WO2005061505A1 - Bicyclic imidazolyl pyrimidin-4-one cannabinoid receptor ligands and uses thereof - Google Patents

Bicyclic imidazolyl pyrimidin-4-one cannabinoid receptor ligands and uses thereof Download PDF

Info

Publication number
WO2005061505A1
WO2005061505A1 PCT/IB2004/004019 IB2004004019W WO2005061505A1 WO 2005061505 A1 WO2005061505 A1 WO 2005061505A1 IB 2004004019 W IB2004004019 W IB 2004004019W WO 2005061505 A1 WO2005061505 A1 WO 2005061505A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
alkyl
salt
substituted
substituents
Prior art date
Application number
PCT/IB2004/004019
Other languages
French (fr)
Inventor
Philip Albert Carpino
Original Assignee
Pfizer Products Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Pfizer Products Inc. filed Critical Pfizer Products Inc.
Publication of WO2005061505A1 publication Critical patent/WO2005061505A1/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
    • A61P25/00Drugs for disorders of the nervous system

Definitions

  • the present invention relates to bicyclic imidazolyl pyrimidinone (e.g., 1 ,9-dihydro-purin-6-one) compounds as cannabinoid receptor ligands, in particular CB1 receptor antagonists, and uses thereof for treating diseases, conditions and/or disorders modulated by cannabinoid receptor antagonists.
  • BACKGROUND Obesity is a major public health concern because of its increasing prevalence and associated health risks. Obesity and overweight are generally defined by body mass index (BMI), which is correlated with total body fat and estimates the relative risk of disease. BMI is calculated by weight in kilograms divided by height in meters squared (kg/m 2 ).
  • Overweight is typically defined as a BMI of 25-29.9 kg/m 2
  • obesity is typically defined as a BMI of 30 kg/m 2 .
  • the increase in obesity is of concern because of the excessive health risks associated with obesity, including coronary heart disease, strokes, hypertension, type 2 diabetes mellitus, dyslipidemia, sleep apnea, osteoarthritis, gall bladder disease, depression, and certain forms of cancer (e.g., endometrial, breast, prostate, and colon).
  • the negative health consequences of obesity make it the second leading cause of preventable death in the United States and impart a significant economic and psychosocial effect on society. See, McGinnis M, Foege WH., "Actual Causes of Death in the United States," JAMA, 270, 2207-12 (1993). Obesity is now recognized as a chronic disease that requires treatment to reduce its associated health risks.
  • weight loss is an important treatment outcome
  • one of the main goals of obesity management is to improve cardiovascular and metabolic values to reduce obesity-related morbidity and mortality. It has been shown that 5-10% loss of body weight can substantially improve metabolic values, such as blood glucose, blood pressure, and lipid concentrations. Hence, it is believed that a 5-10% intentional reduction in body weight may reduce morbidity and mortality.
  • Currently available prescription drugs for managing obesity generally reduce weight by inducing satiety or decreasing dietary fat absorption. Satiety is achieved by increasing synaptic levels of norepinephrine, serotonin, or both. For example, stimulation of serotonin receptor subtypes 1 B, 1 D, and 2C and 1- and 2-adrenergic receptors decreases food intake by regulating satiety.
  • Adrenergic agents e.g., diethylpropion, benzphetamine, phendimetrazine, mazindol, and phentermine
  • Adrenergic agents act by modulating central norepinephrine and dopamine receptors through the promotion of catecholamine release.
  • Older adrenergic weight-loss drugs e.g., amphetamine, methamphetamine, and phenmetrazine
  • CB1 cannabinoid receptor antagonists/inverse agonists have been suggested as potential appetite suppressants. See, e.g., Arnone, M., et al., "Selective Inhibition of Sucrose and Ethanol Intake by SR141716, an Antagonist of Central Cannabinoid (CB1) Receptors," Psvchopharmacol, 132, 104-106 (1997); Colombo, G., et al., "Appetite
  • R 1 is an aryl optionally substituted with one or more substituents, or a heteroaryl optionally substituted with one or more substituents
  • R 3 is hydrogen, (C- ⁇ -C 4 )alkyl, halo-substituted (C ⁇ -C 4 )alkyl, or (C C 4 )alkoxy, or R 3 taken together with
  • R 1 and R 2 are each independently a substituted phenyl. More preferably, R 1 and R 2 are each independently a phenyl substituted with one to three substituents independently selected from the group consisting of halo (preferably, chloro or fluoro), (C ⁇ -C 4 )alkoxy, (CrC 4 )alkyl, halo-substituted (C- ⁇ -C 4 )alkyl (preferably fluoro-substituted alkyl, more preferably, trifluoromethyl) and cyano.
  • halo preferably, chloro or fluoro
  • C ⁇ -C 4 alkoxy
  • CrC 4 halo-substituted alkyl
  • cyano preferably fluoro-substituted alkyl, more preferably, trifluoromethyl
  • R 1 is 2-cyanophenyl, 2- chlorophenyl, 2-fluorophenyl, 2,4-dichlorophenyl, 2-fluoro-4-chlorophenyl, 2- chloro-4-fluorophenyl, 2-methylphenyl or 2,4-difluorophenyl; and R 2 is 4- chlorophenyl, 4-cyanophenyl, 4-ethyIphenyl, 4-isopropylphenyl, 4- ethoxyphenyl, 4-isopropoxyphenyl, 4-trifluoromethylphenyl, or 4-fluorophenyl.
  • R 3 is hydrogen, methyl or trifluoromethyl.
  • R 4 is a chemical moiety selected from the group consisting of (Ci-Csjalkyl, aryl(C ⁇ -C 4 )alkyl, and 3- to 8-membered partially or fully saturated carbocyclic ring(s), and 3- to 8-membered partially or fully saturated heterocycle, where said chemical moiety is optionally substituted with one or more substituent.
  • R 4 is (C ⁇ -C 8 )alkyl, halo- substituted (C ⁇ -Cs)alkyl (preferably, fluoro-substituted (C ⁇ -C 8 )alkyl), cyclopentyl, cyclohexyl, piperidin-1-yl, pyrrolidin-1-yl, or morpholin-1-yl.
  • a compound of Formula (II) is provided.
  • R 1a , R 1b , R 2b , and R 2c are each independently halo, (d-C ⁇ alkoxy, (C C 4 )alkyl, halo-substituted (C ⁇ -C 4 )alkyl, or cyano (preferably, R 1a is chloro, fluoro, methyl, or cyano; R 1b is chloro, fluoro or hydrogen (i.e., m is 0); R 2b is chloro, fluoro, (C- ⁇ -C 4 )alkyl, trifluoromethyl, (CrC 4 )aIkoxy, or cyano; and R 2c is hydrogen (i.e., n is 0)); n and m are each independently 0, 1 or 2; and R 3 and R 4 are as defined above; a pharmaceutically acceptable salt thereof, a prodrug of the compound or the salt, or a solvate or hydrate of the compound, the salt or the prodrug.
  • Preferred compounds of Formula (I) or (II) include: 9-(4- chlorophenyl)-8-(2-chlorophenyl)-1 -isopropyl-1 ,9-dihydropurin-6-one; 8-(2- chloro-phenyl)-1-(2,2,2-trifluoro-ethyl)-9-(4-trifluoromethyl-phenyl)-1 ,9- dihydro-purin-6-one; 8-o-tolyl-1 -(2,2,2-trifluoro-ethyl)-9-(4-trifluoromethyl- phenyl)-1 ,9-dihydro-purin-6-one; 8-(2-chloro-phenyl)-9-(4-ethyl-phenyl)-1 - (2,2,2-trifluoro-ethyl)-1 ,9-dihydro-purin-6-one; 9-(4-tert-butyl-phenyl)-8-(2- chloro
  • compositions that comprises (1 ) a compound of the present invention, and (2) a pharmaceutically acceptable excipient, diluent, or carrier.
  • the composition comprises a therapeutically effective amount of a compound of the present invention.
  • composition may also contain at least one additional pharmaceutical agent (described herein).
  • Preferred agents include nicotine receptor partial agonists, opioid antagonists (e.g., naltrexone and nalmefene), dopaminergic agents (e.g., apomorphine), attention deficit disorder (ADD including attention deficit hyperactivity disorder (ADHD)) agents (e.g., RitalinTM, StratteraTM, ConcertaTM and AdderallTM), and anti- obesity agents (described herein below).
  • a method for treating a disease, condition or disorder modulated by a cannabinoid receptor (preferably, a CB1 receptor) antagonists in animals that includes the step of administering to an animal in need of such treatment a therapeutically effective amount of a compound of the present invention (or a pharmaceutical composition thereof).
  • a cannabinoid receptor preferably, a CB1 receptor
  • Diseases, conditions, and/or disorders modulated by cannabinoid receptor antagonists include eating disorders (e.g., binge eating disorder, anorexia, and bulimia), weight loss or control (e.g., reduction in calorie or food intake, and/or appetite suppression), obesity, depression, atypical depression, bipolar disorders, psychoses, schizophrenia, behavioral addictions, suppression of reward-related behaviors (e.g., conditioned place avoidance, such as suppression of cocaine- and morphine-induced conditioned place preference), substance abuse, addictive disorders, impulsivity, alcoholism (e.g., alcohol abuse, addiction and/or dependence including treatment for abstinence, craving reduction and relapse prevention of alcohol intake), tobacco abuse (e.g., smoking addiction, cessation and/or dependence including treatment for craving reduction and relapse prevention of tobacco smoking), dementia (including memory loss, Alzheimer's disease, dementia of aging, vascular dementia, mild cognitive impairment, age-related cognitive decline, and mild neurocognitive disorder), sexual dysfunction in males (e.g.,
  • the method is used in the treatment of weight loss, obesity, bulimia, ADD/ADHD, dementia, alcoholism, and/or tobacco abuse.
  • Compounds of the present invention may be administered in combination with other pharmaceutical agents.
  • Preferred pharmaceutical agents include nicotine receptor partial agonists, opioid antagonists (e.g., naltrexone (including naltrexone depot), antabuse, and nalmefene), dopaminergic agents (e.g., apomorphine), ADD/ADHD agents (e.g., methylphenidate hydrochloride (e.g., RitalinTM and ConcertaTM), atomoxetine (e.g., StratteraTM), and amphetamines (e.g., AdderallTM)) and anti-obesity agents, such as apo-B/MTP inhibitors, 11 ⁇ -hydroxy steroid dehydrogenase-1 (11 ⁇ -HSD type 1 ) inhibitors, peptide YY 3-36 or analog
  • the combination therapy may be administered as (a) a single pharmaceutical composition which comprises a compound of the present invention, at least one additional pharmaceutical agent described herein and a pharmaceutically acceptable excipient, diluent, or carrier; or (b) two separate pharmaceutical compositions comprising (i) a first composition comprising a compound of the present invention and a pharmaceutically acceptable excipient, diluent, or carrier, and (ii) a second composition comprising at least one additional pharmaceutical agent described herein and a pharmaceutically acceptable excipient, diluent, or carrier.
  • the pharmaceutical compositions may be administered simultaneously or sequentially and in any order. Definitions As used herein, the term "alkyl” refers to a hydrocarbon radical of the general formula C n H 2n + ⁇ .
  • the alkane radical may be straight or branched.
  • (C ⁇ -C 6 )alkyl refers to a monovalent, straight, or branched aliphatic group containing 1 to 6 carbon atoms (e.g., methyl, ethyl, n-propyl, /-propyl, ⁇ -butyl, /-butyl, s-butyl, f-butyl, ⁇ -pentyl, 1-methylbutyl, 2- methylbutyi, 3-methyl butyl, neopentyl, 3,3-dimethylpropyl, hexyl, 2- methylpentyl, and the like).
  • alkyl portion i.e., alkyl moiety
  • acyl e.g., alkanoyl
  • alkylamino dialkylamino
  • alkylthio group alkyl portion of an alkoxy, acyl (e.g., alkanoyl), alkylamino, dialkylamino, and alkylthio group
  • alkane radical or alkyl moiety may be unsubstituted or substituted with one or more substituents (generally, one to three substituents except in the case of halogen substituents such as perchloro or perfluoroalkyls) independently selected from the group of substituents listed below in the definition for "substituted.”
  • Halo-substituted alkyl refers to an alkyl group substituted with one or more halogen atoms (e.g., "fluoro- substituted alkyl” refers to fluoromethyl, difluoromethyl, trifluoromethyl, 1- fluoroethyl, 2-fluoroethyl, 1 ,1-difluoroethyl, 1 ,2-difluoroethyl, 2,2-difluoroethyl, 1 ,1 ,1-trifluoroethyl, 2,2,2-trifluoroethy
  • Preferred halo-substituted alkyls are the chloro- and fluoro-substituted alkyls, more preferably, fluoro-substituted alkyls.
  • the alkane radicals or alkyl moieties are preferably substituted with fluoro substituents (as described above), or 1 or 2 substituents independently selected from (C C 3 )alkyl, (C 3 -C 6 )cycloalkyl, (C 2 -C 3 )alkenyl, aryl, heteroaryl, 3- to 6- membered heterocycle, chloro, cyano, hydroxy, (C ⁇ -C 3 )alkoxy, aryloxy, amino, (C ⁇ -C 6 )alkyl amino, di-(C ⁇ -C 4 )alkyl amino, aminocarboxylate (i.e., (C-i- C )alkyl-0-C(0)-NH-), hydroxy(C 2 -C 3 )alkyla
  • partially or fully saturated carbocyclic ring refers to nonaromatic rings that are either partially or fully hydrogenated and may exist as a single ring, bicyclic ring or a spiral ring. Unless specified otherwise, the carbocyclic ring is generally a 3- to 8-membered ring.
  • partially or fully saturated carbocyclic rings include groups such as cyclopropyl, cyclopropenyl, cyclobutyl, cyclobutenyl, cyclopentyl, cyclpentenyl, cyclopentadienyl, cyclohexyl, cyclohexenyl, cyclohexadienyl, norbomyl (bicyclo[2.2.1]heptyl), norbomenyl, bicyclo[2.2.2]octyl, and the like.
  • the partially saturated or fully saturated cycloalkyl group may be unsubstituted or substituted with one or more substituents (typically, one to three substituents) independently selected from the group of substituents listed below in the definition for "substituted.”
  • a substituted carbocyclic ring also includes groups wherein the carbocyclic ring is fused to a phenyl ring (e.g., indanyl).
  • the carbocyclic group may be attached to the chemical entity or moiety by any one of the carbon atoms within the carbocyclic ring system.
  • the carbocyclic group is preferably substituted with 1 or 2 substituents independently selected from (CrC 3 )alkyl, (C 2 -C 3 )alkenyl, (C ⁇ -C 6 )alkylidenyl, aryl, heteroaryl, 3- to 6-membered heterocycle, chloro, fluoro, cyano, hydroxy, (C 1 -C 3 )alkoxy, aryloxy, amino, (C Ce)al yl amino, di-(CrC 4 )alkyl amino, aminocarboxylate (i.e., (C ⁇ -C 3 )alkyl-O-C(0)-NH-), hydroxy(C 2 - C 3 )alkylamino, or keto (oxo), and more preferably 1 or 2 from substituents independently selected from (CrC 2 )alkyl, 3- to 6-membered heterocycle, fluoro, (C ⁇ -C 3 )alkoxy, (CrC )alkyl amino
  • any cycloalkyl portion of a group (e.g., cycloalkylalkyl, cycloalkylamino, etc.) has the same definition as above.
  • the term "partially saturated or fully saturated heterocyclic ring” (also referred to as “partially saturated or fully saturated heterocycle”) refers to nonaromatic rings that are either partially or fully hydrogenated and may exist as a single ring, bicyclic ring or a spiral ring.
  • the heterocyclic ring is generally a 3- to 6-membered ring containing 1 to 3 heteroatoms (preferably 1 or 2 heteroatoms) independently selected from sulfur, oxygen and/or nitrogen.
  • Partially saturated or fully saturated heterocyclic rings include groups such as epoxy, aziridinyl, tetrahydrofuranyl, dihydrofuranyl, dihydropyridinyl, pyrrolidinyl, N- methylpyrrolidinyl, imidazolidinyl, imidazolinyl, piperidinyl, piperazinyl, pyrazolidinyl, 2H-pyranyl, 4H-pyranyl, 2H-chromenyl, oxazinyl, morpholino, thiomorpholino, tetrahydrothienyl, tetrahydrothienyl 1 ,1 -dioxide, and the like.
  • the partially saturated or fully saturated heterocycle group may be unsubstiuted or substituted with one or more substituents (typically, one to three substituents) independently selected from the group of substituents listed below in the definition for "substituted.”
  • a substituted heterocyclic ring includes groups wherein the heterocyclic ring is fused to an aryl or heteroaryl ring (e.g., 2,3- dihydrobenzofuranyl, 2,3-dihydroindolyl, 2,3-dihydrobenzothiophenyl, 2,3- dihydrobenzothiazolyl, etc.).
  • the heterocycle group is preferably substituted with 1 or 2 substituents independently selected from (C ⁇ -C 3 )alkyl, (C 3 -C 6 )cycloalkyl, (C 2 -C 4 )alkenyl, aryl, heteroaryl, 3- to 6- membered heterocycle, chloro, fluoro, cyano, hydroxy, (C-i-CsJalkoxy, aryloxy, amino, (C ⁇ -C 6 )alkyl amino, di-(C ⁇ -C 3 )alkyl amino, aminocarboxylate (i.e., (CrC 3 )alkyl-0-C(0)-NH-), or keto (oxo), and more preferably with 1 or 2 substituents independently selected from (C ⁇ -C 3 )alkyl, (C 3 -C 6 )cycloalkyl, (C ⁇ jaryl, 6-membered-heteroaryl, 3- to 6-membered heterocycle, or fluoro.
  • substituents independently selected from
  • the heterocyclic group may be attached to the chemical entity or moiety by any one of the ring atoms within the heterocyclic ring system.
  • any heterocycle portion of a group e.g., heterocycle-substituted alkyl, heterocycle carbonyl, etc.
  • aryl or "aromatic carbocyclic ring” refers to aromatic moieties having a single (e.g., phenyl) or a fused ring system (e.g., naphthalene, anthracene, phenanthrene, etc.).
  • a typical aryl group is a 6- to 10-membered aromatic carbocyclic ring(s).
  • the aryl groups When indicated as being “optionally substituted,” the aryl groups may be unsubstituted or substituted with one or more substituents (preferably no more than three substituents) independently selected from the group of substituents listed below in the definition for "substituted.”
  • substituents preferably no more than three substituents
  • Substituted aryl groups include a chain of aromatic moieties (e.g., biphenyl, terphenyl, phenylnaphthalyl, etc.).
  • the aromatic moieties are preferably substituted with 1 or 2 substituents independently selected from (C ⁇ -C )alkyl, (C 2 -C 3 )alkenyl, aryl, heteroaryl, 3- to 6-membered heterocycle, bromo, chloro, fluoro, iodo, cyano, hydroxy, (C ⁇ -C 4 )alkoxy, aryloxy, amino, (C ⁇ -C 6 )alkyl amino, di-(C ⁇ -C 3 )alkyl amino, or aminocarboxylate (i.e., (Ci-C 3 )alkyl-0-C(0)-NH-), and more preferably, 1 or 2 substituents independently selected from (C ⁇ -C 4 )alkyl, chloro, fluoro, cyano, hydroxy, or (C ⁇ -C 4 )alkoxy.
  • 1 or 2 substituents independently selected from (C ⁇ -C 4 )alkyl, chloro, fluoro, cyano, hydroxy,
  • the aryl group may be attached to the chemical entity or moiety by any one of the carbon atoms within the aromatic ring system.
  • the aryl portion (i.e., aromatic moiety) of an aroyl or aroyloxy (i.e., (aryl)-C(O)-O-) has the same definition as above.
  • heteroaryl or “heteroaromatic ring” refers to aromatic moieties containing at least one heteratom (e.g., oxygen, sulfur, nitrogen or combinations thereof) within a 5- to 10-membered aromatic ring system (e.g., pyrrolyl, pyridyl, pyrazolyl, indolyl, indazolyl, thienyl, furanyl, benzofuranyl, oxazolyl, imidazolyl, tetrazolyl, triazinyl, pyrimidyl, pyrazinyl, thiazolyl, purinyl, benzimidazolyl, quinolinyl, isoquinolinyl, benzothiophenyl, benzoxazolyl, etc.).
  • a 5- to 10-membered aromatic ring system e.g., pyrrolyl, pyridyl, pyrazolyl, indolyl, indazolyl,
  • the heteroaromatic moiety may consist of a single or fused ring system.
  • a typical single heteroaryl ring is a 5- to 6-membered ring containing one to three heteroatoms independently selected from oxygen, sulfur and nitrogen and a typical fused heteroaryl ring system is a 9- to 10- membered ring system containing one to four heteroatoms independently selected from oxygen, sulfur and nitrogen.
  • the heteroaryl groups may be unsubstituted or substituted with one or more substituents (preferably no more than three substituents) independently selected from the group of substituents listed below in the definition for "substituted.”
  • the heteroaromatic moieties are preferably substituted with 1 or 2 substituents independently selected from (C ⁇ -C 4 )alkyl, (C 2 -C 3 )alkenyl, aryl, heteroaryl, 3- to 6-membered heterocycle, bromo, chloro, fluoro, iodo, cyano, hydroxy, (C-i- C- alkoxy, aryloxy, amino, (C ⁇ -Ce)alkyl amino, di-(C ⁇ -C 3 )alkyl amino, or aminocarboxylate (i.e., (C ⁇ -C 3 )alkyl-0-C(O)-NH-), and more preferably, 1 or 2 substituents independently selected from (C ⁇ -C 4 )alkyl, (C 2
  • the heteroaryl group may be attached to the chemical entity or moiety by any one of the atoms within the aromatic ring system (e.g., imidazol-1-yl, imidazol-2-yl, imidazol-4-yl, imidazol-5-yl, pyrid-2-yl, pyrid-3-yl, pyrid-4-yl, pyrid-5-yl, or pyrid-6-yl).
  • the heteroaryl portion i.e., heteroaromatic moiety
  • a heteroaroyl or heteroaroyloxy i.e., (heteroaryl)- C(O)-O-
  • acyl refers to hydrogen, alkyl, partially saturated or fully saturated cycloalkyl, partially saturated or fully saturated heterocycle, aryl, and heteroaryl substituted carbonyl groups.
  • acyl includes groups such as (C ⁇ -Ce)alkanoyl (e.g., formyl, acetyl, propionyl, butyryl, valeryl, caproyl, f-butylacetyl, etc.), (C 3 -C 6 )cycloalkylcarbonyl (e.g., cyclopropylcarbonyl, cyclobutylcarbonyl, cyclopentylcarbonyl, cyclohexylcarbonyl, etc.), heterocyclic carbonyl (e.g., pyrrolidinylcarbonyl, pyrrolid-2-one-5-carbonyl, piperidinylcarbonyl, piperazinylcarbonyl, tetrahydrofuranylcarbonyl, etc.
  • alkyl, cycloalkyl, heterocycle, aryl and heteroaryl portion of the acyl group may be any one of the groups described in the respective definitions above.
  • the acyl group may be unsubstituted or optionally substituted with one or more substituents (typically, one to three substituents) independently selected from the group of substituents listed below in the definition for "substituted” or the alkyl, cycloalkyl, heterocycle, aryl and heteroaryl portion of the acyl group may be substituted as described above in the preferred and more preferred list of substituents, respectively.
  • substituted specifically envisions and allows for one or more substitutions that are common in the art. However, it is generally understood by those skilled in the art that the substituents should be selected so as to not adversely affect the pharmacological characteristics of the compound or adversely interfere with the use of the medicament.
  • Suitable substituents for any of the groups defined above include (C C 6 )alkyl, (C 3 -C 7 )cycloalkyl, (C 2 -C 6 )alkenyl, (C ⁇ -C 6 )alkylidenyl, aryl, heteroaryl, 3- to 6-membered heterocycle, halo (e.g., chloro, bromo, iodo and fluoro), cyano, hydroxy, (CrC 6 )alkoxy, aryloxy, sulfhydryl (mercapto), (C ⁇ -C 6 )alkylthio, arylthio, amino, mono- or di-(C ⁇ -Ce)alkyl amino, quaternary ammonium salts, amino(C ⁇ -C 6 )alkoxy, aminocarboxylate (i.e., (CrC 6 )alkyl- O-C(O)-NH-), hydroxy(C 2 -C 6 )alkylamin
  • substituted combinations such as "substituted aryl(C ⁇ -C 6 )alkyl"
  • either the aryl or the alkyl group may be substituted, or both the aryl and the alkyl groups may be substituted with one or more substituents (typically, one to three substituents except in the case of perhalo substitutions).
  • An aryl or heteroaryl substituted carbocyclic or heterocyclic group may be a fused ring (e.g., indanyl, dihydrobenzofuranyl, dihydroindolyl, etc.).
  • solvate refers to a molecular complex of a compound represented by Formula (I) or (II) (including prodrugs and pharmaceutically acceptable salts thereof) with one or more solvent molecules.
  • solvent molecules are those commonly used in the pharmaceutical art, which are known to be innocuous to the recipient, e.g., water, ethanol, and the like.
  • hydrate refers to the complex where the solvent molecule is water.
  • protecting group or “Pg” refers to a substituent that is commonly employed to block or protect a particular functionality while reacting other functional groups on the compound.
  • an “amino- protecting group” is a substituent attached to an amino group that blocks or protects the amino functionality in the compound.
  • Suitable amino-protecting groups include acetyl, trifluoroacetyl, f-butoxycarbonyl (BOC), benzyloxycarbonyl (CBz) and 9-fluorenylmethylenoxycarbonyl (Fmoc).
  • a "hydroxy-protecting group” refers to a substituent of a hydroxy group that blocks or protects the hydroxy functionality.
  • Suitable protecting groups include acetyl and silyl.
  • a “carboxy-protecting group” refers to a substituent of the carboxy group that blocks or protects the carboxy functionality.
  • Common carboxy-protecting groups include -CH 2 CH 2 SO 2 Ph, cyanoethyl, 2-(trimethylsilyl)ethyl, 2-(trimethylsilyl)ethoxymethyl, 2-(p- toluenesulfonyl)ethyl, 2-(p-nitrophenylsulfenyl)ethyl, 2-(diphenylphosphino)- ethyl, nitroethyl and the like.
  • protecting groups and their use see T. W. Greene, Protective Groups in Organic Synthesis, John Wiley & Sons, New York, 1991.
  • terapéuticaally effective amount means an amount of a compound of the present invention that (i) treats or prevents the particular disease, condition, or disorder, (ii) attenuates, ameliorates, or eliminates one or more symptoms of the particular disease, condition, or disorder, or (iii) prevents or delays the onset of one or more symptoms of the particular disease, condition, or disorder described herein.
  • animal refers to humans (male or female), companion animals (e.g., dogs, cats and horses), food-source animals, zoo animals, marine animals, birds and other similar animal species.
  • “Edible animals” refers to food-source animals such as cows, pigs, sheep and poultry.
  • phrases "pharmaceutically acceptable” indicates that the substance or composition must be compatible chemically and/or toxicologically, with the other ingredients comprising a formulation, and/or the mammal being treated therewith.
  • the terms “treating”, “treat”, or “treatment” embrace both preventative, i.e., prophylactic, and palliative treatment.
  • the terms “modulated by a cannabinoid receptor” or “modulation of a cannabinoid receptor” refers to the activation or deactivation of a cannabinoid receptor.
  • a ligand may act as an agonist, partial agonist, inverse agonist, antagonist, or partial antagonist.
  • antagonist includes both full antagonists and partial antagonists, as well as inverse agonists.
  • CB-1 receptor refers to the G-protein coupled type 1 cannabinoid receptor.
  • compounds of the present invention refer to compounds of Formulae (I) and (II), prodrugs thereof, pharmaceutically acceptable salts of the compounds, and/or prodrugs, and hydrates or solvates of the compounds, salts, and/or prodrugs, as well as, all stereoisomers (including diastereoisomers and enantiomers), tautomers and isotopically labeled compounds.
  • DETAILED DESCRIPTION Compounds of the present invention may be synthesized by synthetic routes that include processes analogous to those well-known in the chemical arts, particularly in light of the description contained herein.
  • the starting materials are generally available from commercial sources such as Aldrich Chemicals (Milwaukee, Wl) or are readily prepared using methods well known to those skilled in the art (e.g., prepared by methods generally described in Louis F. Fieser and Mary Fieser, Reagents for Organic Synthesis, v. 1 -19, Wiley, New York (1967-1999 ed.), or Beilsteins Handbuch der organischen Chemie, 4, Aufl. ed. Springer-Verlag, Berlin, including supplements (also available via the Beilstein online database)).
  • the reaction schemes depicted below provide potential routes for synthesizing the compounds of the present invention as well as key intermediates. For a more detailed description of the individual reaction steps, see the Examples section below.
  • Suitable amino-protecting groups include acetyl, trifluoroacetyl, f-butoxycarbonyl (BOC), benzyloxycarbonyl (CBz) and 9- fluorenylmethyleneoxycarbonyl (Fmoc).
