US20080269284A1 - Compounds and Methods for Treating Dyslipidemia - Google Patents

Compounds and Methods for Treating Dyslipidemia Download PDF

Info

Publication number
US20080269284A1
US20080269284A1 US11/570,540 US57054005A US2008269284A1 US 20080269284 A1 US20080269284 A1 US 20080269284A1 US 57054005 A US57054005 A US 57054005A US 2008269284 A1 US2008269284 A1 US 2008269284A1
Authority
US
United States
Prior art keywords
trifluoromethyl
amino
dihydro
quinoline
tetrazol
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US11/570,540
Other languages
English (en)
Inventor
Ana Maria Escribano
Maria Carmen Fernandez
Nathan Bryan Mantlo
Ana Isabel Mateo-Herranz
Eva Maria Martin De La Nava
Xiaodong Wang
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Eli Lilly and Co
Original Assignee
Eli Lilly and Co
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
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=35056992&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20080269284(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Eli Lilly and Co filed Critical Eli Lilly and Co
Priority to US11/570,540 priority Critical patent/US20080269284A1/en
Assigned to ELI LILLY AND COMPANY reassignment ELI LILLY AND COMPANY ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: ESCRIBANO, ANA MARIA, FERNANDEZ, MARIA CARMEN, MARTIN DE LA NAVA, EVA MARIA, MATEO HERRANZ, ANA ISABEL, MANTLO, NATHAN BRYAN, WANG, XIAODONG
Publication of US20080269284A1 publication Critical patent/US20080269284A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic 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 three or more hetero rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/55Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • 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
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • 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
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/14Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/12Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings linked by a chain containing hetero atoms as chain links
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/02Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains two hetero rings
    • C07D491/04Ortho-condensed systems

