WO2005061509A1 - Azabicyclic heterocycles as cannabinoid receptor modulators - Google Patents

Azabicyclic heterocycles as cannabinoid receptor modulators Download PDF

Info

Publication number
WO2005061509A1
WO2005061509A1 PCT/US2004/042542 US2004042542W WO2005061509A1 WO 2005061509 A1 WO2005061509 A1 WO 2005061509A1 US 2004042542 W US2004042542 W US 2004042542W WO 2005061509 A1 WO2005061509 A1 WO 2005061509A1
Authority
WO
WIPO (PCT)
Prior art keywords
agents
compounds
disorders
aryl
group
Prior art date
Application number
PCT/US2004/042542
Other languages
French (fr)
Inventor
William R. Ewing
Guixue Yu
Bruce A. Ellsworth
Original Assignee
Bristol-Myers Squibb Company
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 Bristol-Myers Squibb Company filed Critical Bristol-Myers Squibb Company
Priority to EP04814691A priority Critical patent/EP1699796A4/en
Priority to JP2006545502A priority patent/JP2007514756A/en
Publication of WO2005061509A1 publication Critical patent/WO2005061509A1/en
Priority to IS8501A priority patent/IS8501A/en
Priority to NO20062689A priority patent/NO20062689L/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • A61P15/08Drugs for genital or sexual disorders; Contraceptives for gonadal disorders or for enhancing fertility, e.g. inducers of ovulation or of spermatogenesis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • A61P25/16Anti-Parkinson drugs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/18Antipsychotics, i.e. neuroleptics; Drugs for mania or schizophrenia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/20Hypnotics; Sedatives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • 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
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • 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
    • A61P9/02Non-specific cardiovascular stimulants, e.g. drugs for syncope, antihypotensives
    • 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