  • BOC f-butoxycarbonyl
  • CBz benzyloxycarbonyl
  • Fmoc 9- fluorenylmethyleneoxycarbonyl
  • Suitable amino compounds include those compounds where R 2 is ' aryl (e.g., aniline) or substituted aryl (e.g., 2-chloroaniline, 2-fluoroaniline, 2,4-dichloroaniline, 2-fluoro-4-chloroaniline, 2-chloro-4-flurooaniline, 2,4- difluoroaniline, and other substituted arylamines).
  • R 2 is aryl (e.g., aniline) or substituted aryl (e.g., 2-chloroaniline, 2-fluoroaniline, 2,4-dichloroaniline, 2-fluoro-4-chloroaniline, 2-chloro-4-flurooaniline, 2,4- difluoroaniline, and other substituted arylamines).
  • Intermediate 1 (a) can then be acylated using conventional chemistry well-known to those skilled in the art.
  • intermediate 1 (a) may be reacted with the desired aroyl or heteroaroyl chloride in a basic solvent (e.g., pyridine) to produce intermediate 1 (b).
  • a basic solvent e.g., pyridine
  • intermediate 1 (a) may be reacted with the desired aroyl or heteroaroyl chloride in a reaction inert solvent (e.g., tetrahydrofuran, methylene chloride, ⁇ /, ⁇ /-dimethyl- acetamide).
  • a suitable base e.g., triethylamine, diisopropyl- ethylamine
  • Suitable aroyl chlorides include benzoyl chloride, o-chlorobenzoyl chlorides, o-fluorobenzoyl chloride, p-chlorobenzoyl chloride, p-fluorobenzoyl chloride, 2,4-dichlorobenzoyl chloride, 2,4-difluorobenzoyl chloride, and the like.
  • Intermediate 1(b) may then be cyclized to the hydroxy purine (9H- purin-6-ol) intermediate 1 (c) by refluxing in a weak acid (e.g., acetic acid) or with sulfuric acid in an appropriate solvent (e.g., isopropyl alcohol, toluene).
  • N-Alkylation of intermediate (1c) can then be accomplished using an alkylating agent, preferably an alkyl iodide, alkyl triflate, or a substituted benzyl bromide, and a base, such as lithium hexamethyldisilazide, cesium carbonate, potassium carbonate or lithium hydride, in a polar non-protic solvent such as DMF or THF, at temperatures ranging from about 37°C to about 150 °C to provide compounds of Formula (I) or (II).
  • Scheme II below illustrates an alternative synthesis of intermediate 1(c).
  • Intermediate 2(a) can be first acylated with an acid chloride.
  • Suitable acid chlorides include those compounds where R 1 is aryl (e.g., benzoyl chloride), substituted aryl (e.g., 2-chlorobenzoyl chloride, 4-chlorobenzoyl chloride, and other substituted aryl acid chlorides), heteroaryl, or substituted heteroaryl.
  • Compound 2(b) may be converted to intermediate 2(c) by treatment with dehydrating agents like POCI 3 using procedures described by H.C. Koppel in J. Org. Chem.. 23, 1457 (1958) and in J. Chem. Soc, Perkin Trans. I. 879 (1984).
  • the R 1 group where R 1 is aryl, substituted aryl, heteroaryl or substituted heteroaryl, may then be introduced using reagents like R 2 -B(OH) 2 , R 2 -Br or R 2 -l and Pd catalysts (see Y. Wan et al. in Synthesis, 1597-1600 (2002) , and references contained therein) or copper(ll) catalysts such as cupric acetate or cupric bromide (see, S. Ding et al. in Tetrahedron Lett.. 42, 8751-8755 (2001 ), A. Klapars et al. J. Am. Chem. Soc. 123, 7727-7729 (2001 ), and references contained therein).
  • Scheme III describes the preparation of compounds of formula (I) or (II) where R 4 is alkyl, cycloalkyl, piperidinyl, pyrrolidiny, aryl or heteroaryl and R 3 is hydrogen or (C1-C4) alkyl.
  • Intermediate 3(a) can be prepared by treating an appropriately substituted amine derivative (e.g., R 2 NH 2 ) with trimethylaluminum under inert atmospheric conditions, followed by condensation with the appropriately substituted R 1 CN derivative.
  • an appropriately substituted amine derivative e.g., R 2 NH 2
  • trimethylaluminum under inert atmospheric conditions
  • Suitable amines include substituted phenyl amines (e.g., 4-chlorophenyl amine, 4-fluorophenyl amine, 4-bromophenyl amine, 4-iodophenyl amine, 4-cyanophenyl amine, and the like) pyridin-2-yl amine, pyridin-3-yl amine, pyridin-4-yl amine, substituted pyridinyl amines (e.g., 2-dimethylaminopyridin-5-yl amine, 2-methoxypyridin-5-yl amine, 5- chloropyridin-2-yl amine, 5-methylpyridin-2-yl, 5-methoxypyridin-2-yl amine, 3-chloropyridin-4-yl; amine, 2-N-morpholinylpyridin-5-yl, and the like), and other commercially available or easily synthesized substituted or unsubstituted aryl and heteroaryl
  • Suitable cyano compounds include substituted benzonitriles (e.g., 2-chlorobenzonitrile, 2- fluorobenzonitrile, 2-methoxybenzonitrile, 2-methylbenzonitrile, 2,4- dichlorobenzonitrile, 2,4-difluorobenzonitrile, 2-chloro-4-fluorobenzonitrile, 2- chloro-4-methylbenzonitrile, 2,4-dimethoxybenzonitrile, 2-methyl-4- chlorobenzonitrile, and the like), cya no-substituted pyridines (e.g., 4-cyano- 3-chloropyridine) and other commercially available or easily synthesized substituted or unsubstituted aryl or heteroaryl nitriles.
  • substituted benzonitriles e.g., 2-chlorobenzonitrile, 2- fluorobenzonitrile, 2-methoxybenzonitrile, 2-methylbenzonitrile, 2,4- dichlorobenzonit
  • Intermediate 3(a) can then be condensed with a 3-bromo-2-oxo- propionic acid ester (wherein R is an alkyl group like methyl, ethyl, propyl, benzyl, etc.) to produce the cyclized 4-hydroxy-4,5-dihydro-1 H-imidazole ester 3(b) using procedures analogous to those described by Khanna, I.K., et al., in J. Med. Chem., 40, 1634 (1997).
  • the amidine intermediate 3(a) is refluxed in a polar solvent (e.g., isopropanol) in the presence of a mild base (e.g., sodium bicarbonate).
  • a polar solvent e.g., isopropanol
  • a mild base e.g., sodium bicarbonate
  • the reaction proceeds directly to the desired imidazole ester intermediate 3(c).
  • an acid catalyst e.g., p-toluenesulfonic acid
  • a non-polar solvent e.g., toluene
  • intermediate 3(b) may be treated with methanesulfonyl chloride in the presence of a base (e.g., triethylamine) to give 3(c).
  • Intermediate 3(d), where L 1 is a bromine or iodine can be synthesized from imidazole ester 3(c) using a halogenating agent such as bromine, N-bromosuccinimide, iodine, or N-iodosuccinimide in a suitable protic solvent (e.g., glacial acetic acid or trifluoroacetic acid) or an aprotic solvent (e.g, acetonitrile, ether or THF) at reaction temperatures ranging from 35 °C to 100 °C.
  • a suitable protic solvent e.g., glacial acetic acid or trifluoroacetic acid
  • an aprotic solvent e.g, acetonitrile, ether or THF
  • the halogenated imidizolyl derivative 3(d) can be treated with an ammonia equivalent such as lithium bis(trimethylsilyl)amide in the presence of a catalytic amount of Pd(dba) 2 and a phosphine ligand such as P(t-Bu) 3 in a non-polar solvent (e.g., toluene) at temperatures ranging from 23 °C to reflux to give intermediate 3(e).
  • an ammonia equivalent such as lithium bis(trimethylsilyl)amide
  • Pd(dba) 2 and a phosphine ligand such as P(t-Bu) 3 in a non-polar solvent (e.g., toluene) at temperatures ranging from 23 °C to reflux.
  • a non-polar solvent e.g., toluene
  • a coupling reagent e.g., EDC or O-(7- azabenzotriazol-1-yl)-N,N,N',N'-tetramethyluronium hexafluorophosphate (HATU)
  • a base e.g., triethylamine
  • a suitable aprotic solvent e.g, DMF,
  • intermediate 3(e) can be directly treated with an appropriately substituted amine derivative (e.g., R 4 NH 2 ) in the presence of trimethylaluminum in a non-polar solvent (e.g., toluene) to give intermediate 3(g).
  • an appropriately substituted amine derivative e.g., R 4 NH 2
  • trimethylaluminum e.g., toluene
  • Cyclocondensation of 3(g) with an ortho ester derivative (R 3 C(OR) 3 ) such as triethylorthoformate or triethylorthoacetate in the presence of an acid catalyst such as p- toluenesulfonic acid at temperatures ranging from about 50 °C to about 150 °C can provide compounds of Formula I or II.
  • the imidazolyl ester intermediate 3(e) can also be converted into compounds of formula (I) or (II) where R 3 is not limited to hydrogen and C C alkyl by the methods shown in Scheme IV.
  • 3(e) 5(a) (I) or (II) Scheme IV Intermediate 3(a) can be reacted with an appropriately substituted amidine derivative in the presence of an acid catalyst (e.g., acetic acid) in a polar solvent (e.g., ethanol, 2-ethoxyethanol) to give the 9H-purin-6-ol intermediate 5(a) which can be N-alkylated as previously described to give compounds of formula (I) or (II).
  • Scheme V illustrates another method for preparing compounds of formula (I) or (II) where R 2 is aryl or heteroaryl.
  • Such techniques will be well-known to one of ordinary skill in the art and may include, for example, all types of chromatography (high pressure liquid chromatography (HPLC), column chromatography using common adsorbents such as silica gel, and thin-layer chromatography), recrystallization, and differential (i.e., liquid-liquid) extraction techniques.
  • HPLC high pressure liquid chromatography
  • column chromatography using common adsorbents such as silica gel, and thin-layer chromatography
  • recrystallization i.e., liquid-liquid extraction techniques.
  • differential (i.e., liquid-liquid) extraction techniques i.e., liquid-liquid extraction techniques.
  • the compounds of the present invention may be isolated and used perse or in the form of its pharmaceutically acceptable salt, solvate and/or hydrate.
  • salts refers to inorganic and organic salts of a compound of the present invention.
  • salts can be prepared in situ during the final isolation and purification of a compound, or by separately reacting the compound, N-oxide, or prodrug with a suitable organic or inorganic acid and isolating the salt thus formed.
  • Representative salts include the hydrobromide, hydrochloride, hydroiodide, sulfate, bisulfate, nitrate, acetate, trifluoroacetate, oxalate, besylate, palmitiate, pamoate, malonate, stearate, laurate, malate, borate, benzoate, lactate, phosphate, hexafluorophosphate, benzene sulfonate, tosylate, formate, citrate, maleate, fumarate, succinate, tartrate, naphthylate, mesylate, glucoheptonate, lactobionate, and laurylsulphonate salts, and the like.
  • alkali and alkaline earth metals such as sodium, lithium, potassium, calcium, magnesium, and the like
  • non-toxic ammonium, quaternary ammonium, and amine cations including, but not limited to, ammonium, tetramethylammonium, tetraethylammonium, methylamine, dimethylamine, trimethylamine, triethylamine, ethylamine, and the like. See, e.g., Berge, et al., J. Pharm. Sci., 66, 1-19 (1977).
  • prodrug means a compound that is transformed in vivo to yield a compound of Formula (I) or a pharmaceutically acceptable salt, hydrate or solvate of the compound. The transformation may occur by various mechanisms, such as through hydrolysis in blood.
  • a discussion of the use of prodrugs is provided by T. Higuchi and W. Stella, "Pro-drugs as Novel Delivery Systems," Vol. 14 of the A.C.S. Symposium Series, and in Bioreversible Carriers in Drug Design, ed. Edward B. Roche, American Pharmaceutical Association and Pergamon Press, 1987.
  • a prodrug can comprise an ester formed by the replacement of the hydrogen atom of the acid group with a group such as (Ci-Csjalkyl, (C 2 -C ⁇ 2 )alkanoyloxymethyl, 1-(alkanoyloxy)ethyl having from 4 to 9 carbon atoms, 1-methyl-1-(alkanoyloxy)-ethyl having from 5 to 10 carbon atoms, alkoxycarbonyloxymethyl having from 3 to 6 carbon atoms, 1- (alkoxycarbonyloxy)ethyl having from 4 to 7 carbon atoms, 1-methyl-1- (alkoxycarbonyloxy)ethyl having from 5 to 8 carbon atoms, N- (alkoxycarbonyl)aminomethyl having from 3 to 9 carbon atoms, 1-(N-Csjalkyl, (C 2 -C ⁇ 2 )alkanoyloxymethyl, 1-(alkanoyloxy)ethyl having from 4 to 9 carbon atoms, 1-
  • alkoxycarbonyl)amino)ethyl having from 4 to 10 carbon atoms, 3-phthalidyl, 4-crotonolactonyl, gamma-butyrolacton-4-yl, di-N,N-(C ⁇ -C 2 )alkylamino(C 2 - C 3 )alkyl (such as ⁇ -dimethylaminoethyl), carbamoyl-(C ⁇ -C 2 )alkyl, N,N-di(C- ⁇ - C 2 )alkylcarbamoyl-(C ⁇ -C 2 )alkyl and piperidino-, pyrrolidino- or morpholino(C 2 -C 3 )alkyl.
  • a prodrug can be formed by the replacement of the hydrogen atom of the alcohol group with a group such as (C ⁇ - C 6 )alkanoyloxymethyl, 1-((C ⁇ -C 6 )alkanoyloxy)ethyl, 1-methyl-1-((C C 6 )alkanoyIoxy)ethyl, (C ⁇ -C 6 )alkoxycarbonyloxymethyl, N-(C ⁇ -C 6 )alkanoyloxymethyl, 1-((C ⁇ -C 6 )alkanoyloxy)ethyl, 1-methyl-1-((C C 6 )alkanoyIoxy)ethyl, (C ⁇ -C 6 )alkoxycarbonyloxymethyl, N-(C ⁇ -
  • each ⁇ -aminoacyl group is independently selected from the naturally occurring L-amino acids, P(O)(0H) 2 , P(O)(0(C ⁇ -C 6 )alkyl) 2 or glycosyl (the radical resulting from the removal of a hydroxyl group of the hemiacetal form of a carbohydrate).
  • a prodrug can be formed by the replacement of a hydrogen atom in the amine group with a group such as R-carbonyl, RO-carbonyl, NRR'-carbonyl where R and R' are each independently (C ⁇ -C 0 )alkyl, (C 3 - C 7 )cycloalkyl, benzyl, or R-carbonyl is a natural ⁇ -aminoacyl or natural ⁇ - aminoacyl-natural ⁇ -aminoacyl, -C(OH)C(0)OY' wherein Y' is H, (C ⁇ -C 6 )alkyl or benzyl, -C(OY 0 )Y ⁇ wherein Y 0 is (C C 4 ) alkyl and Y1 is (CrC 6 )alkyl, carboxy(C 1 -C 6 )alkyl, amino(C ⁇ -C 4 )alkyl or mono-N- or di
  • the compounds of the present invention may contain asymmetric or chiral centers, and, therefore, exist in different stereoisomeric forms. It is intended that all stereoisomeric forms of the compounds of the present invention as well as mixtures thereof, including racemic mixtures, form part of the present invention.
  • the present invention embraces all geometric and positional isomers. For example, if a compound of the present invention incorporates a double bond or a fused ring, both the cis- and trans- forms, as well as mixtures, are embraced within the scope of the invention. Diastereomeric mixtures can be separated into their individual diastereoisomers on the basis of their physical chemical differences by methods well known to those skilled in the art, such as by chromatography and/or fractional crystallization.
  • Enantiomers can be separated by converting the enantiomeric mixture into a diastereomeric mixture by reaction with an appropriate optically active compound (e.g., chiral auxiliary such as a chiral alcohol or Mosher's acid chloride), separating the diastereoisomers and converting (e.g., hydrolyzing) the individual diastereoisomers to the corresponding pure enantiomers.
  • an appropriate optically active compound e.g., chiral auxiliary such as a chiral alcohol or Mosher's acid chloride
  • converting e.g., hydrolyzing
  • some of the compounds of the present invention may be atropisomers (e.g., substituted biaryls) and are considered as part of this invention.
  • Enantiomers can also be separated by use of a chiral HPLC column.
  • the compounds of the present invention may exist in unsolvated as well as solvated forms with pharmaceutically acceptable solvents such as water, ethanol, and the like, and it is intended that the invention embrace both solvated and unsolvated forms. It is also possible that the compounds of the present invention may exist in different tautomeric forms, and all such forms are embraced within the scope of the invention.
  • tautomer or "tautomeric form” refers to structural isomers of different energies which are interconvertible via a low energy barrier.
  • proton tautomers also known as prototropic tautomers
  • Valence tautomers include interconversions by reorganization of some of the bonding electrons.
  • the present invention also embraces isotopically-labeled compounds of the present invention which are identical to those recited herein, but for the fact that one or more atoms are replaced by an atom having an atomic mass or mass number different from the atomic mass or mass number usually found in nature.
  • isotopes that can be incorporated into compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, fluorine, iodine, and chlorine, such as 2 H, 3 H, 13 C, 14 C, 15 N, 18 0, 17 0, 31 P, 32 P, 35 S, 18 F, 123 l, and 36 CI, respectively.
  • Certain isotopically-labeled compounds of the present invention are useful in compound and/or substrate tissue distribution assays. Tritiated (i.e., 3 H) and carbon-14 (i.e., 14 C) isotopes are particularly preferred for their ease of preparation and detectability. Further, substitution with heavier isotopes such as deuterium (i.e., 2 H) may afford certain therapeutic advantages resulting from greater metabolic stability (e.g., increased in vivo half-life or reduced dosage requirements) and hence may be preferred in some circumstances.
  • Isotopically labeled compounds of the present invention can generally be prepared by following procedures analogous to those disclosed in the Schemes and/or in the Examples hereinbelow, by substituting an isotopically labeled reagent for a non-isotopically labeled reagent.
  • Compounds of the present invention are useful for treating diseases, conditions and/or disorders modulated by cannabinoid receptor antagonists; therefore, another embodiment of the present invention is a pharmaceutical composition comprising a therapeutically effective amount of a compound of the present invention and a pharmaceutically acceptable excipient, diluent or carrier.
  • a typical formulation is prepared by mixing a compound of the present invention and a carrier, diluent or excipient.
  • Suitable carriers, diluents and excipients are well known to those skilled in the art and include materials such as carbohydrates, waxes, water soluble and/or swellable polymers, hydrophilic or hydrophobic materials, gelatin, oils, solvents, water, and the like.
  • the particular carrier, diluent or excipient used will depend upon the means and purpose for which the compound of the present invention is being applied.
  • Solvents are generally selected based on solvents recognized by persons skilled in the art as safe (GRAS) to be administered to a mammal.
  • safe solvents are non-toxic aqueous solvents such as water and other non-toxic solvents that are soluble or miscible in water.
  • Suitable aqueous solvents include water, ethanol, propylene glycol, polyethylene glycols (e.g., PEG400, PEG300), etc. and mixtures thereof.
  • the formulations may also include one or more buffers, stabilizing agents, surfactants, wetting agents, lubricating agents, emulsifiers, suspending agents, preservatives, antioxidants, opaquing agents, glidants, processing aids, colorants, sweeteners, perfuming agents, flavoring agents and other known additives to provide an elegant presentation of the drug (i.e., a compound of the present invention or pharmaceutical composition thereof) or aid in the manufacturing of the pharmaceutical product (i.e., medicament).
  • the formulations may be prepared using conventional dissolution and mixing procedures.
  • the bulk drug substance i.e., compound of the present invention or stabilized form of the compound (e.g., complex with a cyclodextrin derivative or other known complexation agent)
  • a suitable solvent in the presence of one or more of the excipients described above.
  • the compound of the present invention is typically formulated into pharmaceutical dosage forms to provide an easily controllable dosage of the drug and to give the patient an elegant and easily handleable product.
  • the pharmaceutical composition (or formulation) for application may be packaged in a variety of ways depending upon the method used for administering the drug.
  • an article for distribution includes a container having deposited therein the pharmaceutical formulation in an appropriate form.
  • Suitable containers are well-known to those skilled in the art and include materials such as bottles (plastic and glass), sachets, ampoules, plastic bags, metal cylinders, and the like.
  • the container may also include a tamper-proof assemblage to prevent indiscreet access to the contents of the package.
  • the container has deposited thereon a label that describes the contents of the container. The label may also include appropriate warnings.
  • the present invention further provides a method of treating diseases, conditions and/or disorders modulated by cannabinoid receptor antagonists in an animal that includes administering to an animal in need of such treatment a therapeutically effective amount of a compound of the present invention or a pharmaceutical composition comprising an effective amount of a compound of the present invention and a pharmaceutically acceptable excipient, diluent, or carrier.
  • the method is particularly useful for treating diseases, conditions and/or disorders modulated by cannabinoid receptor (in particular, CB1 receptor) antagonists.
  • eating disorders e.g., binge eating disorder, anorexia, and bulimia
  • weight loss or control e.g., reduction in calorie or food intake, and/or appetite suppression
  • obesity depression, atypical depression, bipolar disorders, psychoses, schizophrenia, behavioral addictions, suppression of reward-related behaviors (e.g., conditioned place avoidance, such as suppression of cocaine- and morphine-induced conditioned place preference)
  • substance abuse e.g., alcohol abuse, addiction and/or dependence including treatment for abstinence, craving reduction and relapse prevention of alcohol intake
  • tobacco abuse e.g., smoking addiction, cessation and/or dependence including treatment for craving reduction and relapse prevention of tobacco smoking
  • dementia including memory loss, Alzheimer's disease, dementia of aging, vascular dementia, mild cognitive impairment, age-related cognitive decline, and mild neurocognitive disorder
  • sexual dysfunction in
  • the compounds of the present invention described herein are useful in treating diseases, conditions, or disorders that are modulated by cannabinoid receptor antagonists. Consequently, the compounds of the present invention (including the compositions and processes used therein) may be used in the manufacture of a medicament for the therapeutic applications described herein.
  • diseases, conditions and/or disorders for which cannabinoid receptor antagonists may be effective include: premenstrual syndrome or late luteal phase syndrome, migraines, panic disorder, anxiety, post- traumatic syndrome, social phobia, cognitive impairment in non-demented individuals, non-amnestic mild cognitive impairment, post operative cognitive decline, disorders associated with impulsive behaviours (such as, disruptive behaviour disorders (e.g., anxiety/depression, executive function improvement, tic disorders, conduct disorder and/or oppositional defiant disorder), adult personality disorders (e.g.
  • borderline personality disorder and antisocial personality disorder diseases associated with impulsive behaviours (e.g., substance abuse, paraphilias and self-mutilation), and impulse control disorders (e.g., intermittene explosive disorder, kleptomania, pyromania, pathological gambling, and trichotillomania)), obsessive compulsive disorder, chronic fatigue synd rome, sexual dysfunction in males (e.g., premature ejaculation), sexual dysfunction in females, disorders of sleep (e.g., sleep apnea), autism, mutism , neurodengenerative movement disorders, spinal cord injury, damage of the central nervous system (e.g., trauma), stroke, neurodegenerative diseases or toxic or infective CNS diseases (e.g., encephalitis or meningitis), cardiovascular disorders (e.g., thrombosis), and diabetes.
  • impulsive behaviours e.g., substance abuse, paraphilias and self-mutilation
  • impulse control disorders e.g.
  • the compounds of the present invention can be administered to a patient at dosage levels in the range of from about 0.7 mg to about 7,000 mg per day.
  • dosage levels in the range of from about 0.7 mg to about 7,000 mg per day.
  • a dosage in the range of from about 0.01 mg to about 100 mg per kilogram body weight is typically sufficient.
  • some variability in the general dosage range may be required depending upon the age and weight of the subject being treated, the intended route of administration, the particular compound being administered and the like.
  • the determination of dosage ranges and optimal dosages for a particular patient is well within the ability of one of ordinary skill in the art having the benefit of the instant disclosure.
  • the compounds of the present invention can be used in sustained release, controlled release, and delayed release formulations, which forms are also well known to one of ordinary skill in the art.
  • Suitable pharmaceutical agents include anti-obesity agents such as apolipoprotein-B secretion/microsomal triglyceride transfer protein (apo-B/MTP) inhibitors, 1 1 ⁇ -hydroxy steroid dehydrogenase-1 (1 1 ⁇ -HSD type 1 ) inhibitors, peptide YY 3 - 36 or analogs thereof, MCR-4 agonists, cholecystokinin-A (CCK-A) agonists, monoamine reuptake inhibitors (such as sibutramine), sympathomimetic agents, ⁇ 3 adrenergic receptor agonists, dopamine agonists (such as bromocriptine), melanocyte-stimulating hormone receptor analogs, 5HT
  • anti-obesity agents such as apolipoprotein-B secretion/microsomal triglyceride transfer protein (apo-B/MTP) inhibitors, 1 1 ⁇ -hydroxy steroid dehydrogenase-1 (1 1 ⁇ -HS
  • anorectic agents such as a bombesin agonist
  • Neuropeptide-Y receptor antagonists e.g., NPY Y5 receptor antagonists, such as the spiro compounds described in US Patent Nos. 6,566,367; 6,649,624; 6,638,942; 6,605,720; 6,495,559; 6,462,053; 6,388,077; 6,335,345; and 6,326,375; US Publication Nos. 2002/0151456 and 2003/036652; and PCT Publication Nos. WO 03/010175.
  • thyromimetic agents dehydroepiandrosterone or an analog thereof, glucocorticoid receptor agonists or antagonists, orexin receptor antagonists, glucagon-like peptide-1 receptor agonists, ciliary neurotrophic factors (such as AxokineTM available from Regeneron Pharmaceuticals, Inc., Tarrytown, NY and Procter & Gamble Company, Cincinnati, OH), human agouti-related proteins (AGRP), ghrelin receptor antagonists, histamine 3 receptor antagonists or inverse agonists, neuromedin U receptor agonists and the like.
  • AxokineTM available from Regeneron Pharmaceuticals, Inc., Tarrytown, NY and Procter & Gamble Company, Cincinnati, OH
  • human agouti-related proteins AGRP
  • ghrelin receptor antagonists histamine 3 receptor antagonists or inverse agonists
  • neuromedin U receptor agonists and the like thyromimetic agents, dehydroepiandrosterone
  • anti-obesity agents including the preferred agents set forth hereinbelow, are well known, or will be readily apparent in light of the instant disclosure, to one of ordinary skill in the art.
  • anti-obesity agents selected from the group consisting of orlistat, sibutramine, bromocriptine, ephedrine, leptin, pseudoephedrine, PYY 3 - 36 or an analog thereof, and 2-oxo-N-(5- phenylpyrazinyl)spiro-[isobenzofuran-1(3H),4'-piperidine]-1 '-carboxamide.
  • compounds of the present invention and combination therapies are administered in conjunction with exercise and a sensible diet.
  • anti-obesity agents for use in the combinations, pharmaceutical compositions, and methods of the invention can be prepared using methods known to one of ordinary skill in the art, for example, sibutramine can be prepared as described in U.S. Pat. No. 4,929,629; bromocriptine can be prepared as described in U.S. Pat. Nos. 3,752,814 and 3,752,888; orlistat can be prepared as described in U.S. Pat. Nos.