Definitions

  • the current invention relates to the fields of medicinal organic chemistry, pharmacology, and medicine. Further, the current invention relates to a group of compounds and methods that demonstrate utility for treating pathological states due to dyslipidemia
  • Coronary heart disease is one of the major causes of morbidity and mortality worldwide. Despite attempts to modify risk factors such as obesity, smoking, lack of exercise, and treatment of dyslipidemia with dietary modification or drug therapy, CHD remains the most common cause of death in the U.S. Over 50% of all CHD deaths are due to underlying atherosclerotic coronary heart disease.
  • Dyslipidemia is a major risk factor for CHD.
  • Low plasma levels of high density lipoprotein (HDL) cholesterol with either normal or elevated levels of low density (LDL) cholesterol is a significant risk factor for developing atherosclerosis and associated coronary artery disease in humans.
  • LDL low density lipoprotein
  • lipoprotein profiles of CHD patients have shown that about 50% of the CHD patients have cholesterol levels that are considered to be in the normal range ( ⁇ 200 mg/dl).
  • these studies found low HDL cholesterol in about 40% of the normo-cholesterolemic CHD patients as compared to the general population reported in the National Health and Nutrition Examination Survey. Since low levels of HDL cholesterol increase the risk of atherosclerosis, methods for elevating plasma HDL cholesterol would be therapeutically beneficial for the treatment of cardiovascular diseases including, but not limited to, atherosclerosis, CHD, stroke, and peripheral vascular disease.
  • CETP Cholesterol ester transfer protein
  • CETP is expressed in multiple tissues and secreted into plasma, where it associates with MDL (X. C. Jiang et al., (1991) Mammalian adipose tissue and muscle are major sources of lipid transfer protein mRNA. J. Biol. Chem. 266:4631-4639). Humans and monkeys, which express CETP, have relatively low HDL cholesterol, whereas mice and rats do not express CETP and carry nearly all their cholesterol in HDL. Furthermore, transgenic expression of CETP in mice results in significantly reduced HDL cholesterol levels and developed severe atherosclerosis compared to control mice (K. R. Marotti et. al., (1993) Severe atherosclerosis in transgenic mice expressing simian cholesteryl ester transfer protein.
  • Antibodies either directly injected into the plasma or generated through vaccine injection can effectively inhibit CETP activity in hamsters and rabbits resulting in elevated HDL cholesterol (C. W. Rittershaus, (1999) Vaccine-induced antibodies inhibit CETP activity in vivo and reduce aortic lesions in a rabbit model of atherosclerosis. Furthermore, antibody neutralization of CETP in rabbits has been shown to be anti-atherogenic ( Arterio. Thromb. Vasc. Biol. 20, 2106-2112; G. F. Evans et al., (1994) Inhibition of cholesteryl ester transfer protein in normocholesterolemic and hypercholesterolemic hamsters: effects on HDL subspecies, quantity, and apolipoprotein distribution. J. Lipid Research. 35, 1634-1645). However, antibody and/or vaccine therapy is not currently a viable option for the treatment of large populations of patients in need of treatment for dyslipidemia and resultant or associated disease state manifestations.
  • 10287662-A describes polycyclic, non-amine containing, polyhydroxylic natural compounds possessing CETP inhibition properties.
  • Lee et al. J. Antibiotics, 49, 693-96 (1996)
  • CETP inhibitors derived from an insect fungus Busch et al. (Lipids, 25, 216-220 (1990)) describe cholesteryl acetyl bromide as a CETP inhibitor.
  • Morton and Zillversmit J. Lipid Res., 35, 836-47 (1982) describe that p-chloromercuriphenyl sulfonate, p-hydroxymercuribenzoate and ethyl mercurithiosalicylate inhibit CETP. Connolly et al. ( Biochem.
  • European Patent Application No. 818448 by Schmidt et al. describes tetrahydroquinoline derivatives as cholesteryl ester transfer protein inhibitors.
  • European Patent Application No. 818197 by Schmek et al describe pyridines with fused heterocycles as cholesteryl ester transfer protein inhibitors. Brandes et al. in German Patent Application No. 19627430 describe bicyclic condensed pyridine derivatives as cholesteryl ester transfer protein inhibitors.
  • U.S. Pat. No. 6,207,671 Schmidt et al. describe substituted pyridine compounds as CETP inhibitors.
  • PCT Patent Application nos. WO 98/39299, by Müller-Gliemann et al. and WO 03/028727 by Gielen et al. certain quinoline derivatives are described as cholesteryl ester transfer protein inhibitors.
  • the present invention provides a compound of Formula I
  • R 1 is selected from a group consisting of: hydroxy, C 1 -C 6 alkyl, aryl, C 2 -C 6 alkenyl, C 1 -C 6 haloalkyl, C 1 -C 6 alkylheterocyclic, C 3 -C 8 cycloalkyl, C 1 -C 6 alkylcycloalkyl; C 1 -C 6 alkylaryl, heterocyclyl, C 1 -C 6 alkylalcohol, C 1 -C 6 alkoxy, aryloxy, —OC 2 -C 6 alkenyl, —OC 1 -C 6 haloalkyl, —OC 1 -C 6 alkylheterocyclic, —OC 3 -C 8 cycloalkyl, —OC 1 -C 6 alkylcycloalkyl, —NR 7 R 8 and —OC 1 -C 6 alkylaryl, —O-heterocyclic, —OC 1 -C
  • R 2a and R 2b are each independently selected from the group consisting of: hydrogen, hydroxy, halo, oxo, C 1 -C 6 alkyl, C 2 -C 6 alkene, C 2 -C 6 alkynyl, C 1 -C 6 alkoxy, C 1 -C 6 haloalkyl, CONR 11 R 12 , NR 11 SO 2 R 12 , NR 11 COR 12 , C 0 -C 6 alkylNR 11 R 12 , C 0 -C 6 alkylCOR 11 , C 0 -C 6 alkylCOOR 11 , cyano, nitro, C 0 -C 6 alkylcycloalkyl, phenyl, C 0 -C 6 alkylaryl, heterocyclyl, C 3 -C 8 cycloalkyl, and C 1 -C 6 haloalkyl; proviced that both R 2a a and R 2b are not simultaneously hydrogen;
  • R 3a and R 3b are independently selected from the group consisting of: hydrogen, halo, C 1 -C 6 alkyl, C 2 -C 6 alkene, C 2 -C 6 alkynyl, C 1 -C 6 alkoxy, and C 1 -C 6 haloalkyl;
  • R 4 is a group represented by the formula —NR 4a R 4b ;
  • R 4a is a heterocyclic group substituted with 1 to 3 groups independently selected from C 3 -C 6 alkyl, C 3 -C 6 alkenyl, C 0 -C 6 alkylCN, C 3 -C 6 alkoxy, C 1 -C 6 alkylalcohol, C 3 -C 6 haloalkyl, OC(O)NR 11 R 12 , C 1 -C 6 alkylNR 11 R 12 wherein the C 1 -C 6 alkyl group (of C 1 -C 6 alkylNR 11 R 12 ) is optionally substituted with —OR 10 or C(O)OR 10 , C 0 -C 6 alkylNR 11 SO 2 R 12 , C 0 -C 6 alkylC(O)NR 11 R 12 , C 1 -C 6 alkylNR 11 C(O)R 12 , C 0 -C 6 alkylNR 11 C(O)OR 12 , C 0 -C 6 alkylNR 11 CHR 10
  • R 4b is selected from the group consisting of C 1 -C 6 alkylaryl, C 2 -C 6 alkenylaryl, C 2 -C 6 alkynylaryl, C 1 -C 6 alkylheterocyclic, C 2 -C 6 alkenylheterocyclic, C 1 -C 6 alkylcycloalkyl, and C 1 -C 6 alkyl-O—C 1 -C 6 alkylaryl, wherein each cycloalkyl, aryl, or heterocyclic group is optionally substituted with 1-3 groups independently selected from the group consisting of hydroxy, oxo, —SC 1 -C 6 alkyl, C 1 -C 6 alkyl, C 1 -C 6 alkenyl, C 1 -C 6 alkynyl, C 1 -C 6 haloalkyl, halogen, C 1 -C 6 alkoxy, aryloxy, C 1 -C 6 alkenyloxy, C 1 -
  • R 5 is selected from a group consisting of: hydrogen, hydroxy, halogen, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 1 -C 6 alkoxy, aryloxy, —OC 2 -C 6 alkenyl, —C 1 -C 6 haloalkyl, C 1 -C 6 haloalkyl, C 3 -C 8 cycloalkyl, C 1 -C 6 alkylaryl, C 1 -C 6 alkylheterocyclic, C 2 -C 6 alkenylaryl, C 2 -C 6 alkenylheterocyclic, aryl, heterocyclic, cyano, nitro, C 0 -C 6 alkylNR 7 R 8 , C 0 -C 6 alkylCOR 7 , C 0 -C 6 alkylCO 2 R 7 , C 0 -C 6 alkylCONR 7 R 8
  • R 6 is independently selected from a group consisting of: hydrogen, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, hydroxy, COR 7 , C 1 -C 6 alkoxy, aryloxy, —OC 2 -C 6 alkenyl, —OC 1 -C 6 haloalkyl, C 1 -C 6 alkylNR 7 R 8 , C 3 -C 8 cycloalkyl, heterocyclic, aryl, C 1 -C 6 alkyl-O—C(O)NR 7 R 8 , C 1 -C 6 alkyl-NR 7 C(O)NR 7 R 5 and C 1 -C 6 alkylcycloalkyl;
  • R 7 and R 8 are each independently selected from a group consisting of: hydrogen, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, —OC 1 -C 6 alkyl, C 1 -C 6 haloalkyl, —O-aryl, —OC 3 -C 8 cycloalkyl, —O-heterocyclic, —NR 7 R 8 , C 1 -C 6 alkylcycloalkyl, —OC 1 -C 6 alkylcycloalkyl, —OC 1 -C 6 alkylheterocyclic, C 1 -C 6 alkylheterocyclic, —OC 1 -C 6 alkylaryl, C 3 -C 8 cycloalkyl, heterocyclic, aryl, and C 1 -C 6 alkylaryl, wherein each alkyl, cycloalkyl, heterocyclic or aryl group is optionally substituted
  • R 10 , R 11 , and R 12 are independently selected from a group consisting of: hydrogen, C 1 -C 6 alkyl, C 1 -C 6 alkenyl, C 3 -C 8 cycloalkyl, heterocyclic, aryl, C 1 -C 6 alkylaryl, wherein each alkyl, aryl, cycloalkyl, and heterocyclic group is optionally substituted with 1-3 groups independently selected from halogen, C 1 -C 6 alkylheterocyclic, and C 1 -C 6 haloalkyl, or R 11 and R 12 combine to form a nitrogen containing heterocyclic ring which may have 0, 1, or 2 additional heteroatoms selected from oxygen, nitrogen or sulfur and is optionally substituted with oxo, C 1 -C 6 alkyl, COR 7 , and —SO 2 R 7 ;
  • the present invention also provides a method for modulating CETP activity comprising the use of a compound of Formula I or a pharmaceutically acceptable salt, solvate, enantiomer, racemate, diastereomer or mixture of diastereomers thereof, for the treatment, prevention or amelioration of CETP mediated diseases.
  • the present invention provides a method for treating or preventing dyslipidemia comprising administering a compound of Formula I, pharmaceutically acceptable salt, solvate, enantiomer, racemate, diastereomer, mixture of diastereomers, or prodrug thereof, to a patient in need thereof.
  • the present invention provides a method for treating or preventing CHD comprising administering a compound of Formula I, pharmaceutically acceptable salt, solvate, enantiomer, racemate, diastereomer, mixture of diastereomers, or prodrug thereof, to a patient in need thereof.
  • the present invention provides a method for treating and/or preventing atherosclerosis comprising administering a compound of Formula I, pharmaceutically acceptable salt, solvate, enantiomer, racemate diastereomer, mixture of diastereomers, or prodrug thereof, to a patient in need thereof.
  • the present invention provides a method for treating and/or preventing diseases related to abnormal CETP activity comprising administering a compound of Formula I, pharmaceutically acceptable salt, solvate, enantiomer, racemate diastereomer, mixture of diastereomers, or prodrug thereof, to a patient in need thereof.
  • the present invention provides a method of raising the ratio of plasma HDL-cholesterol to plasma LDL-cholesterol in a mammal comprising administering a therapeutically effective dose of a compound of Formula I, pharmaceutically acceptable salt, solvate, enantiomer, racemate, diastereomer, mixture of diastereomers, or prodrug thereof, to a patient in need thereof.
  • the present invention provides a method of raising the level of plasma HDL-cholesterol in a mammal comprising administering a therapeutically effective dose of a compound of Formula I, pharmaceutically acceptable salt, solvate, enantiomer, racemate, diastereomer, mixture of diastereomers, or prodrug thereof, to a patient in need thereof.
  • the present invention provides a method of lowering the level of plasma LDL-cholesterol in a mammal comprising administering a therapeutically effective dose of a compound of Formula I, pharmaceutically acceptable salt, solvate, enantiomer, racemate, diastereomer, mixture of diastereomers, or prodrug thereof, to a patient in need thereof.
  • the present invention also provides a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of Formula I or a pharmaceutically acceptable salt, solvate, enantiomer, racemate, diastereomer or mixture of diastereomers thereof, and a carrier.