Definitions

  • Delta-9-tetrahydrocannabinol or Delta-9 THC the principle active component of Cannabis sativa (marijuana) is a member of a large family of lipophilic compounds (i.e., cannabinoids) that mediate physiological and psychotropic effects including regulation of appetite, immunosuppression, analgesia, inflammation, emesis, anti- nocioception, sedation, and intraocular pressure.
  • cannabinoids lipophilic compounds
  • Other members of the cannabinoid family include the endogenous (arachidonic acid-derived) ligands, anandamide, 2- arachidonyl glycerol, and 2-arachidonyl glycerol ether.
  • Cannabinoids work through selective binding to and activation of G-protein coupled cannabinoid receptors.
  • Two types of cannabinoid receptors have been cloned including CB-1 (L. A. Matsuda, et al., Nature, 346, 561-564 (1990)), and CB-2 (S. Munro, et al., Nature, 365, 61-65 (1993)).
  • the CB-1 receptor is highly expressed in the central and peripheral nervous systems (M. Glass, et al., Neuroscience, 11, 299-318 (1997)), while the CB-2 receptor is highly expressed in immune tissue, particularly in spleen and tonsils.
  • the CB-2 receptor is also expressed on other immune system cells, such as lymphoid cells (S.
  • the present application describes compounds according to Formula I, pharmaceutical compositions comprising at least one compound according to Formula I and optionally one or more additional therapeutic agents and methods of treatment using the compounds according to Formula I both alone and in combination with one or more additional therapeutic agents.
  • the compounds have the general Formula I
  • alkyl denotes branched or xmbranched hydrocarbon chains containing 1 to 20 carbons, preferably 1 to 12 carbons-, and more preferably 1 to 8 carbons, in the normal chain, such as, methyl, ethyl, propyl, isopropyl, butyl, sec- butyl, iso-butyl, tert-butyl, pentyl, hexyl, isohexyl, heptyl, 4, -dimethylpentyl, octyl, 2,2,4-trimethylpentyl and the like.
  • alkyl groups may optionally be substituted on any available carbon atom with one or more functional groups commonly attached to such chains, such as, but not limited to hydroxyl, halo, haloalkyl, mercapto or thio, cyano, alkylthio, cycloalkyl, hetesrocyclyl, aryl, heteroaryl, carboxyl, carbalkoyl, carboxamido, carbonyl .
  • alkenyl refers to straight or branched chains of 2 to 20 carbons, preferably 2 to 12 carbons, and more preferably 2 to 8 carbo ⁇ is with one or more double bonds in the nomial chain, such as vinyl, 2-propenyl, 3-butenyl, 2-butenyl, 4- pentenyl, 3-pentenyl, 2-hexenyl, 3-hexenyl, 2-heptenyl, 3-heptenyl, 4-heptenyl, 3- octenyl, 3-nonenyl, 4-decenyl, 3-undecenyl, 4-dodecenyl, 4,5,12-tetradecatrienyl, and the like.
  • alkenyl groups may optionally be substituted on any available carbon atom with one or more functional groups commonly attached to such chains, such as, but not limited to halo, haloalkyl, alkyl., alkoxy, alkynyl, aryl, arylalkyl, cycloalkyl, amino, hydroxyl, heteroaryl, cycloheteroalkyl, alkanoylamino, alkylamido, arylcarbonylamino, nitro, cyano, thiol, alkylthio and/or any of the alkyl substituents set out herein.
  • halo haloalkyl
  • alkyl. alkoxy, alkynyl, aryl, arylalkyl, cycloalkyl, amino, hydroxyl, heteroaryl, cycloheteroalkyl, alkanoylamino, alkylamido, arylcarbonylamino, nitro, cyano, thi
  • alkynyl refers to straight or branched chains of 2 to 20 carbons, preferably 2 to 12 carbons and more preferably 2 to 8 carbons with one or more triple bonds in the normal chain, such as 2-propynyl, 3-butynyl, 2-fc tynyl, 4-pentynyl, 3- pentynyl, 2-hexynyl, 3-hexynyl, 2-heptynyl, 3-heptynyl, 4-hej>tynyl, 3-octynyl, 3- nonynyl, 4-decynyl,3-undecynyl, 4-dodecynyl and the like.
  • alkynyl groups may optionally be substituted on any available carbon atom with one or more functional groups commonly attached to such chains, such as, but not limited to halo, haloalkyl, alkyl, alkoxy, alkenyl, aryl, arylalkyl, cycloalkyl, amino, hydroxyl, heteroaryl, cycloheteroalkyl, alkanoylamino, alkylamido, arylcarbonylamino, nitro, cyano, thiol, alkylthio and/or any of the alkyl substituents set out herein Unless otherwise indicated, the term "cycloalkyl" as employed herein alone or as part of another group includes saturated or partially unsaturated (containing one or more double bonds) cyclic hydrocarbon groups containing 1 to 3 rings, appended or fused, including monocyclicalkyl, bicychcalkyl and tricyclicalkyl, containing a total of 3 to 20 carbons fo ⁇
  • any cycloalkyl may be optionally substituted through any available carbon atoms with one or more groups selected from hydrogen, halo, haloalkyl, alkyl, alkoxy, haloalkyloxy, hydroxyl, alkenyl, alkynyl, aryl, aryloxy, heteroaryl, heteroaryloxy, arylalkyl, heteroarylalkyl, alkylamido, alkanoylamino, oxo, acyl, arylcarbonylamino, amino, nitro, cyano, tliiol and/or alkylthio and/or any of the alkyl substituents.
  • cycloalkylalkyl as used herein alone or as part of another group refers to alkyl groups as defined above having a cycloalkyl substituent, wherein said "cycloalkyl” and/or “alkyl” groups may optionally be substituted as defined above.
  • aryl refers to monocyclic and bicyclic aromatic groups containing 6 to 10 carbons in the ring portion (such as phenyl or naphthyl including 1-naphthyl and 2- naphthyl) and may optionally include one to three additional rings fused to a carbocyclic ring or a heterocyclic ring, for example Further, "aryl", as defined herein, may optionally be substituted with one or more functional groups, such as halo, alkyl, haloalkyl, alkoxy, haloalkoxy, alkenyl, alkynyl, cycloalkyl, cycloalkylalkyl, heterocyclyl, heterocycloalkyl, aryl, heteroaryl, arylalkyl, aryloxy, aryloxyalkyl, arylalkoxy, alkoxycarbonyl, arylcarbonyl, arylalkeny
  • heteroaryl refers to a 5- or 6- membered aromatic ring which includes 1 , 2, 3 or 4 hetero atoms such as nitrogen, oxygen or sulfur.
  • Such rings may be fused to an aryl, cycloalkyl, heteroaryl or heterocyclyl and include possible N-oxides as described in Katritzky, A. R. and Rees, C. W., eds. Comprehensive Heterocyclic Chemistry: The Structure, Reactions, Synthesis and Uses of Heterocyclic Compounds 1984, Pergamon Press, New York, NY; and Katritzky, A. R., Rees, C. W., Scriven, E.
  • heteroaryl as defined herein, may optionally be substituted with one or more substituents such as the substituents included above in the definition of "subsiituted alkyl” and “substituted aryl".
  • substituents include the following:
  • heteroarylalkyl as used herein alone or as part of another group refers to alkyl groups as defined above having a heteroaryl substituent, wherein said heteroaryl and/or alkyl groups may optionally be substituted as defined above.
  • heterocyclo represents an unsubstituted or substituted stable 4 to 7-membered monocyclic ring system which may be saturated or unsaturated, and which consists of carbon atoms, with one to four heteroatoms selected from nitrogen, oxygen ox sulfur, and wherein the nitrogen and sulfur heteroatoms may optionally be oxidized, and the nitrogen heteroatom may optionally be quaternized.
  • the heterocyclic ring may be attached at any heteroatom or carbon atom which results in the creation of a stable structure.
  • heterocyclic groups include, but is not limited to, piperidinyl, piperazinyl, oxopiperazinyl, oxopiperidinyl, oxopyrrolidinyl, oxoazepinyl, azepinyl, pyrrolyl, pyrrolidinyl, furanyl, thienyl, pyrazolyl, pyrazolidinyl, imidazolyl, imidazolinyl, imidazolidinyl, pyridyl, pyrazinyl, pyrimidinyl, pyridazinyl, oxazolyl, oxazolidinyl, isooxazolyl, isoxazolidinyl, morpholinyl, thiazolyl, thiazolidinyl, isothiazolyl, thiadiazolyl, tetrahydropyranyl, thiamo holinyl, thiamorph
  • heterocycloalkyl refers to alkyl groups as defined above having a heterocyclyl substituent, wherein said heterocyclyl and/or alkyl groups may optionally be substituted as defined above.
  • arylalkyl refers to alkyl, alkenyl and alkynyl groups as described above having an aryl substituent.
  • Representative examples of arylalkyl include, but are not limited to, benzyl, 2-phenylethyl, 3-phenylpropyl, phenethyl, benzhydryl and naphthylmethyl and the like.
  • alkoxy means "alkoxy", "aryloxy”, “heteroaryloxy” "arylalkyloxy”, or “heteroarylalkyloxy” as employed herein alone or as part of another group includes an alkyl or aryl group as defined above linked through an oxygen atom.
  • halogen or “halo” as used herein alone or as part of another group refers to chlorine, bromine, fluorine, and iodine, with bromine, chlorine or fluorine being preferred.
  • cyano refers to a -CN group.
  • methylene refers to a -CH 2 - group.
  • nitro refers to a -NO 2 group.
  • the compounds of formula I can be present as salts, which are also within the scope of this invention. Pharmaceutically acceptable (i.e., non-toxic, physiologically acceptable) salts are preferred. If the compounds of formula I have, for example, at least one basic center, they can form acid addition salts.
  • organic carboxylic acids such as alkanecarboxylic acids of 1 to 4 carbon atoms, for example acetic acid, which are unsubstituted or substituted, for example, by halogen as chloroacetic acid, such as saturated or unsaturated dicarboxylic acids, for example oxalic, malonic, succinic, maleic, fumaric, phthalic or terephthalic acid, such as hydroxycarboxylic acids, for example ascorbic, glycolic, lactic, malic, tartaric or citric acid, such as amino acids, (for example aspartic or glutamic acid or lysine or arginine), or benzoic acid, or with organic sulfonic acids, such as (C_-C 4 ) alkyl or arylsulfonic acids which are unsubstituted or substituted, for example
  • Corresponding acid addition salts can also be formed having, if desired, an additionally present basic center.
  • the compounds of formula I having at least one acid group can also form salts with bases.
  • Suitable salts with bases are, for example, metal salts, such as alkali metal or alkaline earth metal salts, for example sodium, potassium or magnesium salts, or salts with ammonia or an organic amine, such as mo ⁇ holine, thiomo ⁇ holine, piperidine, pyrrolidine, a mono, di or tri-lower alkylamine, for example ethyl, tert-butyl, diethyl, diisopropyl, triethyl, tributyl or dimethyl-propylamine, or a mono, di or trihydroxy lower alkylamine, for example mono, di or triethanolamine.
  • Corresponding internal salts may furthermore be formed. Salts which are unsuitable for pharmaceutical uses but which can be employed, for example, for the isolation or purification of free compounds of formula I or their pharmaceutically acceptable salts, are also included.
  • Preferred salts of the compounds of formula I which contain a basic group include monohydrochloride, hydrogensulfate, methanesulfonate, phosphate, nitrate or acetate.
  • Preferred salts of the compounds of formula I which contain an acid group include sodium, potassium and magnesium salts and pharmaceutically acceptable organic amines.
  • modulator refers to a chemical compound with capacity to either enhance (e.g., "agonist” activity) or partially enhance (e.g., “partial agonist” activity) or inhibit (e.g., "antagonist” activity or “inverse agonist” activity) a functional property of biological activity or process (e.g., enzyme activity or receptor binding); such enhancement or inhibition may be contingent on the occurrence of a specific event, such as activation of a signal transduction pathway, and/or may be manifest only in particular cell types.
  • bioactive metabolite as employed herein refers to any functional group contained in a compound of fonnula I with an open valence for further substitution wherein such substitution can, upon biotransformation, generate a compound of formula I.
  • prodrug esters as employed herein includes esters and carbonates formed by reacting one or more hydroxyls of compounds of formula I with alkyl, alkoxy, or aryl substituted acylating agents employing procedures known to those skilled in the art to generate acetates, pivalates, methylcarbonates, benzoates and the like.
  • Prodrug esters may also include- but are not limited to groups such as phosphate esters, phosphonate esters, phosphonamidate esters, sulfate esters, sulfonate esters, and sulfonamidate esters wherein the ester may be further substituted with groups that confer a pharmaceutical advantage such as-but not limited to-favorable aqueous solubility or in vivo exposure to the bioactive component formula I.
  • the term "prodrug” as employed herein includes functionalization of bioactive amine- or hydroxyl-containing compounds of formula I to form alkyl-, acyl-, sulfonyl-, phosphoryl-, or carbohydrate-substituted derivatives.
  • Such derivatives are fonned by reacting compounds of formula I with alkylating-, acylating-, sulfonylating-, or phosphorylating reagents employing procedures known to those skilled in the art.
  • Alkylation of amines of formula I may result in- but are not limited to- derivatives that include spacer units to other prodrug moieties such as substituted alkyoxymethyl-, acyloxymethyl-, phosphoryloxymethyl-, or sulfonyloxymethyl- groups.
  • Alkylation of amines of formula I may result in the generation of quarternary amine salts that act in vivo to provide the bioactive agent (i.e., the compound of formula I).
  • Preferred prodrugs consist of a compound of formula I where a pendant hydroxyl is phosphorylated to generate a phosphate derivative. Such a prodrug may also include a spacer group between the compound of formula I and the phosphate group, such as a methyleneoxy-group. Methods to generate such a prodrug from a compound of formula I are known to those skilled in the art, and are listed in the references below. Preferred prodrugs also consist of a compound of formula I where a pendant amine, such as a pyridine group, is alkylated with a group, such as methyl, to form a quarternary ammonium ion salt.
  • prodrug from a compound of formula I
  • Any compound that can be converted in vivo to provide the bioactive agent i.e., the compound of formula I
  • Various forms of prodrugs are well known in the art. A comprehensive description of prodrugs and prodrug derivatives are described in: The Practice of Medicinal Chemistry, Camille G. Wermuth et al., Ch 31, (Academic Press, 1996); Design of Prodrugs, edited by H. Bundgaard, (Elsevier, 1985); A Textbook of Drug Design and Development, P. Krogsgaard-Larson and H.
  • An administration of a therapeutic agent of the invention includes administration of a therapeutically effective amount of the agent of the invention.
  • therapeutically effective amount refers to an amount of a therapeutic agent to treat or prevent a condition treatable by administration of a composition of the invention. That amount is the amount sufficient to exhibit a detectable therapeutic or preventative or ameliorative effect. The effect may include, for example, treatment or prevention of the conditions listed herein.
  • the precise effective amount for a subject will depend upon the subject's size and health, the nature and extent of the condition being treated, recommendations of the treating physician, and the therapeutics or combination of therapeutics selected for administration.
  • All stereoisomers of the compounds of the instant invention are contemplated, either in mixture or in pure or substantially pure form.
  • the compounds of the present invention can have asymmetric centers at any of the carbon atoms including any one of the R substituents. Consequently, compounds of formula I can exist in enantiomeric or diastereomeric forms or in mixtures thereof.
  • the processes for preparation can utilize racemates, enantiomers or diastereomers as starting materials. When diastereomeric or enantiomeric products are prepared, they can be separated by conventional methods for example, chromatographic techniques, chiral HPLC or fractional crystallization.
  • DIPEA N,N-diisopropylehtylamine
  • EDAC l-(3-Dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride
  • EtOAc ethyl acetate
  • LAH lithium aluminum hydride
  • TFA trifluoroacetic acid
  • h hour(s)
  • PG suitable nitrogen protecting group exemplified by benzyl, methoxymethyl- [MOM], benzyloxymethyl- [BOM], 2-(trimethylsilyl)ethoxymethyl- [SEM], methoxyethoxymethyl- [MEM], or t-butyl groups
  • EE S n 2 or S n l leaving group exemplified by halogen (CI, Br, I) and sulfonates (- OSO 2 -aryl (e.g., -OSO 2 Ph or -OSO 2 PhCH 3 ), or -OSO 2 -alkyl (e.g., -OSO 2 CH 3 or -OSO 2 CF 3 ));
  • Compounds of formula II are either commercially available or available by means known to one skilled in the art.
  • Compounds of formula III can be prepared by reacting compounds of formula II with an appropriate protecting group such as benzyl bromide.
  • an appropriate protecting group such as benzyl bromide.
  • Exemplary nitrogen protecting groups and methods of protecting the nitrogen are similar to those for protecting amines, such as those described in T. W. Greene and P. G. M. Wuts, Protecting Groups in Organic Synthesis, John Wiley & Sons, Inc, New York, 1991.
  • Preferred nitrogen protecting groups are benzyl, tert- butyl, methoxymethyl (MOM), methoxyethoxymethyl (MEM), and 2- (trimethylsilyl)ethoxymethyl (SEM) groups.
  • Compounds of formula IV may be prepared from compounds of formula III via selective displacement of the leaving group (EE) by the conjugate base of an appropriate alcohol, RO-M, wherein R is alkyl or benzyl, and M is a metalloid such as Li, Na, Mg(halide) and the like in solvents such as dioxane. Similar reactions has been described in the literature (Riedl, Z. et. al. Tetrahedron, 2002, 5645-5650).
  • Compounds of formula V can be prepared by the reactions of compounds of formula IV with activated R , such as activated by boronic acids, tin, Grignard reagents, Zinc, Cu, etc in the presence of an appropriate catalyst if needed such as Pd(PPh 3 ) 4 .
  • Compounds of formula V may also be prepared from a compound of formula IV via displacement of the leaving group (EE) by the conjugate base of a 9 9 compound R -H, wherem R is as previously defined, using a base in an inert
  • Exemplary bases include sodium carbonate, potassium carbonate, cesium carbonate, sodium hydride, potassium hydride, or alkyl lithiums.
  • Compounds of formula VI can be prepared by reacting compounds of formula V with a chlorinating agent such as POCl 3 in an inert solvent such as toluene at elevated temperature.
  • Compounds of formula VII, where MM is a metal or a borate ester may be prepared via lithiation of a compound of formula VI wherein EE is hydrogen or a halogen (chloro, bromo, iodo), and reacting the resulting aryl lithium with an appropriate borate derivative or with reagents such as trialkyltin halide.
  • Compounds of formula VIII can be prepared by the reactions of compounds of formula VI with activated R 1 , such as activated by boronic acids, tin, Grignard reagents, Zinc, Cu, etc in the presence of an appropriate catalyst if needed such as Pd(PPh 3 ) 4 .
  • Compounds of formula VIII may also be prepared from a compound of formula VI via displacement of the leaving group (EE) by the conjugate base of a compound R -H, wherein R 1 is as previously defined, using a base in an inert solvent.
  • Compounds of formula VIII can also be prepared by a palladium or nickel catalyzed coupling of a compound of formula VII wherein MM is a borate ester with an appropriately activated R 1 such as halide or mesylate.
  • R 1 such as halide or mesylate.
  • R 1 such as halide or mesylate.
  • R 1 such as halide or borate esters
  • an appropriate catalyst such as tetrakis(triphenylphosphine)palladium(0) and dichlorobis(triphenylphosphine)nickel(II).
  • Compounds of formula IX can be prepared by removing the protecting group
  • PG in compound VIII under acidic (e.g. TFA for t-butyl or Boc-), basic (e.g. NaOH for amide), catalytic hydrogenation (for benzyl-), or Lewis acid (e.g. A1C1 3 for benzyl) conditions.
  • Compounds of formula X can be prepared by reacting compounds of formula IX with a chlorinating agent such as POCl 3 in an inert solvent such as toluene at elevated temperature.
  • Compounds of formula XI can be prepared by reacting compounds of formula X with hydrazine in an inert solvent such as pyridine at elevated temperature.
  • Compounds of formula I can be prepared by reacting compounds of formula XI with an alpha-ketoacid or alpha-ketoacid ester in a protic solvent, such as an alkanoic acid for example, acetic acid in ambient to evelatated temperatures.
  • a protic solvent such as an alkanoic acid for example, acetic acid in ambient to evelatated temperatures.
  • compounds of formula XIII may be prepared from compounds of formula III via selective displacement of the leaving group (EE) by the conjugate base of an appropriate alcohol, RO-M, wherein R is alkyl or benzyl, and M is a metal such as Li, Na, Mg(halide) and the like in solvents such as methanol.
  • RO-M an appropriate alcohol
  • R alkyl or benzyl
  • M a metal such as Li, Na, Mg(halide) and the like in solvents such as methanol.
  • Compounds of formula XIV can be prepared by the reactions of compounds of formula XIII with activated R 1 , such as activated by boronic acids, tin, Grignard reagents, Zinc, Cu, etc in the presence of an appropriate catalyst if needed such as Pd(PPh 3 ) 4 .
  • Compounds of formula XIV may also be prepared from a compound of formula XIII via displacement of the leaving group (EE) by the conjugate base of a compound R ⁇ H, wherein R 1 is as previously defined, using a base in an inert solvent.
  • Compounds of formula XVI, where MM is a metal or a borate ester may be prepared via lithiation of a compound of formula XV wherein EE is hydrogen or a halogen (chloro, bromo, iodo), and reacting the resulting aryl lithium with an appropriate borate derivative or with reagents such as trialkyltin halide.
  • Compounds of formula VIII can be prepared by the reactions of compounds of formula XV with activated R 2 , such as activated by boronic acids, tin, Grignard reagents, Zinc, Cu, etc in the presence of an appropriate catalyst if needed such as Pd(PPh 3 ) 4 .
  • Compounds of formula VIII may also be prepared from a compound of formula XV via displacement of the leaving group (EE) by the conjugate base of a 9 9 compound R -H, wherein R is as previously defined, using a base in an inert solvent.
  • Compounds of formula VIII can also be prepared by a palladium or nickel catalyzed coupling of a compound of formula XVI wherein MM is a borate ester with an appropriately activated R such as halide or mesylate.
  • MM is a metal atom such as tin, zinc, magnesium, and lithium
  • activated R such as halide or borate esters with an appropriate catalyst such as tetrakis(triphenylphosphine)palladium(0) and dichlorobis(triphenylphosphine)nickel(II).
  • activated R such as halide or borate esters
  • an appropriate catalyst such as tetrakis(triphenylphosphine)palladium(0) and dichlorobis(triphenylphosphine)nickel(II).
  • Compounds of formula I may be prepared from a compound of formula VIII as described in Scheme 1.
  • the hydroxyl group in XVII can be activated by reacting with reagents such as trifluromethane sulfonic anhydride in the presence of a suitable base such as triethylamine.
  • This activated moiety can then be selectively coupled with activated R", such as activated, by boronic acids, tin, Grignard reagents, Zinc, Cu, etc in the presence of an appropriate catalyst if needed such as Pd(PPh 3 ) 4 to provide compounds XVIII.
  • Compounds of formula XIX can be prepared from compounds XVIII in a two step sequence: (a) by removing the protecting group (PG) in compound XVIII under acidic (e.g. TFA for t-butyl or Boc-), basic (e.g. NaOH for amide), catalytic hydrogenation (for benzyl-), or Lewis acid (e.g. A1C1 3 for benzyl) conditions, followed by (b) reacting the resulting intermediate with a chlorinating agent such as POCl 3 in an inert solvent such as toluene at elevated temperature.
  • acidic e.g. TFA for t-butyl or Boc-
  • basic e.g. NaOH for amide
  • catalytic hydrogenation for benzyl-
  • Lewis acid e.g. A1C1 3 for benzyl
  • direct treatment of XVIII with chlorinating agents such as POCl 3 at higher temperature may provide compounds of formula XIX in one step from XVTII.
  • Compounds of formula XX can be prepared by reacting compounds of formula XIX with hydrazine in selected solvents such as isobutanol.
  • Compounds of formula XXI can be prepared by reacting compounds of formula XX with an alpha-ketoacid or alpha-ketoacid ester in a protic acid, such as acetic acid at ambient to evelvated temperature. .
  • Compounds of formula I can be prepared by the reactions of compounds of formula XXI with activated R 1 , such as activated by boronic acids, tin, Grignard reagents, Zinc, Cu, etc in the presence of an appropriate catalyst if needed such as
  • Compounds of formula I may also be prepared from a compound of formula XXI via displacement of the leaving group (EE) by the conjugate base of a compound R ⁇ H, wherein R 1 is as previously defined, using a base in an inert solvent.
  • Compounds of formula XXII, where MM is a metal or a borate ester may be prepared via lithiation of a compound of formula XXI wherein EE is hydrogen or a halogen (chloro, bromo, iodo), and reacting the resulting aryl lithium with an appropriate borate derivative or with reagents such as trialkyltin halide.
  • Compounds of formula I can also be prepared by a palladium or nickel catalyzed coupling of a compound of formula XXII wherein MM is a borate ester with an appropriate activated R 1 such as halide or mesylate.
  • R 1 such as halide or mesylate.
  • R 1 such as halide or borate esters
  • an appropriate catalyst such as tetrakis(triphenylphosphine)palladium(0) and dichlorobis(triphenylphosphine)nickel(II).
  • compounds of formula XXIV can be prepared from compounds XV in a two step sequence: (a) by removing the protecting group (PG) in compound XV under acidic (e.g. TFA for t-butyl or Boc-), basic (e.g. NaOH for amide), catalytic hydrogenation (for benzyl-), or Lewis acid (e.g. A1C1 3 for benzyl) conditions, followed by (b) reacting the resulting intermediate with a chlorinating agent such as POCl 3 in an inert solvent such as toluene at elevated temperature
  • a chlorinating agent such as POCl 3
  • an inert solvent such as toluene
  • Compounds of formula XXVII can be prepared by reacting compounds of formula XXV with an alpha-ketoacid or alpha-ketoacid ester in a protic acid, such as acetic acid at ambient to evelated temperatures.
  • Target compounds of formula I can be prepared from compounds of formula XXVII by following an analogous sequence of reactions as described in Scheme 3 via intermediates XI, XXVII and XXVIII.
  • compounds of formula X can be prepared by the reactions of compounds of formula XXIV with activated R , such as activated by boronic acids, tin, Grignard reagents, Zinc, Cu, etc in the presence of an appropriate catalyst if needed such as Pd(PPh 3 ) 4 .
  • activated R such as activated by boronic acids, tin, Grignard reagents, Zinc, Cu, etc.
  • Compounds of formula I can then be prepared from compounds of formula I by following an analogous sequence of reactions as described in Scheme 1.
  • analogs having certain arbitrarily defined subsets of R 1 and R 2 and R 3 can be changed to other analogs having certain other arbitrarily defined subsets of R 1 and R 2 and R 3 by manipulation of the functional groups embedded in these R groups.
  • R la , R 2a , R 3a are groups such as aminoaryl, aminoalkyl, aminoheterocyclyl, or aminoheteroaryl
  • the amino group can be reacted with either carboxylic acids or acid chlorides or sulfonyl chlorides to provide amide or sulfonamide derivatives.
  • Such a manipulation can also be conducted via parallel synthesis.
  • R la , R 2a or R 3a are substituted with an activated group such as a halogen or boronic acid
  • additional metal catalyzed cross-coupling reactions may be performed to provide additional set of analogs as described by Formula I.
  • Such a manipulation can also be conducted via parallel synthesis.
  • Parallel synthesis may be employed in the preparation of compounds, for example, where the intermediates possess an activated reaction center: such as but not limited to a reactive heteroaryl cliloride for Suzuki coupling chemistry or a carboxylic acid for amide coupling chemistry.
  • Analytical HPLC/MS was performed on Shimadzu LC10AS liquid chromatographs and Waters ZMD Mass Spectrometers using the following method:
  • the crude 2-benzyl-4,5-bis(4-chlorophenyl)pyridazin-3(2H)-one from the above reaction was dissolved in toluene (350 mL).
  • Aluminum chloride (A1C1 3 , 46.3 g, 348.0 mmol) was then added to the toluene solution, producing an exotherm.
  • the reaction was then placed in an oil bath preheated at 75°C for 3 h. After this time, the reaction mixture was poured into ice- water (1000 mL), and the resulting mixture was extracted with EtOAc (3 x 500 mL).
  • 4,5-Bis(4-chlorophenyl)pyridazin-3(2H)-one (16.5 g, 52.2 mmol) was suspended in toluene (50 mL).
  • pyridine 8.3 mL, 104.4 mmol
  • POCl 3 14.3 mL, 156.6 mmol
  • the reaction mixture was placed in an oil bath preheated at 110°C. After 4 h, the reaction mixture was cooled to RT, then poured over 500 g ice to quench the excess POCl 3 . The dark mixture was extracted with EtOAc (2 x 300 mL).
  • the title compound was prepared using the method described in Example IE, reacting l-(4,5-bis(4-chlorophenyl)pyridazin-3-yl)hydrazine with 2-(4-fluorophenyl)- 2-oxoacetic acid to give 8,9-bis(4-chlorophenyl)-3-(4-fluorophenyl)-4H- pyridazino[6,l-c][l,2,4]triazin-4-one.
  • Set A consists of compounds that differ from one another only in the identity of R 3 with R 1 and R 2 fixed as 4-chlorophenyl.
  • Set A may be considered a one dimensional library of example compounds. Were one to vary both R and R , a two dimensional library of example compounds would result.
  • Set B is the two dimensional library that consists of all permutations of all of the variants of R 3 represented in Set A and a set of R 2 variants listed below.
  • R 1 is 4- chlorophenyl
  • R 4 is hydrogen
  • the compounds of Set B may be prepared by one skilled in the art by the methods described above.
  • the compounds of Set B are meant to further illustrate the scope of the invention without being limiting in any way.
  • Set B is the two dimensional library that consists of all permutations of all of the variants of R 3 represented in Set A and a set of R 2 variants listed above with R fixed as 4-chlorophenyl.
  • Set C is the three dimensional library that consists of all permutations of all of the variants of R 3 represented in Set A, all of the R 2 variants listed above for Set B, and a set of R 1 variants listed below.
  • R 4 is hydrogen.
  • the compounds of Set C may be prepared by one skilled in the art by the methods described above. The compounds of Set C are meant to further illustrate the scope of the invention without being limiting in any way.
  • Radioligand binding studies were conducted in membranes prepared from Chinese Hamster Ovary (CHO) cells that over-express recombinant human CB-1 (CHO-CB-1 cells). Total assay volume for the binding studies was 100 ⁇ l. 5 ⁇ g of membranes were brought up to a final volume of 95 ⁇ l with Binding Buffer (25 mM HEPES, 150 mM NaCl, 2.5 mM CaCl 2 , 1 mM MgCl 2 , 0.25% BSA). The diluted membranes were preincubated with a compound or DMSO vehicle.
  • Binding Buffer 25 mM HEPES, 150 mM NaCl, 2.5 mM CaCl 2 , 1 mM MgCl 2 , 0.25% BSA.
  • the binding reaction was initiated by the addition of 2 nM final 3 H-CP-55,940 (120 Ci/mmol) and proceeded for 2.5 hours at room temperature.
  • the binding reaction was terminated by transfening the reaction to GF B 96 well plates (presoaked with 0.3% polyethylenimine) using a Packard Cell Harvester.
  • the filter was washed with 0.25x PBS, 30 ⁇ l MicroScint was added per well, and the bound radiolabel was quantitated by scintillation counting on a Packard TopCount Scintillation Counter.
  • the CB-2 radioligand binding assay was conducted identically except that the membranes from CHO-CB-2 cells were used.
  • the compound For a compound to be considered a CB-1 antagonist, the compound must possess a CB-1 receptor binding affinity Ki less than 13000 nM. As determined by the assay described above, the CB-1 receptor binding K, values of working Examples 1-63 fall within the range of 0.01 nM to 10000 nM.
  • CB-1 inverse agonist activity of test compounds was determined in CHO-CB-1 cells using a cAMP accumulation assay.
  • CHO-CB-1 cells were grown in 96 well plates to near confluence.
  • growth medium was aspirated and 100 of Assay Buffer (PBS plus 25 mM HEPES / 0.1 mM 3- isobutyl-1-methylxanthine/ 0.1 % BSA) was added.
  • Assay Buffer PBS plus 25 mM HEPES / 0.1 mM 3- isobutyl-1-methylxanthine/ 0.1 % BSA
  • Compounds were added to the Assay buffer diluted 1:100 from 100% DMSO and allowed to preincubate for 10 minutes prior to addition of 5 uM forskolin.
  • the mixture was allowed to proceed for 15 minutes at room temperature and was terminated by the addition of 0.1 N HC1.
  • the total intracellular cAMP concentration was quantitated using the Amersham cAMP SPA kit.
  • the compounds of the present invention are cannabinoid receptor modulators, and include compounds which are, for example, selective agonists, partial agonists, inverse agonists, antagonists or partial antagonists of the cannabinoid receptor. Accordingly, the compounds of the present invention may be useful for the treatment or prevention of diseases and disorders associated with G-protein coupled cannabinoid receptor activity. Preferably, compounds of the present invention possess activity as antagonists or inverse agonists of the CB-1 receptor, and may be used in the treatment of diseases or disorders associated with the activity of the CB-1 receptor.