  • PYY3-3 6 (including analogs thereof) can be prepared as described in US Publication No. 2002/0141985 and WO 03/027637; and the NPY Y5 receptor antagonist 2-oxo-N-(5- phenylpyrazinyl)spiro[isobenzofuran-1 (3H),4'-piperidine]-1 '-carboxamide can be prepared as described in US Publication No. 2002/0151456.
  • Other useful NPY Y5 receptor antagonists include those described in PCT Publication No.
  • 03/082190 such as 3-oxo-N-(5-phenyl-2-pyrazinyl)-spiro[isobenzofuran- 1 (3H), 4'-piperidine]-1 '-carboxamide; 3-oxo-N-(7-trifluoromethylpyrido[3,2- b]pyridin-2-yl)-spiro-[isobenzofuran-1 (3H), 4'-piperidine]-1 '-carboxamide; N- [5-(3-fluorophenyl)-2-pyrimidinyl]-3-oxospiro-[isobenzofuran-1 (3H), [4'- piperidine]-1 '-carboxamide; fra ⁇ s-3'-oxo-N-(5-phenyl-2-pyrimidinyl)] spiro[cyclohexane-1 ,1 '(3 ⁇ )-isobenzofuran]-4-carboxamide; ⁇ frar?s-3'-
  • agents for reducing alcohol withdrawal symptoms may also be co-administered, such as benzodiazepines, beta-
  • Neuroprotective agents e.g., memantine
  • antipsychotic medications e.g., ziprasidone (GeodonTM), risperidone
  • sulfonylureas and analogs thereof chlorpropamide, glibenclamide, tolbutamide, tolazamide, acetohexamide, Glypizide ® , glimepiride, repaglinide, meglitinide; biguanides: metformin, phenformin, buformin; ⁇ 2- antagonists and imidazolines: midaglizole, isaglidole, deriglidole, idazoxan, efaroxan, fluparoxan; other insulin secretagogues: linogliride, A-4166; glitazones: ciglitazone, Actos ® (pioglitazone), englit
  • the compounds of the present invention may also be administered in combination with a naturally occurring compound that acts to lower plasma cholesterol levels.
  • a naturally occurring compound that acts to lower plasma cholesterol levels.
  • Such naturally occurring compounds are commonly called nutraceuticals and include, for example, garlic extract, Hoodia plant extracts, and niacin.
  • the dosage of the additional pharmaceutical agent will also be generally dependent upon a number of factors including the health of the subject being treated, the extent of treatment desired, the nature and kind of concurrent therapy, if any, and the frequency of treatment and the nature of the effect desired.
  • the dosage range of an anti-obesity agent is in the range of from about O.001 mg to about 100 mg per kilogram body weight of the individual per day, preferably from about 0.1 mg to about 10 mg per kilogram body weight of the individual per day.
  • a compound of the present invention or a combination of a compound of the present invention and at least one additional pharmaceutical agent is administered to a subject in need of such treatment, preferably in the form of a pharmaceutical composition.
  • the compound of the present invention and at least one other pharmaceutical agent may be administered either separately or in the pharmaceutical composition comprising both. It is generally preferred that such administration be oral.
  • parenteral or transdermal administration may be appropriate.
  • a combination of a compound of the present invention and at least one other pharmaceutical agent when administered together, such administration can be sequential in time or simultaneous with the simultaneous method being generally preferred.
  • a compound of the present invention and the additional pharmaceutical agent can be administered in any order. It is generally preferred that such administration be oral. It is especially preferred that such administration be oral and simultaneous.
  • the administration of each can be by the same or by different methods.
  • a compound of the present invention or a combination of a compound of the present invention and at least one additional pharmaceutical agent referred to herein as a
  • compositions suitable for parenteral injection generally include pharmaceutically acceptable sterile aqueous or nonaqueous solutions, dispersions, suspensions, or emulsions, and sterile powders for reconstitution into sterile injectable solutions or dispersions.
  • aqueous and nonaqueous carriers examples include water, ethanol, polyols (propylene glycol, polyethylene glycol, glycerol, and the like), suitable mixtures thereof, vegetable oils (such as olive oil) and injectable organic esters such as ethyl oleate.
  • a coating such as lecithin
  • surfactants such as surfactants.
  • These compositions may also contain adjuvants such as preserving, wetting, emulsifying, and dispersing agents.
  • compositions can be accomplished with various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, and the like. It may also be desirable to include isotonic agents, for example, sugars, sodium chloride, and the like. Prolonged absorption of injectable pharmaceutical compositions can be brought about by the use of agents capable of delaying absorption, for example, aluminum monostearate and gelatin. Solid dosage forms for oral administration include capsules, tablets, powders, and granules.
  • a compound of the present invention or a combination is admixed with at least one inert customary pharmaceutical excipient (or carrier) such as sodium citrate or dicalcium phosphate or
  • fillers or extenders e.g., starches, lactose, sucrose, mannitol, silicic acid and the like
  • binders e.g., carboxymethylcellulose, alginates, gelatin, polyvinylpyrrolidone, sucrose, acacia and the like
  • humectants e.g., glycerol and the like
  • disintegrating agents e.g., agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain complex silicates, sodium carbonate and the like
  • solution retarders e.g., paraffin and the like
  • absorption accelerators e.g., quaternary ammonium compounds and the like
  • the dosage forms may also comprise buffering agents.
  • Solid compositions of a similar type may also be used as fillers in soft or hard filled gelatin capsules using such excipients as lactose or milk sugar, as well as high molecular weight polyethylene glycols, and the like.
  • Solid dosage forms such as tablets, dragees, capsules, and granules can be prepared with coatings and shells, such as enteric coatings and others well known in the art. They may also contain opacifying agents, and can also be of such composition that they release the compound of the present invention and/or the additional pharmaceutical agent in a delayed manner. Examples of embedding compositions that can be used are polymeric substances and waxes.
  • the drug can also be in micro- encapsulated form, if appropriate, with one or more of the above-mentioned excipients.
  • Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, solutions, suspensions, syrups, and elixirs.
  • the liquid dosage form may contain inert diluents commonly used in the art, such as water or other solvents, solubilizing agents and emulsifiers, as for example, ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1 ,3-butylene glycol, dimethylformamide, oils (e.g., cottonseed oil, groundnut oil, corn germ oil, olive oil, castor oil, sesame seed oil and the like), glycerol, tetrahydrofurfuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, or mixtures of these substances, and the like.
  • inert diluents commonly used in the art, such as water or other solvents, solubilizing agents and emulsifiers, as for example, ethyl alcohol, isopropyl alcohol, eth
  • the composition can also include adjuvants, such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents.
  • adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents.
  • Suspensions in addition to the compound of the present invention or the combination, may further comprise suspending agents, e.g., ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar, and tragacanth, or mixtures of these substances, and the like.
  • compositions for rectal or vaginal administration preferably comprise suppositories, which can be prepared by mixing a compound of the present invention or a combination with suitable non-irritating excipients or carriers, such as cocoa butter, polyethylene glycol or a suppository wax which are solid at ordinary room temperature but liquid at body temperature and therefore melt in the rectum or vaginal cavity thereby releasing the active component(s).
  • suitable non-irritating excipients or carriers such as cocoa butter, polyethylene glycol or a suppository wax which are solid at ordinary room temperature but liquid at body temperature and therefore melt in the rectum or vaginal cavity thereby releasing the active component(s).
  • Dosage forms for topical administration of the compounds of the present invention and combinations of the compounds of the present invention with anti-obesity agents may comprise ointments, powders, sprays and inhalants.
  • the drugs are admixed under sterile condition with a pharmaceutically acceptable carrier, and any preservatives, buffers, or propellants that may be required.
  • Ophthalmic formulations, eye ointments, powders, and solutions are also intended to be included within the scope of the present invention. The following paragraphs describe exemplary formulations, dosages, etc. useful for non-human animals.
  • the administration of the compounds of the present invention and combinations of the compounds of the present invention with anti-obesity agents can be effected orally or non-orally (e.g., by injection). An amount of a compound of the present invention or combination of a compound of the present invention with an anti-obesity agent is administered such that an effective dose is received.
  • a daily dose that is administered orally to an animal is between about 0.01 and about 1 ,000 mg/kg of body weight, preferably between about 0.01 and about 300 mg/kg of body weight.
  • a compound of the present invention (or combination) can be carried in the drinking water so that a therapeutic dosage of the compound is ingested with the daily water supply.
  • the compound can be directly metered into drinking water, preferably in the form of a liquid, water- soluble concentrate (such as an aqueous solution of a water-soluble salt).
  • a compound of the present invention (or combination) can also be added directly to the feed, as such, or in the form of an animal feed supplement, also referred to as a premix or concentrate.
  • a premix or concentrate of the compound in a carrier is more commonly employed for the inclusion of the agent in the feed.
  • Suitable carriers are liquid or solid, as desired, such as water, various meals such as alfalfa meal, soybean meal, cottonseed oil meal, linseed oil meal, corncob meal and corn meal, molasses, urea, bone meal, and mineral mixes such as are commonly employed in poultry feeds.
  • a particularly effective carrier is the respective animal feed itself; that is, a small portion of such feed.
  • the carrier facilitates uniform distribution of the compound in the finished feed with which the premix is blended.
  • the compound is thoroughly blended into the premix and, subsequently, the feed.
  • the compound may be dispersed or dissolved in a suitable oily vehicle such as soybean oil, corn oil, cottonseed oil, and the like, or in a volatile organic solvent and then blended with the carrier.
  • a suitable oily vehicle such as soybean oil, corn oil, cottonseed oil, and the like
  • the proportions of compound in the concentrate are capable of wide variation since the amount of the compound in the finished feed may be adjusted by blending the appropriate proportion of premix with the feed to obtain a desired level of compound.
  • High potency concentrates may be blended by the feed manufacturer with proteinaceous carrier such as soybean oil meal and other meals, as described above, to produce concentrated supplements, which are suitable for direct feeding to animals. In such instances, the animals are permitted to consume the usual diet.
  • such concentrated supplements may be added directly to the feed to produce a nutritionally balanced, finished feed containing a therapeutically effective level of a compound of the present invention.
  • the mixtures are thoroughly blended by standard procedures, such as in a twin shell blender, to ensure homogeneity. If the supplement is used as a top dressing for the feed, it likewise helps to ensure uniformity of distribution of the compound across the top of the dressed feed.
  • Drinking water and feed effective for increasing lean meat deposition and for improving lean meat to fat ratio are generally prepared by mixing a compound of the present invention with a sufficient amount of animal feed to provide from about 10 "3 to about 500 ppm of the compound in the feed or water.
  • the preferred medicated swine, cattle, sheep and goat feed generally contain from about 1 to about 400 grams of a compound of the present invention (or combination) per ton of feed, the optimum amount for these animals usually being about 50 to about 3O0 grams per ton of feed.
  • the preferred poultry and domestic pet feeds usually contain about 1 to about 400 grams and preferably about 1 0 to about 400 grams of a compound of the present invention (or combination) per ton of feed.
  • the compounds of the present invention (or combination) may be prepared in the form of a paste or a pellet and administered as an implant, usually under the skin of the head or ear of the animal in which increase in lean meat deposition and improvement in lean meat to fat ratio is sought.
  • parenteral administration involves injection of a sufficient amount of a compound of the present invention (or combination) to provide the animal with about 0.01 to about 20 mg/kg/day of body weight of the drug.
  • the preferred dosage for poultry, swine, cattle, sheep, goats and domestic pets is in the range of from about 0.05 to about 10 mg/kg/day of body weight of drug.
  • Paste formulations can be prepared by dispersing the drug in a pharmaceutically acceptable oil such as peanut oil, sesame oil, corn oil or the like.
  • Pellets containing an effective amount of a compound of the present invention, pharmaceutical composition, or combination can be prepared by admixing a compound of the present invention or combination with a diluent such as carbowax, carnuba wax, and the like, and a lubricant, such as magnesium or calcium stearate, can be added to improve the pelleting process.
  • a diluent such as carbowax, carnuba wax, and the like
  • a lubricant such as magnesium or calcium stearate
  • implants may also be made periodically during the animal treatment period in order to maintain the proper drug level in the animal's body.
  • the present invention has several advantageous veterinary features.
  • the instant invention provides the means by which this may be accomplished.
  • utilization of the method of the present invention yields leaner animals that command higher sale prices from the meat industry.
  • Embodiments of the present invention are illustrated by the following Examples. It is to be understood, however, that the embodiments of the invention are not limited to the specific details of these Examples, as other variations thereof will be known, or apparent in light of the instant disclosure, to one of ordinary skill in the art. EXAMPLES Unless specified otherwise, starting materials are generally available from commercial sources such as Aldrich Chemicals Co. (Milwaukee, Wl), Lancaster Synthesis, Inc.
  • Atmospheric pressure chemical ionization mass spectra were obtained on a FisonsTM Platform II Spectrometer (carrier gas: acetonitrile: available from Micromass Ltd, Manchester, UK).
  • Chemical ionization mass spectra were obtained on a Hewlett-PackardTM 5989 instrument (ammonia ionization, PBMS: available from Hewlett-Packard Company, Palo Alto, CA).
  • Electrospray ionization mass spectra were obtained on a WatersTM ZMD instrument (carrier gas: acetonitrile: available from Waters Corp., Milford, MA). Where the intensity of chlorine or bromine-containing ions are described, the expected intensity ratio was observed (approximately 3:1 for 35 CI/ 37 CI-containing ions and 1:1 for 79 Br/ 81 Br-containing ions) and the intensity of only the lower mass ion is given.
  • the reaction mixture was cooled to room temperature and concentrated in vacuo.
  • the crude residue was taken up in CH 2 CI 2 and the organic solution was washed with H 2 0, sat'd aq. NaHC0 , and saturated aqueous NaCl, dried over MgS0 4 and concentrated in vacuo.
  • the crude residue was purified by silica gel plug filtration using a solvent gradient of 2% EtOAc/CH 2 CI 2 to 10% EtOAc/CH 2 CI 2 .
  • the fractions containing the product were concentrated, and the oily residue was diluted with 1 :3 EtOAc/hexanes (200 ml).
  • a protein assay was performed and 200 ⁇ l of tissue totaling 20 ⁇ g was added to the assay.
  • the test compounds were diluted in drug buffer (0.5% BSA, 10% DMSO and TME) and then 25 ⁇ l were added to a deep well polypropylene plate.
  • [ 3 H]-SR141716A was diluted in a ligand buffer (0.5% BSA plus TME) and 25 ⁇ l were added to the plate.
  • a BCA protein assay was used to determine the appropriate tissue concentration and then 200 ⁇ l of rat brain tissue at the appropriate concentration was added to the plate.
  • the plates were covered and placed in an incubator at 20°C for 60 minutes. At the end of the incubation period 250 ⁇ l of stop buffer (5% BSA plus TME) was added to the reaction plate.
  • a protein assay was performed and 200 ⁇ l of tissue totaling 20 ⁇ g was added to the assay.
  • the test compounds were diluted in drug buffer (0.5% BSA, 10% DMSO and TME) and then 25 ⁇ l were added to a deep well polypropylene plate.
  • [ 3 H]-SR141716A was diluted in a ligand buffer (0.5% BSA plus TME) and 25 ⁇ l were added to the plate. The plates were covered and placed in an incubator at 30°C for 60 minutes.
  • test compounds were diluted in drug buffer (0.5% BSA, 10% DMSO, and 80.5% TME) and then 25 ⁇ l were added to the deep well polypropylene plate.
  • [ 3 H]-5-(1 ,1-Dimethyl-heptyl)-2-[5-hydroxy-2-(3-hydroxy- propyl)-cyclohexyl]-phenol was diluted in ligand buffer (0.5% BSA and 99.5% TME) and then 25 ⁇ l of the solution were added to each well at a concentration of 1 nM.
  • a BCA protein assay was used to determine the appropriate tissue concentration and 200 ⁇ l of the tissue at the appropriate concentration was added to the plate.
  • the plates were covered and placed in an incubator at 30°C for 60 minutes.
  • GTP ⁇ [ 35 S] binding assays were performed in a 96 well FlashPlate TM format in duplicate using 100 pM GTP ⁇ [ 35 S] and 10 ⁇ g membrane per well in assay buffer composed of 50 mM Tris HCI, pH 7.4, 3 mM MgCI 2 , pH 7.4, 10 mM MgCI 2 , 20 mM EGTA, 100 mM NaCl, 30 ⁇ M GDP, 0.1 % bovine serum albumin and the following protease inhibitors: 100 ⁇ g/ml bacitracin, 100 ⁇ g/ml benzamidine, 5 ⁇ g/ml aprotinin, 5 ⁇ g/ml leupeptin.
  • the assay mix was then incubated with increasing concentrations of antagonist (10 "1 ° M to 10 "5 M) for 10 minutes and challenged with the cannabinoid agonist 5-(1 ,1- dimethyl-heptyl)-2-[5-hydroxy-2-(3-hydroxy-propyl)-cyclohexyl]-phenol (10 ⁇ M). Assays were performed at 30°C for one hour. The FlashPlatesTM were then centrifuged at 2000Xg for 10 minutes. Stimulation of GTP ⁇ [ 35 S] binding was then quantified using a Wallac Microbeta. EC 50 calculations were performed using PrismTM by Graphpad. Inverse agonism was measured in the absense of agonist using the protocol described above.
  • CB-1 Receptor FLIPR-based Functional Assay Protocol CHO-K1 cells co-transfected with the human CB-1 receptor cDNA (obtained from Dr. Debra Kendall, University of Connecticut) and the promiscuous G-protein G16 were used for this assay. Cells were plated 48 hours in advance at 12500 cells per well on collagen coated 384 well black clear assay plates. Cells were incubated for one hour with 4 ⁇ M Fluo-4 AM (Molecular Probes) in DMEM (Gibco) containing 2.5 mM probenicid and pluronic acid (.04%). The plates were then washed 3 times with HEPES- buffered saline (containing probenicid; 2.5 mM) to remove excess dye.
  • HEPES- buffered saline containing probenicid; 2.5 mM
  • the plates were incubated for 24 hours in a 37°C incubator. The media was removed and media lacking serum (100 ⁇ l) was added. The plates were then incubated for 18 hours at 37°C. Serum free medium containing 1 mM IBMX was added to each well followed by 10 ⁇ l of test compound (1 :10 stock solution (25 mM compound in DMSO) into 50% DMSO/PBS) diluted 10X in PBS with 0.1 % BSA. After incubating for 20 minutes at 37°C, 2 ⁇ M of Forskolin was added and then incubated for an additional 20 minutes at 37°C. The media was removed, 100 ⁇ l of 0.01 N HCI was added and then incubated for 20 minutes at room temperature.
  • Cannabinoid agoinists such as ⁇ 9 -tetrahydrocannabinol ( ⁇ 9 -THC) and 5-(1 ,1-dimethyl-heptyl)-2-[5-hydroxy-2-(3-hydroxy-propyl)-cyclohexyl]-phenol have been shown to affect four characteristic behaviors in mice, collectively known as the Tetrad.
  • Tetrad mice
  • mice Fifteen minutes later, the mice were challenged with 5-(1 ,1-dimethyl-heptyl)- 2-[5-hydroxy-2-(3-hydroxy-propyl)-cyclohexyl]-phenol (sc administration). Twenty-five minutes after the agonist injection, the mice were placed in clear acrylic cages (431.8 cm x 20.9 cm x 20.3 cm) containing clean wood shavings. The subjects were allowed to explore surroundings for a total of about 5 minutes and the activity was recorded by infrared motion detectors (available from Coulbourn InstrumentsTM, Allentown, PA) that were placed on top of the cages.
  • infrared motion detectors available from Coulbourn InstrumentsTM, Allentown, PA
  • test compound sc, po, ip or icv
  • mice were challenged with 5-(1 ,1-dimethyl-heptyl)-2-[5-hydroxy-2-(3-hydroxy-propyl)- cydohexyl]-phenol (sc).
  • mice were placed on a 6.5
  • test compounds sc, po, ip or iv
  • each mouse was tested for reversal of analgesia using a standard hot plate meter (Columbus Instruments).
  • the hot plate was 10" x 10" x 0.75" with a surrounding clear acrylic wall. Latency to kick, lick or flick hindpaw or jump from the platform was recorded to the nearest tenth of a second. The timer was experimenter activated and each test had a 40 second cut off. Data were presented as a percent reversal of the agonist induced analgesia.
  • Food Intake The following screen was used to evaluate the efficacy of test compounds for inhibiting food intake in Sprague-Dawley rats after an overnight fast. Male Sprague-Dawley rats were obtained from Charles River Laboratories, Inc.
  • the rats were individually housed and fed powdered chow. They were maintained on a 12 hour light/dark cycle and received food and water ad libitum. The animals were acclimated to the vivarium for a period of one week before testing was conducted. Testing was completed during the light portion of the cycle. To conduct the food intake efficacy screen, rats were transferred to individual test cages without food the afternoon prior to testing, and the rats were fasted overnight. After the overnight fast, rats were dosed the following morning with vehicle or test compounds. A known antagonist was dosed (3 mg/kg) as a positive control, and a control group received vehicle alone (no compound). The test compounds were dosed at ranges between 0.1 and 100 mg/kg depending upon the compound.
  • the standard vehicle was 0.5% (w/v) methylcellulose in water and the standard route of administration was oral. However, different vehicles and routes of administration were used to accommodate various compounds when required.
  • Food was provided to the rats 30 minutes after dosing and the Oxymax automated food intake system (Columbus Instruments, Columbus, Ohio) was started. Individual rat food intake was recorded continuously at 10-minute intervals for a period of two hours. When required, food intake was recorded manually using an electronic scale; food was weighed every 30 minutes after food was provided up to four hours after food was provided. Compound efficacy was determined by comparing the food intake pattern of compound-treated rats to vehicle and the standard positive control.
  • Test compounds were generally mixed into a vehicle of 30% (w/v) ⁇ -cyclodextrin in distilled water at a volume of 1-2 ml/kg. Vehicle injections were given to all groups for the first two days of the experiment. This was followed by 2 days of drug injections (to the appropriate groups) and a final day of vehicle injections. On the drug injection days, drugs were given sc 30 minutes prior to a 2-hour alcohol access period.
  • mice (17-19 g) were individually housed upon arrival and given unlimited access to powdered rat chow, water and a 10 % (w/v) alcohol solution. After 2-3 weeks of unlimited access, water was restricted for 2O hours and alcohol was restricted to only 2 hours access daily. This was done in a manner that the access period was the last 2 hours of the dark part of the light cycle. Once drinking behavior stabilized, testing commenced. Mice were considered stable when the average alcohol consumption for 3 days was ⁇ 20% of the average for all 3 days. Day 1 of test consisted of all mice receiving vehicle injection (sc or ip). Thirty to 120 minutes post injection, access was given to alcohol and water.
  • vehicle injection sc or ip
  • mice were injected with vehicle or drug and the same protocol as the previous day was followed. Day 4 was wash out and no injections were given. Data was analyzed using repeated measures ANOVA. Change in water or alcohol consumption was compared back to vehicle for each day of the test. Positive results would be interpreted as a compound that was able to significantly reduce alcohol consumption while having no effect on water Oxygen Consumption Methods: Whole body oxygen consumption is measured using an indirect calorimeter (Oxymax from Columbus Instruments, Columbus, OH) in male Sprague Dawley rats.
  • Rats (300-380 g body weight) are placed in the calorimeter chambers and the chambers are placed in activity monitors. These studies are done during the light cycle. Prior to the measurement of oxygen consumption, the rats are fed standard chow ad libitum. During the measurement of oxygen consumption, food is not available. Basal pre-dose oxygen consumption and ambulatory activity are measured every 10 minutes for 2.5 to 3 hours. At the end of the basal pre-dosing period, the chambers are opened and the animals are administered a single dose of compound (the usual dose range is 0.001 to 10 mg/kg) by oral gavage (or other route of administration as specified, i.e. s.c, i.p., i.v.).
  • Drugs are prepared in methylcellulose, water or other specified vehicle (examples include PEG40O, 30% beta-cyclo dextran and propylene glycol).
  • Oxygen consumption and ambulatory activity are measured every 10 minutes for an additional 1-6 hours post-dosing.
  • the Oxymax calorimeter software calculates the oxygen consumption (ml/kg/h) based on the flow rate of air through the chambers and difference in oxygen content at inlet and output ports.
  • the activity monitors have 15 infrared light beams spaced one inch apart on each axis, ambulatory activity is recorded when two consecutive beams are broken and the results are recorded as counts.

