 2003-08-04 2011-02-16 Bend Research, Inc. Spray drying processes for forming solid amorphous dispersions of drugs and polymers
US7317025B2 (en) 2003-09-24 2008-01-08 Johnson & Johnson Pharmaceutical Research & Development, Llc Non-peptidic NPY Y2 receptor inhibitors
JP2007508301A (en) * 2003-10-08 2007-04-05 イーライ リリー アンド カンパニー Compounds and methods for the treatment of dyslipidemia
EP2098512A1 (en) 2003-10-08 2009-09-09 Eli Lilly & Company Compounds and methods for treating dyslipidemia
US7470705B2 (en) 2004-03-26 2008-12-30 Eli Lilly And Company Compounds and methods for treating dyslipidemia
WO2005097806A1 (en) 2004-03-26 2005-10-20 Eli Lilly And Company Compounds and methods for treating dyslipidemia
US7781443B2 (en) 2004-04-02 2010-08-24 Mitsubishi Tanabe Pharma Corporation Tetrahydronaphthyridine derivatives and a process for preparing the same
US7872126B2 (en) 2004-04-02 2011-01-18 Mitsubishi Tanabe Pharma Corporation Tetrahydroquinoline derivatives and a process for preparing the same
WO2005095409A2 (en) * 2004-04-02 2005-10-13 Tanabe Seiyaku Co., Ltd. Tetrahydroquinoline derivatives and a process for preparing the same
WO2005095409A3 (en) * 2004-04-02 2006-02-09 Tanabe Seiyaku Co Tetrahydroquinoline derivatives and a process for preparing the same
CN100441579C (en) * 2004-04-02 2008-12-10 田边三菱制药株式会社 Tetrahydronaphthyridine derivatives as cholesteryl ester transfer protein inhibitors
US8158640B2 (en) 2004-04-02 2012-04-17 Mitsubishi Tanabe Pharma Corporation Tetrahydroquinoline derivatives and a process for preparing the same
AU2005228291B2 (en) * 2004-04-02 2008-06-26 Mitsubishi Tanabe Pharma Corporation Tetrahydronaphthyridine derivatives and a process for preparing the same
WO2005095395A3 (en) * 2004-04-02 2006-06-01 Tanabe Seiyaku Co Tetrahydronaphthyridine derivatives as cholesteryl ester transfer protein inhibitors
EA011670B1 (en) * 2004-04-02 2009-04-28 Мицубиси Танабе Фарма Корпорейшн Tetrahydroquinoline derivatives and a process for preparing the same
EP2548894A1 (en) 2005-02-03 2013-01-23 Bend Research, Inc. Pharmaceutical compositions with enhanced performance
US7951950B2 (en) 2005-02-24 2011-05-31 Millennium Pharmaceuticals, Inc. PGD2 receptor antagonists for the treatment of inflammatory diseases
US8076364B2 (en) 2006-01-31 2011-12-13 Mitsubishi Tanabe Pharma Corporation Trisubstituted amine compound
WO2007088999A1 (en) 2006-01-31 2007-08-09 Mitsubishi Tanabe Pharma Corporation Trisubstituted amine compound
US7906517B2 (en) 2006-01-31 2011-03-15 Mitsubishi Tanabe Pharma Corporation Trisubstituted amine compound
US7674815B2 (en) 2006-02-07 2010-03-09 Hoffmann-La Roche Inc. Heteroaryl and benzyl amide compounds
WO2007090749A3 (en) * 2006-02-07 2007-11-29 Hoffmann La Roche Benzamide and heteroarene derivatives as cetp inhibitors
WO2007090749A2 (en) * 2006-02-07 2007-08-16 F. Hoffmann-La Roche Ag Benzamide and heteroarene derivatives as cetp inhibitors
KR101069009B1 (en) * 2006-02-07 2011-09-29 에프. 호프만-라 로슈 아게 Benzamide and heteroarene derivatives as cetp inhibitors
WO2007107843A1 (en) * 2006-03-22 2007-09-27 Pfizer Products Inc. Methods of treatment with cetp inhibitors
EP2154132A1 (en) 2006-03-30 2010-02-17 Mitsubishi Tanabe Pharma Corporation A process for preparing tetrahydroquinoline derivatives
WO2007116922A1 (en) 2006-03-30 2007-10-18 Mitsubishi Tanabe Pharma Corporation A process for preparing tetrahydroquinoline derivatives
US8084611B2 (en) 2006-03-30 2011-12-27 Mitsubishi Tanabe Pharma Corporation Process for preparing tetrahydroquinoline derivatives
WO2008018529A1 (en) 2006-08-11 2008-02-14 Kowa Company, Ltd. Novel pyrimidine compound having benzyl(pyridylmethyl)amine structure and pharmaceutical comprising the compound
US7750019B2 (en) 2006-08-11 2010-07-06 Kowa Company, Ltd. Pyrimidine compound having benzyl(pyridylmethyl)amine structure and medicament comprising the same
WO2008060476A2 (en) 2006-11-15 2008-05-22 Schering Corporation Nitrogen-containing heterocyclic compounds and methods of use thereof
US8012989B2 (en) 2007-03-13 2011-09-06 Kowa Company, Ltd. Substituted pyrimidine compounds and their utility as CETP inhibitors
US7790737B2 (en) 2007-03-13 2010-09-07 Kowa Company, Ltd. Substituted pyrimidine compounds and their utility as CETP inhibitors
WO2008111604A1 (en) 2007-03-13 2008-09-18 Kowa Company, Ltd. Novel pyrimidine compound having benzyl(heterocyclicmethyl)amine structure and pharmaceutical product containing the same
WO2008129951A1 (en) 2007-04-13 2008-10-30 Kowa Company, Ltd. Novel pyrimidine compound having dibenzylamine structure, and medicine comprising the compound
US7659271B2 (en) 2007-04-13 2010-02-09 Kowa Company, Ltd. Pyrimidine compound having dibenzylamine structure and medicament comprising the same
WO2010075068A1 (en) 2008-12-16 2010-07-01 Schering Corporation Pyridopyrimidine derivatives and methods of use thereof
WO2010075069A1 (en) 2008-12-16 2010-07-01 Schering Corporation Bicyclic pyranone derivatives as nicotinic acid receptor agonists
US20210393614A1 (en) * 2018-10-17 2021-12-23 Duke University Quinone reductase 2 inhibitors for use as neuroprotective agents