  • the compounds of the present invention can be administered to mammals, preferably humans, for the treatment of a variety of conditions and disorders, including, but not limited to metabolic and eating disorders as well as conditions associated with metabolic disorders, (e.g., obesity, diabetes, arteriosclerosis, hypertension, polycystic ovary disease, cardiovascular disease, osteoarthritis, dermatological disorders, hypertension, insulin resistance, hypercholesterolemia, hypertriglyceridemia, cholelithiasis and sleep disorders, hyperlipidemic conditions, bulimia nervosa and compulsive eating disorders) or psychiatric disorders, such as substance abuse, depression, anxiety, mania and schizophrenia.
  • metabolic disorders e.g., obesity, diabetes, arteriosclerosis, hypertension, polycystic ovary disease, cardiovascular disease, osteoarthritis, dermatological disorders, hypertension, insulin resistance, hypercholesterolemia, hypertriglyceridemia, cholelithiasis and sleep disorders, hyperlipidemic conditions, bulimia nervos
  • Parkinson's Disease cerebral apoplexy and craniocerebral trauma
  • hypotension e.g., hemorrhagic and endotoxin-inducd hypotension
  • cardiac dysfunction e.g., associated with valvular disease, myocardial infarction, cardiac hypertrophy or congestive heart failure
  • improvement of the overall pulmonary function transplant rejection; rheumatoid arthritis; multiple sclerosis; inflammatory bowel disease; lupus; graft vs.
  • appetitive disorders are understood as meaning: disorders associated with a substance and especially abuse of a substance and/or dependency on a substance, disorders of eating behaviors, especially those liable to cause excess weight, irrespective of its origin, for example: bulimia nervosa, craving for sugars.
  • the present invention therefore further relates to the use of a CB-1 receptor antagonist or inverse agonist for the treatment of bulimia and obesity, including obesity associated with type II diabetes (non-insulin-dependent diabetes), or more generally any disease resulting in the patient becoming overweight.
  • Obesity as described herein, is defined by a body mass index (kg/m ) of at least 26. It may be due to any cause, whether genetic or environmental, including overeating and bulemia, polycycstic ovary disease, craniopharyngeoma, Prader-Willi Syndrome, Frohlich's Syndrome, Type II diabetes, growth hormone deficiency, Turner's Syndrome and other pathological states characterized by reduced metabolic activity or reduced energy expenditure.
  • treating encompasses prevention, partial alleviation, or cure of the disease or disorder.
  • treatment of obesity is expected to prevent progression of medical co variants of obesity, such as arteriosclerosis, Type II diabetes, polycystic ovary disease, cardiovascular disease, osteoarthritis, dermatological disorders, hypertension, insulin resistance, hypercholesterolemia, hypertriglyceridemia, cholelithiasis and sleep disorders.
  • Compounds in the present invention may also be useful in treating substance abuse disorders, including substance dependence or abuse without physiological dependence.
  • Substances of abuse include alcohol, amphetamines (or amphetamine- like substances), caffeine, cannabis, cocaine, hallucinogens, inhalents, nicotine, opioids, phencyclidine (or phencyclidine-like compounds), sedative-hypnotics or benzodiazepines, and other (or unknown) substances and combinations of the above.
  • the terms "substance abuse disorders” also includes drug or alcohol withdrawal syndromes and substance-induced anxiety or mood disorder with onset during ⁇ vithdrawal.
  • Compounds in the present invention may be useful in treating memory impainnent and cognitive disorders. The condition of memory impairment is manifested by impaim ⁇ ent of the ability to learn new information and/or the inability to recall previously learned information.
  • Memory impairment is a primary symptom of dementia and can also be a symptom associated with such diseases as Alzheimer's disease, schizophrenia, Parkinson's disease, Huntington's disease, Pick's disease, Creutzfeld- Jakob disease, HIV, cardiovascular disease, and head trauma as well as age-related cognitive decline.
  • Dementias are diseases that include memory loss and additional intellectual impairment separate from memory.
  • Cannabinoid receptor modulators may also be useful in treating cognitive impairments related to attentional deficits, such as attention deficit disorder.
  • Compounds in the present invention may also be useful in treating diseases associated with dysfunction of brain dopaminergic systems, such as Parkinson's Disease and substance abuse disorders. Parkinsons' s Disease is a neurodenerative movement disorder characterized by bradykinesia and tremor.
  • the compounds of the present invention are further useful for the treatment and prevention of respiratory diseases and disorders.
  • Respiratory diseases for which cannabinoid receptor modulators are useful include, but are not limited to, chronic pulmonary obstructive disorder, emphysema, asthma, and bronchitis.
  • cannabinoid receptor modulators block the activation of lung epithelial cells by moeties such as allergic agents, inflammatory cytokines or smoke, thereby limiting release of mucin, cytokines, and chemokines, or selectively inhibiting lung epithelial cell activation.
  • the compounds employed in the present invention may stimulate inhibitory pathways in cells, particularly in leukocytes, lung epithelial cells, or both, and are thus useful in treating such diseases.
  • Leukocyte activation is defined herein as any or all of cell proliferation, cytokine production, adhesion protein expression, and production of inflammatory mediators.
  • Epithelial cell activation is defined herein as the production of any or all of mucins, cytokines, chemokines, and adhesion protein expression.
  • transplant such as organ transplant, acute transplant, xenotransplant or heterograft or homograft (such as is employed in burn treatment)
  • protection from ischemic or reperfusion injury such as ischemic or reperfusion injury incurred during organ transplantation, myocardial infarction, stroke or other causes
  • transplantation tolerance induction arthritis (such as rheumatoid arthritis, psoriatic arthritis or osteoarthritis); multiple sclerosis
  • respiratory and pulmonary diseases including but not limited to chronic obstructive pulmonary disease (COPD), emphysema, bronchitis, and acute respiratory distress syndrome (ARDS); inflammatory bowel disease, including ulcerative colitis and Crohn's disease; lupus (systemic lupus erythematosis); graft vs.
  • COPD chronic obstructive pulmonary disease
  • ARDS acute respiratory distress syndrome
  • T-cell mediated hypersensitivity diseases including contact hypersensitivity, delayed-type hypersensitivity, and gluten-sensitive enteropathy (Celiac disease); psoriasis; contact dermatitis (including that due to poison ivy); Hashimoto's thyroiditis; Sjogren's syndrome; Autoimmune Hyperthyroidism, such as Graves' Disease; Addison's disease (autoimmune disease of the adrenal glands); Autoimmune polyglandular disease (also known as autoimmune polyglandular syndrome); autoimmune alopecia; pernicious anemia; vitiligo; autoimmune hypopituatarism; Guillain-Barre syndrome; other autoimmune diseases; glomerulonephritis; serum sickness; uticaria; allergic diseases such as respiratory allergies (asthma, hayfever, allergic rhinitis) or skin allergies; scleracierma; mycosis fungoides; acute inflammatory and respiratory responses (such as acute respiratory distress syndrome and ishchemia/reperfusion
  • leukocyte activation- associated or ' "leukocyte-activation mediated” disease as used herein includes each of the above referenced diseases or disorders.
  • the compounds of the present invention are useful for treating the aforementioned exemplary disorders irrespective of their etiology.
  • the combined activity of the present compounds towards monocytes, macrophages, T-cells, etc. may be useful in treating any of the above-mentioned disorders.
  • Cannabinoid receptors are important in the regulation of Fc gamma receptor responses of monocytes and macrophages.
  • Compounds of the present invention inhibit the Fc gamma dependent production of TNF alpha in human monocytes/macrophages.
  • the ability to inhibit Fc gamma receptor dependent monocyte and macrophage responses results in additional anti-inflammatory activity for the present compounds.
  • This activity is especially of value, for example, in treating inflammatory diseases such as arthritis or inflammatory bowel disease.
  • the present compounds are useful for treating autoimmune glomerulonephritis and other instances of glomerulonephritis induced by deposition of immune complexes in the kidney that trigger Fc gamma receptor responses leading to kidney damage.
  • Cannabinoid receptors are expressed on lung epithelial cells. These cells are responsible for the secretion of mucins and inflammatory cytokines/chemokines in the lung and are thus intricately involved in the generation and progression of respiratory diseases.
  • Cannabinoid receptor modulators regulate both the spontaneous and the stimulated production of both mucins and cytokines.
  • respiratory and pulmonary diseases including, COPD, ARDS, and bronchitis.
  • cannabinoid receptors may be expressed on gut epithelial cells and hence regulate cytokine and mucin production and may be of clinical use in treating inflammatory diseases related to the gut.
  • Cannabinoid receptors are also expressed on lymphocytes, a subset of leukocytes.
  • cannabinoid receptor modulators will inhibit B and T-cell activation, proliferation and differentiation.
  • such compounds will be useful in treating autoimmune diseases that involve either antibody or cell mediated responses such as multiple sclerosis and lupus.
  • cannabinoid receptors regulate the Fc epsilon receptor and chemokine induced degranulation of mast cells and basophils. These play important roles in asthma, allergic rhinitis, and other allergic disease. Fc epsilon receptors are stimulated by IgE-antigen complexes. Compounds of the present invention inhibit the Fc epsilon induced degranulation responses, including the basophil cell line, RBL. The ability to inhibit Fc epsilon receptor dependent mast cell and basophil responses results in additional anti-inflammatory and anti-allergic activity for the present compounds. In particular, the present compounds are useful for treating asthma, allergic rhinitis, and other instances of allergic disease.
  • compositions comprising, as an active ingredient, a therapeutically effective amount of at least one of the compounds of formula I, alone or in combination with a pharmaceutical earner or diluent.
  • compounds of the present invention can be used alone, in combination with other suitable therapeutic agents useful in the treatment of the aforementioned disorders including: anti-obesity agents; anti-diabetic agents, appetite suppressants; cholesterol/lipid-lowering agents, HDL-raising agents, cognition enhancing agents, agents used to treat neurodegeneration, agents used to treat respiratory conditions, agents used to treat bowel disorders, anti-inflammatory agents; anti-anxiety agents; anti-depressants; anti-hypertensive agents; cardiac glycosides; and anti-tumor agents.
  • Such other therapeutic agent(s) may be administered prior to, simultaneously with, or following the administration of the cannabinoid receptor modulators in accordance with the invention.
  • suitable anti-obesity agents for use in combination with the compounds of the present invention include melanocortin receptor (MC4R) agonists, melanin-concentrating hormone receptor (MCHR) antagonists, growth hormone secretagogue receptor (GHSR) antagonists, galanin receptor modulators, orexin antagonists, CCK agonists, GLP-1 agonists, and other Pre-proglucagon-derived peptides; NPY1 or NPY5 antagonsist, NPY2 and NPY4 modulators, corticotropin releasing factor agonists, histamine receptor-3 (H3) modulators, aP2 inhibitors, PPAR gamma modulators, PPAR delta modulators, acetyl-CoA carboxylase (ACC) inihibitors, 11- ⁇ -HSD-l inhibitors, adinopect
  • Suitable anti-diabetic agents for use in combination with the compounds of the present invention include: insulin secretagogues or insulin sensitizers, which may include biguanides, sulfonyl ureas, glucosidase inhibitors, aldose reductase inhibitors, PPAR ⁇ agonists such as thiazolidinediones, PPAR ⁇ agonists (such as fibric acid derivatives), PPAR ⁇ antagonists or agonists, PPAR ⁇ / ⁇ dual agonists, 11- ⁇ -HSD-l inhibitors, dipeptidyl peptidase IV (DP4) inhibitors, SGLT2 inhibitors, glycogen phosphorylase inhibitors, and/or meglitinides, as well as insulin, and/or glucagon-like peptide- 1 (GLP-1), GLP-1 agonist, and/or a PTP- IB inhibitor (protein tyrosine phosphatase-lB inliibitor).
  • the antidiabetic agent may be an oral antihyperglycemic agent preferably a biguanide such as metformin or phenformin or salts thereof, preferably metformin HC1. Wl ere the antidiabetic agent is a biguanide, the compounds of the present invention will be employed in a weight ratio to biguanide within the range from about 0.001:1 to about 10:1, preferably from about 0.01:1 to about 5:1.
  • the antidiabetic agent may also preferably be a sulfonyl urea such as glyburide
  • the oral antidiabetic agent may also be a glucosidase inhibitor such as acarbose (disclosed in U.S. Patent No. 4,904,769) or miglitol (disclosed in U.S. Patent No.
  • the compounds of the present invention may be employed in combination with a PPAR ⁇ agonist such as a thiazolidinedione oral anti-diabetic agent or other insulin sensitizers (which has an insulin sensitivity effect inNIDDM patients) such as rosiglitazone (SKB), pioglitazone (Takeda), Mitsubishi's MCC-555 (disclosed in U.S. Patent No.
  • a PPAR ⁇ agonist such as a thiazolidinedione oral anti-diabetic agent or other insulin sensitizers (which has an insulin sensitivity effect inNIDDM patients) such as rosiglitazone (SKB), pioglitazone (Takeda), Mitsubishi's MCC-555 (disclosed in U.S. Patent No.
  • Glaxo-Welcome's GL-262570 englitazone (CP-68722, Pfizer) or darglitazone (CP-86325, Pfizer, isaglitazone (MIT/J&J), JTT-501 (JPNT/P&U), L- 895645 (Merck), R-l 19702 (Sankyo/WL), NN-2344 (Dr. Reddy/NN), or YM-440 (Yamanouchi), preferably rosiglitazone and pioglitazone.
  • the compounds of the present invention may be employed with a PPAR ⁇ / ⁇ dual agonist such as MK-767/KRP-297 (Merck/Kyorin; as described in , K. Yajima, et. al, Am. J. Physiol. Endocrinol. Metab., 284: E966-E971 (2003)), AZ-242 (tesaglitazar; Astra-Zeneca; as described in B. Ljung, et. al., J Lipid Res., 43, 1855- 1863 (2002)); muraglitazar; or the compounds described in US patent 6,414,002.
  • MK-767/KRP-297 Merck/Kyorin; as described in , K. Yajima, et. al, Am. J. Physiol. Endocrinol. Metab., 284: E966-E971 (2003)
  • AZ-242 tesaglitazar; Astra-Zeneca; as described in
  • the compounds of the present invention may be employed in combination with anti-Jiyperlipidemia agents, or agents used to treat arteriosclerosis.
  • An example of an hypolipidemic agent would be an HMG CoA reductase inhibitor which includes, but is not limited to, mevastatin and related compounds as disclosed in U.S. Patent No. 3,983,140, lovastatin (mevinolin) and related compounds as disclosed in U.S. Patent No. 4,231,938, pravastatin and related compounds such as disclosed in U.S. Patent No. 4,346,227, simvastatin and related compounds as disclosed in U.S. Patent Nos. 4,448,784 and 4,450,171.
  • HMG CoA reductase inhibitors which may be employed herein include, but are not limited to, fluvastatin, disclosed in U.S. Patent No. 5,354,772, cerivastatin disclosed in U.S. Patent Nos. 5,006,530 and 5,177,080, atorvastatin disclosed in U.S. Patent Nos. 4,681,893, 5,273,995, 5,385,929 and 5,686,104, pitavastatin (Nissan/Sankyo's nisvastatin (NK-104) or itavastatin), disclosed in U.S. Patent No. 5,011,930, Shionogi-Astra/Zeneca rosuvastatin (visastatin (ZD-4522)) disclosed in U.S.
  • phosphinic acid compounds useful in inhibiting HMG CoA reductase suitable for use herein are disclosed in GB 2205837.
  • the squalene synthetase inhibitors suitable for use herein include, but are not limited to, ⁇ -phosphono-sulfonates disclosed in U.S. Patent No. 5,712,396, those disclosed by Biller, et al, J Med.
  • Chem., 31, 1869-1871 (1998) including isoprenoid (phosphinyl-methyl)phosphonates as well as other known squalene synthetase inhibitors, for example, as disclosed in U.S. Patent No. 4,871,721 and 4,924,024 and in Biller, S.A., Neuenschwander, K., Ponpipom, M.M., and Poulter, CD., Current Pharmaceutical Design, 2, 1-40 (1996).
  • other squalene synthetase inhibitors suitable for use herein include the terpenoid pyrophosphates disclosed by P. Ortiz de Montellano, et al., J Med.
  • hypolipidemic agents suitable for use herein include, but are not limited to, fibric acid derivatives, such as fenofibrate, gemfibrozil, clofibrate, bezafibrate, ciprofibrate, clinofibrate and the like, probucol, and related compounds as disclosed in U.S. Patent No.
  • bile acid sequestrants such as cholestyramine 5 colestipol and DEAE-Sephadex (SECHOLEX, POLICEXLDE) and cholestagel (Sankyo/Geltex), as well as lipostabil (Rhone- Poulenc), Eisai E-5050 (an N-substituted ethanolamine derivative), imanixil (HOE- 402), tetrahydrolipstatin (THL), istigmastanylphos-phorylcholine (SPC, Roche), aminocyclodextrin (Tanabe Seiyoku), Ajinomoto AJ-814 (azulene derivative), melinamide (Sumitomo), Sandoz 58-035, American Cyanamid CL-277,082 and CL- 283,546 (disubstituted urea derivatives), nicotinic acid (niacin), acipimo
  • Patent No. 4,759,923, quaternary amine poly(diallyldimethylammonium chloride) and ionenes such as disclosed in U.S. Patent No. 4,027,009, and other known serum cholesterol lowering agents.
  • the other hypolipidemic agent may be an ACAT inhibitor (which also has anti-atherosclerosis activity) such as disclosed in, Drugs of the Future, 24, 9-15
  • the hypolipidemic agent may be an upregulator of LDL receptor activity such as MD-700 (Taisho Phannaceutical Co. Ltd) and LY295427 (Eli Lilly).
  • the hypolipidemic agent may be a cholesterol absorption inhibitor preferably Schering- Plough' s SCH48461 (ezetimibe) as well as those disclosed in Atherosclerosis 115, 45-63 (1995) and J. Med. Chem. 41, 973 (1998).
  • the other lipid agent or lipid-modulating agent may be a cholesteryl transfer protein inhibitor (CETP) such as Pfizer's CP-529,414 as well as those disclosed in WO/0038722 and in EP 818448 (Bayer) and EP 992496, and Pharmacia's SC-744 and SC-795, as well as CETi-1 and JTT-705.
  • CETP cholesteryl transfer protein inhibitor
  • the hypolipidemic agent may be an ileal Na /bile acid cotransporter inhibitor such as disclosed in Drugs of the Future, 24, 425-430 (1999).
  • the ATP citrate lyase inhibitor which may be employed in the combination of the invention may include, for example, those disclosed in U.S. Patent No. 5,447,954.
  • the other lipid agent also includes a phytoestrogen compound such as disclosed in WO 00/30665 including isolated soy bean protein, soy protein concentrate or soy flour as well as an isoflavone such as genistein, daidzein, glycitein or equol, or phytosterols, phytostanol or tocotrienol as disclosed in WO 2000/015201; a beta-lactam cholesterol absorption inhibitor such as disclosed in EP 675714; an HDL upregulator such as an LXR agonist, a PPAR ⁇ -agonist and/or an FXR agonist; an LDL catabolism promoter such as disclosed in EP 1022272; a sodium-proton exchange inhibitor such as disclosed in DE 19622222; an LDL-receptor inducer or a steroidal glycoside such as disclosed in U.S.
  • a phytoestrogen compound such as disclosed in WO 00/30665 including isolated soy bean protein, soy protein concentrate or soy flour as well as
  • Patent No. 5,698,527 and GB 2304106 an anti-oxidant such as beta-carotene, ascorbic acid, ⁇ -tocopherol or retinol as disclosed in WO 94/15592 as well as Vitamin C and an antihomocysteine agent such as folic acid, a folate, Vitamin B6, Vitamin B12 and Vitamin E; isoniazid as disclosed in WO 97/35576; a cholesterol absorption inhibitor, an HMG-CoA synthase inhibitor, or a lanosterol demethylase inhibitor as disclosed in WO 97/48701; a PPAR ⁇ agonist for treating dyslipidemia; or a sterol regulating element binding protein-I (SREBP-1) as disclosed in WO 2000/050574, for example, a sphingolipid, such as ceramide, or neutral sphingomyelenase (N-SMase) or fragment thereof.
  • SREBP-1 sterol regulating element binding protein-I
  • Preferred hypolipidemic agents are pravastatin, lovastatin, simvastatin, atorvastatin, fluvastatin, pitavastatin and rosuvastatin, as well as niacin and/or cholestagel.
  • the compounds of the present invention may be employed in combination with anti-hypertensive agents.
  • suitable anti-hypertensive agents for use in combination with the compounds of the present invention include beta adrenergic blockers, calcium channel blockers (L-type and/or T-type; e.g.
  • diltiazem verapamil, nifedipine, amlodipine and mybefradil
  • diuretics e.g., chlorothiazide, hydrochlor ⁇ thiazide, flumethiazide, hydroflumethiazide, bendroflumethiazide, methylchlorothiazide, trichloromethiazide, polythiazide, benzthiazide, ethacrynic acid tricrynafen, chlorthalidone, furosemide, musolimine, bumetanide, triamtrenene, amiloride, spironolactone), renin inhibitors, ACE inhibitors (e.g., captopril, zofenop il, fosinopril, enalapril, ceranopril, cilazopril, delapril, pentopril, quinapril, ramipril, lisin
  • Dual ET/AII antagonist e.g., compounds disclosed in WO 00/01389
  • neutral endopeptidase (NEP) inhibitors neutral endopeptidase (NEP) inhibitors
  • vasopepsidase inhibitors dual NEP-ACE inhibitors
  • Cannbinoid receptor modulators could be useful in treating other diseases associated with obesity, including sleep disorders. Therefore, the compounds described in the present invention could be used in combination with therapeutics for treating sleep disorders.
  • Suitable therapies for treatment of sleeping disorders for use in combination with the compounds of the present invention include melatonin analogs, melatonin receptor antagonists, ML 1 B agonists, GAB A receptor modulators; NMD A receptor modulators, histamine-3 (H3) receptor modulators, dopamine agonists and orexin receptor modulators.
  • Cannabinoid receptor modulators may reduce or ameliorate substance abuse or addictive disorders. Therefore, combination of cannabinoid receptor modulators with agents used to treat addictive disorders may reduce the dose requirement or improve the efficacy of current addictive disorder therapeutics.
  • agents used to treat substance abuse or addictive disorders are: selective serotonin reuptake inhibitors (SSRI), methadone, buprenorphine, nicotine and bupropion.
  • Cannabinoid receptor modulators may reduce anxiety or depression; therefore, the compounds described in this application may be used in combination with anti- anxiety agents or antidepressants.
  • suitable anti-anxiety agents for use in combination with the compounds of the present invention include benzodiazepines (e.g., diazepam, lorazepam, oxazepam, alprazolam, chlordiazepoxide, clonazepam, chlorazepate, halazepam and prazepam), 5HT1 A receptor agonists (e.g., buspirone, flesinoxan, gepirone and ipsapirone), and corticotropin releasing factor (CRF) antagonists.
  • benzodiazepines e.g., diazepam, lorazepam, oxazepam, alprazolam, chlordiazepoxide, clonazepam, chlorazepate, halazepam and pra
  • SSRIs selective serot
  • the combination of a conventional antipsychotic drug with a CB-1 receptor antagonist could also enhance symptom reduction in the treatment of psychosis or mania. Further, such a combination could enable rapid symptom reduction, reducing the need for chronic treatment with antipsychotic agents. Such a combination could also reduce the effective antipsychotic dose requirement, resulting in reduced probability of developing the motor dysfunction typical of chronic antipsychotic treatment.
  • Suitable antipsychotic agents for use in combination with the compounds of the present invention include the phenothiazine (chlorpromazine, mesoridazine, thioridazine, acetophenazine, fluphenazine, perphenazine and trifluoperazine), thioxanthine (chlorprothixene, thiothixene), heterocyclic dibenzazepine (clozapine, olanzepine and aripiprazole), butyrophenone (haloperidol), dipheyylbutylpiperidine (pimozide) and indolone (molindolone) classes of antipsychotic agents.
  • phenothiazine chlorpromazine, mesoridazine, thioridazine, acetophenazine, fluphenazine, perphenazine and trifluoperazine
  • thioxanthine chlorprothixene, thioth
  • antipsychotic agents with potential therapeutic value in combination with the compounds in the present invention include loxapine, sulpiride and risperidone. Combination of the compounds in the present invention with conventional antipsychotic drugs could also provide an enhanced therapeutic effect for the treatment of schizophrenic disorders, as described above for manic disorders.
  • schizophrenic disorders include paranoid, disorganized, catatonic, undifferentiated and residual schizophrenia, schizophreniform disorder, shcizoaffective disorder, delusional disorder, brief psychotic disorder and psychotic disorder not specified.
  • Suitable antipsychotic drugs for combination with the compounds in the present invention include the antipsychotics mentioned above, as well as dopamine receptor antagonists, muscarinic receptor agonists, 5HT2A receptor antagonists and 5HT2A/dopamine receptor antagonists or partial agonists (e.g., olanzepine, aripiprazole, risperidone, ziprasidone).
  • the compounds described in the present invention could be used to enhance the effects of cognition-enhancing agents, such as acetylcholinesterase inhibitors (e.g., tacrine), muscarinic receptor- 1 agonists (e.g., milameline), nicotinic agonists, glutamic acid receptor (AMPA and NMD A) modulators, and nootropic agents (e.g., piracetam, levetiracetam).
  • cognition-enhancing agents such as acetylcholinesterase inhibitors (e.g., tacrine), muscarinic receptor- 1 agonists (e.g., milameline), nicotinic agonists, glutamic acid receptor (AMPA and NMD A) modulators, and nootropic agents (e.g., piracetam, levetiracetam).
  • the compounds described in the present invention could be used to enhance the effects of agents used in the treatment of Parkinson's Disease.
  • agents used to treat Parkinson's Disease include: levadopa with or without a COMT inhibitor, antiglutamatergic drugs (amantadine, riluzole), alpha-2 adrenergic antagonists such as idazoxan, opiate antagonists, such as naltrexone, other dopamine agonists or transportor modulators, such as ropinirole, or pramipexole or neurotrophic factors such as glial derived neurotrophic factor (GDNF).
  • GDNF glial derived neurotrophic factor
  • Suitable anti- inflammatory agents for use in combination with the compounds of the present invention include prednisone, dexamethasone, cyclooxygenase inhibitors (i.e., COX-1 and/or COX-2 inhibitors such as NSAIDs, aspirin, indomethacin, ibuprofen, piroxicam, Naproxen®, Celebrex®, Vioxx®), CTLA4-Ig agonists/antagonists, CD40 ligand antagonists, IMPDH inhibitors, such as mycophenolate (CellCept®), integrin antagonists, alpha-4 beta-7 integrin antagonists, cell adhesion inhibitors, interferon gamma antagonists, ICAM-1, tumor necrosis factor (TNF) antagonists (e.g., infliximab, OR1384, including TNF-alpha inhibitors, such as tenidap, anti-TNF antibodies or soluble TNF receptor such as etanercept (Enbrel®), rap
  • cyclosporins e.g., cyclosporin A
  • Anti-Tac anti-IL-2 receptor
  • anti-CD45RB anti-CD2, anti-CD3 (OKT-3)
  • anti-CD4, anti-CD80 anti-CD86
  • monoclonal antibody OKT3 agents blocking the interaction between CD40 and gp39, such as antibodies specific for CD40 and/or gp39 (i.e., CD154), fusion proteins constructed from CD40 and gp39 (CD40Ig and CD8gp39), inhibitors, such as nuclear translocation inhibitors, of NF- kappa B function, such as deoxyspergualin (DSG), gold compounds, antiproliferative agents such as methotrexate, FK506 (tacrolimus, Prograf), mycophenolate mofetil, cytotoxic drugs such as azathiprine and
  • the above other therapeutic agents when employed in combination with the compounds of the present invention, may be used, for example, in those amounts indicated in the Physicians' Desk Reference (PDR) or as otherwise determined by one of ordinary skill in the art.
  • the compounds of formula (I) of the invention can be administered orally or parenterally, such as subcutaneously or intravenously, as well as by nasal application, rectally or sublingually to various mammalian species known to be subject to such maladies, e.g., humans, in an effective amount up to 1 gram, preferably up to 200 mg, more preferably up to 100 mg in a regimen of single, two or four divided daily doses.
  • the compounds of the formula (I) can be administered for any of the uses described herein by any suitable means, for example, orally, such as in the form of tablets, capsules, granules or powders; sublingually; bucally; parenterally, such as by subcutaneous, intravenous, intramuscular, or intrasternal injection or infusion techniques (e.g., as sterile injectable aqueous or non-aqueous solutions or suspensions); nasally, including administration to the nasal membranes, such as by inhalation spray; topically, such as in the form of a cream or ointment; or rectally such as in the form of suppositories; in dosage unit formulations containing non-toxic, pharmaceutically acceptable vehicles or diluents.
  • suitable means for example, orally, such as in the form of tablets, capsules, granules or powders; sublingually; bucally; parenterally, such as by subcutaneous, intravenous, intramuscular, or intrasternal injection or infusion techniques (
  • the present compounds can, for example, be administered in a form suitable for immediate release or extended release. Immediate release or extended release can be achieved by the use of suitable pharmaceutical compositions comprising the present compounds, or, particularly in the case of extended release, by the use of devices such as subcutaneous implants or osmotic pumps.
  • the present compounds can also be administered liposomally.
  • compositions for oral administration include suspensions which can contain, for example, microcrystalline cellulose for imparting bulk, alginic acid or sodium alginate as a suspending agent, methylcellulose as a viscosity enhancer, and sweeteners or flavoring agents such as those known in the art; and immediate release tablets which can contain, for example, microcrystalline cellulose, dicalcium phosphate, starch, magnesium stearate and/or lactose and/or other excipients, binders, extenders, disintegrants, diluents and lubricants such as those known in the art.
  • the compounds of formula I can also be delivered through the oral cavity by sublingual and/or buccal administration.
  • Molded tablets, compressed tablets or freeze-dried tablets are exemplary forms which may be used.
  • Exemplary compositions include those formulating the present compound(s) with fast dissolving diluents such as mannitol, lactose, sucrose and/or cyclodextrins. Also included in such formulations may be high molecular weight excipients such as celluloses (avicel) or polyethylene glycols (PEG).
  • Such formulations can also include an excipient to aid mucosal adhesion such as hydroxy propyl cellulose (HPC), hydroxy propyl methyl cellulose (HPMC), sodium carboxy methyl cellulose (SCMC), maleic anhydride copolymer (e.g., Gantrez), and agents to control release such as polyacrylic copolymer (e.g. Carbopol 934).
  • HPC hydroxy propyl cellulose
  • HPMC hydroxy propyl methyl cellulose
  • SCMC sodium carboxy methyl cellulose
  • maleic anhydride copolymer e.g., Gantrez
  • agents to control release such as polyacrylic copolymer (e.g. Carbopol 934).
  • Lubricants, glidants, flavors, coloring agents and stabilizers may also be added for ease of fabrication and use.
  • compositions for nasal aerosol or inhalation administration include solutions in saline which can contain, for example, benzyl alcohol or other suitable preservatives, absorption promoters to enhance bioavailabihty, and/or other solubilizing or dispersing agents such as those known in the art.
  • compositions for parenteral administration include injectable solutions or suspensions which can contain, for example, suitable non-toxic, parenterally acceptable diluents or solvents, such as mannitol, 1,3-butanediol, water, Ringer's solution, an isotonic sodium chloride solution, or other suitable dispersing or wetting and suspending agents, including synthetic mono- or diglycerides, and fatty acids, including oleic acid, or Cremaphor.
  • suitable non-toxic, parenterally acceptable diluents or solvents such as mannitol, 1,3-butanediol, water, Ringer's solution, an isotonic sodium chloride solution, or other suitable dispersing or wetting and suspending agents, including synthetic mono- or diglycerides, and fatty acids, including oleic acid, or Cremaphor.
  • compositions for rectal administration include suppositories which can contain, for example, a suitable non-irritating excipient, such as cocoa butter, synthetic glyceride esters or polyethylene glycols, which are solid at ordinary temperatures, but liquify and/or dissolve in the rectal cavity to release the drug.
  • a suitable non-irritating excipient such as cocoa butter, synthetic glyceride esters or polyethylene glycols, which are solid at ordinary temperatures, but liquify and/or dissolve in the rectal cavity to release the drug.
  • exemplary compositions for topical administration include a topical carrier such as Plastibase (mineral oil gelled with polyethylene).