Abstract

Compounds of Formula (I) are described herein. The compounds have been shown to act as cannabinoid receptor ligands and are therefore useful in the treatment of diseases linked to the mediation of the cannabinoid receptors in animals.

Description

BICYCLIC IMIDAZOLYL PYRIMDIN-4-ONE AS CANNABINOID RECEPTOR LIGANDS AND USES THEREOF
FIELD OF THE INVENTION The present invention relates to bicyclic imidazolyl pyrimidinone (e.g., 1 ,9-dihydro-purin-6-one) compounds as cannabinoid receptor ligands, in particular CB1 receptor antagonists, and uses thereof for treating diseases, conditions and/or disorders modulated by cannabinoid receptor antagonists. BACKGROUND Obesity is a major public health concern because of its increasing prevalence and associated health risks. Obesity and overweight are generally defined by body mass index (BMI), which is correlated with total body fat and estimates the relative risk of disease. BMI is calculated by weight in kilograms divided by height in meters squared (kg/m2). Overweight is typically defined as a BMI of 25-29.9 kg/m2, and obesity is typically defined as a BMI of 30 kg/m2. See, e.g., National Heart, Lung, and Blood Institute, Clinical Guidelines on the Identification, Evaluation, and Treatment of Overweight and Obesity in Adults, The Evidence Report, Washington, DC: U.S. Department of Health and Human Services, NIH publication no. 98-4083 (1998). The increase in obesity is of concern because of the excessive health risks associated with obesity, including coronary heart disease, strokes, hypertension, type 2 diabetes mellitus, dyslipidemia, sleep apnea, osteoarthritis, gall bladder disease, depression, and certain forms of cancer (e.g., endometrial, breast, prostate, and colon). The negative health consequences of obesity make it the second leading cause of preventable death in the United States and impart a significant economic and psychosocial effect on society. See, McGinnis M, Foege WH., "Actual Causes of Death in the United States," JAMA, 270, 2207-12 (1993). Obesity is now recognized as a chronic disease that requires treatment to reduce its associated health risks. Although weight loss is an important treatment outcome, one of the main goals of obesity management is to improve cardiovascular and metabolic values to reduce obesity-related morbidity and mortality. It has been shown that 5-10% loss of body weight can substantially improve metabolic values, such as blood glucose, blood pressure, and lipid concentrations. Hence, it is believed that a 5-10% intentional reduction in body weight may reduce morbidity and mortality. Currently available prescription drugs for managing obesity generally reduce weight by inducing satiety or decreasing dietary fat absorption. Satiety is achieved by increasing synaptic levels of norepinephrine, serotonin, or both. For example, stimulation of serotonin receptor subtypes 1 B, 1 D, and 2C and 1- and 2-adrenergic receptors decreases food intake by regulating satiety. See, Bray GA, "The New Era of Drug Treatment. Pharmacologic Treatment of Obesity: Symposium Overview," Obes Res., 3(suppl 4), 415s-7s (1995). Adrenergic agents (e.g., diethylpropion, benzphetamine, phendimetrazine, mazindol, and phentermine) act by modulating central norepinephrine and dopamine receptors through the promotion of catecholamine release. Older adrenergic weight-loss drugs (e.g., amphetamine, methamphetamine, and phenmetrazine), which strongly engage in dopamine pathways, are no longer recommended because of the risk of their abuse. Fenfluramine and dexfenfluramine, both serotonergic agents used to regulate appetite, are no longer available for use. More recently, CB1 cannabinoid receptor antagonists/inverse agonists have been suggested as potential appetite suppressants. See, e.g., Arnone, M., et al., "Selective Inhibition of Sucrose and Ethanol Intake by SR141716, an Antagonist of Central Cannabinoid (CB1) Receptors," Psvchopharmacol, 132, 104-106 (1997); Colombo, G., et al., "Appetite
Suppression and Weight Loss after the Cannabinoid Antagonist SR141716," Life Sci., 63, PL113-PL117 (1998); Simiand, J., et al., "SR141716, a CB1 Cannabinoid Receptor Antagonist, Selectively Reduces Sweet Food Intake in Marmose." Behav. Pharmacol., 9, 179-181 (1998); and Chaperon, F., et al., "Involvement of Central Cannabinoid (CB1) Receptors in the
Establishment of Place Conditioning in Rats," Psvchopharmacology, 135, 324-332 (1998). For a review of cannabinoid CB1 and CB2 receptor modulators, see Pertwee, R.G., "Cannabinoid Receptor Ligands: Clinical and Neuropharmacological Considerations, Relevant to Future Drug Discovery and Development." Exp. Opin. Invest. Drugs, 9(7), 1553-1571 (2000). Although investigations are on-going, there still exists a need for a more effective and safe therapeutic treatment for reducing or preventing weight-gain. In addition to obesity, there also exists an unmet need for treatment of alcohol abuse. Alcoholism affects approximately 10.9 million men and 4.4 million women in the United States. Approximately 100,000 deaths per year have been attributed to alcohol abuse or dependence. Health risks associated with alcoholism include impaired motor control and decision- making, cancer, liver disease, birth defects, heart disease, drug/drug interactions, pancreatitis and interpersonal problems. Studies have suggested that endogenous cannabinoid tone plays a critical role in the control of ethanol intake. The endogenous CB1 receptor antagonist SR- 141716A has been shown to block voluntary ethanol intake in rats and mice. See, Arnone, M., et al., "Selective Inhibition of Sucrose and Ethanol Intake by SR141716, an Antagonist of Central Cannabinoid (CB1) Receptors," Psychopharmacol, 132, 104-106 (1997). For a review, see Hungund, B.L and B.S. Basavarajappa, "Are Anandamide and Cannabinoid Receptors involved in Ethanol Tolerance? A Review of the Evidence," Alcohol & Alcoholism, 35(2) 126-133, 2000. Current treatments for alcohol abuse or dependence generally suffer from non-compliance or potential hepatotoxicity; therefore, there is a high unmet need for more effective treatment of alcohol abuse/dependence.
SUMMARY The present invention provides compounds of Formula (I):
Figure imgf000005_0001
(I) wherein R1 is an aryl optionally substituted with one or more substituents, or a heteroaryl optionally substituted with one or more substituents; R2 is aryl optionally substituted with one or more substituents, heteroaryl optionally substituted with one or more substituents, -CH=CH- R2a, or -CH2CH2-R2a, where R2a is hydrogen or a chemical moiety selected from (CrC-βJalkyl, 3- to 8-membered partially or fully saturated carbocyclic ring(s), 3- to 6-membered partially or fully saturated heterocycle, aryl, heteroaryl, where the chemical moiety is optionally substituted with one or more substituents; R3 is hydrogen, (C-ι-C4)alkyl, halo-substituted (Cι-C4)alkyl, or (C C4)alkoxy, or R3 taken together with R4 forms a nitrogen containing 5- to 6- membered partially or fully saturated heterocyclic ring optionally substituted with one or more substituents; and R4 is a chemical moiety selected from the group consisting of (Cr Cβ)alkyl, aryl, heteroaryl, aryl(Ci-C )aIkyl, a 3- to 8-membered partially or fully saturated carbocyclic ring(s), heteroaryl(Cι-C3)alkyl, 5-6 membered lactone, 5- to 6-membered lactam, and a 3- to 8-membered partially or fully saturated heterocycle, where said chemical moiety is optionally substituted with one or more substituents, or R4 taken together with R3 forms a nitrogen containing 5- to 6- membered partially or fully saturated heterocyclic ring optionally substituted with one or more substituents; a pharmaceutically acceptable salt thereof, a prodrug of the compound or the salt, or a solvate or hydrate of the compound, the salt or the prodrug. Preferably, R1 and R2 are each independently a substituted phenyl. More preferably, R1 and R2 are each independently a phenyl substituted with one to three substituents independently selected from the group consisting of halo (preferably, chloro or fluoro), (Cι-C4)alkoxy, (CrC4)alkyl, halo-substituted (C-ι-C4)alkyl (preferably fluoro-substituted alkyl, more preferably, trifluoromethyl) and cyano. Most preferably, R1 is 2-cyanophenyl, 2- chlorophenyl, 2-fluorophenyl, 2,4-dichlorophenyl, 2-fluoro-4-chlorophenyl, 2- chloro-4-fluorophenyl, 2-methylphenyl or 2,4-difluorophenyl; and R2 is 4- chlorophenyl, 4-cyanophenyl, 4-ethyIphenyl, 4-isopropylphenyl, 4- ethoxyphenyl, 4-isopropoxyphenyl, 4-trifluoromethylphenyl, or 4-fluorophenyl. Preferably, R3 is hydrogen, methyl or trifluoromethyl. Preferably, R4 is a chemical moiety selected from the group consisting of (Ci-Csjalkyl, aryl(Cι-C4)alkyl, and 3- to 8-membered partially or fully saturated carbocyclic ring(s), and 3- to 8-membered partially or fully saturated heterocycle, where said chemical moiety is optionally substituted with one or more substituent. More preferably, R4 is (Cι-C8)alkyl, halo- substituted (Cι-Cs)alkyl (preferably, fluoro-substituted (Cι-C8)alkyl), cyclopentyl, cyclohexyl, piperidin-1-yl, pyrrolidin-1-yl, or morpholin-1-yl.. In a preferred embodiment of the present invention, a compound of Formula (II) is provided.
Figure imgf000006_0001
(ll) wherein R1a, R1b, R2b, and R2c are each independently halo, (d-C^alkoxy, (C C4)alkyl, halo-substituted (Cι-C4)alkyl, or cyano (preferably, R1a is chloro, fluoro, methyl, or cyano; R1b is chloro, fluoro or hydrogen (i.e., m is 0); R2b is chloro, fluoro, (C-ι-C4)alkyl, trifluoromethyl, (CrC4)aIkoxy, or cyano; and R2c is hydrogen (i.e., n is 0)); n and m are each independently 0, 1 or 2; and R3 and R4 are as defined above; a pharmaceutically acceptable salt thereof, a prodrug of the compound or the salt, or a solvate or hydrate of the compound, the salt or the prodrug. Preferred compounds of Formula (I) or (II) include: 9-(4- chlorophenyl)-8-(2-chlorophenyl)-1 -isopropyl-1 ,9-dihydropurin-6-one; 8-(2- chloro-phenyl)-1-(2,2,2-trifluoro-ethyl)-9-(4-trifluoromethyl-phenyl)-1 ,9- dihydro-purin-6-one; 8-o-tolyl-1 -(2,2,2-trifluoro-ethyl)-9-(4-trifluoromethyl- phenyl)-1 ,9-dihydro-purin-6-one; 8-(2-chloro-phenyl)-9-(4-ethyl-phenyl)-1 - (2,2,2-trifluoro-ethyl)-1 ,9-dihydro-purin-6-one; 9-(4-tert-butyl-phenyl)-8-(2- chloro-phenyl)-1 -(2,2,2-trifluoro-ethyl)-1 ,9-dihydro-purin-6-one; 4-[8-(2- chloro-phenyl)-6-oxo-1-(2,2,2-trifluoro-ethyl)-1 ,6-dihydro-purin-9-yl]- benzonitrile; 2-[9-(4-chloro-phenyl)-6-oxo-1 -(2,2,2-trifluoro-ethyl)-6,9- dihydro-1 H-purin-8-yl]-benzonitrile; 9-(4-bromo-phenyl)-8-(2-chloro-phenyl)- 1 -(2,2,2-trifluoro-ethyl)-1 ,9-dihydro-purin-6-one; 8-(2-bromo-phenyl)-9-(4- chloro-phenyl)-1 -(2,2,2-trifluoro-ethyl)-1 ,9-dihydro-purin-6-one; and 9-(4- chloro-phenyl)-8-(2-chloro-phenyl)-1-(2,2,2-trifluoro-ethyl)-1 ,9-dihydro-purin- 6-one; or a solvate or hydrate of the compound. Some of the compounds described herein may contain at least one chiral center; consequently, those skilled in the art will appreciate that all stereoisomers (e.g., enantiomers and diasteroisomers) of the compounds illustrated and discussed herein are within the scope of the present invention. In addition, tautomeric forms of the compounds are also within the scope of the present invention. Compounds of the present invention have been shown to be useful cannabinoid receptor ligands (in particular, CB1 receptor antagonists). Accordingly, another aspect of the present invention is a pharmaceutical composition that comprises (1 ) a compound of the present invention, and (2) a pharmaceutically acceptable excipient, diluent, or carrier. Preferably, the composition comprises a therapeutically effective amount of a compound of the present invention. The composition may also contain at least one additional pharmaceutical agent (described herein). Preferred agents include nicotine receptor partial agonists, opioid antagonists (e.g., naltrexone and nalmefene), dopaminergic agents (e.g., apomorphine), attention deficit disorder (ADD including attention deficit hyperactivity disorder (ADHD)) agents (e.g., Ritalin™, Strattera™, Concerta™ and Adderall™), and anti- obesity agents (described herein below). In yet another embodiment of the present invention, a method for treating a disease, condition or disorder modulated by a cannabinoid receptor (preferably, a CB1 receptor) antagonists in animals that includes the step of administering to an animal in need of such treatment a therapeutically effective amount of a compound of the present invention (or a pharmaceutical composition thereof). Diseases, conditions, and/or disorders modulated by cannabinoid receptor antagonists include eating disorders (e.g., binge eating disorder, anorexia, and bulimia), weight loss or control (e.g., reduction in calorie or food intake, and/or appetite suppression), obesity, depression, atypical depression, bipolar disorders, psychoses, schizophrenia, behavioral addictions, suppression of reward-related behaviors (e.g., conditioned place avoidance, such as suppression of cocaine- and morphine-induced conditioned place preference), substance abuse, addictive disorders, impulsivity, alcoholism (e.g., alcohol abuse, addiction and/or dependence including treatment for abstinence, craving reduction and relapse prevention of alcohol intake), tobacco abuse (e.g., smoking addiction, cessation and/or dependence including treatment for craving reduction and relapse prevention of tobacco smoking), dementia (including memory loss, Alzheimer's disease, dementia of aging, vascular dementia, mild cognitive impairment, age-related cognitive decline, and mild neurocognitive disorder), sexual dysfunction in males (e.g., erectile difficulty), seizure disorders, epilepsy, inflammation, gastrointestinal disorders (e.g., dysfunction of gastrointestinal motility or intestinal propulsion), attention deficit disorder (ADD/ADHD), Parkinson's disease, and type II diabetes. In a preferred embodiment, the method is used in the treatment of weight loss, obesity, bulimia, ADD/ADHD, dementia, alcoholism, and/or tobacco abuse. Compounds of the present invention may be administered in combination with other pharmaceutical agents. Preferred pharmaceutical agents include nicotine receptor partial agonists, opioid antagonists (e.g., naltrexone (including naltrexone depot), antabuse, and nalmefene), dopaminergic agents (e.g., apomorphine), ADD/ADHD agents (e.g., methylphenidate hydrochloride (e.g., Ritalin™ and Concerta™), atomoxetine (e.g., Strattera™), and amphetamines (e.g., Adderall™)) and anti-obesity agents, such as apo-B/MTP inhibitors, 11β-hydroxy steroid dehydrogenase-1 (11 β-HSD type 1 ) inhibitors, peptide YY3-36 or analogs thereof, MCR-4 agonists, CCK-A agonists, monoamine reuptake inhibitors, sympathomimetic agents, β adrenergic receptor agonists, dopamine receptor agonists, melanocyte-stimulating hormone receptor analogs, 5-HT2c receptor agonists, melanin concentrating hormone receptor antagonists, leptin, leptin receptor agonists, galanin receptor antagonists, lipase inhibitors, bombesin receptor agonists, neuropeptide-Y receptor antagonists (e.g., NPY Y5 antagonists such as those described hereinbelow), thyromimetic agents, dehydroepiandrosterone or analogs thereof, glucocorticoid receptor antagonists, orexin receptor antagonists, glucagon-like peptide-1 receptor agonists, ciliary neurotrophic factors, human agouti-related protein antagonists, ghrelin receptor antagonists, histamine 3 receptor antagonists or inverse agonists, and neuromedin U receptor agonists, and the like. The combination therapy may be administered as (a) a single pharmaceutical composition which comprises a compound of the present invention, at least one additional pharmaceutical agent described herein and a pharmaceutically acceptable excipient, diluent, or carrier; or (b) two separate pharmaceutical compositions comprising (i) a first composition comprising a compound of the present invention and a pharmaceutically acceptable excipient, diluent, or carrier, and (ii) a second composition comprising at least one additional pharmaceutical agent described herein and a pharmaceutically acceptable excipient, diluent, or carrier. The pharmaceutical compositions may be administered simultaneously or sequentially and in any order. Definitions As used herein, the term "alkyl" refers to a hydrocarbon radical of the general formula CnH2n+ι. The alkane radical may be straight or branched. For example, the term "(Cι-C6)alkyl" refers to a monovalent, straight, or branched aliphatic group containing 1 to 6 carbon atoms (e.g., methyl, ethyl, n-propyl, /-propyl, π-butyl, /-butyl, s-butyl, f-butyl, π-pentyl, 1-methylbutyl, 2- methylbutyi, 3-methyl butyl, neopentyl, 3,3-dimethylpropyl, hexyl, 2- methylpentyl, and the like). Similarly, the alkyl portion (i.e., alkyl moiety) of an alkoxy, acyl (e.g., alkanoyl), alkylamino, dialkylamino, and alkylthio group have the same definition as above. When indicated as being "optionally substituted", the alkane radical or alkyl moiety may be unsubstituted or substituted with one or more substituents (generally, one to three substituents except in the case of halogen substituents such as perchloro or perfluoroalkyls) independently selected from the group of substituents listed below in the definition for "substituted." "Halo-substituted alkyl" refers to an alkyl group substituted with one or more halogen atoms (e.g., "fluoro- substituted alkyl" refers to fluoromethyl, difluoromethyl, trifluoromethyl, 1- fluoroethyl, 2-fluoroethyl, 1 ,1-difluoroethyl, 1 ,2-difluoroethyl, 2,2-difluoroethyl, 1 ,1 ,1-trifluoroethyl, 2,2,2-trifluoroethyl, 1 ,1 ,2-trifluoroethyl, 1 ,2,2-trifluoroethyl, 1 ,2,2,2-tetrafluoroethyl, 1 ,1 ,2,2-tetrafluoroethyl, 1 ,1 ,1 ,2-tetrafluoroethyl, 1 ,1 ,2,2,2-pentafluoroethyl, 1 ,1 ,1 ,2,2-pentafluoroethyl, perfluoroethyl, etc.). Preferred halo-substituted alkyls are the chloro- and fluoro-substituted alkyls, more preferably, fluoro-substituted alkyls. When substituted, the alkane radicals or alkyl moieties are preferably substituted with fluoro substituents (as described above), or 1 or 2 substituents independently selected from (C C3)alkyl, (C3-C6)cycloalkyl, (C2-C3)alkenyl, aryl, heteroaryl, 3- to 6- membered heterocycle, chloro, cyano, hydroxy, (Cι-C3)alkoxy, aryloxy, amino, (Cι-C6)alkyl amino, di-(Cι-C4)alkyl amino, aminocarboxylate (i.e., (C-i- C )alkyl-0-C(0)-NH-), hydroxy(C2-C3)alkylamino, or keto (oxo), and more preferably, 1 to 3 fluoro groups, or 1 substituent selected from (Cι-C3)alkyl, (C3-C6)cycloalkyl, (Cβ)aryl, 6-membered-heteroaryl, 3- to 6-membered heterocycle, (C-ι-C3)alkoxy, (C-ι-C )alkyl amino or di-(Cι-C2)alkyl amino. The terms "partially or fully saturated carbocyclic ring" (also referred to as "partially or fully saturated cycloalkyl") refers to nonaromatic rings that are either partially or fully hydrogenated and may exist as a single ring, bicyclic ring or a spiral ring. Unless specified otherwise, the carbocyclic ring is generally a 3- to 8-membered ring. For example, partially or fully saturated carbocyclic rings (or cycloalkyl) include groups such as cyclopropyl, cyclopropenyl, cyclobutyl, cyclobutenyl, cyclopentyl, cyclpentenyl, cyclopentadienyl, cyclohexyl, cyclohexenyl, cyclohexadienyl, norbomyl (bicyclo[2.2.1]heptyl), norbomenyl, bicyclo[2.2.2]octyl, and the like. When designated as being "optionally substituted", the partially saturated or fully saturated cycloalkyl group may be unsubstituted or substituted with one or more substituents (typically, one to three substituents) independently selected from the group of substituents listed below in the definition for "substituted." A substituted carbocyclic ring also includes groups wherein the carbocyclic ring is fused to a phenyl ring (e.g., indanyl). The carbocyclic group may be attached to the chemical entity or moiety by any one of the carbon atoms within the carbocyclic ring system. When substituted, the carbocyclic group is preferably substituted with 1 or 2 substituents independently selected from (CrC3)alkyl, (C2-C3)alkenyl, (Cι-C6)alkylidenyl, aryl, heteroaryl, 3- to 6-membered heterocycle, chloro, fluoro, cyano, hydroxy, (C1-C3)alkoxy, aryloxy, amino, (C Ce)al yl amino, di-(CrC4)alkyl amino, aminocarboxylate (i.e., (Cι-C3)alkyl-O-C(0)-NH-), hydroxy(C2- C3)alkylamino, or keto (oxo), and more preferably 1 or 2 from substituents independently selected from (CrC2)alkyl, 3- to 6-membered heterocycle, fluoro, (Cι-C3)alkoxy, (CrC )alkyl amino or di-(Cι-C2)alkyl amino. Similarly, any cycloalkyl portion of a group (e.g., cycloalkylalkyl, cycloalkylamino, etc.) has the same definition as above. The term "partially saturated or fully saturated heterocyclic ring" (also referred to as "partially saturated or fully saturated heterocycle") refers to nonaromatic rings that are either partially or fully hydrogenated and may exist as a single ring, bicyclic ring or a spiral ring. Unless specified otherwise, the heterocyclic ring is generally a 3- to 6-membered ring containing 1 to 3 heteroatoms (preferably 1 or 2 heteroatoms) independently selected from sulfur, oxygen and/or nitrogen. Partially saturated or fully saturated heterocyclic rings include groups such as epoxy, aziridinyl, tetrahydrofuranyl, dihydrofuranyl, dihydropyridinyl, pyrrolidinyl, N- methylpyrrolidinyl, imidazolidinyl, imidazolinyl, piperidinyl, piperazinyl, pyrazolidinyl, 2H-pyranyl, 4H-pyranyl, 2H-chromenyl, oxazinyl, morpholino, thiomorpholino, tetrahydrothienyl, tetrahydrothienyl 1 ,1 -dioxide, and the like. When indicated as being "optionally substituted", the partially saturated or fully saturated heterocycle group may be unsubstiuted or substituted with one or more substituents (typically, one to three substituents) independently selected from the group of substituents listed below in the definition for "substituted." A substituted heterocyclic ring includes groups wherein the heterocyclic ring is fused to an aryl or heteroaryl ring (e.g., 2,3- dihydrobenzofuranyl, 2,3-dihydroindolyl, 2,3-dihydrobenzothiophenyl, 2,3- dihydrobenzothiazolyl, etc.). When substituted, the heterocycle group is preferably substituted with 1 or 2 substituents independently selected from (Cι-C3)alkyl, (C3-C6)cycloalkyl, (C2-C4)alkenyl, aryl, heteroaryl, 3- to 6- membered heterocycle, chloro, fluoro, cyano, hydroxy, (C-i-CsJalkoxy, aryloxy, amino, (Cι-C6)alkyl amino, di-(Cι-C3)alkyl amino, aminocarboxylate (i.e., (CrC3)alkyl-0-C(0)-NH-), or keto (oxo), and more preferably with 1 or 2 substituents independently selected from (Cι-C3)alkyl, (C3-C6)cycloalkyl, (Cβjaryl, 6-membered-heteroaryl, 3- to 6-membered heterocycle, or fluoro. The heterocyclic group may be attached to the chemical entity or moiety by any one of the ring atoms within the heterocyclic ring system. Similarly, any heterocycle portion of a group (e.g., heterocycle-substituted alkyl, heterocycle carbonyl, etc.) has the same definition as above. The term "aryl" or "aromatic carbocyclic ring" refers to aromatic moieties having a single (e.g., phenyl) or a fused ring system (e.g., naphthalene, anthracene, phenanthrene, etc.). A typical aryl group is a 6- to 10-membered aromatic carbocyclic ring(s). When indicated as being "optionally substituted", the aryl groups may be unsubstituted or substituted with one or more substituents (preferably no more than three substituents) independently selected from the group of substituents listed below in the definition for "substituted." Substituted aryl groups include a chain of aromatic moieties (e.g., biphenyl, terphenyl, phenylnaphthalyl, etc.). When substituted, the aromatic moieties are preferably substituted with 1 or 2 substituents independently selected from (Cι-C )alkyl, (C2-C3)alkenyl, aryl, heteroaryl, 3- to 6-membered heterocycle, bromo, chloro, fluoro, iodo, cyano, hydroxy, (Cι-C4)alkoxy, aryloxy, amino, (Cι-C6)alkyl amino, di-(Cι-C3)alkyl amino, or aminocarboxylate (i.e., (Ci-C3)alkyl-0-C(0)-NH-), and more preferably, 1 or 2 substituents independently selected from (Cι-C4)alkyl, chloro, fluoro, cyano, hydroxy, or (Cι-C4)alkoxy. The aryl group may be attached to the chemical entity or moiety by any one of the carbon atoms within the aromatic ring system. Similarly, the aryl portion (i.e., aromatic moiety) of an aroyl or aroyloxy (i.e., (aryl)-C(O)-O-) has the same definition as above. The term "heteroaryl" or "heteroaromatic ring" refers to aromatic moieties containing at least one heteratom (e.g., oxygen, sulfur, nitrogen or combinations thereof) within a 5- to 10-membered aromatic ring system (e.g., pyrrolyl, pyridyl, pyrazolyl, indolyl, indazolyl, thienyl, furanyl, benzofuranyl, oxazolyl, imidazolyl, tetrazolyl, triazinyl, pyrimidyl, pyrazinyl, thiazolyl, purinyl, benzimidazolyl, quinolinyl, isoquinolinyl, benzothiophenyl, benzoxazolyl, etc.). The heteroaromatic moiety may consist of a single or fused ring system. A typical single heteroaryl ring is a 5- to 6-membered ring containing one to three heteroatoms independently selected from oxygen, sulfur and nitrogen and a typical fused heteroaryl ring system is a 9- to 10- membered ring system containing one to four heteroatoms independently selected from oxygen, sulfur and nitrogen. When indicated as being "optionally substituted", the heteroaryl groups may be unsubstituted or substituted with one or more substituents (preferably no more than three substituents) independently selected from the group of substituents listed below in the definition for "substituted." When substituted, the heteroaromatic moieties are preferably substituted with 1 or 2 substituents independently selected from (Cι-C4)alkyl, (C2-C3)alkenyl, aryl, heteroaryl, 3- to 6-membered heterocycle, bromo, chloro, fluoro, iodo, cyano, hydroxy, (C-i- C- alkoxy, aryloxy, amino, (Cι-Ce)alkyl amino, di-(Cι-C3)alkyl amino, or aminocarboxylate (i.e., (Cι-C3)alkyl-0-C(O)-NH-), and more preferably, 1 or 2 substituents independently selected from (Cι-C4)alkyl, chloro, fluoro, cyano, hydroxy, (Cι-C4)alkoxy, (Cι-C4)alkyl amino or di-(Cι-C2)alkyl amino. The heteroaryl group may be attached to the chemical entity or moiety by any one of the atoms within the aromatic ring system (e.g., imidazol-1-yl, imidazol-2-yl, imidazol-4-yl, imidazol-5-yl, pyrid-2-yl, pyrid-3-yl, pyrid-4-yl, pyrid-5-yl, or pyrid-6-yl). Similarly, the heteroaryl portion (i.e., heteroaromatic moiety) of a heteroaroyl or heteroaroyloxy (i.e., (heteroaryl)- C(O)-O-) has the same definition as above. The term "acyl" refers to hydrogen, alkyl, partially saturated or fully saturated cycloalkyl, partially saturated or fully saturated heterocycle, aryl, and heteroaryl substituted carbonyl groups. For example, acyl includes groups such as (Cι-Ce)alkanoyl (e.g., formyl, acetyl, propionyl, butyryl, valeryl, caproyl, f-butylacetyl, etc.), (C3-C6)cycloalkylcarbonyl (e.g., cyclopropylcarbonyl, cyclobutylcarbonyl, cyclopentylcarbonyl, cyclohexylcarbonyl, etc.), heterocyclic carbonyl (e.g., pyrrolidinylcarbonyl, pyrrolid-2-one-5-carbonyl, piperidinylcarbonyl, piperazinylcarbonyl, tetrahydrofuranylcarbonyl, etc.), aroyl (e.g., benzoyl) and heteroaroyl (e.g., thiophenyl-2-carbonyl, thiophenyl-3-carbonyl, furanyl-2-carbonyl, furanyl-3- carbonyl, 1 H-pyrroyl-2-carbonyl, 1 H-pyrroyl-3-carbonyl, benzo[b]thiophenyl- 2-carbonyl, etc.). In addition, the alkyl, cycloalkyl, heterocycle, aryl and heteroaryl portion of the acyl group may be any one of the groups described in the respective definitions above. When indicated as being "optionally substituted", the acyl group may be unsubstituted or optionally substituted with one or more substituents (typically, one to three substituents) independently selected from the group of substituents listed below in the definition for "substituted" or the alkyl, cycloalkyl, heterocycle, aryl and heteroaryl portion of the acyl group may be substituted as described above in the preferred and more preferred list of substituents, respectively. The term "substituted" specifically envisions and allows for one or more substitutions that are common in the art. However, it is generally understood by those skilled in the art that the substituents should be selected so as to not adversely affect the pharmacological characteristics of the compound or adversely interfere with the use of the medicament. Suitable substituents for any of the groups defined above include (C C6)alkyl, (C3-C7)cycloalkyl, (C2-C6)alkenyl, (Cι-C6)alkylidenyl, aryl, heteroaryl, 3- to 6-membered heterocycle, halo (e.g., chloro, bromo, iodo and fluoro), cyano, hydroxy, (CrC6)alkoxy, aryloxy, sulfhydryl (mercapto), (Cι-C6)alkylthio, arylthio, amino, mono- or di-(Cι-Ce)alkyl amino, quaternary ammonium salts, amino(Cι-C6)alkoxy, aminocarboxylate (i.e., (CrC6)alkyl- O-C(O)-NH-), hydroxy(C2-C6)alkylamino, amino(CrC6)alkylthio, cyanoamino, nitro, (CrC6)carbamyl, keto (oxo), acyl, (Cι-C6)alkyl-CO2-, glycolyl, glycyl, hydrazino, guanyl, sulfamyl, sulfonyl, sulfinyl, thio(Cι-C6)alkyl-C(O)-, thio(C C6)alkyl-C02-, and combinations thereof. In the case of substituted combinations, such as "substituted aryl(Cι-C6)alkyl", either the aryl or the alkyl group may be substituted, or both the aryl and the alkyl groups may be substituted with one or more substituents (typically, one to three substituents except in the case of perhalo substitutions). An aryl or heteroaryl substituted carbocyclic or heterocyclic group may be a fused ring (e.g., indanyl, dihydrobenzofuranyl, dihydroindolyl, etc.). The term "solvate" refers to a molecular complex of a compound represented by Formula (I) or (II) (including prodrugs and pharmaceutically acceptable salts thereof) with one or more solvent molecules. Such solvent molecules are those commonly used in the pharmaceutical art, which are known to be innocuous to the recipient, e.g., water, ethanol, and the like. The term "hydrate" refers to the complex where the solvent molecule is water. The term "protecting group" or "Pg" refers to a substituent that is commonly employed to block or protect a particular functionality while reacting other functional groups on the compound. For example, an "amino- protecting group" is a substituent attached to an amino group that blocks or protects the amino functionality in the compound. Suitable amino-protecting groups include acetyl, trifluoroacetyl, f-butoxycarbonyl (BOC), benzyloxycarbonyl (CBz) and 9-fluorenylmethylenoxycarbonyl (Fmoc). Similarly, a "hydroxy-protecting group" refers to a substituent of a hydroxy group that blocks or protects the hydroxy functionality. Suitable protecting groups include acetyl and silyl. A "carboxy-protecting