  • the present invention also provides a method of treating and/or preventing the pathological sequelae due to low levels of plasma HDL and/or high levels of LDL-cholesterol in a mammal comprising administering an effective dose of a compound of Formula I, pharmaceutically acceptable salt, solvate, enantiomer, racemate, diastereomer, or mixture of diastereomers, thereof, to a patient in need thereof.
  • the present invention also relates to the use of a compound of Formula I for the manufacture of a medicament for treating and/or preventing atherosclerosis in a mammal comprising administering an effective dose of a compound of Formula I, pharmaceutically acceptable salt, solvate, enantiomer, racemate, diastereomer, mixture of diastereomers, or prodrug thereof, to a patient in need thereof.
  • the present invention also provides a combination therapy involving a compound of Formula I and one or more other cardio protective agents such as for example, statins, leptin, and/or other LXR, CETP, ABC A1, and/or lipid regulating agents useful for the treatment and/or prevention of atherosclerosis.
  • cardio protective agents such as for example, statins, leptin, and/or other LXR, CETP, ABC A1, and/or lipid regulating agents useful for the treatment and/or prevention of atherosclerosis.
  • the current invention provides novel compounds of Formula I useful in modulating CETP activity.
  • modulation would include, but not be limited to, up-regulation, down-regulation, inhibition, agonism, antagonism of the CETP receptor as appropriate to achieve HDL raising, or LDL lowering and the resulting biological sequelae from such intervention.
  • diseases or “diseases related to CETP modulation” or “diseases mediated by CETP activity” refers to pathological states where atherosclerosis and cardiovascular diseases are prone because of dyslipidemia and/or other risk factors and are therefore beneficially affected by down-regulation or modulation of CETP activity.
  • diseases include but are not limited to hyperlipidemia and its sequelae such as atherosclerosis, CHD, elevated blood pressure, CHF, stroke, hypertension, hypertriglyceremia, diabetes, obesity, inflammatory diseases including but not limited to dermatitis, arthritis, and pain, and diseases of the central nervous system including but not limited to dementia, cognitive disorders such as Alzheimer's disease.
  • treatment bears its usual meaning which includes prohibiting, inhibiting, ameliorating, halting, restraining, slowing or reversing the progression, or reducing the severity of a pathological symptom related to or resultant from the modulation of CETP activity, especially as related to raising plasma levels of HDL, or lowering LDL-cholesterol levels or raising the HDL/LDL ratio or controlling atherosclerosis, hyperlipidemia and/or hypercholesterolemia.
  • C 1-6 alkyl or “(C 1 -C 6 ) alkyl” or “C 1 -C 6 alkyl” refers to a straight or branched aliphatic chain of 1 to 6 carbon atoms including but not limited to methyl, ethyl, propyl, iso-propyl, n-butyl, tert-butyl, pentyl, and hexyl.
  • alkyl means C 1 -C 6 alkyl.
  • the carbon atom of alkyl groups are attached to the rest of the referenced molecule.
  • C 0 -C 6 alkyl implies an alkyl group as indicated wherein when the term Co applies, the alkyl group is not present, and the remaining groups attach directly to the rest of the referenced molecule.
  • alkenyl and alkynyl for example, a C 2 -C 6 alkenyl group (or a C 2 -C 6 alkynyl group) as used herein mean that the respective groups can include 1, 2, or 3 double bonds (or triple bonds). If more than one double or triple bond is present in the group, the double and triple bonds can be conjugated or non-conjugated.
  • substituted phenyl or “optionally substituted phenyl” refers to a phenyl group having one or more substituents. Preferred substituents are selected from the group consisting of: C 1 -C 6 alkyl, C 1 -C 6 alkoxy, hydroxy, —COOR 7 , C 0 -C 6 alkylNR 7 R 8 , nitro, chloro, fluoro, bromo, iodo, C 1 -C 6 haloalkyl, C 1 -C 6 haloalkoxyalkyl, C 0 -C 6 alkylheterocyclic.
  • optionally substituted 5-7 member carbocyclic or “optionally substituted 5-7 member heterocyclic” whether written in the conjunctive or disjunctive style, or in single or in compound sentences, mean a carbocyclic or heterocyclic 5-7 member ring that is optionally substituted with 1-3 groups independently selected from the group consisting of hydroxy, halo (F, Cl, Br or I), C 1 -C 6 haloalkyl, C 3 -C 8 cycloalkyl, C 1 -C 6 alkylaryl, C 1 -C 6 alkylheterocyclic, aryl, heterocyclic, C 0 -C 3 alkylcyano, nitro, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 1 -C 6 alkoxy, aryloxy, —OC 2 -C 6 alkenyl, —OC 1 -C 6 haloalkyl, C 0 -C 6 alkylNR 7 R 8
  • optionally substituted in general means that the subject group may be substituted, where possible, with 1-3 groups independently selected from the group consisting of: hydroxy, halogen, C 1 -C 6 haloalkyl, C 3 -C 8 cycloalkyl, C 1 -C 6 alkylaryl, C 1 -C 6 alkylheterocyclic, aryl, heterocyclic, CO—C 3 alkylcyano, nitro, C 1 -C 6 alkyl, C 2 -C 6 alkenyl C 1 -C 6 alkoxy, aryloxy, —OC 2 -C 6 alkenyl, —OC 1 -C 6 haloalkyl, C 0 -C 6 alkylNR 7 R 8 , C 0 -C 6 alkylCOR 7 , C 0 -C 6 alkylCO 2 R 7 , C 0 -C 6 alkylCONR 7 R 8 , CONR 7 SO 2 R 8 , —NR
  • aryl refers to a substituted or unsubstituted aromatic or heteroaromatic, or heterocyclic radical.
  • Illustrative aryl groups include but is not limited to napthyl, quinolyl, tetrahydroquinolyl, indazolyl, pyrimidinyl, triazinyl, pyrazine, pyridazinyl, piperidyl, pyrrolidinyl, piperazinyl, morpholinyl, tetrahydrofuranyl, pyranyl, tetrazolyl, imidazolyl, 1,2,3-trazolyl, 1,2,4-triazolyl, oxadiazolyl, thiadiazolyl, thiazolyl, oxazolyl, isoxazolyl, isothiazolyl, pyrazolyl, imidazopyridine, benzimidazolyl, triazolone-yl, imidazolone-yl, imidazo
  • carrier refers to a cyclic group having only carbon and appropriate number of hydrogen atoms.
  • the term encompasses groups such as cycloalkyl, cycloalkene, cycloalkylene, naphthyl, phenyl and the like.
  • heterocycle refers to a 5, 6 or 7 member ring, which may be saturated, partially unsaturated or aromatic mono-cyclic or part of a fused bicyclic ring, and can contain 1-5 heteroatoms selected from N, S, or O, and can optionally be substituted at the ring carbon or nitrogen atom(s) unless otherwise specified.
  • Preferred heterocyclic groups include pyrrolidinyl, piperidinyl, hexamethyleneimino, morpholino, thiomorpholino, benzothiophene, indolyl, quinolyl, isoquinolyl, tetrazolyl, and pyridinyl.
  • alkylheterocyclic or “alkylheterocycle” is understood to mean that the alkyl group is attached to the heterocycle and the point of attachment to the rest of the referenced molecule is the alkyl group.
  • haloalkyl refers to an alkyl (as noted above) substituted with one or more halo atoms selected from F, Br, Cl, and I.
  • haloalkoxyalkyl as used herein include for example trifluoromethoxy, pentafluoroethoxy, trifluoroethoxy (OCH 2 CF 3 ) and the like.
  • Prodrugs describes derivatives of the compounds of the invention that have chemically or metabolically cleavable groups and become by solvolysis or under physiological conditions the compounds of the invention, which are pharmaceutically active, in vivo.
  • Derivatives of the compounds of this invention have activity in both their acid and base derivative forms, but the acid derivative form often offers advantages of solubility, tissue compatibility, or delayed release in a mammalian organism (see, Bundgard, H., Design of Prodrugs, pp. 7-9, 21-24, Elsevier, Amsterdam 1985).
  • Prodrugs include acid derivatives, such as, esters prepared by reaction of the parent acidic compound with a suitable alcohol, or amides prepared by reaction of the parent acid compound with a suitable amine.
  • Simple aliphatic esters e.g., methyl, ethyl, propyl, isopropyl, butyl, sec-butyl, tert-butyl
  • aromatic esters derived from acidic groups pendent on the compounds of this invention are preferred prodrugs.
  • Other preferred esters include morpholinoethyloxy, diethylglycolamide and diethylaminocarbonylmethoxy.
  • double ester type prodrugs such as (acyloxy) alkyl esters or ((alkoxycarbonyl)oxy)alkyl esters.
  • protecting group refers to a group useful for masking reactive sites in a molecule to enhance the reactivity of another group or allow reaction at another desired site or sites following which the protecting group may be removed.
  • Protecting groups are usually used to protect or mask groups including but not limited to —OH, —NH, and —COOH. Suitable protecting groups are known to one of skill in the art and are described in Protecting groups in Organic Synthesis, 3 rd edition, Greene, T. W.; Wuts, P. G. M. Eds., John Wiley and Sons, New York, 1999.
  • solvate refers to a crystal (or crystals) of a compound of the invention formed to include a stoichiometric or non-stoichiometric amount of the compound of Formula I and a solvent molecule.
  • Typical solvating solvents include for example, water, methanol, ethanol, acetone and dimethylformamide.
  • the solvent is water
  • hydrate for a stoichiometric or non-stoichiometric amount of compound and water (or hemi-hydrate for half the stoichiometric amount of water) may optionally be used.
  • salts may be formed which are more water soluble and/or more physiologically suitable than the parent compound.
  • Representative pharmaceutically acceptable salts include but are not limited to, the alkali and alkaline earth salts such as lithium, sodium, potassium, calcium, magnesium, aluminum and the like. Salts are conveniently prepared from the free acid by treating the acid in solution with a base or by exposing the acid to an ion-exchange resin.
  • Base addition salts include for example, ammonium, quaternary ammonium, and amine cations, derived from nitrogenous bases of sufficient basicity to form salts with the compounds of this invention (see, for example, S. M. Berge, et al., “Pharmaceutical Salts,” J. Phar. Sci., 66: 1-19 (1977)).
  • the basic group(s) of the compound of the invention may be reacted with suitable organic or inorganic acids to form salts such as acetate, benzenesulfonate, benzoate, bicarbonate, bisulfate, bitartrate, borate, hydrobromide, camsylate, carbonate, clavulanate, citrate, chloride, edetate, edisylate, estolate, esylate, fluoride, fumarate, gluceptate, gluconate, glutamate, glycolylarsanilate, hexylresorcinate, hydrochloride, hydroxynaphthoate, hydroiodide, isothionate, lactate, lactobionate, laureate, maleate, mandelate, mesylate, methylbromide, methylnitrate, methylsulfate, mucate, napsylate, nitrate, oleate, oxalate, palmitate, pan
  • Preferred salts for the purpose of the invention include the hydrochloride salt, the hydrobromide salt, the bisulfate salt, the methane sulfonic acid salt, the p-toluenesulfonic acid salt, bitartrate, the acetate and the citrate salt.
  • a compound of the invention as illustrated by Formula I may occur as any one of its positional isomers, stereochemical isomers or regio-isomers, all of which are within the scope of the present invention of the invention.
  • Certain compounds of the invention may possess one or more chiral centers, and thus, may exist in optically active forms.
  • the compounds contain an alkenyl or alkenylene group, there exist the possibility of cis and trans isomeric forms of the compounds.
  • the R— and S— isomers and mixtures thereof, including racemic mixtures as well as mixtures of enantiomers or cis- and trans-isomers, are contemplated by this invention. Additional asymmetric carbon atoms can be present in a substituent group such as an alkyl group.
  • stereoisomer it can be prepared by methods well known in the art by using stereo-specific reactions with starting materials that contain the asymmetric centers and are already resolved.