Abstract

The present application describes compounds according to Formula (I), pharmaceutical compositions comprising at least one compound according to Formula (I) and optionally one or more additional therapeutic agents and methods of treatment using the compounds according to Formula (I) both alone and in combination with one or more additional therapeutic agents. The compounds have the general Formula (I) including all prodrugs, pharmaceutically acceptable salts and stereoisomers, R1, R2, R3, and R4 are described herein.

Description

AZABICYCLIC HETEROCYC ES AS CANNABINOID RECEPTOR MODULATORS
RELATED APPLICATION This application claims priority benefit under Title 35 § 119(e) of United
States Provisional Application No. 60/531,451, filed December 19, 2003, the contents of which are herein incorporated by reference.
BACKGROUND OF THE INVENTION Delta-9-tetrahydrocannabinol or Delta-9 THC, the principle active component of Cannabis sativa (marijuana), is a member of a large family of lipophilic compounds (i.e., cannabinoids) that mediate physiological and psychotropic effects including regulation of appetite, immunosuppression, analgesia, inflammation, emesis, anti- nocioception, sedation, and intraocular pressure. Other members of the cannabinoid family include the endogenous (arachidonic acid-derived) ligands, anandamide, 2- arachidonyl glycerol, and 2-arachidonyl glycerol ether. Cannabinoids work through selective binding to and activation of G-protein coupled cannabinoid receptors. Two types of cannabinoid receptors have been cloned including CB-1 (L. A. Matsuda, et al., Nature, 346, 561-564 (1990)), and CB-2 (S. Munro, et al., Nature, 365, 61-65 (1993)). The CB-1 receptor is highly expressed in the central and peripheral nervous systems (M. Glass, et al., Neuroscience, 11, 299-318 (1997)), while the CB-2 receptor is highly expressed in immune tissue, particularly in spleen and tonsils. The CB-2 receptor is also expressed on other immune system cells, such as lymphoid cells (S. Galiegue, et al, Eur J Biochem, 232, 54-61 (1995)). Agonist activation of cannabinoid receptors results in inhibition of cAMP accumulation, stimulation of MAP kinase activity, and closure of calcium channels. There exists substantial evidence that cannabinoids regulate appetitive behavior. Stimulation of CB-1 activity by anandamide or Delta-9 THC results in increased food intake and weight gain in multiple species including humans (Williams and Kirkliam, Psychopharm., 143, 315-317 (1999)). Genetic knock-out of CB-1 result in mice that were hypophagic and lean relative to wild-type litter mates (DiMarzo. et al., Nature, 410, 822- 825 (2001)). Published studies with CB-1 small molecule antagonists have demonstrated decreased food intake and body weight in rats (Trillou, et. al., Am. J. Physiol. Regul. Integr. Comp. Physiol, R345-R353, (2003)). Chronic administration of the CB-1 antagonist AM-251 for two weeks resulted in substantial body weight reduction and decreased adipose tissue mass (Hildebrandt, et. al., Eur. J. Phαrm, 462, 125-132 (2003)). There are multiple studies that have assessed the anorexic effect of the Sanofi CB-1 antagonist, SR-141716 (Rowland, et. al., Pyschophαrm., 159, 111-116 (2001); Colombo, et. al., Life Set, 63, 113-117 (1998)). There are at least two CB-1 antagonists in clinical trials for regulation of appetite, Sanofi's SR-141716 and Solvay's SLV-319. Published Phase Hb data reveal that SR- 141716 dose-dependently reduced body weight in human subj ects over a 16 week trial period. CB-1 antagonists have also been shown to promote cessation of smoking behavior. Phase II clinical data on smoking cessation were presented in September of 2002 at Sanofi-Synthelabo's Information meeting. This data showed that 30.2% of patients treated with the highest dose of SR-141716 stayed abstinent from cigarette smoke relative to 14.8% for placebo.
DETAILED DESCRIPTION OF THE INVENTION The present application describes compounds according to Formula I, pharmaceutical compositions comprising at least one compound according to Formula I and optionally one or more additional therapeutic agents and methods of treatment using the compounds according to Formula I both alone and in combination with one or more additional therapeutic agents. The compounds have the general Formula I
Figure imgf000003_0001
I including all prodrugs, pharmaceutically acceptable salts and stereoisomers, R1, R2, R3, and R4 are described herein: DEFINITIONS The following definitions apply to the terms as used tliroughout this specification, unless otherwise limited in specific instances. As used herein, the tenn "alkyl" denotes branched or xmbranched hydrocarbon chains containing 1 to 20 carbons, preferably 1 to 12 carbons-, and more preferably 1 to 8 carbons, in the normal chain, such as, methyl, ethyl, propyl, isopropyl, butyl, sec- butyl, iso-butyl, tert-butyl, pentyl, hexyl, isohexyl, heptyl, 4, -dimethylpentyl, octyl, 2,2,4-trimethylpentyl and the like. Further, alkyl groups, as defined herein, may optionally be substituted on any available carbon atom with one or more functional groups commonly attached to such chains, such as, but not limited to hydroxyl, halo, haloalkyl, mercapto or thio, cyano, alkylthio, cycloalkyl, hetesrocyclyl, aryl, heteroaryl, carboxyl, carbalkoyl, carboxamido, carbonyl. alkenyl, alkynyl, nitro, amino, alkoxy, aryloxy, arylalkyloxy, heteroaryloxy, amido, -OPO H, -OSO3H, and the like to form alkyl groups such as trifluoromethyl, 3-hydroxyhexyl, 2-carboxypropyl, 2-fluoroethyl, carboxymethyl, cyanobutyl and the like. Unless otherwise indicated, the term "alkenyl" as used herein by itself or as part of another group refers to straight or branched chains of 2 to 20 carbons, preferably 2 to 12 carbons, and more preferably 2 to 8 carboπis with one or more double bonds in the nomial chain, such as vinyl, 2-propenyl, 3-butenyl, 2-butenyl, 4- pentenyl, 3-pentenyl, 2-hexenyl, 3-hexenyl, 2-heptenyl, 3-heptenyl, 4-heptenyl, 3- octenyl, 3-nonenyl, 4-decenyl, 3-undecenyl, 4-dodecenyl, 4,5,12-tetradecatrienyl, and the like. Further, alkenyl groups, as defined herein, may optionally be substituted on any available carbon atom with one or more functional groups commonly attached to such chains, such as, but not limited to halo, haloalkyl, alkyl., alkoxy, alkynyl, aryl, arylalkyl, cycloalkyl, amino, hydroxyl, heteroaryl, cycloheteroalkyl, alkanoylamino, alkylamido, arylcarbonylamino, nitro, cyano, thiol, alkylthio and/or any of the alkyl substituents set out herein. Unless otherwise indicated, the term "alkynyl" as used herein by itself or as part of another group refers to straight or branched chains of 2 to 20 carbons, preferably 2 to 12 carbons and more preferably 2 to 8 carbons with one or more triple bonds in the normal chain, such as 2-propynyl, 3-butynyl, 2-fc tynyl, 4-pentynyl, 3- pentynyl, 2-hexynyl, 3-hexynyl, 2-heptynyl, 3-heptynyl, 4-hej>tynyl, 3-octynyl, 3- nonynyl, 4-decynyl,3-undecynyl, 4-dodecynyl and the like. Further, alkynyl groups, as defined herein, may optionally be substituted on any available carbon atom with one or more functional groups commonly attached to such chains, such as, but not limited to halo, haloalkyl, alkyl, alkoxy, alkenyl, aryl, arylalkyl, cycloalkyl, amino, hydroxyl, heteroaryl, cycloheteroalkyl, alkanoylamino, alkylamido, arylcarbonylamino, nitro, cyano, thiol, alkylthio and/or any of the alkyl substituents set out herein Unless otherwise indicated, the term "cycloalkyl" as employed herein alone or as part of another group includes saturated or partially unsaturated (containing one or more double bonds) cyclic hydrocarbon groups containing 1 to 3 rings, appended or fused, including monocyclicalkyl, bicychcalkyl and tricyclicalkyl, containing a total of 3 to 20 carbons foπning the rings, preferably 3 to 10 carbons, forming the ring and which may be fused to 1 or 2 aromatic rings as described for aryl, which include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, cyclodecyl and cyclododecyl, cyclohexenyl,
Figure imgf000005_0001
Further, any cycloalkyl may be optionally substituted through any available carbon atoms with one or more groups selected from hydrogen, halo, haloalkyl, alkyl, alkoxy, haloalkyloxy, hydroxyl, alkenyl, alkynyl, aryl, aryloxy, heteroaryl, heteroaryloxy, arylalkyl, heteroarylalkyl, alkylamido, alkanoylamino, oxo, acyl, arylcarbonylamino, amino, nitro, cyano, tliiol and/or alkylthio and/or any of the alkyl substituents. The term "cycloalkylalkyl" as used herein alone or as part of another group refers to alkyl groups as defined above having a cycloalkyl substituent, wherein said "cycloalkyl" and/or "alkyl" groups may optionally be substituted as defined above. Unless otherwise indicated, the term "aryl" as employed herein alone or as part of another group refers to monocyclic and bicyclic aromatic groups containing 6 to 10 carbons in the ring portion (such as phenyl or naphthyl including 1-naphthyl and 2- naphthyl) and may optionally include one to three additional rings fused to a carbocyclic ring or a heterocyclic ring, for example
Figure imgf000006_0001
Further, "aryl", as defined herein, may optionally be substituted with one or more functional groups, such as halo, alkyl, haloalkyl, alkoxy, haloalkoxy, alkenyl, alkynyl, cycloalkyl, cycloalkylalkyl, heterocyclyl, heterocycloalkyl, aryl, heteroaryl, arylalkyl, aryloxy, aryloxyalkyl, arylalkoxy, alkoxycarbonyl, arylcarbonyl, arylalkenyl, aminocarbonylaryl, arylthio, arylsulfinyl, arylazo, heteroarylalkyl, heteroarylalkenyl, heteroarylheteroaryl, heteroaryloxy, hydroxyl, nitro, cyano, amino, substituted amino wherein the amino includes 1 or 2 substituents (which are alkyl, aryl or any of the other aryl compounds mentioned in the definitions), thiol, alkylthio, arylthio, heteroarylthio, arylthioalkyl, alkoxyarylthio, alkylcarbonyl, arylcarbonyl, alkylaminocarbonyl, arylaminocarbonyl, alkoxycarbonyl, aminocarbonyl, alkylcarbonyloxy, arylcarbonyloxy, alkylcarbonylamino, arylcarbonylamino, arylsulfinyl, arylsulfinylalkyl, arylsulfonylamino or arylsulfonaminocarbonyl and/or any of the alkyl substituents set out herein. Unless otherwise indicated, the term "heteroaryl" as used herein alone or as part of another group refers to a 5- or 6- membered aromatic ring which includes 1 , 2, 3 or 4 hetero atoms such as nitrogen, oxygen or sulfur. Such rings may be fused to an aryl, cycloalkyl, heteroaryl or heterocyclyl and include possible N-oxides as described in Katritzky, A. R. and Rees, C. W., eds. Comprehensive Heterocyclic Chemistry: The Structure, Reactions, Synthesis and Uses of Heterocyclic Compounds 1984, Pergamon Press, New York, NY; and Katritzky, A. R., Rees, C. W., Scriven, E. F., eds. Comprehensive Heterocyclic Chemistry II: A Review of the Literature 1982-1995 1996, Elsevier Science, Inc., Tarrytown, NY; and references therein. Further, "heteroaryl", as defined herein, may optionally be substituted with one or more substituents such as the substituents included above in the definition of "subsiituted alkyl" and "substituted aryl". Examples of heteroaryl groups include the following:
Figure imgf000007_0001
N. S ' \_
Figure imgf000007_0002
and the like. The term "heteroarylalkyl" as used herein alone or as part of another group refers to alkyl groups as defined above having a heteroaryl substituent, wherein said heteroaryl and/or alkyl groups may optionally be substituted as defined above. The term "heterocyclo", "heterocycle", "heterocyclyl" or "heterocyclic ring", as used herein, represents an unsubstituted or substituted stable 4 to 7-membered monocyclic ring system which may be saturated or unsaturated, and which consists of carbon atoms, with one to four heteroatoms selected from nitrogen, oxygen ox sulfur, and wherein the nitrogen and sulfur heteroatoms may optionally be oxidized, and the nitrogen heteroatom may optionally be quaternized. The heterocyclic ring may be attached at any heteroatom or carbon atom which results in the creation of a stable structure. Examples of such heterocyclic groups include, but is not limited to, piperidinyl, piperazinyl, oxopiperazinyl, oxopiperidinyl, oxopyrrolidinyl, oxoazepinyl, azepinyl, pyrrolyl, pyrrolidinyl, furanyl, thienyl, pyrazolyl, pyrazolidinyl, imidazolyl, imidazolinyl, imidazolidinyl, pyridyl, pyrazinyl, pyrimidinyl, pyridazinyl, oxazolyl, oxazolidinyl, isooxazolyl, isoxazolidinyl, morpholinyl, thiazolyl, thiazolidinyl, isothiazolyl, thiadiazolyl, tetrahydropyranyl, thiamo holinyl, thiamorpholinyl sulfoxide, thiamoφholinyl sulfone, oxadiazolyl and other heterocycles described in Katritzky, A. R. and Rees, C. W., eds. Comprehensive Heterocyclic Chemistry: The Structure, Reactions, Synthesis and Uses of Heterocyclic Compounds 1984, Pergamon Press, New York, NY; and Katritzky, A. R., Rees, C. W., Scriven, E. F., eds. Comprehensive Heterocyclic Chemistry II: A Review of the Literature 1982-1995 1996, Elsevier Science, Inc., Tarrytown, NY; and references therein. The term "heterocycloalkyl" as used herein alone or as part of another group refers to alkyl groups as defined above having a heterocyclyl substituent, wherein said heterocyclyl and/or alkyl groups may optionally be substituted as defined above. The terms "arylalkyl", "arylalkenyl" and "arylalkynyl" as used alone or as part of another group refer to alkyl, alkenyl and alkynyl groups as described above having an aryl substituent. Representative examples of arylalkyl include, but are not limited to, benzyl, 2-phenylethyl, 3-phenylpropyl, phenethyl, benzhydryl and naphthylmethyl and the like. The term "alkoxy", "aryloxy", "heteroaryloxy" "arylalkyloxy", or "heteroarylalkyloxy" as employed herein alone or as part of another group includes an alkyl or aryl group as defined above linked through an oxygen atom. The term "halogen" or "halo" as used herein alone or as part of another group refers to chlorine, bromine, fluorine, and iodine, with bromine, chlorine or fluorine being preferred. The term "cyano," as used herein, refers to a -CN group. The term "methylene," as used herein, refers to a -CH2- group. The term "nitro," as used herein, refers to a -NO2 group. The compounds of formula I can be present as salts, which are also within the scope of this invention. Pharmaceutically acceptable (i.e., non-toxic, physiologically acceptable) salts are preferred. If the compounds of formula I have, for example, at least one basic center, they can form acid addition salts. These are formed, for example, with strong inorganic acids, such as mineral acids, for example sulfuric acid, phosphoric acid or a hydrohalic acid, with organic carboxylic acids, such as alkanecarboxylic acids of 1 to 4 carbon atoms, for example acetic acid, which are unsubstituted or substituted, for example, by halogen as chloroacetic acid, such as saturated or unsaturated dicarboxylic acids, for example oxalic, malonic, succinic, maleic, fumaric, phthalic or terephthalic acid, such as hydroxycarboxylic acids, for example ascorbic, glycolic, lactic, malic, tartaric or citric acid, such as amino acids, (for example aspartic or glutamic acid or lysine or arginine), or benzoic acid, or with organic sulfonic acids, such as (C_-C4) alkyl or arylsulfonic acids which are unsubstituted or substituted, for example by halogen, for example methyl- or p- toluene- sulfonic acid. Corresponding acid addition salts can also be formed having, if desired, an additionally present basic center. The compounds of formula I having at least one acid group (for example COOH) can also form salts with bases. Suitable salts with bases are, for example, metal salts, such as alkali metal or alkaline earth metal salts, for example sodium, potassium or magnesium salts, or salts with ammonia or an organic amine, such as moφholine, thiomoφholine, piperidine, pyrrolidine, a mono, di or tri-lower alkylamine, for example ethyl, tert-butyl, diethyl, diisopropyl, triethyl, tributyl or dimethyl-propylamine, or a mono, di or trihydroxy lower alkylamine, for example mono, di or triethanolamine. Corresponding internal salts may furthermore be formed. Salts which are unsuitable for pharmaceutical uses but which can be employed, for example, for the isolation or purification of free compounds of formula I or their pharmaceutically acceptable salts, are also included. Preferred salts of the compounds of formula I which contain a basic group include monohydrochloride, hydrogensulfate, methanesulfonate, phosphate, nitrate or acetate. Preferred salts of the compounds of formula I which contain an acid group include sodium, potassium and magnesium salts and pharmaceutically acceptable organic amines. The term "modulator" refers to a chemical compound with capacity to either enhance (e.g., "agonist" activity) or partially enhance (e.g., "partial agonist" activity) or inhibit (e.g., "antagonist" activity or "inverse agonist" activity) a functional property of biological activity or process (e.g., enzyme activity or receptor binding); such enhancement or inhibition may be contingent on the occurrence of a specific event, such as activation of a signal transduction pathway, and/or may be manifest only in particular cell types. The term "bioactive metabolite" as employed herein refers to any functional group contained in a compound of fonnula I with an open valence for further substitution wherein such substitution can, upon biotransformation, generate a compound of formula I. Examples of such functional groups of bioactive metabolites include, but are not limited to, -OH, -NH or functional groups wherein the hydrogen can be replaced with a functional group such as -PO3H2 for example, which, upon biotransformation generates an -OH or -NH functional group of a compound of formula I. The term "prodrug esters" as employed herein includes esters and carbonates formed by reacting one or more hydroxyls of compounds of formula I with alkyl, alkoxy, or aryl substituted acylating agents employing procedures known to those skilled in the art to generate acetates, pivalates, methylcarbonates, benzoates and the like. Prodrug esters may also include- but are not limited to groups such as phosphate esters, phosphonate esters, phosphonamidate esters, sulfate esters, sulfonate esters, and sulfonamidate esters wherein the ester may be further substituted with groups that confer a pharmaceutical advantage such as-but not limited to-favorable aqueous solubility or in vivo exposure to the bioactive component formula I. The term "prodrug" as employed herein includes functionalization of bioactive amine- or hydroxyl-containing compounds of formula I to form alkyl-, acyl-, sulfonyl-, phosphoryl-, or carbohydrate-substituted derivatives. Such derivatives are fonned by reacting compounds of formula I with alkylating-, acylating-, sulfonylating-, or phosphorylating reagents employing procedures known to those skilled in the art. Alkylation of amines of formula I may result in- but are not limited to- derivatives that include spacer units to other prodrug moieties such as substituted alkyoxymethyl-, acyloxymethyl-, phosphoryloxymethyl-, or sulfonyloxymethyl- groups. Alkylation of amines of formula I may result in the generation of quarternary amine salts that act in vivo to provide the bioactive agent (i.e., the compound of formula I). Preferred prodrugs consist of a compound of formula I where a pendant hydroxyl is phosphorylated to generate a phosphate derivative. Such a prodrug may also include a spacer group between the compound of formula I and the phosphate group, such as a methyleneoxy-group. Methods to generate such a prodrug from a compound of formula I are known to those skilled in the art, and are listed in the references below. Preferred prodrugs also consist of a compound of formula I where a pendant amine, such as a pyridine group, is alkylated with a group, such as methyl, to form a quarternary ammonium ion salt. Methods to generate such a prodrug from a compound of formula I are known to those skilled in the art, and are listed in the references below. Any compound that can be converted in vivo to provide the bioactive agent (i.e., the compound of formula I) is a prodrug within the scope and spirit of the invention. Various forms of prodrugs are well known in the art. A comprehensive description of prodrugs and prodrug derivatives are described in: The Practice of Medicinal Chemistry, Camille G. Wermuth et al., Ch 31, (Academic Press, 1996); Design of Prodrugs, edited by H. Bundgaard, (Elsevier, 1985); A Textbook of Drug Design and Development, P. Krogsgaard-Larson and H. Bundgaard, eds. Ch 5, pgs 113 - 191 (Harwood Academic Publishers, 1991). Hydrolysis in Drug and Prodrug Metabolism, B. Testa and J. M. Mayer, (Verlag Helvetica Chimica Acta AG, Zurich, Switzerland; Wiley- VCH, Weinheim, Federal Republic of Germany, 2003); Ettmayer, P.; Amidon, G. L.; Clement, B.; Testa, B. "Lessons Learned from Marketed and Investigational Prodrugs" J. Med. Chem. 2004, 47 (10), 2393-2404; and Davidsen, S. K. et al. "N-(Acyloxyalkyl)pyridinium Salts as Soluble Prodrugs of a Potent Platelet Activating Factor Antagonist" J. Med. Chem. 1994, 37 (26), 4423- 4429.
Said references are incoφorated herein by reference. An administration of a therapeutic agent of the invention includes administration of a therapeutically effective amount of the agent of the invention. The term "therapeutically effective amount" as used herein refers to an amount of a therapeutic agent to treat or prevent a condition treatable by administration of a composition of the invention. That amount is the amount sufficient to exhibit a detectable therapeutic or preventative or ameliorative effect. The effect may include, for example, treatment or prevention of the conditions listed herein. The precise effective amount for a subject will depend upon the subject's size and health, the nature and extent of the condition being treated, recommendations of the treating physician, and the therapeutics or combination of therapeutics selected for administration. All stereoisomers of the compounds of the instant invention are contemplated, either in mixture or in pure or substantially pure form. The compounds of the present invention can have asymmetric centers at any of the carbon atoms including any one of the R substituents. Consequently, compounds of formula I can exist in enantiomeric or diastereomeric forms or in mixtures thereof. The processes for preparation can utilize racemates, enantiomers or diastereomers as starting materials. When diastereomeric or enantiomeric products are prepared, they can be separated by conventional methods for example, chromatographic techniques, chiral HPLC or fractional crystallization. The compounds of formula I of the invention can be prepared as shown in the following reaction schemes and description thereof, as well as relevant published literature procedures that may be used by one skilled in the art. Exemplary reagents and procedures for these reactions appear hereinafter and in the working Examples. ABBREVIATIONS The following abbreviations are employed in the Schemes, Examples and elsewhere herein:
Ac = acetyl
AcOH = acetic acid Boc = tert-butoxycarbonyl
DCM = dichloromethane
DIPEA = N,N-diisopropylehtylamine
DMF = N,N-dimethylformamide