group" refers to a substituent of the carboxy group that blocks or protects the carboxy functionality. Common carboxy-protecting groups include -CH2CH2SO2Ph, cyanoethyl, 2-(trimethylsilyl)ethyl, 2-(trimethylsilyl)ethoxymethyl, 2-(p- toluenesulfonyl)ethyl, 2-(p-nitrophenylsulfenyl)ethyl, 2-(diphenylphosphino)- ethyl, nitroethyl and the like. For a general description of protecting groups and their use, see T. W. Greene, Protective Groups in Organic Synthesis, John Wiley & Sons, New York, 1991. The phrase "therapeutically effective amount" means an amount of a compound of the present invention that (i) treats or prevents the particular disease, condition, or disorder, (ii) attenuates, ameliorates, or eliminates one or more symptoms of the particular disease, condition, or disorder, or (iii) prevents or delays the onset of one or more symptoms of the particular disease, condition, or disorder described herein. The term "animal" refers to humans (male or female), companion animals (e.g., dogs, cats and horses), food-source animals, zoo animals, marine animals, birds and other similar animal species. "Edible animals" refers to food-source animals such as cows, pigs, sheep and poultry. The phrase "pharmaceutically acceptable" indicates that the substance or composition must be compatible chemically and/or toxicologically, with the other ingredients comprising a formulation, and/or the mammal being treated therewith. The terms "treating", "treat", or "treatment" embrace both preventative, i.e., prophylactic, and palliative treatment. The terms "modulated by a cannabinoid receptor" or "modulation of a cannabinoid receptor" refers to the activation or deactivation of a cannabinoid receptor. For example, a ligand may act as an agonist, partial agonist, inverse agonist, antagonist, or partial antagonist. The term "antagonist" includes both full antagonists and partial antagonists, as well as inverse agonists. The term "CB-1 receptor" refers to the G-protein coupled type 1 cannabinoid receptor. The term "compounds of the present invention" (unless specifically identified otherwise) refer to compounds of Formulae (I) and (II), prodrugs thereof, pharmaceutically acceptable salts of the compounds, and/or prodrugs, and hydrates or solvates of the compounds, salts, and/or prodrugs, as well as, all stereoisomers (including diastereoisomers and enantiomers), tautomers and isotopically labeled compounds. DETAILED DESCRIPTION Compounds of the present invention may be synthesized by synthetic routes that include processes analogous to those well-known in the chemical arts, particularly in light of the description contained herein. The starting materials are generally available from commercial sources such as Aldrich Chemicals (Milwaukee, Wl) or are readily prepared using methods well known to those skilled in the art (e.g., prepared by methods generally described in Louis F. Fieser and Mary Fieser, Reagents for Organic Synthesis, v. 1 -19, Wiley, New York (1967-1999 ed.), or Beilsteins Handbuch der organischen Chemie, 4, Aufl. ed. Springer-Verlag, Berlin, including supplements (also available via the Beilstein online database)). For illustrative purposes, the reaction schemes depicted below provide potential routes for synthesizing the compounds of the present invention as well as key intermediates. For a more detailed description of the individual reaction steps, see the Examples section below. Those skilled in the art will appreciate that other synthetic routes may be used to synthesize the inventive compounds. Although specific starting materials and reagents are depicted in the schemes and discussed below, other starting materials and reagents can be easily substituted to provide a variety of derivatives and/or reaction conditions. In addition, many of the compounds prepared by the methods described below can be further modified in light of this disclosure using conventional chemistry well known to those skilled in the art. In the preparation of compounds of the present invention, protection of remote functionality (e.g., primary or secondary amine) of intermediates may be necessary. The need for such protection will vary depending on the nature of the remote functionality and the conditions of the preparation methods. Suitable amino-protecting groups (NH-Pg) include acetyl, trifluoroacetyl, f-butoxycarbonyl (BOC), benzyloxycarbonyl (CBz) and 9- fluorenylmethyleneoxycarbonyl (Fmoc). The need for such protection is readily determined by one skilled in the art. For a general description of protecting groups and their use, see T. W. Greene, Protective Groups in Organic Synthesis. John Wiley & Sons, New York, 1991. Compounds of Formula (I) or (II) can be prepared using the general procedures described by R.J. Chorvat, et al. in J. Med. Chem. 42, 833-848 (1999) and depicted in Scheme I below.
Figure imgf000019_0001
(I) or (II) 1 (c) 1 (b) Scheme I Intermediate 1(a) where R3 is hydrogen may be prepared by reacting the desired amino compound (R2-NH2, where R2 is as defined above) with 4,6-dichloro-5-aminopyrimidine (available from Sigma-Aldrich, St. Louis, MO) in refluxing aqueous hydrochloric acid (A. Miyashita et al. in Chem. Pharm. Bull.. 46, 390-399 (1998)) or ethoxyethanol at elevated temperatures. Suitable amino compounds (R2-NH2) include those compounds where R2 is ' aryl (e.g., aniline) or substituted aryl (e.g., 2-chloroaniline, 2-fluoroaniline, 2,4-dichloroaniline, 2-fluoro-4-chloroaniline, 2-chloro-4-flurooaniline, 2,4- difluoroaniline, and other substituted arylamines). Other commercially available derivatives of 4,6-dichloro-5-aminopyrimidine may be used as a starting material for those compounds of Formula (I) or (II) where R3 is other than hydrogen (e.g., 2-methyl-4,6-dichloro-5-aminopyrimidine and 2-ethyl- 4,6-dichloro-5-aminopyrimidine). For representative literature syntheses of 4,6-dichloro-5-aminopyrimidine derivatives see: A. Albert et al. in J. Chem. Soc. 3832 (1954) and W.E. Hymans in J. Heterocvcl. Chem.. 13, 1141 (1976). Intermediate 1 (a) can then be acylated using conventional chemistry well-known to those skilled in the art. For example, intermediate 1 (a) may be reacted with the desired aroyl or heteroaroyl chloride in a basic solvent (e.g., pyridine) to produce intermediate 1 (b). Alternatively, intermediate 1 (a) may be reacted with the desired aroyl or heteroaroyl chloride in a reaction inert solvent (e.g., tetrahydrofuran, methylene chloride, Λ/,Λ/-dimethyl- acetamide). The addition of a suitable base (e.g., triethylamine, diisopropyl- ethylamine) may help facilitate the reaction. Suitable aroyl chlorides include benzoyl chloride, o-chlorobenzoyl chlorides, o-fluorobenzoyl chloride, p-chlorobenzoyl chloride, p-fluorobenzoyl chloride, 2,4-dichlorobenzoyl chloride, 2,4-difluorobenzoyl chloride, and the like. Intermediate 1(b) may then be cyclized to the hydroxy purine (9H- purin-6-ol) intermediate 1 (c) by refluxing in a weak acid (e.g., acetic acid) or with sulfuric acid in an appropriate solvent (e.g., isopropyl alcohol, toluene). N-Alkylation of intermediate (1c) can then be accomplished using an alkylating agent, preferably an alkyl iodide, alkyl triflate, or a substituted benzyl bromide, and a base, such as lithium hexamethyldisilazide, cesium carbonate, potassium carbonate or lithium hydride, in a polar non-protic solvent such as DMF or THF, at temperatures ranging from about 37°C to about 150 °C to provide compounds of Formula (I) or (II). Scheme II below illustrates an alternative synthesis of intermediate 1(c).
Figure imgf000021_0001
Scheme II Intermediate 2(a) can be first acylated with an acid chloride. Suitable acid chlorides (R1-COCI) include those compounds where R1 is aryl (e.g., benzoyl chloride), substituted aryl (e.g., 2-chlorobenzoyl chloride, 4-chlorobenzoyl chloride, and other substituted aryl acid chlorides), heteroaryl, or substituted heteroaryl. Compound 2(b) may be converted to intermediate 2(c) by treatment with dehydrating agents like POCI3 using procedures described by H.C. Koppel in J. Org. Chem.. 23, 1457 (1958) and in J. Chem. Soc, Perkin Trans. I. 879 (1984). The R1 group, where R1 is aryl, substituted aryl, heteroaryl or substituted heteroaryl, may then be introduced using reagents like R2-B(OH)2 , R2-Br or R2-l and Pd catalysts (see Y. Wan et al. in Synthesis, 1597-1600 (2002) , and references contained therein) or copper(ll) catalysts such as cupric acetate or cupric bromide (see, S. Ding et al. in Tetrahedron Lett.. 42, 8751-8755 (2001 ), A. Klapars et al. J. Am. Chem. Soc. 123, 7727-7729 (2001 ), and references contained therein). A SNAr reaction may also be useful for introducing electron deficient heterocycles (see M. Medebielle in New. J. Chem., 19, 349 (1995)). Scheme III describes the preparation of compounds of formula (I) or (II) where R4 is alkyl, cycloalkyl, piperidinyl, pyrrolidiny, aryl or heteroaryl and R3 is hydrogen or (C1-C4) alkyl.
Figure imgf000022_0001
3(a) 3(b)
Figure imgf000022_0002
3(e) 3(d) 3(c)
R NH2, Me3AI aq K2C03
R4NH2, EDC R3C(OR)3
Figure imgf000022_0005
Figure imgf000022_0004
Figure imgf000022_0003
3(f) 3(g) (I) or (II) Scheme III Intermediate 3(a) can be prepared by treating an appropriately substituted amine derivative (e.g., R2NH2) with trimethylaluminum under inert atmospheric conditions, followed by condensation with the appropriately substituted R1CN derivative. Suitable amines include substituted phenyl amines (e.g., 4-chlorophenyl amine, 4-fluorophenyl amine, 4-bromophenyl amine, 4-iodophenyl amine, 4-cyanophenyl amine, and the like) pyridin-2-yl amine, pyridin-3-yl amine, pyridin-4-yl amine, substituted pyridinyl amines (e.g., 2-dimethylaminopyridin-5-yl amine, 2-methoxypyridin-5-yl amine, 5- chloropyridin-2-yl amine, 5-methylpyridin-2-yl, 5-methoxypyridin-2-yl amine, 3-chloropyridin-4-yl; amine, 2-N-morpholinylpyridin-5-yl, and the like), and other commercially available or easily synthesized substituted or unsubstituted aryl and heteroaryl amines. Suitable cyano compounds include substituted benzonitriles (e.g., 2-chlorobenzonitrile, 2- fluorobenzonitrile, 2-methoxybenzonitrile, 2-methylbenzonitrile, 2,4- dichlorobenzonitrile, 2,4-difluorobenzonitrile, 2-chloro-4-fluorobenzonitrile, 2- chloro-4-methylbenzonitrile, 2,4-dimethoxybenzonitrile, 2-methyl-4- chlorobenzonitrile, and the like), cya no-substituted pyridines (e.g., 4-cyano- 3-chloropyridine) and other commercially available or easily synthesized substituted or unsubstituted aryl or heteroaryl nitriles. Intermediate 3(a) can then be condensed with a 3-bromo-2-oxo- propionic acid ester (wherein R is an alkyl group like methyl, ethyl, propyl, benzyl, etc.) to produce the cyclized 4-hydroxy-4,5-dihydro-1 H-imidazole ester 3(b) using procedures analogous to those described by Khanna, I.K., et al., in J. Med. Chem., 40, 1634 (1997). For example, the amidine intermediate 3(a) is refluxed in a polar solvent (e.g., isopropanol) in the presence of a mild base (e.g., sodium bicarbonate). Generally, the reaction (i.e., cyclization followed by dehydration) proceeds directly to the desired imidazole ester intermediate 3(c). In some instances, it may be necessary to dehydrate the carbinol condensation product 3(b) with an acid catalyst (e.g., p-toluenesulfonic acid) in a non-polar solvent (e.g., toluene) at temperatures ranging from room temperature to 150 °C to provide the desired imidazole ester 3(c). Alternatively, intermediate 3(b) may be treated with methanesulfonyl chloride in the presence of a base (e.g., triethylamine) to give 3(c). Intermediate 3(d), where L1 is a bromine or iodine) can be synthesized from imidazole ester 3(c) using a halogenating agent such as bromine, N-bromosuccinimide, iodine, or N-iodosuccinimide in a suitable protic solvent (e.g., glacial acetic acid or trifluoroacetic acid) or an aprotic solvent (e.g, acetonitrile, ether or THF) at reaction temperatures ranging from 35 °C to 100 °C. The halogenated imidizolyl derivative 3(d) can be treated with an ammonia equivalent such as lithium bis(trimethylsilyl)amide in the presence of a catalytic amount of Pd(dba)2 and a phosphine ligand such as P(t-Bu)3 in a non-polar solvent (e.g., toluene) at temperatures ranging from 23 °C to reflux to give intermediate 3(e). Examples of related procedures are described by Lee et al. in Organic Letters, 3, 2729-273 (2001). Hydrolysis of the carboxy-protecting group (R) in 3(e) with aqueous K2CO3 in an alcoholic solvent or with an alkali base such as KOH in a polar solvent (e.g., ethanol) can provide intermediate 3(f). Treatment of the acid 3(f) with an appropriately substituted cycloalkyl, aryl or heteroaryl amine (R4NH2) in the presence of a coupling reagent (e.g., EDC or O-(7- azabenzotriazol-1-yl)-N,N,N',N'-tetramethyluronium hexafluorophosphate (HATU)) and a base (e.g., triethylamine) in a suitable aprotic solvent (e.g, DMF, CH2CI2) can provide intermediate 3(g). Alternatively, intermediate 3(e) can be directly treated with an appropriately substituted amine derivative (e.g., R4NH2) in the presence of trimethylaluminum in a non-polar solvent (e.g., toluene) to give intermediate 3(g). Cyclocondensation of 3(g) with an ortho ester derivative (R3C(OR)3) such as triethylorthoformate or triethylorthoacetate in the presence of an acid catalyst such as p- toluenesulfonic acid at temperatures ranging from about 50 °C to about 150 °C can provide compounds of Formula I or II. The imidazolyl ester intermediate 3(e) can also be converted into compounds of formula (I) or (II) where R3 is not limited to hydrogen and C C alkyl by the methods shown in Scheme IV.
Figure imgf000025_0001
3(e) 5(a) (I) or (II) Scheme IV Intermediate 3(a) can be reacted with an appropriately substituted amidine derivative in the presence of an acid catalyst (e.g., acetic acid) in a polar solvent (e.g., ethanol, 2-ethoxyethanol) to give the 9H-purin-6-ol intermediate 5(a) which can be N-alkylated as previously described to give compounds of formula (I) or (II). Scheme V illustrates another method for preparing compounds of formula (I) or (II) where R2 is aryl or heteroaryl.
Figure imgf000026_0001
5(a) 5(b) 5(c)
Figure imgf000026_0002
5(f) 5(e) 5(d)
Figure imgf000026_0003
5(g) (I) or (II) Scheme V An appropriately substituted thiobenzimidic acid benzyl ester 5(a) can be reacted with amino-cyano-acetic acid alkyl ester 5(b) in non-polar solvents (e.g. CHCI3) to give the imidazolyl intermediate 5(c) (see J. Heyes, N. Ward in DE2142831 ). Protection of the imidazolyl group using methods well known in the art can provide intermediate 5(d). For example, 5(c) can be treated with benzyl bromide or p-methoxybenzyl bromide in an aprotic polar solvent (e.g. DMF) in the presence of a base (e.g., K2CO3) to give 5(d). Cyclocondensation of 5(d) can be accomplished as previously described using R3C(OR)3 to provide the 9H-purin-6-ol derivative 5(e). Reaction of 5(e) with R4-L where L is a leaving group (e.g., Br, CI, OTf) in the presence of a base (e.g., Cs2C03 or lithium hexamethyldisilazide) in a polar solvent (e.g., DMF or THF) can give the N-alkylated 1 ,9-dihydro-purin-6-one 5(f). Removal of the protecting group in 5(f) by standard methods well known in the art can provide 5(g). Intermediate 5(g) can be treated with substituted arylboronic acids and Pd catalysts or aryl iodides (or arylbromides) and Cu catalysts as previously described to provide N-arylated compounds of formula (I) or (II). Conventional methods and/or techniques of separation and purification known to one of ordinary skill in the art can be used to isolate the compounds of the present invention, as well as the various intermediates related thereto. Such techniques will be well-known to one of ordinary skill in the art and may include, for example, all types of chromatography (high pressure liquid chromatography (HPLC), column chromatography using common adsorbents such as silica gel, and thin-layer chromatography), recrystallization, and differential (i.e., liquid-liquid) extraction techniques. The compounds of the present invention may be isolated and used perse or in the form of its pharmaceutically acceptable salt, solvate and/or hydrate. The term "salts" refers to inorganic and organic salts of a compound of the present invention. These salts can be prepared in situ during the final isolation and purification of a compound, or by separately reacting the compound, N-oxide, or prodrug with a suitable organic or inorganic acid and isolating the salt thus formed. Representative salts include the hydrobromide, hydrochloride, hydroiodide, sulfate, bisulfate, nitrate, acetate, trifluoroacetate, oxalate, besylate, palmitiate, pamoate, malonate, stearate, laurate, malate, borate, benzoate, lactate, phosphate, hexafluorophosphate, benzene sulfonate, tosylate, formate, citrate, maleate, fumarate, succinate, tartrate, naphthylate, mesylate, glucoheptonate, lactobionate, and laurylsulphonate salts, and the like. These may include cations based on the alkali and alkaline earth metals, such as sodium, lithium, potassium, calcium, magnesium, and the like, as well as non-toxic ammonium, quaternary ammonium, and amine cations including, but not limited to, ammonium, tetramethylammonium, tetraethylammonium, methylamine, dimethylamine, trimethylamine, triethylamine, ethylamine, and the like. See, e.g., Berge, et al., J. Pharm. Sci., 66, 1-19 (1977). The term "prodrug" means a compound that is transformed in vivo to yield a compound of Formula (I) or a pharmaceutically acceptable salt, hydrate or solvate of the compound. The transformation may occur by various mechanisms, such as through hydrolysis in blood. A discussion of the use of prodrugs is provided by T. Higuchi and W. Stella, "Pro-drugs as Novel Delivery Systems," Vol. 14 of the A.C.S. Symposium Series, and in Bioreversible Carriers in Drug Design, ed. Edward B. Roche, American Pharmaceutical Association and Pergamon Press, 1987. For example, if a compound of the present invention contains a carboxylic acid functional group, a prodrug can comprise an ester formed by the replacement of the hydrogen atom of the acid group with a group such as (Ci-Csjalkyl, (C2-Cι2)alkanoyloxymethyl, 1-(alkanoyloxy)ethyl having from 4 to 9 carbon atoms, 1-methyl-1-(alkanoyloxy)-ethyl having from 5 to 10 carbon atoms, alkoxycarbonyloxymethyl having from 3 to 6 carbon atoms, 1- (alkoxycarbonyloxy)ethyl having from 4 to 7 carbon atoms, 1-methyl-1- (alkoxycarbonyloxy)ethyl having from 5 to 8 carbon atoms, N- (alkoxycarbonyl)aminomethyl having from 3 to 9 carbon atoms, 1-(N-
(alkoxycarbonyl)amino)ethyl having from 4 to 10 carbon atoms, 3-phthalidyl, 4-crotonolactonyl, gamma-butyrolacton-4-yl, di-N,N-(Cι-C2)alkylamino(C2- C3)alkyl (such as β-dimethylaminoethyl), carbamoyl-(Cι-C2)alkyl, N,N-di(C-ι- C2)alkylcarbamoyl-(Cι-C2)alkyl and piperidino-, pyrrolidino- or morpholino(C2-C3)alkyl. Similarly, if a compound of the present invention contains an alcohol functional group, a prodrug can be formed by the replacement of the hydrogen atom of the alcohol group with a group such as (Cι- C6)alkanoyloxymethyl, 1-((Cι-C6)alkanoyloxy)ethyl, 1-methyl-1-((C C6)alkanoyIoxy)ethyl, (Cι-C6)alkoxycarbonyloxymethyl, N-(Cι-
C6)alkoxycarbonylaminomethyl, succinoyl, (Cι-C6)alkanoyl, -amino(d- C4)alkanoyl, arylacyl and α-aminoacyl, or α-aminoacyl-α-aminoacyl, where each α-aminoacyl group is independently selected from the naturally occurring L-amino acids, P(O)(0H)2, P(O)(0(Cι-C6)alkyl)2 or glycosyl (the radical resulting from the removal of a hydroxyl group of the hemiacetal form of a carbohydrate). If a compound of the present invention incorporates an amine functional group, a prodrug can be formed by the replacement of a hydrogen atom in the amine group with a group such as R-carbonyl, RO-carbonyl, NRR'-carbonyl where R and R' are each independently (Cι-C 0)alkyl, (C3- C7)cycloalkyl, benzyl, or R-carbonyl is a natural α-aminoacyl or natural α- aminoacyl-natural α-aminoacyl, -C(OH)C(0)OY' wherein Y' is H, (Cι-C6)alkyl or benzyl, -C(OY0)Yι wherein Y0 is (C C4) alkyl and Y1 is (CrC6)alkyl, carboxy(C1-C6)alkyl, amino(Cι-C4)alkyl or mono-N- or di-N,N-(Cι- C6)alkylaminoalkyl, -C(Y2)Y3 wherein Y2 is H or methyl and Y3 is mono-N- or di-N,N-(Cι-C6)alkylamino, morpholino, piperidin-1-yl or pyrrolidin-1-yl. The compounds of the present invention may contain asymmetric or chiral centers, and, therefore, exist in different stereoisomeric forms. It is intended that all stereoisomeric forms of the compounds of the present invention as well as mixtures thereof, including racemic mixtures, form part of the present invention. In addition, the present invention embraces all geometric and positional isomers. For example, if a compound of the present invention incorporates a double bond or a fused ring, both the cis- and trans- forms, as well as mixtures, are embraced within the scope of the invention. Diastereomeric mixtures can be separated into their individual diastereoisomers on the basis of their physical chemical differences by methods well known to those skilled in the art, such as by chromatography and/or fractional crystallization. Enantiomers can be separated by converting the enantiomeric mixture into a diastereomeric mixture by reaction with an appropriate optically active compound (e.g., chiral auxiliary such as a chiral alcohol or Mosher's acid chloride), separating the diastereoisomers and converting (e.g., hydrolyzing) the individual diastereoisomers to the corresponding pure enantiomers. Also, some of the compounds of the present invention may be atropisomers (e.g., substituted biaryls) and are considered as part of this invention. Enantiomers can also be separated by use of a chiral HPLC column. The compounds of the present invention may exist in unsolvated as well as solvated forms with pharmaceutically acceptable solvents such as water, ethanol, and the like, and it is intended that the invention embrace both solvated and unsolvated forms. It is also possible that the compounds of the present invention may exist in different tautomeric forms, and all such forms are embraced within the scope of the invention. The term "tautomer" or "tautomeric form" refers to structural isomers of different energies which are interconvertible via a low energy barrier. For example, proton tautomers (also known as prototropic tautomers) include interconversions via migration of a proton, such as keto- enol and imine-enamine isomerizations. Valence tautomers include interconversions by reorganization of some of the bonding electrons. The present invention also embraces isotopically-labeled compounds of the present invention which are identical to those recited herein, but for the fact that one or more atoms are replaced by an atom having an atomic mass or mass number different from the atomic mass or mass number usually found in nature. Examples of isotopes that can be incorporated into compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, fluorine, iodine, and chlorine, such as 2H, 3H, 13C, 14C, 15N, 180, 170, 31P, 32P, 35S, 18F, 123l, and 36CI, respectively. Certain isotopically-labeled compounds of the present invention (e.g., those labeled with 3H and 14C) are useful in compound and/or substrate tissue distribution assays. Tritiated (i.e., 3H) and carbon-14 (i.e., 14C) isotopes are particularly preferred for their ease of preparation and detectability. Further, substitution with heavier isotopes such as deuterium (i.e., 2H) may afford certain therapeutic advantages resulting from greater metabolic stability (e.g., increased in vivo half-life or reduced dosage requirements) and hence may be preferred in some circumstances. Isotopically labeled compounds of the present invention can generally be prepared by following procedures analogous to those disclosed in the Schemes and/or in the Examples hereinbelow, by substituting an isotopically labeled reagent for a non-isotopically labeled reagent. Compounds of the present invention are useful for treating diseases, conditions and/or disorders modulated by cannabinoid receptor antagonists; therefore, another embodiment of the present invention is a pharmaceutical composition comprising a therapeutically effective amount of a compound of the present invention and a pharmaceutically acceptable excipient, diluent or carrier. A typical formulation is prepared by mixing a compound of the present invention and a carrier, diluent or excipient. Suitable carriers, diluents and excipients are well known to those skilled in the art and include materials such as carbohydrates, waxes, water soluble and/or swellable polymers, hydrophilic or hydrophobic materials, gelatin, oils, solvents, water, and the like. The particular carrier, diluent or excipient used will depend upon the means and purpose for which the compound of the present invention is being applied. Solvents are generally selected based on solvents recognized by persons skilled in the art as safe (GRAS) to be administered to a mammal. In general, safe solvents are non-toxic aqueous solvents such as water and other non-toxic solvents that are soluble or miscible in water. Suitable aqueous solvents include water, ethanol, propylene glycol, polyethylene glycols (e.g., PEG400, PEG300), etc. and mixtures thereof. The formulations may also include one or more buffers, stabilizing agents, surfactants, wetting agents, lubricating agents, emulsifiers, suspending agents, preservatives, antioxidants, opaquing agents, glidants, processing aids, colorants, sweeteners, perfuming agents, flavoring agents and other known additives to provide an elegant presentation of the drug (i.e., a compound of the present invention or pharmaceutical composition thereof) or aid in the manufacturing of the pharmaceutical product (i.e., medicament). The formulations may be prepared using conventional dissolution and mixing procedures. For example, the bulk drug substance (i.e., compound of the present invention or stabilized form of the compound (e.g., complex with a cyclodextrin derivative or other known complexation agent)) is dissolved in a suitable solvent in the presence of one or more of the excipients described above. The compound of the present invention is typically formulated into pharmaceutical dosage forms to provide an easily controllable dosage of the drug and to give the patient an elegant and easily handleable product. The pharmaceutical composition (or formulation) for application may be packaged in a variety of ways depending upon the method used for administering the drug. Generally, an article for distribution includes a container having deposited therein the pharmaceutical formulation in an appropriate form. Suitable containers are well-known to those skilled in the art and include materials such as bottles (plastic and glass), sachets, ampoules, plastic bags, metal cylinders, and the like. The container may also include a tamper-proof assemblage to prevent indiscreet access to the contents of the package. In addition, the container has deposited thereon a label that describes the contents of the container. The label may also include appropriate warnings. The present invention further provides a method of treating diseases, conditions and/or disorders modulated by cannabinoid receptor antagonists in an animal that includes administering to an animal in need of such treatment a therapeutically effective amount of a compound of the present invention or a pharmaceutical composition comprising an effective amount of a compound of the present invention and a pharmaceutically acceptable excipient, diluent, or carrier. The method is particularly useful for treating diseases, conditions and/or disorders modulated by cannabinoid receptor (in particular, CB1 receptor) antagonists. Preliminary investigations have indicated that the following diseases, conditions, and/or disorders are modulated by cannabinoid receptor antagonists: eating disorders (e.g., binge eating disorder, anorexia, and bulimia), weight loss or control (e.g., reduction in calorie or food intake, and/or appetite suppression), obesity, depression, atypical depression, bipolar disorders, psychoses, schizophrenia, behavioral addictions, suppression of reward-related behaviors (e.g., conditioned place avoidance, such as suppression of cocaine- and morphine-induced conditioned place preference), substance abuse, addictive disorders, impulsivity, alcoholism (e.g., alcohol abuse, addiction and/or dependence including treatment for abstinence, craving reduction and relapse prevention of alcohol intake), tobacco abuse (e.g., smoking addiction, cessation and/or dependence including treatment for craving reduction and relapse prevention of tobacco smoking), dementia (including memory loss, Alzheimer's disease, dementia of aging, vascular dementia, mild cognitive impairment, age-related cognitive decline, and mild neurocognitive disorder), sexual dysfunction in males (e.g., erectile difficulty), seizure disorders, epilepsy, inflammation, gastrointestinal disorders (e.g., dysfunction of gastrointestinal motility or intestinal propulsion), attention deficit disorder (ADD including attention deficit hyperactivity disorder (ADHD)), Parkinson's disease, and type II diabetes. Accordingly, the compounds of the present invention described herein are useful in treating diseases, conditions, or disorders that are modulated by cannabinoid receptor antagonists. Consequently, the compounds of the present invention (including the compositions and processes used therein) may be used in the manufacture of a medicament for the therapeutic applications described herein. Other diseases, conditions and/or disorders for which cannabinoid receptor antagonists may be effective include: premenstrual syndrome or late luteal phase syndrome, migraines, panic disorder, anxiety, post- traumatic syndrome, social phobia, cognitive impairment in non-demented individuals, non-amnestic mild cognitive impairment, post operative cognitive decline, disorders associated with impulsive behaviours (such as, disruptive behaviour disorders (e.g., anxiety/depression, executive function improvement, tic disorders, conduct disorder and/or oppositional defiant disorder), adult personality disorders (e.g. , borderline personality disorder and antisocial personality disorder), diseases associated with impulsive behaviours (e.g., substance abuse, paraphilias and self-mutilation), and impulse control disorders (e.g., intermittene explosive disorder, kleptomania, pyromania, pathological gambling, and trichotillomania)), obsessive compulsive disorder, chronic fatigue synd rome, sexual dysfunction in males (e.g., premature ejaculation), sexual dysfunction in females, disorders of sleep (e.g., sleep apnea), autism, mutism , neurodengenerative movement disorders, spinal cord injury, damage of the central nervous system (e.g., trauma), stroke, neurodegenerative diseases or toxic or infective CNS diseases (e.g., encephalitis or meningitis), cardiovascular disorders (e.g., thrombosis), and diabetes. The compounds of the present invention can be administered to a patient at dosage levels in the range of from about 0.7 mg to about 7,000 mg per day. For a normal adult human having a body weight of about 70 kg, a dosage in the range of from about 0.01 mg to about 100 mg per kilogram body weight is typically sufficient. However, some variability in the general dosage range may be required depending upon the age and weight of the subject being treated, the intended route of administration, the particular compound being administered and the like. The determination of dosage ranges and optimal dosages for a particular patient is well within the ability of one of ordinary skill in the art having the benefit of the instant disclosure. It is also noted that the compounds of the present invention can be used in sustained release, controlled release, and delayed release formulations, which forms are also well known to one of ordinary skill in the art. The compounds of this invention may also be used in conjunction with other pharmaceutical agents for the treatment of the diseases, conditions and/or disorders described herein. Therefore, methods of treatment that include administering compounds of the present invention in combination with other pharmaceutical agents are also provided. Suitable pharmaceutical agents that may be used in combination with the compounds of the present invention include anti-obesity agents such as apolipoprotein-B secretion/microsomal triglyceride transfer protein (apo-B/MTP) inhibitors, 1 1 β-hydroxy steroid dehydrogenase-1 (1 1 β-HSD type 1 ) inhibitors, peptide YY3-36 or analogs thereof, MCR-4 agonists, cholecystokinin-A (CCK-A) agonists, monoamine reuptake inhibitors (such as sibutramine), sympathomimetic agents, β3 adrenergic receptor agonists, dopamine agonists (such as bromocriptine), melanocyte-stimulating hormone receptor analogs, 5HT2c agonists, melanin concentrating hormone antagonists, leptin (the OB protein), leptin analogs, leptin receptor agonists, galanin antagonists, lipase inhibitors (such as tetrahydrolipstatin, i.e. orlistat), anorectic agents (such as a bombesin agonist), Neuropeptide-Y receptor antagonists (e.g., NPY Y5 receptor antagonists, such as the spiro compounds described in US Patent Nos. 6,566,367; 6,649,624; 6,638,942; 6,605,720; 6,495,559; 6,462,053; 6,388,077; 6,335,345; and 6,326,375; US Publication Nos. 2002/0151456 and 2003/036652; and PCT Publication Nos. WO 03/010175. WO 03/082190 and WO 02/048152), thyromimetic agents, dehydroepiandrosterone or an analog thereof, glucocorticoid receptor agonists or antagonists, orexin receptor antagonists, glucagon-like peptide-1 receptor agonists, ciliary neurotrophic factors (such as Axokine™ available from Regeneron Pharmaceuticals, Inc., Tarrytown, NY and Procter & Gamble Company, Cincinnati, OH), human agouti-related proteins (AGRP), ghrelin receptor antagonists, histamine 3 receptor antagonists or inverse agonists, neuromedin U receptor agonists and the like. Other anti-obesity agents, including the preferred agents set forth hereinbelow, are well known, or will be readily apparent in light of the instant disclosure, to one of ordinary skill in the art. Especially preferred are anti-obesity agents selected from the group consisting of orlistat, sibutramine, bromocriptine, ephedrine, leptin, pseudoephedrine, PYY3-36 or an analog thereof, and 2-oxo-N-(5- phenylpyrazinyl)spiro-[isobenzofuran-1(3H),4'-piperidine]-1 '-carboxamide. Preferably, compounds of the present invention and combination therapies are administered in conjunction with exercise and a sensible diet. Representative anti-obesity agents for use in the combinations, pharmaceutical compositions, and methods of the invention can be prepared using methods known to one of ordinary skill in the art, for example, sibutramine can be prepared as described in U.S. Pat. No. 4,929,629; bromocriptine can be prepared as described in U.S. Pat. Nos. 3,752,814 and 3,752,888; orlistat can be prepared as described in U.S. Pat. Nos.