  • desired stereoisomers may be prepared by methods that lead to mixtures of the stereoisomers and subsequent resolution by known methods.
  • a racemic mixture may be reacted with a single enantiomer of some other compound i.e. a chiral resolving agent. This changes the racemic form into a mixture of stereoisomers and diastereomers, because they have different melting points, different boiling points, and different solubilities and can be separated by conventional means, such as crystallization.
  • n is 0, or 1. More preferably, n is 0.
  • q is 0, 1 or 2. More preferably q is 1 or 2. Most preferably, q is 1.
  • Y is a bond or C(O);
  • a preferred R 1 group is selected from the group consisting of: hydroxy, —Oaryl, —OC 1 -C 6 haloalkyl, —OC 1 -C 6 alkylcycloalkyl, —OC 3 -C 8 cycloalkyl, —OC 1 -C 6 alkylcycloalkylNR 7 R 8 , —OC 1 -C 6 alkyl, —OC 0 -C 6 alkylaryl, —OC 1 -C 6 alkylcyano, —OC 1 -C 6 alkylCO 2 R 11 , —OC 3 -C 8 cycloalkylCO 2 R 11 , —OC 1 -C 6 alkylhydroxy, —OC 1 -C 6 alkylNR 7 R 8 and —OC 0 -C 6 alkylheterocyclic; provided that R 1 is not —OH when Y is —S(O) t ; and wherein each alkyl, cycloalkyl,
  • R 3a and R 3b groups are independently selected from the group consisting of: hydrogen, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, and C 2 -C 6 alkynyl. More preferably, R 3a and R 3b are each independently selected from hydrogen and C 1 -C 6 alkyl.
  • R 4 is NR 4a R 4b
  • R 4a is selected from the group consisting of:
  • the R groups are independently selected from the group consisting of: C 3 -C 6 alkyl, C 1 -C 6 alkylalcohol, C 3 -C 6 alkoxy, C 0 -C 6 alkylcycloalkyl, C 0 -C 6 alkylheterocyclic, C 1 -C 6 alkylCN, C 3 -C 6 haloalkyl, C 0 -C 6 alkylNR 11 R 12 wherein the C 1 -C 6 alkyl group (of C 1 -C 6 alkylNR 11 R 12 ) is optionally substituted with —OR 10 or C(O)OR 10 , C 1 -C 6 alkylC(O)NR 11 R 12 , and C 1 -C 6 alkylC(O)OR 11 provided that the R groups are not hydrogen, methyl, or ethyl.
  • the R 11 and R 12 groups are as described below. More preferable the R groups are independently selected from the group consisting of: C 3 -C 6 alkyl, C 2 -C 6 alkylNH 2 , C 2 -C 6 alkylalcohol, C 1 -C 6 alkylcyano, C 1 -C 6 alkylC(O)NH 2 , again provided that the R groups are not hydrogen, methyl or ethyl.
  • R 4b is selected from: C 1 -C 6 alkylaryl, C 1 -C 6 alkylheterocyclic, wherein the heterocyclic and aryl groups are optionally substituted with 1-3 groups selected from the group consisting of: hydroxy, oxo, cyano, —SC 1 -C 6 alkyl, C 1 -C 6 alkyl, C 1 -C 6 alkenyl, C 1 -C 6 alkynyl, C 1 -C 6 haloalkyl, halogen, and —OC 1 -C 6 alkyl.
  • R 4b is benzyl mono or disubstituted with a C 1 -C 6 haloalkyl, halogen and C 1 -C 3 alkyl. Most preferred for R 4b is 3,5-bistrifluorobenzyl.
  • R 5 is preferably selected from a group consisting of hydrogen, halo, C 1 -C 6 alkyl, C 1 -C 6 haloalkyl, —OC 1 -C 6 alkyl, —Oaryl, —OC 2 -C 6 alkenyl, —OC 1 -C 6 haloalkyl, —CH 2 NR 7 R 8 , —NH 2 , —N(C 1 -C 4 alkyl) 2 , —CN, and —NO 2 .
  • any two R 5 groups which combine to form an optionally substituted 5, 6, or 7-member fused ring with the phenyl ring to which they are attached, wherein the 5, 6, or 7-member ring is saturated, partially unsaturated, or fully unsaturated and optionally contains 1, 2, or 3 heteroatoms independently selected from O, N, and S.
  • R 7 and R 8 are independently selected from a group consisting of: hydrogen, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 1 -C 6 alkylaryl, and C 1 -C 6 alkylheterocyclic, wherein each aryl group is optionally substituted with 1-3 groups independently selected from C 1 -C 6 alkyl, halo, and C 1 -C 6 haloalkyl.
  • R 10 , R 11 and R 12 are independently selected from a group consisting of: hydrogen, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 1 -C 6 alkylaryl, and C 1 -C 6 alkylheterocyclic, wherein each alkyl and aryl group is optionally substituted with 1-3 groups independently selected from C 1 -C 6 alkyl, halo, and C 1 -C 6 haloalkyl.
  • Particularly preferred compounds of the invention are selected from the group consisting of: (2R,4S)-4-[(3,5-bistrifluoromethylbenzyl)-(2-propyl-2H-tetrazol-5-yl)amino]-2-ethyl-6-trifluoromethyl-3,4-dihydro-2H-quinoline-1-carboxylic acid isopropyl ester,
  • geometric isomers associated with the double bonds and the optical isomers associated with asymmetric carbon atoms of compounds of Formula I are also contemplated to be within the scope of the current invention as useful for the treatment of diseases related to CETP modulation.
  • the compounds of the instant invention can be synthesized as exemplified in the following Schemes, Examples, and Procedures.
  • Anthranilate intermediates of Formula 1 can be chemically prepared, for example, by following the synthetic routes set forth in the Schemes below.
  • the following discussion is not intended to limit the scope of the present invention in any way because one of skill in the art is able to extrapolate without undue experimentation from the schemes and examples herein to other specific compounds within the scope of the invention.
  • Many of the reagents and starting materials can be readily obtained from commercial suppliers or are readily available to one of ordinary skill in the art.
  • reagents and starting materials may be made by procedures which are selected from standard techniques of organic and heterocyclic chemistry, techniques which are analogous to the syntheses of known similar reagents or starting materials, and the procedures described in the preparations and examples below, including any novel procedures. This includes, but is not limited to, esterification of a carboxylic acid, hydrolysis of a nitrile to a carboxylic acid, and subsequent esterification.
  • R, R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , etc, designations used within immediately following section are for the purpose of illustrating the various methods of synthesizing compounds of the invention and/or illustrating variability of substituents at the pendent position and are not necessarily synonymous in scope or meaning with similar groups used in the generic structure for compounds of Formula I. However, groups in final compounds of the schemes occupying similar positions are co-extensive in scope and meaning compared to groups occupying similar positions as defined for the generic structure of compounds of Formula I.
  • the nucleophilic aromatic substitution occurs by methods known in the art, (Wells, K. M. et al. Tetrahedron Letters, 1996, 37(36), 6439-6442).
  • the reaction proceeds at 0° C. to elevated temperatures (up to or about 150° C.) in anywhere from ten minutes to several days depending on the stability of the starting materials and/or reaction conditions.
  • Synthetic scheme 5 shows preparation of compounds of Formula I.
  • substituted arylamino esters 1 that are either commercially available or prepared as set forth in the literature or in Schemes 1 to 4 can be protected with tosyl chloride, isopropyl chloroformate, or other suitable protecting group to provide 54.
  • the compound 54 may, in turn, be alkylated with appropriately substituted, or unsubstituted 3-bromoethylesters 55 thus affording 56.
  • Dieckmann condensation-cyclization of intermediate 56 yields N-protected tetrahydroquinoline 57, which is subjected to acid hydrolysis and decarboxylation to afford ketone derivatives 58. Removal of the protecting group, if necessary, with acid (e.g.
  • N-acylation of 63 by treatment with an appropriately substituted aryl or alkyl chloroformate in the presence of an organic base such as pyridine affords carbamates of structure 64.
  • treatment of 63 with an acid chloride or an appropriate activated ester affords compounds of formula 64.
  • compound 64 can be obtained as is shown in Scheme 6, by addition of a Grignard reagent to compound 60 followed by hydrolysis in acid media. Or, Michael addition of aniline derivatives 62 to ⁇ , ⁇ -unsaturated carboxylic acid or ester, followed by cyclization in acid media, can afford compound 63.
  • the intermediate tetrahydroquinoline-4-ones 64 may be reduced with a reducing agent such as sodium borohydride in an appropriate solvent, such as tetrahydrofuran or methanol, to achieve the benzylic alcohol 65 as shown in Scheme 6.
  • a reverse procedure for forming the disubstituted amine is shown in Scheme 8. Formation of a Schiff base of a tetrahydroquinoline-4-one (64) with a benzylic amine is followed by treatment with a reducing agent such as sodium borohydride in an appropriate solvent, such as tetrahydrofuran or methanol, to achieve the disubstituted benzylic amine adduct 69. Further elaboration by reaction with an activated heterocylic reagent in the presence of base (or alternatively, Schiff base formation with a heteroaromatic aldehyde followed by reduction) provides a secondary route to disubstituted amine products 67.
  • a reducing agent such as sodium borohydride
  • an appropriate solvent such as tetrahydrofuran or methanol
  • Compounds of Formula I may also be prepared by transformation of pendant functionality as shown in Scheme 9.
  • Scheme 9 shows that disubstituted amine products such as 68 in which the moiety R-A corresponds to reactive functionality such as cyano, carboxylate, or like group may be transformed into heterocyclic moieties such as 71 in intermediate stages of synthesis or at the end of the synthetic preparation. Also, the order of N-substitution may be reversed as shown above.
  • Scheme 9a shows a few examples of transformation reactions to illustrate inter-conversion of functionalities as means of preparing compounds of the invention. Detailed procedures are disclosed in the Examples, known to one of skill in the art, or are readily available from reference sources by one of skill in the art.
  • compound 64 can be treated with a base such as sodium hydride or lithium diisopropylamide or lithium bis(trimethylsilyl)amide in a solvent such as DMF or tetrahydrofuran.
  • a base such as sodium hydride or lithium diisopropylamide or lithium bis(trimethylsilyl)amide
  • a solvent such as DMF or tetrahydrofuran.
  • Alkylation with the appropriately substituted halide or mesylate or tosylate may form compound 79 where R 3a and R 3b can be the same or different.
  • Conversion of 79 to 67 is as described, for example above in Scheme 10.
  • compound 73d may be hydrolyzed to the corresponding amine 80, and may be further acylated using standard procedures by one skilled in the art to provide 73d.
  • 80 can be treated with triphosgene or trichloromethyl-choroformate to provide 81.
  • Compound 81 can afford compound 73d by reaction with the appropriate alcohols.
  • compound 80 can be alkylated by methods known in the art such as treating 80 with base and an alkyl halide, tosylate or the like, to afford 82.
  • compound 82 can be obtained using reductive amination conditions.
  • tetrazole 83 can be alkylated with the appropriate protected aminoalcohol under Mitsunobu conditions or with the appropriate protected aminoalkylbromide, iodide or mesylate, (e.g. where P1 is the protecting group) or the like in the presence of base to provide a protected aminoalkyltetrazole 84. Removal of P1 using methods well known in the art can yield compound 85.
  • tetrazole 83 can be alkylated with the appropriate alkylcyano bromide or with the appropriate acrylonitrile under Michael reaction conditions. Cyano derivative 86 can be then reduced to the corresponding amine 85.
  • Tetrazole 83 can be alkylated using the appropriate alcohol under Mitsunobu conditions, or with the appropriate alkyl halide or the like in the presence of base to provide 87. Removal of P1 using methods well known in the art can yield compound 88. Alternatively hydroxyalkyltetrazole 88 can be obtained by alkylation of 83 with the corresponding halide in the presence of base.