EDAC = l-(3-Dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride EtOAc = ethyl acetate
Et3N = triethylamine
Et2O = diethyl ether HOBt = 1-hydroxybenzotriazole hydrate
HPLC = high performance liquid chromatography
LAH = lithium aluminum hydride
MeOH = methanol MS or Mass Spec = mass spectrometry
NaOH = sodium hydroxide
PG = protecting group
RT = room temperature
TFA = trifluoroacetic acid THF = tetrahydrofuran min = minute(s) h = hour(s)
L = liter mL = milliliter μL = microliter g = gram(s) mg = milligram(s) mol = moles mmol = millimole(s) nM = nanomolar
Compounds of the present invention may be prepared by procedures illustrated in the accompanying schemes. METHODS OF PREPARATION The compounds of the present invention may be prepared by methods such as those illustrated in the following Scheme 1 to 7. Solvents, temperatures, pressures, and other reaction conditions may readily be selected by one of ordinary skill in the art. Starting materials are commercially available or can be readily prepared by one of ordinary skill in the art using known methods. For all of the schemes and compounds described below, R1, R2, R3 and R4 are as described for a compound of formula I. The following are the definitions of symbols used throughout Schemes 1 to 7: PG suitable nitrogen protecting group, exemplified by benzyl, methoxymethyl- [MOM], benzyloxymethyl- [BOM], 2-(trimethylsilyl)ethoxymethyl- [SEM], methoxyethoxymethyl- [MEM], or t-butyl groups; EE Sn2 or Snl leaving group exemplified by halogen (CI, Br, I) and sulfonates (- OSO2-aryl (e.g., -OSO2Ph or -OSO2PhCH3), or -OSO2-alkyl (e.g., -OSO2CH3 or -OSO2CF3)); MM boronate ester or boronic acid, or trialkylstannane; or metal atom such as zinc, magnesium or lithium as part of an organometallic compound used as an intermediate for transition metal mediated coupling reactions.
SCHEME 1
Figure imgf000014_0001
Compounds of formula II are either commercially available or available by means known to one skilled in the art. Compounds of formula III can be prepared by reacting compounds of formula II with an appropriate protecting group such as benzyl bromide. Exemplary nitrogen protecting groups and methods of protecting the nitrogen are similar to those for protecting amines, such as those described in T. W. Greene and P. G. M. Wuts, Protecting Groups in Organic Synthesis, John Wiley & Sons, Inc, New York, 1991. Preferred nitrogen protecting groups are benzyl, tert- butyl, methoxymethyl (MOM), methoxyethoxymethyl (MEM), and 2- (trimethylsilyl)ethoxymethyl (SEM) groups. Compounds of formula IV may be prepared from compounds of formula III via selective displacement of the leaving group (EE) by the conjugate base of an appropriate alcohol, RO-M, wherein R is alkyl or benzyl, and M is a metalloid such as Li, Na, Mg(halide) and the like in solvents such as dioxane. Similar reactions has been described in the literature (Riedl, Z. et. al. Tetrahedron, 2002, 5645-5650). Compounds of formula V can be prepared by the reactions of compounds of formula IV with activated R , such as activated by boronic acids, tin, Grignard reagents, Zinc, Cu, etc in the presence of an appropriate catalyst if needed such as Pd(PPh3)4. Compounds of formula V may also be prepared from a compound of formula IV via displacement of the leaving group (EE) by the conjugate base of a 9 9 compound R -H, wherem R is as previously defined, using a base in an inert solvent.
Exemplary bases include sodium carbonate, potassium carbonate, cesium carbonate, sodium hydride, potassium hydride, or alkyl lithiums. Compounds of formula VI , where EE = CI, can be prepared by reacting compounds of formula V with a chlorinating agent such as POCl3 in an inert solvent such as toluene at elevated temperature. Compounds of formula VII, where MM is a metal or a borate ester, may be prepared via lithiation of a compound of formula VI wherein EE is hydrogen or a halogen (chloro, bromo, iodo), and reacting the resulting aryl lithium with an appropriate borate derivative or with reagents such as trialkyltin halide. Compounds of formula VIII can be prepared by the reactions of compounds of formula VI with activated R1, such as activated by boronic acids, tin, Grignard reagents, Zinc, Cu, etc in the presence of an appropriate catalyst if needed such as Pd(PPh3)4. Compounds of formula VIII may also be prepared from a compound of formula VI via displacement of the leaving group (EE) by the conjugate base of a compound R -H, wherein R1 is as previously defined, using a base in an inert solvent. Compounds of formula VIII can also be prepared by a palladium or nickel catalyzed coupling of a compound of formula VII wherein MM is a borate ester with an appropriately activated R1 such as halide or mesylate. When MM is a metal atom such as tin, zinc, magnesium, and lithium, similar cross-coupling reactions can be performed using activated R1 such as halide or borate esters with an appropriate catalyst such as tetrakis(triphenylphosphine)palladium(0) and dichlorobis(triphenylphosphine)nickel(II). Compounds of formula IX can be prepared by removing the protecting group
(PG) in compound VIII under acidic (e.g. TFA for t-butyl or Boc-), basic (e.g. NaOH for amide), catalytic hydrogenation (for benzyl-), or Lewis acid (e.g. A1C13 for benzyl) conditions. Compounds of formula X can be prepared by reacting compounds of formula IX with a chlorinating agent such as POCl3 in an inert solvent such as toluene at elevated temperature. Compounds of formula XI can be prepared by reacting compounds of formula X with hydrazine in an inert solvent such as pyridine at elevated temperature. Compounds of formula I can be prepared by reacting compounds of formula XI with an alpha-ketoacid or alpha-ketoacid ester in a protic solvent, such as an alkanoic acid for example, acetic acid in ambient to evelatated temperatures.
SCHEME 2
Figure imgf000017_0001
III XIII XIV
as in Scheme 1
Figure imgf000017_0002
Figure imgf000017_0003
As illustrated in Scheme 2, compounds of formula XIII may be prepared from compounds of formula III via selective displacement of the leaving group (EE) by the conjugate base of an appropriate alcohol, RO-M, wherein R is alkyl or benzyl, and M is a metal such as Li, Na, Mg(halide) and the like in solvents such as methanol. Such selective displacements, for example when EE = CI, has been reported in the literature. ( Riedl, Z. et. al. Tetrahedron, 2002, 5645-5650). Compounds of formula XIV can be prepared by the reactions of compounds of formula XIII with activated R1, such as activated by boronic acids, tin, Grignard reagents, Zinc, Cu, etc in the presence of an appropriate catalyst if needed such as Pd(PPh3)4. Compounds of formula XIV may also be prepared from a compound of formula XIII via displacement of the leaving group (EE) by the conjugate base of a compound R^H, wherein R1 is as previously defined, using a base in an inert solvent. Compounds of formula XV , where EE = CI, can be prepared by reacting compounds of formula XIV with a chlorinating agent such as POCl3 in an inert solvent such as toluene at elevated temperature. Alternatively, compounds of formula XV , where EE = CI, can also be prepared by the reactions of compounds of formula III with activated R1, such as activated by boronic acids, tin, Grignard reagents, Zinc, Cu, etc in the presence of an appropriate catalyst if needed such as Pd(PPh3)4. Compounds of formula XVI, where MM is a metal or a borate ester, may be prepared via lithiation of a compound of formula XV wherein EE is hydrogen or a halogen (chloro, bromo, iodo), and reacting the resulting aryl lithium with an appropriate borate derivative or with reagents such as trialkyltin halide. Compounds of formula VIII can be prepared by the reactions of compounds of formula XV with activated R2, such as activated by boronic acids, tin, Grignard reagents, Zinc, Cu, etc in the presence of an appropriate catalyst if needed such as Pd(PPh3)4. Compounds of formula VIII may also be prepared from a compound of formula XV via displacement of the leaving group (EE) by the conjugate base of a 9 9 compound R -H, wherein R is as previously defined, using a base in an inert solvent. Compounds of formula VIII can also be prepared by a palladium or nickel catalyzed coupling of a compound of formula XVI wherein MM is a borate ester with an appropriately activated R such as halide or mesylate. When MM is a metal atom such as tin, zinc, magnesium, and lithium, similar cross-coupling reactions can be performed using activated R such as halide or borate esters with an appropriate catalyst such as tetrakis(triphenylphosphine)palladium(0) and dichlorobis(triphenylphosphine)nickel(II). Compounds of formula I may be prepared from a compound of formula VIII as described in Scheme 1.
SCHEME 3
Figure imgf000019_0001
Figure imgf000019_0002
XVIII XIX
Figure imgf000019_0003
As illustrated in Scheme 3, compounds of formula XVII may be prepared from a compound of formula XIII (from Scheme 2) via displacement of the R group (R = alkyl) using bases such as sodium hydroxide in a suitable solvent such as water. The hydroxyl group in XVII can be activated by reacting with reagents such as trifluromethane sulfonic anhydride in the presence of a suitable base such as triethylamine. This activated moiety can then be selectively coupled with activated R", such as activated, by boronic acids, tin, Grignard reagents, Zinc, Cu, etc in the presence of an appropriate catalyst if needed such as Pd(PPh3)4 to provide compounds XVIII. Compounds of formula XIX can be prepared from compounds XVIII in a two step sequence: (a) by removing the protecting group (PG) in compound XVIII under acidic (e.g. TFA for t-butyl or Boc-), basic (e.g. NaOH for amide), catalytic hydrogenation (for benzyl-), or Lewis acid (e.g. A1C13 for benzyl) conditions, followed by (b) reacting the resulting intermediate with a chlorinating agent such as POCl3 in an inert solvent such as toluene at elevated temperature. In certain instances, direct treatment of XVIII with chlorinating agents such as POCl3 at higher temperature may provide compounds of formula XIX in one step from XVTII. Compounds of formula XX can be prepared by reacting compounds of formula XIX with hydrazine in selected solvents such as isobutanol. Compounds of formula XXI can be prepared by reacting compounds of formula XX with an alpha-ketoacid or alpha-ketoacid ester in a protic acid, such as acetic acid at ambient to evelvated temperature. . Compounds of formula I can be prepared by the reactions of compounds of formula XXI with activated R1, such as activated by boronic acids, tin, Grignard reagents, Zinc, Cu, etc in the presence of an appropriate catalyst if needed such as
Pd(PPh3)4. Compounds of formula I may also be prepared from a compound of formula XXI via displacement of the leaving group (EE) by the conjugate base of a compound R^H, wherein R1 is as previously defined, using a base in an inert solvent. Compounds of formula XXII, where MM is a metal or a borate ester, may be prepared via lithiation of a compound of formula XXI wherein EE is hydrogen or a halogen (chloro, bromo, iodo), and reacting the resulting aryl lithium with an appropriate borate derivative or with reagents such as trialkyltin halide. Compounds of formula I can also be prepared by a palladium or nickel catalyzed coupling of a compound of formula XXII wherein MM is a borate ester with an appropriate activated R1 such as halide or mesylate. When MM is a metal atom such as tin, zinc, magnesium, and lithium, similar cross-coupling reactions can be performed using activated R1 such as halide or borate esters with an appropriate catalyst such as tetrakis(triphenylphosphine)palladium(0) and dichlorobis(triphenylphosphine)nickel(II).
SCHEME 4
Figure imgf000021_0001
XV XXIV XXV
Figure imgf000021_0002
As illustrated in Scheme 4, compounds of formula XXIV can be prepared from compounds XV in a two step sequence: (a) by removing the protecting group (PG) in compound XV under acidic (e.g. TFA for t-butyl or Boc-), basic (e.g. NaOH for amide), catalytic hydrogenation (for benzyl-), or Lewis acid (e.g. A1C13 for benzyl) conditions, followed by (b) reacting the resulting intermediate with a chlorinating agent such as POCl3 in an inert solvent such as toluene at elevated temperature Compounds of formula XXV can be prepared by reacting compounds of formula XXIV with hydrazine in selected solvents such as isobutanol. Compounds of formula XXVII can be prepared by reacting compounds of formula XXV with an alpha-ketoacid or alpha-ketoacid ester in a protic acid, such as acetic acid at ambient to evelated temperatures. Target compounds of formula I can be prepared from compounds of formula XXVII by following an analogous sequence of reactions as described in Scheme 3 via intermediates XI, XXVII and XXVIII.
SCHEME 5 as in Scheme 1
Figure imgf000022_0002
Figure imgf000022_0001
As illustrated in Scheme 5, for compounds of formula I in which R1 and R2 are the same group (equal), compounds of fonnula VIII can prepared by the reactions of compounds of formula III with activated R1 and R2, such as activated by boronic acids, tin, Grignard reagents, Zinc, Cu, etc in the presence of an appropriate catalyst if needed such as Pd(PPh3)4. Compounds of formula VIII may also be prepared from a compound of fonnula III via displacement of the leaving group (EE) by the conjugate 1 1 base of a compound R -H and R -H, wherein R and R is as previously defined, using a base in an inert solvent. Target compounds of formula I can then be prepared from compounds of formula VIII by following an analogous sequence of reactions as described in Scheme 1.
SCHEME 6
As illustrated in Scheme 6, compounds of formula X can be prepared by the reactions of compounds of formula XXIV with activated R , such as activated by boronic acids, tin, Grignard reagents, Zinc, Cu, etc in the presence of an appropriate catalyst if needed such as Pd(PPh3)4. Compounds of formula I can then be prepared from compounds of formula I by following an analogous sequence of reactions as described in Scheme 1.
SCHEME 7
Figure imgf000023_0001
As illustrated in Scheme 7, analogs having certain arbitrarily defined subsets of R1 and R2 and R3 can be changed to other analogs having certain other arbitrarily defined subsets of R1 and R2 and R3 by manipulation of the functional groups embedded in these R groups. For example, when Rla, R2a, R3a are groups such as aminoaryl, aminoalkyl, aminoheterocyclyl, or aminoheteroaryl, the amino group can be reacted with either carboxylic acids or acid chlorides or sulfonyl chlorides to provide amide or sulfonamide derivatives. Such a manipulation can also be conducted via parallel synthesis. In addition, when Rla, R2a or R3a are substituted with an activated group such as a halogen or boronic acid, additional metal catalyzed cross-coupling reactions may be performed to provide additional set of analogs as described by Formula I. Such a manipulation can also be conducted via parallel synthesis. Parallel synthesis may be employed in the preparation of compounds, for example, where the intermediates possess an activated reaction center: such as but not limited to a reactive heteroaryl cliloride for Suzuki coupling chemistry or a carboxylic acid for amide coupling chemistry.
EXAMPLES The following Examples serve to better illustrate, but not limit, some of the prefereed embodiments of the invention.
Analytical HPLC Methods Employed in Characterization of Examples.: Analytical HPLC/MS was performed on Shimadzu LC10AS liquid chromatographs and Waters ZMD Mass Spectrometers using the following method:
Method A. Linear gradient of 0 to 100% solvent B over 4 min, with 1 min hold at 100% B; UV visualization at 220 nm Column: Phenomenex Luna C18 4.6 x 50 mm Flow rate: 4 ml/min Solvent A: 0.2% phosphoric acid, 90% water, 10%) methanol Solvent B: 0.2% phosphoric acid, 90% methanol, 10% water
NMR Employed in Characterization of Examples H NMR spectra were obtained with Bruker or JOEL fourier transform spectrometers operating at the following frequencies: 1H NMR: 400 MHz (Bruker), 400 MHz (JOEL), or 500 MHz (JOEL); 13C NMR: 100 MHz (Bruker), 100 MHz
(JOEL) or 125 MHz (JOEL). Spectra data are reported as Chemical shift (multiplicity, number of hydrogens, coupling constants in Hz) and are reported in ppm(δ units) relative to either an internal standard (tetramethylsilane = 0 ppm) for 1H NMR spectra, or are referenced to the residual solvent peak (2.49 ppm for CD2HSOCD3, 3.30 ppm for CD2HOD, 7.24 ppm for CHC13, 39.7 ppm for CD3SOCD3, 49.0 ppm for CD3OD, 77.0 ppm for CDC13). All 13C NMR spectra were proton decoupled. EXAMPLE 1 Preparation of 8,9-Bis-(4-chIoro-phenyl)-3-isobutyl-pyridazino[6,l- c] [l,2,4]triazin-4-one
Figure imgf000026_0001
1 Preparation of 2-Benzyl-4,5-dibromopyridazin-3(2H)-one
Figure imgf000026_0002
To a solution of dibromopyridazinone (50.0 g, 197.0 mmol) in DMF (200 mL) at RT was added K2CO3 (32.6 g, 236.4 mmol). Benzylbromide (37.0 g, 216.7 mmol) was added via a syringe. The resulting greenish suspension was stireed at RT for 6 h until all the pyridazinone was consumed as judged by HPLC. The reaction mixture was then poured into an Erlenmeyer flask containing water (500 mL) with stirring. A beige colored solid formed. The suspension was stirred for 15 min at RT and then was filtered. The solid was rinsed thoroughly with water until no color was apparent in the filtrate. The solid was dried in a vacuum oven at 50°C overnight. The title compound, 2-benzyl-4,5-dibromopyridazin-3(2H)-one, (68.0 g, 196.5 mmol) was >95 pure as judged by HPLC and was obtained as a solid. MS: M+H = 343. 1H NMR (CDC13, 500 MHz): δ 7.80 (IH, s), 7.45 (2H, d, J=5.0 Hz), 7.29-7.36 (3H, m), 5.31 (2H, s).
IB. Preparation of 2-Benzyl-4,5-bis(4-chlorophenyl)pyridazin-3(2H)-one
Figure imgf000026_0003
To a suspension of 2-benzyl-4,5-dibromopyridazin-3(2H)-one (40 g, 116.0 mmol) in toluene (300 mL) was added Pd(PPh3)4 (4.0 g, 3.5 mmol) under an atmosphere of agron. 4-Chlorophenylboronic acid (40.0 g, 255.2 mmol) was added subsequently portionwise. Under vigorous stirring, Na2CO3 (27.0 g, 255.2 mmol) dissolved in water (50 mL) was added to the suspension. Argon was bubbled through this suspension for 10 min. before the flask was placed in an oil bath preheated at 120°C. The reaction was refluxed for 6 h. The reaction was then allowed to cool to RT and was poured into water (500 mL). The aqueous mixture was extracted with EtOAc (3 x 300 mL). The combined organic layers were washed with NaOH (0.5 N, 200 mL) and water (2 x 500 mL). The organic layer was filtered through a silica gel pad (~50 g) in a sintered glass funnel to remove dark color impurities. Solvents were then evaporated under reduced pressure. The resultant thick syrup contained predominantly the title compound 2-benzyl-4,5-bis(4-chlorophenyl)pyridazin-3(2H)- one which could be used directly in the next reaction, IC. MS: M+H = 407. 1H NMR (CDC13, 500 MHz): δ 7.87 (IH, s), 7.54 (2H, d, J=5.0 Hz), 7.32-7.38 (3H, m), 7.22- 7.28 (4H, m), 7.12 (2H, d, J-10.0 Hz), 7.03 (2H, d, J=10 Hz), 5.40 (2H, s).
IC. Preparation of 4,5-bis(4-chlorophenyl)pyridazin-3(2H)-one
Figure imgf000027_0001
The crude 2-benzyl-4,5-bis(4-chlorophenyl)pyridazin-3(2H)-one from the above reaction was dissolved in toluene (350 mL). Aluminum chloride (A1C13, 46.3 g, 348.0 mmol) was then added to the toluene solution, producing an exotherm. The reaction was then placed in an oil bath preheated at 75°C for 3 h. After this time, the reaction mixture was poured into ice- water (1000 mL), and the resulting mixture was extracted with EtOAc (3 x 500 mL). The combined organic layers were washed with water (500 mL), then filtered through a silica gel pad (-50 g) to remove residual alumina salts. The organic solvents were evaporated under reduced pressure to nearly dryness. Diethyl ether (Et2O, 500 mL) was added with stining. After stirring for 15 min at R.T, hexane (1000 mL) was added subsequently. The resultant beige colored solid was collected by filtration and subsequently washed with a hexane-ether mixture (8:2). The title compound, 4,5-bis(4-chlorophenyl)pyridazin-3(2H)-one, (33.0 g, 92%) was obtained as a solid. MS: M+H = 317. 1H NMR (CDC13, 500 MHz): δ 11.94 (IH, br), 7.89 (IH, s), 7.23-7.32 (4H, m), 7.18 (2H, d, J=10.0 Hz), 7.07 (2H, d, J=10 Hz).
ID. Preparation of 3-Chloro-4,5-bis(4-chlorophenyl)pyridazine
Figure imgf000028_0001
4,5-Bis(4-chlorophenyl)pyridazin-3(2H)-one (16.5 g, 52.2 mmol) was suspended in toluene (50 mL). To the resultant solution was added pyridine (8.3 mL, 104.4 mmol), followed by the addition of POCl3 (14.3 mL, 156.6 mmol). The reaction mixture was placed in an oil bath preheated at 110°C. After 4 h, the reaction mixture was cooled to RT, then poured over 500 g ice to quench the excess POCl3. The dark mixture was extracted with EtOAc (2 x 300 mL). The combined organic layers were washed with water (500 mL), and filtered through a silica gel pad (~ 50 g). The filtrate was concentrated under reduced pressure to give the title compound, 3-chloro-4,5-bis(4-chlorophenyl)pyridazine as a pale colored solid (16.0 g, 92%). MS: M+-H = 335. 1H NMR (CDC13, 500 MHz): δ 9.24(1H, s), 7.35 (2H, d, J=10 Hz), 7.26 (2H, d, J=l 0.0 Hz), 7.08-7.16 (4H, m).
IE. Preparation of l-(4,5-Bis(4-chlorophenyl)pyridazin-3-yl)hydrazine
Figure imgf000028_0002
3-Chloro-4,5-bis(4-chlorophenyl)pyridazine (10.0 g, 30.0 mmol) was dissolved in pyridine (30 mL) and hydrazine mono-hydrate was added (4.5 g, 90.0 mmol). The reaction mixture was refluxed for 3 h and was then added to water (100 mL). The pale colored solid was collected by filtration and rinsed thoroughly with water. The product was dried in a vacuum oven to give the title compound, l-(4,5- bis(4-chlorophenyl)pyridazin-3-yl)hydrazine (9.5 g, 96%). MS: M+H = 331. 1H NMR (DMSO-d6, 500 MHz): δ 8.59(1H, s), 8.56 (IH, br), 7.40 (2H, d, J=10 Hz), 7.35 (2H, d, J=10.0 Hz), 7.14-7.16 (4H, m).
IF. Preparation of 8,9-Bis-(4-chloro-phenyl)-3-isobutyl-pyridazmo[6,l- c] [l,2,4]triazin-4-one
Figure imgf000029_0001
To a solution of l-(4,5-bis(4-chlorophenyl)pyridazin-3-yl)hydrazine (50.0 mg, 0.15 mmol) in AcOH (lmL) was added (CH3)2CH2COCO2H (40.0 mg, 0.3 mmol). The solution was stined at RT for 30 min then was heated at 120°C for 5 h. The reaction mixture was cooled to RT and then poured into water (20 mL). The resultant solution was extracted with EtOAc (20 mL). The organic laer was dried over Na2SO4, filered and concentrate to give the a brown solid. The solid was purified by reverse phase preparative HPLC to give the title compound 9-bis-(4-chloro-phenyl)-3- isobutyl-pyridazino[6,l-c][l,2,4]triazin-4-one, (23.0 mg, 0.054 mmol) as a solid. HPLC retention time = 3.87min; MS observed = 425 (M+H); 1H NMR (CDC13, 500 MHz) δ 8.72 (s, IH), 7.37-7.33 (m, 4H), 7.24 (d, J = 8.8 Hz, 2H), 7.11 (d, J = 8.8 Hz, 2H), 2.96 (d, J = 7.1 Hz, 2H), 2.34 (m, IH), 0.99 (d, J = 9.3 Hz, 6H).
EXAMPLE 2 8,9-Bis(4-chlorophenyl)-3-(4-fluorophenyl)-4H-pyridazino[6,l-c][l,2,4]triazin-4- one
Figure imgf000030_0001
The title compound was prepared using the method described in Example IE, reacting l-(4,5-bis(4-chlorophenyl)pyridazin-3-yl)hydrazine with 2-(4-fluorophenyl)- 2-oxoacetic acid to give 8,9-bis(4-chlorophenyl)-3-(4-fluorophenyl)-4H- pyridazino[6,l-c][l,2,4]triazin-4-one. HPLC retention time = 3.84min; MS observed = 463 (M+H); 1HNMR (CDC13, 500 MHz) δ 8.76 (s, IH), 8.58-8.53 (m, 2H), 7.39- 7.34 (m, 4H), 7.31-7.25 (m, 2H), 7.22-7.13 (m, 4H).