5,274,143; 5,420,305; 5,540,917; and 5,643,874; PYY3-36 (including analogs thereof) can be prepared as described in US Publication No. 2002/0141985 and WO 03/027637; and the NPY Y5 receptor antagonist 2-oxo-N-(5- phenylpyrazinyl)spiro[isobenzofuran-1 (3H),4'-piperidine]-1 '-carboxamide can be prepared as described in US Publication No. 2002/0151456. Other useful NPY Y5 receptor antagonists include those described in PCT Publication No. 03/082190, such as 3-oxo-N-(5-phenyl-2-pyrazinyl)-spiro[isobenzofuran- 1 (3H), 4'-piperidine]-1 '-carboxamide; 3-oxo-N-(7-trifluoromethylpyrido[3,2- b]pyridin-2-yl)-spiro-[isobenzofuran-1 (3H), 4'-piperidine]-1 '-carboxamide; N- [5-(3-fluorophenyl)-2-pyrimidinyl]-3-oxospiro-[isobenzofuran-1 (3H), [4'- piperidine]-1 '-carboxamide; fraπs-3'-oxo-N-(5-phenyl-2-pyrimidinyl)] spiro[cyclohexane-1 ,1 '(3Η)-isobenzofuran]-4-carboxamide; ιfrar?s-3'-oxo-N- [1 -(3-quinolyl)-4-imidazolyl]spiro[cyclohexane-1 , 1 "(3'H)-isobenzofuran]-4- carboxamide; raπs-3-oxo-N-(5-phenyl-2-pyrazinyl)spiro[4-azaiso- benzofuran-1 (3H),1'-cyclohexane]-4'-carboxamide; trans- -[5~(3- fluorophenyl)-2-pyrimidinyl]-3-oxospiro[5-azaisobenzofuran-1 (3H), 1 '- cyclohexane]-4'-carboxamide; tjans-N-[5-(2-fluorophenyl)-2-pyrimidinyl]-3- oxospiro[5-azaisobenzofuran-1 (3H), 1'-cyclohexane]-4'-carboxamide; trans- N-[1-(3,5-difluorophenyl)-4-imidazolyl]-3-oxospiro[7-azaisobenzofuran- 1(3H),1'-cyclohexane]-4'-carboxamide; frans-3-oxo-N-(1-phenyl-4- pyrazolyl)spiro[4-azaisobenzofuran-1(3H),1'-cyclohexane]-4'-carboxamide; frans-N-[1-(2-fluorophenyl)-3-pyrazolyl]-3-oxospiro[6-azaisobenzofuran- 1 (3H),1 '-cyclohexane]-4'-carboxamide; rat?s-3-oxo-N-(l-phenyl-3- pyrazolyl)spiro[6-azaisobenzofuran-1 (3H),1'-cyclohexane]-4'-carboxamide; frans-3-oxo-N-(2-phenyl-1 ,2,3-triazol-4-yl)spiro[6-azaisobenzofuran-1 (3H),1'- cyciohexane]-4'-carboxamide; and pharmaceutically acceptable salts and esters thereof. All of the above recited U.S. patents and publications are incorporated herein by reference. Other suitable pharmaceutical agents that may be administered in combination with the compounds of the present invention include agents designed to treat tobacco abuse (e.g., nicotine receptor partial agonists, bupropion hypochloride (also known under the tradename Zyban™) and nicotine replacement therapies), agents to treat erectile dysfunction (e.g., dopaminergic agents, such as apomorphine), ADD/ADHD agents (e.g., Ritalin™, Strattera™, Concerta™ and Adderall™), and agents to treat alcoholism, such as opioid antagonists (e.g., naltrexone (also known under the tradename ReVia™) and nalmefene), disulfiram (also known under the tradename Antabuse™), and acamprosate (also known under the tradename Campral™)). In addition, agents for reducing alcohol withdrawal symptoms may also be co-administered, such as benzodiazepines, beta- blockers, clonidine, carbamazepine, pregabalin, and gabapentin
(Neurontin™). Treatment for alcoholism is preferably administered in combination with behavioral therapy including such components as motivational enhancement therapy, cognitive behavioral therapy, and referral to self-help groups, including Alcohol Anonymous (AA). Other pharmaceutical agents that may be useful include antihypertensive agents; anti-inflammatory agents (e.g., COX-2 inhibitors); antidepressants (e.g., fluoxetine hydrochloride (Prozac™)); cognitive improvement agents (e.g., donepezil hydrochloride (Aircept™) and other acetylcholinesterase inhibitors); neuroprotective agents (e.g., memantine); antipsychotic medications (e.g., ziprasidone (Geodon™), risperidone
(Risperdal™), and olanzapine (Zyprexa™)); insulin and insulin analogs (e.g., LysPro insulin); GLP-1 (7-37) (insulinotropin) and GLP-1 (7-36)-NH2; sulfonylureas and analogs thereof: chlorpropamide, glibenclamide, tolbutamide, tolazamide, acetohexamide, Glypizide®, glimepiride, repaglinide, meglitinide; biguanides: metformin, phenformin, buformin; α2- antagonists and imidazolines: midaglizole, isaglidole, deriglidole, idazoxan, efaroxan, fluparoxan; other insulin secretagogues: linogliride, A-4166; glitazones: ciglitazone, Actos® (pioglitazone), englitazone, troglitazone, darglitazone, Avandia® (BRL49653); fatty acid oxidation inhibitors: clomoxir, etomoxir; α-glucosidase inhibitors: acarbose, miglitol, emiglitate, voglibose, MDL-25,637, camiglibose, MDL-73,945; β-agonists: BRL 35135, BRL 37344, RO 16-8714, ICI D7114, CL 316,243; phosphodiesterase inhibitors: L- 386,398; lipid-lowering agents: benfluorex: fenfluramine; vanadate and vanadium complexes (e.g., Naglivan®) and peroxovanadium complexes; amylin antagonists; glucagon antagonists; gluconeogenesis inhibitors; somatostatin analogs; antilipolytic agents: nicotinic acid, acipimox, WAG 994, pramlintide (Symlin™), AC 2993, nateglinide, aldose reductase inhibitors (e.g., zopolrestat), glycogen phosphorylase inhibitors, sorbitol dehydrogenase inhibitors, sodium-hydrogen exchanger type 1 (NHE-1) inhibitors and/or cholesterol biosynthesis inhibitors or cholesterol absorption inhibitors, especially a HMG-CoA reductase inhibitor, or a HMG-CoA synthase inhibitor, or a HMG-CoA reductase or synthase gene expression inhibitor, a CETP inhibitor, a bile acid sequesterant, a fibrate, an ACAT inhibitor, a squalene synthetase inhibitor, an anti-oxidant or niacin. The compounds of the present invention may also be administered in combination with a naturally occurring compound that acts to lower plasma cholesterol levels. Such naturally occurring compounds are commonly called nutraceuticals and include, for example, garlic extract, Hoodia plant extracts, and niacin. The dosage of the additional pharmaceutical agent will also be generally dependent upon a number of factors including the health of the subject being treated, the extent of treatment desired, the nature and kind of concurrent therapy, if any, and the frequency of treatment and the nature of the effect desired. In general, the dosage range of an anti-obesity agent is in the range of from about O.001 mg to about 100 mg per kilogram body weight of the individual per day, preferably from about 0.1 mg to about 10 mg per kilogram body weight of the individual per day. However, some variability in the general dosage range may also be required depending upon the age and weight of the subject being treated, the intended route of administration, the particular anti-obesity agent being administered and the like. The determination of dosage ranges and optimal dosages for a particular patient is also well within the ability of one of ordinary skill in the art having the benefit of the instant disclosure. According to the methods of the invention, a compound of the present invention or a combination of a compound of the present invention and at least one additional pharmaceutical agent is administered to a subject in need of such treatment, preferably in the form of a pharmaceutical composition. In the combination aspect of the invention, the compound of the present invention and at least one other pharmaceutical agent may be administered either separately or in the pharmaceutical composition comprising both. It is generally preferred that such administration be oral. However, if the subject being treated is unable to swallow, or oral administration is otherwise impaired or undesirable, parenteral or transdermal administration may be appropriate. According to the methods of the invention, when a combination of a compound of the present invention and at least one other pharmaceutical agent are administered together, such administration can be sequential in time or simultaneous with the simultaneous method being generally preferred. For sequential administration, a compound of the present invention and the additional pharmaceutical agent can be administered in any order. It is generally preferred that such administration be oral. It is especially preferred that such administration be oral and simultaneous. When a compound of the present invention and the additional pharmaceutical agent are administered sequentially, the administration of each can be by the same or by different methods. According to the methods of the invention, a compound of the present invention or a combination of a compound of the present invention and at least one additional pharmaceutical agent (referred to herein as a
"combination") is preferably administered in the form of a pharmaceutical composition. Accordingly, a compound of the present invention or a combination can be administered to a patient separately or together in any conventional oral, rectal, transdermal, parenteral, (for example, intravenous, intramuscular, or subcutaneous) intracisternal, intravaginal, intraperitoneal, intravesical, local (for example, powder, ointment or drop), or buccal, or nasal, dosage form. Compositions suitable for parenteral injection generally include pharmaceutically acceptable sterile aqueous or nonaqueous solutions, dispersions, suspensions, or emulsions, and sterile powders for reconstitution into sterile injectable solutions or dispersions. Examples of suitable aqueous and nonaqueous carriers, diluents, solvents, or vehicles include water, ethanol, polyols (propylene glycol, polyethylene glycol, glycerol, and the like), suitable mixtures thereof, vegetable oils (such as olive oil) and injectable organic esters such as ethyl oleate. Proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersions, and by the use of surfactants. These compositions may also contain adjuvants such as preserving, wetting, emulsifying, and dispersing agents. Prevention of microorganism contamination of the compositions can be accomplished with various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, and the like. It may also be desirable to include isotonic agents, for example, sugars, sodium chloride, and the like. Prolonged absorption of injectable pharmaceutical compositions can be brought about by the use of agents capable of delaying absorption, for example, aluminum monostearate and gelatin. Solid dosage forms for oral administration include capsules, tablets, powders, and granules. In such solid dosage forms, a compound of the present invention or a combination is admixed with at least one inert customary pharmaceutical excipient (or carrier) such as sodium citrate or dicalcium phosphate or (a) fillers or extenders (e.g., starches, lactose, sucrose, mannitol, silicic acid and the like); (b) binders (e.g., carboxymethylcellulose, alginates, gelatin, polyvinylpyrrolidone, sucrose, acacia and the like); (c) humectants (e.g., glycerol and the like); (d) disintegrating agents (e.g., agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain complex silicates, sodium carbonate and the like); (e) solution retarders (e.g., paraffin and the like); (f) absorption accelerators (e.g., quaternary ammonium compounds and the like); (g) wetting agents (e.g., cetyl alcohol, glycerol monostearate and the like); (h) adsorbents (e.g., kaolin, bentonite and the like); and/or (i) lubricants (e.g., talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate and the like). In the case of capsules and tablets, the dosage forms may also comprise buffering agents. Solid compositions of a similar type may also be used as fillers in soft or hard filled gelatin capsules using such excipients as lactose or milk sugar, as well as high molecular weight polyethylene glycols, and the like. Solid dosage forms such as tablets, dragees, capsules, and granules can be prepared with coatings and shells, such as enteric coatings and others well known in the art. They may also contain opacifying agents, and can also be of such composition that they release the compound of the present invention and/or the additional pharmaceutical agent in a delayed manner. Examples of embedding compositions that can be used are polymeric substances and waxes. The drug can also be in micro- encapsulated form, if appropriate, with one or more of the above-mentioned excipients. Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, solutions, suspensions, syrups, and elixirs. In addition to the compound of the present invention or the combination, the liquid dosage form may contain inert diluents commonly used in the art, such as water or other solvents, solubilizing agents and emulsifiers, as for example, ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1 ,3-butylene glycol, dimethylformamide, oils (e.g., cottonseed oil, groundnut oil, corn germ oil, olive oil, castor oil, sesame seed oil and the like), glycerol, tetrahydrofurfuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, or mixtures of these substances, and the like. Besides such inert diluents, the composition can also include adjuvants, such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents. Suspensions, in addition to the compound of the present invention or the combination, may further comprise suspending agents, e.g., ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar, and tragacanth, or mixtures of these substances, and the like. Compositions for rectal or vaginal administration preferably comprise suppositories, which can be prepared by mixing a compound of the present invention or a combination with suitable non-irritating excipients or carriers, such as cocoa butter, polyethylene glycol or a suppository wax which are solid at ordinary room temperature but liquid at body temperature and therefore melt in the rectum or vaginal cavity thereby releasing the active component(s). Dosage forms for topical administration of the compounds of the present invention and combinations of the compounds of the present invention with anti-obesity agents may comprise ointments, powders, sprays and inhalants. The drugs are admixed under sterile condition with a pharmaceutically acceptable carrier, and any preservatives, buffers, or propellants that may be required. Ophthalmic formulations, eye ointments, powders, and solutions are also intended to be included within the scope of the present invention. The following paragraphs describe exemplary formulations, dosages, etc. useful for non-human animals. The administration of the compounds of the present invention and combinations of the compounds of the present invention with anti-obesity agents can be effected orally or non-orally (e.g., by injection). An amount of a compound of the present invention or combination of a compound of the present invention with an anti-obesity agent is administered such that an effective dose is received. Generally, a daily dose that is administered orally to an animal is between about 0.01 and about 1 ,000 mg/kg of body weight, preferably between about 0.01 and about 300 mg/kg of body weight. Conveniently, a compound of the present invention (or combination) can be carried in the drinking water so that a therapeutic dosage of the compound is ingested with the daily water supply. The compound can be directly metered into drinking water, preferably in the form of a liquid, water- soluble concentrate (such as an aqueous solution of a water-soluble salt). Conveniently, a compound of the present invention (or combination) can also be added directly to the feed, as such, or in the form of an animal feed supplement, also referred to as a premix or concentrate. A premix or concentrate of the compound in a carrier is more commonly employed for the inclusion of the agent in the feed. Suitable carriers are liquid or solid, as desired, such as water, various meals such as alfalfa meal, soybean meal, cottonseed oil meal, linseed oil meal, corncob meal and corn meal, molasses, urea, bone meal, and mineral mixes such as are commonly employed in poultry feeds. A particularly effective carrier is the respective animal feed itself; that is, a small portion of such feed. The carrier facilitates uniform distribution of the compound in the finished feed with which the premix is blended. Preferably, the compound is thoroughly blended into the premix and, subsequently, the feed. In this respect, the compound may be dispersed or dissolved in a suitable oily vehicle such as soybean oil, corn oil, cottonseed oil, and the like, or in a volatile organic solvent and then blended with the carrier. It will be appreciated that the proportions of compound in the concentrate are capable of wide variation since the amount of the compound in the finished feed may be adjusted by blending the appropriate proportion of premix with the feed to obtain a desired level of compound. High potency concentrates may be blended by the feed manufacturer with proteinaceous carrier such as soybean oil meal and other meals, as described above, to produce concentrated supplements, which are suitable for direct feeding to animals. In such instances, the animals are permitted to consume the usual diet. Alternatively, such concentrated supplements may be added directly to the feed to produce a nutritionally balanced, finished feed containing a therapeutically effective level of a compound of the present invention. The mixtures are thoroughly blended by standard procedures, such as in a twin shell blender, to ensure homogeneity. If the supplement is used as a top dressing for the feed, it likewise helps to ensure uniformity of distribution of the compound across the top of the dressed feed. Drinking water and feed effective for increasing lean meat deposition and for improving lean meat to fat ratio are generally prepared by mixing a compound of the present invention with a sufficient amount of animal feed to provide from about 10"3 to about 500 ppm of the compound in the feed or water. The preferred medicated swine, cattle, sheep and goat feed generally contain from about 1 to about 400 grams of a compound of the present invention (or combination) per ton of feed, the optimum amount for these animals usually being about 50 to about 3O0 grams per ton of feed. The preferred poultry and domestic pet feeds usually contain about 1 to about 400 grams and preferably about 1 0 to about 400 grams of a compound of the present invention (or combination) per ton of feed. For parenteral administration in animals, the compounds of the present invention (or combination) may be prepared in the form of a paste or a pellet and administered as an implant, usually under the skin of the head or ear of the animal in which increase in lean meat deposition and improvement in lean meat to fat ratio is sought. In general, parenteral administration involves injection of a sufficient amount of a compound of the present invention (or combination) to provide the animal with about 0.01 to about 20 mg/kg/day of body weight of the drug. The preferred dosage for poultry, swine, cattle, sheep, goats and domestic pets is in the range of from about 0.05 to about 10 mg/kg/day of body weight of drug. Paste formulations can be prepared by dispersing the drug in a pharmaceutically acceptable oil such as peanut oil, sesame oil, corn oil or the like. Pellets containing an effective amount of a compound of the present invention, pharmaceutical composition, or combination can be prepared by admixing a compound of the present invention or combination with a diluent such as carbowax, carnuba wax, and the like, and a lubricant, such as magnesium or calcium stearate, can be added to improve the pelleting process. It is, of course, recognized that more than one pellet may be administered to an animal to achieve the desired dose level which will provide the increase in lean meat deposition and improvement in lean meat to fat ratio desired. Moreover, implants may also be made periodically during the animal treatment period in order to maintain the proper drug level in the animal's body. The present invention has several advantageous veterinary features. For the pet owner or veterinarian who wishes to increase leanness and/or trim unwanted fat from pet animals, the instant invention provides the means by which this may be accomplished. For poultry, beef, and swine breeders, utilization of the method of the present invention yields leaner animals that command higher sale prices from the meat industry. Embodiments of the present invention are illustrated by the following Examples. It is to be understood, however, that the embodiments of the invention are not limited to the specific details of these Examples, as other variations thereof will be known, or apparent in light of the instant disclosure, to one of ordinary skill in the art. EXAMPLES Unless specified otherwise, starting materials are generally available from commercial sources such as Aldrich Chemicals Co. (Milwaukee, Wl), Lancaster Synthesis, Inc. (Windham, NH), Acros Organics (Fairlawn, NJ), Maybridge Chemical Company, Ltd. (Cornwall, England), Tyger Scientific (Princeton, NJ), and AstraZeneca Pharmaceuticals (London, England). General Experimental Procedures NMR spectra were recorded on a Varian Unity™ 400 (available from
Varian Inc., Palo Alto, CA) at room temperature at 400 MHz for proton. Chemical shifts are expressed in parts per million (δ) relative to residual solvent as an internal reference. The peak shapes are denoted as follows: s, singlet; d, doublet; t, triplet; q, quartet; m, multiplet; bs, broad singlet; 2s, two singlets. Atmospheric pressure chemical ionization mass spectra (APCI) were obtained on a Fisons™ Platform II Spectrometer (carrier gas: acetonitrile: available from Micromass Ltd, Manchester, UK). Chemical ionization mass spectra (CI) were obtained on a Hewlett-Packard™ 5989 instrument (ammonia ionization, PBMS: available from Hewlett-Packard Company, Palo Alto, CA). Electrospray ionization mass spectra (ES) were obtained on a Waters™ ZMD instrument (carrier gas: acetonitrile: available from Waters Corp., Milford, MA). Where the intensity of chlorine or bromine-containing ions are described, the expected intensity ratio was observed (approximately 3:1 for 35CI/37CI-containing ions and 1:1 for 79Br/81Br-containing ions) and the intensity of only the lower mass ion is given. In some cases only representative 1H NMR peaks are given. MS peaks are reported for all examples. Optical rotations were determined on a PerkinElmer™ 241 polarimeter (available from PerkinElmer Inc., Wellesley, MA) using the sodium D line (λ = 589 nm) at the indicated temperature and are reported as follows [α]D temp, concentration (c = g/100 ml), and solvent. Column chromatography was performed with either Baker™ silica gel (40 μm; J.T. Baker, Phillipsburg, NJ) or Silica Gel 50 (EM Sciences™, Gibbstown, NJ) in glass columns or in Flash 40 Biotage™ columns (ISC, Inc., Shelton, CT) under low nitrogen pressure. Preparation of Key Intermediates
Preparation of Intermediate 6~Chloro-N4-(4-chlorophenyl)-pyrimidine- 4.5-diamine (l-1A-1a):
Figure imgf000047_0001
l-1A-1a 5-Amino-4,6-dichloropyrimidine (5.00 g, 29 mmol) and 4-chloroaniline
(4.71 g, 36 mmol) were suspended in 80 ml H20 and 12 ml ethanol. Concentrated HCI (1.2 ml, 14.5 mmol) was added at room temperature followed by warming reaction to 82 °C. After stirring for 19 hours, the reaction was cooled to room temperature and stirred for 60 hours. The precipitate was collected on a sintered glass funnel and rinsed with water followed by hexanes. After drying under vacuum, l-1A-1a was obtained as an off-white solid (7.38 g, 98%): +ESI MS (M+1 ) 255.3; 1H NMR: (400 MHz, CD3OD): δ 7.87 (s, 1 H), 7.66 (d, J = 8.7 Hz, 2 H), 7.30 (d, J = 8.7 Hz, 2 H).
Preparation of Intermediate 2-Chloro-N-r4-chloro-6-(4-chlorophenylamino)- pyrimidin-5-yll-benzamide (l-1A-1b):
Figure imgf000048_0001
l-1A-1b 6-Chloro-N4-(4-chlorophenyl)-pyrimidine-4,5-diamine l-1A-1a (1.00 g, 3.92 mmol) was dissolved in 6 ml of Λ/,Λ/-dimethylacetamide giving a clear brown solution. After cooling to 5 °C, neat 2-chlorobenzoyl chloride (0.80 g, 4.34 mmol) was added over 1 minute. The solution was warmed to room temperature and stirred for 4 hours. Addition of water (15 ml) caused a white precipitate to come out of solution. The mixture was stirred for an additional 30 minutes at room temperature, then the precipitate was collected by vacuum filtration and rinsed with H2O and then hexanes. The solid was further dried under vacuum to give 1-1 A-1 b as a colorless solid (1.27 g, 82%): +APCI MS (M+1) 393.1; 1H NMR (400 MHz, DMSO-d6) δ 10.02 (s, 1 H), 9.11 (s, 1 H), 8.40 (s, 1 H), 7.93 (dd, J = 7.4, 1.6 Hz, 1 H), 7.66- 7.40 (m, 7H).
Preparation of Intermediate 9-(4-Chlorophenyl)-8-(2-chlorophenyl)-9H-pu n- 6-0I (I-1A-1C-1):
Figure imgf000048_0002
l-1A-1c-1 To a suspension of 2-chloro-N-[4-chloro-6-(4-chlorophenylamino)- pyrimidin-5-yl]-benzamide 1-1 A-1 b (1.00 g, 2.54 mmol) in isopropanol (20 ml) was added neat H2S04 (410 μl, 7.4 mmol) at room temperature. The reaction was refluxed for 8 hours, followed by cooling to room temperature and stirring for 16 hours. To the heterogeneous solution was added 20 ml of water to promote further product precipitation. After stirring for an additional hour at room temperature, the solid was collected on a sintered glass funnel and rinsed with water followed by hexanes. The product was further dried under reduced pressure to give 1-1 A-1 c-1 as a colorless solid (0.72 g, 80%): 1H NMR: (400 MHz, DMSO-d6): δ 12.57 (s, 1 H), 8.08 (d, J = 4.1 Hz, 1 H), 7.66 (dd, J = 7.4, 1.2 Hz, 1 H), 7.51-7.41 (m, 5H), 7.33-7.29 (m, 2H).
The following intermediate was prepared from the appropriately substituted aniline and benzoyl chloride derivatives by the procedures described above: 9-(4-Chloro-phenyl)-8-(2,4-dichloro-phenyl)-9H-purin-6-ol (l-1A-1c-2).
Preparation of Intermediate 2-Chloro-N-(4-chloro-phenyl)-benzamidine (1-1 A- 2a):
Figure imgf000049_0001
(l-1A-2a) Trimethylaluminum (2M in hexanes, 100 ml, 200 mmol) was added dropwise to a solution of 4-chloro-phenylamine (18.2 g, 143 mmol) in toluene (550 ml) under a N2 atmosphere at 0°C. The reaction mixture was warmed to room temperature and stirred for 3.5 hours. A solution of 2- chlorobenzonitrile (23.6 g, 171 mmol) in toluene (140 ml) was slowly added. The heterogeneous mixture was heated at 80°C for 17 hours, cooled to room temperature, diluted with CHCIs/methanol (2:1 ), and poured over a slurry of silica gel. The filter cake was washed with additional CH2CI2/MeOH (2:1). The combined filtrates were concentrated in vacuo, and the resulting yellow solid was triturated with hexanes/ether (2:1 ). The product, 2-chloro-N- (4-chloro-phenyl)-benzamidine l-1A-2a. (25.1 g, 66%) was used without further purification.