  • SPA in vitro Scintillation proximity assay
  • This assay monitors the inhibition of the transfer of [ 3 H]cholesterol esters from HDL (Amersham) to biotinylated LDL (Amersham) by a CETP source.
  • the CETP source for this assay can be produced by AV-12 cells that have been created to express human CETP.
  • the radiolabeled cholesterol ester is transferred in a HEPES-NaCl based buffer, after thirty minutes incubation the reaction is stopped and the biotinylated LDL is bound to streptavidin/scintillant coated SPA beads (Amersham).
  • the radioactive signal is measured in a Packard 96-well scintillation TopCounter with window settings fully open. A decrease in radioactive signal from the LDL relative to a standard indicates the ability of compounds to inhibit the activity of CETP.
  • Preferred compounds of the invention evaluated according to this assay protocol exhibit CETP inhibition at concentrations of less than 100 micromolar.
  • CETP sources can be used to mediate the transfer of radiolabeled cholesterol ester in this assay.
  • endogenous CETP from human plasma, CETP from mice that express human CETP, and endogenous CETP from hamsters can be used as the CETP source in this assay.
  • Buffers other than HEPES-NaCl based buffer can be used in this assay, for example, human plasma, mouse plasma or a Tris-buffer that is high in albumin may be used.
  • radio labeled-LDL may be used in this assay.
  • Test compounds are administered orally in selected aqueous or oil based vehicles for up to one week. At various times after dosing, ranging from 4 h to 48 h, blood/plasma can be obtained.
  • the CETP activity can be determined by a method similar to that described above for the in vitro CETP activity assay, with the modification that plasma from the treated animals is used as the CETP source in the assay.
  • a strain of transgenic mice that express human CETP can also be used to test compounds of this invention.
  • Test compounds can be administered orally in selected aqueous or oil based vehicles for up to one week. At various times after dosing, ranging from 4 h to 48 h, blood/plasma can be obtained.
  • the CETP activity can be determined by a method similar to that described above for the in vitro CETP activity assay, with the modification that plasma from the treated animals is used as the CETP source in the assay.
  • a strain of transgenic mice that express both human CETP and human apolipoprotein A-1 can be used to test compounds of this invention.
  • Test compounds can be administered orally in selected aqueous or oil based vehicles for up to one week. At various times after dosing, ranging from 4 h to 48 h, blood/plasma is obtained.
  • CETP activity can be determined by a method similar to that described for the in vitro CETP activity assay, with the modification that plasma from the treated animals is used as the CETP source in the assay.
  • Activity of compounds of this invention in vivo can be evaluated by comparing the level of elevation of HDL cholesterol relative to a control by a given amount of a compound in a CETP-containing animal species.
  • a strain of transgenic mice that express both human CETP and human apolipoprotein A-1 can be used to evaluate compounds of this invention.
  • Test compounds are administered to the animals once orally in selected aqueous or oil based vehicles. At various times after dosing, ranging from 4 h to 24 h, blood is obtained. The blood is allowed to clot, and serum is obtained from the clotted blood by centrifugation.
  • the HDL cholesterol levels in the serum can be determined by known procedures using HDL-C plus reagents (Roche/Hitachi, Indianapolis, Ind.) with a clinical chemistry analyzer (Roche/Hitachi, Indianapolis, Ind.). Additional serum lipids can be analyzed by enzymatic methods. Lipids in the VLDL, LDL, and HDL fractions are analyzed by enzymatic methods after precipitation or size exclusion chromatography. An example of the elevation of HDL cholesterol levels at 8 hr is summarized in Table 1.
  • the efficacy of compounds of the invention in vivo can also be evaluated utilizing Syrian Golden Hamsters.
  • the compounds can be tested in hamsters made hypercholesterolemic by feeding a high fat high cholesterol diet for a minimum of two weeks or in non-hypercholesterolemic hamsters fed normal chow for two weeks.
  • Test compounds can be administered orally in selected aqueous or oil based vehicles for up to 1 week.
  • Serum from the animals can be obtained, and lipids can be analyzed by enzymatic methods. Lipids in the VLDL, LDL, and HDL fractions can be analyzed by known enzymatic methods after precipitation or size exclusion chromatography.
  • a strain of transgenic mice that expresses human CETP can be used to test the efficacy of the compounds of this invention.
  • the hCETP mice can be made hypercholesterolemic by feeding a high fat chow diet such as TD 88051, as described by Nishina et al. (J Lipid Res., 31, 859-869 (1990)) for at least two weeks before the start of the study.
  • Test compounds can be administered orally to the animals in selected aqueous or oil based vehicles for up to 1 week.
  • Serum can be obtained from the animals.
  • Lipids from the serum can be analyzed by enzymatic methods. Lipids in the VLDL, LDL and HDL fractions are analyzed by enzymatic methods after precipitation or size exclusion chromatography.
  • the term “effective amount” means an amount of compound of the present invention, i.e., Formula I, which is capable of alleviating the symptoms of the various pathological conditions herein described.
  • a specific dose of a compound administered according to this invention will, of course, be determined by the particular circumstances surrounding the case including, for example, but not limited to: the compound administered, the route of administration, the state of being of the patient, and the pathological condition being treated.
  • a typical daily dose will contain a nontoxic dosage level of from about 0.01 mg to about 1000 mg/day of a compound of the present invention.
  • Preferred daily doses generally will be from about 1 mg to about 250 mg/day.
  • the compounds of this invention may be administered by a variety of routes including oral, rectal, transdermal, subcutaneous, intravenous, intramuscular, and intranasal. These compounds preferably are formulated prior to administration, the selection of which will be decided by the attending physician.
  • another aspect of the present invention is a pharmaceutical composition comprising an effective amount of a compound of Formula I, or a pharmaceutically acceptable salt thereof, solvate, prodrug, enantiomer or prodrug thereof, and a pharmaceutically acceptable carrier, diluent, or excipient.
  • the total active ingredients in such formulations comprises from 0.1% to 99.9% by weight of the formulation.
  • pharmaceutically acceptable means that the carrier, diluent, excipients and salt are compatible with the other ingredients of the formulation and not deleterious to the recipient thereof.
  • compositions of the present invention may be prepared by procedures known in the art using well-known and readily available ingredients.
  • the compounds of Formula I can be formulated with common excipients, diluents, or carriers, and formed into tablets, capsules, suspensions, powders, and the like.
  • Non limiting examples of excipients, diluents, and carriers that are suitable for such formulations include the following: fillers and extenders such as starch, sugars, mannitol, and silicic derivatives; binding agents such as carboxymethyl cellulose and other cellulose derivatives, alginates, gelatin, and polyvinyl-pyrrolidone; moisturizing agents such as glycerol; disintegrating agents such as calcium carbonate and sodium bicarbonate; agents for retarding dissolution such as paraffin; resorption accelerators such as quaternary ammonium compounds; surface active agents such as cetyl alcohol, glycerol monostearate; adsorptive carriers such as kaolin and bentonite; and lubricants such as talc, calcium, and magnesium stearate, and solid polyethyl glycols.
  • fillers and extenders such as starch, sugars, mannitol, and silicic derivatives
  • binding agents such as carboxymethyl cellulose and other cellulose derivative
  • the compounds also may be formulated as elixirs or solutions for convenient oral administration or as solutions appropriate for parenteral administration, for example, by intramuscular, subcutaneous or intravenous routes. Additionally, the compounds are well suited to formulation as sustained release dosage forms and the like.
  • the formulations can be so constituted that they release the active ingredient only or preferably in a particular physiological location, possibly over a period of time.
  • the coatings, envelopes, and protective matrices may be made, for example, from polymeric substances or waxes.
  • Compounds of the invention may be formulated following one or more of the formulation examples, procedures, protocols or mixing ratios below.
  • Active Ingredient as used herein means a compound of Formula I, a salt, solvate, racemate, enantiomer diastereomer, mixture of diastereomers, prodrug thereof, or a combination of a compound of Formula I and other effective agents for the treatment or prevention of dyslipidemia, atherosclerosis, or other co-morbid conditions and symptoms.
  • Hard gelatin capsules can be prepared according to the following:
  • Ingredient Quantity (mg/capsule) Active ingredient 0.1-1000 Starch, NF 0-650 Starch flowable powder 0-650 Silicone fluid 350 centistokes 0-15
  • a tablet formulation each tablet containing 2.5-1,000 mgs of active ingredient can be prepared according to the following:
  • Ingredient Quantity (mg/tablet) Active ingredient 2.5-1000 Cellulose, microcrystalline 200-650 Silicon dioxide, fumed 10-650 Stearate acid 5-15 The components are blended and compressed to form tablets.
  • Active ingredient 25-1000 Starch 45 Cellulose, microcrystalline 35 Polyvinylpyrrolidone 4 (as 10% solution in water) Sodium carboxymethyl cellulose 4.5 Magnesium stearate 0.5 Talc 1
  • the active ingredient, starch, and cellulose are passed through a No. 45 mesh U.S. sieve and thoroughly blended.
  • the solution of polyvinylpyrrolidone is mixed with the blended powders.
  • the resulting mixture is 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.
  • the active ingredient is passed through a No. 45 mesh U.S. sieve and then blended with the sodium carboxymethyl cellulose and syrup to form a smooth paste.
  • the benzoic acid solution, flavor, and color are diluted with an amount of purified water and added, with stirring, to the paste. Sufficient purified water is then added to provide the suspension at the desired volume (or concentration).
  • An aerosol solution can be prepared as follows:
  • 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 desired amount is then fed to a stainless steel container and diluted with the remaining propellant.
  • the valve units are then fitted to the container.
  • a solution suitable for intravenous administration can be prepared as follows:
  • a solution comparing the above ingredients can be intravenously administered to a patient at a rate of about 1 mL per minute or as prescribed by a physician.
  • triphenyl phosphine 109 mg, 0.42 mmol
  • diethyl azodicarboxylate 40%, 0.13 mL, 0.42 mmol
  • (+/ ⁇ )-cis 4-[[2-(2-Amino-ethyl)-2H-tetrazol-5-yl]-(3,5-bis-trifluoromethyl-benzyl)-amino]-2-cyclopropyl-6-trifluoromethyl-3,4-dihydro-2H-quinoline-1-carboxylic Acid Isopropyl Ester
  • (+/ ⁇ )-cis 4-[[2-(2-Amino-ethyl)-2H-tetrazol-5-yl]-(3,5-bis-trifluoromethyl-benzyl)-amino]-2-methyl-6-trifluoromethyl-3,4-dihydro-2H-quinoline-1-carboxylic Acid Isopropyl Ester