The compounds of Set A below, in which R3 varies, R1 is 4-chlorophenyl, R2 is 4-chlorophenyl, and R4 is hydrogen may be prepared by one skilled in the art by the methods described above. The compounds of Set A are meant to further illustrate the scope of the invention without being limiting in any way.
Figure imgf000030_0002
Figure imgf000031_0001
Figure imgf000032_0001
As noted above, Set A consists of compounds that differ from one another only in the identity of R3 with R1 and R2 fixed as 4-chlorophenyl. Set A may be considered a one dimensional library of example compounds. Were one to vary both R and R , a two dimensional library of example compounds would result. Set B is the two dimensional library that consists of all permutations of all of the variants of R3 represented in Set A and a set of R2 variants listed below. In Set B, R1 is 4- chlorophenyl, R4 is hydrogen, The compounds of Set B may be prepared by one skilled in the art by the methods described above. The compounds of Set B are meant to further illustrate the scope of the invention without being limiting in any way.
Figure imgf000033_0001
R variants of Set B:
Figure imgf000033_0002
Figure imgf000034_0001
Further, as noted above, Set B is the two dimensional library that consists of all permutations of all of the variants of R3 represented in Set A and a set of R2 variants listed above with R fixed as 4-chlorophenyl. Were one to vary R and R and R , a three dimensional library of example compounds would result. Set C is the three dimensional library that consists of all permutations of all of the variants of R3 represented in Set A, all of the R2 variants listed above for Set B, and a set of R1 variants listed below. In Set C, R4 is hydrogen. The compounds of Set C may be prepared by one skilled in the art by the methods described above. The compounds of Set C are meant to further illustrate the scope of the invention without being limiting in any way.
Rά variants of Set C
Figure imgf000035_0001
Biological Evaluation Cannabinoid Receptor Binding Assay Radioligand binding studies were conducted in membranes prepared from Chinese Hamster Ovary (CHO) cells that over-express recombinant human CB-1 (CHO-CB-1 cells). Total assay volume for the binding studies was 100 μl. 5 μg of membranes were brought up to a final volume of 95 μl with Binding Buffer (25 mM HEPES, 150 mM NaCl, 2.5 mM CaCl2, 1 mM MgCl2, 0.25% BSA). The diluted membranes were preincubated with a compound or DMSO vehicle. The binding reaction was initiated by the addition of 2 nM final 3H-CP-55,940 (120 Ci/mmol) and proceeded for 2.5 hours at room temperature. The binding reaction was terminated by transfening the reaction to GF B 96 well plates (presoaked with 0.3% polyethylenimine) using a Packard Cell Harvester. The filter was washed with 0.25x PBS, 30 μl MicroScint was added per well, and the bound radiolabel was quantitated by scintillation counting on a Packard TopCount Scintillation Counter. The CB-2 radioligand binding assay was conducted identically except that the membranes from CHO-CB-2 cells were used. For a compound to be considered a CB-1 antagonist, the compound must possess a CB-1 receptor binding affinity Ki less than 13000 nM. As determined by the assay described above, the CB-1 receptor binding K, values of working Examples 1-63 fall within the range of 0.01 nM to 10000 nM.
Cannabinoid Receptor Functional Activity Assay Functional CB-1 inverse agonist activity of test compounds was determined in CHO-CB-1 cells using a cAMP accumulation assay. CHO-CB-1 cells were grown in 96 well plates to near confluence. On the day of the functional assay, growth medium was aspirated and 100 of Assay Buffer (PBS plus 25 mM HEPES / 0.1 mM 3- isobutyl-1-methylxanthine/ 0.1 % BSA) was added. Compounds were added to the Assay buffer diluted 1:100 from 100% DMSO and allowed to preincubate for 10 minutes prior to addition of 5 uM forskolin. The mixture was allowed to proceed for 15 minutes at room temperature and was terminated by the addition of 0.1 N HC1. The total intracellular cAMP concentration was quantitated using the Amersham cAMP SPA kit. UTILITIES AND COMBINATIONS
Utilities The compounds of the present invention are cannabinoid receptor modulators, and include compounds which are, for example, selective agonists, partial agonists, inverse agonists, antagonists or partial antagonists of the cannabinoid receptor. Accordingly, the compounds of the present invention may be useful for the treatment or prevention of diseases and disorders associated with G-protein coupled cannabinoid receptor activity. Preferably, compounds of the present invention possess activity as antagonists or inverse agonists of the CB-1 receptor, and may be used in the treatment of diseases or disorders associated with the activity of the CB-1 receptor. Accordingly, the compounds of the present invention can be administered to mammals, preferably humans, for the treatment of a variety of conditions and disorders, including, but not limited to metabolic and eating disorders as well as conditions associated with metabolic disorders, (e.g., obesity, diabetes, arteriosclerosis, hypertension, polycystic ovary disease, cardiovascular disease, osteoarthritis, dermatological disorders, hypertension, insulin resistance, hypercholesterolemia, hypertriglyceridemia, cholelithiasis and sleep disorders, hyperlipidemic conditions, bulimia nervosa and compulsive eating disorders) or psychiatric disorders, such as substance abuse, depression, anxiety, mania and schizophrenia. These compounds could also be used for the improvement of cognitive function (e.g., the treatment of dementia, including Alzheimer's disease, short term memory loss and attention deficit disorders); neurodegenerative disorders (e.g.,
Parkinson's Disease, cerebral apoplexy and craniocerebral trauma) and hypotension (e.g., hemorrhagic and endotoxin-inducd hypotension). These compounds could also be used for treatment of catabolism in connection with pulmonary dysfunction and ventilator dependency; treatment of cardiac dysfunction (e.g., associated with valvular disease, myocardial infarction, cardiac hypertrophy or congestive heart failure); and improvement of the overall pulmonary function; transplant rejection; rheumatoid arthritis; multiple sclerosis; inflammatory bowel disease; lupus; graft vs. host disease; T-cell mediated hypersensitivity disease; psoriasis; asthma; Hashimoto's thyroiditis; Guillain-Barre syndrome; cancer; contact dermatitis; allergic rhinitis; and ischemic or reperfusion injury. Compounds useful in the treatment of appetitive or motivational disorders regulate desires to consume sugars, carbohydrates, alcohol or drugs and more generally to regulate the consumption of ingredients with hedonic value. In the present description and in the claims, appetitive disorders are understood as meaning: disorders associated with a substance and especially abuse of a substance and/or dependency on a substance, disorders of eating behaviors, especially those liable to cause excess weight, irrespective of its origin, for example: bulimia nervosa, craving for sugars. The present invention therefore further relates to the use of a CB-1 receptor antagonist or inverse agonist for the treatment of bulimia and obesity, including obesity associated with type II diabetes (non-insulin-dependent diabetes), or more generally any disease resulting in the patient becoming overweight. Obesity, as described herein, is defined by a body mass index (kg/m ) of at least 26. It may be due to any cause, whether genetic or environmental, including overeating and bulemia, polycycstic ovary disease, craniopharyngeoma, Prader-Willi Syndrome, Frohlich's Syndrome, Type II diabetes, growth hormone deficiency, Turner's Syndrome and other pathological states characterized by reduced metabolic activity or reduced energy expenditure. As used with reference to the utilities described herein, the term "treating" or "treatment" encompasses prevention, partial alleviation, or cure of the disease or disorder. Further, treatment of obesity is expected to prevent progression of medical co variants of obesity, such as arteriosclerosis, Type II diabetes, polycystic ovary disease, cardiovascular disease, osteoarthritis, dermatological disorders, hypertension, insulin resistance, hypercholesterolemia, hypertriglyceridemia, cholelithiasis and sleep disorders. Compounds in the present invention may also be useful in treating substance abuse disorders, including substance dependence or abuse without physiological dependence. Substances of abuse include alcohol, amphetamines (or amphetamine- like substances), caffeine, cannabis, cocaine, hallucinogens, inhalents, nicotine, opioids, phencyclidine (or phencyclidine-like compounds), sedative-hypnotics or benzodiazepines, and other (or unknown) substances and combinations of the above. The terms "substance abuse disorders" also includes drug or alcohol withdrawal syndromes and substance-induced anxiety or mood disorder with onset during Λvithdrawal. Compounds in the present invention may be useful in treating memory impainnent and cognitive disorders. The condition of memory impairment is manifested by impaimαent of the ability to learn new information and/or the inability to recall previously learned information. Memory impairment is a primary symptom of dementia and can also be a symptom associated with such diseases as Alzheimer's disease, schizophrenia, Parkinson's disease, Huntington's disease, Pick's disease, Creutzfeld- Jakob disease, HIV, cardiovascular disease, and head trauma as well as age-related cognitive decline. Dementias are diseases that include memory loss and additional intellectual impairment separate from memory. Cannabinoid receptor modulators may also be useful in treating cognitive impairments related to attentional deficits, such as attention deficit disorder. Compounds in the present invention may also be useful in treating diseases associated with dysfunction of brain dopaminergic systems, such as Parkinson's Disease and substance abuse disorders. Parkinsons' s Disease is a neurodenerative movement disorder characterized by bradykinesia and tremor. As modulators of the cannabinoid receptor, the compounds of the present invention are further useful for the treatment and prevention of respiratory diseases and disorders. Respiratory diseases for which cannabinoid receptor modulators are useful include, but are not limited to, chronic pulmonary obstructive disorder, emphysema, asthma, and bronchitis. In addition, cannabinoid receptor modulators block the activation of lung epithelial cells by moeties such as allergic agents, inflammatory cytokines or smoke, thereby limiting release of mucin, cytokines, and chemokines, or selectively inhibiting lung epithelial cell activation. Moreover, the compounds employed in the present invention may stimulate inhibitory pathways in cells, particularly in leukocytes, lung epithelial cells, or both, and are thus useful in treating such diseases. "Leukocyte activation" is defined herein as any or all of cell proliferation, cytokine production, adhesion protein expression, and production of inflammatory mediators. "Epithelial cell activation" is defined herein as the production of any or all of mucins, cytokines, chemokines, and adhesion protein expression. Use of the compounds of the present invention for treating leukocyte activation-associated disorders is exemplified by, but is not limited to, treating a range of disorders such as: transplant (such as organ transplant, acute transplant, xenotransplant or heterograft or homograft (such as is employed in burn treatment)) rejection; protection from ischemic or reperfusion injury such as ischemic or reperfusion injury incurred during organ transplantation, myocardial infarction, stroke or other causes; transplantation tolerance induction; arthritis (such as rheumatoid arthritis, psoriatic arthritis or osteoarthritis); multiple sclerosis; respiratory and pulmonary diseases including but not limited to chronic obstructive pulmonary disease (COPD), emphysema, bronchitis, and acute respiratory distress syndrome (ARDS); inflammatory bowel disease, including ulcerative colitis and Crohn's disease; lupus (systemic lupus erythematosis); graft vs. host disease; T-cell mediated hypersensitivity diseases, including contact hypersensitivity, delayed-type hypersensitivity, and gluten-sensitive enteropathy (Celiac disease); psoriasis; contact dermatitis (including that due to poison ivy); Hashimoto's thyroiditis; Sjogren's syndrome; Autoimmune Hyperthyroidism, such as Graves' Disease; Addison's disease (autoimmune disease of the adrenal glands); Autoimmune polyglandular disease (also known as autoimmune polyglandular syndrome); autoimmune alopecia; pernicious anemia; vitiligo; autoimmune hypopituatarism; Guillain-Barre syndrome; other autoimmune diseases; glomerulonephritis; serum sickness; uticaria; allergic diseases such as respiratory allergies (asthma, hayfever, allergic rhinitis) or skin allergies; scleracierma; mycosis fungoides; acute inflammatory and respiratory responses (such as acute respiratory distress syndrome and ishchemia/reperfusion injury); dermatomyositis; alopecia areata; chronic actinic dermatitis; eczema; Behcet's disease; Pustulosis palmoplanteris; Pyodemia gangrenum; Sezary's syndrome; atopic dermatitis; systemic schlerosis; and morphea. The term "leukocyte activation- associated" or ' "leukocyte-activation mediated" disease as used herein includes each of the above referenced diseases or disorders. In a particular embodiment, the compounds of the present invention are useful for treating the aforementioned exemplary disorders irrespective of their etiology. The combined activity of the present compounds towards monocytes, macrophages, T-cells, etc. may be useful in treating any of the above-mentioned disorders. Cannabinoid receptors are important in the regulation of Fc gamma receptor responses of monocytes and macrophages. Compounds of the present invention inhibit the Fc gamma dependent production of TNF alpha in human monocytes/macrophages. The ability to inhibit Fc gamma receptor dependent monocyte and macrophage responses results in additional anti-inflammatory activity for the present compounds. This activity is especially of value, for example, in treating inflammatory diseases such as arthritis or inflammatory bowel disease. In particular, the present compounds are useful for treating autoimmune glomerulonephritis and other instances of glomerulonephritis induced by deposition of immune complexes in the kidney that trigger Fc gamma receptor responses leading to kidney damage. Cannabinoid receptors are expressed on lung epithelial cells. These cells are responsible for the secretion of mucins and inflammatory cytokines/chemokines in the lung and are thus intricately involved in the generation and progression of respiratory diseases. Cannabinoid receptor modulators regulate both the spontaneous and the stimulated production of both mucins and cytokines. Thus, such compounds are useful in treating respiratory and pulmonary diseases including, COPD, ARDS, and bronchitis. Further, cannabinoid receptors may be expressed on gut epithelial cells and hence regulate cytokine and mucin production and may be of clinical use in treating inflammatory diseases related to the gut. Cannabinoid receptors are also expressed on lymphocytes, a subset of leukocytes. Thus, cannabinoid receptor modulators will inhibit B and T-cell activation, proliferation and differentiation. Thus, such compounds will be useful in treating autoimmune diseases that involve either antibody or cell mediated responses such as multiple sclerosis and lupus. In addition, cannabinoid receptors regulate the Fc epsilon receptor and chemokine induced degranulation of mast cells and basophils. These play important roles in asthma, allergic rhinitis, and other allergic disease. Fc epsilon receptors are stimulated by IgE-antigen complexes. Compounds of the present invention inhibit the Fc epsilon induced degranulation responses, including the basophil cell line, RBL. The ability to inhibit Fc epsilon receptor dependent mast cell and basophil responses results in additional anti-inflammatory and anti-allergic activity for the present compounds. In particular, the present compounds are useful for treating asthma, allergic rhinitis, and other instances of allergic disease.
Combinations The present invention includes within its scope pharmaceutical compositions comprising, as an active ingredient, a therapeutically effective amount of at least one of the compounds of formula I, alone or in combination with a pharmaceutical earner or diluent. Optionally, compounds of the present invention can be used alone, in combination with other suitable therapeutic agents useful in the treatment of the aforementioned disorders including: anti-obesity agents; anti-diabetic agents, appetite suppressants; cholesterol/lipid-lowering agents, HDL-raising agents, cognition enhancing agents, agents used to treat neurodegeneration, agents used to treat respiratory conditions, agents used to treat bowel disorders, anti-inflammatory agents; anti-anxiety agents; anti-depressants; anti-hypertensive agents; cardiac glycosides; and anti-tumor agents. Such other therapeutic agent(s) may be administered prior to, simultaneously with, or following the administration of the cannabinoid receptor modulators in accordance with the invention. Examples of suitable anti-obesity agents for use in combination with the compounds of the present invention include melanocortin receptor (MC4R) agonists, melanin-concentrating hormone receptor (MCHR) antagonists, growth hormone secretagogue receptor (GHSR) antagonists, galanin receptor modulators, orexin antagonists, CCK agonists, GLP-1 agonists, and other Pre-proglucagon-derived peptides; NPY1 or NPY5 antagonsist, NPY2 and NPY4 modulators, corticotropin releasing factor agonists, histamine receptor-3 (H3) modulators, aP2 inhibitors, PPAR gamma modulators, PPAR delta modulators, acetyl-CoA carboxylase (ACC) inihibitors, 11-β-HSD-l inhibitors, adinopectin receptor modulators; beta 3 adrenergic agonists, such as AJ9677 (Takeda/Dainippon), L750355 (Merck), or CP331648 (Pfizer) or other known beta 3 agonists as disclosed in U.S. Patent Nos. 5,541,204, 5,770,615, 5,491,134, 5,776,983 and 5,488,064, a thyroid receptor beta modulator, such as a thyroid receptor ligand as disclosed in WO 97/21993 (U. Cal SF), WO 99/00353 (KaroBio) and GB98/284425 (KaroBio), a lipase inhibitor, such as orlistat or ATL-962 (Alizyme), serotonin receptor agonists, (e.g., BVT-933 (Biovitrum)), monoamine reuptake inhibitors or releasing agents, such as fenfluramine, dexfenfluramine, fluvoxamine, fluoxetine, paroxetine, sertraline, chlorphentermine, cloforex, clortermine, picilorex, sibutramine, dexamphetamine, phentermine, phenylpropanolamine or mazindol, anorectic agents such as topiramate (Johnson & Johnson), CNTF (ciliary neurotrophic factor) /Axokme® (Regeneron), BDNF (brain- derived neurotrophic factor), leptin and leptin receptor modulators, or cannabinoid- 1 receptor antagonists, such as SR-141716 (Sanofi) or SLV-319 (Solvay). Examples of suitable anti-diabetic agents for use in combination with the compounds of the present invention include: insulin secretagogues or insulin sensitizers, which may include biguanides, sulfonyl ureas, glucosidase inhibitors, aldose reductase inhibitors, PPAR γ agonists such as thiazolidinediones, PPAR α agonists (such as fibric acid derivatives), PPAR δ antagonists or agonists, PPAR α/γ dual agonists, 11-β-HSD-l inhibitors, dipeptidyl peptidase IV (DP4) inhibitors, SGLT2 inhibitors, glycogen phosphorylase inhibitors, and/or meglitinides, as well as insulin, and/or glucagon-like peptide- 1 (GLP-1), GLP-1 agonist, and/or a PTP- IB inhibitor (protein tyrosine phosphatase-lB inliibitor). The antidiabetic agent may be an oral antihyperglycemic agent preferably a biguanide such as metformin or phenformin or salts thereof, preferably metformin HC1. Wl ere the antidiabetic agent is a biguanide, the compounds of the present invention will be employed in a weight ratio to biguanide within the range from about 0.001:1 to about 10:1, preferably from about 0.01:1 to about 5:1. The antidiabetic agent may also preferably be a sulfonyl urea such as glyburide