Also Published As

Publication number Publication date
AR021482A1 (en) 2002-07-24
EP1114033B1 (en) 2004-09-29
ES2228086T3 (en) 2005-04-01
SV1999000149A (en) 2000-07-06
MA26687A1 (en) 2004-12-20
DE69920727D1 (en) 2004-11-04
DK1114033T3 (en) 2004-12-20
ATE277906T1 (en) 2004-10-15
PA8481501A1 (en) 2001-12-14
GT199900149A (en) 2001-03-03
TWI232859B (en) 2005-05-21
SI1114033T1 (en) 2005-02-28
CO5150220A1 (en) 2002-04-29
TNSN99172A1 (en) 2005-11-10
PE20001048A1 (en) 2000-10-17
US6395751B1 (en) 2002-05-28
US6147090A (en) 2000-11-14
BR9913842A (en) 2001-06-12
JP2002526477A (en) 2002-08-20
JP3655193B2 (en) 2005-06-02
EP1114033A1 (en) 2001-07-11
MY121838A (en) 2006-02-28
PT1114033E (en) 2004-12-31
CA2344556A1 (en) 2000-03-30
DZ2889A1 (en) 2003-12-15
AU5439899A (en) 2000-04-10
HN1999000155A (en) 2000-01-12
DE69920727T2 (en) 2006-03-09

Similar Documents

Publication Publication Date Title
EP1114033B1 (en) 4-carboxyamino-2-methyl-1,2,3,4-tetrahydroquinolines as cetp inhibitors
US6489478B1 (en) 4-amino substituted-2-substituted-1,2,3,4-tetrahydroquinolines
AU752566B2 (en) 4-carboxyamino-2-substituted-1,2,3,4-tetrahydroquinolines as CETP inhibitors
US6147089A (en) Annulated 4-carboxyamino-2-methyl-1,2,3,4,-tetrahydroquinolines
US6140342A (en) Oxy substituted 4-carboxyamino-2-methyl-1,2,3,4-tetrahydroquinolines
AU775607B2 (en) 4-carboxyamino-2-substituted-1,2,3,4-tetrahydroquinolines as CETP inhibitors
EP1607389A1 (en) 4-protected-amino-2-substituted-1,2,3,4-tetrahydroquinolines as intermediates for CETP inhibitors
MXPA01002753A (en) 4-carboxyamino-2-methyl-1,2,3,4-tetrahydroquinolines as cetp inhibitors

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AE AL AM AT AU AZ BA BB BG BR BY CA CH CN CR CU CZ DE DK EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MD MG MK MN MW MX NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT UA UG US UZ VN YU ZA ZW

AL Designated countries for regional patents

Kind code of ref document: A1

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

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: 1999940421

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: PA/a/2001/002753

Country of ref document: MX

ENP Entry into the national phase

Ref document number: 2344556

Country of ref document: CA

Ref country code: CA

Ref document number: 2344556

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: 2000 574076

Kind code of ref document: A

Format of ref document f/p: F

WWP Wipo information: published in national office

Ref document number: 1999940421

Country of ref document: EP

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

WWG Wipo information: grant in national office

Ref document number: 1999940421

Country of ref document: EP