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Hematology (AREA)
  • Obesity (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Cardiology (AREA)
  • Diabetes (AREA)
  • Urology & Nephrology (AREA)
  • Vascular Medicine (AREA)
  • Epidemiology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Plural Heterocyclic Compounds (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)
US11/570,540 2004-06-24 2005-06-22 Compounds and Methods for Treating Dyslipidemia Abandoned US20080269284A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US11/570,540 US20080269284A1 (en) 2004-06-24 2005-06-22 Compounds and Methods for Treating Dyslipidemia

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US58270804P 2004-06-24 2004-06-24
US62724104P 2004-11-12 2004-11-12
US66486205P 2005-03-24 2005-03-24
PCT/US2005/021789 WO2006012093A1 (fr) 2004-06-24 2005-06-22 Composés et procédés destinés à traiter la dyslipidémie
US11/570,540 US20080269284A1 (en) 2004-06-24 2005-06-22 Compounds and Methods for Treating Dyslipidemia

Publications (1)

Publication Number Publication Date
US20080269284A1 true US20080269284A1 (en) 2008-10-30

Family

ID=35056992

Family Applications (3)

Application Number Title Priority Date Filing Date
US11/570,540 Abandoned US20080269284A1 (en) 2004-06-24 2005-06-22 Compounds and Methods for Treating Dyslipidemia
US11/570,542 Expired - Fee Related US7786108B2 (en) 2004-06-24 2005-06-23 Compounds and method for treating dyslipidemia
US12/763,344 Abandoned US20100204207A1 (en) 2004-06-24 2010-04-20 Compounds and Methods for Treating Dyslipidemia