(also known as glibenclamide), glimepiride (disclosed in U.S. Patent No. 4,379,785), glipizide, gliclazide or chlorpropamide, other known sulfonylureas or other antihyperglycemic agents which act on the ATP-dependent channel of the beta-cells, with glyburide and glipizide being preferred, which may be administered in the same or in separate oral dosage forms. The oral antidiabetic agent may also be a glucosidase inhibitor such as acarbose (disclosed in U.S. Patent No. 4,904,769) or miglitol (disclosed in U.S. Patent No. 4,639,436), which may be administered in the same or in a separate oral dosage forms. The compounds of the present invention may be employed in combination with a PPAR γ agonist such as a thiazolidinedione oral anti-diabetic agent or other insulin sensitizers (which has an insulin sensitivity effect inNIDDM patients) such as rosiglitazone (SKB), pioglitazone (Takeda), Mitsubishi's MCC-555 (disclosed in U.S. Patent No. 5,594,016), Glaxo-Welcome's GL-262570, englitazone (CP-68722, Pfizer) or darglitazone (CP-86325, Pfizer, isaglitazone (MIT/J&J), JTT-501 (JPNT/P&U), L- 895645 (Merck), R-l 19702 (Sankyo/WL), NN-2344 (Dr. Reddy/NN), or YM-440 (Yamanouchi), preferably rosiglitazone and pioglitazone. The compounds of the present invention may be employed with a PPARα/γ dual agonist such as MK-767/KRP-297 (Merck/Kyorin; as described in , K. Yajima, et. al, Am. J. Physiol. Endocrinol. Metab., 284: E966-E971 (2003)), AZ-242 (tesaglitazar; Astra-Zeneca; as described in B. Ljung, et. al., J Lipid Res., 43, 1855- 1863 (2002)); muraglitazar; or the compounds described in US patent 6,414,002. The compounds of the present invention may be employed in combination with anti-Jiyperlipidemia agents, or agents used to treat arteriosclerosis. An example of an hypolipidemic agent would be an HMG CoA reductase inhibitor which includes, but is not limited to, mevastatin and related compounds as disclosed in U.S. Patent No. 3,983,140, lovastatin (mevinolin) and related compounds as disclosed in U.S. Patent No. 4,231,938, pravastatin and related compounds such as disclosed in U.S. Patent No. 4,346,227, simvastatin and related compounds as disclosed in U.S. Patent Nos. 4,448,784 and 4,450,171. Other HMG CoA reductase inhibitors which may be employed herein include, but are not limited to, fluvastatin, disclosed in U.S. Patent No. 5,354,772, cerivastatin disclosed in U.S. Patent Nos. 5,006,530 and 5,177,080, atorvastatin disclosed in U.S. Patent Nos. 4,681,893, 5,273,995, 5,385,929 and 5,686,104, pitavastatin (Nissan/Sankyo's nisvastatin (NK-104) or itavastatin), disclosed in U.S. Patent No. 5,011,930, Shionogi-Astra/Zeneca rosuvastatin (visastatin (ZD-4522)) disclosed in U.S. Patent No. 5,260,440, and related statin compounds disclosed in U.S. Patent No. 5,753,675, pyrazole analogs of mevalonolactone derivatives as disclosed in U.S. Patent No. 4,613,610, indene analogs of mevalonolactone derivatives as disclosed in PCT application WO 86/03488, 6-[2-(substituted-pyrcol-l-yl)-alkyl)pyran-2-ones and derivatives thereof as disclosed in U.S. Patent No. 4,647,576, Searle's SC-45355 (a 3 -substituted pentanedioic acid derivative) dichloroacetate, imidazole analogs of mevalonolactone as disclosed in PCT application WO 86/07054, 3-carboxy-2-hydroxy-propane- phosphonic acid derivatives as disclosed in French Patent No. 2,596,393, 2,3- disubstituted pyrrole, furan and thiophene derivatives as disclosed in European Patent Application No. 0221025, naphthyl analogs of mevalonolactone as disclosed in U.S. Patent No. 4,686,237, octahydronaphthalenes such as disclosed in U.S. Patent No. 4,499,289, keto analogs of mevinolin (lovastatin) as disclosed in European Patent Application No.0,142,146 A2, and quinoline and pyridine derivatives disclosed in U.S. Patent Nos. 5,506,219 and 5,691,322. In addition, phosphinic acid compounds useful in inhibiting HMG CoA reductase suitable for use herein are disclosed in GB 2205837. The squalene synthetase inhibitors suitable for use herein include, but are not limited to, α-phosphono-sulfonates disclosed in U.S. Patent No. 5,712,396, those disclosed by Biller, et al, J Med. Chem., 31, 1869-1871 (1998) including isoprenoid (phosphinyl-methyl)phosphonates as well as other known squalene synthetase inhibitors, for example, as disclosed in U.S. Patent No. 4,871,721 and 4,924,024 and in Biller, S.A., Neuenschwander, K., Ponpipom, M.M., and Poulter, CD., Current Pharmaceutical Design, 2, 1-40 (1996). In addition, other squalene synthetase inhibitors suitable for use herein include the terpenoid pyrophosphates disclosed by P. Ortiz de Montellano, et al., J Med. Chem., 20, 243-249 (1977), the fa nesyl diphosphate analog A and presqualene pyrophosphate (PSQ-PP) analogs as disclosed by Corey and Volante, J Am. Chem. Soc. , 98, 1291 - 1293 (1976), phosphinylphosphonates reported by McClard, R. W. et al., J Am. Chem. Soc, 109, 5544 (1987) and cyclopropanes reported by Capson, T.L., PhD dissertation, June, 1987, Dept. Med. Chem. U of Utah, Abstract, Table of Contents, pp 16, 17, 40-43, 48-51, Summary. Other hypolipidemic agents suitable for use herein include, but are not limited to, fibric acid derivatives, such as fenofibrate, gemfibrozil, clofibrate, bezafibrate, ciprofibrate, clinofibrate and the like, probucol, and related compounds as disclosed in U.S. Patent No. 3,674,836, probucol and gemfibrozil being prefened, bile acid sequestrants such as cholestyramine5 colestipol and DEAE-Sephadex (SECHOLEX, POLICEXLDE) and cholestagel (Sankyo/Geltex), as well as lipostabil (Rhone- Poulenc), Eisai E-5050 (an N-substituted ethanolamine derivative), imanixil (HOE- 402), tetrahydrolipstatin (THL), istigmastanylphos-phorylcholine (SPC, Roche), aminocyclodextrin (Tanabe Seiyoku), Ajinomoto AJ-814 (azulene derivative), melinamide (Sumitomo), Sandoz 58-035, American Cyanamid CL-277,082 and CL- 283,546 (disubstituted urea derivatives), nicotinic acid (niacin), acipimox, acifran, neomycin, p-aminosalicylic acid, aspirin, poly(diallylmethylamine) derivatives such as disclosed in U.S. Patent No. 4,759,923, quaternary amine poly(diallyldimethylammonium chloride) and ionenes such as disclosed in U.S. Patent No. 4,027,009, and other known serum cholesterol lowering agents. The other hypolipidemic agent may be an ACAT inhibitor (which also has anti-atherosclerosis activity) such as disclosed in, Drugs of the Future, 24, 9-15
(1999), (Avasimibe); "The ACAT inhibitor, Cl-1011 is effective in the prevention and regression of aortic fatty streak area in hamsters", Nicolosi et al., Atherosclerosis (Shannon, Irel), 137 (1), 77-85 (1998); "The pharmacological profile of FCE 27677: a novel ACAT inhibitor with potent hypolipidemic activity mediated by selective suppression of the hepatic secretion of ApoBlOO-containing lipoprotein", Ghiselli, Giancarlo, Cardiovasc. Drug Rev., 16 (1), 16-30 (1998); "RP 73163: a bioavailable alkylsulfinyl-diphenylimidazole ACAT inhibitor", Smith, C, et al, Bioorg. Med. Chem. Lett, 6 (1), 47-50 (1996); "ACAT inhibitors: physiologic mechanisms for hypolipidemic and anti-atherosclerotic activities in experimental animals", Krause et al., Editor(s): Ruffolo, Robert R., Jr.; Hollinger, Marmfred A., Inflammation: Mediators Pathways, 173-98 (1995), Publisher: CRC, Boca Raton, Fla.; "ACAT inhibitors: potential anti-atherosclerotic agents", Sliskovic et al., Curr. Med. Chem., 1 (3), 204-25 (1994); "Inhibitors of acyl-CoA:cholesterol O-acyl transferase (ACAT) as hypocholesterolemic agents. 6. The first water-soluble ACAT inhibitor with lipid- regulating activity. Inhibitors of acyl-CoA:cholesterol acyltransferase (ACAT). 7. Development of a series of substituted N-phenyl-N'-[(l-phenylcyclopentyl)- methyljureas with enhanced hypocholesterolemic activity", Stout et al., Chemt7"acts: Org. Chem., 8 (6), 359-62 (1995), or TS-962 (Taisho Pharmaceutical Co. Ltd), as well as F-1394, CS-505, F-12511, HL-004, K-10085 and YIC-C8-434. The hypolipidemic agent may be an upregulator of LDL receptor activity such as MD-700 (Taisho Phannaceutical Co. Ltd) and LY295427 (Eli Lilly). The hypolipidemic agent may be a cholesterol absorption inhibitor preferably Schering- Plough' s SCH48461 (ezetimibe) as well as those disclosed in Atherosclerosis 115, 45-63 (1995) and J. Med. Chem. 41, 973 (1998). The other lipid agent or lipid-modulating agent may be a cholesteryl transfer protein inhibitor (CETP) such as Pfizer's CP-529,414 as well as those disclosed in WO/0038722 and in EP 818448 (Bayer) and EP 992496, and Pharmacia's SC-744 and SC-795, as well as CETi-1 and JTT-705. The hypolipidemic agent may be an ileal Na /bile acid cotransporter inhibitor such as disclosed in Drugs of the Future, 24, 425-430 (1999). The ATP citrate lyase inhibitor which may be employed in the combination of the invention may include, for example, those disclosed in U.S. Patent No. 5,447,954. The other lipid agent also includes a phytoestrogen compound such as disclosed in WO 00/30665 including isolated soy bean protein, soy protein concentrate or soy flour as well as an isoflavone such as genistein, daidzein, glycitein or equol, or phytosterols, phytostanol or tocotrienol as disclosed in WO 2000/015201; a beta-lactam cholesterol absorption inhibitor such as disclosed in EP 675714; an HDL upregulator such as an LXR agonist, a PPAR α-agonist and/or an FXR agonist; an LDL catabolism promoter such as disclosed in EP 1022272; a sodium-proton exchange inhibitor such as disclosed in DE 19622222; an LDL-receptor inducer or a steroidal glycoside such as disclosed in U.S. Patent No. 5,698,527 and GB 2304106; an anti-oxidant such as beta-carotene, ascorbic acid, α-tocopherol or retinol as disclosed in WO 94/15592 as well as Vitamin C and an antihomocysteine agent such as folic acid, a folate, Vitamin B6, Vitamin B12 and Vitamin E; isoniazid as disclosed in WO 97/35576; a cholesterol absorption inhibitor, an HMG-CoA synthase inhibitor, or a lanosterol demethylase inhibitor as disclosed in WO 97/48701; a PPAR δ agonist for treating dyslipidemia; or a sterol regulating element binding protein-I (SREBP-1) as disclosed in WO 2000/050574, for example, a sphingolipid, such as ceramide, or neutral sphingomyelenase (N-SMase) or fragment thereof. Preferred hypolipidemic agents are pravastatin, lovastatin, simvastatin, atorvastatin, fluvastatin, pitavastatin and rosuvastatin, as well as niacin and/or cholestagel. The compounds of the present invention may be employed in combination with anti-hypertensive agents. Examples of suitable anti-hypertensive agents for use in combination with the compounds of the present invention include beta adrenergic blockers, calcium channel blockers (L-type and/or T-type; e.g. diltiazem, verapamil, nifedipine, amlodipine and mybefradil), diuretics (e.g., chlorothiazide, hydrochlorόthiazide, flumethiazide, hydroflumethiazide, bendroflumethiazide, methylchlorothiazide, trichloromethiazide, polythiazide, benzthiazide, ethacrynic acid tricrynafen, chlorthalidone, furosemide, musolimine, bumetanide, triamtrenene, amiloride, spironolactone), renin inhibitors, ACE inhibitors (e.g., captopril, zofenop il, fosinopril, enalapril, ceranopril, cilazopril, delapril, pentopril, quinapril, ramipril, lisinopril), AT-1 receptor antagonists (e.g., losartan, irbesartan, valsartan), ET receptor antagonists (e.g., sitaxsentan, atrsentan and compounds disclosed in U.S. Patent Nos. 5,612,359 and 6,043,265), Dual ET/AII antagonist (e.g., compounds disclosed in WO 00/01389), neutral endopeptidase (NEP) inhibitors, vasopepsidase inhibitors (dual NEP-ACE inhibitors) (e.g., omapatrilat and gemopatrilat), and nitrates. Cannbinoid receptor modulators could be useful in treating other diseases associated with obesity, including sleep disorders. Therefore, the compounds described in the present invention could be used in combination with therapeutics for treating sleep disorders. Examples of suitable therapies for treatment of sleeping disorders for use in combination with the compounds of the present invention include melatonin analogs, melatonin receptor antagonists, ML 1 B agonists, GAB A receptor modulators; NMD A receptor modulators, histamine-3 (H3) receptor modulators, dopamine agonists and orexin receptor modulators. Cannabinoid receptor modulators may reduce or ameliorate substance abuse or addictive disorders. Therefore, combination of cannabinoid receptor modulators with agents used to treat addictive disorders may reduce the dose requirement or improve the efficacy of current addictive disorder therapeutics. Examples of agents used to treat substance abuse or addictive disorders are: selective serotonin reuptake inhibitors (SSRI), methadone, buprenorphine, nicotine and bupropion. Cannabinoid receptor modulators may reduce anxiety or depression; therefore, the compounds described in this application may be used in combination with anti- anxiety agents or antidepressants. Examples of suitable anti-anxiety agents for use in combination with the compounds of the present invention include benzodiazepines (e.g., diazepam, lorazepam, oxazepam, alprazolam, chlordiazepoxide, clonazepam, chlorazepate, halazepam and prazepam), 5HT1 A receptor agonists (e.g., buspirone, flesinoxan, gepirone and ipsapirone), and corticotropin releasing factor (CRF) antagonists. Examples of suitable classes of anti-depressants for use in combination with the compounds of the present invention include norepinephrine reuptake inhibitors (tertiary and secondary amine tricyclics), selective serotonin reuptake inhibitors (SSRIs) (fluoxetine, fluvoxamine, paroxetine and sertraline), monoamine oxidase inhibitors (MAOIs) (isocarboxazid, phenelzine, tranylcypromine, selegiline), reversible inhibitors of monoamine oxidase (RIMAs) (moclobemide), serotonin and norepinephrine reuptake inhibitors (SNRIs) (venlafaxine), corticotropin releasing factor (CRF) receptor antagonists, alpah-adrenoreceptor antagonists, and atypical antidepressants (bupropion, lithium, nefazodone, trazodone and viloxazine). The combination of a conventional antipsychotic drug with a CB-1 receptor antagonist could also enhance symptom reduction in the treatment of psychosis or mania. Further, such a combination could enable rapid symptom reduction, reducing the need for chronic treatment with antipsychotic agents. Such a combination could also reduce the effective antipsychotic dose requirement, resulting in reduced probability of developing the motor dysfunction typical of chronic antipsychotic treatment. Examples of suitable antipsychotic agents for use in combination with the compounds of the present invention include the phenothiazine (chlorpromazine, mesoridazine, thioridazine, acetophenazine, fluphenazine, perphenazine and trifluoperazine), thioxanthine (chlorprothixene, thiothixene), heterocyclic dibenzazepine (clozapine, olanzepine and aripiprazole), butyrophenone (haloperidol), dipheyylbutylpiperidine (pimozide) and indolone (molindolone) classes of antipsychotic agents. Other antipsychotic agents with potential therapeutic value in combination with the compounds in the present invention include loxapine, sulpiride and risperidone. Combination of the compounds in the present invention with conventional antipsychotic drugs could also provide an enhanced therapeutic effect for the treatment of schizophrenic disorders, as described above for manic disorders. As used here, schizophrenic disorders include paranoid, disorganized, catatonic, undifferentiated and residual schizophrenia, schizophreniform disorder, shcizoaffective disorder, delusional disorder, brief psychotic disorder and psychotic disorder not specified. Examples of suitable antipsychotic drugs for combination with the compounds in the present invention include the antipsychotics mentioned above, as well as dopamine receptor antagonists, muscarinic receptor agonists, 5HT2A receptor antagonists and 5HT2A/dopamine receptor antagonists or partial agonists (e.g., olanzepine, aripiprazole, risperidone, ziprasidone). The compounds described in the present invention could be used to enhance the effects of cognition-enhancing agents, such as acetylcholinesterase inhibitors (e.g., tacrine), muscarinic receptor- 1 agonists (e.g., milameline), nicotinic agonists, glutamic acid receptor (AMPA and NMD A) modulators, and nootropic agents (e.g., piracetam, levetiracetam). Examples of suitable tlierapies for treatment of Alzheimer's disease and cognitive disorders for use in combination with the compounds of the present invention include donepezil, tacrine, revastigraine, 5HT6, gamma secretase inhibitors, beta secretase inhibitors, SK channel blockers, Maxi-K blockers, and KCNQs blockers. The compounds described in the present invention could be used to enhance the effects of agents used in the treatment of Parkinson's Disease. Examples of agents used to treat Parkinson's Disease include: levadopa with or without a COMT inhibitor, antiglutamatergic drugs (amantadine, riluzole), alpha-2 adrenergic antagonists such as idazoxan, opiate antagonists, such as naltrexone, other dopamine agonists or transportor modulators, such as ropinirole, or pramipexole or neurotrophic factors such as glial derived neurotrophic factor (GDNF). The compounds described in the present invention could be used in combination with suitable anti-inflammatory agents. Examples of suitable anti- inflammatory agents for use in combination with the compounds of the present invention include prednisone, dexamethasone, cyclooxygenase inhibitors (i.e., COX-1 and/or COX-2 inhibitors such as NSAIDs, aspirin, indomethacin, ibuprofen, piroxicam, Naproxen®, Celebrex®, Vioxx®), CTLA4-Ig agonists/antagonists, CD40 ligand antagonists, IMPDH inhibitors, such as mycophenolate (CellCept®), integrin antagonists, alpha-4 beta-7 integrin antagonists, cell adhesion inhibitors, interferon gamma antagonists, ICAM-1, tumor necrosis factor (TNF) antagonists (e.g., infliximab, OR1384, including TNF-alpha inhibitors, such as tenidap, anti-TNF antibodies or soluble TNF receptor such as etanercept (Enbrel®), rapamycin (sirolimus or Rapamune) and leflunomide (Arava)), prostaglandin synthesis inhibitors, budesonide, clofazimine, CNI-1493, CD4 antagonists (e.g., priliximab), p38 mitogen-activated protein kinase inhibitors, protein tyrosine kinase (PTK) inhibitors, IKK inhibitors, and therapies for the treatment of irritable bowel syndrome (e.g., Zelnorm® and Maxi-K® openers such as those disclosed in U.S. Patent No. 6,184,231 Bl). Exemplary of such other therapeutic agents which may be used in combination with cannabinoid receptor modulators include the following: cyclosporins (e.g., cyclosporin A), anti-IL-2 receptor (Anti-Tac), anti-CD45RB, anti-CD2, anti-CD3 (OKT-3), anti-CD4, anti-CD80, anti-CD86, monoclonal antibody OKT3, agents blocking the interaction between CD40 and gp39, such as antibodies specific for CD40 and/or gp39 (i.e., CD154), fusion proteins constructed from CD40 and gp39 (CD40Ig and CD8gp39), inhibitors, such as nuclear translocation inhibitors, of NF- kappa B function, such as deoxyspergualin (DSG), gold compounds, antiproliferative agents such as methotrexate, FK506 (tacrolimus, Prograf), mycophenolate mofetil, cytotoxic drugs such as azathiprine and cyclophosphamide, anticytokines such as antiIL-4 or IL-4 receptor fusion proteins and PDE 4 inhibitors such as Ariflo, and the PTK inhibitors disclosed in the following U.S. patent applications, incorporated herein by reference in their entirety: Ser. No. 09/097,338, filed Jun. 15, 1998; Ser. No. 09/094,797, filed Jun. 15, 1998; Ser. No. 09/173,413, filed Oct. 15, 1998; and Ser. No. 09/262,525, filed Mar. 4, 1999. See also the following documents and references cited therein and incorporated herein by reference: Hollenbaugh, D., et al., "Cleavable CD40Ig Fusion Proteins and the Binding to Sgp39", J. Immunol. Methods (Netherlands), 188 (1), pp. 1-7 (Dec. 15, 1995); Hollenbaugh, D., et al., "The Human T Cell Antigen Gp39, A Member of the TNF Gene Family, Is a Ligand for the CD40 Receptor: Expression of a Soluble Form of Gp39 with B Cell Co-Stimulatory Activity", EMBO J (England), 11 (12), pp. 4313-4321 (December 1992); and Moreland, L. W. et al., "Treatment of Rheumatoid Arthritis with a Recombinant Human Tumor Necrosis Factor Receptor (P75)-Fc Fusion Protein, " New England J. of Medicine, 337 (3), pp. 141-147 (1997). ' The above other therapeutic agents, when employed in combination with the compounds of the present invention, may be used, for example, in those amounts indicated in the Physicians' Desk Reference (PDR) or as otherwise determined by one of ordinary skill in the art. The compounds of formula (I) of the invention can be administered orally or parenterally, such as subcutaneously or intravenously, as well as by nasal application, rectally or sublingually to various mammalian species known to be subject to such maladies, e.g., humans, in an effective amount up to 1 gram, preferably up to 200 mg, more preferably up to 100 mg in a regimen of single, two or four divided daily doses. The compounds of the formula (I) can be administered for any of the uses described herein by any suitable means, for example, orally, such as in the form of tablets, capsules, granules or powders; sublingually; bucally; parenterally, such as by subcutaneous, intravenous, intramuscular, or intrasternal injection or infusion techniques (e.g., as sterile injectable aqueous or non-aqueous solutions or suspensions); nasally, including administration to the nasal membranes, such as by inhalation spray; topically, such as in the form of a cream or ointment; or rectally such as in the form of suppositories; in dosage unit formulations containing non-toxic, pharmaceutically acceptable vehicles or diluents. The present compounds can, for example, be administered in a form suitable for immediate release or extended release. Immediate release or extended release can be achieved by the use of suitable pharmaceutical compositions comprising the present compounds, or, particularly in the case of extended release, by the use of devices such as subcutaneous implants or osmotic pumps. The present compounds can also be administered liposomally. Exemplary compositions for oral administration include suspensions which can contain, for example, microcrystalline cellulose for imparting bulk, alginic acid or sodium alginate as a suspending agent, methylcellulose as a viscosity enhancer, and sweeteners or flavoring agents such as those known in the art; and immediate release tablets which can contain, for example, microcrystalline cellulose, dicalcium phosphate, starch, magnesium stearate and/or lactose and/or other excipients, binders, extenders, disintegrants, diluents and lubricants such as those known in the art. The compounds of formula I can also be delivered through the oral cavity by sublingual and/or buccal administration. Molded tablets, compressed tablets or freeze-dried tablets are exemplary forms which may be used. Exemplary compositions include those formulating the present compound(s) with fast dissolving diluents such as mannitol, lactose, sucrose and/or cyclodextrins. Also included in such formulations may be high molecular weight excipients such as celluloses (avicel) or polyethylene glycols (PEG). Such formulations can also include an excipient to aid mucosal adhesion such as hydroxy propyl cellulose (HPC), hydroxy propyl methyl cellulose (HPMC), sodium carboxy methyl cellulose (SCMC), maleic anhydride copolymer (e.g., Gantrez), and agents to control release such as polyacrylic copolymer (e.g. Carbopol 934). Lubricants, glidants, flavors, coloring agents and stabilizers may also be added for ease of fabrication and use. Exemplary compositions for nasal aerosol or inhalation administration include solutions in saline which can contain, for example, benzyl alcohol or other suitable preservatives, absorption promoters to enhance bioavailabihty, and/or other solubilizing or dispersing agents such as those known in the art. Exemplary compositions for parenteral administration include injectable solutions or suspensions which can contain, for example, suitable non-toxic, parenterally acceptable diluents or solvents, such as mannitol, 1,3-butanediol, water, Ringer's solution, an isotonic sodium chloride solution, or other suitable dispersing or wetting and suspending agents, including synthetic mono- or diglycerides, and fatty acids, including oleic acid, or Cremaphor. Exemplary compositions for rectal administration include suppositories which can contain, for example, a suitable non-irritating excipient, such as cocoa butter, synthetic glyceride esters or polyethylene glycols, which are solid at ordinary temperatures, but liquify and/or dissolve in the rectal cavity to release the drug. Exemplary compositions for topical administration include a topical carrier such as Plastibase (mineral oil gelled with polyethylene). It will be understood that the specific dose level and frequency of dosage for any particular subject can be varied and will depend upon a variety of factors including the activity of the specific compound employed, the metabolic stability and length of action of that compound, the species, age, body weight, general health, sex and diet of the subject, the mode and time of administration, rate of excretion, drug combination, and severity of the particular condition. It should be understood that while this invention has been described herein in terms of specific embodiments set forth in detail, such embodiments are presented by way of illustration of the general principles of the invention, and the invention is not necessarily limited thereto. Certain modifications and variations in any given material, process step or chemical formula will be readily apparent to those skilled in the art without departing from the true spirit and scope of the present invention, and all such modifications and variations should be considered within the scope of the claims that follow.