Preparation of lntermediate1-(4-Chloro-phenyl)-2-(2-chloro-phenyl)-4- hvdroxy-4,5-dihydro-1H-imidazole-4-carboxyHc acid ethyl ester (l-1A-2b):
Figure imgf000050_0001
(l-1A-2b) A mixture of 2-chloro-N-(4-chloro-phenyl)-benzamidine (l-1A-2a. 25.1 g, 95 mmol) and NaHC03 (84 g, 189 mmol) in 2-propanol (473 ml) was treated with 3-bromo-2-oxo-propionic acid ethyl ester (14.3 ml, 22 g, 113 mmol). The reaction mixture was heated at 80 °C for 17 hours. After cooling to room temperature, the solvent was removed in vacuo. The residue was diluted with CH2Cl2 and the organic solution was washed with H20, dried over MgSO4, and concentrated in vacuo to provide the product, 1-(4-chloro-phenyl)-2-(2-chloro-phenyl)-4-hydroxy-4,5-dihydro-1 H-imidazole- 4-carboxylic acid ethyl ester l-1A-2b, as a dark red residue (36 g).
Preparation of 1-(4-Chloro-phenyl)-2-(2-chloro-phenyl)-1 H-imidazole-4- carboxylic acid ethyl ester (l-1A-2c):
Figure imgf000051_0001
(l-1A-2c) The crude 1 -(4-chloro-phenyl)-2-(2-chloro-phenyl)-4-hydroxy-4,5- dihydro-1 H-imidazole-4-carboxylic acid ethyl ester (l-1A-2b. 36 g, 94.7 mmol) obtained from the previous step and p-toluenesulfonic acid monohydrate (4.5 g, 24 mmol) in toluene (630 ml) was heated under reflux for 17 hours. The reaction mixture was cooled to room temperature and concentrated in vacuo. The crude residue was taken up in CH2CI2 and the organic solution was washed with H20, sat'd aq. NaHC0 , and saturated aqueous NaCl, dried over MgS04 and concentrated in vacuo. The crude residue was purified by silica gel plug filtration using a solvent gradient of 2% EtOAc/CH2CI2 to 10% EtOAc/CH2CI2. The fractions containing the product were concentrated, and the oily residue was diluted with 1 :3 EtOAc/hexanes (200 ml). After 1 hour, a solid precipitated out of solution and was collected by filtration to provide the product, 1-(4-chloro-phenyl)-2-(2-chloro-phenyl)- 1 H-imidazole-4-carboxylic acid ethyl ester 1-1 A-2c (18.63 g, 69.7%).
Preparation of 5-Bromo-1-(4-chloro-phenyl)-2-(2-chloro-phenyl)-1 H- imidazole-4-carboxylic acid ethyl ester (l-1A-2d):
(l-1A-2d) Bromine (3.6 ml, 0.07 mol) was added to a solution of 1 -(4-chloro-phenyl)-2- (2-chloro-phenyl)-1 H-imidazole-4-carboxylic acid ethyl ester (l-1A-2c. 3.6 g, 0.01 mol) in glacial acetic acid (50 ml) at room temperature. The reaction mixture was stirred for 17 hours, poured over ice-water, and treated with 25% aqueous NaOH until a yellow color was formed. The aqueous solution was extracted with CH2CI2 (3x), and the combined extracts were dried and concentrated in vacuo to give the desired compound, 5-bromo-1-(4-chloro- phenyl)-2-(2-chloro-phenyl)-1 H-imidazole-4-carboxylic acid ethyl ester 1-1 A- 2d, as an oil (4.7 g): +APCI MS (M+1) 441.1 ; 1H NMR (CD3CI) δ 1.41 (3H, t), 4.43 (2H, q), 7.08-7.12 (2H, m), 7.20 - 7.40 (7H, m). Example 1 Preparation of 9-(4-Chlorophenyl)-8-(2-chlorophenyl)-1 -isopropyl-1.9- dihydropurin-6-one (1A-1):
Figure imgf000052_0001
1A-1 9-(4-Chlorophenyl)-8-(2-chlorophenyl)-9H-purin-6-ol (l-1A-1c-1 : 50 mg, 0.14 mmol), 2-iodopropane (26 mg, 0.15 mmol), and cesium carbonate (50 mg, 0.15 mmol) were combined in dimethylformamide (0.7 ml) and stirred overnight. Additional 2-iodopropane (13 mg, 0.76 mmol), and cesium carbonate (25 mg, 0.76 mmol) were added and stirred an additional day. The reaction mixture was diluted with ethyl ether and then washed with saturated aqueous sodium bicarbonate and brine. The organic layer was dried (Na2S04), evaporated to dryness, and then purified on a Biotage™ Flash 12S column using 0-70% ethyl acetate in hexanes as eluant to afford title compound 1A-1 (30 mg, 53%); 1H NMR: +ESI MS (M+1 ) 399.4; (400 MHz, CD2CI2) δ 8.07 (s, 1 H), 7.54 (dd, J = 7.5, 1.2 Hz, 1 H), 7.45-7.35 (m, 5H),
7.19 (d, J = 8.7 Hz, 2H), 5.28 (septuplet, J = 6.8 Hz, 1 H), 1.49 (d, J = 7.1 Hz,
6H). The compounds listed in Table 1 below were prepared using procedures analogous to those described above for the synthesis of Compound 1A-1 using the appropriate starting materials which are available commercially, prepared using preparations well-known to those skilled in the art, or prepared in a manner analogous to routes described above for other intermediates. The compounds listed below were isolated initially as the free base and may be converted to their corresponding hydrochloride salt for testing. Table 1
Figure imgf000053_0001
Figure imgf000053_0002
Figure imgf000054_0001
Figure imgf000055_0002
Example 2 Preparation of 9-(4-Chlorophen yl)-8-(2, 4-dichlorophen yl)-1 -(tetra- hydrofuran-2-ylmethyl)-1,9-dihvdropurin-6-one (2A-1):
Figure imgf000055_0001
2A-1 9-(4-Chlorophenyl)-8-(2,4-dichlorophenyl)-9H-purin-6-ol (l-1A-1c-2. 100 mg, 0.255 mmol), 2-bromomethyl-tetrahydrofuran (42 mg, 0.255 mmol), and cesium carbonate (83 mg, 0.26 mmol) were combined in dimethylformamide (5 ml) and heated to 100 °C overnight. The reaction mixture was diluted with ethyl acetate and washed with saturated aqueous NaCl. The organic layer was dried (Na2S0 ), filtered, and evaporated to dryness. The crude material was purified on a preparative TLC plate using 75% ethyl acetate/hexanes as the eluant to yield the title compound, 9-(4- chlorophenyl)-8-(2,4-dichlorophenyl)-1 -(tetrahydrofuran-2-ylmethyl)-1 ,9- dihydropurin-6-one (2A-1 : 100 mg, 82%): +APCI MS (M+1 ) 512.2; 1H NMR (400 MHz, CD3OD) δ 8.22 (s, 1 H), 7.61 (d, 1 H), 7.50 (s, 1 H), 7.42 (d, 1 H), 7.41 (d, 2H), 7.29 (d, 2H), 4.40 (d, 1 H), 4.21 (d, 1 H), 3.98 (m, 1 H), 3.87 (m, 1 H), 3.75 (m, 1 H), 2.10 (m, 1 H), 1.95 (m, 2H), 1.68 (m, 1 H). A solution of the above material in methylene chloride (~1 ml) was treated with 4 M HCI/dioxane (-0.5 ml), concentrated in vacuo, and dried under high vacuum to yield the hydrochloride salt of compound 2A-1 : +APCI MS (M+1 ) 512.2. The compounds listed in Table 2 below were prepared using procedures analogous to those described above for the synthesis of Compound 2A-1 using the appropriate starting materials which are available commercially, prepared using preparations well-known to those skilled in the art, or prepared in a manner analogous to routes described above for other intermediates. The compounds listed below were isolated initially as the free base and then generally converted to their corresponding hydrochloride salt for testing. Table 2
Figure imgf000056_0001
Figure imgf000056_0002
Figure imgf000057_0001
Figure imgf000058_0001
Figure imgf000059_0001
Figure imgf000060_0001
Figure imgf000061_0002
Example 3 Preparation of 2-r9-(4-Chloro-phenyl)-6-oxo-1-(2.2.2-trifluoro-ethyl)- 6,9-dihvdro-1H-purin-8-yll-benzonitrile (3A-1):
Figure imgf000061_0001
3A-1
A mixture of 8-(2-bromo-phenyl)-9-(4-chloro-phenyl)-1-(2,2,2-trifluoro-ethyl)- 1 ,9-dihydro-purin-6-one (2A-67. 50 mg, 0.103 mmol), Zn(CN)2 (18 mg, 0.155 mmol), Pd(PPh3) (12 mg, 0.01 mmol) in DMF (0.5 ml) was heated in a microwave apparatus (Emrys Optimizer, Personal Chemistry) at 200 °C for 3 minutes. The reaction mixture was quenched with sat'd aqueous NaCl and extracted with EtOAc (2x). The combined organic extracts were dried and concentrated under vacuum. The crude residue was purified via HPLC (Shimadzu) using a solvent gradient of 30% CH3CN/hexanes to 100% CH3CN to give the desired product 2-[9-(4-chloro-phenyl)-6-oxo-1 -(2,2,2- trifluoro-ethyl)-6,9-dihydro-1H-purin-8-yl]-benzonitrile 3A-1 (20 mg): +APCI MS (M+1 ) 430.1 ; 1H NMR (400 MHz, CDCI3) δ 8.05 (s, 1 H), 7.68 (d, 2H, J = 8.7 Hz), 7.62 (dd, 1 H), 7.46 - 7.25 (m, 5H), 4.8 (q, 2H). The compound listed in Table 3 below was prepared using procedures analogous to those described above for the synthesis of Compound 3A-1 using the appropriate starting materials which are available commercially, prepared using preparations well-known to those skilled in the art, or prepared in a manner analogous to routes described above for other intermediates. The compound listed below was isolated initially as the free base and then generally converted to its corresponding hydrochloride salt for testing. Table 3
Figure imgf000062_0001
Figure imgf000062_0002
PHARMACOLOGICAL TESTING The utility of the compounds of the present invention in the practice of the instant invention can be evidenced by activity in at least one of the protocols described hereinbelow. The following acronyms are used in the protocols described below. BSA - bovine serum albumin DMSO - dimethylsulfoxide EDTA - ethylenediamine tetracetic acid PBS - phosphate-buffered saline EGTA - ethylene glycol-b/s(β-aminoethyl ether) N,N,N',N'-tetraacetic acid GDP - guanosine diphosphate sc - subcutaneous po - orally ip - intraperitoneal icv - intra cerebro ventricular iv - intravenous
[3H]-SR141716A - tritiated N-(piperidin-1-yl)-5-(4-chlorophenyl)-1-(2,4- dichlorophenyl)-4-methyl-1 H-pyrazole-3-carboxamide hydrochloride available from Amersham Biosciences, Piscataway, NJ. [3H]-CP-55940 - radiolabled 5-(1 ,1-dimethylheptyl)-2-[5-hydroxy-2-(3- hydroxypropyl)-cyclohexyl]-phenol available from NEN Life Science Products, Boston, MA. AM251 - N -(piperidin-1-yl)-1-(2,4-dichlorophenyl)-5-(4-iodophenyl)-4- methyl-1 H-pyrazole-3-carboxamide available from Tocris™, Ellisville, MO. Compounds having an activity <10 nM are generally tested in the
CB1-R GTPγ [35S] Binding Assay and the CB2-R binding assay described below in the Biological Binding Assays section. Selected compounds are then tested in vivo using one or more of the functional assays described in the Biological Functional Assays section below. The compounds provided a range of binding activities from 0.5 to 5000 nM In Vitro Biological Assays Bioassay systems for determining the CB-1 and CB-2 receptor binding properties and pharmacological activity of cannabinoid receptor ligands are described by Roger G. Pertwee in "Pharmacology of Cannabinoid Receptor Ligands" Current Medicinal Chemistry. 6, 635-664 (1999) and in WO
92/02640 (U.S. Application No. 07/564,075 filed August 8, 1990, incorporated herein by reference). The following assays were designed to detect compounds that inhibit the binding of [3H]-SR141716A (selective radiolabeled CB-1 ligand) and [3H]- 5-(1,1-dimethylheptyl)-2-[5-hydroxy-2-(3-hydroxypropyl)-cyclohexyl]-phenol; radiolabeled CB-1 /CB-2 ligand) to their respective receptors. Rat CB-1 Receptor Binding Protocol PelFreeze brains (available from Pel Freeze Biologicals, Rogers, Arkansas) were cut up and placed in tissue preparation buffer (5 mM Tris HCI, pH = 7.4 and 2 mM EDTA), polytroned at high speed and kept on ice for 15 minutes. The homogenate was then spun at 1 ,000 X g for 5 minutes at 4°C. The supernatant was recovered and centrifuged at 100,000 X G for 1 hour at 4°C. The pellet was then re-suspended in 25 ml of TME (25 nM Tris, pH = 7.4, 5 mM MgCI2, and 1 mM EDTA) per brain used. A protein assay was performed and 200 μl of tissue totaling 20 μg was added to the assay. The test compounds were diluted in drug buffer (0.5% BSA, 10% DMSO and TME) and then 25 μl were added to a deep well polypropylene plate. [3H]-SR141716A was diluted in a ligand buffer (0.5% BSA plus TME) and 25 μl were added to the plate. A BCA protein assay was used to determine the appropriate tissue concentration and then 200 μl of rat brain tissue at the appropriate concentration was added to the plate. The plates were covered and placed in an incubator at 20°C for 60 minutes. At the end of the incubation period 250 μl of stop buffer (5% BSA plus TME) was added to the reaction plate. The plates were then harvested by Skatron onto GF/B filtermats presoaked in BSA (5 mg/ml) plus TME. Each filter was washed twice. The filters were dried overnight. In the morning the filters were counted on a Wallac Betaplate™ counter (available from PerkinElmer Life Sciences™, Boston, MA). Human CB-1 Receptor Binding Protocol Human embryonic kidney 293 (HEK 293) cells transfected with the CB-
1 receptor cDNA (obtained from Dr. Debra Kendall, University of Connecticut) were harvested in homogenization buffer (10 mM EDTA, 10 mM EGTA, 10 mM Na bicarbonate, protease inhibitors; pH = 7.4), and homogenized with a Dounce Homogenizer. The homogenate was then spun at 1 ,000X g for 5 minutes at 4°C. The supernatant was recovered and centrifuged at 25.000X G for 20 minutes at 4°C. The pellet was then re-suspended in 10 ml of homogenization buffer and re-spun at 25.000X G for 20 minutes at 4°C. The final pellet was re-suspended in 1 ml of TME (25 mM Tris buffer (pH = 7.4) containing 5 mM MgCI2 and 1 mM EDTA). A protein assay was performed and 200 μl of tissue totaling 20 μg was added to the assay. The test compounds were diluted in drug buffer (0.5% BSA, 10% DMSO and TME) and then 25 μl were added to a deep well polypropylene plate. [3H]-SR141716A was diluted in a ligand buffer (0.5% BSA plus TME) and 25 μl were added to the plate. The plates were covered and placed in an incubator at 30°C for 60 minutes. At the end of the incubation period 250 μl of stop buffer (5% BSA plus TME) was added to the reaction plate. The plates were then harvested by Skatron onto GF/B filtermats presoaked in BSA (5 mg/ml) plus TME. Each filter was washed twice. The filters were dried overnight. In the morning the filters were counted on a Wallac Betaplate™ counter (available from PerkinElmer Life Sciences™, Boston, MA). CB-2 Receptor Binding Protocol Chinese hamster ovary-K1 (CHO-K1 ) cells transfected with CB-2 cDNA (obtained from Dr. Debra Kendall, University of Connecticut) were harvested in tissue preparation buffer (5 mM Tris-HCI buffer (pH = 7.4) containing 2 mM EDTA), polytroned at high speed and kept on ice for 15 minutes. The homogenate was then spun at 1 ,000X g for 5 minutes at 4°C. The supernatant was recovered and centrifuged at 100.000X G for 1 hour at 4°C. The pellet was then re-suspended in 25 ml of TME (25 mM Tris buffer (pH = 7.4) containing 5 mM MgCI2 and 1 mM EDTA) per brain used. A protein assay was performed and 200 μl of tissue totaling 10 μg was added to the assay. The test compounds were diluted in drug buffer (0.5% BSA, 10% DMSO, and 80.5% TME) and then 25 μl were added to the deep well polypropylene plate. [3H]-5-(1 ,1-Dimethyl-heptyl)-2-[5-hydroxy-2-(3-hydroxy- propyl)-cyclohexyl]-phenol was diluted in ligand buffer (0.5% BSA and 99.5% TME) and then 25 μl of the solution were added to each well at a concentration of 1 nM. A BCA protein assay was used to determine the appropriate tissue concentration and 200 μl of the tissue at the appropriate concentration was added to the plate. The plates were covered and placed in an incubator at 30°C for 60 minutes. At the end of the incubation period 250 μl of stop buffer (5% BSA plus TME) was added to the reaction plate. The plates were then harvested by Skatron format onto GF/B filtermats presoaked in BSA (5 mg/ml) plus TME. Each filter was washed twice. The filters were dried overnight. The filters were then counted on the Wallac Betaplate™ counter. CB-1 Receptor GTPγ [35S1 Binding Assay Membranes were prepared from CHO-K1 cells stably transfected with the human CB-1 receptor cDNA. Membranes were prepared from cells as described by Bass et al., in "Identification and characterization of novel somatostatin antagonists," Molecular Pharmacology. 50, 709-715 (1996). GTPγ [35S] binding assays were performed in a 96 well FlashPlate format in duplicate using 100 pM GTPγ[35S] and 10 μg membrane per well in assay buffer composed of 50 mM Tris HCI, pH 7.4, 3 mM MgCI2, pH 7.4, 10 mM MgCI2, 20 mM EGTA, 100 mM NaCl, 30 μM GDP, 0.1 % bovine serum albumin and the following protease inhibitors: 100 μg/ml bacitracin, 100 μg/ml benzamidine, 5 μg/ml aprotinin, 5 μg/ml leupeptin. The assay mix was then incubated with increasing concentrations of antagonist (10"1° M to 10"5 M) for 10 minutes and challenged with the cannabinoid agonist 5-(1 ,1- dimethyl-heptyl)-2-[5-hydroxy-2-(3-hydroxy-propyl)-cyclohexyl]-phenol (10 μM). Assays were performed at 30°C for one hour. The FlashPlates™ were then centrifuged at 2000Xg for 10 minutes. Stimulation of GTPγ[35S] binding was then quantified using a Wallac Microbeta. EC50 calculations were performed using Prism™ by Graphpad. Inverse agonism was measured in the absense of agonist using the protocol described above. CB-1 Receptor FLIPR-based Functional Assay Protocol CHO-K1 cells co-transfected with the human CB-1 receptor cDNA (obtained from Dr. Debra Kendall, University of Connecticut) and the promiscuous G-protein G16 were used for this assay. Cells were plated 48 hours in advance at 12500 cells per well on collagen coated 384 well black clear assay plates. Cells were incubated for one hour with 4μM Fluo-4 AM (Molecular Probes) in DMEM (Gibco) containing 2.5 mM probenicid and pluronic acid (.04%). The plates were then washed 3 times with HEPES- buffered saline (containing probenicid; 2.5 mM) to remove excess dye. After 20 min, the plates were added to the FLIPR individually and fluorescence levels were continuously monitored over an 80 s period. Compound additions were made simultaneously to all 384 wells after 20 s of baseline. Assays were performed in triplicate and 6 point concentration-response curves generated. Antagonist compounds were subsequently challenged with 3 μM WIN 55,212-2 (CB1 -R agonist). Data were analyzed using Graph Pad Prism. Detection of Inverse Agonists The following cyclic-AMP assay protocol using intact cells was used to determine inverse agonist activity. Cells were plated into a 96-well plate at a plating density of 10,000- 14,000 cells per well at a concentration of 100 μl per well. The plates were incubated for 24 hours in a 37°C incubator. The media was removed and media lacking serum (100 μl) was added. The plates were then incubated for 18 hours at 37°C. Serum free medium containing 1 mM IBMX was added to each well followed by 10 μl of test compound (1 :10 stock solution (25 mM compound in DMSO) into 50% DMSO/PBS) diluted 10X in PBS with 0.1 % BSA. After incubating for 20 minutes at 37°C, 2 μM of Forskolin was added and then incubated for an additional 20 minutes at 37°C. The media was removed, 100 μl of 0.01 N HCI was added and then incubated for 20 minutes at room temperature. Cell lysate (75 μl) along with 25 μl of assay buffer (supplied in FlashPlate™ cAMP assay kit available from NEN Life Science Products Boston, MA) into a Flashplate. cAMP standards and cAMP tracer were added following the kit's protocol. The flashplate was then incubated for 18 hours at 4°C. The content of the wells were aspirated and counted in a Scintillation counter. In Vivo Biological Assays Cannabinoid agoinists such as Δ9-tetrahydrocannabinol (Δ9-THC) and 5-(1 ,1-dimethyl-heptyl)-2-[5-hydroxy-2-(3-hydroxy-propyl)-cyclohexyl]-phenol have been shown to affect four characteristic behaviors in mice, collectively known as the Tetrad. For a description of these behaviors see: Smith, P.B., et al. in "The pharmacological activity of anandamide, a putative endogenous cannabinoid, in mice." J. Pharmacol. Exp. Ther.. 270(1 ), 219- 227 (1994) and Wiley, J., et al. in "Discriminative stimulus effects of anandamide in rats," Eur. J. Pharmacol.. 276(1-2), 49-54 (1995). Reversal of these activities in the Locomotor Activity, Catalepsy, Hypothermia, and Hot Plate assays described below provides a screen for in vivo activity of CB-1 receptor antagonists. All data is presented as % reversal from agonist alone using the following formula: (5-(1 ,1 -dimethyl-heptyl)-2-[5-hydroxy-2-(3-hydroxy-propyI)- cyclohexyl]-phenol/agonist - vehicle/agonist)/(vehicle/vehicle - vehicle/agonist). Negative numbers indicate a potentiation of the agonist activity or non-antagonist activity. Positive numbers indicate a reversal of activity for that particular test. Locomotor Activity Male ICR mice (n=6) (17-19 g, Charles River Laboratories, Inc., Wilmington, MA) were pre-treated with test compound (sc, po, ip, or icv). Fifteen minutes later, the mice were challenged with 5-(1 ,1-dimethyl-heptyl)- 2-[5-hydroxy-2-(3-hydroxy-propyl)-cyclohexyl]-phenol (sc administration). Twenty-five minutes after the agonist injection, the mice were placed in clear acrylic cages (431.8 cm x 20.9 cm x 20.3 cm) containing clean wood shavings. The subjects were allowed to explore surroundings for a total of about 5 minutes and the activity was recorded by infrared motion detectors (available from Coulbourn Instruments™, Allentown, PA) that were placed on top of the cages. The data was computer collected and expressed as "movement units." Catalepsy Male ICR mice (n=6,17-19 g upon arrival) were pre-treated with test compound (sc, po, ip or icv). Fifteen minutes later, the mice were challenged with 5-(1 ,1-dimethyl-heptyl)-2-[5-hydroxy-2-(3-hydroxy-propyl)- cydohexyl]-phenol (sc). Ninety minutes post injection, the mice were placed on a 6.5 cm steel ring attached to a ring stand at a height of about 12 inches. The ring was mounted in a horizontal orientation and the mouse was suspended in the gap of the ring with fore- and hind-paws gripping the perimeter. The duration that the mouse remained completely motionless (except for respiratory movements) was recorded over a 3-minute period. The data were presented as a percent immobility rating. The rating was calculated by dividing the number of seconds the mouse remains motionless by the total time of the observation period and multiplying the result by 100. A percent reversal from the agonist was then calculated. Hypothermia Male ICR mice (n=5) (17-19 g upon arrival) were pretreated with test compounds (sc, po, ip or icv). Fifteen minutes later, mice were challenged with the cannabinoid agonist 5-(1 ,1-dimethyl-heptyl)-2-[5-hydroxy-2-(3- hydroxy-propyl)-cy ohexyl]-phenol (sc). Sixty-five minutes post agonist injection, rectal body temperatures were taken. This was done by inserting a small thermostat probe approximately 2- 2.5 cm into the rectum. Temperatures were recorded to the nearest tenth of a degree Hot Plate Male ICR mice (n=7,17-19 g upon arrival) are pre-treated with test compounds (sc, po, ip or iv). Fifteen minutes later, mice were challenged with a cannabinoid agonist 5-(1 ,1-dimethyl-heptyl)-2-[5-hydroxy-2-(3- hydroxy-propyl)-cyclohexyl]-phenol (sc administration). Forty-five minutes later, each mouse was tested for reversal of analgesia using a standard hot plate meter (Columbus Instruments). The hot plate was 10" x 10" x 0.75" with a surrounding clear acrylic wall. Latency to kick, lick or flick hindpaw or jump from the platform was recorded to the nearest tenth of a second. The timer was experimenter activated and each test had a 40 second cut off. Data were presented as a percent reversal of the agonist induced analgesia. Food Intake The following screen was used to evaluate the efficacy of test compounds for inhibiting food intake in Sprague-Dawley rats after an overnight fast. Male Sprague-Dawley rats were obtained from Charles River Laboratories, Inc. (Wilmington, MA). The rats were individually housed and fed powdered chow. They were maintained on a 12 hour light/dark cycle and received food and water ad libitum. The animals were acclimated to the vivarium for a period of one week before testing was conducted. Testing was completed during the light portion of the cycle. To conduct the food intake efficacy screen, rats were transferred to individual test cages without food the afternoon prior to testing, and the rats were fasted overnight. After the overnight fast, rats were dosed the following morning with vehicle or test compounds. A known antagonist was dosed (3 mg/kg) as a positive control, and a control group received vehicle alone (no compound). The test compounds were dosed at ranges between 0.1 and 100 mg/kg depending upon the compound. The standard vehicle was 0.5% (w/v) methylcellulose in water and the standard route of administration was oral. However, different vehicles and routes of administration were used to accommodate various compounds when required. Food was provided to the rats 30 minutes after dosing and the Oxymax automated food intake system (Columbus Instruments, Columbus, Ohio) was started. Individual rat food intake was recorded continuously at 10-minute intervals for a period of two hours. When required, food intake was recorded manually using an electronic scale; food was weighed every 30 minutes after food was provided up to four hours after food was provided. Compound efficacy was determined by comparing the food intake pattern of compound-treated rats to vehicle and the standard positive control. Alcohol Intake The following protocol evaluates the effects of alcohol intake in alcohol preferring (P) female rats (bred at Indiana University) with an extensive drinking history. The following references provide detailed descriptions of P rats: Li ,T.-K., et al., "Indiana selection studies on alcohol related behaviors" in Development of Animal Models as Pharmacogenetic Tools (eds McClearn C. E., Deitrich R. A. and Erwin V. G.), Research Monograph 6, 171-192 (1981) NIAAA, ADAMHA, Rockville, MD; Lumeng, L, et al., "New strains of rats with alcohol preference and nonpreference" Alcohol And Aldehyde Metabolizing Systems, 3, Academic Press, New York, 537-544 (1977); and Lumeng, L, et al., "Different sensitivities to ethanol in alcohol-preferring and -nonpreferring rats," Pharmacol. Biochem Behav.. 16, 125-130 (1982). Female rats were given 2 hours of access to alcohol (10% v/v and water, 2-bottle choice) daily at the onset of the dark cycle. The rats were maintained on a reverse cycle to facilitate experimenter interactions. The animals were initially assigned to four groups equated for alcohol intakes: Group 1 - vehicle (n =8); Group 2 - positive control (e.g. 5.6 mg/kg AM251 ; n = 8); Group 3 - low dose test compound (n = 8); and Group 4 - high dose of test compound (n = 8). Test compounds were generally mixed into a vehicle of 30% (w/v) β-cyclodextrin in distilled water at a volume of 1-2 ml/kg. Vehicle injections were given to all groups for the first two days of the experiment. This was followed by 2 days of drug injections (to the appropriate groups) and a final day of vehicle injections. On the drug injection days, drugs were given sc 30 minutes prior to a 2-hour alcohol access period. Alcohol intake for all animals was measured during the test period and a comparison was made between drug and vehicle-treated animals to determine effects of the compounds on alcohol drinking behavior. Additional drinking studies were done utilizing female C57BI/6 mice (Charles River). Several studies have shown that this strain of mice will readily consume alcohol with little to no manipulation required (Middaugh et al., "Ethanol Consumption by C57BL/6 Mice: Influence of Gender and Procedural Variables" Alcohol. 17 (3), 175-183, 1999; Le et al., "Alcohol Consumption by C57BL/6, BALA/c, and DBA/2 Mice in a Limited Access Paradigm" Pharmacology Biochemisrtv and Behavior. 47, 375-378, 1994). Mice (17-19 g) were individually housed upon arrival and given unlimited access to powdered rat chow, water and a 10 % (w/v) alcohol solution. After 2-3 weeks of unlimited access, water was restricted for 2O hours and alcohol was restricted to only 2 hours access daily. This was done in a manner that the access period was the last 2 hours of the dark part of the light cycle. Once drinking behavior stabilized, testing commenced. Mice were considered stable when the average alcohol consumption for 3 days was ± 20% of the average for all 3 days. Day 1 of test consisted of all mice receiving vehicle injection (sc or ip). Thirty to 120 minutes post injection, access was given to alcohol and water. Alcohol consumption for that day was calculated (g/kg) and groups were assigned (n=7-10) so that all groups had similar alcohol intake. On day 2 and 3, mice were injected with vehicle or drug and the same protocol as the previous day was followed. Day 4 was wash out and no injections were given. Data was analyzed using repeated measures ANOVA. Change in water or alcohol consumption was compared back to vehicle for each day of the test. Positive results would be interpreted as a compound that was able to significantly reduce alcohol consumption while having no effect on water Oxygen Consumption Methods: Whole body oxygen consumption is measured using an indirect calorimeter (Oxymax from Columbus Instruments, Columbus, OH) in male Sprague Dawley rats. Rats (300-380 g body weight) are placed in the calorimeter chambers and the chambers are placed in activity monitors. These studies are done during the light cycle. Prior to the measurement of oxygen consumption, the rats are fed standard chow ad libitum. During the measurement of oxygen consumption, food is not available. Basal pre-dose oxygen consumption and ambulatory activity are measured every 10 minutes for 2.5 to 3 hours. At the end of the basal pre-dosing period, the chambers are opened and the animals are administered a single dose of compound (the usual dose range is 0.001 to 10 mg/kg) by oral gavage (or other route of administration as specified, i.e. s.c, i.p., i.v.). Drugs are prepared in methylcellulose, water or other specified vehicle (examples include PEG40O, 30% beta-cyclo dextran and propylene glycol). Oxygen consumption and ambulatory activity are measured every 10 minutes for an additional 1-6 hours post-dosing. The Oxymax calorimeter software calculates the oxygen consumption (ml/kg/h) based on the flow rate of air through the chambers and difference in oxygen content at inlet and output ports. The activity monitors have 15 infrared light beams spaced one inch apart on each axis, ambulatory activity is recorded when two consecutive beams are broken and the results are recorded as counts. Resting oxygen consumption, during pre- and post-dosing, is calculated by averaging the 10-min 02 consumption values, excluding periods of high ambulatory activity (ambulatory activity count > 100) and excluding the first 5 values of the pre-dose period and the first value from the post-dose period. Change in oxygen consumption is reported as percent and is calculated by dividing the post-dosing resting oxygen consumption by the pre-dose oxygen consumption *100. Experiments will typically be done with n = 4-6 rats and results reported are mean +/- SEM. Interpretation: An increase in oxygen consumption of >10% is considered a positive result. Historically, vehicle-treated rats have no change in oxygen consumption from pre-dose basal.