Family Applications After (2)

Application Number Title Priority Date Filing Date
US11/570,542 Expired - Fee Related US7786108B2 (en) 2004-06-24 2005-06-23 Compounds and method for treating dyslipidemia
US12/763,344 Abandoned US20100204207A1 (en) 2004-06-24 2010-04-20 Compounds and Methods for Treating Dyslipidemia

Country Status (27)

Country Link
US (3) US20080269284A1 (fr)
EP (3) EP1761521A1 (fr)
JP (2) JP2008504266A (fr)
KR (1) KR20070041452A (fr)
CN (1) CN1972932B (fr)
AT (1) ATE528304T1 (fr)
AU (2) AU2005267436A1 (fr)
BR (2) BRPI0512523A (fr)
CA (2) CA2570688A1 (fr)
CR (1) CR8831A (fr)
CY (1) CY1112124T1 (fr)
DK (1) DK1761522T3 (fr)
EA (2) EA200700119A1 (fr)
EC (2) ECSP067095A (fr)
ES (1) ES2372291T3 (fr)
HK (1) HK1104528A1 (fr)
HR (1) HRP20110761T1 (fr)
IL (2) IL180277A (fr)
MA (2) MA28729B1 (fr)
MX (2) MXPA06014716A (fr)
NO (2) NO20070468L (fr)
NZ (1) NZ550978A (fr)
PL (1) PL1761522T3 (fr)
PT (1) PT1761522E (fr)
RS (1) RS52064B (fr)
SI (1) SI1761522T1 (fr)
WO (2) WO2006012093A1 (fr)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070208003A1 (en) * 2004-03-26 2007-09-06 Bell Michael G Compounds and methods for treating dyslipidemia
US20070254869A1 (en) * 2003-10-08 2007-11-01 Eli Lilly And Company Compounds And Methods For Treating Dislipidemia
WO2013008164A2 (fr) 2011-07-08 2013-01-17 Novartis Ag Méthode de traitement de l'athérosclérose chez des patients présentant un taux élevé de triglycérides
US20170182048A1 (en) * 2014-02-05 2017-06-29 Amgen Inc. Cholesteryl Ester Transfer Protein (CETP) Inhibitor and Pharmaceutical Compositions Comprising Said Inhibitor for Use in the Treatment or Prevention of Cardiovascular Diseases

Families Citing this family (29)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
UA90269C2 (ru) 2004-04-02 2010-04-26 Мицубиси Танабе Фарма Корпорейшн Тетрагидрохинолиновые производные и способ их получения
US20080269284A1 (en) 2004-06-24 2008-10-30 Eli Lilly And Company Compounds and Methods for Treating Dyslipidemia
WO2007107843A1 (fr) * 2006-03-22 2007-09-27 Pfizer Products Inc. Procédés de traitement avec des inhibiteurs de la cetp
TW200808731A (en) 2006-03-30 2008-02-16 Tanabe Seiyaku Co A process for preparing tetrahydroquinoline derivatives
BRPI0715160A2 (pt) 2006-08-08 2013-06-11 Sanofi Aventis imidazolidina-2,4-dionas substituÍdas por arilamimoaril-alquil-, processo para preparÁ-las, medicamentos compeendendo estes compostos, e seu uso
DE102007005045B4 (de) 2007-01-26 2008-12-18 Sanofi-Aventis Phenothiazin Derivate, Verfahren zu ihrer Herstellung und ihre Verwendung als Arzneimittel
WO2008143127A1 (fr) 2007-05-17 2008-11-27 Ajinomoto Co., Inc. Procédé de mesure de l'activité d'une enzyme modifiée par un lipide
EP2025674A1 (fr) 2007-08-15 2009-02-18 sanofi-aventis Tetrahydronaphthaline substituée, son procédé de fabrication et son utilisation en tant que médicament
DE102007054497B3 (de) 2007-11-13 2009-07-23 Sanofi-Aventis Deutschland Gmbh Neue kristalline Diphenylazetidinonhydrate und Verfahren zu deren Herstellung
UY31968A (es) 2008-07-09 2010-01-29 Sanofi Aventis Nuevos derivados heterocíclicos, sus procesos para su preparación, y sus usos terapéuticos
WO2010068601A1 (fr) 2008-12-08 2010-06-17 Sanofi-Aventis Hydrate de fluoroglycoside hétéroaromatique cristallin, ses procédés de fabrication, ses procédés d'utilisation et compositions pharmaceutiques le contenant
TWI450896B (zh) 2009-06-30 2014-09-01 Lilly Co Eli 反式-4-〔〔(5s)-5-〔〔〔3,5-雙(三氟甲基)苯基〕甲基〕(2-甲基-2h-四唑-5-基)胺基〕-2,3,4,5-四氫-7,9-二甲基-1h-1-苯并氮呯-1-基〕甲基〕-環己基羧酸
US8785608B2 (en) 2009-08-26 2014-07-22 Sanofi Crystalline heteroaromatic fluoroglycoside hydrates, pharmaceuticals comprising these compounds and their use
EP2582709B1 (fr) 2010-06-18 2018-01-24 Sanofi Dérivés d'azolopyridin-3-one en tant qu'inhibiteurs de lipases et de phospholipases
EP2683698B1 (fr) 2011-03-08 2017-10-04 Sanofi Dérivés benzyl-oxathiazine substitués avec adamantane ou noradamantane, médicaments contenant ces composés et leur utilisation
US8895547B2 (en) 2011-03-08 2014-11-25 Sanofi Substituted phenyl-oxathiazine derivatives, method for producing them, drugs containing said compounds and the use thereof
US8809324B2 (en) 2011-03-08 2014-08-19 Sanofi Substituted phenyl-oxathiazine derivatives, method for producing them, drugs containing said compounds and the use thereof
WO2012120052A1 (fr) 2011-03-08 2012-09-13 Sanofi Dérivés d'oxathiazine substitués par des carbocycles ou des hétérocycles, leur procédé de préparation, médicaments contenant ces composés et leur utilisation
US8871758B2 (en) 2011-03-08 2014-10-28 Sanofi Tetrasubstituted oxathiazine derivatives, method for producing them, their use as medicine and drug containing said derivatives and the use thereof
WO2012120053A1 (fr) 2011-03-08 2012-09-13 Sanofi Dérivés oxathiazine ramifiés, procédé pour leur préparation, utilisation en tant que médicament, agents pharmaceutiques contenant ces dérivés et leur utilisation
EP2683705B1 (fr) 2011-03-08 2015-04-22 Sanofi Dérivés oxathiazine di- et tri-substitués, procédé pour leur préparation, utilisation en tant que médicament, agent pharmaceutique contenant ces dérivés et utilisation
WO2012120058A1 (fr) 2011-03-08 2012-09-13 Sanofi Dérivés d'oxathiazine substitués par des groupes benzyle ou hétérométhylène, leur procédé de production, leur utilisation comme médicament ainsi que produits pharmaceutiques les contenant et leur utilisation
WO2012120055A1 (fr) 2011-03-08 2012-09-13 Sanofi Dérivés oxathiazine di- et tri-substitués, procédé pour leur préparation, utilisation en tant que médicament, agent pharmaceutique contenant ces dérivés et utilisation
EP2567959B1 (fr) 2011-09-12 2014-04-16 Sanofi Dérivés d'amide d'acide 6-(4-hydroxy-phényl)-3-styryl-1h-pyrazolo[3,4-b]pyridine-4-carboxylique en tant qu'inhibiteurs de kinase
PL2860175T3 (pl) * 2012-06-11 2018-07-31 Tacurion Sposób wytwarzania związku 4,4,7-trifluoro-1,2,3,4-tetrahydro-5h-1-benzazepinowego i związku pośredniego do jego syntezy
WO2014012401A1 (fr) * 2012-07-16 2014-01-23 上海恒瑞医药有限公司 Dérivé benzo-hétérocyclique à sept chaînons, son procédé de préparation et ses applications médicales
CN103833637B (zh) * 2014-03-31 2016-01-06 武汉武药制药有限公司 一种制备依塞曲匹(Evacetrapib)中间体的方法
LT3307734T (lt) 2015-06-09 2020-05-11 Abbvie Inc. Branduolinio receptoriaus moduliatoriai (ror), skirti uždegiminių ir autoimuninių ligų gydymui
WO2016198908A1 (fr) 2015-06-09 2016-12-15 Abbvie Inc. Modulateurs des récepteurs nucléaires ror