Claims

WHAT IS CLAIMED IS:
1. A compound of formula I
Figure imgf000055_0001
I including all prodrugs, pharmaceutically acceptable salts and stereoisomers, wherein: 1 'j R and R are independently selected from the group consisting of aryl and heteroaryl; R is selected from the group consisting of alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkylalkyl, heterocyclylalkyl, aryl, heteroaryl, arylalkyl and heteroarylalkyl; and R4 is selected from the group consisting of hydrogen, alkyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, arylalkyl and heteroarylalkyl.
' 2. The compound according to Claim 1, wherein: R is selected from the group consisting of hydrogen, alkyl, aryl and heteroaryl.
The compound according to Claim 2, wherein R is se ϊlleecctteedd frfroomm ttbhe group consisting of alkyl, cycloalkyl, aryl, heteroaryl, arylalkyl and heteroarylalkyl
4. The compound according to Claim 3, wherein: R4 is selected from the group consisting of hydrogen and alkyl. 5. The compound according to Claim 1, wherein: RR iiss sseelleecctteedd frfroomm tthhee ggrrcoup consisting of alkyl, cycloalkyl, -aryl, heteroaryl, arylalkyl and heteroarylalkyl; and R4 is selected from the group consisting of hydrogen.
6. The compound according to Claim 4, wherein: , R1 is an aryl group that is optionally substituted with one or more alkyl, fluoro, chloro, trifluoromethyl, cyano, methoxy, trifluoromethoxy, aryl, heterocyclyl and heteroaryl.
7. The compound according to Claim 4, wherein: R1 is a heteroaryl group that is optionally substituted with one or more fluoro, chloro, trifluoromethyl, cyano, methoxy, trifluoromethoxy, aryl, heterocyclyl and heteroaryl.
8. The compound according to Claim 4, wherein: R is an aryl group that is optionally substituted with one or more alkyl, fluoro, chloro, trifluoromethyl, cyano, methoxy, trifluoromethoxy, aryl, heterocyclyl and heteroaryl.
9. The compound according to Claim 4, wherein: R1 is selected from the group consisting of pyridine, pyridazine, pyrimidine, pyrzaine, imidazole, isoxazole and oxazole, wherein the group is optionally substituted with one or more fluoro, chloro, trifluoromethyl, cyano, methoxy, trifluoromethoxy, aryl, heterocyclyl and heteroaryl.
10. The compound according to Claim 5, wherein: R3 is selected from the group consisting of aryl, heteroaryl, arylalkyl and heteroarylalkyl that is optionally substituted with one or more fluoro, chloro, bromo, cyano, aryl, heterocyclyl and heteroaryl.
11. The compound of Claim 1 selected from:
Figure imgf000057_0001
Figure imgf000058_0001
Figure imgf000059_0001
2. The compound of Claim 1 selected from:
Figure imgf000059_0002
Figure imgf000060_0001
Figure imgf000060_0002
Figure imgf000060_0003
13. A pharmaceutical composition, comprising: at least one compound according to Claim 1 ; and at least one pharmaceutically acceptable diluent or carrier.
14. The pharmaceutical composition according to Claim 13, further comprising: at least one other therapeutic agent.
15. A pharmaceutical combination comprising a pharmaceutical composition of Claim 13 and a therapeutic agent selected from anti-obesity agents; appetite suppressants; anti-diabetic agents; anti-hyperlipidemia agents; hypolipidemic agents; hypocholesterolemic agents; lipid-modulating agents; cholesterol-lowering agents; lipid-lowering agents; HDL-raising agent, anti-hypertensive agents; agents used to treat sleep disorders; agents used to treat substance abuse and addictive disorders; anti-anxiety agents; anti-depressants; anti-psychotic agents; cognition enhancing agents; agents used to treat cognitive disorders; agents used to treat Alzheimer's disease; agents used to treat Parkinson's disease; anti-inflammatory agents; agents used to treat neurodegeneration; agents used to treat arteriosclerosis; agents used to treat respiratory conditions; agents used to treat bowel disorders; cardiac glycosides; and anti-tumor agents.
16. The pharmaceutical combination according to Claim 15 wherein the other therapeutic agent may be administered prior to, simultaneously with, or following the administration of the pharmaceutical composition of Claim 13.
17. A method for treating a cannabinoid receptor mediated disease or disorder, comprising: administering to a patient in need of treatment a therapeutically effective amount of a compound according to Claim 1. 18. The method according to Claim 17 wherein the diseases or disorders are associated with the activity of the CB-1 receptor.
19. The method according to Claim 18, wherein the diseases or disorders are bulimia, obesity or any disease resulting in the patient becoming overweight.
20. The method according to Claim 18, wherein the diseases or disorders are metabolic disorders, eating disorders and appetitive disorders, including treatment of the conditions associated with those disorders, such as obesity, diabetes, arteriosclerosis, hypertension, polycystic ovary disease, cardiovascular disease, osteoarthritis, dermatological disorders, hypertension, insulin resistance, hypercholesterolemia, hypertriglyceridemia, cholelithiasis and sleep disorders, hyperlipidemic conditions, bulimia nervosa and compulsive eating disorders.
PCT/US2004/042542 2003-12-19 2004-12-20 Azabicyclic heterocycles as cannabinoid receptor modulators WO2005061509A1 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
EP04814691A EP1699796A4 (en) 2003-12-19 2004-12-20 Azabicyclic heterocycles as cannabinoid receptor modulators
JP2006545502A JP2007514756A (en) 2003-12-19 2004-12-20 Azabicyclic heterocycles as cannabinoid receptor modulators
IS8501A IS8501A (en) 2003-12-19 2006-06-08 Azate cyclic heterocycles as cannabinonide receptor antagonists
NO20062689A NO20062689L (en) 2003-12-19 2006-06-12 Azabicyclic heterocycles as modulators of cannabinoid receptors

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US53145103P 2003-12-19 2003-12-19
US60/531,451 2003-12-19

Publications (1)

Publication Number Publication Date
WO2005061509A1 true WO2005061509A1 (en) 2005-07-07

Family

ID=34710226

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2004/042542 WO2005061509A1 (en) 2003-12-19 2004-12-20 Azabicyclic heterocycles as cannabinoid receptor modulators

Country Status (11)

Country Link
US (1) US7037910B2 (en)
EP (1) EP1699796A4 (en)
JP (1) JP2007514756A (en)
CN (2) CN1918164A (en)
AR (2) AR046964A1 (en)
IS (1) IS8501A (en)
NO (1) NO20062689L (en)
PE (2) PE20050756A1 (en)
TW (2) TW200530246A (en)
WO (1) WO2005061509A1 (en)
ZA (2) ZA200604645B (en)

Cited By (27)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1623741A3 (en) * 2004-07-22 2006-06-07 Cadila Healthcare Ltd. Cannabinoid receptor ligands for hair growth modulation
WO2007127474A2 (en) * 2006-04-28 2007-11-08 Northwestern University Compositions and treatments using pyridazine compounds and cholinesterase inhibitors
WO2008017381A1 (en) 2006-08-08 2008-02-14 Sanofi-Aventis Arylaminoaryl-alkyl-substituted imidazolidine-2,4-diones, processes for preparing them, medicaments comprising these compounds, and their use
WO2009021740A2 (en) 2007-08-15 2009-02-19 Sanofis-Aventis Substituted tetrahydronaphthalenes, process for the preparation thereof and the use thereof as medicaments
JP2009532360A (en) * 2006-03-30 2009-09-10 アイアールエム・リミテッド・ライアビリティ・カンパニー Azolopyrimidines as inhibitors of cannabinoid receptor 1 activity
WO2010003624A2 (en) 2008-07-09 2010-01-14 Sanofi-Aventis Heterocyclic compounds, processes for their preparation, medicaments comprising these compounds, and the use thereof
WO2010068601A1 (en) 2008-12-08 2010-06-17 Sanofi-Aventis A crystalline heteroaromatic fluoroglycoside hydrate, processes for making, methods of use and pharmaceutical compositions thereof
US7816357B2 (en) 2003-12-19 2010-10-19 Bristol-Myers Squibb Company Azabicyclic heterocycles as cannabinoid receptor modulators
US7888357B2 (en) 2001-08-31 2011-02-15 Northwestern University Anti-inflammatory and protein kinase inhibitor compositions and related methods for downregulation of detrimental cellular responses and inhibition of cell death
WO2011023754A1 (en) 2009-08-26 2011-03-03 Sanofi-Aventis Novel crystalline heteroaromatic fluoroglycoside hydrates, pharmaceuticals comprising these compounds and their use
WO2011128810A1 (en) * 2010-04-14 2011-10-20 Centre National De La Recherche Scientifique Statins for the prevention or treatment of drug addictions
US8063047B2 (en) 2004-11-02 2011-11-22 Centre National De La Recherche Scientifique (Cnrs) Pyridazine compounds and methods
WO2011157827A1 (en) 2010-06-18 2011-12-22 Sanofi Azolopyridin-3-one derivatives as inhibitors of lipases and phospholipases
WO2012120052A1 (en) 2011-03-08 2012-09-13 Sanofi Oxathiazine derivatives substituted with carbocycles or heterocycles, method for producing same, drugs containing said compounds, and use thereof
WO2012120051A1 (en) 2011-03-08 2012-09-13 Sanofi Benzyl-oxathiazine derivates substituted with adamantane or noradamantane, medicaments containing said compounds and use thereof
WO2012120057A1 (en) 2011-03-08 2012-09-13 Sanofi Novel substituted phenyl-oxathiazine derivatives, method for producing them, drugs containing said compounds and the use thereof
WO2012120050A1 (en) 2011-03-08 2012-09-13 Sanofi Novel substituted phenyl-oxathiazine derivatives, method for producing them, drugs containing said compounds and the use thereof
WO2012120054A1 (en) 2011-03-08 2012-09-13 Sanofi Di- and tri-substituted oxathiazine derivates, method for the production thereof, use thereof as medicine and drug containing said derivatives and use thereof
WO2012120055A1 (en) 2011-03-08 2012-09-13 Sanofi Di- and tri-substituted oxathiazine derivates, method for the production thereof, use thereof as medicine and drug containing said derivatives and use thereof
WO2012120056A1 (en) 2011-03-08 2012-09-13 Sanofi Tetrasubstituted oxathiazine derivatives, method for producing them, their use as medicine and drug containing said derivatives and the use thereof
WO2012120058A1 (en) 2011-03-08 2012-09-13 Sanofi Oxathiazine derivatives which are substituted with benzyl or heteromethylene groups, method for producing them, their use as medicine and drug containing said derivatives and the use thereof
WO2012120053A1 (en) 2011-03-08 2012-09-13 Sanofi Branched oxathiazine derivatives, method for the production thereof, use thereof as medicine and drug containing said derivatives and use thereof
US8367672B2 (en) 2004-11-02 2013-02-05 Universite De Strasbourg Pyridazine compounds, compositions and methods
US8426439B2 (en) 2007-04-11 2013-04-23 Omeros Corporation Compositions and methods for prophylaxis and treatment of addictions
US9408845B2 (en) 2006-04-28 2016-08-09 Northwestern University Formulations containing pyridazine compounds
US10064850B2 (en) 2007-04-11 2018-09-04 Omeros Corporation Compositions and methods for prophylaxis and treatment of addictions
US11241420B2 (en) 2007-04-11 2022-02-08 Omeros Corporation Compositions and methods for prophylaxis and treatment of addictions

Families Citing this family (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100210590A1 (en) * 1995-09-27 2010-08-19 Northwestern University Compositions and treatments for seizure-related disorders
US7317012B2 (en) * 2005-06-17 2008-01-08 Bristol-Myers Squibb Company Bicyclic heterocycles as cannabinoind-1 receptor modulators
DE602007003348D1 (en) * 2006-05-04 2009-12-31 Solvay Pharm Bv MUSCARIN AGONISTS FOR THE TREATMENT OF IMPULSE CONTROL DISORDERS
EP2283159B1 (en) * 2008-05-05 2013-10-09 The Scripps Research Institute Synthesis of (+) cortistatin a and related compounds
WO2011131576A1 (en) * 2010-04-20 2011-10-27 Università Degli Studi Di Roma "La Sapienza" Tranylcypromine derivatives as inhibitors of histone demethylase lsd1 and/or lsd2
JP5437525B1 (en) 2012-12-28 2014-03-12 株式会社ナード研究所 Tyrosine derivative and method for producing tyrosine derivative
CN103933569B (en) * 2013-01-22 2017-01-11 复旦大学 Anti-lung cancer pharmaceutical composition, its application, pill case and package
HUE050964T2 (en) * 2014-01-29 2021-01-28 UCB Biopharma SRL Heteroaryl amides as inhibitors of protein aggregation
CA3026083A1 (en) * 2016-06-03 2017-12-07 The Trustees Of Columbia University In The City Of New York Methods of treating prader-willi syndrome
CN108178759B (en) * 2018-01-05 2020-06-09 上海瑞纷医药科技有限责任公司 Synthesis method of α -adrenoceptor antagonist
CN108498511A (en) * 2018-04-21 2018-09-07 徐晶 A kind of pharmaceutical composition and preparation method thereof for treating knee joint osseous arthritis

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4581449A (en) * 1983-04-28 1986-04-08 I.S.F. S.P.A. Process of making ethyl{6-[ethyl-(2-hydroxypropyl)amino]-3-pyridazinyl}hydrazinecarboxylate

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000020417A1 (en) 1998-10-06 2000-04-13 Takeda Chemical Industries, Ltd. Fused pyridazine compounds, process for the preparation of the same and uses thereof
JP2002205992A (en) 2000-11-08 2002-07-23 Takeda Chem Ind Ltd Bicyclic triazolone derivative and herbicide comprising the same
GB0107751D0 (en) * 2001-03-28 2001-05-16 Pfizer Ltd Pharmaceutically active compounds
BR0307595A (en) * 2002-02-07 2005-02-01 Pfizer Use of pde5 inhibitors such as sildenafil in the treatment of polycystic ovarian syndrome
ATE373660T1 (en) * 2002-03-25 2007-10-15 Merck & Co Inc HETEROCYCLIC BETA-AMINO COMPOUNDS AS INHIBITORS OF DIPEPTIDYLPEPTIDASE FOR TREATMENT OR PREVENTION OF DIABETES
WO2004074259A1 (en) 2003-02-19 2004-09-02 Neurogen Corporation Aryl acid pyrimidinyl methyl amides, pyridazinyl methyl amides and related compounds

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4581449A (en) * 1983-04-28 1986-04-08 I.S.F. S.P.A. Process of making ethyl{6-[ethyl-(2-hydroxypropyl)amino]-3-pyridazinyl}hydrazinecarboxylate

Cited By (35)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7888357B2 (en) 2001-08-31 2011-02-15 Northwestern University Anti-inflammatory and protein kinase inhibitor compositions and related methods for downregulation of detrimental cellular responses and inhibition of cell death
US8088774B2 (en) 2001-08-31 2012-01-03 Northwestern University Anti-inflammatory and protein kinase inhibitor compositions and related methods for downregulation of detrimental cellular responses and inhibition of cell death
US7816357B2 (en) 2003-12-19 2010-10-19 Bristol-Myers Squibb Company Azabicyclic heterocycles as cannabinoid receptor modulators
EP1623741A3 (en) * 2004-07-22 2006-06-07 Cadila Healthcare Ltd. Cannabinoid receptor ligands for hair growth modulation
US8063047B2 (en) 2004-11-02 2011-11-22 Centre National De La Recherche Scientifique (Cnrs) Pyridazine compounds and methods
US9663493B2 (en) 2004-11-02 2017-05-30 Northwestern University Pyridazine compounds, compositions and methods
US9527819B2 (en) 2004-11-02 2016-12-27 Northwestern University Pyridazine compounds, compositions and methods
US8933076B2 (en) 2004-11-02 2015-01-13 Centre National De La Recherche Scientifique (Cnrs) Pyridazine compounds, compositions and methods
US8367672B2 (en) 2004-11-02 2013-02-05 Universite De Strasbourg Pyridazine compounds, compositions and methods
US8722691B2 (en) 2006-03-30 2014-05-13 Irm Llc Azolopyrimidines as inhibitors of cannabinoid 1 activity
JP2009532360A (en) * 2006-03-30 2009-09-10 アイアールエム・リミテッド・ライアビリティ・カンパニー Azolopyrimidines as inhibitors of cannabinoid receptor 1 activity
US8158627B2 (en) 2006-04-28 2012-04-17 Northwestern University Compositions and treatments using pyridazine compounds and cholinesterase inhibitors
WO2007127474A2 (en) * 2006-04-28 2007-11-08 Northwestern University Compositions and treatments using pyridazine compounds and cholinesterase inhibitors
WO2007127474A3 (en) * 2006-04-28 2008-06-19 Univ Northwestern Compositions and treatments using pyridazine compounds and cholinesterase inhibitors
US9408845B2 (en) 2006-04-28 2016-08-09 Northwestern University Formulations containing pyridazine compounds
WO2008017381A1 (en) 2006-08-08 2008-02-14 Sanofi-Aventis Arylaminoaryl-alkyl-substituted imidazolidine-2,4-diones, processes for preparing them, medicaments comprising these compounds, and their use
US11241420B2 (en) 2007-04-11 2022-02-08 Omeros Corporation Compositions and methods for prophylaxis and treatment of addictions
US10064850B2 (en) 2007-04-11 2018-09-04 Omeros Corporation Compositions and methods for prophylaxis and treatment of addictions
US8426439B2 (en) 2007-04-11 2013-04-23 Omeros Corporation Compositions and methods for prophylaxis and treatment of addictions
WO2009021740A2 (en) 2007-08-15 2009-02-19 Sanofis-Aventis Substituted tetrahydronaphthalenes, process for the preparation thereof and the use thereof as medicaments
WO2010003624A2 (en) 2008-07-09 2010-01-14 Sanofi-Aventis Heterocyclic compounds, processes for their preparation, medicaments comprising these compounds, and the use thereof
WO2010068601A1 (en) 2008-12-08 2010-06-17 Sanofi-Aventis A crystalline heteroaromatic fluoroglycoside hydrate, processes for making, methods of use and pharmaceutical compositions thereof
WO2011023754A1 (en) 2009-08-26 2011-03-03 Sanofi-Aventis Novel crystalline heteroaromatic fluoroglycoside hydrates, pharmaceuticals comprising these compounds and their use
FR2958850A1 (en) * 2010-04-14 2011-10-21 Centre Nat Rech Scient DRUGS FOR THE PREVENTION OR TREATMENT OF ADDICTIONS TO DRUGS
WO2011128810A1 (en) * 2010-04-14 2011-10-20 Centre National De La Recherche Scientifique Statins for the prevention or treatment of drug addictions
WO2011157827A1 (en) 2010-06-18 2011-12-22 Sanofi Azolopyridin-3-one derivatives as inhibitors of lipases and phospholipases
WO2012120052A1 (en) 2011-03-08 2012-09-13 Sanofi Oxathiazine derivatives substituted with carbocycles or heterocycles, method for producing same, drugs containing said compounds, and use thereof
WO2012120053A1 (en) 2011-03-08 2012-09-13 Sanofi Branched oxathiazine derivatives, method for the production thereof, use thereof as medicine and drug containing said derivatives and use thereof
WO2012120058A1 (en) 2011-03-08 2012-09-13 Sanofi Oxathiazine derivatives which are substituted with benzyl or heteromethylene groups, method for producing them, their use as medicine and drug containing said derivatives and the use thereof
WO2012120056A1 (en) 2011-03-08 2012-09-13 Sanofi Tetrasubstituted oxathiazine derivatives, method for producing them, their use as medicine and drug containing said derivatives and the use thereof
WO2012120055A1 (en) 2011-03-08 2012-09-13 Sanofi Di- and tri-substituted oxathiazine derivates, method for the production thereof, use thereof as medicine and drug containing said derivatives and use thereof
WO2012120054A1 (en) 2011-03-08 2012-09-13 Sanofi Di- and tri-substituted oxathiazine derivates, method for the production thereof, use thereof as medicine and drug containing said derivatives and use thereof
WO2012120050A1 (en) 2011-03-08 2012-09-13 Sanofi Novel substituted phenyl-oxathiazine derivatives, method for producing them, drugs containing said compounds and the use thereof
WO2012120057A1 (en) 2011-03-08 2012-09-13 Sanofi Novel substituted phenyl-oxathiazine derivatives, method for producing them, drugs containing said compounds and the use thereof
WO2012120051A1 (en) 2011-03-08 2012-09-13 Sanofi Benzyl-oxathiazine derivates substituted with adamantane or noradamantane, medicaments containing said compounds and use thereof

Also Published As

Publication number Publication date
IS8501A (en) 2006-06-08
CN1918165A (en) 2007-02-21
JP2007514756A (en) 2007-06-07
PE20051047A1 (en) 2005-12-16
TW200530246A (en) 2005-09-16
PE20050756A1 (en) 2005-10-06
CN1918164A (en) 2007-02-21
ZA200604778B (en) 2007-12-27
TW200528455A (en) 2005-09-01
US7037910B2 (en) 2006-05-02
ZA200604645B (en) 2007-11-28
AR047063A1 (en) 2006-01-04
NO20062689L (en) 2006-09-12
AR046964A1 (en) 2006-01-04
US20050192278A1 (en) 2005-09-01
EP1699796A4 (en) 2009-03-04
EP1699796A1 (en) 2006-09-13

Similar Documents

Publication Publication Date Title
US7037910B2 (en) Azabicyclic heterocycles as cannabinoid receptor modulators
EP1697371B1 (en) Azabicyclic heterocycles as cannabinoid receptor modulators
EP1697370B1 (en) Azabicyclic heterocycles as cannabinoid receptor modulators
EP1670460B1 (en) Pyrazole derivatives as cannabinoid receptor modulators
EP1891074B1 (en) Azabicyclic heterocycles as cannabinoid receptor modulators
WO2005016286A2 (en) Pyrazine modulators of cannabinoid receptors
WO2005007111A2 (en) Tetrahydroquinoline derivatives as cannabinoid receptor modulators
WO2006138657A1 (en) Bicyclic heterocycles as cannabinoid-1 receptor modulators
US20060160850A1 (en) Bicyclic heterocycles as cannabinoid receptor modulators
US7317012B2 (en) Bicyclic heterocycles as cannabinoind-1 receptor modulators
US20060287342A1 (en) Triazolopyrimidine heterocycles as cannabinoid receptor modulators
US7361766B2 (en) Bicyclic heterocycles as cannabinoid receptor modulators
EP2094705B1 (en) Azabicyclic heterocycles as cannabinoid receptor modulators
US7314882B2 (en) Bicyclic heterocycles as cannabinoid receptor modulators
US7368458B2 (en) Bicyclic heterocycles as cannabinoid receptor modulators
MXPA06006288A (en) Azabicyclic heterocycles as cannabinoid receptor modulators

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BW BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG 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 MA MD MG MK MN MW MX MZ NA NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): GM KE LS MW MZ NA SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LT LU MC NL PL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

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: 2006545502

Country of ref document: JP

NENP Non-entry into the national phase

Ref country code: DE

WWW Wipo information: withdrawn in national office

Country of ref document: DE

WWE Wipo information: entry into national phase

Ref document number: 2004814691

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 2004814691

Country of ref document: EP