Claims

CLAIMS What is claimed is: 1. A compound of Formula (I)
Figure imgf000075_0001
(I) wherein R )1 is an aryl optionally substituted with one or more substituents, or a heteroaryl optionally substituted with one or more substituents; R2 is aryl optionally substituted with one or more substituents, heteroaryl optionally substituted with one or more substituents, -CH=CH- R2a, or -CH2CH2-R2a, where R2a is hydrogen or a chemical moiety selected from (Cι-C8)alkyl, 3- to 8-membered partially or fully saturated carbocyclic ring(s), 3- to 6-membered partially or fully saturated heterocycle, aryl, heteroaryl, where said chemical moiety is optionally substituted with one or more substituents; R3 is hydrogen, (Cι-C )alkyl, halo-substituted (Cι-C4)alkyl, or (Ci- C4)alkoxy, or R3 taken together with R4 forms a nitrogen containing 5- to 6- membered partially or fully saturated heterocyclic ring optionally substituted with one or more substituents; and R4 is a chemical moiety selected from the group consisting of (C-i- Csjalkyl, aryl, heteroaryl, aryl(Cι-C4)alkyl, a 3- to 8-membered partially or fully saturated carbocyclic ring(s), heteroaryl(Cι-C3)alkyl, 5-6 membered lactone, 5- to 6-membered lactam, and a 3- to 8-membered partially or fully saturated heterocycle, where said chemical moiety is optionally substituted with one or more substituents, or R4 taken together with R3 forms a nitrogen containing 5- to 6- membered partially or fully saturated heterocyclic ring optionally substituted with one or more substituents; a pharmaceutically acceptable salt thereof, a prodrug of said compound or said salt, or a solvate or hydrate of said compound, said salt or said prodrug.
2. The compound of Claim 1 wherein R4 is a chemical moiety selected from the group consisting of (Cι-C8)alkyl, aryl(Cι-C4)aIkyl, 3- to 8- membered partially or fully saturated carbocyclic ring(s), and 3- to 8- membered partially or fully saturated heterocycle, where said chemical moiety is optionally substituted with one or more substituents; a pharmaceutically acceptable salt thereof, or a solvate or hydrate of said compound or said salt.
3. The compound of Claim 2 wherein R3 is hydrogen, methyl or trifluoromethyl; a pharmaceutically acceptable salt thereof, or a solvate or hydrate of said compound or said salt.
4. The compound of Claim 2 wherein R4 is (CrC8)alkyl, fluoro- substituted (Cι-C8)alkyl, cyclopentyl, cyclohexyl, piperidin-1-yl, pyrrolidin-1-yl, or morpholin-1-yl; a pharmaceutically acceptable salt thereof, or a solvate or hydrate of said compound or said salt.
5. The compound of Claim 3 wherein R4 is (C-i-CsJalkyl, fluoro- substituted (Cι-C8)alkyl, cyclopentyl, cyclohexyl, piperidin-1-yl, pyrrolidin-1-yl, or morpholin-1-yl; a pharmaceutically acceptable salt thereof, or a solvate or hydrate of said compound or said salt.
6. The compound of Claim 1 , 2, 3, 4 or 5 wherein R1 and R2 are each independently a substituted phenyl; a pharmaceutically acceptable salt thereof, or a solvate or hydrate of said compound or said salt. '
7. The compound of Claim 6 wherein R1 and R2 are each independently a phenyl substituted with one to three substituents independently selected from the group consisting of halo, (CrC4)alkoxy, (C-i- C4)alkyl, halo-substituted (Cι-C4)alkyl, and cyano; a pharmaceutically acceptable salt thereof, or a solvate or hydrate of said compound or said salt.
8. The compound of Claim 7 wherein R1 is 2-cyanophenyl, 2- chlorophenyl, 2-fluorophenyl, 2,4-dichlorophenyl, 2-fluoro-4-chlorophenyl, 2- chloro-4-fluorophenyl, 2-methylphenyl or 2,4-difluorophenyl; and R2 is 4-chlorophenyl, 4-cyanophenyl, 4-ethylphenyl, 4-isopropylphenyl, 4-ethoxyphenyl, 4-isopropoxyphenyl, 4-trifluoromethylphenyl, or 4- fluorophenyl; a pharmaceutically acceptable salt thereof, or a solvate or hydrate of said compound or said salt.
9. A compound of Formula (II)
Figure imgf000077_0001
(ii) wherein R1a, R1b, R2b, and R2c are each independently halo, (Cι-C4)alkoxy, (C C )alkyl, halo-substituted (Cι-C4)alkyl, or cyano; n and m are each independently 0, 1 or 2; and R3 is hydrogen, (Cι-C4)alkyl, halo-substituted (Cι-C4)alkyl, or (C-i-
C )alkoxy, or R3 taken together with R4 forms a nitrogen containing 5- to 6- membered partially or fully saturated heterocyclic ring optionally substituted with one or more substituents; and R4 is a chemical moiety selected from the group consisting of (C-i-
C8)alkyl, aryl, heteroaryl, aryl(Cι -Chalky!, a 3- to 8-membered partially or fully saturated carbocyclic ring(s), heteroaryl(Cι-C3)alkyl, 5-6 membered lactone, 5- to 6-membered lactam, and a 3- to 8-membered partially or fully saturated heterocycle, where said chemical moiety is optionally substituted with one or more substituents, or R4 taken together with R3 forms a nitrogen containing 5- to 6- membered partially or fully saturated heterocyclic ring optionally substituted with one or more substituents, a pharmaceutically acceptable salt thereof, a prodrug of the compound or the salt, or a solvate or hydrate of the compound, the salt or the prodrug.
10. The compound of Claim 9 wherein R3 is hydrogen, methyl or trifluoromethyl; a pharmaceutically acceptable salt thereof, or a solvate or hydrate of said compound or said salt.
11. The compound of Claim 9 wherein R4 is a chemical moiety selected from the group consisting of (Cι-C8)alkyl, aryl(Cι-C4)alkyl, 3- to 8- membered partially or fully saturated carbocyclic ring(s), and 3- to 8- membered partially or fully saturated heterocycle where said chemical moiety is optionally substituted with one or more substituents; a pharmaceutically acceptable salt thereof, or a solvate or hydrate of said compound or said salt.
12. The compound of Claim 10 wherein R4 is a chemical moiety selected from the group consisting of (Ci-Csjaikyl, aryl(Cι-C4)alkyl, 3- to 8- membered partially or fully saturated carbocyclic ring(s), and 3- to 8- membered partially or fully saturated heterocycle where said chemical moiety is optionally substituted with one or more substituents; a pharmaceutically acceptable salt thereof, or a solvate or hydrate of said compound or said salt.
13. The compound of Claim 11 wherein R4 is (CrC8)alkyl, fluoro- substituted (C-i-CsJalkyl, cyclopentyl, cyclohexyl, piperidin-1-yl, pyrrolidin-1-yl, or morpholin-1 -yl; a pharmaceutically acceptable salt thereof, or a solvate or hydrate of said compound or said salt.
14. The compound of Claim 12 wherein R4 is (CrC8)alkyl, fluoro- substituted (Cι-C8)alkyl, cyclopentyl, cyclohexyl, piperidin-1-yl, pyrrolidin-1-yl, or morpholin-1 -yl; a pharmaceutically acceptable salt thereof, or a solvate or hydrate of said compound or said salt.
15. The compound of Claim 9, 10, 11 , 12, 13, or 14 wherein R1a is chloro, fluoro, cyano, or methyl; R1b is chloro or fluoro; R2b is chloro, fluoro, (CrC )alkyl, trifluoromethyl, (C C4)alkoxy, or cyano; m is 0 or 1 ; and n is 0; a pharmaceutically acceptable salt thereof, or a solvate or hydrate of said compound or said salt.
16. A pharmaceutical composition comprising (1 ) a compound of any one of the preceding Claims, or a solvate or hydrate of said compound or said salt; and (2) a pharmaceutically acceptable excipient, diluent, or carrier.
17. The composition of Claim 17 further comprising at least one additional pharmaceutical agent selected from a nicotine receptor partial agonist, an opioid antagonist, a dopaminergic agent, an attention deficit disorder agent, or an anti-obesity agent.
18. A method for treating a disease, condition or disorder which is modulated by a cannabinoid receptor antagonist in animals comprising the step of administering to an animal in need of such treatment a therapeutically effective amount of a compound of Claim 1 or 9, a pharmaceutically acceptable salt thereof, a prodrug of the compound or the salt, or a solvate or hydrate of the compound, the salt or the prodrug.
19. The method of Claim 18 wherein said disease, condition or disorder modulated by a cannabinoid receptor antagonist is selected from the group consisting of eating disorders, weight loss or control, obesity, bulimia, depression, atypical depression, bipolar disorders, psychoses, schizophrenia, behavioral addictions, suppression of reward-related behaviors, alcoholism, tobacco abuse, dementia, seizure disorders, epilepsy, attention deficit disorder, Parkinson's disease, inflammation, gastrointestinal disorders, and type II diabetes.
20. The use of a compound of Claim 1 or 9 in the manufacture of a medicament for treating a disease, condition or disorder which is modulated by a cannabinoid receptor antagonist.
PCT/IB2004/004019 2003-12-16 2004-12-06 Bicyclic imidazolyl pyrimidin-4-one cannabinoid receptor ligands and uses thereof WO2005061505A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US53001203P 2003-12-16 2003-12-16
US60/530,012 2003-12-16

Publications (1)

Publication Number Publication Date
WO2005061505A1 true WO2005061505A1 (en) 2005-07-07

Family

ID=34710151

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2004/004019 WO2005061505A1 (en) 2003-12-16 2004-12-06 Bicyclic imidazolyl pyrimidin-4-one cannabinoid receptor ligands and uses thereof

Country Status (1)

Country Link
WO (1) WO2005061505A1 (en)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
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
US7728141B2 (en) 2003-11-04 2010-06-01 Merck Sharp & Dohme Corp. Substituted naphthyridinone derivatives
US7790730B2 (en) * 2004-07-27 2010-09-07 Gilead Sciences, Inc. Imidazo[4,5-d]pyrimidines, their uses and methods of preparation
WO2013076182A1 (en) * 2011-11-25 2013-05-30 F. Hoffmann-La Roche Ag [1, 2, 3]triazolo [4, 5 -d] pyrimidine derivatives as agonists of the cannabinoid receptor 2 agonists

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040116328A1 (en) * 2002-06-06 2004-06-17 Eisai Co., Ltd. Condensed imidazole derivatives

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040116328A1 (en) * 2002-06-06 2004-06-17 Eisai Co., Ltd. Condensed imidazole derivatives

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
BARTH F: "Cannabinoid receptor agonists and antagonists", EXPERT OPINION ON THERAPEUTIC PATENTS, ASHLEY PUBLICATIONS, GB, vol. 8, no. 3, March 1998 (1998-03-01), pages 301 - 313, XP002150850, ISSN: 1354-3776 *
GOYA P ET AL: "RECENT ADVANCES IN CANNABINOID RECEPTOR AGONISTS AND ANTAGONISTS", EXPERT OPINION ON THERAPEUTIC PATENTS, ASHLEY PUBLICATIONS, GB, vol. 10, no. 10, 2000, pages 1529 - 1538, XP001036546, ISSN: 1354-3776 *
STOIT A R ET AL: "DESIGN, SYNTHESIS AND BIOLOGICAL ACTIVITY OF RIGID CANNABINOID CB RECEPTOR ANTAGONISTS", CHEMICAL AND PHARMACEUTICAL BULLETIN, PHARMACEUTICAL SOCIETY OF JAPAN. TOKYO, JP, vol. 50, no. 8, August 2002 (2002-08-01), pages 1109 - 1113, XP001181236, ISSN: 0009-2363 *

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7728141B2 (en) 2003-11-04 2010-06-01 Merck Sharp & Dohme Corp. Substituted naphthyridinone derivatives
US7790730B2 (en) * 2004-07-27 2010-09-07 Gilead Sciences, Inc. Imidazo[4,5-d]pyrimidines, their uses and methods of preparation
AU2005287407B2 (en) * 2004-07-27 2011-11-17 Gilead Sciences, Inc. Imidazo(4,5-d)pyrimidines, their uses and methods of preparation
KR101238430B1 (en) 2004-07-27 2013-02-28 길리애드 사이언시즈, 인코포레이티드 Imidazo[4,5-d]pyrimidines, their uses and methods of preparation
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
US8722691B2 (en) 2006-03-30 2014-05-13 Irm Llc Azolopyrimidines as inhibitors of cannabinoid 1 activity
WO2013076182A1 (en) * 2011-11-25 2013-05-30 F. Hoffmann-La Roche Ag [1, 2, 3]triazolo [4, 5 -d] pyrimidine derivatives as agonists of the cannabinoid receptor 2 agonists
US9067943B2 (en) 2011-11-25 2015-06-30 Hoffmann-La Roche Inc. [1,2,3]triazolo[4,5-D]pyrimidine derivatives
EA024108B1 (en) * 2011-11-25 2016-08-31 Ф.Хоффманн-Ля Рош Аг [1,2,3]TRIAZOLO[4,5-d]PYRIMIDINE DERIVATIVES AS CANNABINOID RECEPTOR 2 AGONISTS

Similar Documents

Publication Publication Date Title
EP1622903B1 (en) Cannabinoid receptor ligands and uses thereof
US7354929B2 (en) Cannabinoid receptor ligands and uses thereof
US7232823B2 (en) Cannabinoid receptor ligands and uses thereof
US7145012B2 (en) Cannabinoid receptor ligands and uses thereof
US7329658B2 (en) Cannabinoid receptor ligands and uses thereof
US7176210B2 (en) Cannabinoid receptor ligands and uses thereof
US20070027133A1 (en) Bicyclic pyrazolyl and imidazolyl compounds and uses thereof
WO2005103052A1 (en) Pyrazolo [1,5-a] pyrimidin-7-one compounds and uses thereof
US20050026983A1 (en) Imidazole compounds and uses thereof
WO2005061507A1 (en) Bicyclic pyrazol-4-one cannabinoid receptor ligands and uses thereof
WO2005061505A1 (en) Bicyclic imidazolyl pyrimidin-4-one cannabinoid receptor ligands and uses thereof
WO2005049615A1 (en) Pyrazolo`1,5-a!`1,3,5! triazin -4-one derivatives as cb1 receptor antagonists
WO2005061506A1 (en) Bicyclic pyrazolyl and imidazolyl compounds as cannabinoid receptor ligands and uses thereof
WO2005061504A1 (en) Bicyclic pyridazinone cannabinoid receptor ligands and uses thereof
US20070213334A1 (en) Cannabinoid receptor ligands and uses thereof

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

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

Ref country code: DE

WWW Wipo information: withdrawn in national office

Country of ref document: DE

122 Ep: pct application non-entry in european phase