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4204450A (en) * 1977-09-09 1980-05-27 Machines Et Applications Shear for large-sized scrap
US6140343A (en) * 1998-09-17 2000-10-31 Pfizer 4-amino substituted-2-substituted-1,2,3,4-tetrahydroquinolines
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
US6313142B1 (en) * 1999-11-30 2001-11-06 Pfizer Inc. Method for making 4-carboxyamino-2-substituted-1,2,3,4-tetrahydroquinoline
US6689897B2 (en) * 2001-04-30 2004-02-10 Pfizer Inc. Intermediates of CETP inhibitors

Family Cites Families (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CO5271716A1 (es) * 1999-11-30 2003-04-30 Pfizer Prod Inc Cristales de 4- carboxamino 1,2,3,4-tetrahidroquinolina 2- sustituida
SK2332004A3 (en) 2002-08-30 2004-12-01 Japan Tobacco Inc Dibenzylamine compound and medicinal use thereof
MXPA05003456A (es) 2002-10-04 2005-07-05 Millennium Pharm Inc Antagonistas del receptor de pgd2 para el tratamiento de enfermedades inflamatorias.
US20060100239A1 (en) 2003-03-04 2006-05-11 Izuru Nagasaki Method for producing optically active amines
US20040204450A1 (en) * 2003-03-28 2004-10-14 Pfizer Inc Quinoline and quinoxaline compounds
JP4773969B2 (ja) 2003-10-08 2011-09-14 イーライ リリー アンド カンパニー 脂質代謝異常の治療ための化合物及び方法
DK1732933T3 (da) 2004-03-26 2008-10-27 Lilly Co Eli Forobindelser til behandling af dyslipidæmi
UA90269C2 (ru) * 2004-04-02 2010-04-26 Мицубиси Танабе Фарма Корпорейшн Тетрагидрохинолиновые производные и способ их получения
US20080269284A1 (en) 2004-06-24 2008-10-30 Eli Lilly And Company Compounds and Methods for Treating Dyslipidemia
US7700774B2 (en) 2004-12-20 2010-04-20 Dr. Reddy's Laboratories Ltd. Heterocyclic compounds and their pharmaceutical compositions
TWI450896B (zh) * 2009-06-30 2014-09-01 Lilly Co Eli 反式-4-〔〔(5s)-5-〔〔〔3,5-雙(三氟甲基)苯基〕甲基〕(2-甲基-2h-四唑-5-基)胺基〕-2,3,4,5-四氫-7,9-二甲基-1h-1-苯并氮呯-1-基〕甲基〕-環己基羧酸

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4204450A (en) * 1977-09-09 1980-05-27 Machines Et Applications Shear for large-sized scrap
US6140343A (en) * 1998-09-17 2000-10-31 Pfizer 4-amino substituted-2-substituted-1,2,3,4-tetrahydroquinolines
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
US6489478B1 (en) * 1998-09-17 2002-12-03 Pfizer Inc. 4-amino substituted-2-substituted-1,2,3,4-tetrahydroquinolines
US6313142B1 (en) * 1999-11-30 2001-11-06 Pfizer Inc. Method for making 4-carboxyamino-2-substituted-1,2,3,4-tetrahydroquinoline
US6689897B2 (en) * 2001-04-30 2004-02-10 Pfizer Inc. Intermediates of CETP inhibitors

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070254869A1 (en) * 2003-10-08 2007-11-01 Eli Lilly And Company Compounds And Methods For Treating Dislipidemia
US7749992B2 (en) 2003-10-08 2010-07-06 Eli Lilly And Company Compounds and methods for treating dislipidemia
US20070208003A1 (en) * 2004-03-26 2007-09-06 Bell Michael G Compounds and methods for treating dyslipidemia
WO2013008164A2 (fr) 2011-07-08 2013-01-17 Novartis Ag Méthode de traitement de l'athérosclérose chez des patients présentant un taux élevé de triglycérides
US20170182048A1 (en) * 2014-02-05 2017-06-29 Amgen Inc. Cholesteryl Ester Transfer Protein (CETP) Inhibitor and Pharmaceutical Compositions Comprising Said Inhibitor for Use in the Treatment or Prevention of Cardiovascular Diseases
US10653692B2 (en) * 2014-02-05 2020-05-19 Dezima Pharma Bv Cholesteryl ester transfer protein (CETP) inhibitor and pharmaceutical compositions comprising said inhibitor for use in the treatment or prevention of cardiovascular diseases

Also Published As

Publication number Publication date
CA2570673A1 (fr) 2006-01-05
MXPA06014716A (es) 2007-03-12
NO20070468L (no) 2007-03-12
PL1761522T3 (pl) 2012-03-30
CA2570673C (fr) 2013-01-29
WO2006002342A1 (fr) 2006-01-05
JP4836945B2 (ja) 2011-12-14
IL180276A0 (en) 2007-07-04
EA200700120A1 (ru) 2007-06-29
AU2005258241B2 (en) 2011-08-25
CN1972932A (zh) 2007-05-30
NZ550978A (en) 2010-08-27
ECSP067095A (es) 2007-01-26
AU2005267436A1 (en) 2006-02-02
NO20070470L (no) 2007-03-26
WO2006012093A1 (fr) 2006-02-02
EP1761521A1 (fr) 2007-03-14
EA012585B1 (ru) 2009-10-30
BRPI0512516A (pt) 2008-03-11
CA2570688A1 (fr) 2006-02-02
CR8831A (es) 2008-11-13
DK1761522T3 (da) 2012-01-02
IL180277A (en) 2011-08-31
ES2372291T3 (es) 2012-01-18
AU2005258241A1 (en) 2006-01-05
IL180277A0 (en) 2007-07-04
MXPA06014706A (es) 2007-03-12
US7786108B2 (en) 2010-08-31
MA28730B1 (fr) 2007-07-02
US20070244095A1 (en) 2007-10-18
ATE528304T1 (de) 2011-10-15
EA200700119A1 (ru) 2007-10-26
SI1761522T1 (sl) 2012-02-29
CY1112124T1 (el) 2015-11-04
EP1761522B1 (fr) 2011-10-12
RS52064B (en) 2012-06-30
US20100204207A1 (en) 2010-08-12
EP2479175A1 (fr) 2012-07-25
HRP20110761T1 (hr) 2011-11-30
ECSP067098A (es) 2007-01-26
JP2008504277A (ja) 2008-02-14
PT1761522E (pt) 2011-12-21
MA28729B1 (fr) 2007-07-02
KR20070041452A (ko) 2007-04-18
JP2008504266A (ja) 2008-02-14
EP1761522A1 (fr) 2007-03-14
BRPI0512523A (pt) 2008-03-11
CN1972932B (zh) 2011-06-29
HK1104528A1 (en) 2008-01-18

Similar Documents

Publication Publication Date Title
US20080269284A1 (en) Compounds and Methods for Treating Dyslipidemia
US7470705B2 (en) Compounds and methods for treating dyslipidemia
US7173042B2 (en) Quinoline inhibitors of cGMP phosphodiesterase
JP2006528687A (ja) ベンズイミダゾール、こうした化合物を含有する組成物および使用方法
JP2007015928A (ja) 新規オレフィン誘導体
JP2007530648A (ja) Cns障害の処置のための、5−ht6受容体アンタゴニストとしての3−((ヘテロ)アリールスルフォニル)−8−((アミノアルキル)オキシ)キノリン
KR20000005505A (ko) 피페리딘 및 피롤리딘
US7749992B2 (en) Compounds and methods for treating dislipidemia
WO2007025775A2 (fr) Nouveaux inhibiteurs de la cysteine protease
JP4622047B2 (ja) 新規なヘテロ環カルボキサミド誘導体
JP2007517868A (ja) 治療薬i
FR2860792A1 (fr) Derives de thiophene-2-carboxamide, leur preparation et leur application en therapeutique
KR20110136818A (ko) 신경펩티드 y5 수용체 리간드로서의 아미드 유도체
ZA200610093B (en) Compounds and methods for treating dyslipidemia
CN100448869C (zh) 用于治疗糖尿病的苯胺基吡唑衍生物
MXPA06011047A (en) Compounds and methods for treating dyslipidemia
JP2007217407A (ja) 医薬組成物

Legal Events

Date Code Title Description
AS Assignment

Owner name: ELI LILLY AND COMPANY, INDIANA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:MANTLO, NATHAN BRYAN;WANG, XIAODONG;ESCRIBANO, ANA MARIA;AND OTHERS;REEL/FRAME:018699/0974;SIGNING DATES FROM 20050801 TO 20050816

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION